1
|
Agrawal N, Afzal M, Almalki WH, Ballal S, Sharma GC, Krithiga T, Panigrahi R, Saini S, Ali H, Goyal K, Rana M, Abida Khan. Longevity mechanisms in cardiac aging: exploring calcium dysregulation and senescence. Biogerontology 2025; 26:94. [PMID: 40259024 DOI: 10.1007/s10522-025-10229-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Accepted: 03/20/2025] [Indexed: 04/23/2025]
Abstract
Cardiac aging is a multistep process that results in a loss of various structural and functional heart abilities, increasing the risk of heart disease. Since its remarkable discovery in the early 1800s, when limestone is heated, calcium's importance has been defined in numerous ways. It can help stiffen shells and bones, function as a reducing agent in chemical reactions, and play a central role in cellular signalling. The movement of calcium ions in and out of cells and between those is referred to as calcium signalling. It influences the binding of the ligand, enzyme activity, electrochemical gradients, and other cellular processes. Calcium signalling is critical for both contraction and relaxation under the sliding filament model of heart muscle. However, with age, the heart undergoes changes that lead to increases in cardiac dysfunction, such as myocardial fibrosis, decreased cardiomyocyte function, and noxious disturbances in calcium homeostasis. Additionally, when cardiac tissues age, cellular senescence, a state of irreversible cell cycle arrest, accumulates and begins to exacerbate tissue inflammation and fibrosis. This review explores the most recent discoveries regarding the role of senescent cell accumulation and calcium signalling perturbances in cardiac aging. Additionally, new treatment strategies are used to reduce aged-related heart dysfunction by targeting senescent cells and modulating calcium homeostasis.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, UP, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | - Waleed Hassan Almalki
- Department of Pharmacology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Girish Chandra Sharma
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - T Krithiga
- Department of Chemistry, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Rajashree Panigrahi
- Department of Microbiology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Suman Saini
- Department of Chemistry, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, Uttarakhand, 248007, India
| | - Abida Khan
- Center For Health Research, Northern Border University, Arar 73213, Saudi Arabia
| |
Collapse
|
2
|
Wang M, Hou C, Jia F, Zhong C, Xue C, Li J. Aging-associated atrial fibrillation: A comprehensive review focusing on the potential mechanisms. Aging Cell 2024; 23:e14309. [PMID: 39135295 PMCID: PMC11464128 DOI: 10.1111/acel.14309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 10/11/2024] Open
Abstract
Atrial fibrillation (AF) has been receiving a lot of attention from scientists and clinicians because it is an extremely common clinical condition. Due to its special hemodynamic changes, AF has a high rate of disability and mortality. So far, although AF has some therapeutic means, it is still an incurable disease because of its complex risk factors and pathophysiologic mechanisms, which is a difficult problem for global public health. Age is an important independent risk factor for AF, and the incidence of AF increases with age. To date, there is no comprehensive review on aging-associated AF. In this review, we systematically discuss the pathophysiologic evidence for aging-associated AF, and in particular explore the pathophysiologic mechanisms of mitochondrial dysfunction, telomere attrition, cellular senescence, disabled macroautophagy, and gut dysbiosis involved in recent studies with aging-associated AF. We hope that by exploring the various dimensions of aging-associated AF, we can better understand the specific relationship between age and AF, which may be crucial for innovative treatments of aging-associated AF.
Collapse
Affiliation(s)
- Meng‐Fei Wang
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Can Hou
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Fang Jia
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Cheng‐Hao Zhong
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Cong Xue
- The Third Affiliated Hospital of Soochow UniversityThe First People's Hospital of ChangzhouChangzhouChina
| | - Jian‐Jun Li
- State Key Laboratory of Cardiovascular Diseases, Fu Wai Hospital, National Center for Cardiovascular DiseasesChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
3
|
Ninni S, Algalarrondo V, Brette F, Lemesle G, Fauconnier J. Left atrial cardiomyopathy: Pathophysiological insights, assessment methods and clinical implications. Arch Cardiovasc Dis 2024; 117:283-296. [PMID: 38490844 DOI: 10.1016/j.acvd.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 02/01/2024] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
Atrial cardiomyopathy is defined as any complex of structural, architectural, contractile or electrophysiological changes affecting atria, with the potential to produce clinically relevant manifestations. Most of our knowledge about the mechanistic aspects of atrial cardiomyopathy is derived from studies investigating animal models of atrial fibrillation and atrial tissue samples obtained from individuals who have a history of atrial fibrillation. Several noninvasive tools have been reported to characterize atrial cardiomyopathy in patients, which may be relevant for predicting the risk of incident atrial fibrillation and its related outcomes, such as stroke. Here, we provide an overview of the pathophysiological mechanisms involved in atrial cardiomyopathy, and discuss the complex interplay of these mechanisms, including aging, left atrial pressure overload, metabolic disorders and genetic factors. We discuss clinical tools currently available to characterize atrial cardiomyopathy, including electrocardiograms, cardiac imaging and serum biomarkers. Finally, we discuss the clinical impact of atrial cardiomyopathy, and its potential role for predicting atrial fibrillation, stroke, heart failure and dementia. Overall, this review aims to highlight the critical need for a clinically relevant definition of atrial cardiomyopathy to improve treatment strategies.
Collapse
Affiliation(s)
- Sandro Ninni
- CHU de Lille, Université de Lille, 59000 Lille, France.
