1
|
Choi Y, No MH, Heo JW, Cho EJ, Park DH, Kang JH, Kim CJ, Seo DY, Han J, Kwak HB. Resveratrol attenuates aging-induced mitochondrial dysfunction and mitochondria-mediated apoptosis in the rat heart. Nutr Res Pract 2025; 19:186-199. [PMID: 40226768 PMCID: PMC11982693 DOI: 10.4162/nrp.2025.19.2.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 10/03/2024] [Accepted: 10/15/2024] [Indexed: 04/15/2025] Open
Abstract
BACKGROUD/OBJECTIVES Resveratrol, a natural polyphenolic compound, has potent antioxidant and anti-inflammatory properties, leading to beneficial effects against cardiovascular diseases. The purpose of this study was to determine whether resveratrol induces protective effects against aging-induced cardiac remodeling, mitochondrial dysfunction, and mitochondria-mediated apoptosis in the heart. MATERIALS/METHODS Thirty-two male Fischer 344 rats were divided into 4 groups: 2 groups that were orally treated with resveratrol (50 mg/kg/day) for 6 weeks (young and old resveratrol groups), and 2 control groups (young and old control groups). Mitochondrial function and mitochondria-mediated apoptotic pathway were analyzed in cardiac muscle fibers from the left ventricle. RESULTS Resveratrol significantly reduced cardiac hypertrophy and remodeling in aging hearts. In addition, resveratrol significantly ameliorated aging-induced mitochondrial dysfunction (e.g., decreased oxygen respiration and increased hydrogen peroxide emission) and mitochondria-dependent apoptotic signaling (the Bax/Bcl-2 ratio, mitochondrial permeability transition pore opening sensitivity, and cleaved caspase-3 protein levels). Resveratrol also significantly attenuated aging-induced apoptosis (determined via cleaved caspase-3 staining and TUNEL-positive myonuclei) in cardiac muscles. CONCLUSION This study demonstrates that resveratrol treatment has a beneficial effect on aging-induced cardiac remodeling by ameliorating mitochondrial dysfunction and inhibiting mitochondria-mediated apoptosis in the heart.
Collapse
Affiliation(s)
- Youngju Choi
- Institute of Sports and Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
- Institute for Specialized Teaching and Research, Inha University, Incheon 22212, Korea
- Department of Kinesiology, Inha University, Incheon 22212, Korea
| | - Mi-Hyun No
- Institute of Sports and Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
| | - Jun-Won Heo
- Department of Biomedical Science and Engineering, Inha University, Incheon 22212, Korea
| | - Eun-Jeong Cho
- Department of Biomedical Science and Engineering, Inha University, Incheon 22212, Korea
| | - Dong-Ho Park
- Institute of Sports and Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
- Department of Kinesiology, Inha University, Incheon 22212, Korea
- Department of Biomedical Science and Engineering, Inha University, Incheon 22212, Korea
| | - Ju-Hee Kang
- Institute of Sports and Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
- Department of Biomedical Science and Engineering, Inha University, Incheon 22212, Korea
- Department of Pharmacology, College of Medicine, Inha University, Incheon 22212, Korea
| | - Chang-Ju Kim
- Department of Physiology, College of Medicine, Kyung Hee University, Seoul 02447, Korea
| | - Dae Yun Seo
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, College of Medicine, Cardiovascular and Metabolic Disease Center, Inje University, Busan 47392, Korea
| | - Hyo-Bum Kwak
- Institute of Sports and Arts Convergence (ISAC), Inha University, Incheon 22212, Korea
- Department of Kinesiology, Inha University, Incheon 22212, Korea
- Department of Biomedical Science and Engineering, Inha University, Incheon 22212, Korea
| |
Collapse
|
2
|
Nurkolis F, Utami TW, Alatas AI, Wicaksono D, Kurniawan R, Ratmandhika SR, Sukarno KT, Pahu YGP, Kim B, Tallei TE, Tjandrawinata RR, Alhasyimi AA, Surya R, Helen H, Halim P, Muhar AM, Syahputra RA. Can salivary and skin microbiome become a biodetector for aging-associated diseases? Current insights and future perspectives. FRONTIERS IN AGING 2024; 5:1462569. [PMID: 39484071 PMCID: PMC11524912 DOI: 10.3389/fragi.2024.1462569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 10/02/2024] [Indexed: 11/03/2024]
Abstract
Growth and aging are fundamental elements of human development. Aging is defined by a decrease in physiological activities and higher illness vulnerability. Affected by lifestyle, environmental, and hereditary elements, aging results in disorders including cardiovascular, musculoskeletal, and neurological diseases, which accounted for 16.1 million worldwide deaths in 2019. Stress-induced cellular senescence, caused by DNA damage, can reduce tissue regeneration and repair, promoting aging. The root cause of many age-related disorders is inflammation, encouraged by the senescence-associated secretory phenotype (SASP). Aging's metabolic changes and declining immune systems raise illness risk via promoting microbiome diversity. Stable, individual-specific skin and oral microbiomes are essential for both health and disease since dysbiosis is linked with periodontitis and eczema. Present from birth to death, the human microbiome, under the influence of diet and lifestyle, interacts symbiotically with the body. Poor dental health has been linked to Alzheimer's and Parkinson's diseases since oral microorganisms and systemic diseases have important interactions. Emphasizing the importance of microbiome health across the lifetime, this study reviews the understanding of the microbiome's role in aging-related diseases that can direct novel diagnosis and treatment approaches.
Collapse
Affiliation(s)
- Fahrul Nurkolis
- Department of Biological Sciences, Faculty of Sciences and Technology, State Islamic University of Sunan Kalijaga (UIN Sunan Kalijaga), Yogyakarta, Indonesia
| | - Trianna Wahyu Utami
- Department of Dental Biomedical Sciences, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Aiman Idrus Alatas
- Program of Clinical Microbiology Residency, Faculty of Medicine, Universitas Indonesia, Jakarta, Indonesia
| | - Danar Wicaksono
- Alumnus Department of Dermatology and Venereology, Faculty of Medicine, Public Health, and Nursing, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Rudy Kurniawan
- Graduate School of Medicine, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | | | | | | | - Bonglee Kim
- Department of Pathology, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado, Indonesia
| | | | - Ananto Ali Alhasyimi
- Department of Orthodontics, Faculty of Dentistry, Universitas Gadjah Mada, Yogyakarta, Indonesia
| | - Reggie Surya
- Department of Food Technology, Faculty of Engineering, Bina Nusantara University, Jakarta, Indonesia
| | - Helen Helen
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Princella Halim
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| | - Adi Muradi Muhar
- Faculty of Medicine, Universitas Sumatera Utara, Medan, Indonesia
| | - Rony Abdi Syahputra
- Department of Pharmacology, Faculty of Pharmacy, Universitas Sumatera Utara, Medan, Indonesia
| |
Collapse
|
3
|
Mehdipour M, Park S, Wei W, Long JZ, Huang GN. Ophthalmic acid is a bloodborne metabolite that contributes to age-induced cardiomyocyte hypertrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.08.607218. [PMID: 39211274 PMCID: PMC11361124 DOI: 10.1101/2024.08.08.607218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Cardiac aging involves the development of left ventricular hypertrophy alongside a decline in functional capacity. Here, we use neutral blood exchange to demonstrate that the acute removal of age-accumulated blood factors significantly regresses cardiac hypertrophy in aged mice. The reversal of hypertrophy was not attributed to age-associated hemodynamic effects, implicating a role of blood-derived factors. In addition, the overarching paradigm of systemic aging maintains that the age-related overabundance of plasma proteins are largely responsible for causing pathological phenotypes in tissues. Our results suggest that blood metabolites, not proteins, drive cardiac hypertrophy instead. Upon analyzing serum metabolomics data sets, we identified ophthalmic acid as a circulating metabolite whose levels increase with advanced age. Treatment of adult mouse and neonatal rat cardiomyocytes in culture with ophthalmic acid increased their relative surface areas. This study uncovers a non-protein metabolite that may contribute to cardiomyocyte hypertrophy during aging. Identifying a method to counteract ophthalmic acid's hypertrophic effects may reveal novel therapeutic opportunities for cardiac rejuvenation.
Collapse
|
4
|
Westhoff M, Del Villar SG, Voelker TL, Thai PN, Spooner HC, Costa AD, Sirish P, Chiamvimonvat N, Dickson EJ, Dixon RE. BIN1 knockdown rescues systolic dysfunction in aging male mouse hearts. Nat Commun 2024; 15:3528. [PMID: 38664444 PMCID: PMC11045846 DOI: 10.1038/s41467-024-47847-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 04/15/2024] [Indexed: 04/28/2024] Open
Abstract
Cardiac dysfunction is a hallmark of aging in humans and mice. Here we report that a two-week treatment to restore youthful Bridging Integrator 1 (BIN1) levels in the hearts of 24-month-old mice rejuvenates cardiac function and substantially reverses the aging phenotype. Our data indicate that age-associated overexpression of BIN1 occurs alongside dysregulated endosomal recycling and disrupted trafficking of cardiac CaV1.2 and type 2 ryanodine receptors. These deficiencies affect channel function at rest and their upregulation during acute stress. In vivo echocardiography reveals reduced systolic function in old mice. BIN1 knockdown using an adeno-associated virus serotype 9 packaged shRNA-mBIN1 restores the nanoscale distribution and clustering plasticity of ryanodine receptors and recovers Ca2+ transient amplitudes and cardiac systolic function toward youthful levels. Enhanced systolic function correlates with increased phosphorylation of the myofilament protein cardiac myosin binding protein-C. These results reveal BIN1 knockdown as a novel therapeutic strategy to rejuvenate the aging myocardium.
Collapse
Affiliation(s)
- Maartje Westhoff
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Silvia G Del Villar
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Taylor L Voelker
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Phung N Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| | - Heather C Spooner
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Alexandre D Costa
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Padmini Sirish
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
| | - Nipavan Chiamvimonvat
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, USA
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA, USA
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - Eamonn J Dickson
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA
| | - Rose E Dixon
- Department of Physiology and Membrane Biology, University of California Davis, Davis, CA, USA.
| |
Collapse
|
5
|
Wang H, Slotabec L, Didik S, Li Z, Leng L, Zhao B, Bucala R, Li J. A small molecule macrophage migration inhibitory factor agonist ameliorates age-related myocardial intolerance to ischemia-reperfusion insults via metabolic regulation. Metabolism 2024; 153:155792. [PMID: 38232801 PMCID: PMC10932879 DOI: 10.1016/j.metabol.2024.155792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/27/2023] [Accepted: 01/09/2024] [Indexed: 01/19/2024]
Abstract
Macrophage migration inhibitory factor (MIF) is an innate cytokine that regulates both inflammatory and homeostatic responses. MIF is expressed by cardiomyocytes, where it exerts a protective action against ischemia-reperfusion (I/R) injury by activating AMP-activated protein kinase (AMPK). This effect is attenuated in the senescent heart due to an intrinsic, age-related reduction in MIF expression. We hypothesized that treating the aged heart with the small molecule MIF agonist (MIF20) can reinforce protective MIF signaling in cardiomyocytes, leading to a beneficial effect against I/R stress. The administration of MIF20 at the onset of reperfusion was found to not only decrease myocardial infarct size but also preserves systolic function in the aged heart. Protection from I/R injury was reduced in mice with cardiomyocyte-specific Mif deletion, consistent with the mechanism of action of MIF20 to allosterically increase MIF affinity for its cognate receptor CD74. We further found MIF20 to contribute to the maintenance of mitochondrial fitness and to preserve the contractile properties of aged cardiomyocytes under hypoxia/reoxygenation. MIF20 augments protective metabolic responses by reducing the NADH/NAD ratio, leading to a decrease in the accumulation of reactive oxygen species (ROS) in the aged myocardium under I/R stress. We also identify alterations in the expression levels of the downstream effectors PDK4 and LCAD, which participate in the remodeling of the cardiac metabolic profile. Data from this study demonstrates that pharmacologic augmentation of MIF signaling provides beneficial homeostatic actions on senescent myocardium under I/R stress.
