101
|
Gu SX, Tyagi T, Jain K, Gu VW, Lee SH, Hwa JM, Kwan JM, Krause DS, Lee AI, Halene S, Martin KA, Chun HJ, Hwa J. Thrombocytopathy and endotheliopathy: crucial contributors to COVID-19 thromboinflammation. Nat Rev Cardiol 2021; 18:194-209. [PMID: 33214651 PMCID: PMC7675396 DOI: 10.1038/s41569-020-00469-1] [Citation(s) in RCA: 274] [Impact Index Per Article: 68.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/19/2020] [Indexed: 02/06/2023]
Abstract
The core pathology of coronavirus disease 2019 (COVID-19) is infection of airway cells by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) that results in excessive inflammation and respiratory disease, with cytokine storm and acute respiratory distress syndrome implicated in the most severe cases. Thrombotic complications are a major cause of morbidity and mortality in patients with COVID-19. Patients with pre-existing cardiovascular disease and/or traditional cardiovascular risk factors, including obesity, diabetes mellitus, hypertension and advanced age, are at the highest risk of death from COVID-19. In this Review, we summarize new lines of evidence that point to both platelet and endothelial dysfunction as essential components of COVID-19 pathology and describe the mechanisms that might account for the contribution of cardiovascular risk factors to the most severe outcomes in COVID-19. We highlight the distinct contributions of coagulopathy, thrombocytopathy and endotheliopathy to the pathogenesis of COVID-19 and discuss potential therapeutic strategies in the management of patients with COVD-19. Harnessing the expertise of the biomedical and clinical communities is imperative to expand the available therapeutics beyond anticoagulants and to target both thrombocytopathy and endotheliopathy. Only with such collaborative efforts can we better prepare for further waves and for future coronavirus-related pandemics.
Collapse
Affiliation(s)
- Sean X Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Tarun Tyagi
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Kanika Jain
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Vivian W Gu
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Seung Hee Lee
- Division of Cardiovascular Diseases, Center for Biomedical Sciences, National Institute of Health, Cheongju, Chungbuk, Korea
| | - Jonathan M Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Jennifer M Kwan
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Diane S Krause
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Alfred I Lee
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Comprehensive Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Kathleen A Martin
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Hyung J Chun
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - John Hwa
- Yale Cardiovascular Research Center, Section of Cardiovascular Medicine, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
102
|
Saadeh K, Fazmin IT. Mitochondrial Dysfunction Increases Arrhythmic Triggers and Substrates; Potential Anti-arrhythmic Pharmacological Targets. Front Cardiovasc Med 2021; 8:646932. [PMID: 33659284 PMCID: PMC7917191 DOI: 10.3389/fcvm.2021.646932] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/26/2021] [Indexed: 12/31/2022] Open
Abstract
Incidence of cardiac arrhythmias increases significantly with age. In order to effectively stratify arrhythmic risk in the aging population it is crucial to elucidate the relevant underlying molecular mechanisms. The changes underlying age-related electrophysiological disruption appear to be closely associated with mitochondrial dysfunction. Thus, the present review examines the mechanisms by which age-related mitochondrial dysfunction promotes arrhythmic triggers and substrate. Namely, via alterations in plasmalemmal ionic currents (both sodium and potassium), gap junctions, cellular Ca2+ homeostasis, and cardiac fibrosis. Stratification of patients' mitochondrial function status permits application of appropriate anti-arrhythmic therapies. Here, we discuss novel potential anti-arrhythmic pharmacological interventions that specifically target upstream mitochondrial function and hence ameliorates the need for therapies targeting downstream changes which have constituted traditional antiarrhythmic therapy.
Collapse
Affiliation(s)
- Khalil Saadeh
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom
| | - Ibrahim Talal Fazmin
- School of Clinical Medicine, University of Cambridge, Cambridge, United Kingdom.,Faculty of Health and Medical Sciences, University of Surrey, Guildford, United Kingdom.,Royal Papworth Hospital NHS Foundation Trust, Cambridge, United Kingdom
| |
Collapse
|
103
|
Szabó R, Hoffmann A, Börzsei D, Kupai K, Veszelka M, Berkó AM, Pávó I, Gesztelyi R, Juhász B, Turcsán Z, Pósa A, Varga C. Hormone Replacement Therapy and Aging: A Potential Therapeutic Approach for Age-Related Oxidative Stress and Cardiac Remodeling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8364297. [PMID: 33623635 PMCID: PMC7875635 DOI: 10.1155/2021/8364297] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 08/23/2020] [Accepted: 01/10/2021] [Indexed: 12/13/2022]
Abstract
Advanced age is an independent risk factor for cardiovascular diseases, which might be further exacerbated by estrogen deficiency. Hormone replacement therapy (HRT) decreases cardiovascular risks and events in postmenopausal women; however, its effects are not fully elucidated in older individuals. Thus, the aim of our study is to examine the impact of HRT on oxidant/antioxidant homeostasis and cardiac remodeling. In our experiment, control (fertile) and aging (~20-month-old) female Wistar rats were used. Aging rats were further divided into estrogen- (E2, 0.1 mg/kg/day per os) or raloxifene- (RAL, 1.0 mg/kg/day per os) treated subgroups. After 2 weeks of treatment, cardiac heme oxygenase (HO) activity, total glutathione (GSH) content, matrix metalloproteinase-2 (MMP-2) activity, and the concentrations of collagen type I and tissue inhibitor of metalloproteinase (TIMP-2), as well as the infarct size, were determined. The aging process significantly decreased the antioxidant HO activity and GSH content, altered the MMP-2/TIMP-2 signaling, and resulted in an excessive collagen accumulation, which culminated in cardiovascular injury. However, 2 weeks of either E2 or RAL treatment enhanced the antioxidant defense mechanisms and attenuated cardiac remodeling related to aging. Our findings clearly show that 2-week-long HRT is a potential intervention to bias successful cardiovascular aging via reducing oxidative damage and cardiovascular dysfunction.
Collapse
Affiliation(s)
- Renáta Szabó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- Interdisciplinary Excellence Centre, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Alexandra Hoffmann
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Denise Börzsei
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Krisztina Kupai
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- 1st Department of Medicine, University of Szeged, Szeged H-6720, Hungary
| | - Médea Veszelka
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Anikó Magyariné Berkó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Imre Pávó
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Rudolf Gesztelyi
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen H-4032, Hungary
| | - Béla Juhász
- Department of Pharmacology and Pharmacotherapy, University of Debrecen, Debrecen H-4032, Hungary
| | - Zsolt Turcsán
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| | - Anikó Pósa
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
- Interdisciplinary Excellence Centre, Department of Physiology, Anatomy and Neuroscience, University of Szeged, Szeged, Hungary
| | - Csaba Varga
- Department of Physiology, Anatomy and Neuroscience, Faculty of Science and Informatics, University of Szeged, Szeged H-6726, Hungary
| |
Collapse
|
104
|
Cikic S, Chandra PK, Harman JC, Rutkai I, Katakam PV, Guidry JJ, Gidday JM, Busija DW. Sexual differences in mitochondrial and related proteins in rat cerebral microvessels: A proteomic approach. J Cereb Blood Flow Metab 2021; 41:397-412. [PMID: 32241204 PMCID: PMC8370005 DOI: 10.1177/0271678x20915127] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sex differences in mitochondrial numbers and function are present in large cerebral arteries, but it is unclear whether these differences extend to the microcirculation. We performed an assessment of mitochondria-related proteins in cerebral microvessels (MVs) isolated from young, male and female, Sprague-Dawley rats. MVs composed of arterioles, capillaries, and venules were isolated from the cerebrum and used to perform a 3 versus 3 quantitative, multiplexed proteomics experiment utilizing tandem mass tags (TMT), coupled with liquid chromatography/mass spectrometry (LC/MS). MS data and bioinformatic analyses were performed using Proteome Discoverer version 2.2 and Ingenuity Pathway Analysis. We identified a total of 1969 proteins, of which 1871 were quantified by TMT labels. Sixty-four proteins were expressed significantly (p < 0.05) higher in female samples compared with male samples. Females expressed more mitochondrial proteins involved in energy production, mitochondrial membrane structure, anti-oxidant enzyme proteins, and those involved in fatty acid oxidation. Conversely, males had higher expression levels of mitochondria-destructive proteins. Our findings reveal, for the first time, the full extent of sexual dimorphism in the mitochondrial metabolic protein profiles of MVs, which may contribute to sex-dependent cerebrovascular and neurological pathologies.
Collapse
Affiliation(s)
- Sinisa Cikic
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Partha K Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jarrod C Harman
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA, USA.,Department of Physiology, Louisiana State University Health Science Center, New Orleans, LA, USA.,Neuroscience Center of Excellence, Louisiana State University Health Science Center, New Orleans, LA, USA.,Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Ibolya Rutkai
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.,Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Prasad Vg Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.,Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Jessie J Guidry
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA, USA.,Proteomics Core Facility, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Jeffrey M Gidday
- Department of Ophthalmology, Louisiana State University Health Science Center, New Orleans, LA, USA.,Department of Physiology, Louisiana State University Health Science Center, New Orleans, LA, USA.,Neuroscience Center of Excellence, Louisiana State University Health Science Center, New Orleans, LA, USA.,Department of Biochemistry and Molecular Biology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - David W Busija
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA.,Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| |
Collapse
|
105
|
Liu Y, Li M, Du X, Huang Z, Quan N. Sestrin 2, a potential star of antioxidant stress in cardiovascular diseases. Free Radic Biol Med 2021; 163:56-68. [PMID: 33310138 DOI: 10.1016/j.freeradbiomed.2020.11.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 02/06/2023]
Abstract
Physiological reactive oxygen species (ROS) play an important role in cellular signal transduction. However, excessive ROS is an important pathological mechanism in most cardiovascular diseases (CVDs), such as myocardial aging, cardiomyopathy, ischemia/reperfusion injury (e.g., myocardial infarction) and heart failure. Programmed cell death, hypertrophy and fibrosis may be due to oxidative stress. Sestrin 2 (Sesn2), a stress-inducible protein associated with various stress conditions, is a potential antioxidant. Sesn2 can suppress the process of heart damage caused by oxidative stress, promote cell survival and play a key role in a variety of CVDs. This review discusses the effect of Sesn2 on the redox signal, mainly via participation in the signaling pathway of nuclear factor erythroid 2-related factor 2, activation of adenosine monophosphate-activated protein kinase and inhibition of mammalian target of rapamycin complex 1. It also discusses the effect of Sesn2's antioxidant activity on different CVDs. We speculate that Sesn2 plays an important role in CVDs by stimulating the process of antioxidation and promoting the adaptation of cells to stress conditions and/or the environment, opening a new avenue for related therapeutic strategies.
Collapse
Affiliation(s)
- Yunxia Liu
- Department of Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Meina Li
- Department of Infection Control, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xiaoyu Du
- Department of Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Zhehao Huang
- Department of Neurosurgery, The Third Hospital of Jilin University, Changchun, Jilin, 130031, China.
| | - Nanhu Quan
- Department of Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
106
|
Shen ZQ, Huang YL, Teng YC, Wang TW, Kao CH, Yeh CH, Tsai TF. CISD2 maintains cellular homeostasis. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118954. [PMID: 33422617 DOI: 10.1016/j.bbamcr.2021.118954] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023]
Abstract
CDGSH Iron Sulfur Domain 2 (CISD2) is the causative gene for the disease Wolfram syndrome 2 (WFS2; MIM 604928), which is an autosomal recessive disorder showing metabolic and neurodegenerative manifestations. CISD2 protein can be localized on the endoplasmic reticulum (ER), outer mitochondrial membrane (OMM) and mitochondria-associated membrane (MAM). CISD2 plays a crucial role in the regulation of cytosolic Ca2+ homeostasis, ER integrity and mitochondrial function. Here we summarize the most updated publications and discuss the central role of CISD2 in maintaining cellular homeostasis. This review mainly focuses on the following topics. Firstly, that CISD2 has been recognized as a prolongevity gene and the level of CISD2 is a key determinant of lifespan and healthspan. In mice, Cisd2 deficiency shortens lifespan and accelerates aging. Conversely, a persistently high level of Cisd2 promotes longevity. Intriguingly, exercise stimulates Cisd2 gene expression and thus, the beneficial effects offered by exercise may be partly related to Cisd2 activation. Secondly, that Cisd2 is down-regulated in a variety of tissues and organs during natural aging. Three potential mechanisms that may mediate the age-dependent decrease of Cisd2, via regulating at different levels of gene expression, are discussed. Thirdly, the relationship between CISD2 and cell survival, as well as the potential mechanisms underlying the cell death control, are discussed. Finally we discuss that, in cancers, CISD2 may functions as a double-edged sword, either suppressing or promoting cancer development. This review highlights the importance of the CISD2 in aging and age-related diseases and identifies the urgent need for the translation of available genetic evidence into pharmaceutic interventions in order to alleviate age-related disorders and extend a healthy lifespan in humans.
Collapse
Affiliation(s)
- Zhao-Qing Shen
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Yi-Long Huang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan; Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yuan-Chi Teng
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Tai-Wen Wang
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan, Taiwan
| | - Chi-Hsiao Yeh
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Linko, Taiwan; College of Medicine, Chang Gung University, Taoyuan, Taiwan; Community Medicine Research Center, Chang Gung Memorial Hospital, Keelung Branch, Keelung, Taiwan.