| | - Vincent Algalarrondo
- Department of Cardiology, Bichat University Hospital, AP-HP, 75018 Paris, France
| | - Fabien Brette
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | | | - Jérémy Fauconnier
- PhyMedExp, University of Montpellier, INSERM, CNRS, 34093 Montpellier, France
| |
Collapse
|
4
|
Sullivan BP, Collins BC, McMillin SL, Toussaint E, Stein CZ, Spangenburg EE, Lowe DA. Ablation of skeletal muscle estrogen receptor alpha impairs contractility in male mice. J Appl Physiol (1985) 2024; 136:764-773. [PMID: 38328824 PMCID: PMC11286273 DOI: 10.1152/japplphysiol.00714.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Estradiol and estrogen receptor α (ERα) have been shown to be important for the maintenance of skeletal muscle strength in females; however, little is known about the roles of estradiol and ERα in male muscle. The purpose of this study was to determine if skeletal muscle ERα is required for optimal contractility in male mice. We hypothesize that reduced ERα in skeletal muscle impairs contractility in male mice. Skeletal muscle-specific knockout (skmERαKO) male mice exhibited reduced strength across multiple muscles and several contractile parameters related to force generation and kinetics compared with wild-type littermates (skmERαWT). Isolated EDL muscle-specific isometric tetanic force, peak twitch force, peak concentric and peak eccentric forces, as well as the maximal rates of force development and relaxation were 11%-21% lower in skmERαKO compared with skmERαWT mice. In contrast, isolated soleus muscles from skmERαKO mice were not affected. In vivo peak torque of the anterior crural muscles was 20% lower in skmERαKO compared with skmERαWT mice. Muscle masses, contractile protein contents, fiber types, phosphorylation of the myosin regulatory light chain, and caffeine-elicited force did not differ between muscles of skmERαKO and skmERαWT mice, suggesting that strength deficits were not due to size, composition, or calcium release components of muscle contraction. These results indicate that in male mice, reduced skeletal muscle ERα blunts contractility to a magnitude similar to that previously reported in females; however, the mechanism may be sexually dimorphic.NEW & NOTEWORTHY We comprehensively measured in vitro and in vivo contractility of leg muscles with reduced estrogen receptor α (ERα) in male mice and reported that force generation and contraction kinetics are impaired. In contrast to findings in females, phosphorylation of myosin regulatory light chain cannot account for low force production in male skeletal muscle ERα knockout mice. These results indicate that ERα is required for optimal contractility in males and females but via sexually dimorphic means.
Collapse
Affiliation(s)
- Brian P Sullivan
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Brittany C Collins
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Shawna L McMillin
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Elise Toussaint
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Clara Z Stein
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| | - Espen E Spangenburg
- Department of Physiology, Brody School of Medicine, East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, North Carolina, United States
| | - Dawn A Lowe
- Division of Physical Therapy and Rehabilitation Science, Department of Family Medicine and Community Health, University of Minnesota, Minneapolis, Minnesota, United States
| |
Collapse
|
5
|
Linders AN, Dias IB, López Fernández T, Tocchetti CG, Bomer N, Van der Meer P. A review of the pathophysiological mechanisms of doxorubicin-induced cardiotoxicity and aging. NPJ AGING 2024; 10:9. [PMID: 38263284 PMCID: PMC10806194 DOI: 10.1038/s41514-024-00135-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 01/02/2024] [Indexed: 01/25/2024]
Abstract
The population of cancer survivors is rapidly increasing due to improving healthcare. However, cancer therapies often have long-term side effects. One example is cancer therapy-related cardiac dysfunction (CTRCD) caused by doxorubicin: up to 9% of the cancer patients treated with this drug develop heart failure at a later stage. In recent years, doxorubicin-induced cardiotoxicity has been associated with an accelerated aging phenotype and cellular senescence in the heart. In this review we explain the evidence of an accelerated aging phenotype in the doxorubicin-treated heart by comparing it to healthy aged hearts, and shed light on treatment strategies that are proposed in pre-clinical settings. We will discuss the accelerated aging phenotype and the impact it could have in the clinic and future research.
Collapse
Affiliation(s)
- Annet Nicole Linders
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Itamar Braga Dias
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Teresa López Fernández
- Division of Cardiology, Cardiac Imaging and Cardio-Oncology Unit, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Federico II University, Naples, Italy
- Centre for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy
- Interdepartmental Centre of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
- Interdepartmental Hypertension Research Centre (CIRIAPA), Federico II University, Naples, Italy
| | - Nils Bomer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands
| | - Peter Van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Hanzeplein 1, PO Box 30.001, Groningen, The Netherlands.
| |
Collapse
|
6
|
Babini H, Jiménez-Sábado V, Stogova E, Arslanova A, Butt M, Dababneh S, Asghari P, Moore EDW, Claydon TW, Chiamvimonvat N, Hove-Madsen L, Tibbits GF. hiPSC-derived cardiomyocytes as a model to study the role of small-conductance Ca 2+-activated K + (SK) ion channel variants associated with atrial fibrillation. Front Cell Dev Biol 2024; 12:1298007. [PMID: 38304423 PMCID: PMC10830749 DOI: 10.3389/fcell.2024.1298007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 01/05/2024] [Indexed: 02/03/2024] Open
Abstract
Atrial fibrillation (AF), the most common arrhythmia, has been associated with different electrophysiological, molecular, and structural alterations in atrial cardiomyocytes. Therefore, more studies are required to elucidate the genetic and molecular basis of AF. Various genome-wide association studies (GWAS) have strongly associated different single nucleotide polymorphisms (SNPs) with AF. One of these GWAS identified the rs13376333 risk SNP as the most significant one from the 1q21 chromosomal region. The rs13376333 risk SNP is intronic to the KCNN3 gene that encodes for small conductance calcium-activated potassium channels type 3 (SK3). However, the functional electrophysiological effects of this variant are not known. SK channels represent a unique family of K+ channels, primarily regulated by cytosolic Ca2+ concentration, and different studies support their critical role in the regulation of atrial excitability and consequently in the development of arrhythmias like AF. Since different studies have shown that both upregulation and downregulation of SK3 channels can lead to arrhythmias by different mechanisms, an important goal is to elucidate whether the rs13376333 risk SNP is a gain-of-function (GoF) or a loss-of-function (LoF) variant. A better understanding of the functional consequences associated with these SNPs could influence clinical practice guidelines by improving genotype-based risk stratification and personalized treatment. Although research using native human atrial cardiomyocytes and animal models has provided useful insights, each model has its limitations. Therefore, there is a critical need to develop a human-derived model that represents human physiology more accurately than existing animal models. In this context, research with human induced pluripotent stem cells (hiPSC) and subsequent generation of cardiomyocytes derived from hiPSC (hiPSC-CMs) has revealed the underlying causes of various cardiovascular diseases and identified treatment opportunities that were not possible using in vitro or in vivo studies with animal models. Thus, the ability to generate atrial cardiomyocytes and atrial tissue derived from hiPSCs from human/patients with specific genetic diseases, incorporating novel genetic editing tools to generate isogenic controls and organelle-specific reporters, and 3D bioprinting of atrial tissue could be essential to study AF pathophysiological mechanisms. In this review, we will first give an overview of SK-channel function, its role in atrial fibrillation and outline pathophysiological mechanisms of KCNN3 risk SNPs. We will then highlight the advantages of using the hiPSC-CM model to investigate SNPs associated with AF, while addressing limitations and best practices for rigorous hiPSC studies.
