1
|
Sun Y, Zhong N, Zhu X, Fan Q, Li K, Chen Y, Wan X, He Q, Xu Y. Identification of important genes associated with acute myocardial infarction using multiple cell death patterns. Cell Signal 2023; 112:110921. [PMID: 37839544 DOI: 10.1016/j.cellsig.2023.110921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 10/01/2023] [Accepted: 10/09/2023] [Indexed: 10/17/2023]
Abstract
Acute myocardial infarction (AMI) is a global health threat, and programmed cell death (PCD) plays a crucial role in its occurrence and development. In this study, integrated bioinformatics tools were used to explore new biomarkers and therapeutic targets in AMI. Thirteen types of PCD-related genes were identified through literature review, KEGG, and GSEA pathways. Gene expression matrices and clinical data from AMI patients and healthy controls were obtained from the GEO database. Statistical analysis in R identified 377 differentially expressed genes in AMI patients. Intersection analysis between the differentially expressed genes and PCD-related genes revealed 24 genes positively correlated with immune cells such as Neutrophils and Monocytes, while negatively correlated with T cells CD4 memory resting and Plasma cells. Unsupervised clustering analysis divided patients into two groups (C1 and C2) based on the expression levels of these 24 genes. GSVA analysis showed that C2 patients were more active in pathways related to maintaining normal cell morphology and promoting phagocytosis, suggesting a lower programmed cell death rate and a higher tendency to maintain cell survival. Two hub genes, TNFAIP3 and TP53INP2, were identified through LASSO regression analysis and SVM-RFE, and were validated using an external dataset and RT-qPCR、Western blot and ELISA analysis. These hub genes showed significantly higher expression and protein secretion levels in AMI patients compared to healthy individuals. Overall, regulating and controlling PCD, particularly through the identified hub genes, TNFAIP3 and TP53INP2, may provide new therapeutic strategies for improving the prognosis of AMI patients and preventing heart failure.
Collapse
Affiliation(s)
- Yong Sun
- Clifford Hospital, Guangzhou, China.
| | - Nan Zhong
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xianqiong Zhu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | - Keyi Li
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | | | | | - Qi He
- Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Xu
- Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
2
|
Ribeiro ASF, Zerolo BE, López-Espuela F, Sánchez R, Fernandes VS. Cardiac System during the Aging Process. Aging Dis 2023:AD.2023.0115. [PMID: 37163425 PMCID: PMC10389818 DOI: 10.14336/ad.2023.0115] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/15/2023] [Indexed: 05/12/2023] Open
Abstract
The aging process is accompanied by a continuous decline of the cardiac system, disrupting the homeostatic regulation of cells, organs, and systems. Aging increases the prevalence of cardiovascular diseases, thus heart failure and mortality. Understanding the cardiac aging process is of pivotal importance once it allows us to design strategies to prevent age-related cardiac events and increasing the quality of live in the elderly. In this review we provide an overview of the cardiac aging process focus on the following topics: cardiac structural and functional modifications; cellular mechanisms of cardiac dysfunction in the aging; genetics and epigenetics in the development of cardiac diseases; and aging heart and response to the exercise.
Collapse
Affiliation(s)
| | - Blanca Egea Zerolo
- Escuela de Enfermería y Fisioterapia San Juan de Dios. Universidad Pontificia Comillas, Madrid, Spain
| | - Fidel López-Espuela
- Metabolic Bone Diseases Research Group, Nursing and Occupational Therapy College, University of Extremadura, Caceres, Spain
| | - Raúl Sánchez
- Unidad de Cardiopatías Congénitas, Hospital Universitario La Paz, Madrid, Spain
| | - Vítor S Fernandes
- Departamento de Fisiología, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
3
|
Mendoza A, Karch J. Keeping the beat against time: Mitochondrial fitness in the aging heart. FRONTIERS IN AGING 2022; 3:951417. [PMID: 35958271 PMCID: PMC9360554 DOI: 10.3389/fragi.2022.951417] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Accepted: 06/30/2022] [Indexed: 11/21/2022]
Abstract
The process of aging strongly correlates with maladaptive architectural, mechanical, and biochemical alterations that contribute to the decline in cardiac function. Consequently, aging is a major risk factor for the development of heart disease, the leading cause of death in the developed world. In this review, we will summarize the classic and recently uncovered pathological changes within the aged heart with an emphasis on the mitochondria. Specifically, we describe the metabolic changes that occur in the aging heart as well as the loss of mitochondrial fitness and function and how these factors contribute to the decline in cardiomyocyte number. In addition, we highlight recent pharmacological, genetic, or behavioral therapeutic intervention advancements that may alleviate age-related cardiac decline.
Collapse
Affiliation(s)
- Arielys Mendoza
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| | - Jason Karch
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, United States
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
4
|
Naito R, Kasai T, Dohi T, Takaya H, Narui K, Momomura SI. Factors Associated With the Improvement of Left Ventricular Systolic Function by Continuous Positive Airway Pressure Therapy in Patients With Heart Failure With Reduced Ejection Fraction and Obstructive Sleep Apnea. Front Neurol 2022; 13:781054. [PMID: 35359656 PMCID: PMC8960234 DOI: 10.3389/fneur.2022.781054] [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: 09/22/2021] [Accepted: 02/14/2022] [Indexed: 11/25/2022] Open
Abstract
Background Obstructive sleep apnea (OSA) is a potential risk factor in cardiovascular diseases, including arrhythmia, coronary artery disease, and heart failure (HF). Continuous positive airway pressure (CPAP) therapy is an effective therapy for OSA and the underlying HF, partly through a 5–9% increase in the left ventricular ejection fraction (LVEF). However, the data on the factors associated with the efficacy of CPAP on LVEF in patients with HF complicated by OSA are scarce. This study aimed to investigate whether LVEF improves in patients with OSA and HF after 1 month of CPAP therapy, and to clarify which factors are associated with the degree of LVEF improvement. Method This was a prospective, single-arm, open-label study. We enrolled moderate-to-severe patients with OSA and HF who were being followed up at the cardiovascular center of Toranomon Hospital (Tokyo, Japan). The parameters of sleep study and LVEF were assessed at the baseline and after 1 month of CPAP. The multivariate regression analyses, with changes in LVEF as a dependent variable, were performed to determine the factors that were associated with the degree of LVEF improvement. Results We analyzed 55 consecutive patients with OSA and HF (mean age: 60.7 ± 12.2 years, mean LVEF value: 37.2 ± 9.8%). One month of CPAP treatment decreased the apnea-hypopnea index (AHI) from 45.3 ± 16.1 to 5.4 ± 4.1 per hour, and the LVEF improved from 37.2 ± 9.8 to 43.2 ± 11.7%. The multivariate regression analyses demonstrated that age and body mass index (BMI) were significant determinants of LVEF improvement. Conclusion The LVEF improved significantly after 1 month of CPAP therapy in Japanese patients with OSA and HF. Multivariate regression analyses indicated that an improvement in LVEF was likely to be observed in young patients with obesity.
Collapse
Affiliation(s)
- Ryo Naito
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Takatoshi Kasai
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Cardiovascular Respiratory Sleep Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Sleep Center, Toranomon Hospital, Tokyo, Japan
- *Correspondence: Takatoshi Kasai
| | - Tomotaka Dohi
- Department of Cardiovascular Biology and Medicine, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | | | - Koji Narui
- Sleep Center, Toranomon Hospital, Tokyo, Japan
| | | |
Collapse
|
5
|
The Impact of Melatonin Supplementation and NLRP3 Inflammasome Deletion on Age-Accompanied Cardiac Damage. Antioxidants (Basel) 2021; 10:antiox10081269. [PMID: 34439517 PMCID: PMC8389221 DOI: 10.3390/antiox10081269] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 08/06/2021] [Indexed: 12/16/2022] Open
Abstract
To investigate the role of NLRP3 inflammasome in cardiac aging, we evaluate here morphological and ultrastructural age-related changes of cardiac muscles fibers in wild-type and NLRP3-knockout mice, as well as studying the beneficial effect of melatonin therapy. The results clarified the beginning of the cardiac sarcopenia at the age of 12 months, with hypertrophy of cardiac myocytes, increased expression of β-MHC, appearance of small necrotic fibers, decline of cadiomyocyte number, destruction of mitochondrial cristae, appearance of small-sized residual bodies, and increased apoptotic nuclei ratio. These changes were progressed in the cardiac myocytes of 24 old mice, accompanied by excessive collagen deposition, higher expressions of IL-1α, IL-6, and TNFα, complete mitochondrial vacuolation and damage, myofibrils disorganization, multivesicular bodies formation, and nuclear fragmentation. Interestingly, cardiac myocytes of NLRP3-/- mice showed less detectable age-related changes compared with WT mice. Oral melatonin therapy preserved the normal cardiomyocytes structure, restored cardiomyocytes number, and reduced β-MHC expression of cardiac hypertrophy. In addition, melatonin recovered mitochondrial architecture, reduced apoptosis and multivesicular bodies' formation, and decreased expressions of β-MHC, IL-1α, and IL-6. Fewer cardiac sarcopenic changes and highly remarkable protective effects of melatonin treatment detected in aged cardiomyocytes of NLRP3-/- mice compared with aged WT animals, confirming implication of the NLRP3 inflammasome in cardiac aging. Thus, NLRP3 suppression and melatonin therapy may be therapeutic approaches for age-related cardiac sarcopenia.
Collapse
|
6
|
Gebhard C, Maredziak M, Messerli M, Buechel RR, Lin F, Gransar H, Achenbach S, Al-Mallah MH, Andreini D, Bax JJ, Berman DS, Budoff MJ, Cademartiri F, Callister TQ, Chang HJ, Chinnaiyan K, Chow BJW, Cury RC, DeLago A, Feuchtner G, Hadamitzky M, Hausleiter J, Kim YJ, Leipsic J, Maffei E, Marques H, Gonçalves PDA, Pontone G, Raff GL, Rubinshtein R, Shaw LJ, Villines TC, Lu Y, Jones EC, Peña JM, Min JK, Kaufmann PA. Increased long-term mortality in women with high left ventricular ejection fraction: data from the CONFIRM (COronary CT Angiography EvaluatioN For Clinical Outcomes: An InteRnational Multicenter) long-term registry. Eur Heart J Cardiovasc Imaging 2021; 21:363-374. [PMID: 31985803 DOI: 10.1093/ehjci/jez321] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 08/15/2019] [Accepted: 12/23/2019] [Indexed: 12/19/2022] Open
Abstract
AIMS There are significant sex-specific differences in left ventricular ejection fraction (LVEF), with a higher LVEF being observed in women. We sought to assess the clinical relevance of an increased LVEF in women and men. METHODS AND RESULTS A total of 4632 patients from the CONFIRM (COronary CT Angiography EvaluatioN For Clinical Outcomes: An InteRnational Multicenter) registry (44.8% women; mean age 58.7 ± 13.2 years in men and 59.5 ± 13.3 years in women, P = 0.05), in whom LVEF was measured by cardiac computed tomography, were categorized according to LVEF (low <55%, normal 55-65%, and high >65%). The prevalence of high LVEF was similar in both sexes (33.5% in women and 32.5% in men, P = 0.46). After 6 years of follow-up, no difference in mortality was observed in patients with high LVEF in the overall cohort (P = 0.41). When data were stratified by sex, women with high LVEF died more often from any cause as compared to women with normal LVEF (8.6% vs. 7.1%, log rank P = 0.032), while an opposite trend was observed in men (5.8% vs. 6.8% in normal LVEF, log rank P = 0.89). Accordingly, a first order interaction term of male sex and high LVEF was significant (hazard ratios 0.63, 95% confidence intervals 0.41-0.98, P = 0.043) in a Cox regression model of all-cause mortality adjusted for age, cardiovascular risk factors, and severity of coronary artery disease (CAD). CONCLUSION Increased LVEF is highly prevalent in patients referred for evaluation of CAD and is associated with an increased risk of death in women, but not in men. Differentiating between normal and hyperdynamic left ventricles might improve risk stratification in women with CAD. CLINICAL TRIAL REGISTRATION https://clinicaltrials.gov/ct2/show/NCT01443637.