Collapse
Affiliation(s)
- Hao Wang
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Lily Slotabec
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, United States of America
| | - Steven Didik
- Department of Surgery, University of South Florida, FL 33612, United States of America
| | - Zehui Li
- Department of Surgery, University of South Florida, FL 33612, United States of America
| | - Lin Leng
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, United States of America
| | - Bi Zhao
- Genomics Program, College of Public Health, University of South Florida, Tampa, FL 33612, United States of America.
| | - Richard Bucala
- Department of Internal Medicine, Yale School of Medicine, New Haven, CT 06510, United States of America
| | - Ji Li
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, United States of America; G.V. (Sonny) Montgomery VA Medical Center, Jackson, MS 39216, United States of America.
| |
Collapse
|
6
|
Zhang H, Chen C, Liu Y, Chen W, Qi J, Xu Y, Ren L, Yang G, Min D, Liu Z, Cai X, Hao M, Xu G, Hou P. D-galactose causes sinoatrial node dysfunction: from phenotype to mechanism. Aging (Albany NY) 2023; 15:12551-12569. [PMID: 37950730 PMCID: PMC10683603 DOI: 10.18632/aging.205196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/12/2023] [Indexed: 11/13/2023]
Abstract
With the population aging, age-related sinoatrial node dysfunction (SND) has been on the rise. Sinoatrial node (SAN) degeneration is an important factor for the age-related SND development. However, there is no suitable animal modeling method in this field. Here, we investigated whether D-galactose could induce SAN degeneration and explored the associated mechanism. In vivo, twelve C57BL/6 mice were divided into Control and D-galactose group to receive corresponding treatments. Senescence was confirmed by analyzing the hair and weight; cardiac function was evaluated through echocardiography, cerebral blood flux and serum-BNP; the SAN function was evaluated by electrocardiogram; fibrotic change was evaluated by Masson's trichrome staining and oxidative stress was assessed through DHE staining and serum indicators. Mechanism was verified through immunofluorescence-staining and Western blotting. In vitro, mouse-atrial-myocytes were treated with D-galactose, and edaravone was utilized as the ROS scavenger. Senescence, oxidative stress, proliferation ability and mechanism were verified through various methods, and intuitive evidence was obtained through electrophysiological assay. Finally, we concluded that D-galactose can be used to induce age-related SND, in which oxidative stress plays a key role, causing PITX2 ectopic expression and downregulates SHOX2 expression, then through the downstream GATA4/NKX2-5 axis, results in pacing-related ion channels dysfunction, and hence SND development.
Collapse
Affiliation(s)
- Heng Zhang
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Chen Chen
- Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Yue Liu
- Shenyang Medical College, Shenyang 110034, China
| | - Wei Chen
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Jing Qi
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Yue Xu
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Lu Ren
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Guanlin Yang
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Dongyu Min
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Zhuang Liu
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Xintong Cai
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Miao Hao
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Guanzhen Xu
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| | - Ping Hou
- Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang 116600, China
| |
Collapse
|
7
|
Wu Y, Li M, Zhang J, Wang S. Unveiling uterine aging: Much more to learn. Ageing Res Rev 2023; 86:101879. [PMID: 36764360 DOI: 10.1016/j.arr.2023.101879] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/22/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023]
Abstract
Uterine aging is an important factor that impacts fertility, reproductive health, and uterus-related diseases; however, it remains poorly explored. Functionally, these disturbances have been associated with an abnormal hormonal response in the endometrium and decreased endometrial receptivity. Based on emerging evidence, these alterations are mediated via the senescence of endometrial stem cells and impaired decidualization of endometrial stromal cells. Multiple molecular activities may participate in uterine aging, including oxidative stress, inflammation, fibrosis, DNA damage response, and cellular senescence. Over the past decade, several protective strategies targeting these biological processes have afforded promising results, including stem cell therapy, anti-aging drugs, and herbal medicines. However, the currently available evidence is fragmented and scattered. Here, we summarize the most recent findings regarding uterine aging, including functional and structural alterations and potential cellular and molecular mechanisms, and discuss potential protective interventions against uterine aging. Thereby, we hope to provide a comprehensive understanding of the pathophysiological processes and underlying mechanisms associated with uterine aging, as well as improve fecundity and reproductive outcomes in females of advanced reproductive age.
Collapse
Affiliation(s)
- Yaling Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Milu Li
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinjin Zhang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
8
|
Guo Z, Wang M, Ying X, Yuan J, Wang C, Zhang W, Tian S, Yan X. Caloric restriction increases the resistance of aged heart to myocardial ischemia/reperfusion injury via modulating AMPK-SIRT 1-PGC 1a energy metabolism pathway. Sci Rep 2023; 13:2045. [PMID: 36739302 PMCID: PMC9899227 DOI: 10.1038/s41598-023-27611-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 01/04/2023] [Indexed: 02/06/2023] Open
Abstract
A large number of data suggest that caloric restriction (CR) has a protective effect on myocardial ischemia/reperfusion injury (I/R) in the elderly. However, the mechanism is still unclear. In this study, we created the I/R model in vivo by ligating the mice left coronary artery for 45 min followed by reperfusion. C57BL/6J wild-type mice were randomly divided into a young group fed ad libitum (y-AL), aged fed ad libitum (a-AL) and aged calorie restriction group (a-CR, 70% diet restriction), and fed for 6 weeks. The area of myocardial infarction was measured by Evan's blue-TTC staining, plasma cholesterol content quantified by ELISA, fatty acids and glucose measured by Langendorff working system, as well as protein expression of AMPK/SIRT1/PGC1a signaling pathway related factors in myocardial tissue detected by immunoblotting. Our results showed that CR significantly reduced infarct size in elderly mice after I/R injury, promoted glycolysis regardless of I/R injury, and restored myocardial glucose uptake in elderly mice. Compared with a-AL group, CR significantly promoted the expression of p-AMPK, SIRT1, p-PGC1a, and SOD2, but decreased PPARγ expression in aged mice. In conclusion, our results suggest that CR protects elderly mice from I/R injury by altering myocardial substrate energy metabolism via the AMPK/SIRT1/PGC1a pathway.
Collapse
Affiliation(s)
- Zhijia Guo
- 1st Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Meng Wang
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaodong Ying
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jiyu Yuan
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Chenggang Wang
- Shanxi Traditional Chinese Medicine Hospital, Taiyuan, Shanxi, China
| | - Wenjie Zhang
- 1st Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Shouyuan Tian
- 1st Hospital of Shanxi Medical University, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiaoyan Yan
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China.
| |
Collapse
|
9
|
Ashton KJ, Kiessling CJ, Thompson JLM, Aziz AY, Thomas WG, Headrick JP, Reichelt ME. Early cardiac aging linked to impaired stress-resistance and transcriptional control of stress response, quality control and mitochondrial pathways. Exp Gerontol 2023; 171:112011. [PMID: 36347360 DOI: 10.1016/j.exger.2022.112011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Revised: 10/18/2022] [Accepted: 10/30/2022] [Indexed: 11/06/2022]
Abstract
Phenotypic and transcriptomic evidence of early cardiac aging, and associated mechanisms, were investigated in young to middle-aged male mice (C57Bl/6; ages 8, 16, 32, 48 wks). Left ventricular gene expression (profiled via Illumina MouseWG-6 BeadChips), contractile and coronary function, and stress-resistance were assessed in Langendorff perfused hearts under normoxic conditions and following ischemic insult (20 min global ischemia-45 min reperfusion; I-R). Baseline or normoxic contractile function was unaltered by age, while cardiac and coronary 'reserves' (during β-adrenoceptor stimulation; 1 μM isoproterenol) declined by 48 wks. Resistance to I-R injury fell from 16 to 32 wks. Age-dependent transcriptional changes In un-stressed hearts were limited to 104 genes (>1.3-fold; 0.05 FDR), supporting: up-regulated innate defenses (glutathione and xenobiotic metabolism, chemotaxis, interleukins) and catecholamine secretion; and down-regulated extracellular matrix (ECM), growth factor and survival (PI3K/Akt) signaling. In stressed (post-ischemic) myocardium, ∼15-times as many genes (1528) were age-dependent, grouped into 6 clusters (>1.3-fold change; 0.05 FDR): most changing from 16 wks (45 % up/44 % down), a further 5 % declining from 32 wks. Major age-dependent Biological Processes in I-R hearts reveal: declining ATP metabolism, oxidative phosphorylation, cardiac contraction and morphogenesis, phospholipid metabolism and calcineurin signaling; increasing proteolysis and negative control of MAPK; and mixed changes in nuclear transport and angiogenic genes. Pathway analysis supports reductions in: autophagy, stress response, ER protein processing, mRNA surveillance and ribosome/translation genes; with later falls in mitochondrial biogenesis, oxidative phosphorylation and proteasome genes in I-R hearts. Summarizing, early cardiac aging is evident from 16 to 32 wks in male mice, characterized by: declining cardiovascular reserve and stress-resistance, transcriptomic evidence of constitutive stress and altered catecholamine and survival/growth signaling in healthy hearts; and declining stress response, quality control, mitochondrial energy metabolism and cardiac modeling processes in stressed hearts. These very early changes, potentially key substrate for advanced aging, may inform approaches to healthy aging and cardioprotection in the adult heart.
Collapse
Affiliation(s)
- Kevin J Ashton
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Can J Kiessling
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Jamie-Lee M Thompson
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Aliah Y Aziz
- Faculty of Health Sciences and Medicine, Bond University, Gold Coast, QLD, Australia
| | - Walter G Thomas
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - John P Headrick
- School of Medical Science, Griffith University, Southport, QLD, Australia
| | - Melissa E Reichelt
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
10
|
NRF-2/HO-1 Pathway-Mediated SHOX2 Activation Is a Key Switch for Heart Rate Acceleration by Yixin-Fumai Granules. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8488269. [PMID: 36199421 PMCID: PMC9529460 DOI: 10.1155/2022/8488269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Accepted: 09/12/2022] [Indexed: 11/17/2022]
Abstract
Population aging has led to increased sick sinus syndrome (SSS) incidence; however, no effective and safe medical therapy has been reported thus far. Yixin-Fumai granules (YXFMs), a Chinese medicine granule designed for bradyarrhythmia treatment, can effectively increase SSS patients' heart rate. Senescence-induced sinoatrial node (SAN) degeneration is an important part of SSS pathogenesis, and older people often show high levels of oxidative stress; reactive oxygen species (ROS) accumulation in the SAN causes abnormal SAN pacing or conduction functions. The current study observed the protective effects of YXFMs on senescent SAN and explored the relationship between the NRF-2/HO-1 pathway, SHOX2, and T-type calcium channels. We selected naturally senescent C57BL/6 mice with bradycardia to simulate SSS; electrocardiography, Masson's trichrome staining, and DHE staining were used to assess SAN function and tissue damage. Immunofluorescence staining and Western blotting were used to assay related proteins. In vitro, we treated human-induced pluripotent stem cell-derived atrial myocytes (hiPSC-AMs) and mouse atrial myocyte-derived cell line HL-1 with D-galactose to simulate senescent SAN-pacemaker cells. CardioExcyte96 was used to evaluate the pulsatile function of the hiPSC-AMs, and the mechanism was verified by DCFH-DA, immunofluorescence staining, RT-qPCR, and Western blotting. The results demonstrated that YXFMs effectively inhibited senescence-induced SAN hypofunction, and this effect possibly originated from scavenging of ROS and promotion of NRF-2, SHOX2, and T-type calcium channel expression. In vitro experiment results indicated that ML385, si-SHOX2, LDN193189, and Mibefradil reversed YXFMs' effects. Moreover, we, for the first time, found that ROS accumulation may hinder SHOX2 expression; YXFMs can activate SHOX2 through the NRF-2/HO-1 pathway-mediated ROS scavenging and then regulate CACNA1G through the SHOX2/BMP4/GATA4/NKX2-5 axis, improve T-type calcium channel function, and ameliorate the SAN dysfunction. Finally, through network pharmacology and molecular docking, we screened for the most stable YXFMs compound that docks to NRF-2, laying the foundation for future studies.
Collapse
|
11
|
Alharbi KS, Singh Y, Afzal O, Alfawaz Altamimi AS, Kazmi I, Al-Abbasi FA, Alzarea SI, Chellappan DK, Singh SK, Dua K, Gupta G. Molecular explanation of Wnt/βcatenin antagonist pyrvinium mediated calcium equilibrium changes in aging cardiovascular disorders. Mol Biol Rep 2022; 49:11101-11111. [DOI: 10.1007/s11033-022-07863-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/10/2022] [Accepted: 08/11/2022] [Indexed: 10/14/2022]
|
12
|
Hastings MH, Herrera JJ, Guseh JS, Atlason B, Houstis NE, Abdul Kadir A, Li H, Sheffield C, Singh AP, Roh JD, Day SM, Rosenzweig A. Animal Models of Exercise From Rodents to Pythons. Circ Res 2022; 130:1994-2014. [PMID: 35679366 PMCID: PMC9202075 DOI: 10.1161/circresaha.122.320247] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Acute and chronic animal models of exercise are commonly used in research. Acute exercise testing is used, often in combination with genetic, pharmacological, or other manipulations, to study the impact of these manipulations on the cardiovascular response to exercise and to detect impairments or improvements in cardiovascular function that may not be evident at rest. Chronic exercise conditioning models are used to study the cardiac phenotypic response to regular exercise training and as a platform for discovery of novel pathways mediating cardiovascular benefits conferred by exercise conditioning that could be exploited therapeutically. The cardiovascular benefits of exercise are well established, and, frequently, molecular manipulations that mimic the pathway changes induced by exercise recapitulate at least some of its benefits. This review discusses approaches for assessing cardiovascular function during an acute exercise challenge in rodents, as well as practical and conceptual considerations in the use of common rodent exercise conditioning models. The case for studying feeding in the Burmese python as a model for exercise-like physiological adaptation is also explored.