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan; Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan; Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan; Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
107
|
A Plasma Proteomic Signature of Skeletal Muscle Mitochondrial Function. Int J Mol Sci 2020; 21:ijms21249540. [PMID: 33333910 PMCID: PMC7765442 DOI: 10.3390/ijms21249540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/29/2022] Open
Abstract
Although mitochondrial dysfunction has been implicated in aging, physical function decline, and several age-related diseases, an accessible and affordable measure of mitochondrial health is still lacking. In this study we identified the proteomic signature of muscular mitochondrial oxidative capacity in plasma. In 165 adults, we analyzed the association between concentrations of plasma proteins, measured using the SOMAscan assay, and skeletal muscle maximal oxidative phosphorylation capacity assessed as post-exercise phosphocreatine recovery time constant (τPCr) by phosphorous magnetic resonance spectroscopy. Out of 1301 proteins analyzed, we identified 87 proteins significantly associated with τPCr, adjusting for age, sex, and phosphocreatine depletion. Sixty proteins were positively correlated with better oxidative capacity, while 27 proteins were correlated with poorer capacity. Specific clusters of plasma proteins were enriched in the following pathways: homeostasis of energy metabolism, proteostasis, response to oxidative stress, and inflammation. The generalizability of these findings would benefit from replication in an independent cohort and in longitudinal analyses.
Collapse
|
108
|
Florio MC, Magenta A, Beji S, Lakatta EG, Capogrossi MC. Aging, MicroRNAs, and Heart Failure. Curr Probl Cardiol 2020; 45:100406. [PMID: 30704792 PMCID: PMC10544917 DOI: 10.1016/j.cpcardiol.2018.12.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 12/23/2018] [Indexed: 12/12/2022]
Abstract
Aging is a major risk factor for heart failure, one of the leading causes of death in Western society. The mechanisms that underlie the different forms of heart failure have been elucidated only in part and the role of noncoding RNAs is still poorly characterized. Specifically, microRNAs (miRNAs), a class of small noncoding RNAs that can modulate gene expression at the posttranscriptional level in all cells, including myocardial and vascular cells, have been shown to play a role in heart failure with reduced ejection fraction. In contrast, miRNAs role in heart failure with preserved ejection fraction, the predominant form of heart failure in the elderly, is still unknown. In this review, we will focus on age-dependent miRNAs in heart failure and on some other conditions that are prevalent in the elderly and are frequently associated with heart failure with preserved ejection fraction.
Collapse
|
109
|
Martinez PJ, Agudiez M, Molero D, Martin-Lorenzo M, Baldan-Martin M, Santiago-Hernandez A, García-Segura JM, Madruga F, Cabrera M, Calvo E, Ruiz-Hurtado G, Barderas MG, Vivanco F, Ruilope LM, Alvarez-Llamas G. Urinary metabolic signatures reflect cardiovascular risk in the young, middle-aged, and elderly populations. J Mol Med (Berl) 2020; 98:1603-1613. [PMID: 32914213 PMCID: PMC7591416 DOI: 10.1007/s00109-020-01976-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/10/2020] [Accepted: 09/03/2020] [Indexed: 01/09/2023]
Abstract
The predictive value of traditional cardiovascular risk estimators is limited, and young and elderly populations are particularly underrepresented. We aimed to investigate the urine metabolome and its association with cardiovascular risk to identify novel markers that might complement current estimators based on age. Urine samples were collected from 234 subjects categorized into three age-grouped cohorts: 30-50 years (cohort I, young), 50-70 years (cohort II, middle-aged), and > 70 years (cohort III, elderly). Each cohort was further classified into three groups: (a) control, (b) individuals with cardiovascular risk factors, and (c) those who had a previous cardiovascular event. Novel urinary metabolites linked to cardiovascular risk were identified by nuclear magnetic resonance in cohort I and then evaluated by target mass spectrometry quantification in all cohorts. A previously identified metabolic fingerprint associated with atherosclerosis was also analyzed and its potential risk estimation investigated in the three aged cohorts. Three different metabolic signatures were identified according to age: 2-hydroxybutyrate, gamma-aminobutyric acid, hypoxanthine, guanidoacetate, oxaloacetate, and serine in young adults; citrate, cyclohexanol, glutamine, lysine, pantothenate, pipecolate, threonine, and tyramine shared by middle-aged and elderly adults; and trimethylamine N-oxide and glucuronate associated with cardiovascular risk in all three cohorts. The urinary metabolome contains a metabolic signature of cardiovascular risk that differs across age groups. These signatures might serve to complement existing algorithms and improve the accuracy of cardiovascular risk prediction for personalized prevention. KEY MESSAGES: • Cardiovascular risk in the young and elderly is underestimated. • The urinary metabolome reflects cardiovascular risk across all age groups. • Six metabolites constitute a metabolic signature of cardiovascular risk in young adults. • Middle-aged and elderly adults share a cardiovascular risk metabolic signature. • TMAO and glucuronate levels reflect cardiovascular risk across all age groups.
Collapse
Affiliation(s)
- Paula J Martinez
- Department of Immunology, Immunoallergy and Proteomics Laboratory, IIS-Fundación Jiménez Díaz, UAM, Avenida Reyes Católicos 2, 28040, Madrid, Spain
| | - Marta Agudiez
- Department of Immunology, Immunoallergy and Proteomics Laboratory, IIS-Fundación Jiménez Díaz, UAM, Avenida Reyes Católicos 2, 28040, Madrid, Spain
| | - Dolores Molero
- CAI-RMN, Universidad Complutense de Madrid, Madrid, Spain
| | - Marta Martin-Lorenzo
- Department of Immunology, Immunoallergy and Proteomics Laboratory, IIS-Fundación Jiménez Díaz, UAM, Avenida Reyes Católicos 2, 28040, Madrid, Spain
| | | | - Aranzazu Santiago-Hernandez
- Department of Immunology, Immunoallergy and Proteomics Laboratory, IIS-Fundación Jiménez Díaz, UAM, Avenida Reyes Católicos 2, 28040, Madrid, Spain
| | - Juan Manuel García-Segura
- CAI-RMN, Universidad Complutense de Madrid, Madrid, Spain
- Department of Biochemistry and Molecular Biology I, Universidad Complutense, Madrid, Spain
| | - Felipe Madruga
- Departament of Geriatrics, Hospital Virgen del Valle, SESCAM, Toledo, Spain
| | | | | | - Gema Ruiz-Hurtado
- Cardiorenal Translational Laboratory, Instituto de Investigación I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Maria G Barderas
- Department of Vascular Physiopathology, Hospital Nacional de Parapléjicos SESCAM, Toledo, Spain
| | - Fernando Vivanco
- Department of Immunology, Immunoallergy and Proteomics Laboratory, IIS-Fundación Jiménez Díaz, UAM, Avenida Reyes Católicos 2, 28040, Madrid, Spain
- Department of Biochemistry and Molecular Biology I, Universidad Complutense, Madrid, Spain
| | - Luis M Ruilope
- Cardiorenal Translational Laboratory, Instituto de Investigación I+12, Hospital Universitario 12 de Octubre, Madrid, Spain
- CIBER-CV, Hospital Universitario 12 de Octubre, Madrid, Spain
- School of Doctoral Studies and Research, Universidad Europea de Madrid, Madrid, Spain
| | - Gloria Alvarez-Llamas
- Department of Immunology, Immunoallergy and Proteomics Laboratory, IIS-Fundación Jiménez Díaz, UAM, Avenida Reyes Católicos 2, 28040, Madrid, Spain.
- REDINREN, Madrid, Spain.
| |
Collapse
|
110
|
Castellani CA, Longchamps RJ, Sumpter JA, Newcomb CE, Lane JA, Grove ML, Bressler J, Brody JA, Floyd JS, Bartz TM, Taylor KD, Wang P, Tin A, Coresh J, Pankow JS, Fornage M, Guallar E, O'Rourke B, Pankratz N, Liu C, Levy D, Sotoodehnia N, Boerwinkle E, Arking DE. Mitochondrial DNA copy number can influence mortality and cardiovascular disease via methylation of nuclear DNA CpGs. Genome Med 2020; 12:84. [PMID: 32988399 PMCID: PMC7523322 DOI: 10.1186/s13073-020-00778-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 09/04/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Mitochondrial DNA copy number (mtDNA-CN) has been associated with a variety of aging-related diseases, including all-cause mortality. However, the mechanism by which mtDNA-CN influences disease is not currently understood. One such mechanism may be through regulation of nuclear gene expression via the modification of nuclear DNA (nDNA) methylation. METHODS To investigate this hypothesis, we assessed the relationship between mtDNA-CN and nDNA methylation in 2507 African American (AA) and European American (EA) participants from the Atherosclerosis Risk in Communities (ARIC) study. To validate our findings, we assayed an additional 2528 participants from the Cardiovascular Health Study (CHS) (N = 533) and Framingham Heart Study (FHS) (N = 1995). We further assessed the effect of experimental modification of mtDNA-CN through knockout of TFAM, a regulator of mtDNA replication, via CRISPR-Cas9. RESULTS Thirty-four independent CpGs were associated with mtDNA-CN at genome-wide significance (P < 5 × 10- 8). Meta-analysis across all cohorts identified six mtDNA-CN-associated CpGs at genome-wide significance (P < 5 × 10- 8). Additionally, over half of these CpGs were associated with phenotypes known to be associated with mtDNA-CN, including coronary heart disease, cardiovascular disease, and mortality. Experimental modification of mtDNA-CN demonstrated that modulation of mtDNA-CN results in changes in nDNA methylation and gene expression of specific CpGs and nearby transcripts. Strikingly, the "neuroactive ligand receptor interaction" KEGG pathway was found to be highly overrepresented in the ARIC cohort (P = 5.24 × 10- 12), as well as the TFAM knockout methylation (P = 4.41 × 10- 4) and expression (P = 4.30 × 10- 4) studies. CONCLUSIONS These results demonstrate that changes in mtDNA-CN influence nDNA methylation at specific loci and result in differential expression of specific genes that may impact human health and disease via altered cell signaling.
Collapse
Affiliation(s)
- Christina A Castellani
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ryan J Longchamps
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Jason A Sumpter
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Charles E Newcomb
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - John A Lane
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Megan L Grove
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jennifer A Brody
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - James S Floyd
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Traci M Bartz
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
- Department of Biostatistics, University of Washington, Seattle, WA, USA
| | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences, Los Angeles BioMedical Research Institute at Harbor-UCLA Medical Center, Torrance, CA, USA
| | - Penglong Wang
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Adrienne Tin
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Josef Coresh
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - James S Pankow
- Division of Epidemiology & Community Health, School of Public Health, University of Minnesota, Minneapolis, MN, USA
| | - Myriam Fornage
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
- Brown Foundation Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Eliseo Guallar
- Department of Epidemiology and the Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota School of Medicine, Minneapolis, MN, USA
| | - Chunyu Liu
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Daniel Levy
- Framingham Heart Study, Framingham, MA, USA
- Population Sciences Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Nona Sotoodehnia
- Cardiovascular Health Research Unit, Department of Medicine, University of Washington, Seattle, WA, USA
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dan E Arking
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
111
|
Leukocyte mitochondrial DNA copy number and built environment in Mexican Americans: a cross-sectional study. Sci Rep 2020; 10:14988. [PMID: 32917938 PMCID: PMC7486918 DOI: 10.1038/s41598-020-72083-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 08/17/2020] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial DNA (mtDNA) copy number in leukocytes has been regarded as a biomarker for various environmental exposures and chronic diseases. Our previous study showed that certain demographic factors (e.g. age, gender, BMI, etc.) significantly affect levels of leukocyte mtDNA copy number in Mexican Americans. However, the effect of the built environment on leukocyte mtDNA copy number has not been studied previously. In this cross-sectional study, we examined the association between multiple components of the built environment with leukocyte mtDNA copy number among 5,502 Mexican American adults enrolled in Mano-A-Mano, the Mexican American Cohort Study (MACS). Based on the median levels of mtDNA copy number, the study population was stratified into low mtDNA copy number group (< median) and high mtDNA copy number group (≥ median). Among all built environment exposure variables, household density and road/intersection ratio were found to be statistically significant between groups with low and high mtDNA copy number (P < 0.001 and 0.002, respectively). In the multivariate logistic regression analysis, individuals living in areas with elevated levels of household density had 1.24-fold increased odds of having high levels of mtDNA copy number [Odds ratio (OR) = 1.24, 95% confidence interval (CIs) 1.08, 1.36]. Similarly, those living in areas with elevated levels of road/intersection ratio had 1.12-fold increased odds of having high levels of mtDNA copy number (OR = 1.12, 95% CI 1.01, 1.27). In further analysis, when both variables were analyzed together in a multivariate logistic regression model, the significant associations remained. In summary, our results suggest that selected built environment variables (e.g. population density and road/intersection ratio) may influence levels of mtDNA copy number in leukocytes in Mexican Americans.
Collapse
|
112
|
Liu Y, Du X, Huang Z, Zheng Y, Quan N. Sestrin 2 controls the cardiovascular aging process via an integrated network of signaling pathways. Ageing Res Rev 2020; 62:101096. [PMID: 32544433 DOI: 10.1016/j.arr.2020.101096] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 05/03/2020] [Accepted: 06/04/2020] [Indexed: 02/07/2023]
Abstract
As an inevitable biological process, cardiovascular aging is the greatest risk factor for cardiovascular diseases (CVDs). Sestrin 2 (Sesn2), a stress-inducible and age-related protein associated with various stress conditions, plays a pivotal role in slowing this process. It acts as an anti-aging agent, mainly through its antioxidant enzymatic activity and regulation of antioxidant signaling pathways, as well as by activating adenosine monophosphate-activated protein kinase and inhibiting mammalian target of rapamycin complex 1. In this review, we first introduce the biochemical functions of Sesn2 in the cardiovascular aging process, and describe how Sesn2 expression is regulated under various stress conditions. Next, we emphasize the role of Sesn2 signal transduction in a series of age-related CVDs, including hypertension, myocardial ischemia and reperfusion, atherosclerosis, and heart failure, as well as provide potential mechanisms for the association of Sesn2 with CVDs. Finally, we present the potential therapeutic applications of Sesn2-directed therapy and future prospects.
