1
|
Vue Z, Garza‐Lopez E, Neikirk K, Katti P, Vang L, Beasley H, Shao J, Marshall AG, Crabtree A, Murphy AC, Jenkins BC, Prasad P, Evans C, Taylor B, Mungai M, Killion M, Stephens D, Christensen TA, Lam J, Rodriguez B, Phillips MA, Daneshgar N, Koh H, Koh A, Davis J, Devine N, Saleem M, Scudese E, Arnold KR, Vanessa Chavarin V, Daniel Robinson R, Chakraborty M, Gaddy JA, Sweetwyne MT, Wilson G, Zaganjor E, Kezos J, Dondi C, Reddy AK, Glancy B, Kirabo A, Quintana AM, Dai D, Ocorr K, Murray SA, Damo SM, Exil V, Riggs B, Mobley BC, Gomez JA, McReynolds MR, Hinton A. 3D reconstruction of murine mitochondria reveals changes in structure during aging linked to the MICOS complex. Aging Cell 2023; 22:e14009. [PMID: 37960952 PMCID: PMC10726809 DOI: 10.1111/acel.14009] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/01/2023] [Accepted: 09/19/2023] [Indexed: 11/15/2023] Open
Abstract
During aging, muscle gradually undergoes sarcopenia, the loss of function associated with loss of mass, strength, endurance, and oxidative capacity. However, the 3D structural alterations of mitochondria associated with aging in skeletal muscle and cardiac tissues are not well described. Although mitochondrial aging is associated with decreased mitochondrial capacity, the genes responsible for the morphological changes in mitochondria during aging are poorly characterized. We measured changes in mitochondrial morphology in aged murine gastrocnemius, soleus, and cardiac tissues using serial block-face scanning electron microscopy and 3D reconstructions. We also used reverse transcriptase-quantitative PCR, transmission electron microscopy quantification, Seahorse analysis, and metabolomics and lipidomics to measure changes in mitochondrial morphology and function after loss of mitochondria contact site and cristae organizing system (MICOS) complex genes, Chchd3, Chchd6, and Mitofilin. We identified significant changes in mitochondrial size in aged murine gastrocnemius, soleus, and cardiac tissues. We found that both age-related loss of the MICOS complex and knockouts of MICOS genes in mice altered mitochondrial morphology. Given the critical role of mitochondria in maintaining cellular metabolism, we characterized the metabolomes and lipidomes of young and aged mouse tissues, which showed profound alterations consistent with changes in membrane integrity, supporting our observations of age-related changes in muscle tissues. We found a relationship between changes in the MICOS complex and aging. Thus, it is important to understand the mechanisms that underlie the tissue-dependent 3D mitochondrial phenotypic changes that occur in aging and the evolutionary conservation of these mechanisms between Drosophila and mammals.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | | | - Kit Neikirk
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of HealthMarylandBethesdaUSA
| | - Larry Vang
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Heather Beasley
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Jianqiang Shao
- Central Microscopy Research FacilityUniversity of IowaIowaIowa CityUSA
| | - Andrea G. Marshall
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Amber Crabtree
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Alexandria C. Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Brenita C. Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Chantell Evans
- Department of Cell BiologyDuke University School of MedicineNorth CarolinaDurhamUSA
| | - Brittany Taylor
- J. Crayton Pruitt Family Department of Biomedical EngineeringUniversity of FloridaFloridaGainesvilleUSA
| | - Margaret Mungai
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Mason Killion
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Dominique Stephens
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | | | - Jacob Lam
- Department of Internal MedicineUniversity of IowaIowaIowa CityUSA
| | | | - Mark A. Phillips
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Nastaran Daneshgar
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Ho‐Jin Koh
- Department of Biological SciencesTennessee State UniversityTennesseeNashvilleUSA
| | - Alice Koh
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Jamaine Davis
- Department of Biochemistry, Cancer Biology, Neuroscience, and PharmacologyMeharry Medical CollegeTennesseeNashvilleUSA
| | - Nina Devine
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Mohammad Saleem
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO)Rio de JaneiroBrazil
- Sport Sciences and Exercise Laboratory (LaCEE)Catholic University of Petrópolis (UCP)PetrópolisState of Rio de JaneiroBrazil
| | - Kenneth Ryan Arnold
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | - Valeria Vanessa Chavarin
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | - Ryan Daniel Robinson
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | | | - Jennifer A. Gaddy
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
- Department of Medicine Health and SocietyVanderbilt UniversityTennesseeNashvilleUSA
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterTennesseeNashvilleUSA
- Department of Veterans AffairsTennessee Valley Healthcare SystemsTennesseeNashvilleUSA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and PathologyUniversity of WashingtonWashingtonSeattleUSA
| | - Genesis Wilson
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Elma Zaganjor
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - James Kezos
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | - Cristiana Dondi
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | | | - Brian Glancy
- National Heart, Lung and Blood Institute, National Institutes of HealthMarylandBethesdaUSA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of HealthMarylandBethesdaUSA
| | - Annet Kirabo
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research CenterUniversity of Texas at El PasoTexasEl PasoUSA
| | - Dao‐Fu Dai
- Department of PathologyUniversity of Johns Hopkins School of MedicineMarylandBaltimoreUSA
| | - Karen Ocorr
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | - Sandra A. Murray
- Department of Cell Biology, School of MedicineUniversity of PittsburghPennsylvaniaPittsburghUSA
| | - Steven M. Damo
- Department of Life and Physical SciencesFisk UniversityTennesseeNashvilleUSA
- Center for Structural BiologyVanderbilt UniversityTennesseeNashvilleUSA
| | - Vernat Exil
- Department of Pediatrics, Carver College of MedicineUniversity of IowaIowaIowa CityUSA
- Department of Pediatrics, Division of CardiologySt. Louis University School of MedicineMissouriSt. LouisUSA
| | - Blake Riggs
- Department of BiologySan Francisco State UniversityCaliforniaSan FranciscoUSA
| | - Bret C. Mobley
- Department of PathologyVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Jose A. Gomez
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Antentor Hinton
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| |
Collapse
|
2
|
Vue Z, Neikirk K, Vang L, Garza-Lopez E, Christensen TA, Shao J, Lam J, Beasley HK, Marshall AG, Crabtree A, Anudokem J, Rodriguez B, Kirk B, Bacevac S, Barongan T, Shao B, Stephens DC, Kabugi K, Koh HJ, Koh A, Evans CS, Taylor B, Reddy AK, Miller-Fleming T, Actkins KV, Zaganjor E, Daneshgar N, Murray SA, Mobley BC, Damo SM, Gaddy JA, Riggs B, Wanjalla C, Kirabo A, McReynolds M, Gomez JA, Phillips MA, Exil V, Dai DF, Hinton A. Three-dimensional mitochondria reconstructions of murine cardiac muscle changes in size across aging. Am J Physiol Heart Circ Physiol 2023; 325:H965-H982. [PMID: 37624101 PMCID: PMC10977873 DOI: 10.1152/ajpheart.00202.2023] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/26/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
With sparse treatment options, cardiac disease remains a significant cause of death among humans. As a person ages, mitochondria breakdown and the heart becomes less efficient. Heart failure is linked to many mitochondria-associated processes, including endoplasmic reticulum stress, mitochondrial bioenergetics, insulin signaling, autophagy, and oxidative stress. The roles of key mitochondrial complexes that dictate the ultrastructure, such as the mitochondrial contact site and cristae organizing system (MICOS), in aging cardiac muscle are poorly understood. To better understand the cause of age-related alteration in mitochondrial structure in cardiac muscle, we used transmission electron microscopy (TEM) and serial block facing-scanning electron microscopy (SBF-SEM) to quantitatively analyze the three-dimensional (3-D) networks in cardiac muscle samples of male mice at aging intervals of 3 mo, 1 yr, and 2 yr. Here, we present the loss of cristae morphology, the inner folds of the mitochondria, across age. In conjunction with this, the three-dimensional (3-D) volume of mitochondria decreased. These findings mimicked observed phenotypes in murine cardiac fibroblasts with CRISPR/Cas9 knockout of Mitofilin, Chchd3, Chchd6 (some members of the MICOS complex), and Opa1, which showed poorer oxidative consumption rate and mitochondria with decreased mitochondrial length and volume. In combination, these data show the need to explore if loss of the MICOS complex in the heart may be involved in age-associated mitochondrial and cristae structural changes.NEW & NOTEWORTHY This article shows how mitochondria in murine cardiac changes, importantly elucidating age-related changes. It also is the first to show that the MICOS complex may play a role in outer membrane mitochondrial structure.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Trace A Christensen
- Microscopy and Cell Analysis Core Facility, Mayo Clinic, Rochester, Minnesota, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, Iowa, United States
| | - Jacob Lam
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Josephs Anudokem
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benjamin Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Serif Bacevac
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Taylor Barongan
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Dominique C Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, United States
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, Tennessee, United States
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Chantell S Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Brittany Taylor
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
| | - Anilkumar K Reddy
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tyne Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Ky'Era V Actkins
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Nastaran Daneshgar
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, United States
| | - Jennifer A Gaddy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Tennessee Valley Healthcare Systems, United States Department of Veterans Affairs, Nashville, Tennessee, United States
| | - Blake Riggs
- Department of Biology at San Francisco State University, San Francisco, California, United States
| | - Celestine Wanjalla
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Melanie McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States
| | - Jose A Gomez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Mark A Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, Oregon, United States
| | - Vernat Exil
- Division of Cardiology, Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri, United States
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Dao-Fu Dai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
3
|
Bhatti JS, Khullar N, Vijayvergiya R, Navik U, Bhatti GK, Reddy PH. Mitochondrial miRNA as epigenomic signatures: Visualizing aging-associated heart diseases through a new lens. Ageing Res Rev 2023; 86:101882. [PMID: 36780957 DOI: 10.1016/j.arr.2023.101882] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 02/13/2023]
Abstract
Aging bears many hard knocks, but heart disorders earn a particular allusion, being the most widespread. Cardiovascular diseases (CVDs) are becoming the biggest concern to mankind due to sundry health conditions directly or indirectly related to heart-linked abnormalities. Scientists know that mitochondria play a critical role in the pathophysiology of cardiac diseases. Both environment and genetics play an essential role in modulating and controlling mitochondrial functions. Even a minor abnormality may prove detrimental to heart function. Advanced age combined with an unhealthy lifestyle can cause most cardiomyocytes to be replaced by fibrotic tissue which upsets the conducting system and leads to arrhythmias. An aging heart encounters far more heart-associated comorbidities than a young heart. Many state-of-the-art technologies and procedures are already being used to prevent and treat heart attacks worldwide. However, it remains a mystery when this heart bomb would explode because it lacks an alarm. This calls for a novel and effective strategy for timely diagnosis and a sure-fire treatment. This review article provides a comprehensive overture of prospective potentials of mitochondrial miRNAs that predict complicated and interconnected pathways concerning heart ailments and signature compilations of relevant miRNAs as biomarkers to plot the role of miRNAs in epigenomics. This article suggests that analysis of DNA methylation patterns in age-associated heart diseases may determine age-impelled biomarkers of heart disease.