Collapse
Affiliation(s)
- Margaret H Hastings
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Jonathan J Herrera
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor (J.J.H.)
| | - J Sawalla Guseh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Bjarni Atlason
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Nicholas E Houstis
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Azrul Abdul Kadir
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Haobo Li
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Cedric Sheffield
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Anand P Singh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Jason D Roh
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| | - Sharlene M Day
- Cardiovascular Medicine, Perelman School of Medicine' University of Pennsylvania, Philadelphia (S.M.D.)
| | - Anthony Rosenzweig
- Department of Medicine, Division of Cardiology, Cardiovascular Research Center, Corrigan Minehan Heart Center, Massachusetts General Hospital, Harvard Medical School, Boston (M.H.H., J.S.G., B.A., N.E.H., A.A.K., H.L., C.S., A.P.S., J.D.R., A.R.)
| |
Collapse
|
13
|
Dixon RE. Nanoscale Organization, Regulation, and Dynamic Reorganization of Cardiac Calcium Channels. Front Physiol 2022; 12:810408. [PMID: 35069264 PMCID: PMC8769284 DOI: 10.3389/fphys.2021.810408] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2021] [Accepted: 11/30/2021] [Indexed: 12/19/2022] Open
Abstract
The architectural specializations and targeted delivery pathways of cardiomyocytes ensure that L-type Ca2+ channels (CaV1.2) are concentrated on the t-tubule sarcolemma within nanometers of their intracellular partners the type 2 ryanodine receptors (RyR2) which cluster on the junctional sarcoplasmic reticulum (jSR). The organization and distribution of these two groups of cardiac calcium channel clusters critically underlies the uniform contraction of the myocardium. Ca2+ signaling between these two sets of adjacent clusters produces Ca2+ sparks that in health, cannot escalate into Ca2+ waves because there is sufficient separation of adjacent clusters so that the release of Ca2+ from one RyR2 cluster or supercluster, cannot activate and sustain the release of Ca2+ from neighboring clusters. Instead, thousands of these Ca2+ release units (CRUs) generate near simultaneous Ca2+ sparks across every cardiomyocyte during the action potential when calcium induced calcium release from RyR2 is stimulated by depolarization induced Ca2+ influx through voltage dependent CaV1.2 channel clusters. These sparks summate to generate a global Ca2+ transient that activates the myofilaments and thus the electrical signal of the action potential is transduced into a functional output, myocardial contraction. To generate more, or less contractile force to match the hemodynamic and metabolic demands of the body, the heart responds to β-adrenergic signaling by altering activity of calcium channels to tune excitation-contraction coupling accordingly. Recent accumulating evidence suggests that this tuning process also involves altered expression, and dynamic reorganization of CaV1.2 and RyR2 channels on their respective membranes to control the amplitude of Ca2+ entry, SR Ca2+ release and myocardial function. In heart failure and aging, altered distribution and reorganization of these key Ca2+ signaling proteins occurs alongside architectural remodeling and is thought to contribute to impaired contractile function. In the present review we discuss these latest developments, their implications, and future questions to be addressed.
Collapse
Affiliation(s)
- Rose E Dixon
- Department of Physiology and Membrane Biology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
14
|
OUP accepted manuscript. Cardiovasc Res 2022; 118:932-933. [DOI: 10.1093/cvr/cvac017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
15
|
Castillo M. Thoracic Anesthesia for the Geriatric Patient. COHEN'S COMPREHENSIVE THORACIC ANESTHESIA 2022:544-556. [DOI: 10.1016/b978-0-323-71301-6.00038-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
16
|
Rajabi M, Vafaee MS, Hosseini L, Badalzadeh R. Pretreatment with Nicotinamide Mononucleotide Increases the Effect of Ischemic-Postconditioning on Cardioprotection and Mitochondrial Function Following ex vivo Myocardial Reperfusion Injury in Aged Rats. Clin Exp Pharmacol Physiol 2021; 49:474-482. [PMID: 34854121 DOI: 10.1111/1440-1681.13616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 11/11/2021] [Accepted: 11/20/2021] [Indexed: 11/29/2022]
Abstract
The present study aims to evaluate the combined effect of ischemic-postconditioning (IPostC) and nicotinamide mononucleotide (NMN) on cardioprotection and mitochondrial function in aged rats subjected to myocardial ischemia-reperfusion (IR) injury. Sixty aged Wistar rats were randomly divided into 5 groups (n=12), including sham, control, NMN, IPostC, and NMN+IPostC. Regional ischemia was induced by 30-min occlusion of the left anterior descending coronary artery (LAD) followed by 60-min reperfusion. IPostC was applied at the onset of reperfusion, by 6 cycles of 10-s reperfusion/ischemia. NMN (100 mg/kg) was intraperitoneally injected every other day for 28 days before IR. Myocardial hemodynamics and infarct size (IS) were measured, and the left ventricles samples were harvested to assess cardiac mitochondrial function. The results showed that all treatments reduced lactate dehydrogenase release compared to those of the control group. IPostC alone failed to reduce IS and myocardial function. However, NMN and combined therapy could significantly improve myocardial function and decrease the IS compared to the control animals. Moreover, the effects of combined therapy on the decrease of IS, mitochondrial reactive oxygen species (ROS), and improvement of mitochondrial membrane potential (MMP) were greater than those of alone treatments. These results demonstrated that cardioprotection by combined therapy with NMN+IPostC was superior to individual treatments, and pretreatment of aged rats with NMN was able to correct the failure of IPostC in protecting the hearts of aged rats against IR injury.
Collapse
Affiliation(s)
- Mojgan Rajabi
- Aging Research Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Manouchehr S Vafaee
- Psychiatry Research Unit, Southern Denmark Region, Odense, Denmark.,Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Nuclear Medicine, Odense University Hospital, Odense, Denmark
| | - Leila Hosseini
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Reza Badalzadeh
- Molecular Medicine Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Physiology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
17
|
Koutouroushis C, Sarkar O. Role of Autophagy in Cardiovascular Disease and Aging. Cureus 2021; 13:e20042. [PMID: 34873555 PMCID: PMC8631374 DOI: 10.7759/cureus.20042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/29/2021] [Indexed: 11/15/2022] Open
Abstract
Cardiovascular disease is the leading cause of death worldwide and is expected to further increase as people continue to live even longer. Although the life span of the general population is increasing, the con of such a prolonged life span is that aging has certain detrimental effects on the molecular, structural, and functional elements of the cardiovascular system. This review will discuss various molecular pathways linked to longevity, most notably autophagy and its associated mechanisms, and how these pathways can be targeted to promote cardiovascular health through the process of aging. It is to be noted that the process of autophagy decreases with aging; hence, this review concludes that the promotion of autophagy, through implementation of caloric restriction, intermittent fasting, and pharmacologic agents, has proven to be an efficacious means of stimulating cardiovascular health. Therefore, autophagy is an important target for prevention and procrastination of cardiovascular pathologies in the geriatric population.
Collapse
Affiliation(s)
| | - Oiendrila Sarkar
- General Internal Medicine, St. Mary's Hospital, Isle of Wight NHS Trust, Newport, GBR
| |
Collapse
|
18
|
Zhang H, Li L, Hao M, Chen K, Lu Y, Qi J, Chen W, Ren L, Cai X, Chen C, Liu Z, Zhao B, Li Z, Hou P. Yixin-Fumai granules improve sick sinus syndrome in aging mice through Nrf-2/HO-1 pathway: A new target for sick sinus syndrome. JOURNAL OF ETHNOPHARMACOLOGY 2021; 277:114254. [PMID: 34062246 DOI: 10.1016/j.jep.2021.114254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/13/2021] [Accepted: 05/25/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Yixin-Fumai granules (YXFMs)-composed of Ginseng quinquefolium (L.) Alph. Wood, Ophiopogon japonicus (Thunb.) Ker Gawl, Schisandra arisanensis Hayata, Astragalus aaronsohnianus Eig, Salvia cryptantha Montbret & Aucher ex Benth, and Ligusticum striatum DC-are compound granules used in traditional Chinese medicine to increase heart rate and thus treat bradyarrhythmia. It may be effective in treating sick sinus syndrome (SSS). AIM To observe the effect of YXFMs on aging-induced SSS in mice and explore whether this effect is related to the Nrf-2/HO-1 signaling pathway. MATERIALS AND METHODS Mice with a significant decrease in the heart rate due to natural aging were selected to construct an SSS model. After the mice were administered YXFMs, the damage to their sinoartrial node (SAN) was assessed through electrocardiography, Masson's trichrome staining, and terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL). Dihydroethidium staining and immunofluorescence staining were used to assay reactive oxygen species (ROS) content and HCN4, respectively. Moreover, to observe the effects of YXFMs in vitro, the HL-1 cell line, derived from mouse atrial myocytes, was used to simulate SAN pacemaker cells, with H2O2 used as the cellular oxidative stress (OS) inducer. 2,7-Dichlorodihydrofluorescein diacetate staining was used to assay ROS content, whereas immunofluorescence staining and Western blotting were used to elucidate the related protein expression. Finally, mice were injected the Nrf-2 inhibitor ML385 to reversely verify the effects of YXFMs. RESULTS In our in vivo experiments, YXFMs significantly inhibited aging-induced SSS, shortened the R-R interval, increased heart rate, alleviated fibrosis, reduced apoptosis rate and ROS content, and promote HCN4 expression in the SAN. In our in vitro experiments, YXFMs significantly inhibited H2O2-induced cell peroxidation damage, promoted Nrf-2 activation and nuclear metastasis, increased HO-1 expression- thereby inhibiting ROS accumulation-and finally, upregulated HCN4 expression through the inhibition of histone deacetylase 4 (HDAC4) expression and its nuclear metastasis. Finally, injection of the Nrf-2 inhibitor ML385 after YXFMs administration inhibited their protective effect in the mice. CONCLUSION Here, we elaborated on the relationship between aging-induced SSS and the Nrf-2/HO-1 pathway for the first time and proposed that YXFMs improve SSS via the Nrf-2/HO-1 axis. Specifically, YXFMs promoted Nrf-2 activation and plasma-nuclear transfer to enhance HO-1 expression via the Nrf-2/HO-1 axis. This inhibited OS and reduced ROS accumulation in the SAN, and then, through the ROS/HDAC4 axis, reduced HDAC4 expression and plasma-nuclear transfer. Thereby, the OS-induced HCN4 loss in the SAN was inhibited-improving the function of If channel and thus producing SAN protection effect against SSS and improving the heart rate and R-R interval. In the future, we plan to use bioinformatics analysis technology to execute the next step of our research, namely to determine the effect of isolated, purified components of YXFMs in SSS, to increase its efficiency and reduce the toxicity of YXFMs.
Collapse
Affiliation(s)
- Heng Zhang
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Lingkang Li
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Miao Hao
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Keyan Chen
- Department of Laboratory Animal Science, China Medical University, Shenyang, 110000, China
| | - Yongping Lu
- Department of NHC Key Laboratory of Reproductive Health and Medical Genetics, Liaoning Research Institute of Family Planning (The Affiliated Reproductive Hospital of China Medical University), Shenyang, 110000, China
| | - Jing Qi
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Wei Chen
- Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Lu Ren
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China; Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Xintong Cai
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Chen Chen
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Zhuang Liu
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Bin Zhao
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Zhishuang Li
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China
| | - Ping Hou
- Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China; Department of Cardiology, Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110000, China.
| |
Collapse
|
19
|
"Accelerated aging" of the heart as heart failure with preserved ejection fraction-analysis using leg-positive pressure stress echocardiography. Int J Cardiovasc Imaging 2021; 37:2473-2482. [PMID: 33939071 DOI: 10.1007/s10554-021-02258-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 04/26/2021] [Indexed: 01/14/2023]
Abstract
The aging process is a significant risk factor for heart failure. The incidence of heart failure with preserved ejection fraction (HFpEF) dramatically increases with age. Although HFpEF occurs along a continuum of aging of the cardiovascular system, the pathophysiology that differentiates overt HFpEF from physiological aging is not fully understood. A total of 102 subjects were prospectively recruited: 25 patients with HFpEF and 77 healthy controls. Controls were stratified into three age-groups: young (n = 27, 20-40 years), middle aged (n = 25, 40-65 years), and elderly (n = 25, > 65 years). All participants underwent preload stress echocardiography using a leg-positive pressure (LPP) maneuver. With an increase in age, progressive concentric left ventricular (LV) remodeling was observed in healthy controls, resulting in the hemodynamic consequences of an age-dependent increase in the E/e' ratio (ANOVA, P < 0.001). During LPP stress, the E/e' ratio significantly increased in the middle-aged and elderly groups (from 8 ± 2 to 9 ± 3, from 10 ± 2 to 12 ± 3, P < 0.05, respectively), and this was more pronounced in patients with HFpEF (from 16 ± 5 to 17 ± 7, P < 0.05). Forward stroke volume (SV) significantly increased in each healthy group during LPP stress (all P < 0.001) but failed to increase in the HFpEF group (from 43 ± 13 to 44 ± 14 mL/m2, P = 0.65). In a multivariate analysis, LV mass index (odds ratio [OR] 1.051, P < 0.05), E/e' ratio (OR 1.480; P < 0.05), and change in SV (OR 0.780; P < 0.05) were independent parameters that differentiated HFpEF from physiological aging. Structural remodeling and impaired preload reserve may both be critical features that characterize the pathophysiology of HFpEF.