Collapse
Affiliation(s)
- Hosna Babini
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Verónica Jiménez-Sábado
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- IIB SANT PAU, and CIBERCV, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ekaterina Stogova
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Alia Arslanova
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Mariam Butt
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | - Saif Dababneh
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Parisa Asghari
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Edwin D. W. Moore
- Department of Cellular and Physiological Sciences, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Thomas W. Claydon
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
| | | | - Leif Hove-Madsen
- IIB SANT PAU, and CIBERCV, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
- Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Glen F. Tibbits
- Cellular and Regenerative Medicine Centre, BC Children’s Hospital Research Institute, Vancouver, BC, Canada
- Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, BC, Canada
- Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
7
|
Ramos-Mondragón R, Lozhkin A, Vendrov AE, Runge MS, Isom LL, Madamanchi NR. NADPH Oxidases and Oxidative Stress in the Pathogenesis of Atrial Fibrillation. Antioxidants (Basel) 2023; 12:1833. [PMID: 37891912 PMCID: PMC10604902 DOI: 10.3390/antiox12101833] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/26/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
Atrial fibrillation (AF) is the most common type of cardiac arrhythmia and its prevalence increases with age. The irregular and rapid contraction of the atria can lead to ineffective blood pumping, local blood stasis, blood clots, ischemic stroke, and heart failure. NADPH oxidases (NOX) and mitochondria are the main sources of reactive oxygen species in the heart, and dysregulated activation of NOX and mitochondrial dysfunction are associated with AF pathogenesis. NOX- and mitochondria-derived oxidative stress contribute to the onset of paroxysmal AF by inducing electrophysiological changes in atrial myocytes and structural remodeling in the atria. Because high atrial activity causes cardiac myocytes to expend extremely high energy to maintain excitation-contraction coupling during persistent AF, mitochondria, the primary energy source, undergo metabolic stress, affecting their morphology, Ca2+ handling, and ATP generation. In this review, we discuss the role of oxidative stress in activating AF-triggered activities, regulating intracellular Ca2+ handling, and functional and anatomical reentry mechanisms, all of which are associated with AF initiation, perpetuation, and progression. Changes in the extracellular matrix, inflammation, ion channel expression and function, myofibril structure, and mitochondrial function occur during the early transitional stages of AF, opening a window of opportunity to target NOX and mitochondria-derived oxidative stress using isoform-specific NOX inhibitors and mitochondrial ROS scavengers, as well as drugs that improve mitochondrial dynamics and metabolism to treat persistent AF and its transition to permanent AF.
Collapse
Affiliation(s)
- Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
| | - Andrey Lozhkin
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Aleksandr E. Vendrov
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Marschall S. Runge
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| | - Lori L. Isom
- Department of Pharmacology, University of Michigan, 1150 West Medical Center Drive, 2301 Medical Science Research Building III, Ann Arbor, MI 48109, USA; (R.R.-M.); (L.L.I.)
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Nageswara R. Madamanchi
- Department of Internal Medicine, Frankel Cardiovascular Center, University of Michigan, Ann Arbor, MI 48019, USA; (A.L.); (A.E.V.); (M.S.R.)
| |
Collapse
|
8
|
Gökçe Y, Danisman B, Akcay G, Derin N, Yaraş N. L-Carnitine improves mechanical responses of cardiomyocytes and restores Ca 2+ homeostasis during aging. Histochem Cell Biol 2023; 160:341-347. [PMID: 37329457 DOI: 10.1007/s00418-023-02215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2023] [Indexed: 06/19/2023]
Abstract
L-Carnitine (β-hydroxy-γ-trimethylaminobutyric acid, LC) is a crucial molecule for the mitochondrial oxidation of fatty acids. It facilitates the transport of long-chain fatty acids into the mitochondrial matrix. The reduction in LC levels during the aging process has been linked to numerous cardiovascular disorders, including contractility dysfunction, and disrupted intracellular Ca2+ homeostasis. The aim of this study was to examine the effects of long-term (7 months) LC administration on cardiomyocyte contraction and intracellular Ca2+ transients ([Ca2+]i) in aging rats. Male albino Wistar rats were randomly assigned to either the control or LC-treated groups. LC (50 mg/kg body weight/day) was dissolved in distilled water and orally administered for a period of 7 months. The control group received distilled water alone. Subsequently, ventricular single cardiomyocytes were isolated, and the contractility and Ca2+ transients were recorded in aging (18 months) rats. This study demonstrates, for the first time, a novel inotropic effect of long-term LC treatment on rat ventricular cardiomyocyte contraction. LC increased cardiomyocyte cell shortening and resting sarcomere length. Furthermore, LC supplementation led to a reduction in resting [Ca2+]i level and an increase in the amplitude of [Ca2+]i transients, indicative of enhanced contraction. Consistent with these results, decay time of Ca2+ transients also decreased significantly in the LC-treated group. The long-term administration of LC may help restore the Ca2+ homeostasis altered during aging and could be used as a cardioprotective medication in cases where myocyte contractility is diminished.
Collapse
Affiliation(s)
- Yasin Gökçe
- Faculty of Medicine, Department of Biophysics, Harran University, Sanliurfa, Turkey.