Collapse
Affiliation(s)
- Yunxia Liu
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Xiaoyu Du
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Zhehao Huang
- Department of Neurosurgery, China-Japan Union Hospital of Jilin University, Changchun, Jilin, 130031, China
| | - Yang Zheng
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| | - Nanhu Quan
- Cardiovascular Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China.
| |
Collapse
|
113
|
PIP 2: A critical regulator of vascular ion channels hiding in plain sight. Proc Natl Acad Sci U S A 2020; 117:20378-20389. [PMID: 32764146 PMCID: PMC7456132 DOI: 10.1073/pnas.2006737117] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
The phosphoinositide, phosphatidylinositol 4,5-bisphosphate (PIP2), has long been established as a major contributor to intracellular signaling, primarily by virtue of its role as a substrate for phospholipase C (PLC). Signaling by Gq-protein-coupled receptors triggers PLC-mediated hydrolysis of PIP2 into inositol 1,4,5-trisphosphate and diacylglycerol, which are well known to modulate vascular ion channel activity. Often overlooked, however, is the role PIP2 itself plays in this regulation. Although numerous reports have demonstrated that PIP2 is critical for ion channel regulation, how it impacts vascular function has received scant attention. In this review, we focus on PIP2 as a regulator of ion channels in smooth muscle cells and endothelial cells-the two major classes of vascular cells. We further address the concerted effects of such regulation on vascular function and blood flow control. We close with a consideration of current knowledge regarding disruption of PIP2 regulation of vascular ion channels in disease.
Collapse
|
114
|
Wang X, Hart JE, Liu Q, Wu S, Nan H, Laden F. Association of particulate matter air pollution with leukocyte mitochondrial DNA copy number. ENVIRONMENT INTERNATIONAL 2020; 141:105761. [PMID: 32388147 PMCID: PMC7419671 DOI: 10.1016/j.envint.2020.105761] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 04/07/2020] [Accepted: 04/22/2020] [Indexed: 05/22/2023]
Abstract
BACKGROUND Ambient particulate matter (PM) has been associated with mitochondrial damage and dysfunction caused by excessive oxidative stress, but the associations between long-term PM exposure and leukocyte mitochondrial DNA copy number (mtDNAcn), a biomarker of mitochondrial dysfunction due to oxidative stress, are less studied. OBJECTIVES To investigate the associations between short-, intermediate- and long-term exposure (1-, 3- and 12-months) to different size fractions of PM (PM2.5, PM2.5-10 and PM10) and leukocyte mtDNAcn in a cross-sectional study. METHODS The associations between each of the PM exposure metrics with z scores of log-transformed mtDNAcn were examined using generalized linear regression models in 2758 female participants from the Nurses' Health Study (NHS). Monthly exposures to PM were estimated from spatio-temporal prediction models matched to each participants' address history. Potential effect modification by selected covariates was examined using multiplicative interaction terms and subgroup analyses. RESULTS In single-size fraction models, increases in all size fractions of PM were associated with decreases in mtDNAcn, although only models with longer averages of PM2.5 reached statistical significance. For example, an interquartile range (IQR) increase in 12-month average ambient PM2.5 (5.5 μg/m3) was associated with a 0.07 [95% confidence interval (95% CI): -0.13, -0.01; p-value = 0.02] decrease in mtDNAcn z score in both basic- and multivariable-adjusted models. Associations for PM2.5 were stronger after controlling for PM2.5-10 in two size-fraction models. CONCLUSIONS Our study suggests that long-term exposure to ambient PM2.5 is associated with decreased mtDNAcn in healthy women.
Collapse
Affiliation(s)
- Xinmei Wang
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Jaime E Hart
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Exposure, Epidemiology and Risk Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Qisijing Liu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China
| | - Shaowei Wu
- Department of Occupational and Environmental Health Sciences, School of Public Health, Peking University, Beijing, China; Key Laboratory of Molecular Cardiovascular Sciences, Peking University, Ministry of Education, China.
| | - Hongmei Nan
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, IN, USA; Indiana University Melvin and Bren Simon Cancer Center, Indianapolis, IN, USA
| | - Francine Laden
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Exposure, Epidemiology and Risk Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, MA, USA; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| |
Collapse
|
115
|
DeConne TM, Muñoz ER, Sanjana F, Hobson JC, Martens CR. Cardiometabolic risk factors are associated with immune cell mitochondrial respiration in humans. Am J Physiol Heart Circ Physiol 2020; 319:H481-H487. [PMID: 32678706 DOI: 10.1152/ajpheart.00434.2020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Modifiable cardiometabolic risk factors induce the release of proinflammatory cytokines and reactive oxygen species from circulating peripheral blood mononuclear cells (PBMCs), resulting in increased cardiovascular disease risk and compromised immune health. These changes may be driven by metabolic reprogramming of PBMCs, resulting in reduced mitochondrial respiration; however, this has not been fully tested. We aimed to determine the independent associations between cardiometabolic risk factors including BMI, blood pressure, fasting glucose, and plasma lipids with mitochondrial respiration in PBMCs isolated from generally healthy individuals (n = 21) across the adult lifespan (12 men/9 women; age, 56 ± 21 yr; age range, 22-78 yr; body mass index, 27.9 ± 5.7 kg/m2; blood pressure, 123 ± 16/72 ± 10 mmHg; glucose, 90 ± 14 mg/dL; low-density lipoprotein cholesterol (LDL-C), 111 ± 22 mg/dL; and high-density lipoprotein cholesterol (HDL-C), 62 ± 16 mg/dL). PBMCs were isolated from whole blood by density-dependent centrifugation and used to assess mitochondrial function by respirometry. Primary outcomes included basal and maximal oxygen consumption rate (OCR), which were subsequently used to determine spare respiratory capacity and OCR metabolic potential. After we corrected for systolic blood pressure (SBP), diastolic blood pressure (DBP), and blood glucose, LDL-C was negatively associated with maximal respiration (r = -0.56, P = 0.016), spare respiratory capacity (r = -0.58, P = 0.012), and OCR metabolic potential (r = -0.71, P = 0.0011). In addition, SBP was negatively associated with OCR metabolic potential (r = -0.62, P = 0.0056) after we corrected for DBP, blood glucose, and LDL-C. These data suggest a link between blood cholesterol, SBP, and mitochondrial health that may provide insight into how cardiometabolic risk factors contribute to impaired immune cell function.NEW & NOTEWORTHY Independent of other cardiometabolic risk factors, low-density lipoprotein cholesterol, and systolic blood pressure were found to be negatively associated with several parameters of mitochondrial respiration in peripheral blood mononuclear cells of healthy adults. These data suggest that low-density lipoprotein cholesterol and systolic blood pressure may induce metabolic reprogramming of immune cells, contributing to increased cardiovascular disease risk and impaired immune health.
Collapse
Affiliation(s)
- Theodore M DeConne
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - Eric R Muñoz
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - Faria Sanjana
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - Joshua C Hobson
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| | - Christopher R Martens
- Department of Kinesiology and Applied Physiology, University of Delaware, Newark, Delaware
| |
Collapse
|
116
|
Castellani CA, Longchamps RJ, Sun J, Guallar E, Arking DE. Thinking outside the nucleus: Mitochondrial DNA copy number in health and disease. Mitochondrion 2020; 53:214-223. [PMID: 32544465 DOI: 10.1016/j.mito.2020.06.004] [Citation(s) in RCA: 211] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 05/19/2020] [Accepted: 06/08/2020] [Indexed: 02/07/2023]
Abstract
Mitochondrial DNA copy number (mtDNA-CN) is a biomarker of mitochondrial function and levels of mtDNA-CN have been reproducibly associated with overall mortality and a number of age-related diseases, including cardiovascular disease, chronic kidney disease, and cancer. Recent advancements in techniques for estimating mtDNA-CN, in particular the use of DNA microarrays and next-generation sequencing data, have led to the comprehensive assessment of mtDNA-CN across these and other diseases and traits. The importance of mtDNA-CN measures to disease and these advancing technologies suggest the potential for mtDNA-CN to be a useful biomarker in the clinic. While the exact mechanism(s) underlying the association of mtDNA-CN with disease remain to be elucidated, we review the existing literature which supports roles for inflammatory dynamics, immune function and alterations to cell signaling as consequences of variation in mtDNA-CN. We propose that future studies should focus on characterizing longitudinal, cell-type and cross-tissue profiles of mtDNA-CN as well as improving methods for measuring mtDNA-CN which will expand the potential for its use as a clinical biomarker.
Collapse
Affiliation(s)
- Christina A Castellani
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Ryan J Longchamps
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jing Sun
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Eliseo Guallar
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States; The Welch Center for Prevention, Epidemiology and Clinical Research, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Dan E Arking
- McKusick-Nathans Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, United States.
| |
Collapse
|
117
|
Pauls AD, Sandhu V, Young D, Nevay DL, Yeung DF, Sirrs S, Tsang MY, Tsang TSM, Lehman A, Mezei MM, Poburko D. High rate of hypertension in patients with m.3243A>G MELAS mutations and POLG variants. Mitochondrion 2020; 53:194-202. [PMID: 32502631 DOI: 10.1016/j.mito.2020.05.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 05/22/2020] [Accepted: 05/26/2020] [Indexed: 12/14/2022]
Abstract
Animal studies suggest that decreased vascular mitochondrial DNA copy number can promote hypertension. We conducted a chart review of blood pressure and hemodynamics in patients with either mitochondrial encephalopathy, lactic acidosis, and stroke-like episodes (MELAS, n = 36) or individuals with variants in the mitochondrial DNA polymerase gamma (POLG, n = 26). The latter included both pathogenic variants and variants of unknown significance (VUS). Hypertension rates (MELAS 50%, POLG 50%) were elevated relative to Canadian norms in 20-39 (MELAS) and 40-59 (MELAS and POLG) years of age groups. Peripheral resistance was high in the hypertensive versus normotensive patients, potentially indicative of microvascular disease. Despite antihypertensive treatment, systolic blood pressure remained elevated in the POLG versus MELAS group. The risk of hypertension was not associated with MELAS heteroplasmy. Hypertension rates were not different between individuals with known pathogenic POLG variants and those with VUS, including common variants. Hypertension (HT) also did not differ between patients with POLG variants with (n = 17) and without chronic progressive external opthalmoplegia (n = 9) (CPEO). HT was associated with variants in all three functional domains of POLG. These findings suggest that both pathogenic variants and several VUS in the POLG gene may promote human hypertension and extend our past reports that increased risk of HT is associated with MELAS.
Collapse
Affiliation(s)
- Andrew D Pauls
- Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Vikrant Sandhu
- Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, Canada
| | - Dana Young
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada
| | - Dayna-Lynn Nevay
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada
| | - Darwin F Yeung
- Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Sandra Sirrs
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada
| | - Michael Y Tsang
- Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Teresa S M Tsang
- Division of Cardiology, University of British Columbia, Vancouver, BC, Canada
| | - Anna Lehman
- Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michelle M Mezei
- Adult Metabolic Diseases Unit, Vancouver General Hospital, Vancouver, BC, Canada; Division of Neurology, University of British Columbia, Vancouver, BC, Canada
| | - Damon Poburko
- Biomedical Physiology & Kinesiology, Simon Fraser University, Burnaby, Canada; Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
118
|
Forte M, Stanzione R, Cotugno M, Bianchi F, Marchitti S, Rubattu S. Vascular ageing in hypertension: Focus on mitochondria. Mech Ageing Dev 2020; 189:111267. [PMID: 32473170 DOI: 10.1016/j.mad.2020.111267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 12/25/2022]
Abstract
Hypertension is a common age-related disease, along with vascular and neurodegenerative diseases. Vascular ageing increases during hypertension, but hypertension itself accelerates vascular ageing, thus creating a vicious circle. Vascular stiffening, endothelial dysfunction, impaired contractility and vasorelaxation are the main alterations related to vascular ageing, as a consequence of vascular smooth muscle and endothelial cells senescence. Several molecular mechanisms have been involved into the functional and morphological changes of the aged vessels. Among them, oxidative stress, inflammation, extracellular matrix deregulation and mitochondrial dysfunction are the best characterized. In the present review, we discuss relevant literature about the biology of vascular and cerebrovascular ageing with a particular focus on mitochondria signalling. We underline the therapeutic strategies, able to improve mitochondrial health, which may represent a promising tool to decrease vascular dysfunction associated with ageing and hypertension-related complications.
Collapse
Affiliation(s)
- Maurizio Forte
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | | | - Maria Cotugno
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | - Franca Bianchi
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy
| | | | - Speranza Rubattu
- IRCCS Neuromed, Via Atinense, 18, 86077 Pozzilli IS, Italy; Department of Clinical and Molecular Medicine, School of Medicine and Psychology, Sapienza University of Rome, 00189 Rome, Italy.
| |
Collapse
|
119
|
Hydrogen Sulfide Promotes Cardiomyocyte Proliferation and Heart Regeneration via ROS Scavenging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1412696. [PMID: 32566074 PMCID: PMC7261318 DOI: 10.1155/2020/1412696] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 02/26/2020] [Accepted: 04/27/2020] [Indexed: 12/13/2022]
Abstract
Neonatal mouse hearts can regenerate completely in 21 days after cardiac injury, providing an ideal model to exploring heart regenerative therapeutic targets. The oxidative damage by Reactive Oxygen Species (ROS) is one of the critical reasons for the cell cycle arrest of cardiomyocytes (CMs), which cause mouse hearts losing the capacity to regenerate in 7 days or shorter after birth. As an antioxidant, hydrogen sulfide (H2S) plays a protective role in a variety of diseases by scavenging ROS produced during the pathological processes. In this study, we found that blocking H2S synthesis by PAG (H2S synthase inhibitor) suspended heart regeneration and CM proliferation with ROS deposition increase after cardiac injury (myocardial infarction or apex resection) in 2-day-old mice. NaHS (a H2S donor) administration improved heart regeneration with CM proliferation and ROS elimination after myocardial infarction in 7-day-old mice. NaHS protected primary neonatal mouse CMs from H2O2-induced apoptosis and promoted CM proliferation via SOD2-dependent ROS scavenging. The oxidative DNA damage in CMs was reduced with the elimination of ROS by H2S. Our results demonstrated for the first time that H2S promotes heart regeneration and identified NaHS as a potent modulator for cardiac repair.