Collapse
Affiliation(s)
- Catherine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Monika Maredziak
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland.,Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Michael Messerli
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| | - Fay Lin
- Dalio Institute of Cardiovascular Imaging, Weill Cornell Medical College and New York Presbyterian Hospital, 1300 York Avenue, New York, NY 10065, USA
| | - Heidi Gransar
- Department of Imaging, Cedars-Sinai Medical Center, 8705 Gracie Allen Dr, Los Angeles, CA 90048, USA
| | - Stephan Achenbach
- Department of Cardiology, Friedrich-Alexander-University Erlangen-Nuremberg, Maximiliansplatz 2, 91054 Erlangen, Germany
| | - Mouaz H Al-Mallah
- King Saud bin Abdulaziz University for Health Sciences, King Abdullah International Medical Research Center, King Abdulaziz Cardiac Center, Ministry of National Guard, Health Affairs, Ar Rimayah, Riyadh 14611, Saudi Arabia
| | - Daniele Andreini
- Centro Cardiologico Monzino, IRCCS Milan, Via Carlo Parea, 4, 20138 Milan, Italy
| | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Center, Albinusdreef 2, 2333 Leiden, The Netherlands
| | - Daniel S Berman
- Department of Imaging and Medicine, Cedars Sinai Medical Center, 8705 Gracie Allen Dr, Los Angeles, CA, USA
| | - Matthew J Budoff
- Department of Medicine, Los Angeles Biomedical Research Institute, 1124 W Carson St, Torrance, CA 90502, USA
| | - Filippo Cademartiri
- Cardiovascular Imaging Center, SDN IRCCS, via Gianturco 113, 80143 Naples, Italy
| | - Tracy Q Callister
- Tennessee Heart and Vascular Institute, 353 New Shackle Island Rd, Hendersonville, TN 37075, USA
| | - Hyuk-Jae Chang
- Division of Cardiology, Severance Cardiovascular Hospital, Severance Biomedical Science Institute, Yonsei University College of Medicine, Yonsei University Health System, 50-1 Yonsei-Ro, Seodaemun-Gu, Seoul 03722, South Korea
| | - Kavitha Chinnaiyan
- Department of Cardiology, William Beaumont Hospital, 3601 W 13 Mile Rd, Royal Oak, MI 48073, USA
| | - Benjamin J W Chow
- Department of Medicine and Radiology, University of Ottawa, 501 Smyth Road, Ottawa, Ontario K1H 8L6, Canada
| | - Ricardo C Cury
- Department of Radiology, Miami Cardiac and Vascular Institute, 8900 N Kendall Dr, Miami, FL 33176, USA
| | - Augustin DeLago
- Capitol Cardiology Associates, Corporate Woods 7 Southwoods Blvd., Albany, NY 12211, USA
| | - Gudrun Feuchtner
- Department of Radiology, Medical University of Innsbruck, Christoph-Probst-Platz 1, Innrain 52, 6020 Innsbruck, Austria
| | - Martin Hadamitzky
- Department of Radiology and Nuclear Medicine, German Heart Center Munich, Lazarettstraße 36, 80636 Munich, Germany
| | - Joerg Hausleiter
- Medizinische Klinik I der Ludwig-Maximilians-Universität München, Marchioninistr. 15, 81377 Munich, Germany
| | - Yong-Jin Kim
- Seoul National University Hospital, 101 Daehak-ro Jongno-gu, Seoul, South Korea
| | - Jonathon Leipsic
- Department of Medicine and Radiology, University of British Columbia, 2775 Laurel St, Vancouver, BC V5Z 1M9, Canada
| | - Erica Maffei
- Department of Radiology, Area Vasta 1/ASUR Marche, Viale Federico Comandino, 70, 61029 Urbino, Italy
| | - Hugo Marques
- UNICA, Unit of Cardiovascular Imaging, Hospital da Luz, Avenida Lusíada, 100, 1500-650 Lisboa, Portugal
| | - Pedro de Araújo Gonçalves
- UNICA, Unit of Cardiovascular Imaging, Hospital da Luz, Avenida Lusíada, 100, 1500-650 Lisboa, Portugal
| | - Gianluca Pontone
- Centro Cardiologico Monzino, IRCCS Milan, Via Carlo Parea, 4, 20138 Milan, Italy
| | - Gilbert L Raff
- Department of Cardiology, William Beaumont Hospital, 3601 W 13 Mile Rd, Royal Oak, MI 48073, USA
| | - Ronen Rubinshtein
- Department of Cardiology at the Lady Davis Carmel Medical Center, The Ruth and Bruce Rappaport School of Medicine, Technion-Israel Institute of Technology, Haifa 34362, Israel
| | - Leslee J Shaw
- Dalio Institute of Cardiovascular Imaging, Weill Cornell Medical College and New York Presbyterian Hospital, 1300 York Avenue, New York, NY 10065, USA
| | - Todd C Villines
- Cardiology Service, Walter Reed National Military Center, 8901 Rockville Pike, Bethesda, MD 20889, USA
| | - Yao Lu
- Department of Healthcare Policy and Research, New York-Presbyterian Hospital and the Weill Cornell Medical College, 402 E. 67th Street, New York, NY 10065, USA
| | - Erica C Jones
- Dalio Institute of Cardiovascular Imaging, Weill Cornell Medical College and New York Presbyterian Hospital, 1300 York Avenue, New York, NY 10065, USA
| | - Jessica M Peña
- Dalio Institute of Cardiovascular Imaging, Weill Cornell Medical College and New York Presbyterian Hospital, 1300 York Avenue, New York, NY 10065, USA
| | - James K Min
- Dalio Institute of Cardiovascular Imaging, Weill Cornell Medical College and New York Presbyterian Hospital, 1300 York Avenue, New York, NY 10065, USA
| | - Philipp A Kaufmann
- Department of Nuclear Medicine, University Hospital Zurich, Raemistrasse 100, 8091 Zurich, Switzerland
| |
Collapse
|
7
|
El-Far AH, Elewa YHA, Abdelfattah EZA, Alsenosy AWA, Atta MS, Abou-Zeid KM, Al Jaouni SK, Mousa SA, Noreldin AE. RETRACTED: Thymoquinone and Curcumin Defeat Aging-Associated Oxidative Alterations Induced by D-Galactose in Rats' Brain and Heart. Int J Mol Sci 2021; 22:6839. [PMID: 34202112 PMCID: PMC8268720 DOI: 10.3390/ijms22136839] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/09/2021] [Accepted: 06/18/2021] [Indexed: 01/17/2023] Open
Abstract
D-galactose (D-gal) administration causes oxidative disorder and is widely utilized in aging animal models. Therefore, we subcutaneously injected D-gal at 200 mg/kg BW dose to assess the potential preventive effect of thymoquinone (TQ) and curcumin (Cur) against the oxidative alterations induced by D-gal. Other than the control, vehicle, and D-gal groups, the TQ and Cur treated groups were orally supplemented at 20 mg/kg BW of each alone or combined. TQ and Cur effectively suppressed the oxidative alterations induced by D-gal in brain and heart tissues. The TQ and Cur combination significantly decreased the elevated necrosis in the brain and heart by D-gal. It significantly reduced brain caspase 3, calbindin, and calcium-binding adapter molecule 1 (IBA1), heart caspase 3, and BCL2. Expression of mRNA of the brain and heart TP53, p21, Bax, and CASP-3 were significantly downregulated in the TQ and Cur combination group along with upregulation of BCL2 in comparison with the D-gal group. Data suggested that the TQ and Cur combination is a promising approach in aging prevention.
Collapse
Affiliation(s)
- Ali H. El-Far
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| | - Yaser H. A. Elewa
- Department of Histology, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44519, Egypt
- Laboratory of Anatomy, Faculty of Veterinary Medicine, Basic Veterinary Sciences, Hokkaido University, Sapporo 060-0818, Japan
| | | | - Abdel-Wahab A. Alsenosy
- Department of Biochemistry, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| | - Mustafa S. Atta
- Department of Physiology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh 33516, Egypt;
| | - Khalid M. Abou-Zeid
- Animal Care Unit, Medical Research Institute, Alexandria University, Alexandria 21544, Egypt; (E.-Z.A.A.); (K.M.A.-Z.)
| | - Soad K. Al Jaouni
- Department of Hematology/Pediatric Oncology, Yousef Abdulatif Jameel Scientific Chair of Prophetic Medicine Application, Faculty of Medicine, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
| | - Shaker A. Mousa
- Pharmaceutical Research Institute, Albany College of Pharmacy and Health Sciences, Rensselaer, NY 12144, USA;
| | - Ahmed E. Noreldin
- Histology and Cytology Department, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511, Egypt;
| |
Collapse
|
8
|
Woulfe KC, Walker LA. Physiology of the Right Ventricle Across the Lifespan. Front Physiol 2021; 12:642284. [PMID: 33737888 PMCID: PMC7960651 DOI: 10.3389/fphys.2021.642284] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/05/2021] [Indexed: 01/27/2023] Open
Abstract
The most common cause of heart failure in the United States is ischemic left heart disease; accordingly, a vast amount of work has been done to elucidate the molecular mechanisms underlying pathologies of the left ventricle (LV) as a general model of heart failure. Until recently, little attention has been paid to the right ventricle (RV) and it has commonly been thought that the mechanical and biochemical properties of the RV are similar to those of the LV. However, therapies used to treat LV failure often fail to improve ventricular function in RV failure underscoring, the need to better understand the unique physiologic and pathophysiologic properties of the RV. Importantly, hemodynamic stresses (such as pressure overload) often underlie right heart failure further differentiating RV failure as unique from LV failure. There are significant structural, mechanical, and biochemical properties distinctive to the RV that influences its function and it is likely that adaptations of the RV occur uniquely across the lifespan. We have previously reviewed the adult RV compared to the LV but there is little known about differences in the pediatric or aged RV. Accordingly, in this mini-review, we will examine the subtle distinctions between the RV and LV that are maintained physiologically across the lifespan and will highlight significant knowledge gaps in our understanding of pediatric and aging RV. Consideration of how RV function is altered in different disease states in an age-specific manner may enable us to define RV function in health and importantly, in response to pathology.
Collapse
Affiliation(s)
- Kathleen C Woulfe
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Lori A Walker
- Division of Cardiology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
9
|
Izzo C, Vitillo P, Di Pietro P, Visco V, Strianese A, Virtuoso N, Ciccarelli M, Galasso G, Carrizzo A, Vecchione C. The Role of Oxidative Stress in Cardiovascular Aging and Cardiovascular Diseases. Life (Basel) 2021; 11:60. [PMID: 33467601 PMCID: PMC7829951 DOI: 10.3390/life11010060] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
Aging can be seen as process characterized by accumulation of oxidative stress induced damage. Oxidative stress derives from different endogenous and exogenous processes, all of which ultimately lead to progressive loss in tissue and organ structure and functions. The oxidative stress theory of aging expresses itself in age-related diseases. Aging is in fact a primary risk factor for many diseases and in particular for cardiovascular diseases and its derived morbidity and mortality. Here we highlight the role of oxidative stress in age-related cardiovascular aging and diseases. We take into consideration the molecular mechanisms, the structural and functional alterations, and the diseases accompanied to the cardiovascular aging process.
Collapse
Affiliation(s)
- Carmine Izzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paolo Vitillo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Valeria Visco
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Andrea Strianese
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Nicola Virtuoso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Michele Ciccarelli
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Gennaro Galasso
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, Baronissi, 84081 Salerno, Italy; (C.I.); (P.V.); (P.D.P.); (V.V.); (A.S.); (N.V.); (M.C.); (G.G.); (A.C.)