Collapse
Affiliation(s)
- Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda, India.
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India.
| | - Rajesh Vijayvergiya
- Department of Cardiology, Post Graduate Institute of Medical Education and Research, Chandigarh, India.
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Bathinda, India.
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali, India.
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Neuroscience & Pharmacology, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Departments of Neurology, School of Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Public Health Department of Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Department of Speech, Language and Hearing Sciences, School Health Professions, Texas Tech University Health Sciences Center, Lubbock, TX, USA; Nutritional Sciences Department, College of Human Sciences, Texas Tech University, Lubbock, TX 79409, USA.
| |
Collapse
|
4
|
Protective roles of MITOL against myocardial senescence and ischemic injury partly via Drp1 regulation. iScience 2022; 25:104582. [PMID: 35789860 PMCID: PMC9249672 DOI: 10.1016/j.isci.2022.104582] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 11/30/2021] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
Abnormal mitochondrial fragmentation by dynamin-related protein1 (Drp1) is associated with the progression of aging-associated heart diseases, including heart failure and myocardial infarction (MI). Here, we report a protective role of outer mitochondrial membrane (OMM)-localized E3 ubiquitin ligase MITOL/MARCH5 against cardiac senescence and MI, partly through Drp1 clearance by OMM-associated degradation (OMMAD). Persistent Drp1 accumulation in cardiomyocyte-specific MITOL conditional-knockout mice induced mitochondrial fragmentation and dysfunction, including reduced ATP production and increased ROS generation, ultimately leading to myocardial senescence and chronic heart failure. Furthermore, ischemic stress-induced acute downregulation of MITOL, which permitted mitochondrial accumulation of Drp1, resulted in mitochondrial fragmentation. Adeno-associated virus-mediated delivery of the MITOL gene to cardiomyocytes ameliorated cardiac dysfunction induced by MI. Our findings suggest that OMMAD activation by MITOL can be a therapeutic target for aging-associated heart diseases, including heart failure and MI. MITOL is essential for maintaining cardiac function partly via Drp1 clearance MITOL deficiency causes cardiac aging partly via Drp1 accumulation Ischemic stress induces a rapid downregulation of MITOL MITOL expression attenuates cardiac dysfunction in acute myocardial infarction
Collapse
|
5
|
Abstract
Sarcopenia is common in aging and in patients with heart failure (HF) who may experience worse outcomes. Patients with muscle wasting are more likely to experience falls and can have serious complications when undergoing cardiac procedures. While intensive nutritional support and exercise rehabilitation can help reverse some of these changes, they are often under-prescribed in a timely manner, and we have limited insights into who would benefit. Mechanistic links between gut microbial metabolites (GMM) have been identified and may contribute to adverse clinical outcomes in patients with cardio-renal diseases and aging. This review will examine the emerging evidence for the influence of the gut microbiome-derived metabolites and notable signaling pathways involved in both sarcopenia and HF, especially those linked to dietary intake and mitochondrial metabolism. This provides a unique opportunity to gain mechanistic and clinical insights into developing novel therapeutic strategies that target these GMM pathways or through tailored nutritional modulation to prevent progressive muscle wasting in elderly patients with heart failure.
Collapse
Affiliation(s)
- Chia-Feng Liu
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, OH 44195, USA
| | - W H Wilson Tang
- Center for Microbiome and Human Health, Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland, OH 44195, USA.,Department of Cardiovascular Medicine, Heart, Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| |
Collapse
|
6
|
Cardioprotective effects of early intervention with sacubitril/valsartan on pressure overloaded rat hearts. Sci Rep 2021; 11:16542. [PMID: 34400686 PMCID: PMC8368201 DOI: 10.1038/s41598-021-95988-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 08/03/2021] [Indexed: 12/11/2022] Open
Abstract
Left ventricular remodeling due to pressure overload is associated with poor prognosis. Sacubitril/valsartan is the first-in-class Angiotensin Receptor Neprilysin Inhibitor and has been demonstrated to have superior beneficial effects in the settings of heart failure. The aim of this study was to determine whether sacubitril/valsartan has cardioprotective effect in the early intervention of pressure overloaded hearts and whether it is superior to valsartan alone. We induced persistent left ventricular pressure overload in rats by ascending aortic constriction surgery and orally administrated sacubitril/valsartan, valsartan, or vehicle one week post operation for 10 weeks. We also determined the effects of sacubitril/valsartan over valsartan on adult ventricular myocytes and fibroblasts that were isolated from healthy rats and treated in culture. We found that early intervention with sacubitril/valsartan is superior to valsartan in reducing pressure overload-induced ventricular fibrosis and in reducing angiotensin II-induced adult ventricular fibroblast activation. While neither sacubitril/valsartan nor valsartan changes cardiac hypertrophy development, early intervention with sacubitril/valsartan protects ventricular myocytes from mitochondrial dysfunction and is superior to valsartan in reducing mitochondrial oxidative stress in response to persistent left ventricular pressure overload. In conclusion, our findings demonstrate that sacubitril/valsartan has a superior cardioprotective effect over valsartan in the early intervention of pressure overloaded hearts, which is independent of the reduction of left ventricular afterload. Our study provides evidence in support of potential benefits of the use of sacubitril/valsartan in patients with resistant hypertension or in patients with severe aortic stenosis.
Collapse
|
7
|
Di Fonso A, Pietrangelo L, D’Onofrio L, Michelucci A, Boncompagni S, Protasi F. Ageing Causes Ultrastructural Modification to Calcium Release Units and Mitochondria in Cardiomyocytes. Int J Mol Sci 2021; 22:8364. [PMID: 34445071 PMCID: PMC8395047 DOI: 10.3390/ijms22168364] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/20/2021] [Accepted: 07/28/2021] [Indexed: 12/12/2022] Open
Abstract
Ageing is associated with an increase in the incidence of heart failure, even if the existence of a real age-related cardiomyopathy remains controversial. Effective contraction and relaxation of cardiomyocytes depend on efficient production of ATP (handled by mitochondria) and on proper Ca2+ supply to myofibrils during excitation-contraction (EC) coupling (handled by Ca2+ release units, CRUs). Here, we analyzed mitochondria and CRUs in hearts of adult (4 months old) and aged (≥24 months old) mice. Analysis by confocal and electron microscopy (CM and EM, respectively) revealed an age-related loss of proper organization and disposition of both mitochondria and EC coupling units: (a) mitochondria are improperly disposed and often damaged (percentage of severely damaged mitochondria: adults 3.5 ± 1.1%; aged 16.5 ± 3.5%); (b) CRUs that are often misoriented (longitudinal) and/or misplaced from the correct position at the Z line. Immunolabeling with antibodies that mark either the SR or T-tubules indicates that in aged cardiomyocytes the sarcotubular system displays an extensive disarray. This disarray could be in part caused by the decreased expression of Cav-3 and JP-2 detected by western blot (WB), two proteins involved in formation of T-tubules and in docking SR to T-tubules in dyads. By WB analysis, we also detected increased levels of 3-NT in whole hearts homogenates of aged mice, a product of nitration of protein tyrosine residues, recognized as marker of oxidative stress. Finally, a detailed EM analysis of CRUs (formed by association of SR with T-tubules) points to ultrastructural modifications, i.e., a decrease in their frequency (adult: 5.1 ± 0.5; aged: 3.9 ± 0.4 n./50 μm2) and size (adult: 362 ± 40 nm; aged: 254 ± 60 nm). The changes in morphology and disposition of mitochondria and CRUs highlighted by our results may underlie an inefficient supply of Ca2+ ions and ATP to the contractile elements, and possibly contribute to cardiac dysfunction in ageing.
Collapse
Affiliation(s)
- Alessia Di Fonso
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Laura Pietrangelo
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Laura D’Onofrio
- IZSAM, Istituto Zooprofilattico Sperimentale dell’Abruzzo e del Molise G. Caporale of Teramo, 64100 Teramo, Italy;
| | - Antonio Michelucci
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
| | - Simona Boncompagni
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| | - Feliciano Protasi
- CAST, Center for Advanced Studies and Technology, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy; (A.D.F.); (A.M.); (S.B.); (F.P.)