Collapse
|
120
|
Forte M, Schirone L, Ameri P, Basso C, Catalucci D, Modica J, Chimenti C, Crotti L, Frati G, Rubattu S, Schiattarella GG, Torella D, Perrino C, Indolfi C, Sciarretta S. The role of mitochondrial dynamics in cardiovascular diseases. Br J Pharmacol 2020; 178:2060-2076. [DOI: 10.1111/bph.15068] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/31/2020] [Accepted: 04/01/2020] [Indexed: 12/22/2022] Open
Affiliation(s)
- Maurizio Forte
- Department of AngioCardioNeurology IRCCS Neuromed Pozzili Italy
| | - Leonardo Schirone
- Department of Medical and Surgical Sciences and Biotechnologies Sapienza University of Rome Latina Italy
- Department of Internal, Anesthetic and Cardiovascular Clinical Sciences “La Sapienza” University of Rome Rome Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit IRCCS Ospedale Policlinico Genova Italy
- Department of Internal Medicine University of Genova Genova Italy
| | - Cristina Basso
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health University of Padua Medical School Padova Italy
| | - Daniele Catalucci
- Humanitas Clinical and Research Center IRCCS Rozzano Italy
- National Research Council Institute of Genetic and Biomedical Research ‐ UOS Milan Italy
| | - Jessica Modica
- Humanitas Clinical and Research Center IRCCS Rozzano Italy
- National Research Council Institute of Genetic and Biomedical Research ‐ UOS Milan Italy
| | - Cristina Chimenti
- Department of Cardiovascular, Respiratory, Nephrologic, and Geriatric Sciences Sapienza University of Rome Rome Italy
| | - Lia Crotti
- Istituto Auxologico Italiano IRCCS, Center for Cardiac Arrhythmias of Genetic Origin and Laboratory of Cardiovascular Genetics Milan Italy
- Istituto Auxologico Italiano, IRCCS, Department of Cardiovascular, Neural and Metabolic Sciences San Luca Hospital Milan Italy
- Department of Medicine and Surgery Università Milano‐Bicocca Milan Italy
| | - Giacomo Frati
- Department of AngioCardioNeurology IRCCS Neuromed Pozzili Italy
- Department of Medical and Surgical Sciences and Biotechnologies Sapienza University of Rome Latina Italy
| | - Speranza Rubattu
- Department of AngioCardioNeurology IRCCS Neuromed Pozzili Italy
- Department of Clinical and Molecular Medicine, School of Medicine and Psychology Sapienza University of Rome Rome Italy
| | - Gabriele Giacomo Schiattarella
- Department of Internal Medicine (Cardiology) University of Texas Southwestern Medical Center Dallas TX USA
- Division of Cardiology, Department of Advanced Biomedical Sciences Federico II University of Naples Naples Italy
| | - Daniele Torella
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine Magna Graecia University Catanzaro Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences Federico II University of Naples Naples Italy
| | - Ciro Indolfi
- Molecular and Cellular Cardiology Laboratory, Department of Experimental and Clinical Medicine Magna Graecia University Catanzaro Italy
| | - Sebastiano Sciarretta
- Department of AngioCardioNeurology IRCCS Neuromed Pozzili Italy
- Department of Medical and Surgical Sciences and Biotechnologies Sapienza University of Rome Latina Italy
| | | |
Collapse
|
121
|
Xie Y, Gao Y, Gao R, Yang W, Dong Z, Moses RE, Sun A, Li X, Ge J. The proteasome activator REGγ accelerates cardiac hypertrophy by declining PP2Acα-SOD2 pathway. Cell Death Differ 2020; 27:2952-2972. [PMID: 32424140 PMCID: PMC7494903 DOI: 10.1038/s41418-020-0554-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/05/2022] Open
Abstract
Pathological cardiac hypertrophy eventually leads to heart failure without adequate treatment. REGγ is emerging as 11S proteasome activator of 20S proteasome to promote the degradation of cellular proteins in a ubiquitin- and ATP-independent manner. Here, we found that REGγ was significantly upregulated in the transverse aortic constriction (TAC)-induced hypertrophic hearts and angiotensin II (Ang II)-treated cardiomyocytes. REGγ deficiency ameliorated pressure overload-induced cardiac hypertrophy were associated with inhibition of cardiac reactive oxygen species (ROS) accumulation and suppression of protein phosphatase 2A catalytic subunit α (PP2Acα) decay. Mechanistically, REGγ interacted with and targeted PP2Acα for degradation directly, thereby leading to increase of phosphorylation levels and nuclear export of Forkhead box protein O (FoxO) 3a and subsequent of SOD2 decline, ROS accumulation, and cardiac hypertrophy. Introducing exogenous PP2Acα or SOD2 to human cardiomyocytes significantly rescued the REGγ-mediated ROS accumulation of Ang II stimulation in vitro. Furthermore, treatment with superoxide dismutase mimetic, MnTBAP prevented cardiac ROS production and hypertrophy features that REGγ caused in vivo, thereby establishing a REGγ–PP2Acα–FoxO3a–SOD2 pathway in cardiac oxidative stress and hypertrophy, indicates modulating the REGγ-proteasome activity may be a potential therapeutic approach in cardiac hypertrophy-associated disorders.
Collapse
Affiliation(s)
- Yifan Xie
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Institutes of Biomedical Science, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Yang Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Rifeng Gao
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Wenlong Yang
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Zheng Dong
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China.,Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China
| | - Robb E Moses
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA
| | - Aijun Sun
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Institutes of Biomedical Science, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China.
| | - Xiaotao Li
- Department of Molecular and Cellular Biology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, 77030, USA. .,Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, 500 Dongchuan Road, Shanghai, 200241, China.
| | - Junbo Ge
- Department of Cardiology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Institutes of Biomedical Science, Fudan University, 180 Fenglin Road, Shanghai, 200032, China. .,Shanghai Institute of Cardiovascular Diseases, 180 Fenglin Road, Shanghai, 200032, China.
| |
Collapse
|
122
|
Chen YF, Hebert VY, Stadler K, Xue SY, Slaybaugh K, Luttrell-Williams E, Glover MC, Krzywanski DM, Dugas TR. Coenzyme Q10 Alleviates Chronic Nucleoside Reverse Transcriptase Inhibitor-Induced Premature Endothelial Senescence. Cardiovasc Toxicol 2020; 19:500-509. [PMID: 31020509 DOI: 10.1007/s12012-019-09520-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Human immunodeficiency virus (HIV)-infected patients undergoing antiretroviral therapy are afforded an increased lifespan but also exhibit an elevated incidence of cardiovascular disease. HIV therapy uses a combination drug approach, and nucleoside reverse transcriptase inhibitors (NRTI) are a backbone of this therapy. Endothelial dysfunction is an initiating event in cardiovascular disease etiology, and in our prior studies, NRTIs induced an endothelial dysfunction that was dependent upon mitochondrial oxidative stress. Moreover, short-term NRTI administration induced a mitophagy-associated endothelial toxicity and increased reactive oxygen species (ROS) production that was rescued by coenzyme Q10 (Q10) or overexpression of a mitochondrial antioxidant enzyme. Thus, our objective was to examine mitochondrial toxicity in endothelial cells after chronic NRTI treatment and evaluate Q10 as a potential adjunct therapy for preventing NRTI-induced mitochondrial toxicity. Human aortic endothelial cells (HAEC) were exposed to chronic NRTI treatment, with or without Q10. ROS production, cell proliferation rate, levels of senescence, and mitochondrial bioenergetic function were determined. Chronic NRTI increased ROS production but decreased population doubling. In addition, NRTI increased the accumulation of β-galactosidase, indicative of an accelerated rate of senescence. Moreover, ATP-linked respiration was diminished. Co-treatment with Q10 delayed the onset of NRTI-induced senescence, decreased ROS production and rescued the cells' mitochondrial respiration rate. Thus, our findings may suggest antioxidant enrichment approaches for reducing the cardiovascular side effects of NRTI therapy.
Collapse
Affiliation(s)
- Yi-Fan Chen
- Comparative Biomedical Sciences, LSU School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, USA
| | - Valeria Y Hebert
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, 70808, USA
| | - Stephen Y Xue
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Kate Slaybaugh
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Elliot Luttrell-Williams
- Pharmacology, Toxicology and Neuroscience, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Mitzi C Glover
- Clinical Laboratory Sciences, School of Allied Health Professions, LSU Health Sciences Center, 1900 Gravier Street, New Orleans, LA, 70112, USA
| | - David M Krzywanski
- Cellular Biology and Anatomy, LSU Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Tammy R Dugas
- Comparative Biomedical Sciences, LSU School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, USA.
| |
Collapse
|
123
|
Cumulative Effect of Cardiovascular Risk Factors on Regulation of AMPK/SIRT1-PGC-1 α-SIRT3 Pathway in the Human Erectile Tissue. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1525949. [PMID: 32377289 PMCID: PMC7195659 DOI: 10.1155/2020/1525949] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/02/2020] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease risk factors (CVDRF), especially diabetes mellitus (DM), disrupt oxidative stress response. This condition underlies endothelial dysfunction, early manifested in men as erectile dysfunction. The current study is aimed at elucidating the impact of CVDRF in the oxidation responsive AMPK/SIRT1-PGC-1α-SIRT3 pathway and related miRNAs in the human corpus cavernosum. Human penile tissue fragments from individuals submitted to programmed urological surgeries (n = 27), aged 43-63 years, were clustered depending on the presence of CVDRF; the control group included samples from patients without CVDRF, and groups A and B included samples from patients with DM and additional CVDRF, totalizing ≤2 CVDRF (group A) and ≥3 CVDRF (group B). Dual-immunolabelling of SIRT3, SOD2, or GPX1 with α-actin in tissue sections was carried out. The assessment of expression levels of NOX1, phospho-AMPKα, total AMPKα, SIRT1, PGC-1α, SIRT3, SOD2, and GPX1 was performed by western blotting and of miR-200a, miR-34a, miR-421, and miR-206 by real-time PCR. Phospho-AMPKα and SIRT3 expression was found significantly increased in group B relative to other groups, suggesting a marked influence of CVDRF, additional to DM, in the regulation of these enzymes. NOX1 was also increased in group B relative to controls. Only an increasing tendency was observed in the phospho-AMPKα/total AMPKα ratio, SIRT1, and PGC-1α expression in groups A and B when compared with controls. Concerning antioxidant enzymes, GPX1 expression was found incremented in group A, but SOD2 expression was decreased in groups A and B, comparative with controls. Group B presented significantly diminished levels of miR-421 and miR-200a, but only a decreasing trend on miR-34 and miR-206 expression was observed. Taken together, our findings demonstrated that besides DM, additional CVDRF presented a cumulative effect in the cellular response to oxidative unbalance, contributing to AMPK/SIRT1-PGC-1α-SIRT3 pathway activation. SOD2, a major mitochondrial antioxidant defence, did not follow the same variation.
Collapse
|
124
|
Warraich UEA, Hussain F, Kayani HUR. Aging - Oxidative stress, antioxidants and computational modeling. Heliyon 2020; 6:e04107. [PMID: 32509998 PMCID: PMC7264715 DOI: 10.1016/j.heliyon.2020.e04107] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/12/2020] [Accepted: 05/27/2020] [Indexed: 12/22/2022] Open
Abstract
Aging is a degenerative, biological, time-dependent, universally conserved process thus designed as one of the highest known risk factors for morbidity and mortality. Every individual has its own aging mechanisms as both environmental conditions (75%) and genetics (25%) account for aging. Several theories have been proposed until now but not even a single theory solves this mystery. There are still some queries un-answered to the scientific community regarding mechanisms behind aging. However, oxidative stress theory (OST) is considered one of the famous theories that sees mitochondria as one of the leading organelles which largely contribute to the aging process. Many reactive oxygen species (ROS) are produced endogenously and exogenously that are associated with aging. But the mitochondrial ROS contribute largely to the aging process as mitochondrial dysfunction due to oxidative stress is considered one of the contributors toward aging. Although ROS is known to damage cell machinery, new evidence suggests their role in signal transduction to regulate biological and physiological processes. Moreover, besides mitochondria, other important cell organelles such as peroxisome and endoplasmic reticulum also produce ROS that contribute to aging. However, nature has provided humans with free radical scavengers called antioxidants that protect from harmful effects of ROS. Future predictions regarding aging, biochemical mechanisms involved, biomarkers internal and external factors can be easily done with machine learning algorithms and other computational models. This review explains important aspects of aging, the contribution of ROS producing organelles in aging, importance of antioxidants fighting against ROS, different computational models developed to understand the complexities of the aging.