- Department of Angio-Cardio-Neurology, Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Isernia, Italy
| |
Collapse
|
10
|
Chang YM, Shibu MA, Chen CS, Tamilselvi S, Tsai CT, Tsai CC, Kumar KA, Lin HJ, Mahalakshmi B, Kuo WW, Huang CY. Adipose derived mesenchymal stem cells along with Alpinia oxyphylla extract alleviate mitochondria-mediated cardiac apoptosis in aging models and cardiac function in aging rats. JOURNAL OF ETHNOPHARMACOLOGY 2021; 264:113297. [PMID: 32841691 DOI: 10.1016/j.jep.2020.113297] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 08/04/2020] [Accepted: 08/18/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Fructus (Alpinia oxyphylla MIQ) known as Yi Zhi Ren in Chinese medicine has been used as a food and herbal medicinal substance in China for centuries; in the year 2015 Chinese Pharmacopoeia Commission reported water extracts of Alpinia oxyphyllae Fructus (AoF) as a popular medication for aging-related diseases in the form of tonic, aphrodisiac, and health-care food in south China. AIM OF THE STUDY Adipose mesenchymal stem cells are physiologically and therapeutically associated with healthy vascular function and cardiac health. However aging conditions hinder stem cell function and increases the vulnerability to cardiovascular diseases. In this study, the effect of the anti-aging herbal medicine AoF to enhance the cardiac restorative function of adipose-derived mesenchymal stem cells (ADMSCs) in aging condition was investigated. MATERIALS AND METHODS Low dose (0.1 μM) Doxorubicin and D-galactose (150 mg/kg/day for 8 weeks) were used to respectively induce aging in vitro and in vivo. For In vivo studies, 20 week old WKY rats were divided into Control, Aging induced (AI), AI + AoF, AI + ADMSC, AI + AoF Oral + ADMSC, and AI + AoF treated ADMSC groups. AoF (100 mg/kg/day) was administered orally and ADMSCs (1 × 106 cells) were injected (IV). RESULTS AoF preconditioned ADMSC showed reduction in low dose Dox induced mitochondrial apoptosis and improved DNA replication in H9c2 cardiomyoblasts. In vivo experiments confirmed that both a combined treatment with AoF-ADMSCs and with AoF preconditioned ADMSCs reduced aging associated cardiac damages which was correlated with reduction in apoptosis and expression of senescence markers (P21 and β-gal). Survival and longevity markers were upregulated up on combined administration of AoF and ADMSCs. The cardiac performance of the aging-induced rats was improved significantly in the treatment groups. AoF along with ADMSCs might activate paracrine factors to restore the performance of an aging heart. CONCLUSION Hence, we propose that ADMSCs combined with AoF have promising therapeutic properties in the treatment of healthy aging heart.
Collapse
Affiliation(s)
- Yung-Ming Chang
- The School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, 840, Taiwan; Chinese Medicine Department, E-DA Hospital, Kaohsiung, 824, Taiwan; 1PT Biotechnology Co., Ltd., Taichung, 433, Taiwan
| | - Marthandam Asokan Shibu
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
| | - Chih-Sheng Chen
- Graduate Institute of Chinese Medicine, China Medical University, Taiwan; Division of Chinese Medicine Asia University Hospital Taichung, Taiwan
| | - Shanmugam Tamilselvi
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
| | | | - Chin-Chuan Tsai
- The School of Chinese Medicine for Post-Baccalaureate, I-Shou University, Kaohsiung, 840, Taiwan; Chinese Medicine Department, E-DA Hospital, Kaohsiung, 824, Taiwan
| | - Kannan Ashok Kumar
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan
| | - Hung-Jen Lin
- Department of Chinese Medicine, China Medical University Hospital, Taichung, 40447, Taiwan; School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - B Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang, 550000, Viet Nam
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Cardiovascular and Mitochondrial Related Disease Research Center, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, 970, Taiwan; Graduate Institute of Biomedical Sciences, China Medical University, Taichung, 404, Taiwan; Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, 970, Taiwan; Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, 404, Taiwan; Department of Biotechnology, Asia University, Taichung, 413, Taiwan.
| |
Collapse
|
11
|
Lin WT, Nithiyanantham S, Hsieh DJY, Chen RJ, Day CH, Liao JY, Kuo CH, Mahalakshmi B, Kuo WW, Huang CY. Bioactive peptides attenuate cardiac apoptosis in spontaneously hypertensive rat hearts through activation of autophagy and mitochondrial biogenesis pathway. ENVIRONMENTAL TOXICOLOGY 2020; 35:804-810. [PMID: 32141235 DOI: 10.1002/tox.22916] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 02/20/2020] [Accepted: 02/23/2020] [Indexed: 06/10/2023]
Abstract
Alcalase potato protein hydrolysate (APPH) might have a very important role in therapeutic effects. This study aims to examine the beneficial effects of bioactive peptides (DIKTNKPVIF [DI] and IF) from APPH supplement in the regulation of cardiac apoptosis, autophagy, and mitochondrial biogenesis pathway in spontaneously hypertensive rats (SHR). We have investigated ejection fraction, fractional shortening, Tunel assay, apoptosis, autophagy, and mitochondrial biogenesis pathway marker expression to show the efficacy of bioactive peptides in an SHR model. Bioactive peptides significantly upregulate ejection fraction and fractional shortening in SHR rats. SHR rats exhibited higher protein expression of apoptotic markers such as BAD, cytochrome c, and caspase 3. Finally, the bioactive peptides upregulate survival proteins (p-AKT/p-PI3K), autophagy (Beclin1/LC3B), and mitochondrial biogenesis (p-AMPKα/SIRT1/PGC1α/p-Foxo3a/Nrf2/CREB) marker expressions compared with the SHR groups. In summary, the bioactive peptides protect the heart tissues through the activation of autophagy and mitochondrial biogenesis pathway and thereby attenuate cardiac apoptosis in a spontaneously hypertensive rat model.
Collapse
Affiliation(s)
- Wan Teng Lin
- Department of Hospitality Management, College of Agriculture, Tunghai University, Taichung, Taiwan
| | | | - Dennis Jine-Yuan Hsieh
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Ray-Jade Chen
- Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | | | - Jia Ying Liao
- Department of Hospitality Management, College of Agriculture, Tunghai University, Taichung, Taiwan
| | - Chia-Hua Kuo
- Department of Sports Sciences, University of Taipei, Taipei, Taiwan
| | - B Mahalakshmi
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam
| | - Wei-Wen Kuo
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Cardiovascular and Mitochondrial Related Diseases Research Center, Buddhist Tzu Chi Medical Foundation, Hualien Tzu Chi Hospital, Hualien, Taiwan
- Center of General Education, Buddhist Tzu Chi Medical Foundation, Tzu Chi University of Science and Technology, Hualien, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
12
|
The impact of cardiovascular risk factors on global longitudinal strain over a decade in the general population: the copenhagen city heart study. Int J Cardiovasc Imaging 2020; 36:1907-1916. [DOI: 10.1007/s10554-020-01906-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 06/01/2020] [Indexed: 11/26/2022]
|
13
|
Protein and Mitochondria Quality Control Mechanisms and Cardiac Aging. Cells 2020; 9:cells9040933. [PMID: 32290135 PMCID: PMC7226975 DOI: 10.3390/cells9040933] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/07/2020] [Accepted: 04/07/2020] [Indexed: 12/31/2022] Open
Abstract
Cardiovascular disease (CVD) is the number one cause of death in the United States. Advancing age is a primary risk factor for developing CVD. Estimates indicate that 20% of the US population will be ≥65 years old by 2030. Direct expenditures for treating CVD in the older population combined with indirect costs, secondary to lost wages, are predicted to reach $1.1 trillion by 2035. Therefore, there is an eminent need to discover novel therapeutic targets and identify new interventions to delay, lessen the severity, or prevent cardiovascular complications associated with advanced age. Protein and organelle quality control pathways including autophagy/lysosomal and the ubiquitin-proteasome systems, are emerging contributors of age-associated myocardial dysfunction. In general, two findings have sparked this interest. First, strong evidence indicates that cardiac protein degradation pathways are altered in the heart with aging. Second, it is well accepted that damaged and misfolded protein aggregates and dysfunctional mitochondria accumulate in the heart with age. In this review, we will: (i) define the different protein and mitochondria quality control mechanisms in the heart; (ii) provide evidence that each quality control pathway becomes dysfunctional during cardiac aging; and (iii) discuss current advances in targeting these pathways to maintain cardiac function with age.
Collapse
|
14
|
Loredo-Mendoza ML, Ramirez-Sanchez I, Bustamante-Pozo MM, Ayala M, Navarrete V, Garate-Carrillo A, Ito BR, Ceballos G, Omens J, Villarreal F. The role of inflammation in driving left ventricular remodeling in a pre-HFpEF model. Exp Biol Med (Maywood) 2020; 245:748-757. [PMID: 32183553 DOI: 10.1177/1535370220912699] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
IMPACT STATEMENT The incidence of HFpEF continues to increase and ∼2/3 of the patient population are post-menopausal women. Unfortunately, most studies focus on the use of male animal models of remodeling. In this study, however, using female rats to set a model of pre-HFpEF, we provide insights to possible mechanisms that contribute to HFpEF development in humans that will lead us to a better understanding of the underlying pathophysiology of HFpEF.
Collapse
Affiliation(s)
- Maria L Loredo-Mendoza
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Histopathology, School of Medicine, Universidad Panamericana, Ciudad de Mexico 03920, Mexico
| | - Israel Ramirez-Sanchez
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Moises Muratt Bustamante-Pozo
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Marcos Ayala
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
| | - Viridiana Navarrete
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
| | - Alejandra Garate-Carrillo
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Bruce R Ito
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Guillermo Ceballos
- Seccion de Estudios de Posgrado e Investigacion, Escuela Superior de Medicina, Instituto Politecnico Nacional, Mexico 07738, Mexico
| | - Jeffrey Omens
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
| | - Francisco Villarreal
- Department of Medicine, School of Medicine, University of California, San Diego, CA 92093-0021, USA
- VA San Diego Healthcare, San Diego, CA 92161, USA
| |
Collapse
|
15
|
Stokar J, Leibowitz D, Durst R, Shaham D, Zwas DR. Echocardiography overestimates LV mass in the elderly as compared to cardiac CT. PLoS One 2019; 14:e0224104. [PMID: 31648248 PMCID: PMC6812823 DOI: 10.1371/journal.pone.0224104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 10/05/2019] [Indexed: 01/19/2023] Open
Abstract
PURPOSE Echocardiographic studies have shown an increase in LV mass with advanced age. However, autopsy and MRI studies demonstrate that with aging, LV mass is unchanged or slightly decreased, with a decrease in LV volume and an increase in wall thickness consistent with concentric remodeling. LV structural remodeling with aging may lead to an overestimation of LV mass in older adults when using standard echocardiography measurements and calculations. This study compared CT and echocardiographic LV mass calculation in younger and older patients and parameters associated with age-related LV remodeling. METHODS Same subject modality comparison of echocardiographic and cardiac CT LV measurement with derivation of LV mass was performed retrospectively. Echocardiographic measurements were performed by a single observer in accordance with European Association of Cardiovascular Imaging (EACI)/American Society of Echocardiography (ASE) guidelines. CT measurements were performed in end-diastole on multiplanar reformatted image planes corresponding to those typically used in echocardiography. Calculated CT measurements were based on automatic segmentation of heart chambers via edge-tracing algorithms. RESULTS 129 patients were identified. In patients age 65 and older, LV mass was significantly higher when calculated using echocardiographic measurements compared to CT. Patients 65 years of age and older were found to have increased average wall thickness measurements with echocardiography but not with CT. The discrepancy between calculated echo and CT LV mass was reduced when using the mid-septal instead of proximal wall width for the EACI convention. CONCLUSION In the elderly, increased echo-derived LV mass may reflect remodeling rather than a true increase in LV mass.
Collapse
Affiliation(s)
- Joshua Stokar
- Division of Endocrinology, Hadassah University Medical Center, Jerusalem, Israel
| | - David Leibowitz
- Division of Cardiology, Hadassah University Medical Center, Jerusalem, Israel
| | - Ronen Durst
- Division of Cardiology, Hadassah University Medical Center, Jerusalem, Israel
| | - Dorith Shaham
- Division of Radiology, Hadassah University Medical Center, Jerusalem, Israel
| | - Donna R. Zwas
- Division of Cardiology, Hadassah University Medical Center, Jerusalem, Israel
| |
Collapse
|
16
|
Kovina MV, Karnaukhov AV, Krasheninnikov ME, Kovin AL, Gazheev ST, Sergievich LA, Karnaukhova EV, Bogdanenko EV, Balyasin MV, Khodarovich YM, Dyuzheva TG, Lyundup AV. Extension of Maximal Lifespan and High Bone Marrow Chimerism After Nonmyeloablative Syngeneic Transplantation of Bone Marrow From Young to Old Mice. Front Genet 2019; 10:310. [PMID: 31031800 PMCID: PMC6473025 DOI: 10.3389/fgene.2019.00310] [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: 07/10/2018] [Accepted: 03/21/2019] [Indexed: 02/01/2023] Open
Abstract
The goal of this work was to determine the effect of nonablative syngeneic transplantation of young bone marrow (BM) to laboratory animals (mice) of advanced age upon maximum duration of their lifespan. To do this, transplantation of 100 million nucleated cells from BM of young syngeneic donors to an old nonablated animal was performed at the time when half of the population had already died. As a result, the maximum lifespan (MLS) increased by 28 ± 5%, and the survival time from the beginning of the experiment increased 2.8 ± 0.3-fold. The chimerism of the BM 6 months after the transplantation was 28%.