- DMSI, Department of Medicine and Aging Sciences, University G. d’Annunzio (Ud’A) of Chieti-Pescara, 66100 Chieti, Italy
| |
Collapse
|
8
|
Brandão SR, Reis-Mendes A, Domingues P, Duarte JA, Bastos ML, Carvalho F, Ferreira R, Costa VM. Exploring the aging effect of the anticancer drugs doxorubicin and mitoxantrone on cardiac mitochondrial proteome using a murine model. Toxicology 2021; 459:152852. [PMID: 34246718 DOI: 10.1016/j.tox.2021.152852] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/31/2022]
Abstract
Current cancer therapies are successfully increasing the lifespan of cancer patients. Nevertheless, cardiotoxicity is a serious chemotherapy-induced adverse side effect. Doxorubicin (DOX) and mitoxantrone (MTX) are cardiotoxic anticancer agents, whose toxicological mechanisms are still to be identified. This study focused on DOX and MTX's cardiac mitochondrial damage and their molecular mechanisms. As a hypothesis, we also sought to compare the cardiac modulation caused by 9 mg/kg of DOX or 6 mg/kg of MTX in young adult mice (3 months old) with old control mice (aged control, 18-20 months old) to determine if DOX- and MTX-induced damage had common links with the aging process. Cardiac homogenates and enriched mitochondrial fractions were prepared from treated and control animals and analyzed by immunoblotting and enzymatic assays. Enriched mitochondrial fractions were also characterized by mass spectrometry-based proteomics. Data obtained showed a decrease in mitochondrial density in young adults treated with DOX or MTX and aged control, as assessed by citrate synthase (CS) activity. Furthermore, aged control had increased expression of the peroxisome proliferator-activated receptor γ coactivator 1 α (PGC1α) and manganese superoxide dismutase (MnSOD). Regarding the enriched mitochondrial fractions, DOX and MTX led to downregulation of proteins related to oxidative phosphorylation, fatty acid oxidation, amino acid metabolic process, and tricarboxylic acid cycle. MTX had a greater impact on malate dehydrogenase (MDH2) and pyruvate dehydrogenase E1 component subunit α (PDHA1). No significant proteomic changes were observed in the enriched mitochondrial fractions of aged control when compared to young control. To conclude, DOX and MTX promoted changes in several mitochondrial-related proteins in young adult mice, but none resembling the aged phenotype.
Collapse
Affiliation(s)
- Sofia Reis Brandão
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal; LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Portugal
| | - Ana Reis-Mendes
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Portugal
| | - Pedro Domingues
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal
| | - José Alberto Duarte
- CIAFEL, Faculty of Sports, University of Porto, Portugal; TOXRUN - Toxicology Research Unit, University Institute of Health Sciences, CESPU, CRL, Gandra, Portugal
| | - Maria Lourdes Bastos
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Portugal
| | - Félix Carvalho
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Portugal
| | - Rita Ferreira
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, Portugal
| | - Vera Marisa Costa
- UCIBIO, REQUIMTE, Laboratory of Toxicology, Department of Biological Sciences, Faculty of Pharmacy, University of Porto, Portugal; Associate Laboratory i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, Portugal.
| |
Collapse
|
9
|
Aged Monkeys Fed a High-Fat/High-Sugar Diet Recapitulate Metabolic Disorders and Cardiac Contractile Dysfunction. J Cardiovasc Transl Res 2021; 14:799-815. [PMID: 33591467 DOI: 10.1007/s12265-021-10105-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Accepted: 01/27/2021] [Indexed: 12/28/2022]
Abstract
Aged nonhuman primate (NHP) models are of great value for studying the pathology of metabolic heart diseases and developing therapeutic strategies. In this study, aged male cynomolgus monkeys were fed a regular diet or a high-fat/high-sugar diet (HFSD) for 8 months. Metabolic disorders were diagnosed by 1H-NMR and serum biochemistry, and cardiac function was evaluated by echocardiography. Our results showed that serum metabolic profiles were altered in aged monkeys fed a HFSD, in line with aortic tissue damage, cardiac remodeling, and contractile dysfunction. This aged monkey model significantly increased expression of proinflammatory cytokines and altered expression and phosphorylation of intracellular signaling proteins in the heart, as compared to aged monkeys on a regular diet. Furthermore, the animals demonstrated increased phosphorylation of cardiac myofilament proteins which are causatively associated with decreased myofilament contractility. We conclude that the aged monkey model fed a HFSD exhibits metabolic disorders and cardiac contractile dysfunction.
Collapse
|
10
|
The mKATP Channels and protein-kinase C Are Involved in the Cardioprotective Effects of Genistein on Estrogen-Deficient Rat Hearts Exposed to Ischemia/Reperfusion: Energetic Study. J Cardiovasc Pharmacol 2020; 75:460-474. [PMID: 32195757 DOI: 10.1097/fjc.0000000000000816] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Estrogenic deficiency is considered a risk of coronary disease in women. The phytoestrogen genistein could be a safe preventive strategy. The first aim of this work was to validate a model of cardiac stunning in which natural estrogenic deficiency rats, ie, adult young male (YM) and aged female (AgF), are compared with young female rats (YF). The second aim was to study whether the in vivo administration of genistein prevents the stunning in estrogenic deficiency rats. The third aim was to evaluate whether in our estrogenic deficiency model exists a synergy between genistein and estradiol. The fourth aim was to characterize the underlying mechanisms of genistein. Stunning was induced by ischemia/reperfusion (I/R) in isolated hearts inside a calorimeter. The left ventricular pressure (P) and total heat rate (Ht) were simultaneously measured, while diastolic contracture and muscle economy (P/Ht) were calculated. During R, P/Ht and P recovered less in AgF and YM than in YF rat hearts. Genistein through i.p. (GST-ip) improved P and P/Ht in AgF and YM, but not in YF. In YM, the cardioprotections of GST-ip and estradiol were synergistic. After ischemia, GST-ip increased SR Ca leak causing diastolic contracture. The GST-ip cardioprotection neither was affected by blockade of PI3K-Akt, NO synthases, or phosphatases, but it was sensitive to blockade of protein-kinase C and mKATP channels. Results suggest that (1) estrogenic deficiency worsens cardiac stunning, (2) GST-ip was more cardioprotective in estrogenic deficiency and synergistic with estradiol, and (3) cardioprotection of GST-ip depends on the protein-kinase C and mKATP channel pathway activation.
Collapse
|
11
|
Wang M, Scott SR, Koniaris LG, Zimmers TA. Pathological Responses of Cardiac Mitochondria to Burn Trauma. Int J Mol Sci 2020; 21:ijms21186655. [PMID: 32932869 PMCID: PMC7554938 DOI: 10.3390/ijms21186655] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/04/2020] [Accepted: 09/08/2020] [Indexed: 12/18/2022] Open
Abstract
Despite advances in treatment and care, burn trauma remains the fourth most common type of traumatic injury. Burn-induced cardiac failure is a key factor for patient mortality, especially during the initial post-burn period (the first 24 to 48 h). Mitochondria, among the most important subcellular organelles in cardiomyocytes, are a central player in determining the severity of myocardial damage. Defects in mitochondrial function and structure are involved in pathogenesis of numerous myocardial injuries and cardiovascular diseases. In this article, we comprehensively review the current findings on cardiac mitochondrial pathological changes and summarize burn-impaired mitochondrial respiration capacity and energy supply, induced mitochondrial oxidative stress, and increased cell death. The molecular mechanisms underlying these alterations are discussed, along with the possible influence of other biological variables. We hope this review will provide useful information to explore potential therapeutic approaches that target mitochondria for cardiac protection following burn injury.
Collapse
Affiliation(s)
- Meijing Wang
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
- Correspondence:
| | - Susan R. Scott
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
| | - Leonidas G. Koniaris
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
- Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianopolis, IN 46202, USA
- Center for Cachexia Research Innovation and Therapy, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| | - Teresa A. Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; (S.R.S.); (L.G.K.); (T.A.Z.)
- Simon Cancer Center, Indiana University, Indianapolis, IN 46202, USA
- Indiana Center for Musculoskeletal Health, Indianopolis, IN 46202, USA
- Center for Cachexia Research Innovation and Therapy, Indiana University Purdue University Indianapolis, Indianapolis, IN 46202, USA
| |
Collapse
|
12
|
Castillo EC, Vázquez-Garza E, Yee-Trejo D, García-Rivas G, Torre-Amione G. What Is the Role of the Inflammation in the Pathogenesis of Heart Failure? Curr Cardiol Rep 2020; 22:139. [PMID: 32910299 PMCID: PMC7481763 DOI: 10.1007/s11886-020-01382-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE OF REVIEW In heart failure, whether it is associated with reduced or preserved ejection fraction, the immune system is activated and contributes to heart remodeling and impaired function. RECENT FINDINGS Studies indicate that cells of the immune system not only play a role in the pathology but are also critical regulators of heart function. Knowledge about the role of the immune system driving heart failure will lead to the development of new targets to this system, particularly in those patients that, despite the apparent wellness, relapse and worsen. In this review, we will address the diverse mechanisms that trigger inflammation and their impact on heart failure progression.
Collapse
Affiliation(s)
- Elena C. Castillo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
| | - Eduardo Vázquez-Garza
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
| | - David Yee-Trejo
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, 66278 San Pedro Garza García, NL Mexico
- Tecnologico de Monterrey, Centro de Medicina Funcional, Hospital Zambrano Hellion, TecSalud, 66278 San Pedro Garzar García, NL Mexico
| | - Guillermo Torre-Amione
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Ave. Morones Prieto 3000, 64710 Monterrey, NL Mexico
- Tecnologico de Monterrey, Centro de Investigación Biomédica, Hospital Zambrano Hellion, TecSalud, 66278 San Pedro Garza García, NL Mexico
- De Bakey CRC, The Methodist Hospital, Cornell University, Houston, TX USA
| |
Collapse
|
13
|
Jamieson KL, Keshavarz-Bahaghighat H, Darwesh AM, Sosnowski DK, Seubert JM. Age and Sex Differences in Hearts of Soluble Epoxide Hydrolase Null Mice. Front Physiol 2020; 11:48. [PMID: 32116760 PMCID: PMC7019103 DOI: 10.3389/fphys.2020.00048] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 01/21/2020] [Indexed: 12/19/2022] Open
Abstract
Biological aging is an inevitable part of life that has intrigued individuals for millennia. The progressive decline in biological systems impacts cardiac function and increases vulnerability to stress contributing to morbidity and mortality in aged individuals. Yet, our understanding of the molecular, biochemical and physiological mechanisms of aging as well as sex differences is limited. There is growing evidence indicating CYP450 epoxygenase-mediated metabolites of n-3 and n-6 polyunsaturated fatty acids (PUFAs) are active lipid mediators regulating cardiac homeostasis. These epoxy metabolites are rapidly hydrolyzed and inactivated by the soluble epoxide hydrolase (sEH). The current study characterized cardiac function in young and aged sEH null mice compared to the corresponding wild-type (WT) mice. All aged mice had significantly increased cardiac hypertrophy, except in aged female sEH null mice. Cardiac function as assessed by echocardiography demonstrated a marked decline in aged WT mice, notably significant decreases in ejection fraction and fractional shortening in both sexes. Interestingly, aged female sEH null mice had preserved systolic function, while aged male sEH null mice had preserved diastolic function compared to aged WT mice. Assessment of cardiac mitochondria demonstrated an increased expression of acetyl Mn-SOD levels that correlated with decreased Sirt-3 activity in aged WT males and females. Conversely, aged sEH null mice had preserved Sirt-3 activity and better mitochondrial ultrastructure compared to WT mice. Consistent with these changes, the activity level of SOD significantly decreased in WT animals but was preserved in aged sEH null animals. Markers of oxidative stress demonstrated age-related increase in protein carbonyl levels in WT and sEH null male mice. Together, these data highlight novel cardiac phenotypes from sEH null mice demonstrating a sexual dimorphic pattern of aging in the heart.
