151
|
Mester-Tonczar J, Winkler J, Einzinger P, Hasimbegovic E, Kastner N, Lukovic D, Zlabinger K, Spannbauer A, Traxler D, Batkai S, Thum T, Gyöngyösi M. Association between Circular RNA CDR1as and Post-Infarction Cardiac Function in Pig Ischemic Heart Failure: Influence of the Anti-Fibrotic Natural Compounds Bufalin and Lycorine. Biomolecules 2020; 10:E1180. [PMID: 32823854 PMCID: PMC7463784 DOI: 10.3390/biom10081180] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
Anti-fibrotic therapies are of increasing interest to combat cardiac remodeling and heart failure progression. Recently, anti-fibrotic circular RNAs (circRNAs) have been identified in human and rodent cardiac tissue. In vivo (rodent) experiments proved cardiac anti-fibrotic effects of the natural compounds bufalin and lycorine by downregulating miRNA-671-5p, associated with a theoretic increase in the tissue level of circRNA CDR1as. Accordingly, we hypothesized that both anti-fibrotic drugs may inhibit focal myocardial fibrosis of the remodeled left ventricle (LV) also in a translational large animal model of heart failure (HF). Domestic pigs were repeatedly treated with subcutaneous injections of either bufalin, lycorine, or saline, (n = 5/group) between days 7-21 post acute myocardial infarction (AMI). At the 2-month follow-up, both bufalin and lycorine led to significantly reduced cardiac fibrosis. Bufalin treatment additionally led to smaller end-diastolic volumes, higher LV ejection fraction (EF), and increased expression of CDR1as of the AMI region. Elevated tissue levels of the circRNA CDR1as in the AMI region of the pig heart correlated significantly with LV and right ventricular EF, LV stroke volume, and negatively with infarct size. In conclusion, we successfully identified the circRNA CDR1as in pig hearts and show a significant association with improved LV and RV function by anti-fibrotic therapies in a translational animal model of HF.
Collapse
Affiliation(s)
- Julia Mester-Tonczar
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Johannes Winkler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Patrick Einzinger
- Institute of Information Systems Engineering, Research Unit of Information and Software Engineering, Vienna University of Technology, 1040 Vienna, Austria;
| | - Ena Hasimbegovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Nina Kastner
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Dominika Lukovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Katrin Zlabinger
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Andreas Spannbauer
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Denise Traxler
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| | - Sandor Batkai
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (S.B.); (T.T.)
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, 30625 Hannover, Germany; (S.B.); (T.T.)
- REBIRTH Center of Translational Regenerative Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, 1090 Vienna, Austria; (J.M.-T.); (J.W.); (E.H.); (N.K.); (D.L.); (K.Z.); (A.S.); (D.T.)
| |
Collapse
|
152
|
Groenendyk J, Wang Q, Wagg C, Lee D, Robinson A, Barr A, Light PE, Lopaschuk GD, Agellon LB, Michalak M. Selective enhancement of cardiomyocyte efficiency results in a pernicious heart condition. PLoS One 2020; 15:e0236457. [PMID: 32790682 PMCID: PMC7425937 DOI: 10.1371/journal.pone.0236457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 07/05/2020] [Indexed: 12/24/2022] Open
Abstract
Transgenic mice with selective induction of calreticulin transgene expression in cardiomyocytes (CardiacCRT+) were analyzed. CardiacCRT+ cardiomyocytes showed increased contractility and Ca2+ transients. Yet, in vivo assessment of cardiac performance, and ischemic tolerance of CardiacCRT+ mice demonstrated right ventricle dilation and reduced cardiac output, increased QT interval and decreased P amplitude. Paradoxically, ex vivo working hearts from CardiacCRT+ mice showed enhanced ischemic cardio-protection and cardiac efficiency. Under aerobic conditions, CardiacCRT+ hearts showed less efficient cardiac function than sham control hearts due to an increased ATP production from glycolysis relative to glucose oxidation. During reperfusion, this inefficiency was reversed, with CardiacCRT+ hearts exhibiting better functional recovery and increased cardiac efficiency compared to sham control hearts. On the other hand, mechanical stretching of isolated cardiac fibroblasts activated the IRE1α branch of the unfolded protein response pathway as well as induction of Col1A2 and TGFβ gene expression ex vivo, which were all suppressed by tauroursodeoxycholic acid.
Collapse
Affiliation(s)
- Jody Groenendyk
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Qian Wang
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Cory Wagg
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Dukgyu Lee
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, Canada
| | - Alison Robinson
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Amy Barr
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, Canada
| | - Peter E. Light
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
| | - Gary D. Lopaschuk
- Department of Pharmacology, University of Alberta, Edmonton, Alberta, Canada
- Department of Pediatrics, University of Alberta, Edmonton, Alberta, Canada
| | - Luis B. Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, Quebec, Canada
- * E-mail: (MM); (LBA)
| | - Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
- * E-mail: (MM); (LBA)
| |
Collapse
|
153
|
Park SK, Oh CM, Kang JG, Jung JY. Association of Sugar-Sweetened Carbonated Beverage with the Alteration in Left Ventricular Structure and Diastolic Function. J Am Coll Nutr 2020; 40:496-501. [PMID: 32779979 DOI: 10.1080/07315724.2020.1800534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND High consumption of sugar-sweetened carbonated beverage (SSCB) has been associated with multiple metabolic risk factors for cardiovascular disease. However, published data is scarce regarding the influence of SSCB consumption on left ventricular (LV) structure and diastolic function. The present study is to investigate the association of SSCB consumption with alteration in LV structure and diastolic function. METHOD Study subjects were 46,417 Koreans who received echocardiography as an item of health checkup. They were categorized into 4 groups by SSCB consumption based on one serving dose (200 ml) with never/almost never, <1 serving/week, 1 ≤ serving/week <3 and ≥3 serving/week. Multivariate logistic regression analysis was used in calculating adjusted odd ratio (OR) and 95% confidence interval (CI) (adjusted OR [95% CI]) for left ventricular hypertrophy, increased relative wall thickness (IRWT) and impaired LV relaxation with each group (reference: never/almost never consumption). Subgroup analysis was conducted by age of ≥40 and <40. RESULT Compared with never/almost never consumption, SSCB consumption ≥3 serving/week had the higher mean levels in body mass index, blood pressure and triglyceride despite of younger age. In fully adjusted analysis, SSCB consumption ≥3 serving/week was associated with IRWT (1.14 [1.02-1.27]) and impaired LV relaxation (1.23 [1.08-1.41]). This association was reinforced in age subgroup ≥40 years, but not statistically significant in age subgroup <40 years. CONCLUSION SSCB consumption ≥3 serving/week was associated with the increased probability of IRWT and impaired LV relaxation.
Collapse
Affiliation(s)
- Sung Keun Park
- Center for Cohort Studies, Total Healthcare Center, School of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul, Korea
| | - Chang-Mo Oh
- Departments of Preventive Medicine, School of Medicine, Kyung Hee University, Seoul, Korea
| | - Jeong Gyu Kang
- Total Healthcare Center, School of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul, Korea
| | - Ju Young Jung
- Total Healthcare Center, School of Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University, Seoul, Korea
| |
Collapse
|
154
|
Wesseling M, de Poel JH, de Jager SC. Growth differentiation factor 15 in adverse cardiac remodelling: from biomarker to causal player. ESC Heart Fail 2020; 7:1488-1501. [PMID: 32424982 PMCID: PMC7373942 DOI: 10.1002/ehf2.12728] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 03/06/2020] [Accepted: 04/03/2020] [Indexed: 12/13/2022] Open
Abstract
Heart failure is a growing health issue as a negative consequence of improved survival upon myocardial infarction, unhealthy lifestyle, and the ageing of our population. The large and complex pathology underlying heart failure makes diagnosis and especially treatment very difficult. There is an urgent demand for discriminative biomarkers to aid disease management of heart failure. Studying cellular pathways and pathophysiological mechanisms contributing to disease initiation and progression is crucial for understanding the disease process and will aid to identification of novel biomarkers and potential therapeutic targets. Growth differentiation factor 15 (GDF15) is a proven valuable biomarker for different pathologies, including cancer, type 2 diabetes, and cardiovascular diseases. Although the prognostic value of GDF15 in heart failure is robust, the biological function of GDF15 in adverse cardiac remodelling is not fully understood. GDF15 is a distant member of the transforming growth factor-β family and involved in various biological processes including inflammation, cell cycle, and apoptosis. However, more research is suggesting a role in fibrosis, hypertrophy, and endothelial dysfunction. As GDF15 is a pleiotropic protein, elucidating the exact role of GDF15 in complex disease processes has proven to be a challenge. In this review, we provide an overview of the role GDF15 plays in various intracellular and extracellular processes underlying heart failure, and we touch upon crucial points that need consideration before GDF15 can be integrated as a biomarker in standard care or when considering GDF15 for therapeutic intervention.
Collapse
Affiliation(s)
- Marian Wesseling
- Laboratory for Experimental CardiologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
- Laboratory for Clinical Chemistry and HematologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Julius H.C. de Poel
- Laboratory for Experimental CardiologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
| | - Saskia C.A. de Jager
- Laboratory for Experimental CardiologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
- Laboratory for Translational ImmunologyUniversity Medical Centre UtrechtUtrechtThe Netherlands
| |
Collapse
|
155
|
Rabe E, Partsch H, Morrison N, Meissner MH, Mosti G, Lattimer CR, Carpentier PH, Gaillard S, Jünger M, Urbanek T, Hafner J, Patel M, Wu S, Caprini J, Lurie F, Hirsch T. Risks and contraindications of medical compression treatment - A critical reappraisal. An international consensus statement. Phlebology 2020; 35:447-460. [PMID: 32122269 PMCID: PMC7383414 DOI: 10.1177/0268355520909066] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVES Medical compression therapy is used for non-invasive treatment of venous and lymphatic diseases. Medical compression therapy-associated adverse events and contraindications have been reported, although some contraindications are theoretically based. This consensus statement provides recommendations on medical compression therapy risks and contraindications. METHODS A systematic literature search of medical compression therapy publications reporting adverse events up until November 2017 was performed. A consensus panel comprising 15 international experts critically reviewed the publications and formulated the recommendations. RESULTS Sixty-two publications reporting medical compression therapy adverse events were identified. The consensus panel issued 21 recommendations on medical compression therapy contraindications and adverse event risk mitigation, in addition to reviewing medical compression therapy use in borderline indications. The most frequently reported non-severe medical compression therapy-associated adverse events included skin irritation, discomfort and pain. Very rare but severe adverse events, including soft tissue and nerve injury, were also identified. CONCLUSION This consensus statement summarises published medical compression therapy-associated adverse events and contraindications, and provides guidance on medical compression therapy. Severe medical compression therapy-associated adverse events are very rarely encountered if compression is used correctly and contraindications are considered.
Collapse
Affiliation(s)
- Eberhard Rabe
- Department of Dermatology, University of Bonn, Bonn, Germany
| | - Hugo Partsch
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Mark H Meissner
- Department of Surgery, Harborview Medical Center, Seattle, WA, USA
| | - Giovanni Mosti
- Angiology Department, Clinica MD Barbantini, Lucca, Italy
| | - Christopher R Lattimer
- Josef Pflug Vascular Laboratory, West London Vascular and Interventional Centre, Ealing Hospital & Imperial College, London, UK
| | - Patrick H Carpentier
- Centre de Recherche Universitaire de La Léchère, Equipe THEMAS, Université Joseph Fourier, Grenoble, France
| | | | - Michael Jünger
- Department of Dermatology, University of Greifswald, Greifswald, Germany
| | - Tomasz Urbanek
- Department of General Surgery, Vascular Surgery, Angiology and Phlebology, Medical University of Silesia, Katowice, Poland
| | - Juerg Hafner
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland
| | - Malay Patel
- First Choice Vascular, Swastik Society, Ahmedabad, India
| | - Stephanie Wu
- Department of Podiatric Surgery & Applied Biomechanics, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Joseph Caprini
- Department of Vascular Surgery, The University of Chicago, Chicago, IL, USA
| | - Fedor Lurie
- Department of Vascular Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Tobias Hirsch
- Practice for Internal Medicine and Vascular Diseases, Halle, Germany
| |
Collapse
|
156
|
Impact of Modality and Intensity of Early Exercise Training on Ventricular Remodeling after Myocardial Infarction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5041791. [PMID: 32765807 PMCID: PMC7387991 DOI: 10.1155/2020/5041791] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 11/18/2022]
Abstract
The objective of this study was to analyze the impact of different modalities and intensities of exercise training on cardiac remodeling started early after experimental myocardial infarction (MI). Male Wistar rats, weighing 200-250 g, were subjected to experimental MI. After 5 days, the animals were allocated into three experimental groups and observed for three months: S (sedentary control animals), C (animals subjected to continuous low-intensity training), and HIT (animals subjected to high-intensity interval training). Low-intensity exercise training was performed at a treadmill speed corresponding to 40% VO2 max, which was kept unchanged throughout the entire session (i.e., continuous low-intensity training). High-intensity interval training was performed in such a way that rats run during 3 min at 60% VO2 max, followed by 4-minute intervals at 85% VO2 max (i.e., high-intensity interval training). After the follow-up period, we studied hypertrophy and ventricular geometry, functional alterations in vivo and in vitro, oxidative stress, apoptosis, and cardiac energetic metabolism. Our data showed that both high-intensity interval and continuous low-intensity modalities improved cardiac energetic metabolism variables in comparison with sedentary infarcted animals. In addition, high-intensity interval training decreased cardiac oxidative stress, associated with improved diastolic function. On the other hand, the continuous low-intensity group showed impairment of cardiac function. Therefore, altogether, our data suggest that high-intensity interval training could be the best modality for early physical exercise after MI and should be better studied in this clinical scenario.