Collapse
Affiliation(s)
- Marina V Kovina
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Alexey V Karnaukhov
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | | | - Artem L Kovin
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Sarul T Gazheev
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Larisa A Sergievich
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Elena V Karnaukhova
- Institute of Cell Biophysics, Russian Academy of Sciences, Pushchino, Russia
| | - Elena V Bogdanenko
- Institute of General Pathology and Pathophysiology, Russian Academy of Sciences, Moscow, Russia
| | - Maxim V Balyasin
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| | - Yury M Khodarovich
- Department of Molecular Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | | | - Alexey V Lyundup
- Institute for Regenerative Medicine, Sechenov University, Moscow, Russia
| |
Collapse
|
17
|
Alpinate Oxyphyllae
extracts enhance the longevity and homing of mesenchymal stem cells and augment their protection against senescence in H9c2 cells. J Cell Physiol 2018; 234:12042-12050. [DOI: 10.1002/jcp.27867] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Accepted: 11/13/2018] [Indexed: 12/19/2022]
|
18
|
Garvin AM, Jackson MA, Korzick DH. Inhibition of programmed necrosis limits infarct size through altered mitochondrial and immune responses in the aged female rat heart. Am J Physiol Heart Circ Physiol 2018; 315:H1434-H1442. [PMID: 29957016 DOI: 10.1152/ajpheart.00595.2017] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Both advancing age and estrogen loss exacerbate acute myocardial infarction in the female heart. However, the mechanistic underpinnings of age-related differences in cell death after ischemia-reperfusion (I/R) injury in female subjects and reductions in cardioprotective reserve capacity remain largely unexplored. The aim of the present study was to determine the efficacy of programmed necrosis inhibition on infarct size reduction and preservation of left ventricular (LV) function after I/R injury with female aging. Fischer 344 rats were ovariectomized (OVX) at 15 mo and studied at 24 mo (MO OVX) versus adult rats with intact ovaries (6 mo). After in vivo coronary artery ligation (55-min ischemia and 2- or 6-h reperfusion), necrostatin-1 (Nec-1; 3.5 or 5.7 mg/kg) delivered upon reperfusion significantly reduced infarct size by 37% and improved LV function in the MO OVX group ( P < 0.01). Although age-associated elevations in cyclophilin D and mitochondrial acetylation ( P < 0.001) were unaffected by Nec-1, profound reductions in IL-1, IL-6, and TNF-α ( P < 0.05) as well as cardiac immune cell infiltration were observed in MO OVX but not adult rats. We conclude that chronic inflammation and postmenopausal estrogen deficiency conspire to exacerbate acute infarction through a mechanism involving exaggerated mitochondria-mediated programmed necrosis through receptor-interacting protein 1 signaling. Modulatory effects of programmed necrosis inhibition on proinflammatory cytokine production after I/R reveal a potentially important mechanistic target to restore and preserve cardiac function in the OVX aged female heart. NEW & NOTEWORTHY Myocardial infarct size reduction by inhibition of programmed necrosis in aged female subjects suggests a dominant cell death pathway. Alterations in mitochondrial protein levels and acetylation underscore a mitochondria-dependent mechanism, whereas the profound cytokine reduction in aged subjects alone points to a divergent role for immune modulation of programmed necrosis and viable therapeutic target.
Collapse
Affiliation(s)
- Alexandra M Garvin
- Intercollege Graduate Degree Program in Physiology, The Pennsylvania State University , University Park, Pennsylvania
| | - Morgan A Jackson
- Intercollege Graduate Degree Program in Physiology, The Pennsylvania State University , University Park, Pennsylvania
| | - Donna H Korzick
- Intercollege Graduate Degree Program in Physiology, The Pennsylvania State University , University Park, Pennsylvania.,Department of Kinesiology, The Pennsylvania State University , University Park, Pennsylvania
| |
Collapse
|
19
|
Spurthi KM, Sarikhani M, Mishra S, Desingu PA, Yadav S, Rao S, Maity S, Tamta AK, Kumar S, Majumdar S, Jain A, Raghuraman A, Khan D, Singh I, Samuel RJ, Ramachandra SG, Nandi D, Sundaresan NR. Toll-like receptor 2 deficiency hyperactivates the FoxO1 transcription factor and induces aging-associated cardiac dysfunction in mice. J Biol Chem 2018; 293:13073-13089. [PMID: 29929978 DOI: 10.1074/jbc.ra118.001880] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 06/09/2018] [Indexed: 12/17/2022] Open
Abstract
Toll-like receptors (TLRs) are a family of pattern-recognition receptors involved in innate immunity. Previous studies have shown that TLR2 inhibition protects the heart from acute stress, including myocardial infarction and doxorubicin-induced cardiotoxicity in animal models. However, the role of TLR2 in the development of aging-associated heart failure is not known. In this work, we studied aging-associated changes in structure and function of TLR2-deficient mice hearts. Whereas young TLR2-KO mice did not develop marked cardiac dysfunction, 8- and 12-month-old TLR2-KO mice exhibited spontaneous adverse cardiac remodeling and cardiac dysfunction in an age-dependent manner. The hearts of the 8-month-old TLR2-KO mice had increased fibrosis, cell death, and reactivation of fetal genes. Moreover, TLR2-KO hearts displayed reduced infiltration by macrophages, increased numbers of myofibroblasts and atrophic cardiomyocytes, and higher levels of the atrophy-related ubiquitin ligases MuRF-1 and atrogin-1. Mechanistically, TLR2 deficiency impaired the PI3K/Akt signaling pathway, leading to hyperactivation of the transcription factor Forkhead box protein O1 (FoxO1) and, in turn, to elevated expression of FoxO target genes involved in the regulation of muscle wasting and cell death. AS1842856-mediated chemical inhibition of FoxO1 reduced the expression of the atrophy-related ubiquitin ligases and significantly reversed the adverse cardiac remodeling while improving the contractile functions in the TLR2-KO mice. Interestingly, TLR2 levels decreased in hearts of older mice, and the activation of TLR1/2 signaling improved cardiac functions in these mice. These findings suggest that TLR2 signaling is essential for protecting the heart against aging-associated adverse remodeling and contractile dysfunction in mice.
Collapse
Affiliation(s)
- Kondapalli Mrudula Spurthi
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Mohsen Sarikhani
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sneha Mishra
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Perumal Arumugam Desingu
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shikha Yadav
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Swathi Rao
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Sangeeta Maity
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ankit Kumar Tamta
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shweta Kumar
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Shamik Majumdar
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Aditi Jain
- the Centre for Biosystems Science and Engineering, Indian Institute of Science, Bengaluru, Karnataka 560012, India, and
| | - Aishwarya Raghuraman
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Danish Khan
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Ishwar Singh
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Rosa J Samuel
- the Central Animal Facility, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Subbaraya G Ramachandra
- the Central Animal Facility, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Dipankar Nandi
- the Department of Biochemistry, Indian Institute of Science, Bengaluru, Karnataka 560012, India
| | - Nagalingam R Sundaresan
- From the Department of Microbiology and Cell Biology, Indian Institute of Science, Bengaluru, Karnataka 560012, India,
| |
Collapse
|
20
|
Aging-Induced Biological Changes and Cardiovascular Diseases. BIOMED RESEARCH INTERNATIONAL 2018; 2018:7156435. [PMID: 29984246 PMCID: PMC6015721 DOI: 10.1155/2018/7156435] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 04/23/2018] [Accepted: 05/03/2018] [Indexed: 12/12/2022]
Abstract
Aging is characterized by functional decline in homeostatic regulation and vital cellular events. This process can be linked with the development of cardiovascular diseases (CVDs). In this review, we discussed aging-induced biological alterations that are associated with CVDs through the following aspects: (i) structural, biochemical, and functional modifications; (ii) autonomic nervous system (ANS) dysregulation; (iii) epigenetic alterations; and (iv) atherosclerosis and stroke development. Aging-mediated structural and biochemical modifications coupled with gradual loss of ANS regulation, vascular stiffening, and deposition of collagen and calcium often disrupt cardiovascular system homeostasis. The structural and biochemical adjustments have been consistently implicated in the progressive increase in mechanical burden and functional breakdown of the heart and vessels. In addition, cardiomyocyte loss in this process often reduces adaptive capacity and cardiovascular function. The accumulation of epigenetic changes also plays important roles in the development of CVDs. In summary, the understanding of the aging-mediated changes remains promising towards effective diagnosis, discovery of new drug targets, and development of new therapies for the treatment of CVDs.
Collapse
|
21
|
Nrf2 Deficiency Unmasks the Significance of Nitric Oxide Synthase Activity for Cardioprotection. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:8309698. [PMID: 29854098 PMCID: PMC5952436 DOI: 10.1155/2018/8309698] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/28/2017] [Revised: 01/17/2018] [Accepted: 02/27/2018] [Indexed: 12/14/2022]
Abstract
The transcription factor nuclear factor (erythroid-derived 2)-like 2 (Nrf2) is a key master switch that controls the expression of antioxidant and cytoprotective enzymes, including enzymes catalyzing glutathione de novo synthesis. In this study, we aimed to analyze whether Nrf2 deficiency influences antioxidative capacity, redox state, NO metabolites, and outcome of myocardial ischemia reperfusion (I/R) injury. In Nrf2 knockout (Nrf2 KO) mice, we found elevated eNOS expression and preserved NO metabolite concentrations in the aorta and heart as compared to wild types (WT). Unexpectedly, Nrf2 KO mice have a smaller infarct size following myocardial ischemia/reperfusion injury than WT mice and show fully preserved left ventricular systolic function. Inhibition of NO synthesis at onset of ischemia and during early reperfusion increased myocardial damage and systolic dysfunction in Nrf2 KO mice, but not in WT mice. Consistent with this, infarct size and diastolic function were unaffected in eNOS knockout (eNOS KO) mice after ischemia/reperfusion. Taken together, these data suggest that eNOS upregulation under conditions of decreased antioxidant capacity might play an important role in cardioprotection against I/R. Due to the redundancy in cytoprotective mechanisms, this fundamental antioxidant property of eNOS is not evident upon acute NOS inhibition in WT mice or in eNOS KO mice until Nrf2-related signaling is abrogated.
Collapse
|
22
|
Takemoto Y, Horiba M, Harada M, Sakamoto K, Takeshita K, Murohara T, Kadomatsu K, Kamiya K. Midkine Promotes Atherosclerotic Plaque Formation Through Its Pro-Inflammatory, Angiogenic and Anti-Apoptotic Functions in Apolipoprotein E-Knockout Mice. Circ J 2017; 82:19-27. [PMID: 28781288 DOI: 10.1253/circj.cj-17-0043] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND A recent study suggested that midkine (MK), a heparin-binding growth factor, is associated with atherosclerosis progression in patients with artery disease. It has previously been reported that MK plays a critical role in neointima formation in a restenosis model, whereas the role of MK in the development of atherosclerosis has not been investigated. The present study assessed the effect of MK administration on the process of atherosclerotic plaque formation in apolipoprotein E-knockout (ApoE-/-) mice. METHODS AND RESULTS Using an osmotic pump, human recombinant MK protein was intraperitoneally administered for 12 weeks in C57BL/6 ApoE-/-(ApoE-/--MK) and ApoE+/+mice fed a high-fat diet. Saline was administered to the control groups of ApoE-/-(ApoE-/--saline) and ApoE+/+mice. The atherosclerotic lesion areas in longitudinal aortic sections were significantly larger in ApoE-/--MK mice than in ApoE-/--saline mice. The aortic mRNA levels of pro-inflammatory and angiogenic factors, and the percentage of macrophages in aortic root lesions, were significantly higher in ApoE-/--MK mice than in ApoE-/--saline mice, whereas the percentage of apoptotic cells was significantly lower in ApoE-/--MK mice than in ApoE-/--saline mice. CONCLUSIONS The systemic administration of MK in ApoE-/-mice promoted atherosclerotic plaque formation through pro-inflammatory, angiogenic, and anti-apoptotic effects. MK may serve as a potential therapeutic target for the prevention of atherosclerosis under atherogenic conditions.