Collapse
Affiliation(s)
- K Lockhart Jamieson
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | | | - Ahmed M Darwesh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Deanna K Sosnowski
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - John M Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada.,Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
14
|
Hamilton S, Terentyev D. Altered Intracellular Calcium Homeostasis and Arrhythmogenesis in the Aged Heart. Int J Mol Sci 2019; 20:ijms20102386. [PMID: 31091723 PMCID: PMC6566636 DOI: 10.3390/ijms20102386] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 04/29/2019] [Accepted: 05/08/2019] [Indexed: 12/17/2022] Open
Abstract
Aging of the heart is associated with a blunted response to sympathetic stimulation, reduced contractility, and increased propensity for arrhythmias, with the risk of sudden cardiac death significantly increased in the elderly population. The altered cardiac structural and functional phenotype, as well as age-associated prevalent comorbidities including hypertension and atherosclerosis, predispose the heart to atrial fibrillation, heart failure, and ventricular tachyarrhythmias. At the cellular level, perturbations in mitochondrial function, excitation-contraction coupling, and calcium homeostasis contribute to this electrical and contractile dysfunction. Major determinants of cardiac contractility are the intracellular release of Ca2+ from the sarcoplasmic reticulum by the ryanodine receptors (RyR2), and the following sequestration of Ca2+ by the sarco/endoplasmic Ca2+-ATPase (SERCa2a). Activity of RyR2 and SERCa2a in myocytes is not only dependent on expression levels and interacting accessory proteins, but on fine-tuned regulation via post-translational modifications. In this paper, we review how aberrant changes in intracellular Ca2+ cycling via these proteins contributes to arrhythmogenesis in the aged heart.
Collapse
Affiliation(s)
- Shanna Hamilton
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| | - Dmitry Terentyev
- Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
- Department of Physiology and Cell Biology, College of Medicine, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
15
|
No MH, Heo JW, Yoo SZ, Jo HS, Park DH, Kang JH, Seo DY, Han J, Kwak HB. Effects of aging on mitochondrial hydrogen peroxide emission and calcium retention capacity in rat heart. J Exerc Rehabil 2018; 14:920-926. [PMID: 30656149 PMCID: PMC6323348 DOI: 10.12965/jer.1836550.275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Accepted: 11/03/2018] [Indexed: 01/04/2023] Open
Abstract
Aging is a risk factor for heart disease and heart failure, which result from a progressive impairment of cardiac functions, including stroke volume, cardiac output, blood flow, and oxygen consumption. Age-related cardiac dysfunction is associated with impaired cardiac structures, such as the loss of myocytes, structural remodeling, altered calcium (Ca2+) handling, and contractile dysfunction. However, the mechanism by which aging affects mitochondrial function in the heart is poorly understood. The purpose of this study was to determine the effects of aging on mitochondrial function in the rat heart. Male Fischer 344 rats were randomly assigned to very young sedentary (VYS, 1 month), young sedentary (YS, 4 months), middle-aged sedentary (MS, 10 months), and old sedentary (OS, 20 months) groups. mitochondrial complex protein levels and mitochondrial function (e.g., mitochondrial hydrogen peroxide (H2O2) emission and Ca2+ retention capacity) were analyzed in the left ventricle. Aging was associated with decreased levels of OXPHOS (oxidative phosphorylation) protein expression of complex I to IV in the function of the electron transport chain. Aging increased the mitochondrial H2O2 emitting potential in the heart. In contrast, mitochondrial Ca2+ retention capacity gradually decreased with age. These data demonstrate that aging impairs mitochondrial function in cardiac muscle, suggesting that mitochondrial dysfunction with aging may be a primary factor for aging-induced cardiac dysfunction in the heart.
Collapse
Affiliation(s)
- Mi-Hyun No
- Department of Kinesiology, Inha University, Incheon, Korea
| | - Jun-Won Heo
- Department of Kinesiology, Inha University, Incheon, Korea
| | - Su-Zi Yoo
- Department of Kinesiology, Inha University, Incheon, Korea
| | - Han-Sam Jo
- Department of Kinesiology, Inha University, Incheon, Korea
| | - Dong-Ho Park
- Department of Kinesiology, Inha University, Incheon, Korea
| | - Ju-Hee Kang
- Department of Pharmacology and Medicinal Toxicology Research Center, Inha University School of Medicine, Incheon, Korea
| | - Dae-Yun Seo
- Department of Physiology and Cardiovascular and Metabolic Disease Center, Inje University School of Medicine, Busan, Korea
| | - Jin Han
- Department of Physiology and Cardiovascular and Metabolic Disease Center, Inje University School of Medicine, Busan, Korea
| | - Hyo-Bum Kwak
- Department of Kinesiology, Inha University, Incheon, Korea
| |
Collapse
|
16
|
Wang Y, Li Y, He C, Gou B, Song M. Mitochondrial regulation of cardiac aging. Biochim Biophys Acta Mol Basis Dis 2018; 1865:1853-1864. [PMID: 30593894 DOI: 10.1016/j.bbadis.2018.12.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 12/03/2018] [Accepted: 12/04/2018] [Indexed: 12/13/2022]
Abstract
Aging is associated with progressive decline in cardiac structure and function. Accumulating evidence in model organisms and humans links cardiac aging to mitochondrial regulation, encompassing a complex interplay of mitochondrial morphology, mitochondrial ROS, mitochondrial DNA mutations, mitochondrial unfolded protein response, nicotinamide adenine dinucleotide levels and sirtuins, as well as mitophagy. This review summarizes the recent discoveries on the mitochondrial regulation of cardiac aging and the possible molecular mechanisms underlying the anti-aging effects, as well as the potential interventions that alleviate aging-related cardiac diseases and attenuate cardiac aging via the regulation of mitochondria.
Collapse
Affiliation(s)
- Yuhan Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Forestry University, Beijing 100083, China
| | - Yujing Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Science and Technology of China, Anhui 230026, China
| | - Chuting He
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Bo Gou
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Moshi Song
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China; Institute of Stem cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
17
|
Perera NCN, Godahewa GI, Nam BH, Park JY, Lee J. Two metalloenzymes from rockfish (Sebastes schligellii): Deciphering their potential involvement in redox homeostasis against oxidative stress. FISH & SHELLFISH IMMUNOLOGY 2018; 80:31-45. [PMID: 29859306 DOI: 10.1016/j.fsi.2018.05.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/21/2018] [Accepted: 05/29/2018] [Indexed: 06/08/2023]
Abstract
Disturbance in the balance between pro-oxidants and anti-oxidants result oxidative stress in aerobic organisms. However, oxidative stress can be inhibited by enzymatic and non-enzymatic defense mechanisms. Superoxide dismutases (SODs) are well-known scavengers of superoxide radicals, and they protect cells by detoxifying hazardous reactive oxygen species. Here, we have identified and characterized two different SODs, CuZnSOD and MnSOD, from black rockfish (RfCuZnSOD and RfMnSOD, respectively). In silico analysis revealed the well-conserved molecular structures comprising all essential properties of CuZnSOD and MnSOD. Phylogenetic analysis revealed that both RfCuZnSOD and RfMnSOD cladded with their fish counterparts. The recombinant RfSOD proteins demonstrated their potential superoxide scavenging abilities through a xanthine oxidase assay. The optimum temperature and pH conditions for both rRfSODs were 25 °C and pH 8, respectively. Moreover, the potential peroxidation function of rRfCuZnSOD was observed in the presence of HCO3-. The highest peroxidation activity was observed at 100 μg/mL of rRfCuZnSOD using the MTT cell viability assay and flow cytometry. The analogous tissue-specific expression profile indicated ubiquitous expression of both RfCuZnSOD and RfMnSOD in selected tissues of healthy juvenile rockfish. An immune challenge experiment illustrated the altered expression profiles of both RfCuZnSOD and RfMnSOD against lipopolysaccharide, Streptococcus iniae, and polyinosinic-polycytidylic acid (poly I:C). Collectively, these results strengthen the general understanding of the structural and functional characteristics of SODs within the host defense system.
Collapse
Affiliation(s)
- N C N Perera
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - G I Godahewa
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Bo-Hye Nam
- Biotechnology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Jung Youn Park
- Biotechnology Research Division, National Institute of Fisheries Science, 408-1 Sirang-ri, Gijang-up, Gijang-gun, Busan, 46083, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea.
| |
Collapse
|
18
|
Whitehead N, Gill JF, Brink M, Handschin C. Moderate Modulation of Cardiac PGC-1α Expression Partially Affects Age-Associated Transcriptional Remodeling of the Heart. Front Physiol 2018; 9:242. [PMID: 29618980 PMCID: PMC5871735 DOI: 10.3389/fphys.2018.00242] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/06/2018] [Indexed: 01/09/2023] Open
Abstract
Aging is associated with a decline in cardiac function due to a decreased myocardial reserve. This adverse cardiac remodeling comprises of a variety of changes, including a reduction in mitochondrial function and a decline in the expression of the peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α), a central regulator of mitochondrial biogenesis and metabolic adaptation in the myocardium. To study the etiological involvement of PGC-1α in cardiac aging, we used mouse models mimicking the modest down- and upregulation of this coactivator in the old and the exercised heart, respectively. Young mice with reduced cardiac expression of PGC-1α recapitulated part of the age-related impairment in mitochondrial gene expression, but otherwise did not aggravate the aging process. Inversely however, moderate overexpression of PGC-1α counteracts numerous key age-related remodeling changes, e.g., by improving blood pressure, age-associated apoptosis, and collagen accumulation, as well as in the expression of many, but not all cardiac genes involved in mitochondrial biogenesis, dynamics, metabolism, calcium handling and contractility. Thus, while the reduction of PGC-1α in the heart is insufficient to cause an aging phenotype, moderate overexpression reduces pathological remodeling of older hearts and could thereby contribute to the beneficial effects of exercise on cardiac function in aging.