Collapse
|
157
|
Assessment of Cardiac Remodeling-A Chance for Novel Cardiac Biomarkers? J Clin Med 2020; 9:jcm9072087. [PMID: 32635167 PMCID: PMC7408733 DOI: 10.3390/jcm9072087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 06/28/2020] [Accepted: 06/29/2020] [Indexed: 01/31/2023] Open
|
158
|
Kumar M, Haghighi K, Kranias EG, Sadayappan S. Phosphorylation of cardiac myosin-binding protein-C contributes to calcium homeostasis. J Biol Chem 2020; 295:11275-11291. [PMID: 32554466 DOI: 10.1074/jbc.ra120.013296] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/17/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiac myosin-binding protein-C (cMyBP-C) is highly phosphorylated under basal conditions. However, its phosphorylation level is decreased in individuals with heart failure. The necessity of cMyBP-C phosphorylation for proper contractile function is well-established, but the physiological and pathological consequences of decreased cMyBP-C phosphorylation in the heart are not clear. Herein, using intact adult cardiomyocytes from mouse models expressing phospho-ablated (AAA) and phosphomimetic (DDD) cMyBP-C as well as controls, we found that cMyBP-C dephosphorylation is sufficient to reduce contractile parameters and calcium kinetics associated with prolonged decay time of the calcium transient and increased diastolic calcium levels. Isoproterenol stimulation reversed the depressive contractile and Ca2+-kinetic parameters. Moreover, caffeine-induced calcium release yielded no difference between AAA/DDD and controls in calcium content of the sarcoplasmic reticulum. On the other hand, sodium-calcium exchanger function and phosphorylation levels of calcium-handling proteins were significantly decreased in AAA hearts compared with controls. Stress conditions caused increases in both spontaneous aftercontractions in AAA cardiomyocytes and the incidence of arrhythmias in vivo compared with the controls. Treatment with omecamtiv mecarbil, a positive cardiac inotropic drug, rescued the contractile deficit in AAA cardiomyocytes, but not the calcium-handling abnormalities. These findings indicate a cascade effect whereby cMyBP-C dephosphorylation causes contractile defects, which then lead to calcium-cycling abnormalities, resulting in aftercontractions and increased incidence of cardiac arrhythmias under stress conditions. We conclude that improvement of contractile deficits alone without improving calcium handling may be insufficient for effective management of heart failure.
Collapse
Affiliation(s)
- Mohit Kumar
- Heart, Lung, and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA.,Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kobra Haghighi
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Evangelia G Kranias
- Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| | - Sakthivel Sadayappan
- Heart, Lung, and Vascular Institute, Division of Cardiovascular Health and Disease, Department of Internal Medicine, University of Cincinnati, Cincinnati, Ohio, USA .,Department of Pharmacology and Systems Physiology, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
159
|
Guevorkian AG. The effect of hypothalamic peptides, neurohormone C and proline-rich peptide-1on the Ca 2+-handling system in heartin pathophysiological conditions. Heliyon 2020; 6:e04360. [PMID: 32637717 PMCID: PMC7330072 DOI: 10.1016/j.heliyon.2020.e04360] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 05/26/2020] [Accepted: 06/26/2020] [Indexed: 11/21/2022] Open
Abstract
Atthe Institute of Biochemistry named after H. Buniatyan we discovered and studied hypothalamic peptides with coronary dilatory and antioxidant activities:neurohormone C (NC) and proline-rich peptide-1 (PRP-1). Both NC and PRP-1 exhibit cardioprotective effects, in part by restoring the calcium affinity for calcium-binding membrane proteins in cardiomyocytes. This affinity is diminished in the sarcoplasmic reticulum and mitochondriawith myocardial damage, heart failure, pancreatic necrosis and crush syndrome caused by isoproterenol. The peptides can also destroy the four detected toxic peptides and myocardial depressant factor, and protect against ischemia-reperfusion injury. Further studies of these peptides may be promising for the treatment of patients at high risk of cardiovascular disease, regardless of pathology.
Collapse
Affiliation(s)
- Artashes G. Guevorkian
- Department of Biochemistry, Yerevan State Medical University after MkhitarHeratsi, 2 Koryun St., Yerevan 0025, Armenia
| |
Collapse
|
160
|
Pereira BLB, Rodrigue A, Arruda FCDO, Bachiega TF, Lourenço MAM, Correa CR, Azevedo PS, Polegato BF, Okoshi K, Fernandes AAH, de Paiva SAR, Zornoff LAM, Power KA, Minicucci MF. Spondias mombin L. attenuates ventricular remodelling after myocardial infarction associated with oxidative stress and inflammatory modulation. J Cell Mol Med 2020; 24:7862-7872. [PMID: 32468694 PMCID: PMC7348186 DOI: 10.1111/jcmm.15419] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 05/02/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
The objective of this study was to evaluate Spondias mombin L. (SM) pulp and its influence on cardiac remodelling after myocardial infarction (MI). Male Wistar rats were assigned to four groups: a sham group (animals underwent simulated surgery) that received standard chow (S; n = 20), an infarcted group that received standard chow (MI; n = 24), an infarcted group supplemented with 100 mg of SM/kg bodyweight/d, (MIS100; n = 23) and an infarcted group supplemented with 250 mg of SM/kg bodyweight/d (MIS250; n = 22). After 3 months of treatment, morphological, functional and biochemical analyses were performed. MI induced structural and functional changes in the left ventricle with worsening systolic and diastolic function, and SM supplementation at different doses did not influence these variables as analysed by echocardiography and an isolated heart study (P > .05). However, SM supplementation attenuated cardiac remodelling after MI, reducing fibrosis (P = .047) and hypertrophy (P = .006). Biomarkers of oxidative stress, inflammatory processes and energy metabolism were further investigated in the myocardial tissue. SM supplementation improved the efficiency of energy metabolism and decreased lipid hydroperoxide in the myocardium [group S (n = 8): 267.26 ± 20.7; group MI (n = 8): 330.14 ± 47.3; group MIS100 (n = 8): 313.8 ± 46.2; group MIS250: 294.3 ± 38.0 nmol/mg tissue; P = .032], as well as decreased the activation of the inflammatory pathway after MI. In conclusion, SM supplementation attenuated cardiac remodelling processes after MI. We also found that energy metabolism, oxidative stress and inflammation are associated with this effect. In addition, SM supplementation at the highest dose is more effective.
Collapse
Affiliation(s)
| | - Alexane Rodrigue
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | | | - Tatiana Fernanda Bachiega
- Internal Medicine Department, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | | | - Camila Renata Correa
- Internal Medicine Department, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Paula Schmidt Azevedo
- Internal Medicine Department, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Bertha Furlan Polegato
- Internal Medicine Department, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | - Katashi Okoshi
- Internal Medicine Department, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| | | | | | | | - Krista Anne Power
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Canada
| | - Marcos Ferreira Minicucci
- Internal Medicine Department, Botucatu Medical School, São Paulo State University (UNESP), Botucatu, Brazil
| |
Collapse
|
161
|
De Majo F, Hegenbarth JC, Rühle F, Bär C, Thum T, de Boer M, Duncker DJ, Schroen B, Armand AS, Stoll M, De Windt LJ. Dichotomy between the transcriptomic landscape of naturally versus accelerated aged murine hearts. Sci Rep 2020; 10:8136. [PMID: 32424227 PMCID: PMC7235007 DOI: 10.1038/s41598-020-65115-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 04/29/2020] [Indexed: 11/12/2022] Open
Abstract
We investigated the transcriptomic landscape of the murine myocardium along the course of natural aging and in three distinct mouse models of premature aging with established aging-related cardiac dysfunction. Genome-wide total RNA-seq was performed and the expression patterns of protein-coding genes and non-coding RNAs were compared between hearts from naturally aging mice, mice with cardiac-specific deficiency of a component of the DNA repair machinery, mice with reduced mitochondrial antioxidant capacity and mice with reduced telomere length. Our results demonstrate that no dramatic changes are evident in the transcriptomes of naturally senescent murine hearts until two years of age, in contrast to the transcriptome of accelerated aged mice. Additionally, these mice displayed model-specific alterations of the expression levels of protein-coding and non-coding genes with hardly any overlap with age-related signatures. Our data demonstrate very limited similarities between the transcriptomes of all our murine aging models and question their reliability to study human cardiovascular senescence.
Collapse
Affiliation(s)
- Federica De Majo
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands.,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Jana-Charlotte Hegenbarth
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands.,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Frank Rühle
- Bioinformatics Core Facility, Institute of Molecular Biology (IMB), Mainz, Germany.,Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany
| | - Christian Bär
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany.,REBIRTH Excellence Cluster, Hannover Medical School, Hannover, Germany
| | - Martine de Boer
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Blanche Schroen
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands
| | - Anne-Sophie Armand
- Institut Necker Enfants Malades, Inserm U1151, Paris, France; Universite Paris Descartes, Sorbonne Paris Cite, Paris, France
| | - Monika Stoll
- Department of Genetic Epidemiology, Institute of Human Genetics, University Hospital Münster, Münster, Germany.,Department of Biochemistry, Cardiovascular Research Institute Maastricht, Maastricht University, Maastricht, The Netherlands
| | - Leon J De Windt
- Department of Molecular Genetics, Faculty of Science and Engineering; Maastricht University, Maastricht, The Netherlands. .,CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences; Maastricht University, Maastricht, The Netherlands.
| |
Collapse
|
162
|
Tarbit E, Singh I, Peart JN, Rose'Meyer RB. Biomarkers for the identification of cardiac fibroblast and myofibroblast cells. Heart Fail Rev 2020; 24:1-15. [PMID: 29987445 DOI: 10.1007/s10741-018-9720-1] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Experimental research has recognized the importance of cardiac fibroblast and myofibroblast cells in heart repair and function. In a normal healthy heart, the cardiac fibroblast plays a central role in the structural, electrical, and chemical aspects within the heart. Interestingly, the transformation of cardiac fibroblast cells to cardiac myofibroblast cells is suspected to play a vital part in the development of heart failure. The ability to differentiate between the two cells types has been a challenge. Myofibroblast cells are only expressed in the stressed or failing heart, so a better understanding of cell function may identify therapies that aid repair of the damaged heart. This paper will provide an outline of what is currently known about cardiac fibroblasts and myofibroblasts, the physiological and pathological roles within the heart, and causes for the transition of fibroblasts into myoblasts. We also reviewed the potential markers available for characterizing these cells and found that there is no single-cell specific marker that delineates fibroblast or myofibroblast cells. To characterize the cells of fibroblast origin, vimentin is commonly used. Cardiac fibroblasts can be identified using discoidin domain receptor 2 (DDR2) while α-smooth muscle actin is used to distinguish myofibroblasts. A known cytokine TGF-β1 is well established to cause the transformation of cardiac fibroblasts to myofibroblasts. This review will also discuss clinical treatments that inhibit or reduce the actions of TGF-β1 and its contribution to cardiac fibrosis and heart failure.
Collapse
Affiliation(s)
- Emiri Tarbit
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia
| | - Indu Singh
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia
| | - Jason N Peart
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia
| | - Roselyn B Rose'Meyer
- School of Medical Sciences, Griffith University, Griffith, QLD, 4222, Australia.
| |
Collapse
|
163
|
Wang Y, Wang X, Wang J, Li B, Yu R, Hu Y, Li X, Peng G, Zhang M, Zhu M. Tongmai Yangxin intervening in myocardial remodeling after PCI for coronary heart disease: study protocol for a double-blind, randomized controlled trial. Trials 2020; 21:287. [PMID: 32197640 PMCID: PMC7085139 DOI: 10.1186/s13063-020-4208-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 02/25/2020] [Indexed: 12/11/2022] Open
Abstract
Background Coronary heart disease (CHD) has become a common cardiovascular disease that seriously threatens the health of people. As reperfusion in the early phase and drug therapy, especially percutaneous coronary intervention (PCI), have become widely used in the clinic, the mortality of acute myocardial infarction in the short term has been reduced significantly. In addition, in 40%–56% of patients who experience myocardial infarction, cardiac dysfunction occurs and about 25%–33% develop heart failure. Methods This study was designed as a multicenter, double-blind, randomized, placebo-controlled, parallel-group, superiority trial. Participants were randomly assigned in a 1:1 ratio through a centrally controlled, computer-generated, simple randomization schedule. The primary outcome was left ventricular end-diastolic volume index = left ventricular end-diastolic volume/body surface area. The combined secondary outcomes include traditional Chinese medicine syndrome score, echocardiogram results, 6-minute walk test results, Seattle Angina Questionnaire score, cardiac magnetic resonance imaging results, biological indicators, dynamic electrocardiogram results, and experiment event rate. Assessments will be performed at baseline and at 4, 8, and 12 weeks after randomization. Discussion This trial will demonstrate that the addition of a Tongmai Yangxin pill (TMYX) to conventional treatment will intervene in the development of cardiac remodeling and cardiac dysfunction. Trial registration This study was registered in the Chinese Clinical Trial Registry on 7 May 2019. The registration number is ChiCRT1900023023 (http://www.chictr.org.cn/showproj.aspx?proj=12370).