Collapse
|
20
|
Zhao Q, Liu F, Zhao Q, Zhang J, Luo J, Li X, Yang Y. Constitutive activation of ERK1/2 signaling protects against myocardial ischemia via inhibition of mitochondrial fragmentation in the aging heart. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:479. [PMID: 33850876 PMCID: PMC8039677 DOI: 10.21037/atm-21-503] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background Studies have shown that the ability of the myocardium to tolerate ischemia becomes significantly compromised with age. During ischemia, several endogenous protective signals are activated to protect the heart from injury, among which extracellular-signal regulated kinase (ERK) 1/2 signaling has been established as playing a pivotal role. However, in aging hearts, the activation of ERK1/2 is compromised. Mitogen-activated protein kinase/ERK kinase (MEK) is a major regulator of ERK1/2 signaling. In the present study, we investigated whether transduction of CaMEK, a constitutively activated MEK, using adeno-associated virus serotype 9 (AAV9) could protect the aging heart against ischemia. Methods Myocardial ischemia models were established in aging mice and senescent cardiomyocytes, and AAV9-mediated delivery of CaMEK was applied. Echocardiography, fluorescent staining, transmission electron microscopy, flow cytometry, and immunoblotting were used to explore the effects of CaMEK and their underlying mechanism. Results AAV9-CaMEK activated ERK1/2 signaling and exerted cardioprotective effects against ischemia in aging hearts. Specifically, CaMEK transduction decreased dynamin-related protein-1 (Drp1) expression and phosphorylation at serine 616, resulting in improved mitochondrial morphology and function in aging ischemic hearts. Furthermore, CaMEK transduction exerted similar protective effects in senescent cardiomyocytes under hypoxia. Meanwhile, with the inhibition of ERK1/2 signaling in senescent cardiomyocytes under hypoxia, the opposite effects were observed, including an increase in mitochondrial fragmentation and aggravation of mitochondrial dysfunction and cell apoptosis. Conclusions Our results suggested that AAV9-CaMEK alleviated ischemia-induced myocardium injury in the aging heart, at least in part, through inhibition of mitochondrial fragmentation. Therefore, AAV9-CaMEK is a potential intervention for prevention of ischemia-induced injury of the aging myocardium.
Collapse
Affiliation(s)
- Qiang Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Fen Liu
- Xinjiang Key Laboratory of Cardiovascular Disease Research, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Qian Zhao
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Jinyu Zhang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Junyi Luo
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Xiaomei Li
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Yining Yang
- Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China.,Department of Cardiology, People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, China
| |
Collapse
|
21
|
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: 57] [Impact Index Per Article: 14.3] [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
|
22
|
Greenig M, Melville A, Huntley D, Isalan M, Mielcarek M. Cross-Sectional Transcriptional Analysis of the Aging Murine Heart. Front Mol Biosci 2020; 7:565530. [PMID: 33102519 PMCID: PMC7545256 DOI: 10.3389/fmolb.2020.565530] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Accepted: 08/14/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease accounts for millions of deaths each year and is currently the leading cause of mortality worldwide. The aging process is clearly linked to cardiovascular disease, however, the exact relationship between aging and heart function is not fully understood. Furthermore, a holistic view of cardiac aging, linking features of early life development to changes observed in old age, has not been synthesized. Here, we re-purpose RNA-sequencing data previously-collected by our group, investigating gene expression differences between wild-type mice of different age groups that represent key developmental milestones in the murine lifespan. DESeq2's generalized linear model was applied with two hypothesis testing approaches to identify differentially-expressed (DE) genes, both between pairs of age groups and across mice of all ages. Pairwise comparisons identified genes associated with specific age transitions, while comparisons across all age groups identified a large set of genes associated with the aging process more broadly. An unsupervised machine learning approach was then applied to extract common expression patterns from this set of age-associated genes. Sets of genes with both linear and non-linear expression trajectories were identified, suggesting that aging not only involves the activation of gene expression programs unique to different age groups, but also the re-activation of gene expression programs from earlier ages. Overall, we present a comprehensive transcriptomic analysis of cardiac gene expression patterns across the entirety of the murine lifespan.
Collapse
Affiliation(s)
- Matthew Greenig
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Andrew Melville
- Department of Mathematics, Imperial College London, London, United Kingdom
| | - Derek Huntley
- Department of Life Sciences, Imperial College London, London, United Kingdom
| | - Mark Isalan
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Imperial College Center for Synthetic Biology, Imperial College London, London, United Kingdom
| | - Michal Mielcarek
- Department of Life Sciences, Imperial College London, London, United Kingdom
- Imperial College Center for Synthetic Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
23
|
Vaz Fragoso CA, McAvay GJ. Antihypertensive medications and physical function in older persons. Exp Gerontol 2020; 138:111009. [PMID: 32593771 PMCID: PMC7395796 DOI: 10.1016/j.exger.2020.111009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 05/18/2020] [Accepted: 06/18/2020] [Indexed: 01/08/2023]
Abstract
BACKGROUND To further inform benefits and risks of medications on physical function in aging populations, we have evaluated the associations of antihypertensive (antiHTN) class and number used with skeletal muscle function, mobility, sedentary time, and symptoms in older persons. METHODS Using baseline data from the Lifestyle Interventions and Independence in Elder (LIFE) study (N = 1567, mean age 78.9 years) and multivariable models, we evaluated cross-sectional associations of antiHTN class and number used with physical measures and symptom questionnaires. AntiHTN class included diuretics, angiotensin converting enzyme inhibitors (ACEi), angiotensin receptor blockers (ARB), calcium channel blockers (CCB), and beta blockers (BB). Physical measures included respiratory muscle weakness (maximal inspiratory pressure), grip weakness (dynamometer), impaired lower extremity proximal muscle strength (chair stands), impaired balance (three-stage test), slow gait (400 m walk), mobility impairment (Short Physical Performance Battery), and high sedentary time (accelerometry). Symptoms included dyspnea and fatigue. Covariates included clinical characteristics and non-antiHTNs. RESULTS Use of any antiHTN was highly prevalent (n = 1248 [79.6%]). In the antiHTN subgroup, each antiHTN class was well represented (ranging 36.6%-62.7%) and included use of three or more antiHTNs (32.0%). In adjusted models, the only statistically significant associations were use of BB and three or more antiHTNs with high sedentary time: odds ratios (95% confidence intervals) 1.44 (1.12, 1.85) and 1.52 (1.04, 2.23), respectively. CONCLUSION Use of BB and three or more antiHTNs yielded 44% and 52% increased odds of accelerometry-defined high sedentary time, respectively. Notably, high sedentary time is a risk factor for adverse health outcomes. Thus, future work should evaluate whether high sedentary time mitigates benefits or increases risks, regarding antiHTN use in aging populations.
Collapse
Affiliation(s)
- Carlos A Vaz Fragoso
- VA Connecticut, West Haven, CT, United States of America; Yale School of Medicine, Department of Medicine, New Haven, CT, United States of America.
| | - Gail J McAvay
- Yale School of Medicine, Department of Medicine, New Haven, CT, United States of America
| |
Collapse
|
24
|
Dong M, Yang Z, Fang H, Xiang J, Xu C, Zhou Y, Wu Q, Liu J. Aging Attenuates Cardiac Contractility and Affects Therapeutic Consequences for Myocardial Infarction. Aging Dis 2020; 11:365-376. [PMID: 32257547 PMCID: PMC7069457 DOI: 10.14336/ad.2019.0522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 05/22/2019] [Indexed: 12/21/2022] Open
Abstract
Cardiac function of the human heart changes with age. The age-related change of systolic function is subtle under normal conditions, but abrupt under stress or in a pathogenesis state. Aging decreases the cardiac tolerance to stress and increases susceptibility to ischemia, which caused by aging-induced Ca2+ transient impairment and metabolic dysfunction. The changes of contractility proteins and the relative molecules are in a non-linear fashion. Specifically, the expression and activation of cMLCK increase first then fall during ischemia and reperfusion (I/R). This change is responsible for the nonmonotonic contractility alteration in I/R which the underlying mechanism is still unclear. Contractility recovery in I/R is also attenuated by age. The age-related change in cardiac contractility influences the therapeutic effect and intervention timepoint. For most cardiac ischemia therapies, the therapeutic result in the elderly is not identical to the young. Anti-aging treatment has the potential to prevent the development of ischemic injury and improves cardiac function. In this review we discuss the mechanism underlying the contractility changes in the aged heart and age-induced ischemic injury. The potential mechanism underlying the increased susceptibility to ischemic injury in advanced age is highlighted. Furthermore, we discuss the effect of age and the administration time for intervention in cardiac ischemia therapies.
Collapse
Affiliation(s)
- Ming Dong
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Ziyi Yang
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Hongcheng Fang
- Shenzhen Shajing Hospital, Affiliated of Guangzhou Medical University, Shenzhen, Guangdong, China
| | - Jiaqing Xiang
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Cong Xu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Yanqing Zhou
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Qianying Wu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| | - Jie Liu
- Department of Pathophysiology, Guangdong Key Laboratory of Genome Stability and Human Disease Prevention, Shenzhen University Health Science Center, Guangdong, China
| |
Collapse
|
25
|
Li X, Liu J, Hu H, Lu S, Lu Q, Quan N, Rousselle T, Patel MS, Li J. Dichloroacetate Ameliorates Cardiac Dysfunction Caused by Ischemic Insults Through AMPK Signal Pathway-Not Only Shifts Metabolism. Toxicol Sci 2020; 167:604-617. [PMID: 30371859 DOI: 10.1093/toxsci/kfy272] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dichloroacetate (DCA), an inhibitor of pyruvate dehydrogenase kinase (PDK), regulates substrate metabolism in the heart. AMP-activated protein kinase (AMPK) is an age-related energy sensor that protects the heart from ischemic injury. This study aims to investigate whether DCA can protect the heart from ischemic injury through the AMPK signaling pathway. Young (3-4 months) and aged (20-24 months) male C57BL/6J mice were subjected to ligation of the left anterior descending coronary artery (LAD) for an in vivo ischemic model. The systolic function of the hearts was significantly decreased in both young and aged mice after 45 min of ischemia and 24 h of reperfusion. DCA treatment significantly improved cardiac function in both young and aged mice. The myocardial infarction analysis demonstrated that DCA treatment significantly reduced the infarction size caused by ischemia/reperfusion (I/R) in both young and aged mice. The isolated-cardiomyocyte experiments showed that DCA treatment ameliorated contractile dysfunction and improved the intracellular calcium signal of cardiomyocytes under hypoxia/reoxygenation (H/R) conditions. These cardioprotective functions of DCA can be attenuated by inhibiting AMPK activation. Furthermore, the metabolic measurements with an ex vivo working heart system demonstrated that the effects of DCA treatment on modulating the metabolic shift response to ischemia and reperfusion stress can be attenuated by inhibiting AMPK activity. The immunoblotting results showed that DCA treatment triggered cardiac AMPK signaling pathway by increasing the phosphorylation of AMPK's upstream kinase liver kinase B1 (LKB1) under both sham operations and I/R conditions. Thus, except from modulating metabolism in hearts, the cardioprotective function of DCA during I/R was mediated by the LKB1-AMPK pathway.
Collapse
Affiliation(s)
- Xuan Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Jia Liu
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216.,Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
| | - Haiyan Hu
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Shaoxin Lu
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Qingguo Lu
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Nanhu Quan
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216.,Department of Geriatrics, The First Hospital of Jilin University, Changchun 130021, China
| | - Thomas Rousselle
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| | - Mulchand S Patel
- Department of Biochemistry, University at Buffalo, The State University of New York, Buffalo New York 14203
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi 39216
| |
Collapse
|
26
|
Dokuchaev A, Khamzin S, Solovyova O. In-silico study of age-related ionic remodeling in human ventricular cardiomyocytes. BIO WEB OF CONFERENCES 2020. [DOI: 10.1051/bioconf/20202201024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Ageing is one of the dominant risk factors for cardiovascular diseases. A large number of experimental data is collected on the cellular remodeling in the ageing myocardium from mammals, but very little is known about the human cardiomyocytes. We used a combined electro-mechanical model of human ventricular cardiomyocytes and a population of models approach to investigate the variability in the response of cardiomyocytes to age-related changes in model parameters of the ionic currents.