Collapse
Affiliation(s)
| | | | - Marijke Brink
- Department of Biomedicine, University of Basel and University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
19
|
Hassanpour SH, Dehghani MA, Karami SZ. Study of respiratory chain dysfunction in heart disease. J Cardiovasc Thorac Res 2018; 10:1-13. [PMID: 29707171 PMCID: PMC5913686 DOI: 10.15171/jcvtr.2018.01] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 12/25/2017] [Indexed: 02/06/2023] Open
Abstract
The relentlessly beating heart has the greatest oxygen consumption of any organ in the body at rest reflecting its huge metabolic turnover and energetic demands. The vast majority of its energy is produced and cycled in form of ATP which stems mainly from oxidative phosphorylation occurring at the respiratory chain in the mitochondria. A part from energy production, the respiratory chain is also the main source of reactive oxygen species and plays a pivotal role in the regulation of oxidative stress. Dysfunction of the respiratory chain is therefore found in most common heart conditions. The pathophysiology of mitochondrial respiratory chain dysfunction in hereditary cardiac mitochondrial disease, the aging heart, in LV hypertrophy and heart failure, and in ischaemia-reperfusion injury is reviewed. We introduce the practicing clinician to the complex physiology of the respiratory chain, highlight its impact on common cardiac disorders and review translational pharmacological and non-pharmacological treatment strategies.
Collapse
Affiliation(s)
| | - Mohammad Amin Dehghani
- Department of Toxicology, School of Pharmacy, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | | |
Collapse
|
20
|
Lee SR, Nilius B, Han J. Gaseous Signaling Molecules in Cardiovascular Function: From Mechanisms to Clinical Translation. Rev Physiol Biochem Pharmacol 2018; 174:81-156. [PMID: 29372329 DOI: 10.1007/112_2017_7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon monoxide (CO), hydrogen sulfide (H2S), and nitric oxide (NO) constitute endogenous gaseous molecules produced by specific enzymes. These gases are chemically simple, but exert multiple effects and act through shared molecular targets to control both physiology and pathophysiology in the cardiovascular system (CVS). The gases act via direct and/or indirect interactions with each other in proteins such as heme-containing enzymes, the mitochondrial respiratory complex, and ion channels, among others. Studies of the major impacts of CO, H2S, and NO on the CVS have revealed their involvement in controlling blood pressure and in reducing cardiac reperfusion injuries, although their functional roles are not limited to these conditions. In this review, the basic aspects of CO, H2S, and NO, including their production and effects on enzymes, mitochondrial respiration and biogenesis, and ion channels are briefly addressed to provide insight into their biology with respect to the CVS. Finally, potential therapeutic applications of CO, H2S, and NO with the CVS are addressed, based on the use of exogenous donors and different types of delivery systems.
Collapse
Affiliation(s)
- Sung Ryul Lee
- Department of Convergence Biomedical Science, Cardiovascular and Metabolic Disease Center, College of Medicine, Inje University, Busan, Republic of Korea
| | - Bernd Nilius
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea.
| |
Collapse
|
21
|
Song M, Franco A, Fleischer JA, Zhang L, Dorn GW. Abrogating Mitochondrial Dynamics in Mouse Hearts Accelerates Mitochondrial Senescence. Cell Metab 2017; 26:872-883.e5. [PMID: 29107503 PMCID: PMC5718956 DOI: 10.1016/j.cmet.2017.09.023] [Citation(s) in RCA: 229] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 04/18/2017] [Accepted: 09/27/2017] [Indexed: 12/30/2022]
Abstract
Mitochondrial fusion and fission are critical to heart health; genetically interrupting either is rapidly lethal. To understand whether it is loss of, or the imbalance between, fusion and fission that underlies observed cardiac phenotypes, we engineered mice in which Mfn-mediated fusion and Drp1-mediated fission could be concomitantly abolished. Compared to fusion-defective Mfn1/Mfn2 cardiac knockout or fission-defective Drp1 cardiac knockout mice, Mfn1/Mfn2/Drp1 cardiac triple-knockout mice survived longer and manifested a unique pathological form of cardiac hypertrophy. Over time, however, combined abrogation of fission and fusion provoked massive progressive mitochondrial accumulation that severely distorted cardiomyocyte sarcomeric architecture. Mitochondrial biogenesis was not responsible for mitochondrial superabundance, whereas mitophagy was suppressed despite impaired mitochondrial proteostasis. Similar but milder defects were observed in aged hearts. Thus, cardiomyopathies linked to dynamic imbalance between fission and fusion are temporarily mitigated by forced mitochondrial adynamism at the cost of compromising mitochondrial quantity control and accelerating mitochondrial senescence.
Collapse
Affiliation(s)
- Moshi Song
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Antonietta Franco
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Julie A Fleischer
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Lihong Zhang
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Gerald W Dorn
- Center for Pharmacogenomics, Department of Internal Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
22
|
Lee SR, Han J. Mitochondrial Mutations in Cardiac Disorders. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 982:81-111. [PMID: 28551783 DOI: 10.1007/978-3-319-55330-6_5] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondria individually encapsulate their own genome, unlike other cellular organelles. Mitochondrial DNA (mtDNA) is a circular, double-stranded, 16,569-base paired DNA containing 37 genes: 13 proteins of the mitochondrial respiratory chain, two ribosomal RNAs (rRNAs; 12S and 16S), and 22 transfer RNAs (tRNAs). The mtDNA is more vulnerable to oxidative modifications compared to nuclear DNA because of its proximity to ROS-producing sites, limited presence of DNA damage repair systems, and continuous replication in the cell. mtDNA mutations can be inherited or sporadic. Simple mtDNA mutations are point mutations, which are frequently found in mitochondrial tRNA loci, causing mischarging of mitochondrial tRNAs or deletion, duplication, or reduction in mtDNA content. Because mtDNA has multiple copies and a specific replication mechanism in cells or tissues, it can be heterogenous, resulting in characteristic phenotypic presentations such as heteroplasmy, genetic drift, and threshold effects. Recent studies have increased the understanding of basic mitochondrial genetics, providing an insight into the correlations between mitochondrial mutations and cardiac manifestations including hypertrophic or dilated cardiomyopathy, arrhythmia, autonomic nervous system dysfunction, heart failure, or sudden cardiac death with a syndromic or non-syndromic phenotype. Clinical manifestations of mitochondrial mutations, which result from structural defects, functional impairment, or both, are increasingly detected but are not clear because of the complex interplay between the mitochondrial and nuclear genomes, even in homoplasmic mitochondrial populations. Additionally, various factors such as individual susceptibility, nutritional state, and exposure to chemicals can influence phenotypic presentation, even for the same mtDNA mutation.In this chapter, we summarize our current understanding of mtDNA mutations and their role in cardiac involvement. In addition, epigenetic modifications of mtDNA are briefly discussed for future elucidation of their critical role in cardiac involvement. Finally, current strategies for dealing with mitochondrial mutations in cardiac disorders are briefly stated.
Collapse
Affiliation(s)
- Sung Ryul Lee
- Department of Integrated Biomedical Science, Cardiovascular and Metabolic Disease Center, College of Medicine, Inje University, Busan, 47392, South Korea
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Cardiovascular and Metabolic Disease Center, Department of Physiology, College of Medicine, Inje University, Busan, 47392, South Korea.
| |
Collapse
|
23
|
El-Sikhry HE, Alsaleh N, Dakarapu R, Falck JR, Seubert JM. Novel Roles of Epoxyeicosanoids in Regulating Cardiac Mitochondria. PLoS One 2016; 11:e0160380. [PMID: 27494529 PMCID: PMC4975494 DOI: 10.1371/journal.pone.0160380] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/18/2016] [Indexed: 11/18/2022] Open
Abstract
Maintenance of a healthy pool of mitochondria is important for the function and survival of terminally differentiated cells such as cardiomyocytes. Epoxyeicosatrienoic acids (EETs) are epoxy lipids derived from metabolism of arachidonic acid by cytochrome P450 epoxygenases. We have previously shown that EETs trigger a protective response limiting mitochondrial dysfunction and reducing cellular death. The aim of this study was to investigate whether EET-mediated effects influence mitochondrial quality in HL-1 cardiac cells during starvation. HL-1 cells were subjected to serum- and amino acid free conditions for 24h. We employed a dual-acting synthetic analog UA-8 (13-(3-propylureido)tridec-8-enoic acid), possessing both EET-mimetic and soluble epoxide hydrolase (sEH) inhibitory properties, or 14,15-EET as model EET molecules. We demonstrated that EET-mediated events significantly improved mitochondrial function as assessed by preservation of the ADP/ATP ratio and oxidative respiratory capacity. Starvation induced mitochondrial hyperfusion observed in control cells was attenuated by UA-8. However, EET-mediated events did not affect the expression of mitochondrial dynamic proteins Fis1, DRP-1 or Mfn2. Rather we observed increased levels of OPA-1 oligomers and increased mitochondrial cristae density, which correlated with the preserved mitochondrial function. Increased DNA binding activity of pCREB and Nrf1/2 and increased SIRT1 activity together with elevated mitochondrial proteins suggest EET-mediated events led to preserved mitobiogenesis. Thus, we provide new evidence for EET-mediated events that preserve a healthier pool of mitochondria in cardiac cells following starvation-induced stress.