| | - Betul Danisman
- Faculty of Medicine, Department of Biophysics, Ataturk University, Erzurum, Turkey
| | - Guven Akcay
- Faculty of Medicine, Department of Biophysics, Hitit University, Corum, Turkey
| | - Narin Derin
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| | - Nazmi Yaraş
- Faculty of Medicine, Department of Biophysics, Akdeniz University, Antalya, Turkey
| |
Collapse
|
9
|
Shi S, Mao X, Lv J, Wang Y, Zhang X, Shou X, Zhang B, Li Y, Wu H, Song Q, Hu Y. Qi-Po-Sheng-Mai granule ameliorates Ach-CaCl 2 -induced atrial fibrillation by regulating calcium homeostasis in cardiomyocytes. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 119:155017. [PMID: 37597360 DOI: 10.1016/j.phymed.2023.155017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/15/2023] [Accepted: 08/06/2023] [Indexed: 08/21/2023]
Abstract
BACKGROUND Atrial fibrillation (AF) is one of the most common arrhythmias encountered in clinical settings. Currently, the pathophysiology of AF remains unclear, which severely limits the effectiveness and safety of medical therapies. The Chinese herbal formula Qi-Po-Sheng-Mai Granule (QPSM) has been widely used in China to treat AF. However, its pharmacological and molecular mechanisms remain unknown. PURPOSE The purpose of this study was to investigate the molecular mechanisms and potential targets of QPSM for AF. STUDY DESIGN AND METHODS The AF model was induced by Ach (66 μg/ml) and CaCl2 (10 mg/kg), and the dose of 0.1 ml/100 g was injected into the tail vein for 5 weeks. QPSM was administered daily at doses of 4.42 and 8.84 g/kg, and amiodarone (0.18 g/kg) was used as the positive control. The effect of QPSM on AF was assessed by electrocardiogram, echocardiography, and histopathological analysis. Then, we employed network pharmacology with single nucleus RNA sequencing (snRNA-Seq) to investigate the molecular mechanisms and potential targets of QPSM for AF. Furthermore, high performance liquid chromatography (HPLC) method was used for component analysis of QPSM, and molecular docking was used to verify the potential targets. Using the IonOptix single cell contraction and ion synchronization test equipment, single myocyte length and calcium ion variations were observed in real time. The expression levels of calcium Transporter-related proteins were detected by western blot and immunohistochemistry. RESULTS Based on an Ach-CaCl2-induced AF model, we found that QPSM treatment significantly reduced atrial electrical remodeling-related markers, such as AF inducibility and duration, and attenuated atrial dilation and fibrosis. Network pharmacology identified 52 active ingredients and 119 potential targets for QPSM in the treatment of AF, and 45 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways were enriched, among which calcium pathway had the greatest impact. Using single nucleus sequencing (snRNA-seq), we identified cardiomyocytes as the most differentially expressed in response to drug treatment, with nine differentially expressed genes enriched in calcium signaling pathways. High performance liquid chromatography and molecular docking confirmed that the core components of QPSM strongly bind to the key factors in the calcium signaling pathway. Additional experiments have shown that QPSM increases calcium transients (CaT) and contractility in the individual cardiomyocyte. This was accomplished by increasing the expression of CACNA1C and SERCA2a and decreasing the expression of CAMK2B and NCX1. CONCLUSION The present study has systematically elucidated the role of QPSM in maintaining calcium homeostasis in cardiomyocytes through the regulation of calcium transporters, which could lead to new drug development ideas for AF.
Collapse
Affiliation(s)
- Shuqing Shi
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Xinxin Mao
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Jiayu Lv
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Yajiao Wang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Xuesong Zhang
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xintian Shou
- China Academy of Chinese Medical Sciences, Beijing, China
| | - Bingxuan Zhang
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Yumeng Li
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China
| | - Huaqin Wu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qingqiao Song
- Department of Internal Medicine, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, 5 Beixiange Street Xicheng District, Beijing 100053, China.
| | - Yuanhui Hu
- Department of Cardiovascular, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
10
|
Tarifa C, Jiménez-Sábado V, Franco R, Montiel J, Guerra J, Ciruela F, Hove-Madsen L. Expression and Impact of Adenosine A 3 Receptors on Calcium Homeostasis in Human Right Atrium. Int J Mol Sci 2023; 24:ijms24054404. [PMID: 36901835 PMCID: PMC10003044 DOI: 10.3390/ijms24054404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 02/16/2023] [Accepted: 02/20/2023] [Indexed: 02/25/2023] Open
Abstract
Increased adenosine A2A receptor (A2AR) expression and activation underlies a higher incidence of spontaneous calcium release in atrial fibrillation (AF). Adenosine A3 receptors (A3R) could counteract excessive A2AR activation, but their functional role in the atrium remains elusive, and we therefore aimed to address the impact of A3Rs on intracellular calcium homeostasis. For this purpose, we analyzed right atrial samples or myocytes from 53 patients without AF, using quantitative PCR, patch-clamp technique, immunofluorescent labeling or confocal calcium imaging. A3R mRNA accounted for 9% and A2AR mRNA for 32%. At baseline, A3R inhibition increased the transient inward current (ITI) frequency from 0.28 to 0.81 events/min (p < 0.05). Simultaneous stimulation of A2ARs and A3Rs increased the calcium spark frequency seven-fold (p < 0.001) and the ITI frequency from 0.14 to 0.64 events/min (p < 0.05). Subsequent A3R inhibition caused a strong additional increase in the ITI frequency (to 2.04 events/min; p < 0.01) and increased phosphorylation at s2808 1.7-fold (p < 0.001). These pharmacological treatments had no significant effects on L-type calcium current density or sarcoplasmic reticulum calcium load. In conclusion, A3Rs are expressed and blunt spontaneous calcium release at baseline and upon A2AR-stimulation in human atrial myocytes, pointing to A3R activation as a means to attenuate physiological and pathological elevations of spontaneous calcium release events.
Collapse
Affiliation(s)
- Carmen Tarifa
- Biomedical Research Institute of Barcelona, IIBB-CSIC, 08036 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain
| | - Verónica Jiménez-Sábado
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Rafael Franco
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biología, Universitat de Barcelona, 08028 Barcelona, Spain
| | - José Montiel
- Cardiac Surgery Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
| | - José Guerra
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, 08907 L’Hospitalet de Llobregat, Spain
- Neuropharmacology & Pain Group, Neuroscience Program, Institut d’Investigació Biomèdica de Bellvitge, IDIBELL, 08907 L’Hospitalet de Llobregat, Spain
| | - Leif Hove-Madsen
- Biomedical Research Institute of Barcelona, IIBB-CSIC, 08036 Barcelona, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB Sant Pau), 08025 Barcelona, Spain
- Cardiology Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34-935565620
| |
Collapse
|
11
|
Weinberg SH, King DR. When it comes to the heart, age, and sex matter (sometimes). Am J Physiol Heart Circ Physiol 2023; 324:H226-H228. [PMID: 36607802 PMCID: PMC9886353 DOI: 10.1152/ajpheart.00719.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 12/24/2022] [Indexed: 01/07/2023]
Affiliation(s)
- Seth H Weinberg
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| | - D Ryan King
- Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio
| |
Collapse
|
12
|
Jiménez-Sábado V, Casabella-Ramón S, Llach A, Gich I, Casellas S, Ciruela F, Chen SRW, Guerra JM, Ginel A, Benítez R, Cinca J, Tarifa C, Hove-Madsen L. Beta-blocker treatment of patients with atrial fibrillation attenuates spontaneous calcium release-induced electrical activity. Biomed Pharmacother 2023; 158:114169. [PMID: 36592495 DOI: 10.1016/j.biopha.2022.114169] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/21/2022] [Accepted: 12/28/2022] [Indexed: 01/01/2023] Open
Abstract
AIMS Atrial fibrillation (AF) has been associated with excessive spontaneous calcium release, linked to cyclic AMP (cAMP)-dependent phosphorylation of calcium regulatory proteins. Because β-blockers are expected to attenuate cAMP-dependent signaling, we aimed to examine whether the treatment of patients with β-blockers affected the incidence of spontaneous calcium release events or transient inward currents (ITI). METHODS The impact of treatment with commonly used β-blockers was analyzed in human atrial myocytes from 371 patients using patch-clamp technique, confocal calcium imaging or immunofluorescent labeling. Data were analyzed using multivariate regression analysis taking into account potentially confounding effects of relevant clinical factors RESULTS: The L-type calcium current (ICa) density was diminished significantly in patients with chronic but not paroxysmal AF and the treatment of patients with β-blockers did not affect ICa density in any group. By contrast, the ITI frequency was elevated in patients with either paroxysmal or chronic AF that did not receive treatment, and β-blocker treatment reduced the frequency to levels observed in patients without AF. Confocal calcium imaging showed that β-blocker treatment also reduced the calcium spark frequency in patients with AF to levels observed in those without AF. Furthermore, phosphorylation of the ryanodine receptor (RyR2) at Ser-2808 and phospholamban at Ser-16 was significantly lower in patients with AF that received β-blockers. CONCLUSION Together, our findings demonstrate that β-blocker treatment may be of therapeutic utility to prevent spontaneous calcium release-induced atrial electrical activity; especially in patients with a history of paroxysmal AF displaying preserved ICa density.