Collapse
Affiliation(s)
- Yongxia Wang
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Xinlu Wang
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Jianru Wang
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Bin Li
- Graduate Division, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Rui Yu
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Yucai Hu
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Xingyuan Li
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Guangcao Peng
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China
| | - Mengmeng Zhang
- Henan University of Chinese Medicine, 156 Jinshui East Road, Zhengdong New District, Zhengzhou, 450046, Henan Province, China
| | - Mingjun Zhu
- Center of Cardiology, The First Affiliated Hospital of Henan University of TCM, 19 Renmin Road, Jinshui District, Zhengzhou, 450100, Henan Province, China.
| |
Collapse
|
164
|
Luchi TC, Coelho PM, Cordeiro JP, Assis ALEM, Nogueira BV, Marques VB, Dos Santos L, Lima-Leopoldo AP, Lunz W, Leopoldo AS. Chronic aerobic exercise associated to low-dose L-NAME improves contractility without changing calcium handling in rat cardiomyocytes. ACTA ACUST UNITED AC 2020; 53:e8761. [PMID: 32159612 PMCID: PMC7076801 DOI: 10.1590/1414-431x20198761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 12/11/2019] [Indexed: 11/22/2022]
Abstract
Nitric oxide (NO) inhibition by high-dose NG-nitro-L-arginine methyl ester (L-NAME) is associated with several detrimental effects on the cardiovascular system. However, low-dose L-NAME increases NO synthesis, which in turn induces physiological cardiovascular benefits, probably by activating a protective negative feedback mechanism. Aerobic exercise, likewise, improves several cardiovascular functions in healthy hearts, but its effects are not known when chronically associated with low-dose L-NAME. Thus, we tested whether the association between low-dose L-NAME administration and chronic aerobic exercise promotes beneficial effects to the cardiovascular system, evaluating the cardiac remodeling process. Male Wistar rats were randomly assigned to control (C), L-NAME (L), chronic aerobic exercise (Ex), and chronic aerobic exercise associated to L-NAME (ExL). Aerobic training was performed with progressive intensity for 12 weeks; L-NAME (1.5 mg·kg-1·day-1) was administered by orogastric gavage. Low-dose L-NAME alone did not change systolic blood pressure (SBP), but ExL significantly increased SBP at week 8 with normalization after 12 weeks. Furthermore, ExL promoted the elevation of left ventricle (LV) end-diastolic pressure without the presence of cardiac hypertrophy and fibrosis. Time to 50% shortening and relaxation were reduced in ExL, suggesting a cardiomyocyte contractile improvement. In addition, the time to 50% Ca2+ peak was increased without alterations in Ca2+ amplitude and time to 50% Ca2+ decay. In conclusion, the association of chronic aerobic exercise and low-dose L-NAME prevented cardiac pathological remodeling and induced cardiomyocyte contractile function improvement; however, it did not alter myocyte affinity and sensitivity to intracellular Ca2+ handling.
Collapse
Affiliation(s)
- T C Luchi
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - P M Coelho
- Programa de Pós-Graduação em Nutrição e Saúde, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - J P Cordeiro
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - A L E M Assis
- Departamento de Morfologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - B V Nogueira
- Departamento de Morfologia, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - V B Marques
- Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - L Dos Santos
- Departamento de Ciências Fisiológicas, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - A P Lima-Leopoldo
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo, Vitória, ES, Brasil.,Programa de Pós-Graduação em Nutrição e Saúde, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - W Lunz
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| | - A S Leopoldo
- Departamento de Desportos, Centro de Educação Física e Desportos, Universidade Federal do Espírito Santo, Vitória, ES, Brasil.,Programa de Pós-Graduação em Nutrição e Saúde, Centro de Ciências da Saúde, Universidade Federal do Espírito Santo, Vitória, ES, Brasil
| |
Collapse
|
165
|
Binas S, Knyrim M, Hupfeld J, Kloeckner U, Rabe S, Mildenberger S, Quarch K, Strätz N, Misiak D, Gekle M, Grossmann C, Schreier B. miR-221 and -222 target CACNA1C and KCNJ5 leading to altered cardiac ion channel expression and current density. Cell Mol Life Sci 2020; 77:903-918. [PMID: 31312877 PMCID: PMC7058603 DOI: 10.1007/s00018-019-03217-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 06/14/2019] [Accepted: 07/02/2019] [Indexed: 12/26/2022]
Abstract
MicroRNAs (miRs) contribute to different aspects of cardiovascular pathology, among others cardiac hypertrophy and atrial fibrillation. The aim of our study was to evaluate the impact of miR-221/222 on cardiac electrical remodeling. Cardiac miR expression was analyzed in a mouse model with altered electrocardiography parameters and severe heart hypertrophy. Next generation sequencing revealed 14 differentially expressed miRs in hypertrophic hearts, with miR-221 and -222 being the strongest regulated miR-cluster. This increase was restricted to cardiomyocytes and not observed in cardiac fibroblasts. Additionally, we evaluated the change of miR-221/222 in vivo in two models of pharmacologically induced heart hypertrophy (angiotensin II, isoprenaline), thereby demonstrating a stimulus-induced increase in miR-221/222 in vivo by angiotensin II but not by isoprenaline. Whole transcriptome analysis by RNA-seq and qRT-PCR validation revealed an enriched number of downregulated mRNAs coding for proteins located in the T-tubule, which are also predicted targets for miR-221/222. Among those, mRNAs were the L-type Ca2+ channel subunits as well as potassium channel subunits. We confirmed that both miRs target the 3'-untranslated regions of Cacna1c and Kcnj5. Furthermore, enhanced expression of these miRs reduced L-type Ca2+ channel and Kcnj5 channel abundance and function, which was analyzed by whole-cell patch clamp recordings or Western blot and flux measurements, respectively. miR-221 and -222 contribute to the regulation of L-type Ca2+ channels as well as Kcnj5 channels and, therefore, potentially contribute to disturbed cardiac excitation generation and propagation. Future studies will have to evaluate the pathophysiological and clinical relevance of aberrant miR-221/222 expression for electrical remodeling.
Collapse
Affiliation(s)
- Stephanie Binas
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Maria Knyrim
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Julia Hupfeld
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Udo Kloeckner
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Sindy Rabe
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Sigrid Mildenberger
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Katja Quarch
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Nicole Strätz
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Danny Misiak
- Institute of Molecular Medicine, Martin-Luther-University Halle-Wittenberg, Heinrich-Damerow-Str. 1, 06120, Halle/Saale, Germany
| | - Michael Gekle
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Claudia Grossmann
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany
| | - Barbara Schreier
- Julius-Bernstein-Institute of Physiology, Martin Luther University Halle-Wittenberg, Magdeburger Str. 6, 06110, Halle/Saale, Germany.
| |
Collapse
|
166
|
Ravalli F, Kossar AP, Takayama H, Grau JB, Ferrari G. Aortic Valve Regurgitation: Pathophysiology and Implications for Surgical Intervention in the Era of TAVR. STRUCTURAL HEART : THE JOURNAL OF THE HEART TEAM 2020; 4:87-98. [PMID: 32529168 PMCID: PMC7288848 DOI: 10.1080/24748706.2020.1719446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 10/25/2022]
Abstract
Aortic insufficiency (AI) or regurgitation is caused by the malcoaptation of the aortic valve (AV) cusps due to intrinsic abnormalities of the valve itself, a dilatation or geometric distortion of the aortic root, or by some combination thereof. In recent years, there has been an increase in the number of studies suggesting that AI is an active disease process caused by a combination of factors including but not limited to alteration of specific molecular pathways, genetic predisposition, and changes in the mechanotransductive properties of the AV apparatus. As the surgical management of AV disease continues to evolve, increasingly sophisticated surgical and percutaneous techniques for AV repair and replacement, including transcatheter aortic valve replacement (TAVR), have become more commonplace and will likely continue to expand as new devices are introduced. However, these techniques necessitate frequent reappraisal of the biological and mechanobiological mechanisms underlying AV regurgitation to better understand the risk factors for AI development and recurrence following surgical intervention as well as expand our limited knowledge on patient selection for such procedures. The aim of this review is to describe some of the putative mechanisms implicated in the development of AI, dissect some of the cross-talk among known and possible signaling pathways leading to valve remodeling, identify association between these pathways and pharmacological approaches, and discuss the implications for surgical and percutaneous approaches to AV repair in replacement in the TAVR era.
Collapse
|
167
|
Mohammadzadeh N, Melleby AO, Palmero S, Sjaastad I, Chakravarti S, Engebretsen KVT, Christensen G, Lunde IG, Tønnessen T. Moderate Loss of the Extracellular Matrix Proteoglycan Lumican Attenuates Cardiac Fibrosis in Mice Subjected to Pressure Overload. Cardiology 2020; 145:187-198. [PMID: 31968347 DOI: 10.1159/000505318] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Accepted: 12/05/2019] [Indexed: 12/20/2022]
Abstract
INTRODUCTION The heart undergoes myocardial remodeling during progression to heart failure following pressure overload. Myocardial remodeling is associated with structural and functional changes in cardiac myocytes, fibroblasts, and the extracellular matrix (ECM) and is accompanied by inflammation. Cardiac fibrosis, the accumulation of ECM molecules including collagens and collagen cross-linking, contributes both to impaired systolic and diastolic function. Insufficient mechanistic insight into what regulates cardiac fibrosis during pathological conditions has hampered therapeutic so-lutions. Lumican (LUM) is an ECM-secreted proteoglycan known to regulate collagen fibrillogenesis. Its expression in the heart is increased in clinical and experimental heart failure. Furthermore, LUM is important for survival and cardiac remodeling following pressure overload. We have recently reported that total lack of LUM increased mortality and left ventricular dilatation, and reduced collagen expression and cross-linking in LUM knockout mice after aortic banding (AB). Here, we examined the effect of LUM on myocardial remodeling and function following pressure overload in a less extreme mouse model, where cardiac LUM level was reduced to 50% (i.e., moderate loss of LUM). METHODS AND RESULTS mRNA and protein levels of LUM were reduced to 50% in heterozygous LUM (LUM+/-) hearts compared to wild-type (WT) controls. LUM+/- mice were subjected to AB. There was no difference in survival between LUM+/- and WT mice post-AB. Echocardiography revealed no striking differences in cardiac geometry between LUM+/- and WT mice 2, 4, and 6 weeks post-AB, although markers of diastolic dysfunction indicated better function in LUM+/- mice. LUM+/- hearts revealed reduced cardiac fibrosis assessed by histology. In accordance, the expression of collagen I and III, the main fibrillar collagens in the heart, and other ECM molecules central to fibrosis, i.e. including periostin and fibronectin, was reduced in the hearts of LUM+/- compared to WT 6 weeks post-AB. We found no differences in collagen cross-linking between LUM+/- and WT mice post-AB, as assessed by histology and qPCR. CONCLUSIONS Moderate lack of LUM attenuated cardiac fibrosis and improved diastolic dysfunction following pressure overload in mice, adding to the growing body of evidence suggesting that LUM is a central profibrotic molecule in the heart that could serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Naiyereh Mohammadzadeh
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Arne Olav Melleby
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Sheryl Palmero
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ivar Sjaastad
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Shukti Chakravarti
- Department of Medicine, Johns Hopkins University, Baltimore, Maryland, USA.,Department of Ophthalmology and Pathology, NYU Langone Health, New York, New York, USA
| | | | - Geir Christensen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo, Oslo, Norway
| | - Ida G Lunde
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway.,KG Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo, Oslo, Norway.,Center for Molecular Medicine Norway, Oslo University Hospital and University of Oslo, Oslo, Norway
| | - Theis Tønnessen
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway, .,KG Jebsen Center for Cardiac Research and Center for Heart Failure Research, University of Oslo, Oslo, Norway, .,Department of Cardiothoracic Surgery, Oslo University Hospital, Oslo, Norway,
| |
Collapse
|
168
|
Melo AB, Damiani APL, Coelho PM, de Assis ALEM, Nogueira BV, Guimarães Ferreira L, Leite RD, Ribeiro Júnior RF, Lima-Leopoldo AP, Leopoldo AS. Resistance training promotes reduction in Visceral Adiposity without improvements in Cardiomyocyte Contractility and Calcium handling in Obese Rats. Int J Med Sci 2020; 17:1819-1832. [PMID: 32714085 PMCID: PMC7378665 DOI: 10.7150/ijms.42612] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 03/23/2020] [Indexed: 11/05/2022] Open
Abstract
Resistance training (RT) improves the cardiomyocyte calcium (Ca2+) cycling during excitation-contraction coupling. However, the role of RT in cardiomyocyte contractile function associated with Ca2+ handling in obesity is unclear. Wistar rats were distributed into four groups: control, sedentary obese, control plus RT, and obesity plus RT. The 10-wk RT protocol was used (4-5 vertical ladder climbs, 60-second interval, 3× a week, 50-100% of maximum load). Metabolic, hormonal, cardiovascular and biochemical parameters were determined. Reduced leptin levels, epididymal, retroperitoneal and visceral fat pads, lower body fat, and adiposity index were observed in RT. Obesity promoted elevation of collagen, but RT did not promote modifications of LV collagen in ObRT. RT induced elevation in maximum rates of contraction and relaxation, and reduction of time to 50% relaxation. ObRT group did not present improvement in the cardiomyocyte contractile function in comparison to Ob group. Reduced cardiac PLB serine16 phosphorylation (pPLB Ser16) and pPLB Ser16/PLB ratio with no alterations in sarcoplasmic reticulum Ca2+ ATPase (SERCA2a) and phospholamban (PLB) expression were observed in Ob groups. Resistance training improved body composition reduced fat pads and plasma leptin levels but did not promote positive alterations in cardiomyocyte contractile function, Ca2+ handling and phospholamban phosphorylation.