Collapse
Affiliation(s)
- Umm-e-Ammara Warraich
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | - Fatma Hussain
- Department of Biochemistry, Faculty of Sciences, University of Agriculture, Faisalabad, Pakistan
| | | |
Collapse
|
125
|
Kiss T, Tarantini S, Csipo T, Balasubramanian P, Nyúl-Tóth Á, Yabluchanskiy A, Wren JD, Garman L, Huffman DM, Csiszar A, Ungvari Z. Circulating anti-geronic factors from heterochonic parabionts promote vascular rejuvenation in aged mice: transcriptional footprint of mitochondrial protection, attenuation of oxidative stress, and rescue of endothelial function by young blood. GeroScience 2020; 42:727-748. [PMID: 32172434 PMCID: PMC7205954 DOI: 10.1007/s11357-020-00180-6] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 03/04/2020] [Indexed: 12/15/2022] Open
Abstract
Aging-induced functional and phenotypic alterations of the vasculature (e.g., endothelial dysfunction, oxidative stress) have a central role in morbidity and mortality of older adults. It has become apparent in recent years that cell autonomous mechanisms alone are inadequate to explain all aspects of vascular aging. The present study was designed to test the hypothesis that age-related changes in circulating anti-geronic factors contribute to the regulation of vascular aging processes in a non-cell autonomous manner. To test this hypothesis, through heterochronic parabiosis we determined the extent, if any, to which endothelial function, vascular production of ROS, and shifts in the vascular transcriptome (RNA-seq) are modulated by the systemic environment. We found that in aortas isolated from isochronic parabiont aged (20-month-old) C57BL/6 mice [A-(A); parabiosis for 8 weeks] acetylcholine-induced endothelium-dependent relaxation was impaired and ROS production (dihydroethidium fluorescence) was increased as compared with those in aortas from young isochronic parabiont (6-month-old) mice [Y-(Y)]. The presence of young blood derived from young parabionts significantly improved endothelium-dependent vasorelaxation and attenuated ROS production in vessels of heterochronic parabiont aged [A-(Y)] mice. In aortas derived from heterochronic parabiont young [Y-(A)] mice, acetylcholine-induced relaxation and ROS production were comparable with those in aortas derived from Y-(Y) mice. Using RNA-seq we assessed transcriptomic changes in the aortic arch associated with aging and heterochronic parabiosis. We identified 347 differentially expressed genes in A-(A) animals compared with Y-(Y) controls. We have identified 212 discordant genes, whose expression levels differed in the aged phenotype, but have shifted back toward the young phenotype by the presence of young blood in aged A-(Y) animals. Pathway analysis shows that vascular protective effects mediated by young blood-regulated genes include mitochondrial rejuvenation. In conclusion, a relatively short-term exposure to young blood can rescue vascular aging phenotypes, including attenuation of oxidative stress, mitochondrial rejuvenation, and improved endothelial function. Our findings provide additional evidence supporting the significant plasticity of vascular aging and evidence for the existence of anti-geronic factors capable of exerting rejuvenating effects on the aging vasculature.
Collapse
Affiliation(s)
- Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Department of Cardiology, Division of Clinical Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
| | - Jonathan D. Wren
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK USA
| | - Lori Garman
- Oklahoma Medical Research Foundation, Genes & Human Disease Research Program, Oklahoma City, OK USA
| | - Derek M. Huffman
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461 USA
- Institute for Aging Research, Albert Einstein College of Medicine, Bronx, NY USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY USA
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Center for Geroscience and Healthy Brain Aging, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK 73104 USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School/Departments of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104 USA
| |
Collapse
|
126
|
Mitochondrial ROS-Modulated mtDNA: A Potential Target for Cardiac Aging. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:9423593. [PMID: 32308810 PMCID: PMC7139858 DOI: 10.1155/2020/9423593] [Citation(s) in RCA: 112] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 03/05/2020] [Accepted: 03/16/2020] [Indexed: 02/05/2023]
Abstract
Mitochondrial DNA (mtDNA) damage is associated with the development of cardiovascular diseases. Cardiac aging plays a central role in cardiovascular diseases. There is accumulating evidence linking cardiac aging to mtDNA damage, including mtDNA mutation and decreased mtDNA copy number. Current wisdom indicates that mtDNA is susceptible to damage by mitochondrial reactive oxygen species (mtROS). This review presents the cellular and molecular mechanisms of cardiac aging, including autophagy, chronic inflammation, mtROS, and mtDNA damage, and the effects of mitochondrial biogenesis and oxidative stress on mtDNA. The importance of nucleoid-associated proteins (Pol γ), nuclear respiratory factors (NRF1 and NRF2), the cGAS-STING pathway, and the mitochondrial biogenesis pathway concerning the development of mtDNA damage during cardiac aging is discussed. Thus, the repair of damaged mtDNA provides a potential clinical target for preventing cardiac aging.
Collapse
|
127
|
Zinovkin RA, Zamyatnin AA. Mitochondria-Targeted Drugs. Curr Mol Pharmacol 2020; 12:202-214. [PMID: 30479224 PMCID: PMC6875871 DOI: 10.2174/1874467212666181127151059] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 11/05/2018] [Accepted: 11/19/2018] [Indexed: 01/06/2023]
Abstract
Background: Targeting of drugs to the subcellular compartments represents one of the modern trends in molecular pharmacology. The approach for targeting mitochondria was developed nearly 50 years ago, but only in the last decade has it started to become widely used for delivering drugs. A number of pathologies are associated with mitochondrial dysfunction, including cardiovascular, neurological, inflammatory and metabolic conditions. Objective: This mini-review aims to highlight the role of mitochondria in pathophysiological conditions and diseases, to classify and summarize our knowledge about targeting mitochondria and to review the most important preclinical and clinical data relating to the antioxidant lipophilic cations MitoQ and SkQ1. Methods: This is a review of available information in the PubMed and Clinical Trials databases (US National Library of Medicine) with no limiting period. Results and Conclusion: Mitochondria play an important role in the pathogenesis of many diseases and possibly in aging. Both MitoQ and SkQ1 have shown many beneficial features in animal models and in a few completed clinical trials. More clinical trials and research efforts are needed to understand the signaling pathways influenced by these compounds. The antioxidant lipophilic cations have great potential for the treatment of a wide range of pathologies.
Collapse
Affiliation(s)
- Roman A Zinovkin
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russian Federation.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Institute of Mitoengineering, Moscow State University, Moscow, Russian Federation
| | - Andrey A Zamyatnin
- Belozersky Institute of Physico-Chemical Biology, Moscow State University, Moscow, Russian Federation.,Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russian Federation
| |
Collapse
|
128
|
Yang Y, Li T, Li Z, Liu N, Yan Y, Liu B. Role of Mitophagy in Cardiovascular Disease. Aging Dis 2020; 11:419-437. [PMID: 32257551 PMCID: PMC7069452 DOI: 10.14336/ad.2019.0518] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/18/2019] [Indexed: 12/17/2022] Open
Abstract
Cardiovascular disease is the leading cause of mortality worldwide, and mitochondrial dysfunction is the primary contributor to these disorders. Recent studies have elaborated on selective autophagy-mitophagy, which eliminates damaged and dysfunctional mitochondria, stabilizes mitochondrial structure and function, and maintains cell survival and growth. Numerous recent studies have reported that mitophagy plays an important role in the pathogenesis of various cardiovascular diseases. This review summarizes the mechanisms underlying mitophagy and advancements in studies on the role of mitophagy in cardiovascular disease.
Collapse
Affiliation(s)
- Yibo Yang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Tianyi Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Youyou Yan
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130041, China
| |
Collapse
|
129
|
Park SH, Kwon OS, Park SY, Weavil JC, Hydren JR, Reese V, Andtbacka RHI, Hyngstrom JR, Richardson RS. Vasodilatory and vascular mitochondrial respiratory function with advancing age: evidence of a free radically mediated link in the human vasculature. Am J Physiol Regul Integr Comp Physiol 2020; 318:R701-R711. [PMID: 32022597 DOI: 10.1152/ajpregu.00268.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recognizing the age-related decline in skeletal muscle feed artery (SMFA) vasodilatory function, this study examined the link between vasodilatory and mitochondrial respiratory function in the human vasculature. Twenty-four SMFAs were harvested from young (35 ± 6 yr, n = 9) and old (71 ± 9 yr, n = 15) subjects. Vasodilation in SMFAs was assessed, by pressure myography, in response to flow-induced shear stress, acetylcholine (ACh), and sodium nitroprusside (SNP) while mitochondrial respiration was measured, by respirometry, in permeabilized SMFAs. Endothelium-dependent vasodilation was significantly attenuated in the old, induced by both flow (young: 92 ± 3, old: 45 ± 4%) and ACh (young: 92 ± 3, old: 54 ± 5%), with no significant difference in endothelium-independent vasodilation. Complex I and I + II state 3 respiration was significantly lower in the old (CI young: 10.1 ± 0.8, old: 7.0 ± 0.4 pmol·s-1·mg-1; CI + II young: 12.3 ± 0.6, old: 7.6 ± 0.4 pmol·s-1·mg-1). The respiratory control ratio (RCR) was also significantly attenuated in the old (young: 2.2 ± 0.1, old: 1.1 ± 0.1). Furthermore, state 3 (CI + II) and 4 respiration, as well as RCR, were significantly correlated (r = 0.49-0.86) with endothelium-dependent, but not endothelium-independent, function. Finally, the direct intervention with mitochondrial-targeted antioxidant (MitoQ) significantly improved endothelium-dependent vasodilation in the old but not in the young. Thus, the age-related decline in vasodilatory function is linked to attenuated vascular mitochondrial respiratory function, likely by augmented free radicals.NEW & NOTEWORTHY In human skeletal muscle feed arteries, the well-recognized age-related fall in endothelium-dependent vasodilatory function is strongly linked to a concomitant fall in vascular mitochondrial respiratory function. The direct intervention with the mitochondrial-targeted antioxidant restored vasodilatory function in the old but not in the young, supporting the concept that exacerbated mitochondrial-derived free radical production is linked to age-related vasodilatory dysfunction. Age-related vasodilatory dysfunction in humans is linked to attenuated vascular mitochondrial respiratory function, likely a consequence of augmented free radical production.
Collapse
Affiliation(s)
- Soung Hun Park
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans' Affairs Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah Salt Lake City, Utah
| | - Oh Sung Kwon
- Department of Kinesiology, University of Connecticut, Storrs, Connecticut
| | - Song-Young Park
- School of Health and Kinesiology, University of Nebraska, Omaha, Nebraska
| | - Joshua C Weavil
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans' Affairs Medical Center, Salt Lake City, Utah.,Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Jay R Hydren
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans' Affairs Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah Salt Lake City, Utah
| | - Van Reese
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans' Affairs Medical Center, Salt Lake City, Utah.,Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Robert H I Andtbacka
- Department of Surgery, Huntsman Cancer Hospital, University of Utah, Salt Lake City, Utah
| | - John R Hyngstrom
- Department of Surgery, Huntsman Cancer Hospital, University of Utah, Salt Lake City, Utah
| | - Russell S Richardson
- Geriatric Research, Education, and Clinical Center, George E. Whalen Veterans' Affairs Medical Center, Salt Lake City, Utah.,Department of Nutrition and Integrative Physiology, University of Utah Salt Lake City, Utah.,Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| |
Collapse
|
130
|
Hydrogen Sulfide as a Potential Alternative for the Treatment of Myocardial Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4105382. [PMID: 32064023 PMCID: PMC6998763 DOI: 10.1155/2020/4105382] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
Harmful, stressful conditions or events in the cardiovascular system result in cellular damage, inflammation, and fibrosis. Currently, there is no targeted therapy for myocardial fibrosis, which is highly associated with a large number of cardiovascular diseases and can lead to fatal heart failure. Hydrogen sulfide (H2S) is an endogenous gasotransmitter similar to nitric oxide and carbon monoxide. H2S is involved in the suppression of oxidative stress, inflammation, and cellular death in the cardiovascular system. The level of H2S in the body can be boosted by stimulating its synthesis or supplying it exogenously with a simple H2S donor with a rapid- or slow-releasing mode, an organosulfur compound, or a hybrid with known drugs (e.g., aspirin). Hypertension, myocardial infarction, and inflammation are exaggerated when H2S is reduced. In addition, the exogenous delivery of H2S mitigates myocardial fibrosis caused by various pathological conditions, such as a myocardial infarct, hypertension, diabetes, or excessive β-adrenergic stimulation, via its involvement in a variety of signaling pathways. Numerous experimental findings suggest that H2S may work as a potential alternative for the management of myocardial fibrosis. In this review, the antifibrosis role of H2S is briefly addressed in order to gain insight into the development of novel strategies for the treatment of myocardial fibrosis.
Collapse
|
131
|
Wiedenhoeft T, Tarantini S, Nyúl-Tóth Á, Yabluchanskiy A, Csipo T, Balasubramanian P, Lipecz A, Kiss T, Csiszar A, Csiszar A, Ungvari Z. Fusogenic liposomes effectively deliver resveratrol to the cerebral microcirculation and improve endothelium-dependent neurovascular coupling responses in aged mice. GeroScience 2019; 41:711-725. [PMID: 31654270 PMCID: PMC6925096 DOI: 10.1007/s11357-019-00102-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 09/10/2019] [Indexed: 12/11/2022] Open
Abstract
Adjustment of cerebral blood flow (CBF) to the increased oxygen and nutrient demands of active brain regions via neurovascular coupling (NVC) has an essential role in maintenance of healthy cognitive function. In advanced age, cerebromicrovascular oxidative stress and endothelial dysfunction impair neurovascular coupling, contributing to age-related cognitive decline. Recently we developed a resveratrol (3,4',5-trihydroxystilbene)-containing fusogenic liposome (FL-RSV)-based molecular delivery system that can effectively target cultured cerebromicrovascular endothelial cells, attenuating age-related oxidative stress. To assess the cerebromicrovascular protective effects of FL-RSV in vivo, aged (24-month-old) C57BL/6 mice were treated with FL-RSV for four days. To demonstrate effective cellular uptake of FL-RSV, accumulation of the lipophilic tracer dyes in cells of the neurovascular unit was confirmed using two-photon imaging (through a chronic cranial window). NVC was assessed by measuring CBF responses (laser speckle contrast imaging) evoked by contralateral whisker stimulation. We found that NVC responses were significantly impaired in aged mice. Treatment with FL-RSV significantly improved NVC responses by increasing NO-mediated vasodilation. These findings are paralleled by the protective effects of FL-RSV on endothelium-dependent relaxation in the aorta. Thus, treatment with FL-RSV rescues endothelial function and NVC responses in aged mice. We propose that resveratrol containing fusogenic liposomes could also be used for combined delivery of various anti-geronic factors, including proteins, small molecules, DNA vectors and mRNAs targeting key pathways involved in microvascular aging and neurovascular dysfunction for the prevention/treatment of age-related cerebromicrovascular pathologies and development of vascular cognitive impairment (VCI) in aging.