Collapse
Affiliation(s)
- Verónica Jiménez-Sábado
- CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Sergi Casabella-Ramón
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain; Department of Cell Biology, Physiology and Immunology and Neuroscience Institute, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anna Llach
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Ignasi Gich
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; CIBER Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain
| | - Sandra Casellas
- Servicio de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Ciruela
- Pharmacology Unit, Dept. Pathology and Experimental Therapeutics, School of Medicine and Health Sciences, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Spain; Neuropharmacology and Pain Group, Neuroscience Program, Institut d'Investigació Biomèdica de Bellvitge, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - S R Wayne Chen
- Department of Physiology and Pharmacology, The Libin Cardiovascular Institute, University of Calgary, Canada
| | - José M Guerra
- CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Servicio de Cardiología and Univ. Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Antonino Ginel
- Servicio de Cirugía Cardíaca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Raúl Benítez
- Dept. d'Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Univ. Politècnica de Catalunya, Barcelona, Spain
| | - Juan Cinca
- Servicio de Cardiología and Univ. Autònoma de Barcelona, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Carmen Tarifa
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain
| | - Leif Hove-Madsen
- CIBER Enfermedades Cardiovasculares (CIBERCV), Instituto de Salud Carlos III, Madrid, Spain; IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Instituto de Investigaciones Biomédicas de Barcelona (IIBB-CSIC), Barcelona, Spain.
| |
Collapse
|
13
|
Tarifa C, Vallmitjana A, Jiménez-Sábado V, Marchena M, Llach A, Herraiz-Martínez A, Godoy-Marín H, Nolla-Colomer C, Ginel A, Viñolas X, Montiel J, Ciruela F, Echebarria B, Benítez R, Cinca J, Hove-Madsen L. Spatial Distribution of Calcium Sparks Determines Their Ability to Induce Afterdepolarizations in Human Atrial Myocytes. JACC. BASIC TO TRANSLATIONAL SCIENCE 2022; 8:1-15. [PMID: 36777175 PMCID: PMC9911326 DOI: 10.1016/j.jacbts.2022.07.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 07/18/2022] [Accepted: 07/18/2022] [Indexed: 11/25/2022]
Abstract
Analysis of the spatio-temporal distribution of calcium sparks showed a preferential increase in sparks near the sarcolemma in atrial myocytes from patients with atrial fibrillation (AF), linked to higher ryanodine receptor (RyR2) phosphorylation at s2808 and lower calsequestrin-2 levels. Mathematical modeling, incorporating modulation of RyR2 gating, showed that only the observed combinations of RyR2 phosphorylation and calsequestrin-2 levels can account for the spatio-temporal distribution of sparks in patients with and without AF. Furthermore, we demonstrate that preferential calcium release near the sarcolemma is key to a higher incidence and amplitude of afterdepolarizations in atrial myocytes from patients with AF.
Collapse
Affiliation(s)
- Carmen Tarifa
- Instituto de Investigaciones Biomédicas de Barcelona, IIBB-CSIC, Barcelona, Spain,IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Alexander Vallmitjana
- Department d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, Spain
| | - Verónica Jiménez-Sábado
- Instituto de Investigaciones Biomédicas de Barcelona, IIBB-CSIC, Barcelona, Spain,IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares, Madrid, Spain
| | - Miquel Marchena
- Department Physics, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Anna Llach
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Adela Herraiz-Martínez
- Instituto de Investigaciones Biomédicas de Barcelona, IIBB-CSIC, Barcelona, Spain,IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Héctor Godoy-Marín
- Department Pathology and Experimental Therapeutics, IDIBELL, University of Barcelona, Barcelona, Spain,Neuroscience Institute, University of Barcelona, Barcelona, Spain
| | - Carme Nolla-Colomer
- Department d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, Spain
| | - Antonino Ginel
- Servicio de Cirugía Cardiaca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Xavier Viñolas
- Servicio de Cardiología, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - José Montiel
- Servicio de Cirugía Cardiaca, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Francisco Ciruela
- Department Pathology and Experimental Therapeutics, IDIBELL, University of Barcelona, Barcelona, Spain,Neuroscience Institute, University of Barcelona, Barcelona, Spain
| | - Blas Echebarria
- Department Physics, Universitat Politècnica de Catalunya, Barcelona, Spain
| | - Raúl Benítez
- Department d’Enginyeria de Sistemes, Automàtica i Informàtica Industrial, Universitat Politècnica de Catalunya, Spain
| | - Juan Cinca
- IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares, Madrid, Spain,Servicio de Cardiología, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Leif Hove-Madsen
- Instituto de Investigaciones Biomédicas de Barcelona, IIBB-CSIC, Barcelona, Spain,IIB Sant Pau, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain,Centro de Investigación Biomédica en Red Enfermedades Cardiovaculares, Madrid, Spain,Address for correspondence: Dr Leif Hove-Madsen, Cardiac Rhythm and Contraction Group, Biomedical Research Institute Barcelona, Hospital de la Santa Creu i Sant Pau, St Antoni Ma Claret 167, 08025 Barcelona, Spain.