To generate a control model population, we varied 9 ionic parameters and excluded model samples with biomarkers of cellular action potential (AP) and Ca2+ transient (CT) falling outside the physiological ranges. Using the control population of models, we evaluated the response to age-related reduction in the K+ transient outward current, SERCA pump, and an increase in the Na+Ca2+ exchange current and L-type Ca2+ current. Then, we randomly generated 60 age-related sets of the 4 parameters and applied each set to every model in the control population. We showed an increase in the frequency of repolarization anomalies (RA) and critical AP prolongation in the ageing model populations suggesting arrhythmogenic effects of the ionic remodeling. The population based approach allowed us to assess the pro-arrhythmic contribution of the ionic parameters in ageing cardiomyocytes.
Collapse
|
27
|
Salcan S, Bongardt S, Monteiro Barbosa D, Efimov IR, Rassaf T, Krüger M, Kötter S. Elastic titin properties and protein quality control in the aging heart. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2019; 1867:118532. [PMID: 31421188 DOI: 10.1016/j.bbamcr.2019.118532] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 07/12/2019] [Accepted: 08/12/2019] [Indexed: 01/09/2023]
Abstract
Cardiac aging affects the heart on the functional, structural, and molecular level and shares characteristic hallmarks with the development of chronic heart failure. Apart from age-dependent left ventricular hypertrophy and fibrosis that impairs diastolic function, diminished activity of cardiac protein-quality-control systems increases the risk of cytotoxic accumulation of defective proteins. Here, we studied the impact of cardiac aging on the sarcomeric protein titin by analyzing titin-based cardiomyocyte passive tension, titin modification and proteasomal titin turnover. We analyzed left ventricular samples from young (6 months) and old (20 months) wild-type mice and healthy human donor patients grouped according to age in young (17-50 years) and aged hearts (51-73 years). We found no age-dependent differences in titin isoform composition of mouse or human hearts. In aged hearts from mice and human we determined altered titin phosphorylation at serine residues S4010 and S4099 in the elastic N2B domain, but no significant changes in phosphorylation of S11878 and S12022 in the elastic PEVK region. Importantly, overall titin-based cardiomyocyte passive tension remained unchanged. In aged hearts, the calcium-activated protease calpain-1, which provides accessibility to ubiquitination by releasing titin from the sarcomere, showed decreased proteolytic activity. In addition, we observed a reduction in the proteasomal activities. Taken together, our data indicate that cardiac aging does not affect titin-based passive properties of the cardiomyocytes, but impairs protein-quality control, including titin, which may result in a diminished adaptive capacity of the aged myocardium.
Collapse
Affiliation(s)
- Senem Salcan
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Sabine Bongardt
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - David Monteiro Barbosa
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany
| | - Igor R Efimov
- George Washington University, Department of Biomedical Engineering, Science and Engineering Hall, Washington DC-20052, USA
| | - Tienush Rassaf
- University Hospital Essen, Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, 45147 Essen, Germany
| | - Martina Krüger
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany.
| | - Sebastian Kötter
- Department of Cardiovascular Physiology, Medical Faculty, Heinrich Heine-University Düsseldorf, D-40225 Düsseldorf, Germany.
| |
Collapse
|
28
|
Blice-Baum AC, Guida MC, Hartley PS, Adams PD, Bodmer R, Cammarato A. As time flies by: Investigating cardiac aging in the short-lived Drosophila model. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1831-1844. [PMID: 30496794 PMCID: PMC6527462 DOI: 10.1016/j.bbadis.2018.11.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 11/05/2018] [Accepted: 11/13/2018] [Indexed: 02/06/2023]
Abstract
Aging is associated with a decline in heart function across the tissue, cellular, and molecular levels. The risk of cardiovascular disease grows significantly over time, and as developed countries continue to see an increase in lifespan, the cost of cardiovascular healthcare for the elderly will undoubtedly rise. The molecular basis for cardiac function deterioration with age is multifaceted and not entirely clear, and there is a limit to what investigations can be performed on human subjects or mammalian models. Drosophila melanogaster has emerged as a useful model organism for studying aging in a short timeframe, benefitting from a suite of molecular and genetic tools and displaying highly conserved traits of cardiac senescence. Here, we discuss recent advances in our understanding of cardiac aging and how the fruit fly has aided in these developments.
Collapse
Affiliation(s)
| | - Maria Clara Guida
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Paul S Hartley
- Bournemouth University, Department of Life and Environmental Science, Talbot Campus, Fern Barrow, Poole, Dorset BH12 5BB, UK.
| | - Peter D Adams
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Rolf Bodmer
- Development, Aging and Regeneration Program, Sanford-Burnham-Prebys Medical Discovery Institute, La Jolla, CA, USA.
| | - Anthony Cammarato
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
29
|
Alterations of protein expression of phospholamban, ZASP and plakoglobin in human atria in subgroups of seniors. Sci Rep 2019; 9:5610. [PMID: 30948763 PMCID: PMC6449388 DOI: 10.1038/s41598-019-42141-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 03/22/2019] [Indexed: 01/20/2023] Open
Abstract
The mature mammalian myocardium contains composite junctions (areae compositae) that comprise proteins of adherens junctions as well as desmosomes. Mutations or deficiency of many of these proteins are linked to heart failure and/or arrhythmogenic cardiomyopathy in patients. We firstly wanted to address the question whether the expression of these proteins shows an age-dependent alteration in the atrium of the human heart. Right atrial biopsies, obtained from patients undergoing routine bypass surgery for coronary heart disease were subjected to immunohistology and/or western blotting for the plaque proteins plakoglobin (γ-catenin) and plakophilin 2. Moreover, the Z-band protein cypher 1 (Cypher/ZASP) and calcium handling proteins of the sarcoplasmic reticulum (SR) like phospholamban, SERCA and calsequestrin were analyzed. We noted expression of plakoglobin, plakophilin 2 and Cypher/ZASP in these atrial preparations on western blotting and/or immunohistochemistry. There was an increase of Cypher/ZASP expression with age. The present data extend our knowledge on the expression of anchoring proteins and SR regulatory proteins in the atrium of the human heart and indicate an age-dependent variation in protein expression. It is tempting to speculate that increased expression of Cypher/ZASP may contribute to mechanical changes in the aging human myocardium.
Collapse
|
30
|
Song M, Belmonte JCI, Liu GH. Age-related cardiopathies gene editing. Aging (Albany NY) 2019; 11:1327-1328. [PMID: 30853663 PMCID: PMC6428092 DOI: 10.18632/aging.101853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 03/05/2019] [Indexed: 11/30/2022]
Affiliation(s)
- Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Institute for Stem cell and Regeneration, CAS, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | | | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China.,Institute for Stem cell and Regeneration, CAS, Beijing 100101, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.,National Clinical Research Center for Geriatric Disorders, Advanced Innovation Center for Human Brain Protection, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| |
Collapse
|
31
|
Francis Stuart SD, Wang L, Woodard WR, Ng GA, Habecker BA, Ripplinger CM. Age-related changes in cardiac electrophysiology and calcium handling in response to sympathetic nerve stimulation. J Physiol 2018; 596:3977-3991. [PMID: 29938794 DOI: 10.1113/jp276396] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 06/22/2018] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Ageing results in changes to cardiac electrophysiology, Ca2+ handling, and β-adrenergic responsiveness. Sympathetic neurodegeneration also occurs with age, yet detailed action potential and Ca2+ handling responses to physiological sympathetic nerve stimulation (SNS) in the aged heart have not been assessed. Optical mapping in mouse hearts with intact sympathetic innervation revealed reduced responsiveness to SNS in the aged atria (assessed by heart rate) and aged ventricles (assessed by action potentials and Ca2+ transients). Sympathetic nerve density and noradrenaline content were reduced in aged ventricles, but noradrenaline content was preserved in aged atria. These results demonstrate that reduced responsiveness to SNS in the atria may be primarily due to decreased β-adrenergic receptor responsiveness, whereas reduced responsiveness to SNS in the ventricles may be primarily due to neurodegeneration. ABSTRACT The objective of this study was to determine how age-related changes in sympathetic structure and function impact cardiac electrophysiology and intracellular Ca2+ handling. Innervated hearts from young (3-4 months, YWT, n = 10) and aged (20-24 months, AGED, n = 11) female mice (C57Bl6) were optically mapped using the voltage (Vm ,)- and calcium (Ca2+ )-sensitive indicators Rh237 and Rhod2-AM. Sympathetic nerve stimulation (SNS) was performed at the spinal cord (T1-T3). β-Adrenergic responsiveness was assessed with isoproterenol (1 μM, ISO). Sympathetic nerve density and noradrenaline content were also quantified. Stimulation thresholds necessary to produce a defined increase in heart rate (HR) with SNS were higher in AGED vs. YWT hearts (5.4 ± 0.4 vs. 3.8 ± 0.4 Hz, P < 0.05). Maximal HR with SNS was lower in AGED vs. YWT (20.5 ± 3.41% vs. 73.0 ± 7.63% increase, P < 0.05). β-Adrenergic responsiveness of the atria (measured as percentage increase in HR with ISO) was decreased in AGED vs. YWT hearts (75.3 ± 22.5% vs. 148.5 ± 19.8%, P < 0.05). SNS significantly increased action potential duration (APD) in YWT but not AGED. Ca2+ transient durations and rise times were unchanged by SNS, yet AGED hearts had an increased susceptibility to Ca2+ alternans and ventricular arrhythmias. β-Adrenergic responsiveness of all ventricular parameters were similar between AGED and YWT. Sympathetic nerve density and noradrenaline content were decreased in the AGED ventricle, but not atria, compared to YWT. These data suggest that decreased responsiveness to SNS in the aged atria may be primarily due to decreased β-adrenergic responsiveness, whereas decreased responsiveness to SNS in the aged ventricles may be primarily due to nerve degeneration.
Collapse
Affiliation(s)
| | - Lianguo Wang
- Department of Pharmacology, University of California Davis, Davis, CA, USA
| | - William R Woodard
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA
| | - G Andre Ng
- Department of Cardiovascular Sciences, University of Leicester, NIHR Leicester Biomedical Research Centre, Leicester, UK
| | - Beth A Habecker
- Department of Physiology and Pharmacology, Oregon Health and Science University, Portland, OR, USA
| | | |
Collapse
|
32
|
Lam A, Karekar P, Shah K, Hariharan G, Fleyshman M, Kaur H, Singh H, Gururaja Rao S. Drosophila Voltage-Gated Calcium Channel α1-Subunits Regulate Cardiac Function in the Aging Heart. Sci Rep 2018; 8:6910. [PMID: 29720608 PMCID: PMC5932002 DOI: 10.1038/s41598-018-25195-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 04/17/2018] [Indexed: 12/20/2022] Open
Abstract
Ion channels maintain numerous physiological functions and regulate signaling pathways. They are the key targets for cellular reactive oxygen species (ROS), acting as signaling switches between ROS and ionic homeostasis. We have carried out a paraquat (PQ) screen in Drosophila to identify ion channels regulating the ROS handling and survival in Drosophila melanogaster. Our screen has revealed that α1-subunits (D-type, T-type, and cacophony) of voltage-gated calcium channels (VGCCs) handle PQ-mediated ROS stress differentially in a gender-based manner. Since ROS are also involved in determining the lifespan, we discovered that the absence of T-type and cacophony decreased the lifespan while the absence of D-type maintained a similar lifespan to that of the wild-type strain. VGCCs are also responsible for electrical signaling in cardiac cells. The cardiac function of each mutant was evaluated through optical coherence tomography (OCT), which revealed that α1-subunits of VGCCs are essential in maintaining cardiac rhythmicity and cardiac function in an age-dependent manner. Our results establish specific roles of α1-subunits of VGCCs in the functioning of the aging heart.
Collapse
Affiliation(s)
- Alexander Lam
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Priyanka Karekar
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Kajol Shah
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Girija Hariharan
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Michelle Fleyshman
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Harmehak Kaur
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - Harpreet Singh
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA. .,Division of Cardiology, Department of Medicine, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| | - Shubha Gururaja Rao
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
| |
Collapse
|
33
|
Inoue H, Yamashita T, Akao M, Atarashi H, Ikeda T, Okumura K, Koretsune Y, Shimizu W, Tsutsui H, Toyoda K, Hirayama A, Yasaka M, Yamaguchi T, Akishita M, Hasebe N, Kario K, Mizokami Y, Nagata K, Nakamura M, Terauchi Y, Yamamoto T, Teramukai S, Kimura T, Kaburagi J, Takita A. Prospective observational study in elderly patients with non-valvular atrial fibrillation: Rationale and design of the All Nippon AF In the Elderly (ANAFIE) Registry. J Cardiol 2018; 72:300-306. [PMID: 29625717 DOI: 10.1016/j.jjcc.2018.02.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 02/16/2018] [Accepted: 02/26/2018] [Indexed: 01/16/2023]
Abstract
BACKGROUND Although anticoagulation effectively prevents stroke in patients with atrial fibrillation (AF), it has been underused in elderly AF patients for many reasons, mainly because of knowledge gaps regarding cardiovascular treatment of these populations with multiple comorbidities and poor prognosis. The objectives of the All Nippon AF In the Elderly (ANAFIE) Registry are to collect real-world information about the clinical status of patients with non-valvular AF (NVAF) aged ≥75 years, current status of anticoagulant therapy, and prognosis with/without anticoagulation to establish a database for this specific patient population that is increasing remarkably worldwide. METHODS AND DESIGN The ANAFIE Registry is an observational, multicenter, prospective study of Japanese patients with NVAF aged ≥75 years that will include 30,000 patients and have the primary endpoint of composite of stroke and systemic embolism over a 2-year follow-up period. In parallel with the main study, seven sub-cohort studies will be conducted with assessments including coagulation-fibrinolysis markers, echocardiography, heart rate, hypertension, cognitive function, frailty, and medication adherence. Subgroup analyses will be performed, and stratified by renal function, HbA1c, and maximum number of drugs used. The study was started in October 2016, with a planned 2-year recruitment period. As of January 31, 2018, 33,213 patients were enrolled; the recruitment was therefore ended 8 months earlier than the original plan. CONCLUSIONS The ANAFIE Registry will provide a valuable database for the clinical status, management, and outcomes of mortality, stroke, systemic embolism, and hemorrhagic events with/without anticoagulation in the increasing population of elderly NVAF patients, and will identify risk factors associated with these clinical events.