Collapse
Affiliation(s)
- Alexandre Barroso Melo
- Centre for Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Andressa Prata Leite Damiani
- Centre for Health Sciences, Department of Nutrition, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Priscila Murucci Coelho
- Centre for Health Sciences, Department of Nutrition, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | | | - Breno Valentim Nogueira
- Center of Health Sciences, Department of Morphology, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Lucas Guimarães Ferreira
- Centre for Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Richard Diego Leite
- Centre for Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Rogério Faustino Ribeiro Júnior
- Center of Health Sciences, Department of Physiological Sciences, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - Ana Paula Lima-Leopoldo
- Centre for Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| | - André Soares Leopoldo
- Centre for Physical Education and Sports, Department of Sports, Federal University of Espírito Santo, Vitória, Espírito Santo, Brazil
| |
Collapse
|
169
|
Si LYN, Ramalingam A, Ali SS, Aminuddin A, Ng PY, Latip J, Kamisah Y, Budin SB, Zainalabidin S. Roselle attenuates cardiac hypertrophy after myocardial infarction in vivo and in vitro. EXCLI JOURNAL 2019; 18:876-892. [PMID: 31645847 PMCID: PMC6806200 DOI: 10.17179/excli2019-1792] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 09/23/2019] [Indexed: 11/24/2022]
Abstract
Roselle (Hibiscus sabdariffa Linn) has been traditionally used as folk medicine for hypertension and maintaining cardiovascular health, with therapeutic potential in protecting against numerous cardiovascular diseases. However, it remains unclear whether roselle can be used for management of cardiac hypertrophy seen after myocardial infarction (MI). This study therefore investigated the effects of aqueous roselle extract on cardiac hypertrophy arising from myocardial infarction both in vivo and in vitro. For in vivo study, male Sprague-Dawley rats were divided into control or MI groups (receiving 85 mg/kg isoproterenol s.c. for 2 days) and were given roselle extract (100 mg/kg, p.o daily) for 28 days. Cardiac structure and functional changes were evaluated at study end-point using histology, Langendorff analysis and gene expression analysis. In vitro effects of roselle were also assessed on ANG II-induced cardiomyocytes hypertrophy using H9c2 cells, simulating cardiac hypertrophy evident after MI. Roselle significantly ameliorated MI-induced cardiac systolic and diastolic dysfunction, as seen across improvement in left ventricular developed pressure (LVDP) and its derivative (LVdP/dtmax) and isovolumic relaxation (Tau). Oxidative stress evident across elevated pro-oxidant markers (NOX2 subunit of NADPH oxidase and 8-isoprostane) as well as reduced antioxidant markers (superoxide dismutase and glutathione) were also significantly attenuated by roselle. Furthermore, roselle treatment markedly reduced markers of cardiac remodeling (cardiac hypertrophy and fibrosis) compared to the untreated MI rats. On in vitro analysis, roselle significantly attenuated ANG II-induced cardiomyoycte hypertrophy in dose-dependent manner. This study demonstrated that roselle attenuates cardiac hypertrophy and dysfunction seen after MI both in vivo and in vitro, and these effects are likely mediated by phenolic compounds found in roselle extract.
Collapse
Affiliation(s)
- Lislivia-Yiang-Nee Si
- Programme of Biomedical Science, Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Anand Ramalingam
- Programme of Biomedical Science, Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Shafreena Shaukat Ali
- Programme of Biomedical Science, Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Amnani Aminuddin
- Drug and Herbal Research Center, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Pei-Yuen Ng
- Drug and Herbal Research Center, Faculty of Pharmacy, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Jalifah Latip
- School of Chemical Sciences and Food Technology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, 43600, Bangi, Selangor, Malaysia
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, 56000 Kuala Lumpur, Malaysia
| | - Siti Balkis Budin
- Programme of Biomedical Science, Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| | - Satirah Zainalabidin
- Programme of Biomedical Science, Centre of Applied and Health Sciences, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, 50300, Kuala Lumpur, Malaysia
| |
Collapse
|
170
|
Pereira-Silva DC, Machado-Silva RP, Castro-Pinheiro C, Fernandes-Santos C. Does gender influence cardiovascular remodeling in C57BL/6J mice fed a high-fat, high-sucrose and high-salt diet? Int J Exp Pathol 2019; 100:153-160. [PMID: 31321834 DOI: 10.1111/iep.12318] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 11/11/2018] [Accepted: 04/02/2019] [Indexed: 12/23/2022] Open
Abstract
Animal models are widely used to study the physiopathology of human diseases. However, the influence of gender on modern society diet style-induced cardiovascular disease has not thus far been explored in these models. Thus, this study investigated cardiovascular remodelling in C57BL/6J mice fed a diet rich in saturated fat, sucrose and salt, evaluating gender effect on this process. Male and female C57BL/6J mice were fed AIN93M diet or a modified AIN93M rich in fat, sucrose and salt (HFSS) for 12 weeks. Body mass, water and food intake and cardiovascular remodelling were assessed. The HFSS diet did not lead to body mass gain or glucose metabolism disturbance as assessed by serum glucose, insulin and oral glucose tolerance test. However, female mice on a HFSS diet had increased visceral and subcutaneous adiposity. Only male mice displayed heart hypertrophy. The left ventricle was not hypertrophied in either male or female mice, but its lumen was dilated. Intramyocardial arteries and the thoracic aorta showed media thickening in male mice, but in the female it was only observed in the thoracic aorta. Finally, intramyocardial artery dilation was present in both genders, but not in the aorta. Therefore changes in LV dimensions and arterial remodelling were influenced by both gender and the HFSS diet. In conclusion, male and female C57BL/6J mice suffered cardiovascular remodelling after 12 weeks of HFSS feeding, although they did not develop obesity or diabetes. Sexual dimorphism occurred in response to diet for body adiposity, heart hypertrophy and intramyocardial artery remodelling.
Collapse
Affiliation(s)
- Debora Cristina Pereira-Silva
- Laboratorio Multiusuario de Pesquisa Biomedica, Departamento de Ciencias Basicas, Instituto de Saude de Nova Friburgo, Universidade Federal Fluminense, Nova Fribrugo, RJ, Brazil
| | - Rayane Paula Machado-Silva
- Laboratorio Multiusuario de Pesquisa Biomedica, Departamento de Ciencias Basicas, Instituto de Saude de Nova Friburgo, Universidade Federal Fluminense, Nova Fribrugo, RJ, Brazil
| | - Camila Castro-Pinheiro
- Laboratorio Multiusuario de Pesquisa Biomedica, Departamento de Ciencias Basicas, Instituto de Saude de Nova Friburgo, Universidade Federal Fluminense, Nova Fribrugo, RJ, Brazil
| | - Caroline Fernandes-Santos
- Laboratorio Multiusuario de Pesquisa Biomedica, Departamento de Ciencias Basicas, Instituto de Saude de Nova Friburgo, Universidade Federal Fluminense, Nova Fribrugo, RJ, Brazil
| |
Collapse
|
171
|
Ma S, Ma J, Mai X, Zhao X, Guo L, Zhang M. Danqi soft capsule prevents infarct border zone remodelling and reduces susceptibility to ventricular arrhythmias in post-myocardial infarction rats. J Cell Mol Med 2019; 23:5454-5465. [PMID: 31232519 PMCID: PMC6653321 DOI: 10.1111/jcmm.14428] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Revised: 04/11/2019] [Accepted: 04/19/2019] [Indexed: 12/15/2022] Open
Abstract
Danqi soft capsule (DQ) is a traditional Chinese medicine containing Salvia miltiorrhiza and Panax notoginseng; it is safe and efficient in treating ischaemic heart diseases. The purpose of the present study was to assess whether DQ could prevent infarct border zone (IBZ) remodelling and decrease ventricular arrhythmias occurrence in post‐myocardial infarction (MI) stage. MI was induced by a ligation of the left anterior descending coronary artery. DQ was administered to the post‐MI rats started from 1 week after MI surgery for 4 weeks. The results showed that DQ treatment significantly attenuated tachyarrhythmia induction rates and arrhythmia score in post‐MI rats. In echocardiography, DQ improved left ventricular (LV) systolic and diastolic function. Histological assessment revealed that DQ significantly reduced fibrotic areas and myocyte areas, and increased connexin (Cx) 43 positive areas in IBZ. Western blot revealed that DQ treatment significantly reduced the protein expression levels of type I and III collagens, α‐smooth muscle actin (α‐SMA), transforming growth factor‐β1 (TGF‐β1) and Smad3 phosphorylation, while increasing Cx43 amounts. Overall, these findings mainly indicated that DQ intervention regulates interstitial fibrosis, Cx43 expression and myocyte hypertrophy by TGF‐β1/Smad3 pathway in IBZ, inhibits LV remodelling and reduces vulnerability to tachyarrhythmias after MI. This study presents a proof of concept for novel antiarrhythmic strategies in preventing IBZ remodelling, modifying the healed arrhythmogenic substrate and thus reducing susceptibility to ventricular arrhythmias in the late post‐MI period.
Collapse
Affiliation(s)
- Shiyu Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Myocardial Infarction, Guangzhou, China
| | - Jin Ma
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xiaoyi Mai
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Myocardial Infarction, Guangzhou, China
| | - Xujie Zhao
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Liheng Guo
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Minzhou Zhang
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China.,Guangzhou Key Laboratory of Chinese Medicine for Prevention and Treatment of Myocardial Infarction, Guangzhou, China
| |
Collapse
|
172
|
Saucerman JJ, Tan PM, Buchholz KS, McCulloch AD, Omens JH. Mechanical regulation of gene expression in cardiac myocytes and fibroblasts. Nat Rev Cardiol 2019; 16:361-378. [PMID: 30683889 PMCID: PMC6525041 DOI: 10.1038/s41569-019-0155-8] [Citation(s) in RCA: 133] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
The intact heart undergoes complex and multiscale remodelling processes in response to altered mechanical cues. Remodelling of the myocardium is regulated by a combination of myocyte and non-myocyte responses to mechanosensitive pathways, which can alter gene expression and therefore function in these cells. Cellular mechanotransduction and its downstream effects on gene expression are initially compensatory mechanisms during adaptations to the altered mechanical environment, but under prolonged and abnormal loading conditions, they can become maladaptive, leading to impaired function and cardiac pathologies. In this Review, we summarize mechanoregulated pathways in cardiac myocytes and fibroblasts that lead to altered gene expression and cell remodelling under physiological and pathophysiological conditions. Developments in systems modelling of the networks that regulate gene expression in response to mechanical stimuli should improve integrative understanding of their roles in vivo and help to discover new combinations of drugs and device therapies targeting mechanosignalling in heart disease.
Collapse
Affiliation(s)
- Jeffrey J Saucerman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Philip M Tan
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Kyle S Buchholz
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| | - Andrew D McCulloch
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA.
| | - Jeffrey H Omens
- Departments of Bioengineering and Medicine, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
173
|
Chacar S, Hajal J, Saliba Y, Bois P, Louka N, Maroun RG, Faivre J, Fares N. Long-term intake of phenolic compounds attenuates age-related cardiac remodeling. Aging Cell 2019; 18:e12894. [PMID: 30680911 PMCID: PMC6413651 DOI: 10.1111/acel.12894] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 10/26/2018] [Accepted: 11/27/2018] [Indexed: 01/14/2023] Open
Abstract
With the onset of advanced age, cardiac-associated pathologies have increased in prevalence. The hallmarks of cardiac aging include cardiomyocyte senescence, fibroblast proliferation, inflammation, and hypertrophy. The imbalance between levels of reactive oxygen species (ROS) and antioxidant enzymes is greatly enhanced in aging cells, promoting cardiac remodeling. In this work, we studied the long-term impact of phenolic compounds (PC) on age-associated cardiac remodeling. Three-month-old Wistar rats were treated for 14 months till middle-age with either 2.5, 5, 10, or 20 mg kg-1 day-1 of PC. PC treatment showed a dose-dependent preservation of cardiac ejection fraction and fractional shortening as well as decreased hypertrophy reflected by left ventricular chamber diameter and posterior wall thickness as compared to untreated middle-aged control animals. Analyses of proteins from cardiac tissue showed that PC attenuated several hypertrophic pathways including calcineurin/nuclear factor of activated T cells (NFATc3), calcium/calmodulin-dependent kinase II (CAMKII), extracellular regulated kinase 1/2 (ERK1/2), and glycogen synthase kinase 3ß (GSK 3ß). PC-treated groups exhibited reduced plasma inflammatory and fibrotic markers and revealed as well ameliorated extracellular matrix remodeling and interstitial inflammation by a downregulated p38 pathway. Myocardia from PC-treated middle-aged rats presented less fibrosis with suppression of profibrotic transforming growth factor-ß1 (TGF-ß1) Smad pathway. Additionally, reduction of apoptosis and oxidative damage in the PC-treated groups was reflected by elevated antioxidant enzymes and reduced RNA/DNA damage markers. Our findings pinpoint that a daily consumption of phenolic compounds could preserve the heart from the detrimental effects of aging storm.