| |
Collapse
|
14
|
Peyton MP, Yang TY, Higgins L, Markowski TW, Vue C, Parker LL, Lowe DA. Global phosphoproteomic profiling of skeletal muscle in ovarian hormone-deficient mice. Physiol Genomics 2022; 54:417-432. [PMID: 36062884 PMCID: PMC9639773 DOI: 10.1152/physiolgenomics.00104.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/05/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
Protein phosphorylation is important in skeletal muscle development, growth, regeneration, and contractile function. Alterations in the skeletal muscle phosphoproteome due to aging have been reported in males; however, studies in females are lacking. We have demonstrated that estrogen deficiency decreases muscle force, which correlates with decreased myosin regulatory light chain phosphorylation. Thus, we questioned whether the decline of estrogen in females that occurs with aging might alter the skeletal muscle phosphoproteome. C57BL/6J female mice (6 mo) were randomly assigned to a sham-operated (Sham) or ovariectomy (Ovx) group to investigate the effects of estrogen deficiency on skeletal muscle protein phosphorylation in a resting, noncontracting condition. After 16 wk of estrogen deficiency, the tibialis anterior muscle was dissected and prepped for label-free nano-liquid chromatography-tandem mass spectrometry phosphoproteomic analysis. We identified 4,780 phosphopeptides in tibialis anterior muscles of ovariectomized (Ovx) and Sham-operated (Sham) control mice. Further analysis revealed 647 differentially regulated phosphopeptides (Benjamini-Hochberg adjusted P value < 0.05 and 1.5-fold change ratio) that corresponded to 130 proteins with 22 proteins differentially phosphorylated (3 unique to Ovx, 2 unique to Sham, 6 upregulated, and 11 downregulated). Differentially phosphorylated proteins associated with the sarcomere, cytoplasm, and metabolic and calcium signaling pathways were identified. Our work provides the first global phosphoproteomic analysis in females and how estrogen deficiency impacts the skeletal muscle phosphoproteome.
Collapse
Affiliation(s)
- Mina P Peyton
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
- Department of Computer Science, Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, Minnesota
| | - Tzu-Yi Yang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - LeeAnn Higgins
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Todd W Markowski
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Cha Vue
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Laurie L Parker
- Department of Computer Science, Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, Minnesota
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| | - Dawn A Lowe
- Division of Rehabilitation Science, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
- Division of Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
15
|
Changes in cAMP signaling are associated with age-related downregulation of spontaneously beating atrial tissue energetic indices. GeroScience 2022; 45:209-219. [PMID: 35790659 PMCID: PMC9886694 DOI: 10.1007/s11357-022-00609-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 06/15/2022] [Indexed: 02/03/2023] Open
Abstract
The prevalence of atria-related diseases increases exponentially with age and is associated with ATP supply-to-demand imbalances. Because evidence suggests that cAMP regulates ATP supply-to-demand, we explored aged-associated alterations in atrial ATP supply-to-demand balance and its correlation with cAMP levels. Right atrial tissues driven by spontaneous sinoatrial node impulses were isolated from aged (22-26 months) and adult (3-4 months) C57/BL6 mice. ATP demand increased by addition of isoproterenol or 3-Isobutyl-1-methylxanthine (IBMX) and decreased by application of carbachol. Each drug was administrated at the dose that led to a maximal change in beating rate (Xmax) and to 50% of that maximal change in adult tissue (X50). cAMP, NADH, NAD + NADH, and ATP levels were measured in the same tissue. The tight correlation between cAMP levels and the beating rate (i.e., the ATP demand) demonstrated in adult atria was altered in aged atria. cAMP levels were lower in aged compared to adult atrial tissue exposed to X50 of ISO or IBMX, but this difference narrowed at Xmax. Neither ATP nor NADH levels correlated with ATP demand in either adult or aged atria. Baseline NADH levels were lower in aged as compared to adult atria, but were restored by drug perturbations that increased cAMP levels. Reduction in Ca2+-activated adenylyl cyclase-induced decreased cAMP and prolongation of the spontaneous beat interval of adult atrial tissue to their baseline levels in aged tissue, brought energetics indices to baseline levels in aged tissue. Thus, cAMP regulates right atrial ATP supply-to-demand matching and can restore age-associated ATP supply-to-demand imbalance.
Collapse
|
16
|
Regulation of Cr(VI)-Induced Premature Senescence in L02 Hepatocytes by ROS-Ca2+-NF-κB Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7295224. [PMID: 35222804 PMCID: PMC8881123 DOI: 10.1155/2022/7295224] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/18/2022] [Accepted: 01/29/2022] [Indexed: 02/07/2023]
Abstract
Stress-induced premature senescence may be involved in the pathogeneses of acute liver injury. Hexavalent chromium [Cr(VI)], a common environmental pollutant related to liver injury, likely leads to premature senescence in L02 hepatocytes. However, the underlying mechanisms regarding hepatocyte premature senility in Cr(VI) exposure remain poorly understood. In this study, we found that chronic exposure of L02 hepatocytes to Cr(VI) led to premature senescence characterized by increased β-galactosidase activity, senescence-associated heterochromatin foci, G1 phase arrest, and decreased cell proliferation. Additionally, Cr(VI)-induced senescent L02 hepatocytes showed upregulated inflammation-related factors, such as IL-6 and fibroblast growth factor 23 (FGF23), which also exhibited reactive oxygen species (ROS) accumulation derived from mitochondria accompanied with increased concentration of intracellular calcium ions (Ca2+) and activity of nuclear factor kappa B (NF-κB). Of note is that ROS inhibition by N-acetyl-Lcysteine pretreatment not only alleviated Cr(VI)-induced premature senescence but also reduced the elevated intracellular Ca2+, activated NF-κB, and secretion of IL-6/FGF23. Intriguingly, the toxic effect of Cr(VI) upon premature senescence of L02 hepatocytes and increased levels of IL-6/FGF23 could be partially reversed by the intracellular Ca2+ chelator BAPTA-AM pretreatment. Furthermore, by utilizing the NF-κB inhibitor pyrrolidine dithiocarbamate (PDTC), we confirmed that NF-κB mediated IL-6/FGF23 to regulate the Cr(VI)-induced L02 hepatocyte premature senescence, whilst the concentration of intracellular Ca2+ was not influenced by PDTC. To the best of our knowledge, our data reports for the first time the role of ROS-Ca2+-NF-κB signaling pathway in Cr(VI)-induced premature senescence. Our results collectively shed light on further exploration of innovative intervention strategies and treatment targeting Cr(VI)-induced chronic liver damage related to premature senescence.