Collapse
Affiliation(s)
| | | | - Masaharu Akao
- Department of Cardiology, National Hospital Organization Kyoto Medical Center, Kyoto, Japan
| | | | - Takanori Ikeda
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, Tokyo, Japan
| | - Ken Okumura
- Division of Cardiology, Saiseikai Kumamoto Hospital Cardiovascular Center, Kumamoto, Japan
| | - Yukihiro Koretsune
- Institute for Clinical Research, National Hospital Organization Osaka National Hospital, Osaka, Japan
| | - Wataru Shimizu
- Division of Cardiology, Nippon Medical School Department of Medicine, Tokyo, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Science, Fukuoka, Japan
| | - Kazunori Toyoda
- Department of Cerebrovascular Medicine, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Atsushi Hirayama
- Department of Cardiology, Nihon University Itabashi Hospital, Tokyo, Japan
| | - Masahiro Yasaka
- Department of Cerebrovascular Medicine and Neurology, Cerebrovascular Center, National Hospital Organization Kyushu Medical Center, Fukuoka, Japan
| | | | - Masahiro Akishita
- Department of Geriatric Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Naoyuki Hasebe
- First Department of Internal Medicine, Asahikawa Medical College, Hokkaido, Japan
| | - Kazuomi Kario
- Division of Cardiovascular Medicine, Department of Medicine, Jichi Medical University School of Medicine, Tochigi, Japan
| | - Yuji Mizokami
- Division of Gastroenterology, University of Tsukuba Hospital, Ibaraki, Japan
| | - Ken Nagata
- Clinical Research Institute, Yokohama General Hospital, Kanagawa, Japan
| | - Masato Nakamura
- Department of Cardiovascular Medicine, Ohashi Hospital Medical Center, Toho University, Tokyo, Japan
| | - Yasuo Terauchi
- Department of Endocrinology and Metabolism, Graduate School of Medicine, Yokohama City University, Kanagawa, Japan
| | - Takatsugu Yamamoto
- Department of Internal Medicine, Teikyo University School of Medicine, Tokyo, Japan
| | - Satoshi Teramukai
- Department of Biostatistics, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tetsuya Kimura
- Medical Science Department, Daiichi Sankyo, Tokyo, Japan
| | | | - Atsushi Takita
- Safety and Risk Management Department, Daiichi Sankyo, Tokyo, Japan
| |
Collapse
|
34
|
Izzo C, Carrizzo A, Alfano A, Virtuoso N, Capunzo M, Calabrese M, De Simone E, Sciarretta S, Frati G, Oliveti M, Damato A, Ambrosio M, De Caro F, Remondelli P, Vecchione C. The Impact of Aging on Cardio and Cerebrovascular Diseases. Int J Mol Sci 2018; 19:E481. [PMID: 29415476 PMCID: PMC5855703 DOI: 10.3390/ijms19020481] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 01/29/2018] [Accepted: 02/01/2018] [Indexed: 01/03/2023] Open
Abstract
A growing number of evidences report that aging represents the major risk factor for the development of cardio and cerebrovascular diseases. Understanding Aging from a genetic, biochemical and physiological point of view could be helpful to design a better medical approach and to elaborate the best therapeutic strategy to adopt, without neglecting all the risk factors associated with advanced age. Of course, the better way should always be understanding risk-to-benefit ratio, maintenance of independence and reduction of symptoms. Although improvements in treatment of cardiovascular diseases in the elderly population have increased the survival rate, several studies are needed to understand the best management option to improve therapeutic outcomes. The aim of this review is to give a 360° panorama on what goes on in the fragile ecosystem of elderly, why it happens and what we can do, right now, with the tools at our disposal to slow down aging, until new discoveries on aging, cardio and cerebrovascular diseases are at hand.
Collapse
Affiliation(s)
- Carmine Izzo
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Albino Carrizzo
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| | - Antonia Alfano
- Heart Department, A.O.U. “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (A.A.); (E.D.S.)
| | - Nicola Virtuoso
- Department of Cardiovascular Medicine, A.O.U. Federico II, 80131 Naples, Italy;
| | - Mario Capunzo
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Mariaconsiglia Calabrese
- Rehabilitation Department, A.O.U. “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy;
| | - Eros De Simone
- Heart Department, A.O.U. “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy; (A.A.); (E.D.S.)
| | - Sebastiano Sciarretta
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy
| | - Giacomo Frati
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Polo Pontino, 04100 Latina, Italy
| | - Marco Oliveti
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Antonio Damato
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| | - Mariateresa Ambrosio
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| | - Francesco De Caro
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Paolo Remondelli
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
| | - Carmine Vecchione
- Departement of Medicine and Surgery, University of Salerno, 84081 Salerno, Italy; (C.I.); (M.C.); (M.O.); (F.D.C.); (P.R.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy; (A.C.); (S.S.); (G.F.); (A.D.); (M.A.)
| |
Collapse
|
35
|
Sestrin2 prevents age-related intolerance to post myocardial infarction via AMPK/PGC-1α pathway. J Mol Cell Cardiol 2018; 115:170-178. [PMID: 29325933 DOI: 10.1016/j.yjmcc.2018.01.005] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 12/12/2017] [Accepted: 01/07/2018] [Indexed: 12/21/2022]
Abstract
We have revealed that a novel stress-inducible protein, Sestrin2, declines in the heart with aging. Moreover, there is an interaction between Sestrin2 and energy sensor AMPK in the heart in response to ischemic stress. The objective of this study is to determine whether Sestrin2-AMPK complex modulates PGC-1α in the heart and protects the heart from ischemic insults. In order to characterize the role of cardiac Sestrin2-AMPK signaling cascade in aging, C57BL/6 wild type young mice (3-4months), aged mice (24-26months) and young Sestrin2 KO mice were subjected to left anterior descending coronary artery occlusion for in vivo regional ischemia. Intriguingly, ischemic AMPK activation was blunted in aged WT and young Sesn2 KO hearts as compared with young WT hearts. In addition, the AMPK downstream PGC-1α was down-regulated in the aged and Sestrin2 KO hearts during post myocardial infarction. To further determine the regulation of AMPK on mitochondrial functions in aging, the downstream of mitochondrial biogenesis PGC-1α transcriptional factor were measured. The results demonstrated that the PGC-1α downstream effectors TFAM and UCP2 were impaired in the aged and Sestrin2 KO post-MI hearts as compared to the young hearts. While the apoptotic flux markers such as AIF, Bax/Bcl-2 were up-regulated in both aged and Sestrin2 KO hearts versus young hearts. Furthermore, both Sestrin2 KO and aged hearts demonstrated more susceptible to ischemic insults as compared to young hearts. Additionally, the adeno-associated virus (AAV9)-Sestrin2 delivered to the aged hearts via a coronary delivery approach significantly rescued the ischemic tolerance of aged hearts. Taken together, the decreased Sestrin2 levels in aging lead to an impaired AMPK/PGC-1α signaling cascade and an increased sensitivity to ischemic insults.
Collapse
|
36
|
Senatus LM, Schmidt AM. The AGE-RAGE Axis: Implications for Age-Associated Arterial Diseases. Front Genet 2017; 8:187. [PMID: 29259621 PMCID: PMC5723304 DOI: 10.3389/fgene.2017.00187] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 11/10/2017] [Indexed: 12/20/2022] Open
Abstract
The process of advanced glycation leads to the generation and accumulation of an heterogeneous class of molecules called advanced glycation endproducts, or AGEs. AGEs are produced to accelerated degrees in disorders such as diabetes, renal failure, inflammation, neurodegeneration, and in aging. Further, AGEs are present in foods and in tobacco products. Hence, through both endogenous production and exogenous consumption, AGEs perturb vascular homeostasis by a number of means; in the first case, AGEs can cause cross-linking of long-lived molecules in the basement membranes such as collagens, thereby leading to “vascular stiffening” and processes that lead to hyperpermeability and loss of structural integrity. Second, AGEs interaction with their major cell surface signal transduction receptor for AGE or RAGE sets off a cascade of events leading to modulation of gene expression and loss of vascular and tissue homeostasis, processes that contribute to cardiovascular disease. In addition, it has been shown that an enzyme, which plays key roles in the detoxification of pre-AGE species, glyoxalase 1 (GLO1), is reduced in aged and diabetic tissues. In the diabetic kidney devoid of Ager (gene encoding RAGE), higher levels of Glo1 mRNA and GLO1 protein and activity were observed, suggesting that in conditions of high AGE accumulation, natural defenses may be mitigated, at least in part through RAGE. AGEs are a marker of arterial aging and may be detected by both biochemical means, as well as measurement of “skin autofluorescence.” In this review, we will detail the pathobiology of the AGE-RAGE axis and the consequences of its activation in the vasculature and conclude with potential avenues for therapeutic interruption of the AGE-RAGE ligand-RAGE pathways as means to forestall the deleterious consequences of AGE accumulation and signaling via RAGE.
Collapse
Affiliation(s)
- Laura M Senatus
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
37
|
Bakovic M, Filipovic N, Ferhatovic Hamzic L, Kunac N, Zdrilic E, Vitlov Uljevic M, Kostic S, Puljak L, Vukojevic K. Changes in neurofilament 200 and tyrosine hydroxylase expression in the cardiac innervation of diabetic rats during aging. Cardiovasc Pathol 2017; 32:38-43. [PMID: 29175663 DOI: 10.1016/j.carpath.2017.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 11/10/2017] [Accepted: 11/10/2017] [Indexed: 11/19/2022] Open
Abstract
Changes in sensory and sympathetic innervation during diabetes mellitus (DM) can be a predictor of arrhythmias, silent myocardial ischemia, and chronic heart failure, but knowledge about these changes is still unsatisfactory. We analyzed whether prolonged DM induces changes in density of sensory and sympathetic nerve terminals of rat's heart and whether it contributes to cardiomyopathy during aging. DM was induced by i/p injecting 55 mg/kg streptozotocin to male Sprague-Dawley rats, while a control group received a citrate buffer. DM in the rats was validated by measuring blood glucose level. Animals were sacrificed after 2 weeks, 2 months, 6 months, and 12 months. Five areas of cardiac sections were analyzed. Antibodies raised against tyrosine hydroxylase (TH) and neurofilament 200 kDa (NF 200) were used to detect sympathetic and sensory fibers. TH immunoreactive fiber density increased in DM groups 2 weeks after induction, reaching a peek after 2 months, while in the later stages of DM (6 and 12 months), there was no significant difference compared to control. NF 200 immunoreactive fiber density increased 2 weeks after induction compared to control. There was no consistent pattern of change during the given period in both the DM or control groups. In the DM group, we found thickening of the left ventricle wall (P<.05) as the sign of cardiomyopathy. Our findings suggest that hyperglycemia as a hallmark of DM in early stages can lead to proliferation of sympathetic and sensory nerve terminals. This finding can contribute to a better understanding of the occurrence of arrhythmias and silent myocardial ischemia in DM.
Collapse
Affiliation(s)
- Marija Bakovic
- Laboratory for Neurocardiology, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Natalija Filipovic
- Laboratory for Neurocardiology, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Lejla Ferhatovic Hamzic
- Laboratory for Pain Reaserch, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Nenad Kunac
- Department of Pathology, Citology and Forensic medicine, University Hospital Split, Spinciceva 1, 21000 Split, Croatia
| | - Elena Zdrilic
- Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Marija Vitlov Uljevic
- Laboratory for Neurocardiology, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Sandra Kostic
- Laboratory for Microscopy, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Livia Puljak
- Laboratory for Pain Reaserch, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia
| | - Katarina Vukojevic
- Laboratory for Neurocardiology, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; Laboratory for Early Human Development, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia; Laboratory for Microscopy, Department of Anatomy, Histology and Embryology, University of Split School of Medicine, Soltanska 2, 21000 Split, Croatia.
| |
Collapse
|
38
|
Valli H, Ahmad S, Fraser JA, Jeevaratnam K, Huang CLH. Pro-arrhythmic atrial phenotypes in incrementally paced murine Pgc1β -/- hearts: effects of age. Exp Physiol 2017; 102:1619-1634. [PMID: 28960529 PMCID: PMC5725712 DOI: 10.1113/ep086589] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/26/2017] [Indexed: 12/13/2022]
Abstract
New Findings What is the central question of this study? Can we experimentally replicate atrial pro‐arrhythmic phenotypes associated with important chronic clinical conditions, including physical inactivity, obesity, diabetes mellitus and metabolic syndrome, compromising mitochondrial function, and clarify their electrophysiological basis? What is the main finding and its importance? Electrocardiographic and intracellular cardiomyocyte recording at progressively incremented pacing rates demonstrated age‐dependent atrial arrhythmic phenotypes in Langendorff‐perfused murine Pgc1β−/− hearts for the first time. We attributed these to compromised action potential conduction and excitation wavefronts, whilst excluding alterations in recovery properties or temporal electrophysiological instabilities, clarifying these pro‐arrhythmic changes in chronic metabolic disease.