Collapse
Affiliation(s)
- Yoshio Takemoto
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University
- Department of Cardiovascular Medicine, Gifu Prefectural Tajimi Hospital
| | - Mitsuru Horiba
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University
- Department of Biochemistry, Nagoya University Graduate School of Medicine
- Suizawa Hospital
| | - Masahide Harada
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University
- Department of Cardiology, Fujita Health University
| | - Kazuma Sakamoto
- Department of Biochemistry, Nagoya University Graduate School of Medicine
| | - Kyosuke Takeshita
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Kenji Kadomatsu
- Department of Biochemistry, Nagoya University Graduate School of Medicine
| | - Kaichiro Kamiya
- Department of Cardiovascular Research, Research Institute of Environmental Medicine, Nagoya University
| |
Collapse
|
23
|
The Role of Nrf2 in Cardiovascular Function and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9237263. [PMID: 29104732 PMCID: PMC5618775 DOI: 10.1155/2017/9237263] [Citation(s) in RCA: 193] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 07/27/2017] [Indexed: 02/07/2023]
Abstract
Free radicals, reactive oxygen/nitrogen species (ROS/RNS), hydrogen sulphide, and hydrogen peroxide play an important role in both intracellular and intercellular signaling; however, their production and quenching need to be closely regulated to prevent cellular damage. An imbalance, due to exogenous sources of free radicals and chronic upregulation of endogenous production, contributes to many pathological conditions including cardiovascular disease and also more general processes involved in aging. Nuclear factor erythroid 2-like 2 (NFE2L2; commonly known as Nrf2) is a transcription factor that plays a major role in the dynamic regulation of a network of antioxidant and cytoprotective genes, through binding to and activating expression of promoters containing the antioxidant response element (ARE). Nrf2 activity is regulated by many mechanisms, suggesting that tight control is necessary for normal cell function and both hypoactivation and hyperactivation of Nrf2 are indicated in playing a role in different aspects of cardiovascular disease. Targeted activation of Nrf2 or downstream genes may prove to be a useful avenue in developing therapeutics to reduce the impact of cardiovascular disease. We will review the current status of Nrf2 and related signaling in cardiovascular disease and its relevance to current and potential treatment strategies.
Collapse
|
24
|
Alfaras I, Di Germanio C, Bernier M, Csiszar A, Ungvari Z, Lakatta EG, de Cabo R. Pharmacological Strategies to Retard Cardiovascular Aging. Circ Res 2017; 118:1626-42. [PMID: 27174954 DOI: 10.1161/circresaha.116.307475] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 04/08/2016] [Indexed: 01/10/2023]
Abstract
Aging is the major risk factor for cardiovascular diseases, which are the leading cause of death in the United States. Traditionally, the effort to prevent cardiovascular disease has been focused on addressing the conventional risk factors, including hypertension, hyperglycemia, hypercholesterolemia, and high circulating levels of triglycerides. However, recent preclinical studies have identified new approaches to combat cardiovascular disease. Calorie restriction has been reproducibly shown to prolong lifespan in various experimental model animals. This has led to the development of calorie restriction mimetics and other pharmacological interventions capable to delay age-related diseases. In this review, we will address the mechanistic effects of aging per se on the cardiovascular system and focus on the prolongevity benefits of various therapeutic strategies that support cardiovascular health.
Collapse
Affiliation(s)
- Irene Alfaras
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Clara Di Germanio
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Michel Bernier
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Anna Csiszar
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Zoltan Ungvari
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Edward G Lakatta
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.)
| | - Rafael de Cabo
- From the Experimental Gerontology Section, Translational Gerontology Branch (I.A., C.D.G., M.B., R.d.C.) and Laboratory of Cardiovascular Science (E.G.L.), National Institute on Aging, National Institutes of Health, Baltimore, MD; Faculty of Veterinary Medicine, University of Teramo, Teramo, Italy (C.D.G.); and Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK (A.C., Z.U.).
| |
Collapse
|
25
|
Meschiari CA, Ero OK, Pan H, Finkel T, Lindsey ML. The impact of aging on cardiac extracellular matrix. GeroScience 2017; 39:7-18. [PMID: 28299638 PMCID: PMC5352584 DOI: 10.1007/s11357-017-9959-9] [Citation(s) in RCA: 153] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/05/2017] [Indexed: 12/24/2022] Open
Abstract
Age-related changes in cardiac homeostasis can be observed at the cellular, extracellular, and tissue levels. Progressive cardiomyocyte hypertrophy, inflammation, and the gradual development of cardiac fibrosis are hallmarks of cardiac aging. In the absence of a secondary insult such as hypertension, these changes are subtle and result in slight to moderate impaired myocardial function, particularly diastolic function. While collagen deposition and cross-linking increase during aging, extracellular matrix (ECM) degradation capacity also increases due to increased expression of matrix metalloproteinases (MMPs). Of the MMPs elevated with cardiac aging, MMP-9 has been extensively evaluated and its roles are reviewed here. In addition to proteolytic activity on ECM components, MMPs oversee cell signaling during the aging process by modulating cytokine, chemokine, growth factor, hormone, and angiogenic factor expression and activity. In association with elevated MMP-9, macrophage numbers increase in an age-dependent manner to regulate the ECM and angiogenic responses. Understanding the complexity of the molecular interactions between MMPs and the ECM in the context of aging may provide novel diagnostic indicators for the early detection of age-related fibrosis and cardiac dysfunction.
Collapse
Affiliation(s)
- Cesar A Meschiari
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Room G351-04, Jackson, MS, USA
| | - Osasere Kelvin Ero
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Room G351-04, Jackson, MS, USA
| | - Haihui Pan
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Toren Finkel
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, Bethesda, MD, USA
| | - Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Room G351-04, Jackson, MS, USA.
- G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, 39216-4505, USA.
| |
Collapse
|
26
|
Narasimhan M, Rajasekaran NS. Exercise, Nrf2 and Antioxidant Signaling in Cardiac Aging. Front Physiol 2016; 7:241. [PMID: 27378947 PMCID: PMC4911351 DOI: 10.3389/fphys.2016.00241] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 06/03/2016] [Indexed: 12/16/2022] Open
Abstract
Aging is represented by a progressive decline in cellular functions. The age-related deformities in cardiac behaviors are the loss of cardiac myocytes through apoptosis or programmed cell death. Oxidative stress (OS) and its deleterious consequence contribute to age-related mechanical remodeling, reduced regenerative capacity, and apoptosis in cardiac tissue. The pathogenesis of OS in the elderly can predispose the heart to other cardiac complications such as atherosclerosis, hypertension, ischemic heart disease, cardiac myopathy, and so on. At the molecular level, oxidant-induced activation of Nrf2 (Nuclear erythroid-2-p45-related factor-2), a transcription factor, regulates several genes containing AREs (Antioxidant Response Element) and bring the respective translates to counteract the reactive radicals and establish homeostasis. Myriad of Nrf2 gene knockout studies in various organs such as lung, liver, kidney, brain, etc. have shown that dysregulation of Nrf2 severely affects the oxidant/ROS sensitivity and predispose the system to several pathological changes with aberrant cellular lesions. On the other hand, its gain of function chemical interventions exhibited oxidant stress resistance and cytoprotection. However, thus far, only a few investigations have shown the potential role of Nrf2 and its non-pharmacological induction in cardiac aging. Therefore, here we review the involvement of Nrf2 signaling along with its responses and ramifications on the cascade of OS under acute exercise stress (AES), moderate exercise training (MET), and endurance exercise stress (EES) conditions in the aging heart.
Collapse
Affiliation(s)
- Madhusudhanan Narasimhan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center Lubbock, TX, USA
| | - Namakkal S Rajasekaran
- Cardiac Aging and Redox Signaling Laboratory, Center for Free Radical Biology, Division of Molecular and Cellular Pathology, Department of Pathology, University of Alabama at BirminghamBirmingham, AL, USA; Division of Cardiovascular Medicine, Department of Medicine, University of Utah School of MedicineSalt Lake City, UT, USA; Department of Exercise Physiology, College of Health, University of Utah School of MedicineSalt Lake City, UT, USA
| |
Collapse
|
27
|
Shirakabe A, Ikeda Y, Sciarretta S, Zablocki DK, Sadoshima J. Aging and Autophagy in the Heart. Circ Res 2016; 118:1563-76. [PMID: 27174950 PMCID: PMC4869999 DOI: 10.1161/circresaha.116.307474] [Citation(s) in RCA: 330] [Impact Index Per Article: 36.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 02/15/2016] [Indexed: 12/15/2022]
Abstract
The aging population is increasing in developed countries. Because the incidence of cardiac disease increases dramatically with age, it is important to understand the molecular mechanisms through which the heart becomes either more or less susceptible to stress. Cardiac aging is characterized by the presence of hypertrophy, fibrosis, and accumulation of misfolded proteins and dysfunctional mitochondria. Macroautophagy (hereafter referred to as autophagy) is a lysosome-dependent bulk degradation mechanism that is essential for intracellular protein and organelle quality control. Autophagy and autophagic flux are generally decreased in aging hearts, and murine autophagy loss-of-function models develop exacerbated cardiac dysfunction that is accompanied by the accumulation of misfolded proteins and dysfunctional organelles. On the contrary, stimulation of autophagy generally improves cardiac function in mouse models of protein aggregation by removing accumulated misfolded proteins, dysfunctional mitochondria, and damaged DNA, thereby improving the overall cellular environment and alleviating aging-associated pathology in the heart. Increasing lines of evidence suggest that autophagy is required for many mechanisms that mediate lifespan extension, such as caloric restriction, in various organisms. These results raise the exciting possibility that autophagy may play an important role in combating the adverse effects of aging in the heart. In this review, we discuss the role of autophagy in the heart during aging, how autophagy alleviates age-dependent changes in the heart, and how the level of autophagy in the aging heart can be restored.
Collapse
Affiliation(s)
- Akihiro Shirakabe
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Yoshiyuki Ikeda
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Sebastiano Sciarretta
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Daniela K Zablocki
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.)
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Rutgers-New Jersey Medical School, Newark (A.S., Y.I., S.S., D.K.Z., J.S.); Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan (Y.I.); Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Rome, Italy (S.S.); and Department of AngioCardioNeurology, IRCCS Neuromed, Pozzilli, Italy (S.S.).
| |
Collapse
|
28
|
Horn MA, Trafford AW. Aging and the cardiac collagen matrix: Novel mediators of fibrotic remodelling. J Mol Cell Cardiol 2016; 93:175-85. [PMID: 26578393 PMCID: PMC4945757 DOI: 10.1016/j.yjmcc.2015.11.005] [Citation(s) in RCA: 173] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Revised: 11/02/2015] [Accepted: 11/04/2015] [Indexed: 01/05/2023]
Abstract
Cardiovascular disease is a leading cause of death worldwide and there is a pressing need for new therapeutic strategies to treat such conditions. The risk of developing cardiovascular disease increases dramatically with age, yet the majority of experimental research is executed using young animals. The cardiac extracellular matrix (ECM), consisting predominantly of fibrillar collagen, preserves myocardial integrity, provides a means of force transmission and supports myocyte geometry. Disruptions to the finely balanced control of collagen synthesis, post-synthetic deposition, post-translational modification and degradation may have detrimental effects on myocardial functionality. It is now well established that the aged heart is characterized by fibrotic remodelling, but the mechanisms responsible for this are incompletely understood. Furthermore, studies using aged animal models suggest that interstitial remodelling with disease may be age-dependent. Thus with the identification of new therapeutic strategies targeting fibrotic remodelling, it may be necessary to consider age-dependent mechanisms. In this review, we discuss remodelling of the cardiac collagen matrix as a function of age, whilst highlighting potential novel mediators of age-dependent fibrotic pathways.
Collapse
Affiliation(s)
- Margaux A Horn
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, 3.06 Core Technology Facility, 46 Grafton Street, Manchester M13 9NT, United Kingdom.
| | - Andrew W Trafford
- Institute of Cardiovascular Sciences, Manchester Academic Health Sciences Centre, 3.06 Core Technology Facility, 46 Grafton Street, Manchester M13 9NT, United Kingdom
| |
Collapse
|
29
|
Tower J. Programmed cell death in aging. Ageing Res Rev 2015; 23:90-100. [PMID: 25862945 DOI: 10.1016/j.arr.2015.04.002] [Citation(s) in RCA: 280] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 03/15/2015] [Accepted: 04/01/2015] [Indexed: 02/08/2023]
Abstract
Programmed cell death (PCD) pathways, including apoptosis and regulated necrosis, are required for normal cell turnover and tissue homeostasis. Mis-regulation of PCD is increasingly implicated in aging and aging-related disease. During aging the cell turnover rate declines for several highly-mitotic tissues. Aging-associated disruptions in systemic and inter-cell signaling combined with cell-autonomous damage and mitochondrial malfunction result in increased PCD in some cell types, and decreased PCD in other cell types. Increased PCD during aging is implicated in immune system decline, skeletal muscle wasting (sarcopenia), loss of cells in the heart, and neurodegenerative disease. In contrast, cancer cells and senescent cells are resistant to PCD, enabling them to increase in abundance during aging. PCD pathways limit life span in fungi, but whether PCD pathways normally limit adult metazoan life span is not yet clear. PCD is regulated by a balance of negative and positive factors, including the mitochondria, which are particularly subject to aging-associated malfunction.