Collapse
|
17
|
Adili A, Zhu X, Cao H, Tang X, Wang Y, Wang J, Shi J, Zhou Q, Wang D. Atrial Fibrillation Underlies Cardiomyocyte Senescence and Contributes to Deleterious Atrial Remodeling during Disease Progression. Aging Dis 2022; 13:298-312. [PMID: 35111375 PMCID: PMC8782549 DOI: 10.14336/ad.2021.0619] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 06/19/2021] [Indexed: 11/01/2022] Open
Affiliation(s)
| | | | | | | | | | | | | | - Qing Zhou
- Correspondence should be addressed to: Dr. Qing Zhou (), Dr. Dongjin Wang (), The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Dongjin Wang
- Correspondence should be addressed to: Dr. Qing Zhou (), Dr. Dongjin Wang (), The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
18
|
Baghaiee B, Bayatmakoo R, Karimi P, Pescatello LS. Moderate Aerobic Training Inhibits Middle-Aged Induced Cardiac Calcineurin-NFAT Signaling by Improving TGF-ß, NPR-A, SERCA2, and TRPC6 in Wistar Rats. CELL JOURNAL 2021; 23:756-762. [PMID: 34979065 PMCID: PMC8753105 DOI: 10.22074/cellj.2021.7531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 04/15/2020] [Indexed: 12/03/2022]
Abstract
OBJECTIVE The purpose of this study was to investigate the effect of moderate-intensity training on the calcineurin/ nuclear factor of activated t-cells (NFAT) pathway and factors affecting it in the middle-age Wistar rats. MATERIALS AND METHODS In this experimental study, 40 young (n=10, 4-month-old) and middle-aged (n=30, 13-15 months old) Wistar rats were included in this experimental study. All young and 10 middle-aged rats did not training and served as a control comparision; while the remaining 20 middle-aged rats were trained at moderate intensity for 4-weeks (n=10) or 8-weeks (n=10) on a treadmill (speed: 16 m/minutes, slope: 0%, distance: 830 m, duration: 54 minutes). RESULTS Calcineurin tissue expression was increased in the middle-aged control rats compared to the young control rats (P=0.001). Expression of sarco/endoplasmic reticulum Ca2+-ATPase (SERC2A), natriuretic peptide receptor-A (NPR-A), phospholamban (PLB), plasma membrane Ca2+ ATPase (PMCA4b), and p-AKT was significantly decreased in the heart tissue of middle-aged control compared to the young control rats (P=0.001). Furthermore, transforming growth factor beta (TGF-β), including transient receptor potential canonical 6 (TRPC6), were up-regulated in the heart tissue of middle-aged control compared to the young control rats (P=0.001). However, aerobic training inhibited this pathway and reversed all changes in the trained middle-aged rats. CONCLUSION Aerobic training effectively inhibited the calcineurin/NFATc pathway and modulated intracellular Ca2+ levels at least partially by restoring NPR-A, SERCA2, p-PLB, and p-AKT, and decreasing TRPC6 and TGF-β levels.
Collapse
Affiliation(s)
- Behrouz Baghaiee
- Department of Physical Education and Sport Science, Jolfa Branch, Islamic Azad University, Jolfa, Iran
| | - Roshanak Bayatmakoo
- Department of Biochemistry, School of Medicine, Tabriz Branch, Islamic Azad University, Tabriz, Iran,P.O.Box: 5157944533Department of BiochemistrySchool of MedicineTabriz BranchIslamic Azad UniversityTabrizIran
| | - Pouran Karimi
- Neuroscience Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Linda Shannon Pescatello
- Department of Kinesiology, College of Agriculture, Health and Natural Resources, University of Connecticut, Connecticut, USA
| |
Collapse
|
19
|
Fong SPT, Agrawal S, Gong M, Zhao J. Modulated Calcium Homeostasis and Release Events Under Atrial Fibrillation and Its Risk Factors: A Meta-Analysis. Front Cardiovasc Med 2021; 8:662914. [PMID: 34355025 PMCID: PMC8329373 DOI: 10.3389/fcvm.2021.662914] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 06/11/2021] [Indexed: 12/26/2022] Open
Abstract
Background: Atrial fibrillation (AF) is associated with calcium (Ca2+) handling remodeling and increased spontaneous calcium release events (SCaEs). Nevertheless, its exact mechanism remains unclear, resulting in suboptimal primary and secondary preventative strategies. Methods: We searched the PubMed database for studies that investigated the relationship between SCaEs and AF and/or its risk factors. Meta-analysis was used to examine the Ca2+ mechanisms involved in the primary and secondary AF preventative groups. Results: We included a total of 74 studies, out of the identified 446 publications from inception (1982) until March 31, 2020. Forty-five were primary and 29 were secondary prevention studies for AF. The main Ca2+ release events, calcium transient (standardized mean difference (SMD) = 0.49; I2 = 35%; confidence interval (CI) = 0.33–0.66; p < 0.0001), and spark amplitude (SMD = 0.48; I2 = 0%; CI = −0.98–1.93; p = 0.054) were enhanced in the primary diseased group, while calcium transient frequency was increased in the secondary group. Calcium spark frequency was elevated in both the primary diseased and secondary AF groups. One of the key cardiac currents, the L-type calcium current (ICaL) was significantly downregulated in primary diseased (SMD = −1.07; I2 = 88%; CI = −1.94 to −0.20; p < 0.0001) and secondary AF groups (SMD = −1.28; I2 = 91%; CI = −2.04 to −0.52; p < 0.0001). Furthermore, the sodium–calcium exchanger (INCX) and NCX1 protein expression were significantly enhanced in the primary diseased group, while only NCX1 protein expression was shown to increase in the secondary AF studies. The phosphorylation of the ryanodine receptor at S2808 (pRyR-S2808) was significantly elevated in both the primary and secondary groups. It was increased in the primary diseased and proarrhythmic subgroups (SMD = 0.95; I2 = 64%; CI = 0.12–1.79; p = 0.074) and secondary AF group (SMD = 0.66; I2 = 63%; CI = 0.01–1.31; p < 0.0001). Sarco/endoplasmic reticulum Ca2+-ATPase (SERCA) expression was elevated in the primary diseased and proarrhythmic drug subgroups but substantially reduced in the secondary paroxysmal AF subgroup. Conclusions: Our study identified that ICaL is reduced in both the primary and secondary diseased groups. Furthermore, pRyR-S2808 and NCX1 protein expression are enhanced. The remodeling leads to elevated Ca2+ functional activities, such as increased frequencies or amplitude of Ca2+ spark and Ca2+ transient. The main difference identified between the primary and secondary diseased groups is SERCA expression, which is elevated in the primary diseased group and substantially reduced in the secondary paroxysmal AF subgroup. We believe our study will add new evidence to AF mechanisms and treatment targets.