Collapse
Affiliation(s)
- Haitham E. El-Sikhry
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Nasser Alsaleh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Rambabu Dakarapu
- Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - John R. Falck
- Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- * E-mail:
| |
Collapse
|
24
|
Emelyanova L, Ashary Z, Cosic M, Negmadjanov U, Ross G, Rizvi F, Olet S, Kress D, Sra J, Tajik AJ, Holmuhamedov EL, Shi Y, Jahangir A. Selective downregulation of mitochondrial electron transport chain activity and increased oxidative stress in human atrial fibrillation. Am J Physiol Heart Circ Physiol 2016; 311:H54-63. [PMID: 27199126 PMCID: PMC4967212 DOI: 10.1152/ajpheart.00699.2015] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Accepted: 04/27/2016] [Indexed: 12/19/2022]
Abstract
Mitochondria are critical for maintaining normal cardiac function, and a deficit in mitochondrial energetics can lead to the development of the substrate that promotes atrial fibrillation (AF) and its progression. However, the link between mitochondrial dysfunction and AF in humans is still not fully defined. The aim of this study was to elucidate differences in the functional activity of mitochondrial oxidative phosphorylation (OXPHOS) complexes and oxidative stress in right atrial tissue from patients without (non-AF) and with AF (AF) who were undergoing open-heart surgery and were not significantly different for age, sex, major comorbidities, and medications. The overall functional activity of the electron transport chain (ETC), NADH:O2 oxidoreductase activity, was reduced by 30% in atrial tissue from AF compared with non-AF patients. This was predominantly due to a selective reduction in complex I (0.06 ± 0.007 vs. 0.09 ± 0.006 nmol·min(-1)·citrate synthase activity(-1), P = 0.02) and II (0.11 ± 0.012 vs. 0.16 ± 0.012 nmol·min(-1)·citrate synthase activity(-1), P = 0.003) functional activity in AF patients. Conversely, complex V activity was significantly increased in AF patients (0.21 ± 0.027 vs. 0.12 ± 0.01 nmol·min(-1)·citrate synthase activity(-1), P = 0.005). In addition, AF patients exhibited a higher oxidative stress with increased production of mitochondrial superoxide (73 ± 17 vs. 11 ± 2 arbitrary units, P = 0.03) and 4-hydroxynonenal level (77.64 ± 30.2 vs. 9.83 ± 2.83 ng·mg(-1) protein, P = 0.048). Our findings suggest that AF is associated with selective downregulation of ETC activity and increased oxidative stress that can contribute to the progression of the substrate for AF.
Collapse
Affiliation(s)
- Larisa Emelyanova
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Zain Ashary
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Milanka Cosic
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Ulugbek Negmadjanov
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Gracious Ross
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Farhan Rizvi
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Susan Olet
- Patient-Centered Research, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin; and
| | - David Kress
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Jasbir Sra
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - A Jamil Tajik
- Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Ekhson L Holmuhamedov
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Yang Shi
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| | - Arshad Jahangir
- Sheikh Khalifa bin Hamad Al Thani Center for Integrative Research on Cardiovascular Aging, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin; Patient-Centered Research, Aurora Research Institute, Aurora Health Care, Milwaukee, Wisconsin; and Aurora Cardiovascular Services, Aurora Sinai/Aurora St. Luke's Medical Centers, Milwaukee, Wisconsin
| |
Collapse
|
25
|
Effect of Cumulating Exposure to Abacavir on the Risk of Cardiovascular Disease Events in Patients From the Swiss HIV Cohort Study. J Acquir Immune Defic Syndr 2015; 69:413-21. [PMID: 25932884 DOI: 10.1097/qai.0000000000000662] [Citation(s) in RCA: 65] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Patients with HIV exposed to the antiretroviral drug abacavir may have an increased risk of cardiovascular disease (CVD). There is concern that this association arises because of a channeling bias. Even if exposure is a risk, it is not clear how that risk changes as exposure cumulates. METHODS We assess the effect of exposure to abacavir on the risk of CVD events in the Swiss HIV Cohort Study. We use a new marginal structural Cox model to estimate the effect of abacavir as a flexible function of past exposures while accounting for risk factors that potentially lie on a causal pathway between exposure to abacavir and CVD. RESULTS A total of 11,856 patients were followed for a median of 6.6 years; 365 patients had a CVD event (4.6 events per 1000 patient-years). In a conventional Cox model, recent--but not cumulative--exposure to abacavir increased the risk of a CVD event. In the new marginal structural Cox model, continued exposure to abacavir during the past 4 years increased the risk of a CVD event (hazard ratio = 2.06; 95% confidence interval: 1.43 to 2.98). The estimated function for the effect of past exposures suggests that exposure during the past 6-36 months caused the greatest increase in risk. CONCLUSIONS Abacavir increases the risk of a CVD event: the effect of exposure is not immediate, rather the risk increases as exposure cumulates over the past few years. This gradual increase in risk is not consistent with a rapidly acting mechanism, such as acute inflammation.
Collapse
|
26
|
da Silva Pedroza AA, Lopes A, Mendes da Silva RF, Braz GR, Nascimento LP, Ferreira DS, dos Santos ÂA, Batista-de-Oliveira-Hornsby M, Lagranha CJ. Can fish oil supplementation and physical training improve oxidative metabolism in aged rat hearts? Life Sci 2015; 137:133-41. [DOI: 10.1016/j.lfs.2015.07.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Revised: 07/17/2015] [Accepted: 07/20/2015] [Indexed: 01/17/2023]
|
27
|
Emre E, Ural E, Kahraman G, Bildirici U, Kilic T, Bozyel S, Onuk R, Akbulut T, Ural D. Effect of thrombus aspiration on postprocedural outcomes in elderly patients with acute ST-elevation myocardial ınfarction. Geriatr Gerontol Int 2015; 16:722-8. [PMID: 26246152 DOI: 10.1111/ggi.12545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2015] [Indexed: 11/29/2022]
Abstract
AIM The effects of thrombus aspiration (TA) during primary percutaneous coronary intervention (PCI) for ST-elevation myocardial infarction (STEMI) have been evaluated in several studies. The aim of the present study was to evaluate postprocedural outcomes in elderly STEMI patients who have a tendency for vasoconstruction and decreased coronary flow reserve. METHODS A total of 124 patients (aged ≥65 years) with STEMI who underwent primary PCI (71.2% men, 29.8% women, mean age 74 ± 7 years) were enrolled in the study. Patients were divided into two groups according to intervention with and without TA. Acute angiographic, electrocardiographic and echocardiographic results were compared between the two groups. RESULTS TA was carried out in 42 patients (33.8%). Baseline clinical characteristics and predischarge echocardiographic features did not differ between TA(+) and TA(-) patients (ejection fraction 37.26 ± 8.91 vs 38.53 ± 11.18, P = 0.558, wall motion index 1.69 ± 0.38 vs 1.76 ± 0.37, P = 0.316, septal E' 0.058 ± 0.022 vs 0.053 ± 0.015, P = 0.267, E/E' 11.82 ± 4.30 vs 13.12 ± 5.09, P = 0.370). Acute angiographic and electrocardiographic results did not differ between the two groups, but were slightly better in the thrombectomy group than those without TA corrected TIMI frame count (31.63 ± 16.33 vs 34.97 ± 15.81, P = 0.197, TIMI-3 88.1% vs 79.3%, P = 0.223, ST segment resolution 81.3% vs 70.3%, P = 0.250). CONCLUSIONS Thrombectomy during primary PCI has no effect on postprocedural outcomes in an elderly group with STEMI. Geriatr Gerontol Int 2015; ●●: ●●-●●.
Collapse
Affiliation(s)
- Ender Emre
- Department of Cardiology, Çorlu State Hospital, Tekirdağ, Turkey
| | - Ertan Ural
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Göksel Kahraman
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Ulas Bildirici
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Teoman Kilic
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Serdar Bozyel
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Raşik Onuk
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Tayyar Akbulut
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Dilek Ural
- Department of Cardiology, School of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
28
|
Abstract
SIGNIFICANCE The molecular mechanism of aging is still vigorously debated, although a general consensus exists that mitochondria are significantly involved in this process. However, the previously postulated role of mitochondrial-derived reactive oxygen species (ROS) as the damaging agents inducing functional loss in aging has fallen out of favor in the recent past. In this review, we critically examine the role of ROS in aging in the light of recent advances on the relationship between mitochondrial structure and function. RECENT ADVANCES The functional mitochondrial respiratory chain is now recognized as a reflection of the dynamic association of respiratory complexes in the form of supercomplexes (SCs). Besides providing kinetic advantage (channeling), SCs control ROS generation by the respiratory chain, thus providing a means to regulate ROS levels in the cell. Depending on their concentration, these ROS are either physiological signals essential for the life of the cell or toxic species that damage cell structure and functions. CRITICAL ISSUES We propose that under physiological conditions the dynamic nature of SCs reversibly controls the generation of ROS as signals involved in mitochondrial-nuclear communication. During aging, there is a progressive loss of control of ROS generation so that their production is irreversibly enhanced, inducing a vicious circle in which signaling is altered and structural damage takes place. FUTURE DIRECTIONS A better understanding on the forces affecting SC association would allow the manipulation of ROS generation, directing these species to their physiological signaling role.
Collapse
Affiliation(s)
- Maria Luisa Genova
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| | - Giorgio Lenaz
- Dipartimento di Scienze Biomediche e Neuromotorie, Alma Mater Studiorum-Università di Bologna , Bologna, Italy
| |
Collapse
|
29
|
van Empel VPM, Kaye DM, Borlaug BA. Effects of healthy aging on the cardiopulmonary hemodynamic response to exercise. Am J Cardiol 2014; 114:131-5. [PMID: 24852914 DOI: 10.1016/j.amjcard.2014.04.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 04/03/2014] [Accepted: 04/03/2014] [Indexed: 11/27/2022]
Abstract
This study aimed to define the influence of healthy aging on the central hemodynamic response to exercise. Advancing age results in numerous alterations to the cardiovascular system and is a major risk factor to develop heart failure. In patients with suspected heart failure with preserved ejection fraction, there is an increasing interest in the incorporation of stress hemodynamic studies into the diagnostic evaluation pathway. However, many patients with suspected heart failure with preserved ejection fraction are older, and there are few data regarding the effect of aging on the normal central hemodynamic responses to exercise. Therefore, we examined 55 healthy patients using right-sided cardiac catheterization with exercise. Mean age was 49.6 years, with 36% older than 55 years. On exercise, the mean pulmonary artery pressure was higher with advancing age (r = 0.412, p = 0.002). Additionally, age was negatively associated with cardiac index (r = 0.407, p = 0.005). The exercise-induced rise in pulmonary capillary wedge pressure (r = 0.378, p = 0.004) was greater with advancing age. Pulse pressure measured during exercise (r = 0.541, p <0.01) increased with age, as did diastolic dysfunction assessed by E/A ratio (r = 0.569, p <0.001). In conclusion, aging was associated with decreased cardiac output and increased pulmonary artery pressure during exercise, which developed as the consequence of both increased pulmonary vasculature resistance and higher left ventricular filling pressures.