Collapse
Affiliation(s)
- Tabea Wiedenhoeft
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Institute of Complex Systems, ICS-7: Biomechanics, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Ádám Nyúl-Tóth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Csipo
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Division of Clinical Physiology, Department of Cardiology/ Kalman Laki Doctoral School, Faculty of Medicine, University of Debrecen, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Agnes Lipecz
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience,Theoretical Medicine Doctoral School/Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- International Training Program in Geroscience,Theoretical Medicine Doctoral School/Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- Doctoral School of Basic and Translational Medicine/Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Agnes Csiszar
- Institute of Complex Systems, ICS-7: Biomechanics, Forschungszentrum Jülich GmbH, 52425, Jülich, Germany
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Oklahoma Center for Geroscience, Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Department of Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral School of Basic and Translational Medicine/Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary.
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
132
|
Arauna D, Furrianca M, Espinosa-Parrilla Y, Fuentes E, Alarcón M, Palomo I. Natural Bioactive Compounds As Protectors Of Mitochondrial Dysfunction In Cardiovascular Diseases And Aging. Molecules 2019; 24:molecules24234259. [PMID: 31766727 PMCID: PMC6930637 DOI: 10.3390/molecules24234259] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2019] [Revised: 09/05/2019] [Accepted: 09/08/2019] [Indexed: 01/04/2023] Open
Abstract
Diet, particularly the Mediterranean diet, has been considered as a protective factor against the development of cardiovascular diseases, the main cause of death in the world. Aging is one of the major risk factors for cardiovascular diseases, which have an oxidative pathophysiological component, being the mitochondria one of the key organelles in the regulation of oxidative stress. Certain natural bioactive compounds have the ability to regulate oxidative phosphorylation, the production of reactive oxygen species and the expression of mitochondrial proteins; but their efficacy within the mitochondrial physiopathology of cardiovascular diseases has not been clarified yet. The following review has the purpose of evaluating several natural compounds with evidence of mitochondrial effect in cardiovascular disease models, ascertaining the main cellular mechanisms and their potential use as functional foods for prevention of cardiovascular disease and healthy aging.
Collapse
Affiliation(s)
- Diego Arauna
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
| | - María Furrianca
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Departamento de enfermería, Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Yolanda Espinosa-Parrilla
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Laboratory of Molecular Medicine —LMM, Center for Education, Healthcare and Investigation—CADI, Universidad de Magallanes, Punta Arenas 6200000, Chile
- School of Medicine, Universidad de Magallanes, Punta Arenas 6200000, Chile
| | - Eduardo Fuentes
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Correspondence: (E.F.); (I.P.)
| | - Marcelo Alarcón
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
| | - Iván Palomo
- Thrombosis Research Center, Medical Technology School, Department of Clinical Biochemistry and Immunohaematology, Faculty of Health Sciences, Interdisciplinary Center on Aging, Universidad de Talca, Talca 3460000, Chile; (D.A.); (M.A.)
- Thematic Task Force on Aging, CUECH Research Network, Santiago 8320000, Chile; (M.F.); (Y.E.-P.)
- Correspondence: (E.F.); (I.P.)
| |
Collapse
|
133
|
Wang X, Liu Y, Jia Y, Liu H, Bao X, He Z, Ge W. Proteome Profiling of Cerebral Vessels in Rhesus Macaques: Dysregulation of Antioxidant Activity and Extracellular Matrix Proteins Contributes to Cerebrovascular Aging in Rhesus Macaques. Front Aging Neurosci 2019; 11:293. [PMID: 31708766 PMCID: PMC6819311 DOI: 10.3389/fnagi.2019.00293] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 10/10/2019] [Indexed: 12/28/2022] Open
Abstract
Aging is a major risk factor for cerebrovascular disease; however, the molecular mechanisms of cerebrovascular aging remain to be clarified. The aim of this study was to reveal the molecular signaling pathways involved in cerebrovascular aging. This study used high-resolution liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS), in combination with quantitative 6-plex tandem mass tag labeling, to profile protein changes in brain vessels from three groups of healthy rhesus macaques (3-years, 6-years, and 20-years). Western blot analyses were used to validate the proteomic data. A total of 2,934 proteins were identified and analyzed. Twenty-two proteins were continuously downregulated with increasing age, while three proteins were continuously upregulated. When comparing Group C vs. Group B, 270 proteins were downregulated, while 73 proteins were upregulated. All these 368 significantly changed proteins were used for further analysis. Bioinformatic analysis showed that the changed proteins were involved in several signaling pathways during cerebrovascular aging. Proteins in the NRF2 pathway, such as Glutathione S-transferase Mu (GSTM), were consistently downregulated especially after 6-years old, whereas proteins related to miRNA targets in the extracellular matrix (ECM) and membrane receptors were upregulated. Protein-protein interaction networks demonstrated that disorders of energy pathways and serine/threonine kinases were critical during cerebrovascular aging. Data are available via ProteomeXchange under the identifier PXD012306. Our results indicated that during aging, the disorders of energy metabolism and dysfunction of antioxidant activity caused over-production of reactive oxygen species (ROS) may exacerbate cerebrovascular aging. In addition, accumulation of ECM proteins during aging might be closely associated with age-related arterial stiffening and decreased compliance.
Collapse
Affiliation(s)
- Xia Wang
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yifan Liu
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Yangjie Jia
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Haotian Liu
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Zhanlong He
- Yunnan Key Laboratory of Vaccine Research and Development on Severe Infectious Disease, Institute of Medical Biology, Chinese Academy of Medical Sciences, Peking Union Medical College, Kunming, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology, Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, School of Basic Medicine, Peking Union Medical College, Beijing, China
- Department of Neurosurgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
134
|
Murphy KR, Baggett B, Cooper LL, Lu Y, O-Uchi J, Sedivy JM, Terentyev D, Koren G. Enhancing Autophagy Diminishes Aberrant Ca 2+ Homeostasis and Arrhythmogenesis in Aging Rabbit Hearts. Front Physiol 2019; 10:1277. [PMID: 31636573 PMCID: PMC6787934 DOI: 10.3389/fphys.2019.01277] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022] Open
Abstract
Aim Aging in humans is associated with a 10–40-fold greater incidence of sudden cardiac death from malignant tachyarrhythmia. We have reported that thiol oxidation of ryanodine receptors (RyR2s) by mitochondria-derived reactive oxygen species (mito-ROS) contributes to defective Ca2+ homeostasis in cardiomyocytes (CMs) from aging rabbit hearts. However, mechanisms responsible for the increase in mito-ROS in the aging heart remain poorly understood. Here we test the hypothesis that age-associated decrease in autophagy is a major contributor to enhanced mito-ROS production and thereby pro-arrhythmic disturbances in Ca2+ homeostasis. Methods and Results Ventricular tissues from aged rabbits displayed significant downregulation of proteins involved in mitochondrial autophagy compared with tissues from young controls. Blocking autophagy with chloroquine increased total ROS production in primary rabbit CMs and mito-ROS production in HL-1 CMs. Furthermore, chloroquine treatment of HL-1 cells depolarized mitochondrial membrane potential (Δψm) to 50% that of controls. Blocking autophagy significantly increased oxidation of RyR2, resulting in enhanced propensity to pro-arrhythmic spontaneous Ca2+ release under β-adrenergic stimulation. Aberrant Ca2+ release was abolished by treatment with the mito-ROS scavenger mito-TEMPO. Importantly, the autophagy enhancer Torin1 and ATG7 overexpression reduced the rate of mito-ROS production and restored both Δψm and defective Ca2+ handling in CMs derived from aged rabbit hearts. Conclusion Decreased autophagy is a major cause of increased mito-ROS production in the aging heart. Our data suggest that promoting autophagy may reduce pathologic mito-ROS during normal aging and reduce pro-arrhythmic spontaneous Ca2+ release via oxidized RyR2s.
Collapse
Affiliation(s)
- Kevin R Murphy
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States.,Department of Molecular Biology, Cellular Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Brett Baggett
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States.,Department of Molecular Biology, Cellular Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Leroy L Cooper
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States.,Department of Biology, Vassar College, Poughkeepsie, NY, United States
| | - Yichun Lu
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Jin O-Uchi
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - John M Sedivy
- Department of Molecular Biology, Cellular Biology and Biochemistry, Brown University, Providence, RI, United States
| | - Dmitry Terentyev
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Gideon Koren
- Cardiovascular Research Center at the Cardiovascular Institute, Division of Cardiology, Warren Alpert Medical School of Brown University, Providence, RI, United States
| |
Collapse
|
135
|
Csiszar A, Yabluchanskiy A, Ungvari A, Ungvari Z, Tarantini S. Overexpression of catalase targeted to mitochondria improves neurovascular coupling responses in aged mice. GeroScience 2019; 41:609-617. [PMID: 31643012 PMCID: PMC6885076 DOI: 10.1007/s11357-019-00111-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 09/20/2019] [Indexed: 12/22/2022] Open
Abstract
Moment-to-moment adjustment of cerebral blood flow (CBF) to neuronal activity via the homeostatic mechanism known as neurovascular coupling (NVC) has an essential role in maintenance of normal brain function. In advanced age cerebromicrovascular endothelial dysfunction impairs NVC responses, which contribute to age-related cognitive decline. Recently, we have shown that pharmacological treatments that attenuate mitochondrial production of reactive oxygen species (ROS) provide significant neurovascular protection, improving NVC responses in aged mice. Transgenic mice that overexpress human catalase localized to the mitochondria (mCAT) are protected from age-related mitochondrial oxidative stress and exhibit a longevity phenotype associated with resistance to several age-related pathologies. The present study was designed to test the hypothesis that mitochondria-targeted overexpression of catalase also confers protection against age-related impairment of NVC responses. To achieve this goal, NVC responses were assessed in aged (24 months old) mCAT mice and compared with those in age-matched wild-type mice and young control mice by measuring CBF responses (laser speckle contrast imaging) evoked by contralateral whisker stimulation. We found that mitochondrial overexpression of catalase resulted in improved NVC in aged mice due to preserved NO-mediated (L-NAME inhibitable) component of the response. Thus, our present and previous findings demonstrate that interventions that boost mitochondrial antioxidative defenses confer significant cerebromicrovascular protective effects, which preserve NVC responses in aged mice. Our findings provide additional proof-of-concept for the potential use of mitochondria-targeted antioxidants as therapy for prevention of vascular cognitive impairment associated with aging.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Anna Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Oklahoma Center for Geroscience, Department of Biochemistry, University of Oklahoma Health Sciences Center, 975 NE 10th Street, BRC 1311, Oklahoma City, OK, 73104, USA.
- International Training Program in Geroscience, Theoretical Medicine Doctoral School, Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary.
- International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine, Department of Public Health, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
136
|
Abstract
Cardiovascular diseases are the most prominent maladies in aging societies. Indeed, aging promotes the structural and functional declines of both the heart and the blood circulation system. In this review, we revise the contribution of known longevity pathways to cardiovascular health and delineate the possibilities to interfere with them. In particular, we evaluate autophagy, the intracellular catabolic recycling system associated with life- and health-span extension. We present genetic models, pharmacological interventions, and dietary strategies that block, reduce, or enhance autophagy upon age-related cardiovascular deterioration. Caloric restriction or caloric restriction mimetics like metformin, spermidine, and rapamycin (all of which trigger autophagy) are among the most promising cardioprotective interventions during aging. We conclude that autophagy is a fundamental process to ensure cardiac and vascular health during aging and outline its putative therapeutic importance.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.)
| | - Simon Sedej
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.).,BioTechMed Graz, Austria (S.S., D.C.-G., F.M.)
| | - Didac Carmona-Gutierrez
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Frank Madeo
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France (G.K.).,Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France (G.K.).,INSERM, U1138, Paris, France (G.K.).,Université Paris Descartes, Sorbonne Paris Cité, France (G.K.).,Université Pierre et Marie Curie, Paris, France (G.K.).,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France (G.K.).,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden (G.K.)
| |
Collapse
|
137
|
Abstract
Aging of the vasculature plays a central role in morbidity and mortality of older people. To develop novel treatments for amelioration of unsuccessful vascular aging and prevention of age-related vascular pathologies, it is essential to understand the cellular and functional changes that occur in the vasculature during aging. In this review, the pathophysiological roles of fundamental cellular and molecular mechanisms of aging, including oxidative stress, mitochondrial dysfunction, impaired resistance to molecular stressors, chronic low-grade inflammation, genomic instability, cellular senescence, epigenetic alterations, loss of protein homeostasis, deregulated nutrient sensing, and stem cell dysfunction in the vascular system are considered in terms of their contribution to the pathogenesis of both microvascular and macrovascular diseases associated with old age. The importance of progeronic and antigeronic circulating factors in relation to development of vascular aging phenotypes are discussed. Finally, future directions and opportunities to develop novel interventions to prevent/delay age-related vascular pathologies by targeting fundamental cellular and molecular aging processes are presented.
Collapse
Affiliation(s)
- Zoltan Ungvari
- From the Vascular Cognitive Impairment Laboratory, Reynolds Oklahoma Center on Aging (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Geriatric Medicine, Translational Geroscience Laboratory (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Medical Physics and Informatics, University of Szeged, Hungary (Z.U., A.C.)
- Department of Pulmonology, Semmelweis University of Medicine, Budapest, Hungary (Z.U.)
| | - Stefano Tarantini
- From the Vascular Cognitive Impairment Laboratory, Reynolds Oklahoma Center on Aging (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Geriatric Medicine, Translational Geroscience Laboratory (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
| | - Anthony J Donato
- Division of Geriatrics, Department of Internal Medicine, University of Utah, Salt Lake City (A.J.D.)