Collapse
Affiliation(s)
- Stéphanie Chacar
- Faculté de Médecine, Laboratoire de Recherche en Physiologie et Physiopathologie, LRPP, Pôle Technologie SantéUniversité Saint JosephBeyrouthLiban
- Faculté des Sciences, Centre d’Analyses et de Recherche, UR GPF, Laboratoire CTAUniversité Saint‐JosephBeyrouthLiban
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM)Université de PoitiersPoitiersFrance
| | - Joelle Hajal
- Faculté de Médecine, Laboratoire de Recherche en Physiologie et Physiopathologie, LRPP, Pôle Technologie SantéUniversité Saint JosephBeyrouthLiban
| | - Youakim Saliba
- Faculté de Médecine, Laboratoire de Recherche en Physiologie et Physiopathologie, LRPP, Pôle Technologie SantéUniversité Saint JosephBeyrouthLiban
| | - Patrick Bois
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM)Université de PoitiersPoitiersFrance
| | - Nicolas Louka
- Faculté des Sciences, Centre d’Analyses et de Recherche, UR GPF, Laboratoire CTAUniversité Saint‐JosephBeyrouthLiban
| | - Richard G. Maroun
- Faculté des Sciences, Centre d’Analyses et de Recherche, UR GPF, Laboratoire CTAUniversité Saint‐JosephBeyrouthLiban
| | - Jean‐François Faivre
- Laboratoire Signalisation et Transports Ioniques Membranaires (STIM)Université de PoitiersPoitiersFrance
| | - Nassim Fares
- Faculté de Médecine, Laboratoire de Recherche en Physiologie et Physiopathologie, LRPP, Pôle Technologie SantéUniversité Saint JosephBeyrouthLiban
| |
Collapse
|
174
|
Vatnikov YA, Rudenko AA, Usha BV, Kulikov EV, Notina EA, Bykova IA, Khairova NI, Bondareva IV, Grishin VN, Zharov AN. Left ventricular myocardial remodeling in dogs with mitral valve endocardiosis. Vet World 2019; 13:731-738. [PMID: 32546918 PMCID: PMC7245723 DOI: 10.14202/vetworld.2020.731-738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2019] [Accepted: 03/06/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND AND AIM Left ventricular myocardial remodeling could play an important role in the progression of chronic heart failure (CHF) syndrome in dogs with mitral valve endocardiosis. The aim of this study was to evaluate the left ventricular myocardial remodeling in dogs with mitral valve endocardiosis and to study the dependence of the incidence of this pathological phenomenon on the functional class (FC) of progression of the CHF syndrome. MATERIALS AND METHODS A total of 108 afflicted dogs and 36 clinically healthy dogs were examined using transthoracic echocardiography. The following structural and geometric parameters of the left ventricular remodeling were evaluated: Myocardial mass and its index, sphericity index at the end of systole and diastole, end-systolic and end-diastolic relative wall thickness, and integral remodeling index. RESULTS In all clinically healthy dogs, a normal type of the left ventricular chamber geometry was revealed, whereas, in dogs with mitral valve endocardiosis, the normal geometry of the left ventricle occurred in 56.4%, eccentric hypertrophy in 24.1%, concentric remodeling in 10.2%, and concentric hypertrophy in 9.3% of the cases. In patients with endocardiosis, there was no dilatation type of cardiac remodeling observed. CONCLUSION When compared to the clinically healthy animals, the dogs with mitral valve endocardiosis presented with indicators of structural and geometric remodeling, such as increased myocardial mass, myocardial mass index, and sphericity index at the end of systole and diastole, as well as relatively reduced integral systolic index of remodeling and systolic relative thickness of the walls of the heart. The parameters of the left ventricular myocardial remodeling correlated significantly with the FC of CHF syndrome.
Collapse
Affiliation(s)
- Yury A. Vatnikov
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Andrey A. Rudenko
- Department of Veterinary Medicine, Moscow State University of Food Production, 125080 Moscow, Russia
| | - Boris V. Usha
- Department of Veterinary Medicine, Moscow State University of Food Production, 125080 Moscow, Russia
| | - Evgeny V. Kulikov
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Elena A. Notina
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Irina A. Bykova
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Nadiya I. Khairova
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Irina V. Bondareva
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Victor N. Grishin
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Andrey N. Zharov
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
175
|
Contti MM, Barbosa MF, del Carmen Villanueva Mauricio A, Nga HS, Valiatti MF, Takase HM, Bravin AM, de Andrade LGM. Kidney transplantation is associated with reduced myocardial fibrosis. A cardiovascular magnetic resonance study with native T1 mapping. J Cardiovasc Magn Reson 2019; 21:21. [PMID: 30917836 PMCID: PMC6437926 DOI: 10.1186/s12968-019-0531-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 03/07/2019] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The measurement of native T1 through cardiovascular magnetic resonance (CMR) is a noninvasive method of assessing myocardial fibrosis without gadolinium contrast. No studies so far have evaluated native T1 after renal transplantation. The primary aim of the current study is to assess changes in the myocardium native T1 6 months after renal transplantation. METHODS We prospectively evaluated 44 renal transplant patients with 3 T CMR exams: baseline at the beginning of transplantation and at 6 months after transplantation. RESULTS The native T1 time was measured in the midventricular septum and decreased significantly from 1331 ± 52 ms at the baseline to 1298 ± 42 ms 6 months after transplantation (p = 0.001). The patients were split into two groups through a two-step cluster algorithm: In cluster-1 (n = 30) the left ventricular (LV) mass index and the prevalence of diabetes were lower. In cluster-2 (n = 14) the LV mass index and diabetes prevalence were higher. Decrease in native T1 values was significant only in the patients in cluster-1 (p = 0.001). CONCLUSIONS The native myocardial T1 time decreased significantly 6 months after renal transplant, which may be associated with the regression of the reactive fibrosis. The patients with greater baseline LV mass index and the diabetic group did not reach a significant decrease in T1.
Collapse
Affiliation(s)
- Mariana Moraes Contti
- Department of Internal Medicine, UNESP, Univ Estadual Paulista, Rubião Jr, s/n, Botucatu/SP, 18.618-970 Brazil
| | | | | | - Hong Si Nga
- Department of Internal Medicine, UNESP, Univ Estadual Paulista, Rubião Jr, s/n, Botucatu/SP, 18.618-970 Brazil
| | - Mariana Farina Valiatti
- Department of Internal Medicine, UNESP, Univ Estadual Paulista, Rubião Jr, s/n, Botucatu/SP, 18.618-970 Brazil
| | - Henrique Mochida Takase
- Department of Internal Medicine, UNESP, Univ Estadual Paulista, Rubião Jr, s/n, Botucatu/SP, 18.618-970 Brazil
| | - Ariane Moyses Bravin
- Department of Internal Medicine, UNESP, Univ Estadual Paulista, Rubião Jr, s/n, Botucatu/SP, 18.618-970 Brazil
| | | |
Collapse
|
176
|
Engineering a naturally-derived adhesive and conductive cardiopatch. Biomaterials 2019; 207:89-101. [PMID: 30965152 DOI: 10.1016/j.biomaterials.2019.03.015] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/08/2019] [Accepted: 03/13/2019] [Indexed: 12/11/2022]
Abstract
Myocardial infarction (MI) leads to a multi-phase reparative process at the site of damaged heart that ultimately results in the formation of non-conductive fibrous scar tissue. Despite the widespread use of electroconductive biomaterials to increase the physiological relevance of bioengineered cardiac tissues in vitro, there are still several limitations associated with engineering biocompatible scaffolds with appropriate mechanical properties and electroconductivity for cardiac tissue regeneration. Here, we introduce highly adhesive fibrous scaffolds engineered by electrospinning of gelatin methacryloyl (GelMA) followed by the conjugation of a choline-based bio-ionic liquid (Bio-IL) to develop conductive and adhesive cardiopatches. These GelMA/Bio-IL adhesive patches were optimized to exhibit mechanical and conductive properties similar to the native myocardium. Furthermore, the engineered patches strongly adhered to murine myocardium due to the formation of ionic bonding between the Bio-IL and native tissue, eliminating the need for suturing. Co-cultures of primary cardiomyocytes and cardiac fibroblasts grown on GelMA/Bio-IL patches exhibited comparatively better contractile profiles compared to pristine GelMA controls, as demonstrated by over-expression of the gap junction protein connexin 43. These cardiopatches could be used to provide mechanical support and restore electromechanical coupling at the site of MI to minimize cardiac remodeling and preserve normal cardiac function.
Collapse
|
177
|
Badae NM, El Naggar AS, El Sayed SM. Is the cardioprotective effect of the ACE2 activator diminazene aceturate more potent than the ACE inhibitor enalapril on acute myocardial infarction in rats? Can J Physiol Pharmacol 2019; 97:638-646. [PMID: 30840489 DOI: 10.1139/cjpp-2019-0078] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Myocardial infarction is a major cause of cardiac dysfunction. All components of the cardiac renin-angiotensin system (RAS) are upregulated in myocardial infarction. Angiotensin-converting enzyme (ACE) and ACE2 are key enzymes involved in synthesis of components of RAS and provide a counter-regulatory mechanism within RAS. We compared the cardioprotective effect of the ACE2 activator diminazene aceturate (DIZE) versus the ACE inhibitor enalapril on post acute myocardial infarction (AMI) ventricular dysfunction in rats. Adult male rats received subcutaneous injections of either saline (control) or isoproterenol (85 mg/kg) to induce AMI. Rats with AMI confirmed biochemically and by ECG, were either left untreated (AMI) or administered DIZE (AMI + DIZE) or enalapril (AMI + enalapril) daily for 4 weeks. DIZE caused a significant activation of cardiac ACE2 compared with enalapril. DIZE caused a significantly greater enhancement of cardiac hemodynamics. DIZE also caused greater reductions in heart-type fatty acid binding protein (H-FABP), β-myosin heavy chain (β-MYH), and in heart mass to total body mass ratio. These results indicated that activation of cardiac ACE2 by DIZE enhanced the protective axis of RAS and improved myocardial function following AMI, whereas enalapril was not sufficient to restore all cardiac parameters back to normal.
Collapse
Affiliation(s)
- Noha Mohamed Badae
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.,Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Asmaa Samy El Naggar
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.,Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Samiha Mahmoud El Sayed
- Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.,Department of Medical Physiology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| |
Collapse
|
178
|
Maksuti E, Westerhof BE, Ugander M, Donker DW, Carlsson M, Broomé M. Cardiac remodeling in aortic and mitral valve disease: a simulation study with clinical validation. J Appl Physiol (1985) 2019; 126:1377-1389. [PMID: 30730809 DOI: 10.1152/japplphysiol.00791.2018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Remodeling is an important long-term determinant of cardiac function throughout the progression of heart disease. Numerous biomolecular pathways for mechanosensing and transduction are involved. However, we hypothesize that biomechanical factors alone can explain changes in myocardial volume and chamber size in valve disease. A validated model of the human vasculature and the four cardiac chambers was used to simulate aortic stenosis, mitral regurgitation, and aortic regurgitation. Remodeling was simulated with adaptive feedback preserving myocardial fiber stress and wall shear stress in all four cardiac chambers. Briefly, the model used myocardial fiber stress to determine wall thickness and cardiac chamber wall shear stress to determine chamber volume. Aortic stenosis resulted in the development of concentric left ventricular hypertrophy. Aortic and mitral regurgitation resulted in eccentric remodeling and eccentric hypertrophy, with more pronounced hypertrophy for aortic regurgitation. Comparisons with published clinical data showed the same direction and similar magnitudes of changes in end-diastolic volume index and left ventricular diameters. Changes in myocardial wall volume and wall thickness were within a realistic range in both stenotic and regurgitant valvular disease. Simulations of remodeling in left-sided valvular disease support, in both a qualitative and quantitative manner, that left ventricular chamber size and hypertrophy are primarily determined by preservation of wall shear stress and myocardial fiber stress. NEW & NOTEWORTHY Cardiovascular simulations with adaptive feedback that normalizes wall shear stress and fiber stress in the cardiac chambers could predict, in a quantitative and qualitative manner, remodeling patterns seen in patients with left-sided valvular disease. This highlights how mechanical stress remains a fundamental aspect of cardiac remodeling. This in silico study validated with clinical data paves the way for future patient-specific predictions of remodeling in valvular disease.
Collapse
Affiliation(s)
- Elira Maksuti
- Extracorporeal Membrane Oxygenation Department, Karolinska University Hospital, Stockholm, Sweden
| | - Berend E Westerhof
- Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam Universitair Medische Centra, Vrije Universiteit Amsterdam , Amsterdam , The Netherlands
| | - Martin Ugander
- Department of Clinical Physiology, Karolinska Institutet and Karolinska University Hospital , Stockholm , Sweden
| | - Dirk W Donker
- Department of Intensive Care Medicine, University Medical Center Utrecht, Utrecht University , Utrecht , The Netherlands
| | - Marcus Carlsson
- Clinical Physiology, Department of Clinical Sciences Lund, Lund University, Skane University Hospital , Lund , Sweden
| | - Michael Broomé
- Extracorporeal Membrane Oxygenation Department, Karolinska University Hospital, Stockholm, Sweden.,Anesthesia and Intensive Care, Department of Physiology and Pharmacology, Karolinska Institutet , Stockholm , Sweden
| |
Collapse
|
179
|
Atrial Fibrillation, Ventricular Arrhythmia, and Left Ventricular Remodeling in the ICU – First Results of the Single- Center RHYTHM-ACC Registry. JOURNAL OF CARDIOVASCULAR EMERGENCIES 2019. [DOI: 10.2478/jce-2018-0026] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Abstract
The aim of this study was to investigate the association between left ventricular remodeling, atrial fibrillation (AF), and the severity of ventricular tachycardia (VT) in patients with ventricular rhythm disturbances admitted in a level 3 facility of acute cardiac care. Material and Methods: The RHYTHM-ACC registry was a single-center observational study, including 150 consecutive patients with sustained or non-sustained ventricular tachycardia (sVT and nsVT, respectively) admitted in an intensive cardiac care unit (ICCU), separated in: group 1 - 29 patients (21.01%) with dilated cardiomyopathy (DCM), and group 2 - 109 patients (78.99%) with normal ventricular performance. We investigated the difference between clinical characteristics of patients with sVT versus those with nsVT in each study group, and the association between AF and different forms of ventricular arrhythmia in 38 (25.33%) patients with AF and 112 (74.66%) patients in sinus rhythm. Results: There were no significant differences between the study groups with respect to type of ventricular arrhythmia: sVT (46.87% vs. 36.44%, p = 0.2), nsVT (43.75% vs. 55.93%, p = 0.2), or ventricular fibrillation (VF) (9.37% vs. 7.62%, p = 0.7). However, patients with DCM presented a significantly higher incidence of AF (43.75% vs. 20.33%, p = 0.01) and bundle branch block (37.5% vs. 11.86%, p = 0.0007). VF occurred more frequently in patients with AF compared to those in sinus rhythm (18.42% vs. 4.46%, p = 0.006). Multivariate analysis identified the co-existence of AF (OR = 4.8, p = 0.01) and the presence of a bundle branch block (BBB) (OR = 3.9, p = 0.03) as the most powerful predictors for the degeneration of VT into VF in patients admitted with sVT or nsVT in an ICCU unit. Conclusions: In patients with any type of VT admitted in an ICCU, the presence of ventricular remodeling is associated with a higher incidence of AF and conduction abnormalities, but not with a more severe pattern of ventricular arrhythmia. At the same time, AF and BBB seem to represent the most powerful predictors for degeneration of VT into VF, independent of the type of VT.