Collapse
|
30
|
Quarles EK, Dai DF, Tocchi A, Basisty N, Gitari L, Rabinovitch PS. Quality control systems in cardiac aging. Ageing Res Rev 2015; 23:101-15. [PMID: 25702865 PMCID: PMC4686341 DOI: 10.1016/j.arr.2015.02.003] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Revised: 02/02/2015] [Accepted: 02/12/2015] [Indexed: 12/31/2022]
Abstract
Cardiac aging is an intrinsic process that results in impaired cardiac function, along with cellular and molecular changes. These degenerative changes are intimately associated with quality control mechanisms. This review provides a general overview of the clinical and cellular changes which manifest in cardiac aging, and the quality control mechanisms involved in maintaining homeostasis and retarding aging. These mechanisms include autophagy, ubiquitin-mediated turnover, apoptosis, mitochondrial quality control and cardiac matrix homeostasis. Finally, we discuss aging interventions that have been observed to impact cardiac health outcomes. These include caloric restriction, rapamycin, resveratrol, GDF11, mitochondrial antioxidants and cardiolipin-targeted therapeutics. A greater understanding of the quality control mechanisms that promote cardiac homeostasis will help to understand the benefits of these interventions, and hopefully lead to further improved therapeutic modalities.
Collapse
Affiliation(s)
- Ellen K Quarles
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Dao-Fu Dai
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Autumn Tocchi
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Nathan Basisty
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Lemuel Gitari
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| | - Peter S Rabinovitch
- University of Washington School of Medicine, Department of Pathology, Box 357470, Seattle, WA 98195-7470, United States.
| |
Collapse
|
31
|
Matsushima S, Sadoshima J. The role of sirtuins in cardiac disease. Am J Physiol Heart Circ Physiol 2015; 309:H1375-89. [PMID: 26232232 DOI: 10.1152/ajpheart.00053.2015] [Citation(s) in RCA: 266] [Impact Index Per Article: 26.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 07/27/2015] [Indexed: 12/25/2022]
Abstract
Modification of histones is one of the important mechanisms of epigenetics, in which genetic control is determined by factors other than an individual's DNA sequence. Sirtuin family proteins, which are class III histone deacetylases, were originally identified as gene silencers that affect the mating type of yeast, leading to the name "silent mating-type information regulation 2" (SIR2). They are characterized by their requirement of nicotinamide adenine dinucleotide for their enzyme activity, unlike other classes of histone deacetylases. Sirtuins have been traditionally linked to longevity and the beneficial effects of calorie restriction and DNA damage repair. Recently, sirtuins have been shown to be involved in a wide range of physiological and pathological processes, including aging, energy responses to low calorie availability, and stress resistance, as well as apoptosis and inflammation. Sirtuins can also regulate mitochondrial biogenesis and circadian clocks. Seven sirtuin family proteins (Sirt1-7) have been identified as mammalian SIR2 orthologs, localized in different subcellular compartments, namely, the cytoplasm (Sirt1, 2), the mitochondria (Sirt3, 4, 5), and the nucleus (Sirt1, 2, 6, 7). Sirt1 is evolutionarily close to yeast SIR2 and has been the most intensively investigated in the cardiovascular system. Endogenous Sirt1 plays a pivotal role in mediating the cell death/survival process and has been implicated in the pathogenesis of cardiovascular disease. Downregulation of Sirt2 is protective against ischemic-reperfusion injury. Increased Sirt3 expression has been shown to correlate with longevity in humans. In addition, Sirt3 protects cardiomyocytes from aging and oxidative stress and suppresses cardiac hypertrophy. Sirt6 has also recently been demonstrated to attenuate cardiac hypertrophy, and Sirt7 is known to regulate apoptosis and stress responses in the heart. On the other hand, the roles of Sirt4 and Sirt5 in the heart remain largely uncharacterized.
Collapse
Affiliation(s)
- Shouji Matsushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; and Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey; and
| |
Collapse
|
32
|
Siagian M, Lousiana M, Santoso DI, Endardjo S. Effects of anaerobic exercise and detraining on the caspase-3 expression of rat ventricular cardiomyocyte. MEDICAL JOURNAL OF INDONESIA 2015. [DOI: 10.13181/mji.v24i2.1220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
Background: Anaerobic physical exercise is a high intensity physical exercise performed in a short time. This exercise can stimulate apoptosis in left ventricular cardiomyocytes. The aim of this study is to analyze the apoptosis of cardiomyocytes after anaerobic exercise and detraining.Methods: Thirty two wistar rats Ratus Novergicus 250-350 grams (8-10 weeks old) were divided into the following groups (n = 4) and given naerobic physical exercise four and 12 weeks (group Exc-4, Exc-12) and anaerobic exercise followed by four weeks of detraining (Exc-4-D, Exc-12-D). The control groups were only observed in the same period (group CTL-4, CTL-12, CTL-4-D, CTL-12-D). At the end of observation, the rats were sacrificed and examination of the expression of caspase-3 as an indicator of apoptosis was done using immunohistochemical staining. Data were analyzed with ANOVA test.Results: An increase in expression of caspase-3 in the group Exc-4 (72.03%) compared to the CTL-4 (27.22%), (p < 0,001); and Exc-12 (79.30%) compared to the CTL-12 (30.53%) (p = 0.027). Detraining process showed a significant decline Capase-3 expression (31.12% in exc-4-D and 30.44% in the exc-12-D).Conclusion: Anaerobic physical exercise can increase apoptosis in rat left ventricle cardiomyocyte characterized by increased expression of caspase-3. Detraining can improve heart condition characterized by decreased expression of caspase-3.
Collapse
|
33
|
Isgaard J, Arcopinto M, Karason K, Cittadini A. GH and the cardiovascular system: an update on a topic at heart. Endocrine 2015; 48:25-35. [PMID: 24972804 PMCID: PMC4328125 DOI: 10.1007/s12020-014-0327-6] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Accepted: 06/04/2014] [Indexed: 01/06/2023]
Abstract
In this review, the importance of growth hormone (GH) for the maintenance of normal cardiac function in adult life is discussed. Physiological effects of GH and underlying mechanisms for interactions between GH and insulin-like growth factor I (IGF-I) and the cardiovascular system are covered as well as the cardiac dysfunction caused both by GH excess (acromegaly) and by GH deficiency in adult hypopituitary patients. In both acromegaly and adult GH deficiency, there is also increased cardiovascular morbidity and mortality possibly linked to aberrations in GH status. Finally, the status of the GH/IGF-I system in relation to heart failure and the potential of GH as a therapeutic tool in the treatment of heart failure are reviewed in this article.
Collapse
Affiliation(s)
- Jörgen Isgaard
- Laboratory of Experimental Endocrinology, Department of Internal Medicine, Sahlgrenska Academy, University of Gothenburg, Gröna Stråket 8, 413 45, Göteborg, Sweden,
| | | | | | | |
Collapse
|
34
|
Shukla SK, Sharma SB, Singh UR. Pre-treatment with α-tocopherol and Terminalia arjuna ameliorates, pro-inflammatory cytokines, cardiac and apoptotic markers in myocardial infracted rats. Redox Rep 2014; 20:49-59. [PMID: 25180938 DOI: 10.1179/1351000214y.0000000104] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022] Open
Abstract
OBJECTIVE This study was aimed to evaluate the cardioprotective potential of combination of T. arjuna and α-tocopherol in isoproterenol induced myocardial injury. METHODS Wistar albino rats were pre-treated with hydroalcoholic extract of T. arjuna (HETA) and α-tocopherol (100 mg/kg b. w) daily for 30 days. Isoproterenol (ISP, 85 mg/kg b.w) was administered on 28th and 29th days at an interval of 24 hr. RESULTS ISP treated rats showed significant increase in lipid peroxidation (MDA), cardiac markers (CK-MB, SGOT, Trop I and LDH), pro-inflammatory cytokine (IL-6, CRP, TNF-α) levels and apoptotic markers (Bcl-2/Bax) as compared to healthy group. Pre-treatment with HETA 100 mg/kg b. w, reduced the elevated levels of these markers and significant effect (p<0.05) were observed with the combination of HETA and α-tocopherol at a dose of 100 mg/kg b. w, which was further confirmed by histopathological studies. CONCLUSION The present study concluded that the combination of α-tocopherol (100 mg/kg b. w) and hydroalcoholic extract of T. arjuna (100 mg/kg b. w) augments endogenous antioxidant compounds of rat heart and also prevents the myocardium from ISP-induced myocardial injury and it may have therapeutic and prophylactic value in the treatment of ischemic heart disease.
Collapse
|
35
|
Kwak HB, Lee Y, Kim JH, Van Remmen H, Richardson AG, Lawler JM. MnSOD overexpression reduces fibrosis and pro-apoptotic signaling in the aging mouse heart. J Gerontol A Biol Sci Med Sci 2014; 70:533-44. [PMID: 25016531 DOI: 10.1093/gerona/glu090] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 05/20/2014] [Indexed: 01/15/2023] Open
Abstract
Contractility of the heart is impaired with advancing age via mechanical remodeling, as myocytes are lost through apoptosis and collagenous fibers accumulate. Exercise training confers protection against fibrosis and apoptosis in the aging heart, but the mechanisms remain poorly understood. We recently reported that exercise training elevates Mn isoform of superoxide dismutase (MnSOD) in the aging heart, concomitant with reduction in oxidative stress and fibrosis. Here, we tested the hypothesis that overexpression of MnSOD would be causal in protection against fibrosis and apoptosis in the aging heart. Hearts were extracted from young (8 months) wild-type, young mice overexpressing the Sod2 (MnSOD) gene, old (28 months) wild-type, and old transgenic mice. Left ventricle MnSOD protein levels were elevated in young mice overexpressing the Sod2 (MnSOD) gene and old transgenic mice. MnSODTg mice exhibited lower oxidative stress (total hydroperoxides, 4-hydroxynonenal, and 8-isoprostane) in the old group. Age-related cardiac remodeling and fibrosis was mitigated in MnSOD Tg mice with reductions in extramyocyte space (-65%), collagen-I, and transforming growth factor-β. Pro-apoptotic markers Bax (-38%) and caspase-3 cleavage (-41%) were reduced and apoptosis (terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling-positive nuclei, DNA laddering) was mitigated in MnSOD Tg hearts compared with old wild-type. We conclude that MnSOD elevation is indeed protective against oxidative stress, fibrosis, and apoptosis in the aging heart.
Collapse
Affiliation(s)
- Hyo-Bum Kwak
- Department of Health and Kinesiology, Redox Biology and Cell Signaling Laboratory, Texas A&M University, College Station. Department of Kinesiology, Inha University, Incheon, South Korea
| | - Yang Lee
- Department of Health and Kinesiology, Redox Biology and Cell Signaling Laboratory, Texas A&M University, College Station
| | - Jong-Hee Kim
- Department of Health and Kinesiology, Redox Biology and Cell Signaling Laboratory, Texas A&M University, College Station. Department of Health and Human Performance, University of Houston, Texas
| | - Holly Van Remmen
- Free Radical Biology and Aging Research Program, Oklahoma Medical Research Foundation
| | - Arlan G Richardson
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Science Center
| | - John M Lawler
- Department of Health and Kinesiology, Redox Biology and Cell Signaling Laboratory, Texas A&M University, College Station.
| |
Collapse
|
36
|
Knowlton AA, Korzick DH. Estrogen and the female heart. Mol Cell Endocrinol 2014; 389:31-9. [PMID: 24462775 PMCID: PMC5709037 DOI: 10.1016/j.mce.2014.01.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/04/2014] [Accepted: 01/05/2014] [Indexed: 12/24/2022]
Abstract
Estrogen has a plethora of effects in the cardiovascular system. Studies of estrogen and the heart span human clinical trials and basic cell and molecular investigations. Greater understanding of cell and molecular responses to estrogens can provide further insights into the findings of clinical studies. Differences in expression and cellular/intracellular distribution of the two main receptors, estrogen receptor (ER) α and β, are thought to account for the specificity and differences in responses to estrogen. Much remains to be learned in this area, but cellular distribution within the cardiovascular system is becoming clearer. Identification of GPER as a third ER has introduced further complexity to the system. 17β-estradiol (E2), the most potent human estrogen, clearly has protective properties activating a signaling cascade leading to cellular protection and also influencing expression of the protective heat shock proteins (HSP). E2 protects the heart from ischemic injury in basic studies, but the picture is more involved in the whole organism and clinical studies. Here the complexity of E2's widespread effects comes into play and makes interpretation of findings more challenging. Estrogen loss occurs primarily with aging, but few studies have used aged models despite clear evidence of differences between the response to estrogen deficiency in adult and aged animals. Thus more work is needed focusing on the effects of aging vs. estrogen loss on the cardiovascular system.