Collapse
Affiliation(s)
- Vanessa P M van Empel
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia; Department of Cardiology, Maastricht University Medical Centre, Maastricht, The Netherlands
| | - David M Kaye
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia; Department of Cardiovascular Medicine, Alfred Hospital, Melbourne, Australia; Department of Medicine, Monash University, Melbourne, Australia.
| | - Barry A Borlaug
- Division of Cardiovascular Diseases, Department of Medicine, Mayo Clinic and Foundation, Rochester
| |
Collapse
|
30
|
Bitto A, Lerner CA, Nacarelli T, Crowe E, Torres C, Sell C. P62/SQSTM1 at the interface of aging, autophagy, and disease. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9626. [PMID: 24557832 PMCID: PMC4082582 DOI: 10.1007/s11357-014-9626-3] [Citation(s) in RCA: 107] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Accepted: 01/28/2014] [Indexed: 06/02/2023]
Abstract
Advanced age is characterized by increased incidence of many chronic, noninfectious diseases that impair the quality of living of the elderly and pose a major burden on the healthcare systems of developed countries. These diseases are characterized by impaired or altered function at the tissue and cellular level, which is a hallmark of the aging process. Age-related impairments are likely due to loss of homeostasis at the cellular level, which leads to the accumulation of dysfunctional organelles and damaged macromolecules, such as proteins, lipids, and nucleic acids. Intriguingly, aging and age-related diseases can be delayed by modulating nutrient signaling pathways converging on the target of rapamycin (TOR) kinase, either by genetic or dietary intervention. TOR signaling influences aging through several potential mechanisms, such as autophagy, a degradation pathway that clears the dysfunctional organelles and damaged macromolecules that accumulate with aging. Autophagy substrates are targeted for degradation by associating with p62/SQSTM1, a multidomain protein that interacts with the autophagy machinery. p62/SQSTM1 is involved in several cellular processes, and its loss has been linked to accelerated aging and to age-related pathologies. In this review, we describe p62/SQSTM1, its role in autophagy and in signaling pathways, and its emerging role in aging and age-associated pathologies. Finally, we propose p62/SQSTM1 as a novel target for aging studies and age-extending interventions.
Collapse
Affiliation(s)
- Alessandro Bitto
- />Department of Pathology, University of Washington, Health Science Building D-514, Box 357470, Seattle, WA USA
| | | | - Timothy Nacarelli
- />Department of Pathology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102 USA
| | - Elizabeth Crowe
- />Department of Pathology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102 USA
| | - Claudio Torres
- />Department of Pathology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102 USA
| | - Christian Sell
- />Department of Pathology, Drexel University College of Medicine, 245 N 15th Street, Philadelphia, PA 19102 USA
| |
Collapse
|
31
|
Boudina S. Cardiac aging and insulin resistance: could insulin/insulin-like growth factor (IGF) signaling be used as a therapeutic target? Curr Pharm Des 2014; 19:5684-94. [PMID: 23448491 DOI: 10.2174/1381612811319320004] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 02/18/2013] [Indexed: 01/02/2023]
Abstract
Intrinsic cardiac aging is an independent risk factor for cardiovascular disease and is associated with structural and functional changes that impede cardiac responses to stress and to cardio-protective mechanisms. Although systemic insulin resistance and the associated risk factors exacerbate cardiac aging, cardiac-specific insulin resistance without confounding systemic alterations, could prevent cardiac aging. Thus, strategies aimed to reduce insulin/insulin-like growth factor (IGF) signaling in the heart prevent cardiac aging in lower organisms and in mammals but the mechanisms underlying this protection are not fully understood. In this review, we describe the impact of aging on the cardiovascular system and discuss the mounting evidence that reduced insulin/IGF signaling in the heart could alleviate age-associated alterations and preserve cardiac performance.
Collapse
Affiliation(s)
- Sihem Boudina
- Division of Endocrinology, Metabolism and Diabetes, Program in Human Molecular Biology & Genetics, 15 N 2030 E Bldg # 533 Rm. 3410B, Salt Lake City, Utah 84112, USA.
| |
Collapse
|
32
|
Mulligan CM, Le CH, deMooy AB, Nelson CB, Chicco AJ. Inhibition of delta-6 desaturase reverses cardiolipin remodeling and prevents contractile dysfunction in the aged mouse heart without altering mitochondrial respiratory function. J Gerontol A Biol Sci Med Sci 2014; 69:799-809. [PMID: 24418793 DOI: 10.1093/gerona/glt209] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Aging results in a redistribution of polyunsaturated fatty acids (PUFAs) in myocardial phospholipids. In particular, a selective loss of linoleic acid (18:2n6) with reciprocal increases of long-chain PUFAs (eg, arachidonic and docosahexaenoic acids) in the mitochondrial phospholipid cardiolipin correlates with cardiac mitochondrial dysfunction and contractile impairment in aging and related pathologies. In this study, we demonstrate a reversal of this aged-related PUFA redistribution pattern in cardiac mitochondria from aged (25 months) C57Bl/6 mice by inhibition of delta-6 desaturase, the rate limiting enzyme in long-chain PUFA biosynthesis. Interestingly, delta-6 desaturase inhibition had no effect on age-related mitochondrial respiratory dysfunction, H2O2 release, or lipid peroxidation but markedly attenuated cardiac dilatation, hypertrophy, and contractile dysfunction in aged mice. Taken together, our studies indicate that PUFA metabolism strongly influences phospholipid remodeling and cardiac function but dissociates these processes from mitochondrial respiratory dysfunction and oxidant production in the aged mouse heart.
Collapse
|
33
|
Schwarz K, Siddiqi N, Singh S, Neil CJ, Dawson DK, Frenneaux MP. The breathing heart - mitochondrial respiratory chain dysfunction in cardiac disease. Int J Cardiol 2013; 171:134-43. [PMID: 24377708 DOI: 10.1016/j.ijcard.2013.12.014] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 11/04/2013] [Accepted: 12/11/2013] [Indexed: 01/20/2023]
Abstract
The relentlessly beating heart has the greatest oxygen consumption of any organ in the body at rest reflecting its huge metabolic turnover and energetic demands. The vast majority of its energy is produced and cycled in form of ATP which stems mainly from oxidative phosphorylation occurring at the respiratory chain in the mitochondria. Apart from energy production, the respiratory chain is also the main source of reactive oxygen species and plays a pivotal role in the regulation of oxidative stress. Dysfunction of the respiratory chain is therefore found in most common heart conditions. The pathophysiology of mitochondrial respiratory chain dysfunction in hereditary cardiac mitochondrial disease, the ageing heart, in LV hypertrophy and heart failure, and in ischaemia-reperfusion injury is reviewed. We introduce the practising clinician to the complex physiology of the respiratory chain, highlight its impact on common cardiac disorders and review translational pharmacological and non-pharmacological treatment strategies.
Collapse
Affiliation(s)
| | | | | | - Christopher J Neil
- University of Aberdeen, United Kingdom; Western Health, Victoria, Australia
| | | | | |
Collapse
|
34
|
Gao J, Ding XS, Zhang YM, Dai DZ, Liu M, Zhang C, Dai Y. Hypoxia/oxidative stress alters the pharmacokinetics of CPU86017-RS through mitochondrial dysfunction and NADPH oxidase activation. Acta Pharmacol Sin 2013; 34:1575-84. [PMID: 24122013 DOI: 10.1038/aps.2013.94] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Accepted: 06/24/2013] [Indexed: 12/15/2022] Open
Abstract
AIM Hypoxia/oxidative stress can alter the pharmacokinetics (PK) of CPU86017-RS, a novel antiarrhythmic agent. The aim of this study was to investigate the mechanisms underlying the alteration of PK of CPU86017-RS by hypoxia/oxidative stress. METHODS Male SD rats exposed to normal or intermittent hypoxia (10% O2) were administered CPU86017-RS (20, 40 or 80 mg/kg, ig) for 8 consecutive days. The PK parameters of CPU86017-RS were examined on d 8. In a separate set of experiments, female SD rats were injected with isoproterenol (ISO) for 5 consecutive days to induce a stress-related status, then CPU86017-RS (80 mg/kg, ig) was administered, and the tissue distributions were examined. The levels of Mn-SOD (manganese containing superoxide dismutase), endoplasmic reticulum (ER) stress sensor proteins (ATF-6, activating transcription factor 6 and PERK, PRK-like ER kinase) and activation of NADPH oxidase (NOX) were detected with Western blotting. Rat liver microsomes were incubated under N2 for in vitro study. RESULTS The Cmax, t1/2, MRT (mean residence time) and AUC (area under the curve) of CPU86017-RS were significantly increased in the hypoxic rats receiving the 3 different doses of CPU86017-RS. The hypoxia-induced alteration of PK was associated with significantly reduced Mn-SOD level, and increased ATF-6, PERK and NOX levels. In ISO-treated rats, the distributions of CPU86017-RS in plasma, heart, kidney, and liver were markedly increased, and NOX levels in heart, kidney, and liver were significantly upregulated. Co-administration of the NOX blocker apocynin eliminated the abnormalities in the PK and tissue distributions of CPU86017-RS induced by hypoxia/oxidative stress. The metabolism of CPU86017-RS in the N2-treated liver microsomes was significantly reduced, addition of N-acetylcysteine (NAC), but not vitamin C, effectively reversed this change. CONCLUSION The altered PK and metabolism of CPU86017-RS induced by hypoxia/oxidative stress are produced by mitochondrial abnormalities, NOX activation and ER stress; these abnormalities are significantly alleviated by apocynin or NAC.