Collapse
|
180
|
Langmia IM, Kräker K, Weiss SE, Haase N, Schütte T, Herse F, Dechend R. Cardiovascular Programming During and After Diabetic Pregnancy: Role of Placental Dysfunction and IUGR. Front Endocrinol (Lausanne) 2019; 10:215. [PMID: 31024453 PMCID: PMC6466995 DOI: 10.3389/fendo.2019.00215] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/18/2019] [Indexed: 12/31/2022] Open
Abstract
Intrauterine growth restriction (IUGR) is a condition whereby a fetus is unable to achieve its genetically determined potential size. IUGR is a global health challenge due to high mortality and morbidity amongst affected neonates. It is a multifactorial condition caused by maternal, fetal, placental, and genetic confounders. Babies born of diabetic pregnancies are usually large for gestational age but under certain conditions whereby prolonged uncontrolled hyperglycemia leads to placental dysfunction, the outcome of the pregnancy is an intrauterine growth restricted fetus with clinical features of malnutrition. Placental dysfunction leads to undernutrition and hypoxia, which triggers gene modification in the developing fetus due to fetal adaptation to adverse utero environmental conditions. Thus, in utero gene modification results in future cardiovascular programming in postnatal and adult life. Ongoing research aims to broaden our understanding of the molecular mechanisms and pathological pathways involved in fetal programming due to IUGR. There is a need for the development of effective preventive and therapeutic strategies for the management of growth-restricted infants. Information on the mechanisms involved with in utero epigenetic modification leading to development of cardiovascular disease in adult life will increase our understanding and allow the identification of susceptible individuals as well as the design of targeted prevention strategies. This article aims to systematically review the latest molecular mechanisms involved in the pathogenesis of IUGR in cardiovascular programming. Animal models of IUGR that used nutrient restriction and hypoxia to mimic the clinical conditions in humans of reduced flow of nutrients and oxygen to the fetus will be discussed in terms of cardiac remodeling and epigenetic programming of cardiovascular disease. Experimental evidence of long-term fetal programming due to IUGR will also be included.
Collapse
Affiliation(s)
- Immaculate M. Langmia
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Alexander von Humboldt Foundation, Bonn, Germany
| | - Kristin Kräker
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Sara E. Weiss
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Nadine Haase
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Till Schütte
- Berlin Institute of Health (BIH), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Center for Cardiovascular Research, Institute of Pharmacology, Charité -Universitätsmedizin Berlin, Berlin, Germany
| | - Florian Herse
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Max-Delbrueck Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralf Dechend
- Experimental and Clinical Research Center, A Joint Cooperation Between the Max-Delbrueck Center for Molecular Medicine and the Charité Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Charité–Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin, Berlin Institute of Health, Humboldt-Universität zu Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- HELIOS-Klinikum, Berlin, Germany
- *Correspondence: Ralf Dechend
| |
Collapse
|
181
|
Aboelkasem Ali Mousa M, Abdelsabour Abdallah M, Shamseddin Mohammad H, Ahmad Aly Youssef A. Early predictors of left ventricular remodeling after primary percutaneous coronary intervention. Egypt Heart J 2018; 70:403-407. [PMID: 30591763 PMCID: PMC6303352 DOI: 10.1016/j.ehj.2018.05.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/21/2018] [Indexed: 11/24/2022] Open
|
182
|
Michalak M, Agellon LB. Stress Coping Strategies in the Heart: An Integrated View. Front Cardiovasc Med 2018; 5:168. [PMID: 30519562 PMCID: PMC6258784 DOI: 10.3389/fcvm.2018.00168] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 11/02/2018] [Indexed: 12/15/2022] Open
Abstract
The heart is made up of an ordered amalgam of cardiac cell types that work together to coordinate four major processes, namely energy production, electrical conductance, mechanical work, and tissue remodeling. Over the last decade, a large body of information has been amassed regarding how different cardiac cell types respond to cellular stress that affect the functionality of their elaborate intracellular membrane networks, the cellular reticular network. In the context of the heart, the manifestations of stress coping strategies likely differ depending on the coping strategy outcomes of the different cardiac cell types, and thus may underlie the development of distinct cardiac disorders. It is not clear whether all cardiac cell types have similar sensitivity to cellular stress, how specific coping response strategies modify their unique roles, and how their metabolic status is communicated to other cells within the heart. Here we discuss our understanding of the roles of specialized cardiac cells that together make the heart function as an organ with the ability to pump blood continuously and follow a regular rhythm.
Collapse
Affiliation(s)
- Marek Michalak
- Department of Biochemistry, University of Alberta, Edmonton, AB, Canada
| | - Luis B Agellon
- School of Human Nutrition, McGill University, Ste. Anne de Bellevue, QC, Canada
| |
Collapse
|
183
|
Many Cells Make Life Work-Multicellularity in Stem Cell-Based Cardiac Disease Modelling. Int J Mol Sci 2018; 19:ijms19113361. [PMID: 30373227 PMCID: PMC6274721 DOI: 10.3390/ijms19113361] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 10/23/2018] [Accepted: 10/24/2018] [Indexed: 12/22/2022] Open
Abstract
Cardiac disease causes 33% of deaths worldwide but our knowledge of disease progression is still very limited. In vitro models utilising and combining multiple, differentiated cell types have been used to recapitulate the range of myocardial microenvironments in an effort to delineate the mechanical, humoral, and electrical interactions that modulate the cardiac contractile function in health and the pathogenesis of human disease. However, due to limitations in isolating these cell types and changes in their structure and function in vitro, the field is now focused on the development and use of stem cell-derived cell types, most notably, human-induced pluripotent stem cell-derived CMs (hiPSC-CMs), in modelling the CM function in health and patient-specific diseases, allowing us to build on the findings from studies using animal and adult human CMs. It is becoming increasingly appreciated that communications between cardiomyocytes (CMs), the contractile cell of the heart, and the non-myocyte components of the heart not only regulate cardiac development and maintenance of health and adult CM functions, including the contractile state, but they also regulate remodelling in diseases, which may cause the chronic impairment of the contractile function of the myocardium, ultimately leading to heart failure. Within the myocardium, each CM is surrounded by an intricate network of cell types including endothelial cells, fibroblasts, vascular smooth muscle cells, sympathetic neurons, and resident macrophages, and the extracellular matrix (ECM), forming complex interactions, and models utilizing hiPSC-derived cell types offer a great opportunity to investigate these interactions further. In this review, we outline the historical and current state of disease modelling, focusing on the major milestones in the development of stem cell-derived cell types, and how this technology has contributed to our knowledge about the interactions between CMs and key non-myocyte components of the heart in health and disease, in particular, heart failure. Understanding where we stand in the field will be critical for stem cell-based applications, including the modelling of diseases that have complex multicellular dysfunctions.
Collapse
|
184
|
Sung PH, Chua S, Chen KH, Sun CK, Li YC, Huang CR, Luo CW, Chai HT, Lu HI, Yip HK. Role of double knockdown of tPA and MMP-9 on regulating the left ventricular function and remodeling followed by transverse aortic constriction-induced hypertrophic cardiomyopathy in mice. Am J Transl Res 2018; 10:2781-2795. [PMID: 30323866 PMCID: PMC6176237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 08/09/2018] [Indexed: 06/08/2023]
Abstract
This study tested the hypothesis that extracellular matrix accumulation in tPA-/-/MMP-9-/- [double-knockout (DKO)] may be protective against left ventricular (LV) remodeling and dysfunction following transverse aortic constriction (TAC)-induced hypertrophic cardiomyopathy in mice. Wild-type C57BL/6 mice (n = 20) were equally categorized into sham-control (SC1) and TAC1. Similarly, DKO mice (n = 20) were equally divided into two groups (i.e., SC2 and ATC2). By days 28/60 after TAC, LV ejection fraction (LVEF) was significantly higher in TAC2 than TAC1, whereas LV end-systolic/diastolic dimensions displayed an opposite pattern to LVEF between the two groups (all P < 0.05). By day 90, LVEF was significantly higher in SC groups than that in TAC1 and TAC2 without notable difference between the latter two groups, whereas LV end-systolic/diastolic dimensions, cardiomyocyte size and right-ventricular systolic pressure showed an opposite pattern compared with LVEF in all groups (all P < 0.01). Total heart weight was highest in TAC1 and significantly higher in TAC2 than those in the SC groups (P < 0.01). LV myocardial protein expressions of inflammation (TNF-α/NF-κβ), apoptosis (mitochondrial-Bax/cleaved caspase-3/PARP), oxidative stress (NOX-1/NOX-2/oxidized protein), fibrosis (Smad3/TGF-β), DNA/mitochondrial damage (γ-H2AX/cytosolic-cytochrome-C) and LV hypertrophy/pressure-overload (β-MHC/BNP) biomarkers were significantly increased in TAC2 compared to TAC1 and SC groups, and significantly increased in TAC1 compared to SC groups (all P < 0.001). Histopathology demonstrated that the fibrotic/collagen-deposition areas and sarcomere length exhibited an identical pattern to inflammation among the four groups (all P < 0.0001). In conclusion, although tPA-/-/MMP-9-/- seemed to preserve cardiac function in an experimental setting of hypertrophic cardiomyopathy at an early stage, it failed to exert long-term protective effect.
Collapse
Affiliation(s)
- Pei-Hsun Sung
- Division of Cardiology, Department of Internl Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
| | - Sarah Chua
- Division of Cardiology, Department of Internl Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Kuan-Hung Chen
- Department of Anesthesiology, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Cheuk-Kwan Sun
- Department of Emergency Medicine, E-Da Hospital, I-Shou University School of Medicine for International StudentsKaohsiung 82445, Taiwan
| | - Yi-Chen Li
- Division of Cardiology, Department of Internl Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Chi-Ruei Huang
- Division of Cardiology, Department of Internl Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Chi-Wen Luo
- Division of Cardiology, Department of Internl Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Han-Tan Chai
- Division of Cardiology, Department of Internl Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Hung-I Lu
- Division of Thoracic and Cardiovascular Surgery, Department of Surgery, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
| | - Hon-Kan Yip
- Division of Cardiology, Department of Internl Medicine, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of MedicineKaohsiung 83301, Taiwan
- Institute for Translational Research in Biomedicine, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
- Center for Shockwave Medicine and Tissue Engineering, Kaohsiung Chang Gung Memorial HospitalKaohsiung 83301, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical UniversityTaichung 40402, Taiwan
- Department of Nursing, Asia UniversityTaichung 41354, Taiwan
| |
Collapse
|
185
|
Sun S, Kee HJ, Jin L, Ryu Y, Choi SY, Kim GR, Jeong MH. Gentisic acid attenuates pressure overload-induced cardiac hypertrophy and fibrosis in mice through inhibition of the ERK1/2 pathway. J Cell Mol Med 2018; 22:5964-5977. [PMID: 30256522 PMCID: PMC6237595 DOI: 10.1111/jcmm.13869] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/09/2018] [Accepted: 07/30/2018] [Indexed: 01/06/2023] Open
Abstract
We previously reported that gentisic acid (2,5‐dihydroxybenzoic acid) is the third most abundant phenolic component of Dendropanax morbifera branch extracts. Here, we investigated its effects on cardiac hypertrophy and fibrosis in a mouse model of pressure overload and compared them to those of the beta blocker bisoprolol and calcium channel blocker diltiazem. Cardiac hypertrophy was induced in mice by transverse aortic constriction (TAC). Beginning 2 weeks after this procedure, the mice were given daily intraperitoneal injections of gentisic acid (100 mg/kg/d), bisoprolol (5 mg/kg/d) or diltiazem (10 mg/kg/d) for 3 weeks. Cardiac hypertrophy was evaluated by the heart weight‐to‐body weight ratio, the cardiomyocyte cross‐sectional area after haematoxylin and eosin staining, and echocardiography. Markers of cardiac hypertrophy and fibrosis were tested by reverse transcription‐quantitative real‐time polymerase chain reaction, western blotting and Masson's trichrome staining. The suppressive effects of gentisic acid treatment on TAC‐induced cardiac hypertrophy and fibrosis were comparable to those of bisoprolol administration. Cardiac hypertrophy was reversed and left ventricular septum and posterior wall thickness were restored by gentisic acid, bisoprolol and diltiazem treatment. Cardiac hypertrophic marker gene expression and atrial and brain natriuretic peptide levels were decreased by gentisic acid and bisoprolol, as were cardiac (interstitial and perivascular) fibrosis and fibrosis‐related gene expression. Cardiac hypertrophy‐associated upregulation of the transcription factors GATA4 and Sp1 and activation of extracellular signal‐regulated kinase 1/2 were also negated by these drugs. These results suggest that gentisic acid could serve as a therapeutic agent for cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Simei Sun
- Heart Research Center, Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea.,Molecular Medicine, Brain Korea 21 PLUS, Chonnam National University Graduate School, Gwangju, Korea
| | - Hae Jin Kee
- Heart Research Center, Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Li Jin
- The Second Affiliated Hospital & Yuying Children's Hospital Wenzhou Medical University, Wenzhou, China
| | - Yuhee Ryu
- Heart Research Center, Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Sin Young Choi
- Heart Research Center, Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Gwi Ran Kim
- Heart Research Center, Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| | - Myung Ho Jeong
- Heart Research Center, Chonnam National University Hospital, Gwangju, Korea.,Hypertension Heart Failure Research Center, Chonnam National University Hospital, Gwangju, Korea
| |
Collapse
|
186
|
Hermans-Beijnsberger S, van Bilsen M, Schroen B. Long non-coding RNAs in the failing heart and vasculature. Noncoding RNA Res 2018; 3:118-130. [PMID: 30175285 PMCID: PMC6114261 DOI: 10.1016/j.ncrna.2018.04.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 04/09/2018] [Accepted: 04/09/2018] [Indexed: 02/06/2023] Open
Abstract
Following completion of the human genome, it became evident that the majority of our DNA is transcribed into non-coding RNAs (ncRNAs) instead of protein-coding messenger RNA. Deciphering the function of these ncRNAs, including both small- and long ncRNAs (lncRNAs), is an emerging field of research. LncRNAs have been associated with many disorders and a number have been identified as key regulators in the development and progression of disease, including cardiovascular disease (CVD). CVD causes millions of deaths worldwide, annually. Risk factors include coronary artery disease, high blood pressure and ageing. In this review, we will focus on the roles of lncRNAs in the cellular and molecular processes that underlie the development of CVD: cardiomyocyte hypertrophy, fibrosis, inflammation, vascular disease and ageing. Finally, we discuss the biomarker and therapeutic potential of lncRNAs.