Atrial arrhythmias, most commonly manifesting as atrial fibrillation, represent a major clinical problem. The incidence of atrial fibrillation increases with both age and conditions associated with energetic dysfunction. Atrial arrhythmic phenotypes were compared in young (12–16 week) and aged (>52 week) wild‐type (WT) and peroxisome proliferative activated receptor, gamma, coactivator 1 beta (Ppargc1b)‐deficient (Pgc1β−/−) Langendorff‐perfused hearts, previously used to model mitochondrial energetic disorder. Electrophysiological explorations were performed using simultaneous whole‐heart ECG and intracellular atrial action potential (AP) recordings. Two stimulation protocols were used: an S1S2 protocol, which imposed extrasystolic stimuli at successively decremented intervals following regular pulse trains; and a regular pacing protocol at successively incremented frequencies. Aged Pgc1β−/− hearts showed greater atrial arrhythmogenicity, presenting as atrial tachycardia and ectopic activity. Maximal rates of AP depolarization (dV/dtmax) were reduced in Pgc1β−/− hearts. Action potential latencies were increased by the Pgc1β−/− genotype, with an added interactive effect of age. In contrast, AP durations to 90% recovery (APD90) were shorter in Pgc1β−/− hearts despite similar atrial effective recovery periods amongst the different groups. These findings accompanied paradoxical decreases in the incidence and duration of alternans in the aged and Pgc1β−/− hearts. Limiting slopes of restitution curves of APD90 against diastolic interval were correspondingly reduced interactively by Pgc1β−/− genotype and age. In contrast, reduced AP wavelengths were associated with Pgc1β−/− genotype, both independently and interacting with age, through the basic cycle lengths explored, with the aged Pgc1β−/− hearts showing the shortest wavelengths. These findings thus implicate AP wavelength in possible mechanisms for the atrial arrhythmic changes reported here.
Collapse
Affiliation(s)
- Haseeb Valli
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Shiraz Ahmad
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - James A Fraser
- Physiological Laboratory, University of Cambridge, Cambridge, UK
| | - Kamalan Jeevaratnam
- Physiological Laboratory, University of Cambridge, Cambridge, UK.,PU-RCSI School of Medicine, Perdana University, Serdang, Selangor Darul Ehsan, Malaysia.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, UK
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Cambridge, UK.,Department of Biochemistry, University of Cambridge, Cambridge, UK
| |
Collapse
|
39
|
Barton GP, de Lange WJ, Ralphe JC, Aiken J, Diffee G. Linking metabolic and contractile dysfunction in aged cardiac myocytes. Physiol Rep 2017; 5:5/20/e13485. [PMID: 29084842 PMCID: PMC5661240 DOI: 10.14814/phy2.13485] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 10/02/2017] [Accepted: 10/03/2017] [Indexed: 02/06/2023] Open
Abstract
Aging is associated with declining cardiac contractile function as well as changes in metabolism and mitochondrial function. The relationship between age‐related changes in cardiac metabolism and declining cardiac contractile function has not been determined. In order to define the role energetics play in changes in contractile function, we measured mitochondrial NADH, [NADH]m, during continuous contractions of isolated left ventricular myocytes from young (Y) and old (O) FBN rats. Second, we explored the role of metabolic disruption with rotenone and increased workload with isoproterenol (ISO) had on age‐related changes in myocytes shortening. Single, intact myocytes were stimulated for 10 min of continuous contraction at either 2 Hz or 4 Hz while being perfused with Ringer's solution. Properties of shortening (peak shortening and rate of shortening) were measured at the onset (T0) and after 10 min (T10) of continuous contraction, and the decline in shortening over time (T10/T0) was determined. Although young and old myocytes had similar contractile function under resting conditions, old myocytes demonstrated decrements in [NADH]m during continuous stimulation, while young myocytes maintained constant [NADH]m over this time. In addition, old myocytes exhibited impaired contractile function to a workload (ISO) and metabolic (rotenone) stress compared to young myocytes. Taken together, these results demonstrated that old myocytes are susceptible to stress‐induced contractile dysfunction which may be related to altered cellular energetics.
Collapse
Affiliation(s)
- Gregory P Barton
- Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Willem J de Lange
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin
| | - John C Ralphe
- Department of Pediatrics, University of Wisconsin-Madison, Madison, Wisconsin
| | - Judd Aiken
- Department of Agriculture, Food, and Nutritional Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Gary Diffee
- Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
40
|
Pisano C, Balistreri CR, Ricasoli A, Ruvolo G. Cardiovascular Disease in Ageing: An Overview on Thoracic Aortic Aneurysm as an Emerging Inflammatory Disease. Mediators Inflamm 2017; 2017:1274034. [PMID: 29203969 PMCID: PMC5674506 DOI: 10.1155/2017/1274034] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 09/16/2017] [Accepted: 09/28/2017] [Indexed: 02/07/2023] Open
Abstract
Medial degeneration associated with thoracic aortic aneurysm and acute aortic dissection was originally described by Erdheim as a noninflammatory lesion related to the loss of smooth muscle cells and elastic fibre fragmentation in the media. Recent evidences propose the strong role of a chronic immune/inflammatory process in aneurysm evocation and progression. The coexistence of inflammatory cells with markers of apoptotic vascular cell death in the media of ascending aorta with aneurysms and type A dissections raises the possibility that activated T cells and macrophages may contribute to the elimination of smooth muscle cells and degradation of the matrix. On the other hand, several inflammatory pathways (including TGF-β, TLR-4 interferon-γ, chemokines, and interferon-γ) seem to be involved in the medial degeneration related to aged and dilated aorta. This is an overview on thoracic aortic aneurysm as an emerging inflammatory disease.
Collapse
Affiliation(s)
- Calogera Pisano
- Cardiac Surgery Unit, “P. Giaccone” University Hospital, Palermo, Italy
| | - Carmela Rita Balistreri
- Department of Pathobiology and Medical and Forensic Biotechnologies, University of Palermo, Palermo, Italy
| | | | - Giovanni Ruvolo
- Cardiac Surgery Unit, Tor Vergata University Hospital, Rome, Italy
| |
Collapse
|
41
|
Stöckigt F, Beiert T, Knappe V, Baris OR, Wiesner RJ, Clemen CS, Nickenig G, Andrié RP, Schrickel JW. Aging-related mitochondrial dysfunction facilitates the occurrence of serious arrhythmia after myocardial infarction. Biochem Biophys Res Commun 2017; 493:604-610. [PMID: 28867191 DOI: 10.1016/j.bbrc.2017.08.145] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2017] [Accepted: 08/25/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND During aging a mosaic of normal cells and cells with mitochondrial deficiency develops in various tissues including the heart. Whether this contributes to higher susceptibility for arrhythmia following myocardial infarction (MI) is unknown. METHODS AND RESULTS Myocardial cryoinfarction was performed in 12-month-old transgenic mice with accelerated accumulation of deletions in mitochondrial DNA. Occurrence and pathogenesis of arrhythmia was investigated after two weeks. Holter-ECG recordings revealed higher rates of premature ventricular complexes (incidence > 10/24 h: 100% vs. 20%; p = 0.048) and more severe spontaneous arrhythmia during stress test in mutant mice with MI as compared to control mice with MI. Mice with mitochondrial dysfunction exhibited longer spontaneous AV-blocks (467 ± 26 ms vs. 377 ± 24 ms; p = 0.013), an increased probability for induction of ventricular tachycardia during in vivo electrophysiological investigation (22% vs. 9%; p = 0.044), and a reduced conduction velocity in the infarct borderzone (38.5 ± 0.5 cm/s vs. 55.3 ± 0.9 cm/s; p = 0.001). Furthermore, mutant mice exhibited a significant reduction of the phospho-Cx43/Cx43 ratio in right (0.59 ± 0.04 vs. 0.85 ± 0.01; p = 0.027) and left ventricular myocardium (0.72 ± 0.01 vs. 0.86 ± 0.02; p = 0.023). CONCLUSIONS Aging-related cardiac mosaic respiratory chain dysfunction facilitates the occurrence of spontaneous and inducible cardiac arrhythmia after myocardial infarction and is associated with slowing of electrical impulse propagation in the infarct borderzone.
Collapse
Affiliation(s)
- Florian Stöckigt
- Department of Internal Medicine II, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany.
| | - Thomas Beiert
- Department of Internal Medicine II, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Vincent Knappe
- Department of Internal Medicine II, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Olivier R Baris
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Rudolf J Wiesner
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
| | - Christoph S Clemen
- Center for Biochemistry, Institute of Biochemistry I, Medical Faculty, University of Cologne, Cologne, Germany; Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Georg Nickenig
- Department of Internal Medicine II, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - René P Andrié
- Department of Internal Medicine II, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| | - Jan W Schrickel
- Department of Internal Medicine II, University Hospital Bonn, Rheinische Friedrich-Wilhelms University, Bonn, Germany
| |
Collapse
|
42
|
Quan N, Sun W, Wang L, Chen X, Bogan JS, Zhou X, Cates C, Liu Q, Zheng Y, Li J. Sestrin2 prevents age-related intolerance to ischemia and reperfusion injury by modulating substrate metabolism. FASEB J 2017; 31:4153-4167. [PMID: 28592638 PMCID: PMC5572689 DOI: 10.1096/fj.201700063r] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 05/22/2017] [Indexed: 12/24/2022]
Abstract
A novel stress-inducible protein, Sestrin2 (Sesn2), declines in the heart with aging. AMPK has emerged as a pertinent stress-activated kinase that has been shown to have cardioprotective capabilities against myocardial ischemic injury. We identified the interaction between Sesn2 and AMPK in the ischemic heart. To determine whether ischemic AMPK activation-modulated by the Sesn2-AMPK complex in the heart-is impaired in aging that sensitizes the heart to ischemic insults, young C57BL/6 mice (age 3-4 mo), middle-aged mice (age 10-12 mo), and aged mice (age 24-26 mo) were subjected to left anterior descending coronary artery occlusion for in vivo regional ischemia. The ex vivo working heart system was used for measuring substrate metabolism. The protein level of Sesn2 in hearts was gradually decreased with aging. Of interest, ischemic AMPK activation was blunted in aged hearts compared with young hearts (P < 0.05); the AMPK downstream glucose uptake and the rate of glucose oxidation were significantly impaired in aged hearts during ischemia and reperfusion (P < 0.05 vs. young hearts). Myocardial infarction size was larger in aged hearts (P < 0.05 vs. young hearts). Immunoprecipitation with Sesn2 Ab revealed that cardiac Sesn2 forms a complex with AMPK and upstream liver kinase B1 (LKB1) during ischemia. Of interest, the binding affinity between Sesn2 and AMPK upstream LKB1 is impaired in aged hearts during ischemia (P < 0.05 vs. young hearts). Furthermore, Sesn2-knockout hearts demonstrate a cardiac phenotype and response to ischemic stress that is similar to wild-type aged hearts (i.e., impaired ischemic AMPK activation and higher sensitivity to ischemia- and reperfusion- induced injury). Adeno-associated virus-Sesn2 was delivered to aged hearts via a coronary delivery approach and significantly rescued the protein level of Sesn2 and the ischemic tolerance of aged hearts; therefore, Sesn2 is a scaffold protein that mediates AMPK activation in the ischemic myocardium via an interaction with AMPK upstream LKB1. Decreased Sesn2 levels in aging lead to a blunted ischemic AMPK activation, alterations in substrate metabolism, and an increased sensitivity to ischemic insults-Quan, N., Sun, W., Wang, L., Chen, X., Bogan, J. S., Zhou, X., Cates, C., Liu, Q., Zheng, Y., Li J. Sestrin2 prevents age-related intolerance to ischemia and reperfusion injury by modulating substrate metabolism.