Collapse
|
35
|
Constant-Urban C, Charif M, Goffin E, Van Heugen JC, Elmoualij B, Chiap P, Mouithys-Mickalad A, Serteyn D, Lebrun P, Pirotte B, De Tullio P. Triphenylphosphonium salts of 1,2,4-benzothiadiazine 1,1-dioxides related to diazoxide targeting mitochondrial ATP-sensitive potassium channels. Bioorg Med Chem Lett 2013; 23:5878-81. [DOI: 10.1016/j.bmcl.2013.08.091] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2013] [Revised: 08/22/2013] [Accepted: 08/26/2013] [Indexed: 01/03/2023]
|
36
|
Samokhvalov V, Alsaleh N, El-Sikhry HE, Jamieson KL, Chen CB, Lopaschuk DG, Carter C, Light PE, Manne R, Falck JR, Seubert JM. Epoxyeicosatrienoic acids protect cardiac cells during starvation by modulating an autophagic response. Cell Death Dis 2013; 4:e885. [PMID: 24157879 PMCID: PMC3920965 DOI: 10.1038/cddis.2013.418] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 09/21/2013] [Accepted: 09/26/2013] [Indexed: 12/25/2022]
Abstract
Epoxyeicosatrienoic acids (EETs) are cytochrome P450 epoxygenase metabolites of arachidonic acid involved in regulating pathways promoting cellular protection. We have previously shown that EETs trigger a protective response limiting mitochondrial dysfunction and reducing cellular death. Considering it is unknown how EETs regulate cell death processes, the major focus of the current study was to investigate their role in the autophagic response of HL-1 cells and neonatal cardiomyocytes (NCMs) during starvation. We employed a dual-acting synthetic analog UA-8 (13-(3-propylureido)tridec-8-enoic acid), possessing both EET-mimetic and soluble epoxide hydrolase (sEH) inhibitory properties, or 14,15-EET as model EET molecules. We demonstrated that EETs significantly improved viability and recovery of starved cardiac cells, whereas they lowered cellular stress responses such as caspase-3 and proteasome activities. Furthermore, treatment with EETs resulted in preservation of mitochondrial functional activity in starved cells. The protective effects of EETs were abolished by autophagy-related gene 7 (Atg7) short hairpin RNA (shRNA) or pharmacological inhibition of autophagy. Mechanistic evidence demonstrated that sarcolemmal ATP-sensitive potassium channels (pmKATP) and enhanced activation of AMP-activated protein kinase (AMPK) played a crucial role in the EET-mediated effect. Our data suggest that the protective effects of EETs involve regulating the autophagic response, which results in a healthier pool of mitochondria in the starved cardiac cells, thereby representing a novel mechanism of promoting survival of cardiac cells. Thus, we provide new evidence highlighting a central role of the autophagic response in linking EETs with promoting cell survival during deep metabolic stress such as starvation.
Collapse
Affiliation(s)
- V Samokhvalov
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Cooper LL, Li W, Lu Y, Centracchio J, Terentyeva R, Koren G, Terentyev D. Redox modification of ryanodine receptors by mitochondria-derived reactive oxygen species contributes to aberrant Ca2+ handling in ageing rabbit hearts. J Physiol 2013; 591:5895-911. [PMID: 24042501 DOI: 10.1113/jphysiol.2013.260521] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Ageing is associated with a blunted response to sympathetic stimulation and an increased risk of arrhythmia and sudden cardiac death. Aberrant calcium (Ca(2+)) handling is an important contributor to the electrical and contractile dysfunction associated with ageing. Yet, the specific molecular mechanisms underlying abnormal Ca(2+) handling in ageing heart remain poorly understood. In this study, we used ventricular myocytes isolated from young (5-9 months) and old (4-6 years) rabbit hearts to test the hypothesis that changes in Ca(2+) homeostasis are caused by post-translational modification of ryanodine receptors (RyRs) by mitochondria-derived reactive oxygen species (ROS) generated in the ageing heart. Changes in parameters of Ca(2+) handling were determined by measuring cytosolic and intra-sarcoplasmic reticulum (SR) Ca(2+) dynamics in intact and permeabilized ventricular myocytes using confocal microscopy. We also measured age-related changes in ROS production and mitochondria membrane potential using a ROS-sensitive dye and a mitochondrial voltage-sensitive fluorescent indicator, respectively. In permeablized myocytes, ageing did not change SERCA activity and spark frequency but decreased spark amplitude and SR Ca(2+) load suggesting increased RyR activity. Treatment with the antioxidant dithiothreitol reduced RyR-mediated SR Ca(2+) leak in permeabilized myocytes from old rabbit hearts to the level comparable to young. Moreover, myocytes from old rabbits had more depolarized mitochondria membrane potential and increased rate of ROS production. Under β-adrenergic stimulation, Ca(2+) transient amplitude, SR Ca(2+) load, and latency of pro-arrhythmic spontaneous Ca(2+) waves (SCWs) were decreased while RyR-mediated SR Ca(2+) leak was increased in cardiomyocytes from old rabbits. Additionally, with β-adrenergic stimulation, scavenging of mitochondrial ROS in myocytes from old rabbit hearts restored redox status of RyRs, which reduced SR Ca(2+) leak, ablated most SCWs, and increased latency to levels comparable to young. These data indicate that an age-associated increase of ROS production by mitochondria leads to the thiol-oxidation of RyRs, which underlies the hyperactivity of RyRs and thereby shortened refractoriness of Ca(2+) release in cardiomyocytes from the ageing heart. This mechanism probably plays an important role in the increased incidence of arrhythmia and sudden death in the ageing population.
Collapse
Affiliation(s)
- Leroy L Cooper
- D. Terentyev: Cardiovascular Research Center, Rhode Island Hospital, Brown University School of Medicine, 1 Hoppin Street, West Coro-5, Providence, RI 02903, USA.
| | | | | | | | | | | | | |
Collapse
|
38
|
Chaudhary KR, Zordoky BNM, Edin ML, Alsaleh N, El-Kadi AOS, Zeldin DC, Seubert JM. Differential effects of soluble epoxide hydrolase inhibition and CYP2J2 overexpression on postischemic cardiac function in aged mice. Prostaglandins Other Lipid Mediat 2012; 104-105:8-17. [PMID: 22922020 DOI: 10.1016/j.prostaglandins.2012.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Revised: 08/01/2012] [Accepted: 08/03/2012] [Indexed: 01/24/2023]
Abstract
Cardioprotective effects of epoxyeicosatrienoic acids (EETs) have been demonstrated in models of young mice with either the cardiomyocyte specific over-expression of cytochrome P450 2J2 (CYP2J2 Tr) or deletion of soluble epoxide hydrolase (sEH null). In this study we examined differences in EET-induced cardioprotection in young (2 months) and aged (12 months) CYP2J2 Tr and sEHnull mice using Langendorff isolated perfused heart model. Improved postischemic functional recovery was observed in both young and aged sEH null mice compared to age matched WT. Conversely, the cardioprotective effect observed in young CYP2J2 Tr was lost in aged CYP2J2 Tr mice. The loss of cardioprotection in aged CYP2J2 Tr was regained following perfusion with the sEH inhibitor t-AUCB. Data demonstrated increased levels of leukotoxin diol (DiHOME) and oxidative stress as well decreased protein phosphatase 2A (PP2A) activation in aged CYP2J2 Tr. In conclusion, inhibition of sEH and EET-induced cardioprotection is maintained in aged mice. However, the loss of protective effects observed in aged CYP2J2 Tr might be attributed to increased levels of DiHOME, oxidative stress and/or decreased PP2A activity.
Collapse
Affiliation(s)
- Ketul R Chaudhary
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | | | | | | | | | | | | |
Collapse
|
39
|
Cardiac arrhythmia and heart failure: From bench to bedside. J Geriatr Cardiol 2012; 8:131-2. [PMID: 22783298 PMCID: PMC3390068 DOI: 10.3724/sp.j.1263.2011.00131] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2011] [Revised: 09/20/2011] [Accepted: 09/27/2011] [Indexed: 11/25/2022] Open
|
40
|
Age-related changes in mitochondrial membrane composition of rainbow trout (Oncorhynchus mykiss) heart and brain. Comp Biochem Physiol B Biochem Mol Biol 2012; 163:129-37. [PMID: 22634369 DOI: 10.1016/j.cbpb.2012.05.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 05/15/2012] [Accepted: 05/15/2012] [Indexed: 12/21/2022]
Abstract
Membrane composition, particularly of mitochondria, could be a critical factor by determining the propagation of reactions involved in mitochondrial function during periods of high oxidative stress such as rapid growth and aging. Considering that phospholipids not only contribute to the structural and physical properties of biological membranes, but also participate actively in cell signaling and apoptosis, changes affecting either class or fatty acid compositions could affect phospholipid properties and, thus, alter mitochondrial function and cell viability. In the present study, heart and brain mitochondrial membrane phospholipid compositions were analyzed in rainbow trout during the four first years of life, a period characterized by rapid growth and a sustained high metabolic rate. Specifically, farmed fish of three ages (1-, 2- and 4-years) were studied, and phospholipid class compositions of heart and brain mitochondria, and fatty acid compositions of individual phospholipid classes were determined. Rainbow trout heart and brain mitochondria showed different phospholipid compositions (class and fatty acid), likely related to tissue-specific functions. Furthermore, changes in phospholipid class and fatty acid compositions with age were also tissue-dependent. Heart mitochondria had lower proportions of cardiolipin (CL), phosphatidylserine (PS) and phosphatidylinositol, and higher levels of phosphatidylcholine (PC) and phosphatidylethanolamine (PE) with age. Heart mitochondrial membranes became more unsaturated with age, with a significative increase of peroxidation index in CL, PS and sphingomyelin (SM). Therefore, heart mitochondria became more susceptible to oxidative damage with age. In contrast, brain mitochondrial PC and PS content decreased in 4-year-old animals while there was an increase in the proportion of SM. The three main phospholipid classes in brain (PC, PE and PS) showed decreased n-3 polyunsaturated fatty acids, docosahexaenoic acid and peroxidation index, which indicate a different response of brain mitochondrial lipids to rapid growth and maturation.
Collapse
|