Collapse
Affiliation(s)
- A A Knowlton
- The Department of Veteran's Affairs, Northern California VA, Sacramento, CA, USA; Molecular & Cellular Cardiology, Departments of Medicine and Pharmacology, University of California, Davis, USA.
| | - D H Korzick
- Intercollege Program in Physiology and Department of Kinesiology, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
37
|
Kovina MV, Zuev VA, Kagarlitskiy GO, Khodarovich YM. Effect on lifespan of high yield non-myeloablating transplantation of bone marrow from young to old mice. Front Genet 2013; 4:144. [PMID: 23967009 PMCID: PMC3736046 DOI: 10.3389/fgene.2013.00144] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Accepted: 07/15/2013] [Indexed: 11/30/2022] Open
Abstract
Tissue renewal is a well-known phenomenon by which old and dying-off cells of various tissues of the body are replaced by progeny of local or circulating stem cells (SCs). An interesting question is whether donor SCs are capable to prolong the lifespan of an aging organism by tissue renewal. In this work, we investigated the possible use of bone marrow (BM) SC for lifespan extension. To this purpose, chimeric C57BL/6 mice were created by transplanting BM from young 1.5-month-old donors to 21.5-month-old recipients. Transplantation was carried out by means of a recently developed method which allowed to transplant without myeloablation up to 1.5 × 108 cells, that is, about 25% of the total BM cells of the mouse. As a result, the mean survival time, counting from the age of 21.5 months, the start of the experiment, was +3.6 and +5.0 (±0.1) months for the control and experimental groups, respectively, corresponding to a 39 ± 4% increase in the experimental group over the control. In earlier studies on BM transplantation, a considerably smaller quantity of donor cells (5 × 106) was used, about 1% of the total own BM cells. The recipients before transplantation were exposed to a lethal (for control animals) X-ray dose which eliminated the possibility of studying the lifespan extension by this method.
Collapse
|
38
|
Kwak HB. Aging, exercise, and extracellular matrix in the heart. J Exerc Rehabil 2013; 9:338-47. [PMID: 24278882 PMCID: PMC3836529 DOI: 10.12965/jer.130049] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 06/13/2013] [Accepted: 06/20/2013] [Indexed: 11/22/2022] Open
Abstract
Aging is characterized by a progressive impairment of (a) cardiac structure including fibrosis and cardiomyocyte density, and (b) cardiac function including stroke volume, ejection fraction, and cardiac output. The cardiac remodeling involves loss of cardiac myocytes, reactive hypertrophy of the remaining cells, and increased extracellular matrix (ECM) and fibrosis in the aging heart, especially left ventricles. Fibrosis (i.e., accumulation of collagen) with aging is very critical in impairing cardiac function associated with increased myocardial stiffness. The balance of ECM remodeling via ECM synthesis and degradation is essential for normal cardiac structure and function. Thus an understanding of upstream ECM regulatory factors such as matrix metalloproteinases (MMPs), tissue inhibitors of metalloproteinases (TIMPs), tumor necrosis factor-α (TNF-α), transforming growth factor-β (TGF-β), and myofibroblasts is necessary for gaining new insights into managing cardiac remodeling and dysfunction with aging. In contrast, exercise training effectively improves cardiac function in both young and older individuals. Exercise training also improves maximal cardiovascular function by increasing stroke volume and cardiac output. However, limited data indicate that exercise training might attenuate collagen content and remodeling in the aging heart. We recently found that 12 weeks of exercise training protected against geometric changes of collagen ECM in the aging heart and ameliorated age-associated dysregulation of ECM in the heart, as indicated by up-regulation of active MMPs as well as down-regulation of TIMPs and TGF-β. This review will provide a summary and discussion of aging and exercise effects on fibrosis and upstream regulators of ECM in the heart.
Collapse
Affiliation(s)
- Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon, Korea
| |
Collapse
|
39
|
Kwak HB. Effects of aging and exercise training on apoptosis in the heart. J Exerc Rehabil 2013; 9:212-9. [PMID: 24278863 PMCID: PMC3836520 DOI: 10.12965/jer.130002] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Revised: 03/04/2013] [Accepted: 04/02/2013] [Indexed: 12/30/2022] Open
Abstract
Aging is characterized by a progressive decline in cardiac function. A critical contributor to the age-related impairment in cardiac function is the loss of cardiac myocytes through “apoptosis”, or programmed cell death. Structural remodeling in the heart with advancing age includes (a) loss of cardiomyocytes, (b) reactive hypertrophy of the remaining cardiomyocytes, and (c) increased connective tissue and altered geometry. The loss of cardiomyocytes with aging occurs through apoptosis. Particularly, mitochondrial-mediated apoptotic pathway is the best characterized and believed critical in regulating apoptosis with aging, suggesting that mitochondria are very important sites of programmed cell death. It has been also reported that mitochondrial dysfunction, oxidative stress, and impaired stress response contribute to age-induced mechanical remodeling as well as apoptosis. In contrast, exercise training not only improves cardiac function, but also reduces the risk of heart disease. We recently found that aging increased mitochondrial-mediated apoptotic signaling and apoptosis in the left ventricle, while chronic exercise training was effective in diminishing mitochondrial-mediated apoptotic signaling pathways in the aging heart, as indicated by lower DNA fragmentation, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling (TUNEL)-positive staining, and caspase-3 cleavage, when compared with left ventricles from the age-matched sedentary group. In this review, we will provide a comprehensive update regarding the effects of aging and exercise training on apoptosis in the heart.
Collapse
Affiliation(s)
- Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon, Korea
| |
Collapse
|
40
|
Korzick DH, Lancaster TS. Age-related differences in cardiac ischemia-reperfusion injury: effects of estrogen deficiency. Pflugers Arch 2013; 465:669-85. [PMID: 23525672 DOI: 10.1007/s00424-013-1255-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2013] [Revised: 02/23/2013] [Accepted: 02/25/2013] [Indexed: 01/17/2023]
Abstract
Despite conflicting evidence for the efficacy of hormone replacement therapy in cardioprotection of postmenopausal women, numerous studies have demonstrated reductions in ischemia/reperfusion (I/R) injury following chronic or acute exogenous estradiol (E2) administration in adult male and female, gonad-intact and gonadectomized animals. It has become clear that ovariectomized adult animals may not accurately represent the combined effects of age and E2 deficiency on reductions in ischemic tolerance seen in the postmenopausal female. E2 is known to regulate the transcription of several cardioprotective genes. Acute, non-genomic E2 signaling can also activate many of the same signaling pathways recruited in cardioprotection. Alterations in cardioprotective gene expression or cardioprotective signal transduction are therefore likely to result within the context of aging and E2 deficiency and may help explain the reduced ischemic tolerance and loss of cardioprotection in the senescent female heart. Quantification of the mitochondrial proteome as it adapts to advancing age and E2 deficiency may also represent a key experimental approach to uncover proteins associated with disruptions in cardiac signaling contributing to age-associated declines in ischemic tolerance. These alterations have important ramifications for understanding the increased morbidity and mortality due to ischemic cardiovascular disease seen in postmenopausal females. Functional perturbations that occur in mitochondrial respiration and Ca(2+) sensitivity with age-associated E2 deficiency may also allow for the identification of alternative therapeutic targets for reducing I/R injury and treatment of the leading cause of death in postmenopausal women.
Collapse
Affiliation(s)
- Donna H Korzick
- Department of Kinesiology, The Pennsylvania State University, University Park, PA 16802, USA.
| | | |
Collapse
|
41
|
Maejima Y, Galeotti J, Molkentin JD, Sadoshima J, Zhai P. Constitutively active MEK1 rescues cardiac dysfunction caused by overexpressed GSK-3α during aging and hemodynamic pressure overload. Am J Physiol Heart Circ Physiol 2012; 303:H979-88. [PMID: 22904158 DOI: 10.1152/ajpheart.00415.2012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Expression of GSK-3α is increased in aging hearts and those subjected to hemodynamic overload. Overexpressed GSK-3α inhibits ERK and enhances pressure overload (PO)-induced cardiac dysfunction. We studied whether suppression of the MEK1/ERK pathway contributes to cardiac responses induced by overexpressed GSK-3α using constitutively active MEK1 (CA-MEK1)/GSK-3α bigenic mice (bigenic mice), which were obtained by crossing cardiac-specific GSK-3α transgenic mice (Tg-GSK) and cardiac-specific CA-MEK1 transgenic mice (Tg-MEK1). The suppression of ERK phosphorylation observed in Tg-GSK was eliminated in bigenic mice. At 12 mo, left ventricular (LV) weight/tibia length, LV weight/body weight, and cardiac myocyte size were significantly smaller in Tg-GSK than in nontransgenic mice (NTg), but were not significantly different between Tg-MEK1 and bigenic mice. The LV ejection fraction (LVEF), fractional shortening (FS), and change in pressure over time were significantly lower in Tg-GSK than in NTg, but were not significantly different between bigenic mice and Tg-MEK1. The increase in apoptosis in Tg-GSK was abolished in bigenic mice, although the increase in fibrosis was not. After PO, the decrease in cardiac hypertrophy and the enhancement of apoptosis seen in Tg-GSK were abrogated in bigenic mice. After PO, the LVEF and FS were significantly reduced in Tg-GSK compared with its sham, but not in NTg, Tg-MEK1, or bigenic mice compared with their respective shams. There was no significant difference in LVEF and FS between bigenic mice and Tg-MEK1 after PO. In conclusion, inhibition of the MEK1/ERK pathway mediates the hypertrophy suppression and cardiac dysfunction caused by GSK-3α overexpression in cardiac myocytes.
Collapse
Affiliation(s)
- Yasuhiro Maejima
- Cardiovascular Research Institute, Department of Cell Biology and Molecular Medicine, University of Medicine and Dentistry, New Jersey, New Jersey Medical School, Newark, New Jersey 07103, USA
| | | | | | | | | |
Collapse
|
42
|
Picard M, Wright KJ, Ritchie D, Thomas MM, Hepple RT. Mitochondrial function in permeabilized cardiomyocytes is largely preserved in the senescent rat myocardium. PLoS One 2012; 7:e43003. [PMID: 22912774 PMCID: PMC3415432 DOI: 10.1371/journal.pone.0043003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Accepted: 07/16/2012] [Indexed: 01/19/2023] Open
Abstract
The aging heart is characterized by a progressive decline in contractile function and diastolic relaxation. Amongst the factors implicated in these changes is a progressive replacement fibrosis secondary to cardiomyocyte death, oxidative damage, and energetic deficit, each of which may be secondary to impaired mitochondrial function. Here, we performed an in-depth examination of mitochondrial function in saponin-permeabilized cardiomyocyte bundles, a preparation where all mitochondria are represented and their structure intact, from young adult (YA) and senescent (SEN) rats (n = 8 per group). When accounting for increased fibrosis (+19%, P<0.01) and proportional decrease in citrate synthase activity in the SEN myocardium (-23%, P<0.05), mitochondrial respiration and reactive oxygen species (H(2)O(2)) emission across a range of energized states was similar between age groups. Accordingly, the abundance of electron transport chain proteins was also unchanged. Likewise, except for CuZnSOD (-37%, P<0.05), the activity of antioxidant enzymes was unaltered with aging. Although time to mitochondrial permeability transition pore (mPTP) opening was decreased (-25%, P<0.05) in the SEN heart, suggesting sensitization to apoptotic stimuli, this was not associated with a difference in apoptotic index measured by ELISA. Collectively, our results suggest that the function of existing cardiac ventricular mitochondria is relatively preserved in SEN rat heart when measured in permeabilized cells.