Collapse
Affiliation(s)
- Nanhu Quan
- Cardiovascular Center, First Hospital of Jilin University, Changchun, China
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Wanqing Sun
- Cardiovascular Center, First Hospital of Jilin University, Changchun, China
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Lin Wang
- Cardiovascular Center, First Hospital of Jilin University, Changchun, China
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Xu Chen
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Jonathan S Bogan
- Section of Endocrinology, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA; and
- Department of Cell Biology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Xinchun Zhou
- Department of Pathology, Cancer Institute, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Courtney Cates
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Quan Liu
- Cardiovascular Center, First Hospital of Jilin University, Changchun, China
| | - Yang Zheng
- Cardiovascular Center, First Hospital of Jilin University, Changchun, China;
| | - Ji Li
- Department of Physiology and Biophysics, Mississippi Center for Heart Research, University of Mississippi Medical Center, Jackson, Mississippi, USA;
| |
Collapse
|
43
|
Politano G, Logrand F, Brancaccio M, Di Carlo S. In-silico cardiac aging regulatory model including microRNA post-transcriptional regulation. Methods 2017; 124:57-68. [DOI: 10.1016/j.ymeth.2017.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Revised: 05/26/2017] [Accepted: 06/02/2017] [Indexed: 12/28/2022] Open
|
44
|
Plotnikov EY, Silachev DN, Popkov VA, Zorova LD, Pevzner IB, Zorov SD, Jankauskas SS, Babenko VA, Sukhikh GT, Zorov DB. Intercellular Signalling Cross-Talk: To Kill, To Heal and To Rejuvenate. Heart Lung Circ 2017; 26:648-659. [DOI: 10.1016/j.hlc.2016.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Revised: 11/22/2016] [Accepted: 12/06/2016] [Indexed: 12/16/2022]
|
45
|
Steenman M, Lande G. Cardiac aging and heart disease in humans. Biophys Rev 2017; 9:131-137. [PMID: 28510085 DOI: 10.1007/s12551-017-0255-9] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 03/05/2017] [Indexed: 02/06/2023] Open
Abstract
The world population continues to grow older rapidly, mostly because of declining fertility and increasing longevity. Since age represents the largest risk factor for cardiovascular disease, the prevalence of these pathologies increases dramatically with increasing age. In order to improve patient care and prevention for age-related cardiac diseases, insight should be gained from the analysis of processes involved in and leading to cardiac aging. It is from this perspective that we provide here an overview of changes associated with age in the heart on four levels: functional, structural, cellular and molecular. We highlight those changes that are in common with the development of the two major age-associated cardiac pathologies: heart failure and atrial fibrillation. These commonly affected processes in aging and cardiac pathophysiology may provide an explanation for the age risk factor in cardiac disease.
Collapse
Affiliation(s)
- Marja Steenman
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France.
| | - Gilles Lande
- l'institut du thorax, INSERM, CNRS, UNIV Nantes, CHU Nantes, Nantes, France
| |
Collapse
|
46
|
Ryu T, Song SY. Perioperative management of left ventricular diastolic dysfunction and heart failure: an anesthesiologist's perspective. Korean J Anesthesiol 2017; 70:3-12. [PMID: 28184260 PMCID: PMC5296384 DOI: 10.4097/kjae.2017.70.1.3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/19/2016] [Accepted: 12/19/2016] [Indexed: 12/12/2022] Open
Abstract
Anesthesiologists frequently see asymptomatic patients with diastolic dysfunction or heart failure for various surgeries. These patients typically show normal systolic function but abnormal diastolic parameters in their preoperative echocardiographic evaluations. The symptoms that are sometimes seen are similar to those of chronic obstructive pulmonary disease. Patients with diastolic dysfunction, and even with diastolic heart failure, have the potential to develop a hypertensive crisis or pulmonary congestion. Thus, in addition to conventional perioperative risk quantification, it may be important to consider the results of diastolic assessment for predicting the postoperative outcome and making better decisions. If anesthesiologists see female patients older than 70 years of age who have hypertension, diabetes, chronic renal disease, recent weight gain, or exercise intolerance, they should focus on the patient's diastologic echocardiography indicators such as left atrial enlargement or left ventricular hypertrophy. In addition, there is a need for perioperative strategies to mitigate diastolic dysfunction-related morbidity. Specifically, hypertension should be controlled, keeping pulse pressure below diastolic blood pressure, maintaining a sinus rhythm and normovolemia, and avoiding tachycardia and myocardial ischemia. There is no need to classify these diastolic dysfunction, but it is important to manage this condition to avoid worsening outcomes.
Collapse
Affiliation(s)
- Taeha Ryu
- Department of Anesthesiology and Pain Medicine, School of Medicine, Catholic University of Daegu, Daegu, Korea
| | - Seok-Young Song
- Department of Anesthesiology and Pain Medicine, School of Medicine, Catholic University of Daegu, Daegu, Korea
| |
Collapse
|
47
|
Hsu YJ, Hsu SC, Hsu CP, Chen YH, Chang YL, Sadoshima J, Huang SM, Tsai CS, Lin CY. Sirtuin 1 protects the aging heart from contractile dysfunction mediated through the inhibition of endoplasmic reticulum stress-mediated apoptosis in cardiac-specific Sirtuin 1 knockout mouse model. Int J Cardiol 2016; 228:543-552. [PMID: 27875732 DOI: 10.1016/j.ijcard.2016.11.247] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/08/2016] [Accepted: 11/10/2016] [Indexed: 11/19/2022]
Abstract
BACKGROUND The longevity regulator Sirtuin 1 is an NAD+-dependent histone deacetylase that regulates endoplasmic reticulum stress and influences cardiomyocyte apoptosis during cardiac contractile dysfunction induced by aging. The mechanism underlying Sirtuin 1 function in cardiac contractile dysfunction related to aging has not been completely elucidated. METHODS We evaluated cardiac contractile function, endoplasmic reticulum stress, apoptosis, and oxidative stress in 6- and 12month-old cardiac-specific Sirtuin 1 knockout (Sirt1-/-) and control (Sirt1f/f) mice using western blotting and immunohistochemistry. Mice were injected with a protein disulphide isomerase inhibitor. For in vitro analysis, cultured H9c2 cardiomyocytes were exposed to either a Sirtuin 1 inhibitor or activator, with or without a mitochondrial inhibitor, to evaluate the effects of Sirtuin 1 on endoplasmic reticulum stress, nitric oxide synthase expression, and apoptosis. The effects of protein disulphide isomerase inhibition on oxidative stress and ER stress-related apoptosis were also investigated. RESULTS Compared with 6-month-old Sirt1f/f mice, marked impaired contractility was observed in 12-month-old Sirt1-/- mice. These findings were consistent with increased endoplasmic reticulum stress and apoptosis in the myocardium. Measures of oxidative stress and nitric oxide synthase expression were significantly higher in Sirt1-/- mice compared with those in Sirt1f/f mice at 6months. In vitro experiments revealed increased endoplasmic reticulum stress-mediated apoptosis in H9c2 cardiomyocytes treated with a Sirtuin 1 inhibitor; the effects were ameliorated by a Sirtuin 1 activator. Moreover, consistent with the in vitro findings, impaired cardiac contractility was demonstrated in Sirt1-/- mice injected with a protein disulphide isomerase inhibitor. CONCLUSION The present study demonstrates that the aging heart is characterized by contractile dysfunction associated with increased oxidative stress and endoplasmic reticulum stress and Sirtuin 1 might have the ability to protect the aging hearts from the inhibition of endoplasmic reticulum-mediated apoptosis.
Collapse
Affiliation(s)
- Yu-Juei Hsu
- Division of Nephrology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan; Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Shih-Che Hsu
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chiao-Po Hsu
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yen-Hui Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Yung-Lung Chang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, University of Medicine and Dentistry of New Jersey, New Jersey Medical School, Newark, NJ, USA
| | - Shih-Ming Huang
- Department of Biochemistry, National Defense Medical Center, Taipei, Taiwan
| | - Chien-Sung Tsai
- Superintendent's Office, Taoyuan Armed Forces General Hospital, Taoyuan, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chih-Yuan Lin
- Division of Cardiovascular Surgery, Department of Surgery, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan.
| |
Collapse
|
48
|
Barton GP, Sepe JJ, McKiernan SH, Aiken JM, Diffee GM. Mitochondrial and Metabolic Gene Expression in the Aged Rat Heart. Front Physiol 2016; 7:352. [PMID: 27601998 PMCID: PMC4993773 DOI: 10.3389/fphys.2016.00352] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 08/02/2016] [Indexed: 02/05/2023] Open
Abstract
Aging is associated with a decline in cardiac function. Exercise intervention has been suggested as a way to improve this decrement. Age-related decline in cardiac function is associated with decreases in fatty acid oxidation, mitochondrial function, and AMP-activated protein kinase (AMPK) activity. The molecular mechanisms involved with age-related changes in mitochondrial function and substrate metabolism are poorly understood. We determined gene expression differences in hearts of Young (6 mo), Old (33 mo), and old exercise trained (Old + EXE) (34 mo) FBN rats, using Qiagen PCR arrays for Glucose, Fatty acid, and Mitochondrial metabolism. Old rats demonstrated decreased (p < 0.05) expression for key genes in fatty acid oxidation, mitochondrial function, and AMPK signaling. There were no differences in the expression of genes involved in glucose metabolism with age. These gene expression changes occurred prior to altered protein translation as we found no differences in the protein content of peroxisome proliferator activated receptor gamma, coactivators 1 alpha (PGC-1α), peroxisome proliferator activated receptor alpha (PPARα), and AMPKα2 between young and old hearts. Four months of exercise training did not attenuate the decline in the gene expression in aged hearts. Despite this lack of change in gene expression, exercise-trained rats demonstrated increased exercise capacity compared to their sedentary counterparts. Taken together, our results show that differential expression of genes associated with fatty acid metabolism, AMPK signaling and mitochondrial function decrease in the aging heart which may play a role in age-related declines in fatty acid oxidation, AMPK activity, and mitochondrial function in the heart.
Collapse
Affiliation(s)
- Gregory P Barton
- Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison Madison, WI, USA
| | - Joseph J Sepe
- Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison Madison, WI, USA
| | - Susan H McKiernan
- Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison Madison, WI, USA
| | - Judd M Aiken
- Departments of Agriculture, Food, and Nutritional Sciences, University of Alberta-Edmonton Edmonton, AB, Canada
| | - Gary M Diffee
- Balke Biodynamics Laboratory, Department of Kinesiology, University of Wisconsin-Madison Madison, WI, USA
| |
Collapse
|
49
|
Menting ME, McGhie JS, Koopman LP, Vletter WB, Helbing WA, van den Bosch AE, Roos-Hesselink JW. Normal myocardial strain values using 2D speckle tracking echocardiography in healthy adults aged 20 to 72 years. Echocardiography 2016; 33:1665-1675. [DOI: 10.1111/echo.13323] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Affiliation(s)
| | | | - Laurens P. Koopman
- Department of Pediatrics; Division of Pediatric Cardiology; Erasmus MC - Sophia Children's Hospital; Rotterdam The Netherlands
| | - Wim B. Vletter
- Department of Cardiology; Erasmus MC; Rotterdam The Netherlands
| | - Willem A. Helbing
- Department of Pediatrics; Division of Pediatric Cardiology; Erasmus MC - Sophia Children's Hospital; Rotterdam The Netherlands
| | | | | |
Collapse
|
50
|
Tepp K, Timohhina N, Puurand M, Klepinin A, Chekulayev V, Shevchuk I, Kaambre T. Bioenergetics of the aging heart and skeletal muscles: Modern concepts and controversies. Ageing Res Rev 2016; 28:1-14. [PMID: 27063513 DOI: 10.1016/j.arr.2016.04.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/01/2016] [Accepted: 04/04/2016] [Indexed: 01/03/2023]
Abstract
Age-related alterations in the bioenergetics of the heart and oxidative skeletal muscle tissues are of crucial influence on their performance. Until now the prevailing concept of aging was the mitochondrial theory, the increased production of reactive oxygen species, mediated by deficiency in the activity of respiratory chain complexes. However, studies with mitochondria in situ have presented results which, to some extent, disagree with previous ones, indicating that the mitochondrial theory of aging may be overestimated. The studies reporting age-related decline in mitochondrial function were performed using mainly isolated mitochondria. Measurements on this level are not able to take into account the system level properties. The relevant information can be obtained only from appropriate studies using cells or tissue fibers. The functional interactions between the components of Intracellular Energetic Unit (ICEU) regulate the energy production and consumption in oxidative muscle cells. The alterations of these interactions in ICEU should be studied in order to find a more effective protocol to decelerate the age-related changes taking place in the energy metabolism. In this article, an overview is given of the present theories and controversies of causes of age-related alterations in bioenergetics. Also, branches of study, which need more emphasis, are indicated.
Collapse
Affiliation(s)
- Kersti Tepp
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia.
| | - Natalja Timohhina
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Marju Puurand
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Aleksandr Klepinin
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Vladimir Chekulayev
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Igor Shevchuk
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia
| | - Tuuli Kaambre
- Laboratory of Bioenergetics, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia; Faculty of Science, Tallinn University, Narva mnt. 25, 10120, Estonia
| |
Collapse
|