Collapse
Affiliation(s)
- Steffie Hermans-Beijnsberger
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - Marc van Bilsen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
- Department of Physiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| | - Blanche Schroen
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, 6200 MD, Maastricht, The Netherlands
| |
Collapse
|
187
|
Awad MA, Aldosari SR, Abid MR. Genetic Alterations in Oxidant and Anti-Oxidant Enzymes in the Vascular System. Front Cardiovasc Med 2018; 5:107. [PMID: 30140678 PMCID: PMC6095034 DOI: 10.3389/fcvm.2018.00107] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular diseases (CVD) are one of the prime causes of mortality worldwide. Experimental animal models have become a valuable tool to investigate and further advance our knowledge on etiology, pathophysiology and intervention. They also provide a great opportunity to understand the contribution of different genes and effector molecules in the pathogenesis and development of diseases at the sub-cellular levels. High levels of reactive oxygen species (ROS) have been associated with the progression of CVD such as ischemic heart disease (IHD), myocardial infarction, hypertension, atherosclerosis, aortic aneurysm, aortic dissection and others. On the contrary, low levels of antioxidants were associated with exacerbated cardiovascular event. Major focus of this review is on vascular pathogenesis that leads to CVD, with special emphasis on the roles of oxidant/antioxidant enzymes in health and disease progression in vascular cells including vascular endothelium. The major oxidant enzymes that have been implicated with the progression of CVD include NADPH Oxidase, nitric oxide synthase, monoamine oxidase, and xanthine oxidoreductase. The major antioxidant enzymes that have been attributed to normalizing the levels of oxidative stress include superoxide dismutases, catalase and glutathione peroxidases (GPx), and thioredoxin. Cardiovascular phenotypes of major oxidants and antioxidants knockout and transgenic animal models are discussed here.
Collapse
Affiliation(s)
- Maan A Awad
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - Sarah R Aldosari
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| |
Collapse
|
188
|
A Reduction in ADAM17 Expression Is Involved in the Protective Effect of the PPAR- α Activator Fenofibrate on Pressure Overload-Induced Cardiac Hypertrophy. PPAR Res 2018; 2018:7916953. [PMID: 30105051 PMCID: PMC6076894 DOI: 10.1155/2018/7916953] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/27/2018] [Accepted: 07/08/2018] [Indexed: 12/21/2022] Open
Abstract
The peroxisome proliferator-activated receptor-α (PPAR-α) agonist fenofibrate ameliorates cardiac hypertrophy; however, its mechanism of action has not been completely determined. Our previous study indicated that a disintegrin and metalloproteinase-17 (ADAM17) is required for angiotensin II-induced cardiac hypertrophy. This study aimed to determine whether ADAM17 is involved in the protective action of fenofibrate against cardiac hypertrophy. Abdominal artery constriction- (AAC-) induced hypertensive rats were used to observe the effects of fenofibrate on cardiac hypertrophy and ADAM17 expression. Primary cardiomyocytes were pretreated with fenofibrate (10 μM) for 1 hour before being stimulated with angiotensin II (100 nM) for another 24 hours. Fenofibrate reduced the ratios of left ventricular weight to body weight (LVW/BW) and heart weight to body weight (HW/BW), left ventricular anterior wall thickness (LVAW), left ventricular posterior wall thickness (LVPW), and ADAM17 mRNA and protein levels in left ventricle in AAC-induced hypertensive rats. Similarly, in vitro experiments showed that fenofibrate significantly attenuated angiotensin II-induced cardiac hypertrophy and diminished ADAM17 mRNA and protein levels in primary cardiomyocytes stimulated with angiotensin II. In summary, a reduction in ADAM17 expression is associated with the protective action of PPAR-α agonists against pressure overload-induced cardiac hypertrophy.
Collapse
|
189
|
Maciejak A, Kostarska-Srokosz E, Gierlak W, Dluzniewski M, Kuch M, Marchel M, Opolski G, Kiliszek M, Matlak K, Dobrzycki S, Lukasik A, Segiet A, Sygitowicz G, Sitkiewicz D, Gora M, Burzynska B. Circulating miR-30a-5p as a prognostic biomarker of left ventricular dysfunction after acute myocardial infarction. Sci Rep 2018; 8:9883. [PMID: 29959359 PMCID: PMC6026144 DOI: 10.1038/s41598-018-28118-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 06/12/2018] [Indexed: 02/06/2023] Open
Abstract
Left ventricular (LV) dysfunction after acute myocardial infarction (AMI) is associated with an increased risk of heart failure (HF) development. Diverse microRNAs (miRNAs) have been shown to appear in the bloodstream following various cardiovascular events. The aim of this study was to identify prognostic miRNAs associated with LV dysfunction following AMI. Patients were divided into subgroups comprising patients who developed or not LV dysfunction within six months of the infarction. miRNA profiles were determined in plasma and serum samples of the patients on the first day of AMI. Levels of 14 plasma miRNAs and 16 serum miRNAs were significantly different in samples from AMI patients who later developed LV dysfunction compared to those who did not. Two miRNAs were up-regulated in both types of material. Validation in an independent group of patients, using droplet digital PCR (ddPCR) confirmed that miR-30a-5p was significantly elevated on admission in those patients who developed LV dysfunction and HF symptoms six months after AMI. A bioinformatics analysis indicated that miR-30a-5p may regulate genes involved in cardiovascular pathogenesis. This study demonstrates, for the first time, a prognostic value of circulating miR-30a-5p and its association with LV dysfunction and symptoms of HF after AMI.
Collapse
Affiliation(s)
- Agata Maciejak
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Edyta Kostarska-Srokosz
- Chair and Department of Cardiology, Hypertension and Internal Medicine, Second Faculty of Medicine, Medical University of Warsaw, Mazovian Bródnowski Hospital, Warsaw, Poland
| | - Wlodzimierz Gierlak
- Chair and Department of Cardiology, Hypertension and Internal Medicine, Second Faculty of Medicine, Medical University of Warsaw, Mazovian Bródnowski Hospital, Warsaw, Poland
| | - Miroslaw Dluzniewski
- Cardiology Department, Midtown Medical Center, Mazovia Brodno Hospital, Warsaw, Poland
| | - Marek Kuch
- Chair and Department of Cardiology, Hypertension and Internal Medicine, Second Faculty of Medicine, Medical University of Warsaw, Mazovian Bródnowski Hospital, Warsaw, Poland
| | - Michal Marchel
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Grzegorz Opolski
- 1st Chair and Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Marek Kiliszek
- Department of Cardiology and Internal Diseases, Military Institute of Medicine, Warsaw, Poland
| | - Krzysztof Matlak
- Department of Cardiac Surgery, Medical University of Bialystok, Bialystok, Poland
| | - Slawomir Dobrzycki
- Department of Invasive Cardiology, Medical University of Bialystok, Bialystok, Poland
| | - Anna Lukasik
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Agnieszka Segiet
- 1st Faculty of Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Grazyna Sygitowicz
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Dariusz Sitkiewicz
- Department of Clinical Chemistry and Laboratory Diagnostics, Medical University of Warsaw, Warsaw, Poland
| | - Monika Gora
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| | - Beata Burzynska
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland.
| |
Collapse
|
190
|
Chiang JT, Badrealam KF, Shibu MA, Cheng SF, Shen CY, Chang CF, Lin YM, Viswanadha VP, Liao SC, Huang CY. Anti-Apoptosis and Anti-Fibrosis Effects of Eriobotrya Japonica in Spontaneously Hypertensive Rat Hearts. Int J Mol Sci 2018; 19:ijms19061638. [PMID: 29857545 PMCID: PMC6032044 DOI: 10.3390/ijms19061638] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 05/20/2018] [Accepted: 05/22/2018] [Indexed: 01/02/2023] Open
Abstract
Myocardial apoptosis and fibrosis represent important contributing factors for development of hypertension-induced heart failure. The present study aims to investigate the potential effects of Eriobotrya japonica leaf extract (EJLE) against hypertension-induced cardiac apoptosis and fibrosis in spontaneously hypertensive rats (SHRs). Twelve-week-old male rats were randomly divided into four different groups; control Wistar Kyoto (WKY) rats, hypertensive SHR rats, SHR rats treated with a low dose (100 mg/kg body weight) of EJLE and SHR rats treated with a high dose (300 mg/kg body weight) of EJLE. Animals were acclimatized for 4 weeks and thereafter were gastric fed for 8 weeks with two doses of EJLE per week. The rats were then euthanized following cardiac functional analysis by echocardiography. The cardiac tissue sections were examined by Terminal Deoxynucleotidyl Transferase-Mediated Deoxyuridine Triphosphate (dUTP) Nick End-Labeling (TUNEL) assay, histological staining and Western blotting to assess the cardio-protective effects of EJ in SHR animals. Echocardiographic measurements provided convincing evidence to support the ability of EJ to ameliorate crucial cardiac functional characteristics. Furthermore, our results reveal that supplementation of EJLE effectively attenuated cardiac apoptosis and fibrosis and also enhanced cell survival in hypertensive SHR hearts. Thus, the present study concludes that EJLE potentially provides cardio-protective effects against hypertension-induced cardiac apoptosis and fibrosis in SHR animals.
Collapse
Affiliation(s)
- Jui-Ting Chiang
- Graduate Institute of Aging Medicine, China Medical University, Taichung 40402, Taiwan.
| | - Khan Farheen Badrealam
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Marthandam Asokan Shibu
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
| | - Sue-Fei Cheng
- Department of Pharmacy, Taiwan Adventist Hospital, Taipei 10556, Taiwan.
- Mackay Junior College of Medicine, Nursing, and Management, New Taipei City 11260, Taiwan.
| | - Chia-Yao Shen
- Department of Nursing, Mei Ho University, 23 Pingguang Road, Pingtung 91202, Taiwan.
| | - Chih-Feng Chang
- Department of Internal Medicine, Division of Cardiology, Taichung Armed Forces Taichung General Hospital, Taichung 40402, Taiwan.
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua 500, Taiwan.
| | | | - Shih-Chieh Liao
- School of Medicine, College of Medicine, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
| | - Chih-Yang Huang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 40402, Taiwan.
- Graduate Institute of Chinese Medical Science, China Medical University, 91 Hsueh-Shih Road, Taichung 40402, Taiwan.
- Department of Biological Science, Asia University, Taichung 40402, Taiwan.
| |
Collapse
|
191
|
Lourenço MAM, Braz MG, Aun AG, Pereira BLB, Figueiredo AM, da Silva RAC, Kazmarek EM, Alegre PHC, Bachiega TF, Zanati SG, Azevedo PS, Polegato BF, Fernandes AAH, de Paiva SAR, Zornoff LAM, Minicucci MF. Spondias mombin supplementation attenuated cardiac remodelling process induced by tobacco smoke. J Cell Mol Med 2018; 22:3996-4004. [PMID: 29808581 PMCID: PMC6050477 DOI: 10.1111/jcmm.13683] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 04/10/2018] [Indexed: 01/31/2023] Open
Abstract
The objective of this study was to investigate the influence of Spondias mombin (SM) supplementation on the cardiac remodelling process induced by exposure to tobacco smoke (ETS) in rats. Male Wistar rats were divided into 4 groups: group C (control, n = 20) comprised animals not exposed to cigarette smoke and received standard chow; group ETS (n = 20) comprised animals exposed to cigarette smoke and received standard chow; group ETS100 (n = 20) received standard chow supplemented with 100 mg/kg body weight/d of SM; and group ETS250 (n = 20) received standard chow supplemented with 250 mg/kg body weight/d of SM. The observation period was 2 months. The ETS animals had higher values of left cardiac chamber diameters and of left ventricular mass index. SM supplementation attenuated these changes. In addition, the myocyte cross‐sectional area (CSA) was lower in group C compared with the ETS groups; however, the ETS250 group had lower values of CSA compared with the ETS group. The ETS group also showed higher cardiac levels of lipid hydroperoxide (LH) compared with group C; and, groups ETS100 and ETS250 had lower concentrations of LH compared with the ETS group. Regarding energy metabolism, SM supplementation decreased glycolysis and increased the β‐oxidation and the oxidative phosphorylation. There were no differences in the expression of Nrf‐2, SIRT‐1, NF‐κB, interferon‐gamma and interleukin 10. In conclusion, our results suggest that ETS induced the cardiac remodelling process. In addition, SM supplementation attenuated this process, along with oxidative stress reduction and energy metabolism modulation.