- Veterans Affairs Medical Center-Salt Lake City, Geriatrics Research Education and Clinical Center, UT (A.J.D.)
| | - Veronica Galvan
- Barshop Institute for Longevity and Aging Studies (V.G.), University of Texas Health Science Center at San Antonio
- Department of Physiology (V.G.), University of Texas Health Science Center at San Antonio
| | - Anna Csiszar
- From the Vascular Cognitive Impairment Laboratory, Reynolds Oklahoma Center on Aging (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Geriatric Medicine, Translational Geroscience Laboratory (Z.U., S.T., A.C.), University of Oklahoma Health Sciences Center, Oklahoma City
- Department of Medical Physics and Informatics, University of Szeged, Hungary (Z.U., A.C.)
| |
Collapse
|
138
|
Chen YF, Dugas TR. Endothelial mitochondrial senescence accelerates cardiovascular disease in antiretroviral-receiving HIV patients. Toxicol Lett 2019; 317:13-23. [PMID: 31562912 DOI: 10.1016/j.toxlet.2019.09.018] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 09/12/2019] [Accepted: 09/21/2019] [Indexed: 02/06/2023]
Abstract
Combination antiretroviral therapy (cART) has been hugely successful in reducing the mortality associated with human immunodeficiency virus (HIV) infection, resulting in a growing population of people living with HIV (PLWH). Since PLWH now have a longer life expectancy, chronic comorbidities have become the focus of the clinical management of HIV. For example, cardiovascular complications are now one of the most prevalent causes of death in PLWH. Numerous epidemiological studies show that antiretroviral treatment increases cardiovascular disease (CVD) risk and early onset of CVD in PLWH. Nucleoside reverse transcriptase inhibitors (NRTIs) are the backbone of cART, and two NRTIs are typically used in combination with one drug from another drug class, e.g., a fusion inhibitor. NRTIs are known to induce mitochondrial dysfunction, contributing to toxicity in numerous tissues, such as myopathy, lipoatrophy, neuropathy, and nephropathy. In in vitro studies, short-term NRTI treatment induces an endothelial dysfunction with an increased reactive oxygen species (ROS) production; long-term NRTI treatment decreases cell replication capacity, while increasing mtROS production and senescent cell accumulation. These findings suggest that a mitochondrial oxidative stress is involved in the pathogenesis of NRTI-induced endothelial dysfunction and premature senescence. Mitochondrial dysfunction, defined by a compromised mitochondrial quality control via biogenesis and mitophagy, has a causal role in premature endothelial senescence and can potentially initiate early cardiovascular disease (CVD) development in PLWH. In this review, we explore the hypothesis and present literature supporting that long-term NRTI treatment induces vascular dysfunction by interfering with endothelial mitochondrial homeostasis and provoking mitochondrial genomic instability, resulting in premature endothelial senescence.
Collapse
Affiliation(s)
- Yi-Fan Chen
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, United States
| | - Tammy R Dugas
- Comparative Biomedical Sciences, Louisiana State University School of Veterinary Medicine, Skip Bertman Drive, Baton Rouge, LA, 70808, United States.
| |
Collapse
|
139
|
Increased mitochondrial NADPH oxidase 4 (NOX4) expression in aging is a causative factor in aortic stiffening. Redox Biol 2019; 26:101288. [PMID: 31419754 PMCID: PMC6831838 DOI: 10.1016/j.redox.2019.101288] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/09/2019] [Accepted: 07/29/2019] [Indexed: 12/21/2022] Open
Abstract
Aging is characterized by increased aortic stiffness, an early, independent predictor and cause of cardiovascular disease. Oxidative stress from excess reactive oxygen species (ROS) production increases with age. Mitochondria and NADPH oxidases (NOXs) are two major sources of ROS in cardiovascular system. We showed previously that increased mitochondrial ROS levels over a lifetime induce aortic stiffening in a mouse oxidative stress model. Also, NADPH oxidase 4 (NOX4) expression and ROS levels increase with age in aortas, aortic vascular smooth muscle cells (VSMCs) and mitochondria, and are correlated with age-associated aortic stiffness in hypercholesterolemic mice. The present study investigated whether young mice (4 months-old) with increased mitochondrial NOX4 levels recapitulate vascular aging and age-associated aortic stiffness. We generated transgenic mice with low (Nox4TG605; 2.1-fold higher) and high (Nox4TG618; 4.9-fold higher) mitochondrial NOX4 expression. Young Nox4TG618 mice showed significant increase in aortic stiffness and decrease in phenylephrine-induced aortic contraction, but not Nox4TG605 mice. Increased mitochondrial oxidative stress increased intrinsic VSMC stiffness, induced aortic extracellular matrix remodeling and fibrosis, a leftward shift in stress-strain curves, decreased volume compliance and focal adhesion turnover in Nox4TG618 mice. Nox4TG618 VSMCs phenocopied other features of vascular aging such as increased DNA damage, increased premature and replicative senescence and apoptosis, increased proinflammatory protein expression and decreased respiration. Aortic stiffening in young Nox4TG618 mice was significantly blunted with mitochondrial-targeted catalase overexpression. This demonstration of the role of mitochondrial oxidative stress in aortic stiffness will galvanize search for new mitochondrial-targeted therapeutics for treatment of age-associated vascular dysfunction. Aortic stiffness in aging is associated with increased mitochondrial NOX4 levels. Young mitochondrial Nox4 overexpressing transgenic mice phenocopy aortic stiffness. Nox4 transgenic mice show increased VSMC stiffness, aortic remodeling and fibrosis. Nox4 transgenic mouse VSMC show DNA damage, senescence, apoptosis and inflammation. High mitochondrial catalase levels blunt aortic stiffness in Nox4 transgenic mice.
Collapse
|
140
|
Czigler A, Toth L, Szarka N, Berta G, Amrein K, Czeiter E, Lendvai-Emmert D, Bodo K, Tarantini S, Koller A, Ungvari Z, Buki A, Toth P. Hypertension Exacerbates Cerebrovascular Oxidative Stress Induced by Mild Traumatic Brain Injury: Protective Effects of the Mitochondria-Targeted Antioxidative Peptide SS-31. J Neurotrauma 2019; 36:3309-3315. [PMID: 31266393 DOI: 10.1089/neu.2019.6439] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Traumatic brain injury (TBI) induces cerebrovascular oxidative stress, which is associated with neurovascular uncoupling, autoregulatory dysfunction, and persisting cognitive decline in both pre-clinical models and patients. However, single mild TBI (mTBI), the most frequent form of brain trauma, increases cerebral generation of reactive oxygen species (ROS) only transiently. We hypothesized that comorbid conditions might exacerbate long-term ROS generation in cerebral arteries after mTBI. Because hypertension is the most important cerebrovascular risk factor in populations prone to mild brain trauma, we induced mTBI in normotensive and spontaneously hypertensive rats (SHR) and assessed changes in cytoplasmic and mitochondrial superoxide (O2-) production by confocal microscopy in isolated middle cerebral arteries (MCA) 2 weeks after mTBI using dihydroethidine (DHE) and the mitochondria-targeted redox-sensitive fluorescent indicator dye MitoSox. We found that mTBI induced a significant increase in long-term cytoplasmic and mitochondrial O2- production in MCAs of SHRs and increased expression of the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase subunit Nox4, which were reversed to the normal level by treating the animals with the cell-permeable, mitochondria-targeted antioxidant peptide SS-31 (5.7 mg kg-1 day-1, i.p.). Persistent mTBI-induced oxidative stress in MCAs of SHRs was significantly decreased by inhibiting vascular NADPH oxidase (apocyinin). We propose that hypertension- and mTBI-induced cerebrovascular oxidative stress likely lead to persistent dysregulation of cerebral blood flow (CBF) and cognitive dysfunction, which might be reversed by SS-31 treatment.
Collapse
Affiliation(s)
- Andras Czigler
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary
| | - Luca Toth
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary
| | - Nikolett Szarka
- Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary
| | - Gergely Berta
- Medical Biology and University of Pecs, Medical School, Pecs, Hungary
| | - Kriszitina Amrein
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Endre Czeiter
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Immunology and Biotechnology, University of Pecs, Medical School, Pecs, Hungary
| | - Dominika Lendvai-Emmert
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Kornelia Bodo
- Immunology and Biotechnology, University of Pecs, Medical School, Pecs, Hungary
| | - Stefano Tarantini
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Akos Koller
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Department of Morphology and Physiology, Semmelweis University, Budapest, Hungary.,Sport-Physiology Research Center, University of Physical Education, Budapest, Hungary.,Department of Physiology, New York Medical College, Valhalla, New York
| | - Zoltan Ungvari
- Reynolds Oklahoma Center on Aging, Donald W. Reynolds Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Andras Buki
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary
| | - Peter Toth
- Department of Neurosurgery and Szentagothai Research Center, University of Pecs, Medical School, Pecs, Hungary.,Institute for Translational Medicine, Departments of University of Pecs, Medical School, Pecs, Hungary.,MTA-PTE Clinical Neuroscience MR Research Group, Pecs, Hungary
| |
Collapse
|
141
|
Sujkowski A, Spierer AN, Rajagopalan T, Bazzell B, Safdar M, Imsirovic D, Arking R, Rand DM, Wessells R. Mito-nuclear interactions modify Drosophila exercise performance. Mitochondrion 2019; 47:188-205. [PMID: 30408593 PMCID: PMC7035791 DOI: 10.1016/j.mito.2018.11.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 10/19/2018] [Accepted: 11/03/2018] [Indexed: 02/08/2023]
Abstract
Endurance exercise has received increasing attention as a broadly preventative measure against age-related disease and dysfunction. Improvement of mitochondrial quality by enhancement of mitochondrial turnover is thought to be among the important molecular mechanisms underpinning the benefits of exercise. Interactions between the mitochondrial and nuclear genomes are important components of the genetic basis for variation in longevity, fitness and the incidence of disease. Here, we examine the effects of replacing the mitochondrial genome (mtDNA) of several Drosophila strains with mtDNA from other strains, or from closely related species, on exercise performance. We find that mitochondria from flies selected for longevity increase the performance of flies from a parental strain. We also find evidence that mitochondria from other strains or species alter exercise performance, with examples of both beneficial and deleterious effects. These findings suggest that both the mitochondrial and nuclear genomes, as well as interactions between the two, contribute significantly to exercise capacity.
Collapse
Affiliation(s)
- Alyson Sujkowski
- Department of Physiology, Wayne State University, Detroit, MI, United States
| | - Adam N Spierer
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, United States
| | - Thiviya Rajagopalan
- Department of Physiology, Wayne State University, Detroit, MI, United States
| | - Brian Bazzell
- Department of Physiology, Wayne State University, Detroit, MI, United States
| | - Maryam Safdar
- Department of Physiology, Wayne State University, Detroit, MI, United States
| | - Dinko Imsirovic
- Department of Physiology, Wayne State University, Detroit, MI, United States
| | - Robert Arking
- Department of Biological Sciences, Wayne State University, Detroit, MI, United States
| | - David M Rand
- Department of Ecology and Evolutionary Biology, Brown University, Providence, RI, United States
| | - Robert Wessells
- Department of Physiology, Wayne State University, Detroit, MI, United States.