Collapse
Affiliation(s)
- Martin Picard
- Department of Kinesiology, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Kathryn J. Wright
- Muscle & Aging Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Darmyn Ritchie
- Muscle & Aging Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Melissa M. Thomas
- Muscle & Aging Laboratory, Faculty of Kinesiology, University of Calgary, Calgary, Alberta, Canada
| | - Russell T. Hepple
- Department of Kinesiology, Department of Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
43
|
Abstract
Regulation of organ growth is critical during embryogenesis. At the cellular level, mechanisms controlling the size of individual embryonic organs include cell proliferation, differentiation, migration, and attrition through cell death. All these mechanisms play a role in cardiac morphogenesis, but experimental studies have shown that the major determinant of cardiac size during prenatal development is myocyte proliferation. As this proliferative capacity becomes severely restricted after birth, the number of cell divisions that occur during embryogenesis limits the growth potential of the postnatal heart. We summarize here current knowledge concerning regional control of myocyte proliferation as related to cardiac morphogenesis and dysmorphogenesis. There are significant spatial and temporal differences in rates of cell division, peaking during the preseptation period and then gradually decreasing toward birth. Analysis of regional rates of proliferation helps to explain the mechanics of ventricular septation, chamber morphogenesis, and the development of the cardiac conduction system. Proliferation rates are influenced by hemodynamic loading, and transduced by autocrine and paracrine signaling by means of growth factors. Understanding the biological response of the developing heart to such factors and physical forces will further our progress in engineering artificial myocardial tissues for heart repair and designing optimal treatment strategies for congenital heart disease.
Collapse
Affiliation(s)
- David Sedmera
- Charles University in Prague, First Faculty of Medicine, Institute of Anatomy, Prague, Czech Republic.
| | | |
Collapse
|
44
|
Anversa P, Olivetti G. Cellular Basis of Physiological and Pathological Myocardial Growth. Compr Physiol 2011. [DOI: 10.1002/cphy.cp020102] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
45
|
Ungvari Z, Kaley G, de Cabo R, Sonntag WE, Csiszar A. Mechanisms of vascular aging: new perspectives. J Gerontol A Biol Sci Med Sci 2010; 65:1028-41. [PMID: 20576649 DOI: 10.1093/gerona/glq113] [Citation(s) in RCA: 369] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
This review focuses on molecular, cellular, and functional changes that occur in the vasculature during aging; explores the links between mitochondrial oxidative stress, inflammation, and development of vascular disease in the elderly patients; and provides a landscape of molecular mechanisms involved in cellular oxidative stress resistance, which could be targeted for the prevention or amelioration of unsuccessful vascular aging. Practical interventions for prevention of age-associated vascular dysfunction and disease in old age are considered here based on emerging knowledge of the effects of anti-inflammatory treatments, regular exercise, dietary interventions, and caloric restriction mimetics.
Collapse
Affiliation(s)
- Zoltan Ungvari
- Department of Geriatric Medicine, Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1303, Oklahoma City, OK 73104, USA.
| | | | | | | | | |
Collapse
|
46
|
Gallogly MM, Shelton MD, Qanungo S, Pai HV, Starke DW, Hoppel CL, Lesnefsky EJ, Mieyal JJ. Glutaredoxin regulates apoptosis in cardiomyocytes via NFkappaB targets Bcl-2 and Bcl-xL: implications for cardiac aging. Antioxid Redox Signal 2010; 12:1339-53. [PMID: 19938943 PMCID: PMC2864653 DOI: 10.1089/ars.2009.2791] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Cardiomyocyte apoptosis is a well-established contributor to irreversible injury following myocardial infarction (MI). Increased cardiomyocyte apoptosis is associated also with aging in animal models, exacerbated by MI; however, mechanisms for this increased sensitivity to oxidative stress are unknown. Protein mixed-disulfide formation with glutathione (protein glutathionylation) is known to change the function of intermediates that regulate apoptosis. Since glutaredoxin (Grx) specifically catalyzes protein deglutathionylation, we examined its status with aging and its influence on regulation of apoptosis. Grx1 content and activity are decreased by approximately 40% in elderly (24-mo) Fischer 344 rat hearts compared to adult (6-mo) controls. A similar extent of Grx1 knockdown in H9c2 cardiomyocytes led to increased apoptosis, decreased NFkappaB-dependent transcriptional activity, and decreased production (mRNA and protein) of anti-apoptotic NFkappaB target genes, Bcl-2 and Bcl-xL. Knockdown of Bcl-2 and/or Bcl-xL in wild-type H9c2 cells to the same extent ( approximately 50%) as observed in Grx1-knockdown cells increased baseline apoptosis; and knockdown of Bcl-xL, but not Bcl-2, also increased oxidant-induced apoptosis analogous to Grx1-knockdown cells. Natural Grx1-deficient cardiomyocytes isolated from elderly rats also displayed diminished NFkappaB activity and Bcl-xL content. Taken together, these data indicate diminution of Grx1 in elderly animals contributes to increased apoptotic susceptibility via regulation of NFkappaB function.
Collapse
Affiliation(s)
- Molly M Gallogly
- Department of Pharmacology, Case Western Reserve University, School of Medicine, Cleveland, Ohio 44106-4965, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Characterization of an animal model of postmenopausal cardiac hypertrophy and novel mechanisms responsible for cardiac decompensation using ovariectomized pressure-overloaded rats. Menopause 2010; 17:213-21. [PMID: 19741553 DOI: 10.1097/gme.0b013e3181b57489] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The development of animal models of cardiovascular disease are critical to define pathophysiological mechanisms and to advance diagnosis and therapy. The lack of a suitable animal model represents a failure to define the mechanisms responsible for postmenopausal myocardial hypertrophy in hypertension and adverse cardiac remodeling. METHODS In this review, we presented a rat model of postmenopausal myocardial hypertrophy, with particular focus on the similarities between the animal model and postmenopausal women regarding myocardial function as well as molecular and subcellular mechanisms. To elucidate the molecular mechanism of left ventricular (LV) hypertrophy and remodeling in postmenopausal women, we analyzed myocardial hypertrophy as well as cardiac function and hypertrophy-related protein expression in ovariectomized (OVX) and pressure overloaded (PO) rats. RESULTS The model is characterized by depletion of serum estrogen and increased heart-to-body weight and lung-to-body weight ratios. Moreover, the OVX-PO rats also show increased mean arterial blood pressure, LV end-diastolic pressure, LV developed pressure, and maximal rates of LV contraction and relaxation compared with the OVX group. Importantly, Akt activity was largely attenuated, and both endothelial nitric oxide synthase expression and activity were markedly reduced in the OVX-PO group. Finally, significant increased mortality was observed in the OVX-PO group after chronic isoproterenol administration. CONCLUSIONS Our results demonstrate that rats subject to OVX are unable to compensate for hypertrophy partly due to impaired Akt-endothelial nitric oxide synthase signaling along with deteriorated heart function and demonstrated increased mortality. In this review, we discussed the mechanisms of cardiac injury, which could play a critical role in postmenopausal hypertrophy, as well as the characteristics of the OVX-PO female rats as a model to test cardioprotective drugs in postmenopausal women.
Collapse
|
48
|
Howlett SE. Age-associated changes in excitation-contraction coupling are more prominent in ventricular myocytes from male rats than in myocytes from female rats. Am J Physiol Heart Circ Physiol 2009; 298:H659-70. [PMID: 19966062 DOI: 10.1152/ajpheart.00214.2009] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We evaluated effects of age on components of excitation-contraction (EC) coupling in ventricular myocytes from male and female rats to examine sex differences in mechanisms responsible for age-related contractile dysfunction. Myocytes were isolated from anesthetized young adult (approximately 3 mo) and aged (approximately 24 mo) Fischer 344 rats. Ca(2+) concentrations and contractions were measured simultaneously (37 degrees C, 2 Hz). Fractional shortening declined with age in males (6.7 +/- 0.6% to 2.4 +/- 0.4%; P < 0.05), as did peak Ca(2+) transients (47.7 +/- 4.6 to 28.1 +/- 2.1 nM; P < 0.05) and Ca(2+) current densities (-7.7 +/- 0.7 to -6.2 +/- 0.5 pA/pF; P < 0.05). Although sarcoplasmic reticulum (SR) Ca(2+) content was similar regardless of age in males, EC coupling gain declined significantly with age to 55.8 +/- 7.8% of values in younger males. In contrast with results in males, contraction and Ca(2+) transient amplitudes were unaffected by age in females. Ca(2+) current density declined with age in females (-7.5 +/- 0.5 to -5.1 +/- 0.7 pA/pF; P < 0.05), but SR Ca(2+) content actually increased dramatically (49.0 +/- 7.5 to 147.3 +/- 28.5 nM; P < 0.05). Even so, EC coupling gain was not affected by age in female myocytes. Age also promoted hypertrophy of male myocytes more than female myocytes. Age and sex differences in EC coupling were largely maintained when conditioning pulse frequency was increased to 4 Hz. Contractions, Ca(2+) transients, and EC coupling gain were also smaller in young females than in young males. Thus age-dependent changes are more prominent in myocytes from males than females. Increased SR Ca(2+) content may compensate for reduced Ca(2+) current to preserve contractile function in aged females, which may limit the detrimental effects of age on cardiac contractile function.
Collapse
Affiliation(s)
- Susan E Howlett
- Department of Pharmacology and Division of Geriatric Medicine, Dalhousie University, Halifax, Nova Scotia, Canada.
| |
Collapse
|
49
|
Lee J, Cho HS, Park S, Kim WK. Regular Exercise Produced Cardioprotective Effects on Rat's Heart with Hypertension Induced by L-NAME Administration. Clin Exp Hypertens 2009; 31:364-75. [DOI: 10.1080/10641960902977924] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
50
|
Hofer T, Servais S, Seo AY, Marzetti E, Hiona A, Upadhyay SJ, Wohlgemuth SE, Leeuwenburgh C. Bioenergetics and permeability transition pore opening in heart subsarcolemmal and interfibrillar mitochondria: effects of aging and lifelong calorie restriction. Mech Ageing Dev 2009; 130:297-307. [PMID: 19428447 PMCID: PMC2680750 DOI: 10.1016/j.mad.2009.01.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Revised: 01/12/2009] [Accepted: 01/21/2009] [Indexed: 10/21/2022]
Abstract
Loss of cardiac mitochondrial function with age may cause increased cardiomyocyte death through mitochondria-mediated release of apoptogenic factors. We investigated ventricular subsarcolemmal (SSM) and interfibrillar (IFM) mitochondrial bioenergetics and susceptibility towards Ca(2+)-induced permeability transition pore (mPTP) opening with aging and lifelong calorie restriction (CR). Cardiac mitochondria were isolated from 8-, 18-, 29- and 37-month-old male Fischer 344 x Brown Norway rats fed either ad libitum (AL) or 40% calorie restricted diets. With age, H(2)O(2) generation did not increase and oxygen consumption did not significantly decrease in either SSM or IFM. Strikingly, IFM displayed an increased susceptibility towards mPTP opening during senescence. In contrast, Ca(2+) retention capacity of SSM was not affected by age, but SSM tolerated much less Ca(2+) than IFM. Only modest age-dependent increases in cytosolic caspase activities and cytochrome c levels were observed and were not affected by CR. Levels of putative mPTP-modulating components: cyclophilin-D, the adenine nucleotide translocase (ANT), and the voltage-dependent ion channel (VDAC) were not affected by aging or CR. In summary, the age-related reduction of Ca(2+) retention capacity in IFM may explain the increased susceptibility to stress-induced cell death in the aged myocardium.
Collapse
Affiliation(s)
- Tim Hofer
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
| | - Stephane Servais
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
| | - Arnold Young Seo
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
| | - Emanuele Marzetti
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
- Department of Gerontology, Geriatrics and Physiatrics, Catholic University of the Sacret Heart, Rome 00168, Italy
| | - Asimina Hiona
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
| | - Shashank Jagdish Upadhyay
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
| | - Stephanie Eva Wohlgemuth
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
| | - Christiaan Leeuwenburgh
- Department of Aging and Geriatric Research, College of Medicine, Institute on Aging, Division of Biology of Aging, University of Florida, Gainesville 32610, USA
| |
Collapse
|