Collapse
Affiliation(s)
| | - Mariana Gobbo Braz
- Department of Anesthesiology, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | - Aline Garcia Aun
- Department of Anesthesiology, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | | | - Amanda Menezes Figueiredo
- Internal Medicine Department, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | | | - Elisa Moya Kazmarek
- Internal Medicine Department, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | | | - Tatiana Fernanda Bachiega
- Internal Medicine Department, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | - Silmeia Garcia Zanati
- Internal Medicine Department, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | - Paula Schmidt Azevedo
- Internal Medicine Department, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | - Bertha Furlan Polegato
- Internal Medicine Department, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | - Ana Angélica Henrique Fernandes
- Chemistry and Biochemistry Department, Institute of Biological Sciences, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| | | | | | - Marcos Ferreira Minicucci
- Internal Medicine Department, Botucatu Medical School, UNESP-São Paulo State University, Botucatu, Brazil
| |
Collapse
|
192
|
Ghosh N, Katare R. Molecular mechanism of diabetic cardiomyopathy and modulation of microRNA function by synthetic oligonucleotides. Cardiovasc Diabetol 2018; 17:43. [PMID: 29566757 PMCID: PMC5863891 DOI: 10.1186/s12933-018-0684-1] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Accepted: 03/10/2018] [Indexed: 02/06/2023] Open
Abstract
Diabetic cardiomyopathy (DCM) is a chronic complication in individuals with diabetes and is characterized by ventricular dilation and hypertrophy, diastolic dysfunction, decreased or preserved systolic function and reduced ejection fraction eventually resulting in heart failure. Despite being well characterized, the fundamental mechanisms leading to DCM are still elusive. Recent studies identified the involvement of small non-coding small RNA molecules such as microRNAs (miRs) playing a key role in the etiology of DCM. Therefore, miRs associated with DCM represents a new class of targets for the development of mechanistic therapeutics, which may yield marked benefits compared to other therapeutic approaches. Indeed, few miRs currently under active clinical investigation, with many expressing cautious optimism that miRs based therapies will succeed in the coming years. The major caution in using miRs based therapy is the need to improve the stability and specificity following systemic injection, which can be achieved through chemical and structural modification. In this review, we first discuss the established role of miRs in DCM and the advances in miRs based therapeutic strategies for the prevention/treatment of DCM. We next discuss the currently employed chemical modification of miR oligonucleotides and their utility in therapies specifically focusing on the DCM. Finally, we summarize the commonly used delivery system and approaches for assessment of miRNA modulation and potential off-target effects.
Collapse
Affiliation(s)
- Nilanjan Ghosh
- Department of Physiology-HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010 New Zealand
| | - Rajesh Katare
- Department of Physiology-HeartOtago, University of Otago, 270, Great King Street, Dunedin, 9010 New Zealand
| |
Collapse
|
193
|
Inoue T, Sonoda N, Hiramatsu S, Kimura S, Ogawa Y, Inoguchi T. Serum Bilirubin Concentration is Associated with Left Ventricular Remodeling in Patients with Type 2 Diabetes Mellitus: A Cohort Study. Diabetes Ther 2018; 9:331-338. [PMID: 29335891 PMCID: PMC5801254 DOI: 10.1007/s13300-018-0368-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Previous studies have shown that serum bilirubin concentration is inversely associated with the risk of cardiovascular disease. The relationship between serum bilirubin concentration and left ventricular geometry, however, has not been investigated in patients with diabetes mellitus. METHODS In this cohort study, 158 asymptomatic patients with type 2 diabetes mellitus without overt heart disease were enrolled. Left ventricular structure and function were assessed using echocardiography. Serum bilirubin concentration, glycemic control, lipid profile, and other clinical characteristics were evaluated, and their association with left ventricular geometry was determined. Patients with New York Heart Association Functional Classification greater than I, left ventricular ejection fraction less than 50%, history of coronary artery disease, severe valvulopathy, chronic atrial fibrillation, or creatinine clearance less than 30 ml/min, and those receiving insulin treatment, were excluded. RESULTS Univariate analyses showed that relative wall thickness (RWT) was significantly correlated with diastolic blood pressure (P = 0.003), HbA1c (P = 0.024), total cholesterol (P = 0.043), urinary albumin (P = 0.023), and serum bilirubin concentration (P = 0.009). There was no association between left ventricular mass index and serum bilirubin concentration. Multivariate linear regression analysis showed that log RWT was positively correlated with diastolic blood pressure (P = 0.010) and that log RWT was inversely correlated with log bilirubin (P = 0.003). In addition, the patients with bilirubin less than 0.8 mg/dl had a higher prevalence of concentric left ventricular remodeling compared with those with bilirubin 0.8 mg/dl or more. CONCLUSION Our study shows that the serum bilirubin concentration may be associated with the progression of concentric left ventricular remodeling in patients with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Tomoaki Inoue
- Department of Diabetes Mellitus and Endocrinology, Tagawa Municipal Hospital, Fukuoka, Japan.
- Department of Diabetes Mellitus and Endocrinology, Hamanomachi Hospital, Fukuoka, Japan.
| | - Noriyuki Sonoda
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinsuke Hiramatsu
- Department of Diabetes Mellitus and Endocrinology, Hamanomachi Hospital, Fukuoka, Japan
| | - Shinichiro Kimura
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Molecular and Cellular Metabolism, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Toyoshi Inoguchi
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
194
|
Ianni A, Yuan X, Bober E, Braun T. Sirtuins in the Cardiovascular System: Potential Targets in Pediatric Cardiology. Pediatr Cardiol 2018; 39:983-992. [PMID: 29497772 PMCID: PMC5958173 DOI: 10.1007/s00246-018-1848-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 02/24/2018] [Indexed: 12/14/2022]
Abstract
Cardiovascular diseases represent a major cause of death and morbidity. Cardiac and vascular pathologies develop predominantly in the aged population in part due to lifelong exposure to numerous risk factors but are also found in children and during adolescence. In comparison to adults, much has to be learned about the molecular pathways driving cardiovascular diseases in the pediatric population. Sirtuins are highly conserved enzymes that play pivotal roles in ensuring cardiac homeostasis under physiological and stress conditions. In this review, we discuss novel findings about the biological functions of these molecules in the cardiovascular system and their possible involvement in pediatric cardiovascular diseases.
Collapse
Affiliation(s)
- Alessandro Ianni
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Ludwig Strasse 43, 61231, Bad Nauheim, Germany.
| | - Xuejun Yuan
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Ludwig Strasse 43, 61231, Bad Nauheim, Germany
| | - Eva Bober
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Ludwig Strasse 43, 61231, Bad Nauheim, Germany
| | - Thomas Braun
- Department of Cardiac Development and Remodeling, Max-Planck-Institute for Heart and Lung Research, Ludwig Strasse 43, 61231, Bad Nauheim, Germany.
| |
Collapse
|
195
|
Muscular Atrophy in Cardiovascular Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:369-391. [DOI: 10.1007/978-981-13-1435-3_17] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
196
|
Wang Y, Wang Q, Yu W, Du H. Crocin Attenuates Oxidative Stress and Myocardial Infarction Injury in Rats. Int Heart J 2018; 59:387-393. [DOI: 10.1536/ihj.17-114] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- Yongwang Wang
- Department of Anesthesiology, Tianjin First Center Hospital
| | - Qingping Wang
- Department of Anesthesiology, Tianjin First Center Hospital
| | - Wenli Yu
- Department of Anesthesiology, Tianjin First Center Hospital
| | - Hongyin Du
- Department of Anesthesiology, Tianjin First Center Hospital
| |
Collapse
|
197
|
Li F, Zong J, Zhang H, Zhang P, Xu L, Liang K, Yang L, Yong H, Qian W. Orientin Reduces Myocardial Infarction Size via eNOS/NO Signaling and Thus Mitigates Adverse Cardiac Remodeling. Front Pharmacol 2017; 8:926. [PMID: 29311930 PMCID: PMC5742593 DOI: 10.3389/fphar.2017.00926] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/06/2017] [Indexed: 01/04/2023] Open
Abstract
Orientin is a flavonoid extracted from Chinese traditional herb, Polygonum orientale L. Previous study has reported that orientin protected myocardial from ischemia reperfusion injury. However, whether orientin could protect against cardiac remodeling after myocardial injury remains unclear. The aim of our study is to investigate the effects of orientin in the progression of cardiac remodeling after myocardial infarction (MI). Mice cardiac remodeling model was established by left coronary artery ligation surgery. Experimental groups were as follows: vehicle-sham, orientin-sham, vehicle-MI, and orientin-MI. Animals were treated with vehicle or orientin (40 mg/kg) for 25 days starting 3 days after surgery. After 4 weeks of MI, mice with orientin treatment had decreased mortality and improved cardiac function. Significantly, at 4 weeks post-MI, orientin treatment decreased fibrosis, inflammatory response, and cardiomyocyte apoptosis. Furthermore, orientin treatment attenuated the hypoxia-induced neonatal rat cardiomyocyte apoptosis and increased cell viability. Additionally, orientin supplementation mitigated oxidative stress in remodeling heart tissue and cardiomyocytes exposed to hypoxia as measured by 2′,7′-dichlorodihydrofluorescein diacetate fluorescent probe. Mechanistically, orientin promotes cardioprotection by activating the eNOS/NO signaling cascades, which was confirmed by eNOS inhibitor (L-NAME) in vitro and in vivo. Inhibition of oxidative stress by orientin via eNOS/NO signaling cascades in the heart may represent a potential therapy for cardiac remodeling.
Collapse
Affiliation(s)
- Fangfang Li
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Jing Zong
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Hao Zhang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Peijie Zhang
- Emergency Department, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Luhong Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Kai Liang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Lu Yang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Hui Yong
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| | - Wenhao Qian
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.,Institute of Cardiovascular Disease Research, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
198
|
Roever L, Palandri Chagas AC. Editorial: Cardiac Remodeling: New Insights in Physiological and Pathological Adaptations. Front Physiol 2017; 8:751. [PMID: 29018366 PMCID: PMC5622945 DOI: 10.3389/fphys.2017.00751] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/14/2017] [Indexed: 12/20/2022] Open
Affiliation(s)
| | - Antonio C. Palandri Chagas
- Heart Institute (InCor), HCFMUSP University of São Paulo Medical School, São Paulo, Brazil
- Faculty of Medicine ABC, Santo André, Brazil
| |
Collapse
|
199
|
Garcia LR, Polegato BF, Zornoff LAM. Challenges of Translational Science. Arq Bras Cardiol 2017; 108:388-389. [PMID: 28591317 PMCID: PMC5444883 DOI: 10.5935/abc.20170061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 04/12/2017] [Indexed: 01/12/2023] Open
Affiliation(s)
- Leonardo R Garcia
- Departamento de Clínica Médica - Faculdade de Medicina de Botucatu, Botucatu, SP - Brazil
| | - Bertha F Polegato
- Departamento de Clínica Médica - Faculdade de Medicina de Botucatu, Botucatu, SP - Brazil
| | - Leonardo A M Zornoff
- Departamento de Clínica Médica - Faculdade de Medicina de Botucatu, Botucatu, SP - Brazil
| |
Collapse
|
200
|
Minicucci M, Oliveira F, Santos P, Polegato B, Roscani M, Fernandes AA, Lustosa B, Paiva S, Zornoff L, Azevedo P. Pentoxifylline Attenuates Cardiac Remodeling Induced by Tobacco Smoke Exposure. Arq Bras Cardiol 2016; 106:396-403. [PMID: 27096523 PMCID: PMC4914004 DOI: 10.5935/abc.20160057] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 02/19/2016] [Indexed: 12/25/2022] Open
Abstract
Background Tobacco smoke exposure is an important risk factor for cardiac remodeling.
Under this condition, inflammation, oxidative stress, energy metabolism
abnormalities, apoptosis, and hypertrophy are present. Pentoxifylline has
anti‑inflammatory, anti-apoptotic, anti-thrombotic and anti-proliferative
properties. Objective The present study tested the hypothesis that pentoxifylline would attenuate
cardiac remodeling induced by smoking. Methods Wistar rats were distributed in four groups: Control (C), Pentoxifylline
(PX), Tobacco Smoke (TS), and PX-TS. After two months, echocardiography,
invasive blood pressure measurement, biochemical, and histological studies
were performed. The groups were compared by two-way ANOVA with a
significance level of 5%. Results TS increased left atrium diameter and area, which was attenuated by PX. In
the isolated heart study, TS lowered the positive derivate (+dp/dt), and
this was attenuated by PX. The antioxidants enzyme superoxide dismutase and
glutathione peroxidase were decreased in the TS group; PX recovered these
activities. TS increased lactate dehydrogenase (LDH) and decreased
3-hydroxyacyl Coenzyme A dehydrogenases (OH-DHA) and citrate synthase (CS).
PX attenuated LDH, 3-OH-DHA and CS alterations in TS-PX group. TS increased
IL-10, ICAM-1, and caspase-3. PX did not influence these variables. Conclusion TS induced cardiac remodeling, associated with increased inflammation,
oxidative stress, apoptosis, and changed energy metabolism. PX attenuated
cardiac remodeling by reducing oxidative stress and improving cardiac
bioenergetics, but did not act upon cardiac cytokines and apoptosis.
Collapse
Affiliation(s)
- Marcos Minicucci
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Fernando Oliveira
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Priscila Santos
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Bertha Polegato
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Meliza Roscani
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Ana Angelica Fernandes
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Beatriz Lustosa
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Sergio Paiva
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Leonardo Zornoff
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| | - Paula Azevedo
- Faculdade de Medicina de Botucatu, Universidade Estadual Paulista, São Paulo, SP, Brazil
| |
Collapse
|