| |
Collapse
|
142
|
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
|
143
|
Caporarello N, Meridew JA, Jones DL, Tan Q, Haak AJ, Choi KM, Manlove LJ, Prakash YS, Tschumperlin DJ, Ligresti G. PGC1α repression in IPF fibroblasts drives a pathologic metabolic, secretory and fibrogenic state. Thorax 2019; 74:749-760. [PMID: 31182654 DOI: 10.1136/thoraxjnl-2019-213064] [Citation(s) in RCA: 61] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/16/2019] [Accepted: 05/18/2019] [Indexed: 12/11/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal ageing-related disease linked to mitochondrial dysfunction. The present study aimed to determine whether peroxisome proliferator activated receptor gamma co-activator 1-alpha (PPARGC1A, encoding PGC1α), a master regulator of mitochondrial biogenesis, is diminished in IPF and controls pathologic fibroblast activation. Primary human IPF, control lung fibroblasts and fibroblasts sorted from bleomycin-injured mice were used to evaluate the expression and function of PGC1α. In vitro PGC1α manipulation was performed by small interfering RNA knockdown or overexpression. Fibroblast activation was assessed by quantitative PCR, Western blotting, matrix deposition, secreted cytokine array, immunofluorescence and traction force microscopy. Mitochondrial function was assessed by Seahorse analyzer and mitochondria mass and number by flow cytometry, mitochondrial DNA quantification and transmission electron microscopy (TEM). We found that PGC1α levels are stably repressed in IPF fibroblasts. After bleomycin injury in young mice, PGC1α expression drops transiently but then increases prior to fibrosis resolution. In contrast, PGC1α expression fails to recover in aged mice with persistent fibrosis. PGC1α knockdown alone in normal human lung fibroblasts reduces mitochondrial mass and function while enhancing contractile and matrix synthetic fibroblast activation, senescence-related gene expression and soluble profibrotic and prosenescence signalling. Re-expression of PGC1α in IPF fibroblasts ameliorates all of these pathological cellular functions. Pharmacological treatment of IPF fibroblasts with rosiglitazone, but not thyroid hormone, elevated PGC1α expression and attenuated fibroblast activation. The sustained repression of PGC1α and beneficial effects of its rescue in IPF fibroblasts identifies PGC1α as an important regulator of the fibroblast's pathological state in IPF.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Jeffrey A Meridew
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Dakota L Jones
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Qi Tan
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Andrew J Haak
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Kyoung M Choi
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Logan J Manlove
- Anesthesiology and Perioperative Medicine, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Y S Prakash
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA.,Anesthesiology and Perioperative Medicine, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Daniel J Tschumperlin
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| | - Giovanni Ligresti
- Physiology & Biomedical Engineering, Mayo Clinic Minnesota, Rochester, Minnesota, USA
| |
Collapse
|
144
|
Ashar FN, Zhang Y, Longchamps RJ, Lane J, Moes A, Grove ML, Mychaleckyj JC, Taylor KD, Coresh J, Rotter JI, Boerwinkle E, Pankratz N, Guallar E, Arking DE. Association of Mitochondrial DNA Copy Number With Cardiovascular Disease. JAMA Cardiol 2019; 2:1247-1255. [PMID: 29049454 DOI: 10.1001/jamacardio.2017.3683] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Importance Mitochondrial dysfunction is a core component of the aging process and may play a key role in atherosclerotic cardiovascular disease. Mitochondrial DNA copy number (mtDNA-CN), which represents the number of mitochondria per cell and number of mitochondrial genomes per mitochondrion, is an indirect biomarker of mitochondrial function. Objective To determine whether mtDNA-CN, measured in an easily accessible tissue (buffy coat/circulating leukocytes), can improve risk classification for cardiovascular disease (CVD) and help guide initiation of statin therapy for primary prevention of CVD. Design, Setting, and Participants Prospective, population-based cohort analysis including 21 870 participants (20 163 free from CVD at baseline) from 3 studies: Cardiovascular Health Study (CHS), Atherosclerosis Risk in Communities Study (ARIC), and Multiethnic Study of Atherosclerosis (MESA). The mean follow-up was 13.5 years. The study included 11 153 participants from ARIC, 4830 from CHS, and 5887 from MESA. Analysis of the data was conducted from March 10, 2014, to January 29, 2017. Exposures Mitochondrial DNA-CN measured from buffy coat/circulating leukocytes. Main Outcomes and Measures Incident CVD, which combines coronary heart disease, defined as the first incident myocardial infarction or death owing to coronary heart disease, and stroke, defined as the first nonfatal stroke or death owing to stroke. Results Of the 21 870 participants, the mean age was 62.4 years (ARIC, 57.9 years; MESA, 62.4 years; and CHS, 72.5 years), and 54.7% of participants were women. The hazard ratios for incident coronary heart disease, stroke, and CVD associated with a 1-SD decrease in mtDNA-CN were 1.29 (95% CI, 1.24-1.33), 1.11 (95% CI, 1.06-1.16), and 1.23 (95% CI, 1.19-1.26). The associations persisted after adjustment for traditional CVD risk factors. Addition of mtDNA-CN to the 2013 American College of Cardiology/American Heart Association Pooled Cohorts Equations for estimating 10-year hard atherosclerosis CVD risk was associated with improved risk classification (continuous net reclassification index, 0.194; 95% CI, 0.130-0.258; P < .001). Mitochondrial DNA-CN further improved sensitivity and specificity for the 2013 American College of Cardiology/American Heart Association recommendations on initiating statin therapy for primary prevention of ASCVD (net 221 individuals appropriately downclassified and net 15 individuals appropriately upclassified). Conclusions and Relevance Mitochondrial DNA-CN was independently associated with incident CVD in 3 large prospective studies and may have potential clinical utility in improving CVD risk classification.
Collapse
Affiliation(s)
- Foram N Ashar
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Yiyi Zhang
- Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Ryan J Longchamps
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John Lane
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Anna Moes
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Megan L Grove
- School of Public Health, Human Genetics Center, The University of Texas Health Science Center at Houston
| | | | - Kent D Taylor
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor, University of California, Los Angeles Medical Center, Torrance
| | - Josef Coresh
- Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Jerome I Rotter
- Institute for Translational Genomics and Population Sciences and Department of Pediatrics, Los Angeles Biomedical Research Institute at Harbor, University of California, Los Angeles Medical Center, Torrance.,Division of Genomic Outcomes, Departments of Pediatrics and Medicine, Harbor, University of California, Los Angeles Medical Center, Torrance
| | - Eric Boerwinkle
- School of Public Health, Human Genetics Center, The University of Texas Health Science Center at Houston
| | - Nathan Pankratz
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis
| | - Eliseo Guallar
- Departments of Epidemiology and Medicine, and Welch Center for Prevention, Epidemiology, and Clinical Research, Johns Hopkins University Bloomberg School of Public Health, Baltimore, Maryland
| | - Dan E Arking
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
145
|
Csiszar A, Tarantini S, Yabluchanskiy A, Balasubramanian P, Kiss T, Farkas E, Baur JA, Ungvari Z. Role of endothelial NAD + deficiency in age-related vascular dysfunction. Am J Physiol Heart Circ Physiol 2019; 316:H1253-H1266. [PMID: 30875255 PMCID: PMC6620681 DOI: 10.1152/ajpheart.00039.2019] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 02/26/2019] [Accepted: 03/12/2019] [Indexed: 12/23/2022]
Abstract
Age-related alterations in endothelium and the resulting vascular dysfunction critically contribute to a range of pathological conditions associated with old age. To develop therapies rationally that improve vascular health and thereby increase health span and life span in older adults, it will be essential to understand the cellular and molecular mechanisms contributing to vascular aging. Preclinical studies in model organisms demonstrate that NAD+ availability decreases with age in multiple tissues and that supplemental NAD+ precursors can ameliorate many age-related cellular impairments. Here, we provide a comprehensive overview of NAD+-dependent pathways [including the NAD+-using silent information regulator-2-like enzymes and poly(ADP-ribose) polymerase enzymes] and the potential consequences of endothelial NAD+ deficiency in vascular aging. The multifaceted vasoprotective effects of treatments that reverse the age-related decline in cellular NAD+ levels, as well as their potential limitations, are discussed. The preventive and therapeutic potential of NAD+ intermediates as effective, clinically relevant interventions in older adults at risk for ischemic heart disease, vascular cognitive impairment, and other common geriatric conditions and diseases that involve vascular pathologies (e.g., sarcopenia, frailty) are critically discussed. We propose that NAD+ precursors [e.g., nicotinamide (Nam) riboside, Nam mononucleotide, niacin] should be considered as critical components of combination therapies to slow the vascular aging process and increase cardiovascular health span.
Collapse
Affiliation(s)
- Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
| | - Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Priya Balasubramanian
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
- Theoretical Medicine Doctoral School, University of Szeged , Szeged , Hungary
| | - Eszter Farkas
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging, Department of Geriatric Medicine, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
- Department of Medical Physics and Informatics, University of Szeged , Szeged , Hungary
- Theoretical Medicine Doctoral School, University of Szeged , Szeged , Hungary
- Department of Pulmonology, Semmelweis University , Budapest , Hungary
- Department of Health Promotion Sciences, Hudson College of Public Health, University of Oklahoma Health Sciences Center , Oklahoma City, Oklahoma
| |
Collapse
|
146
|
Han C, Zhou J, Liu B, Liang C, Pan X, Zhang Y, Zhang Y, Wang Y, Shao L, Zhu B, Wang J, Yin Q, Yu XY, Li Y. Delivery of miR-675 by stem cell-derived exosomes encapsulated in silk fibroin hydrogel prevents aging-induced vascular dysfunction in mouse hindlimb. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 99:322-332. [DOI: 10.1016/j.msec.2019.01.122] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 01/25/2019] [Accepted: 01/26/2019] [Indexed: 01/10/2023]
|
147
|
Bukiya AN. Fetal Cerebral Artery Mitochondrion as Target of Prenatal Alcohol Exposure. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:ijerph16091586. [PMID: 31067632 PMCID: PMC6539770 DOI: 10.3390/ijerph16091586] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 04/29/2019] [Accepted: 05/03/2019] [Indexed: 12/14/2022]
Abstract
Prenatal alcohol exposure results in an array of developmental abnormalities known as fetal alcohol spectrum disorders (FASDs). Despite the high prevalence of FASDs, therapeutic interventions against accidental or intended exposure of developing fetuses to alcohol are limited. This review outlines current knowledge about mitochondria in cerebral blood vessels as a potential target for anti-FASDs intervention. First, it describes the multifaceted role of mitochondria in maintaining the cerebral artery diameter as shown in adult tissue. Second, current literature on alcohol-driven damage of mitochondrial morphology and function in several fetal tissues, including liver, heart, and brain is summarized. The functional consequences of alcohol exposure in these organs include morphological enlargement of mitochondria, increased oxidative stress, and alteration of cellular respiration. These studies point to a tissue-specific effect of alcohol on mitochondrial function and a particular vulnerability of fetal mitochondria to alcohol exposure when compared to adult counterparts. Third, recent work from our group describing persistent changes in fetal baboon cerebral artery proteome following three episodes of prenatal alcohol exposure is reviewed. In conclusion, the consequences of prenatal alcohol exposure on cerebral artery mitochondria constitute an open field of investigation and, eventually, a point of therapeutic intervention against FASDs.
Collapse
Affiliation(s)
- Anna N Bukiya
- Department Pharmacology, College of Medicine, The University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
148
|
Wang XB, Cui NH, Zhang S, Liu ZJ, Ma JF, Ming L. Leukocyte telomere length, mitochondrial DNA copy number, and coronary artery disease risk and severity: A two-stage case-control study of 3064 Chinese subjects. Atherosclerosis 2019; 284:165-172. [DOI: 10.1016/j.atherosclerosis.2019.03.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Revised: 03/03/2019] [Accepted: 03/12/2019] [Indexed: 01/29/2023]
|
149
|
Tarantini S, Valcarcel-Ares MN, Toth P, Yabluchanskiy A, Tucsek Z, Kiss T, Hertelendy P, Kinter M, Ballabh P, Süle Z, Farkas E, Baur JA, Sinclair DA, Csiszar A, Ungvari Z. Nicotinamide mononucleotide (NMN) supplementation rescues cerebromicrovascular endothelial function and neurovascular coupling responses and improves cognitive function in aged mice. Redox Biol 2019; 24:101192. [PMID: 31015147 PMCID: PMC6477631 DOI: 10.1016/j.redox.2019.101192] [Citation(s) in RCA: 216] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/02/2019] [Accepted: 04/07/2019] [Indexed: 01/17/2023] Open
Abstract
Adjustment of cerebral blood flow (CBF) to neuronal activity via neurovascular coupling (NVC) has an essential role in maintenance of healthy cognitive function. In aging increased oxidative stress and cerebromicrovascular endothelial dysfunction impair NVC, contributing to cognitive decline. There is increasing evidence showing that a decrease in NAD+ availability with age plays a critical role in a range of age-related cellular impairments but its role in impaired NVC responses remains unexplored. The present study was designed to test the hypothesis that restoring NAD+ concentration may exert beneficial effects on NVC responses in aging. To test this hypothesis 24-month-old C57BL/6 mice were treated with nicotinamide mononucleotide (NMN), a key NAD+ intermediate, for 2 weeks. NVC was assessed by measuring CBF responses (laser Doppler flowmetry) evoked by contralateral whisker stimulation. We found that NVC responses were significantly impaired in aged mice. NMN supplementation rescued NVC responses by increasing endothelial NO-mediated vasodilation, which was associated with significantly improved spatial working memory and gait coordination. These findings are paralleled by the sirtuin-dependent protective effects of NMN on mitochondrial production of reactive oxygen species and mitochondrial bioenergetics in cultured cerebromicrovascular endothelial cells derived from aged animals. Thus, a decrease in NAD+ availability contributes to age-related cerebromicrovascular dysfunction, exacerbating cognitive decline. The cerebromicrovascular protective effects of NMN highlight the preventive and therapeutic potential of NAD+ intermediates as effective interventions in patients at risk for vascular cognitive impairment (VCI).
Collapse
Affiliation(s)
- Stefano Tarantini
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Marta Noa Valcarcel-Ares
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Toth
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Neurosurgery, Medical School, University of Pecs, Hungary
| | - Andriy Yabluchanskiy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Zsuzsanna Tucsek
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Tamas Kiss
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Hertelendy
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Michael Kinter
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, 73104, USA
| | - Praveen Ballabh
- Division of Neonatology, Department of Pediatrics, Albert Einstein College of Medicine, USA
| | - Zoltán Süle
- Department of Anatomy, University of Szeged, Szeged, Hungary
| | - Eszter Farkas
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Joseph A Baur
- Department of Physiology and Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Anna Csiszar
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment and Neurodegeneration Program, Reynolds Oklahoma Center on Aging/Department of Geriatric Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary; Department of Public Health, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
150
|
Stem Cell-Derived Exosomes Prevent Aging-Induced Cardiac Dysfunction through a Novel Exosome/lncRNA MALAT1/NF- κB/TNF- α Signaling Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9739258. [PMID: 31089420 PMCID: PMC6476062 DOI: 10.1155/2019/9739258] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/12/2018] [Indexed: 12/22/2022]
Abstract
Aging is a risk factor for cardiovascular disease, and there is no effective therapeutic approach to alleviate this condition. NF-κB and TNF-α have been implicated in the activation of the aging process, but the signaling molecules responsible for the inactivation of NF-κB and TNF-α remain unknown. Exosomes have been reported to improve heart functions by releasing miRNA. Recent studies suggest that lncRNAs are more tissue-specific and developmental stage-specific compared to miRNA. However, the role of lncRNA in exosome-mediated cardiac repair has not been explored. In the present study, we focused on metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), which is an lncRNA associated with cell senescence. We discovered that human umbilical cord mesenchymal stem cell- (UMSC-) derived exosomes prevent aging-induced cardiac dysfunction. Silencer RNA against lncRNA MALAT1 blocked the beneficial effects of exosomes. In summary, we discovered that UMSC-derived exosomes prevent aging-induced cardiac dysfunction by releasing novel lncRNA MALAT1, which in turn inhibits the NF-κB/TNF-α signaling pathway. These findings will lead to the development of therapies that delay aging and progression of age-related diseases.
Collapse
|