Collapse
Affiliation(s)
- Sarah Pei Ting Fong
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Shaleka Agrawal
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| | - Mengqi Gong
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jichao Zhao
- Auckland Bioengineering Institute, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
20
|
Remodeling of t-system and proteins underlying excitation-contraction coupling in aging versus failing human heart. NPJ Aging Mech Dis 2021; 7:16. [PMID: 34050186 PMCID: PMC8163749 DOI: 10.1038/s41514-021-00066-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 04/26/2021] [Indexed: 11/14/2022] Open
Abstract
It is well established that the aging heart progressively remodels towards a senescent phenotype, but alterations of cellular microstructure and their differences to chronic heart failure (HF) associated remodeling remain ill-defined. Here, we show that the transverse tubular system (t-system) and proteins underlying excitation-contraction coupling in cardiomyocytes are characteristically remodeled with age. We shed light on mechanisms of this remodeling and identified similarities and differences to chronic HF. Using left ventricular myocardium from donors and HF patients with ages between 19 and 75 years, we established a library of 3D reconstructions of the t-system as well as ryanodine receptor (RyR) and junctophilin 2 (JPH2) clusters. Aging was characterized by t-system alterations and sarcolemmal dissociation of RyR clusters. This remodeling was less pronounced than in HF and accompanied by major alterations of JPH2 arrangement. Our study indicates that targeting sarcolemmal association of JPH2 might ameliorate age-associated deficiencies of heart function.
Collapse
|
21
|
Hatem SN. Revealing the molecular history of the transition from paroxysmal to permanent atrial fibrillation. Cardiovasc Res 2021; 117:1612-1613. [PMID: 33878181 DOI: 10.1093/cvr/cvab106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Stéphane N Hatem
- INSERM UMRS1166, ICAN-Institute of Cardiometabolism and Nutrition, Sorbonne University, Institute of Cardiology, Pitié-Salpêtrière Hospital, 91, boulevard de l'hôpital, 75013 Paris, France
| |
Collapse
|
22
|
Herraiz-Martínez A, Tarifa C, Jiménez-Sábado V, Llach A, Godoy-Marín H, Colino H, Nolla-Colomer C, Casabella S, Izquierdo-Castro P, Benítez I, Benítez R, Roselló-Díez E, Rodríguez-Font E, Viñolas X, Ciruela F, Cinca J, Hove-Madsen L. Influence of sex on intracellular calcium homeostasis in patients with atrial fibrillation. Cardiovasc Res 2021; 118:1033-1045. [PMID: 33788918 PMCID: PMC8930070 DOI: 10.1093/cvr/cvab127] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/30/2021] [Indexed: 11/22/2022] Open
Abstract
Aims Atrial fibrillation (AF) has been associated with intracellular calcium disturbances in human atrial myocytes, but little is known about the potential influence of sex and we here aimed to address this issue. Methods and results Alterations in calcium regulatory mechanisms were assessed in human atrial myocytes from patients without AF or with long-standing persistent or permanent AF. Patch-clamp measurements revealed that L-type calcium current (ICa) density was significantly smaller in males with than without AF (−1.15 ± 0.37 vs. −2.06 ± 0.29 pA/pF) but not in females with AF (−1.88 ± 0.40 vs. −2.21 ± 0.0.30 pA/pF). In contrast, transient inward currents (ITi) were more frequent in females with than without AF (1.92 ± 0.36 vs. 1.10 ± 0.19 events/min) but not in males with AF. Moreover, confocal calcium imaging showed that females with AF had more calcium spark sites than those without AF (9.8 ± 1.8 vs. 2.2 ± 1.9 sites/µm2) and sparks were wider (3.0 ± 0.3 vs. 2.2 ± 0.3 µm) and lasted longer (79 ± 6 vs. 55 ± 8 ms), favouring their fusion into calcium waves that triggers ITIs and afterdepolarizations. This was linked to higher ryanodine receptor phosphorylation at s2808 in women with AF, and inhibition of adenosine A2A or beta-adrenergic receptors that modulate s2808 phosphorylation was able to reduce the higher incidence of ITI in women with AF. Conclusion Perturbations of the calcium homoeostasis in AF is sex-dependent, concurring with increased spontaneous SR calcium release-induced electrical activity in women but not in men, and with diminished ICa density in men only.
Collapse
Affiliation(s)
| | - Carmen Tarifa
- Biomedical Research Institute Barcelona Centre IIBB-CSIC.,IIB Sant Pau
| | | | | | - Hector Godoy-Marín
- Dept. Pathology and Experimental Therapeutics, IDIBELL, Univ. Barcelona, L'Hospitalet de Llobregat, Spain.,Neuroscience Institute, Univ. Barcelona, Spain
| | - Hildegard Colino
- Biomedical Research Institute Barcelona Centre IIBB-CSIC.,IIB Sant Pau
| | | | - Sergi Casabella
- Biomedical Research Institute Barcelona Centre IIBB-CSIC.,IIB Sant Pau
| | | | - Iván Benítez
- Biostatistic Unit, Biomedical Research Institute, IRBLleida, Spain
| | - Raul Benítez
- Dept. Automatic Control, Univ. Politècnica de Catalunya, Barcelona
| | - Elena Roselló-Díez
- Dept. Cardiac Surgery, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain.,Univ. Autònoma de Barcelona, Spain
| | | | - Xavier Viñolas
- Dept. Cardiology, Hospital de la Santa Creu i Sant Pau, Barcelona
| | - Francisco Ciruela
- Dept. Pathology and Experimental Therapeutics, IDIBELL, Univ. Barcelona, L'Hospitalet de Llobregat, Spain.,Neuroscience Institute, Univ. Barcelona, Spain
| | - Juan Cinca
- Dept. Cardiology, Hospital de la Santa Creu i Sant Pau, Barcelona.,CIBERCV.,Univ. Autònoma de Barcelona, Spain
| | - Leif Hove-Madsen
- Biomedical Research Institute Barcelona Centre IIBB-CSIC.,IIB Sant Pau.,CIBERCV
| |
Collapse
|
23
|
Liao HY, Liao B, Zhang HH. CISD2 plays a role in age-related diseases and cancer. Biomed Pharmacother 2021; 138:111472. [PMID: 33752060 DOI: 10.1016/j.biopha.2021.111472] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 03/03/2021] [Accepted: 03/04/2021] [Indexed: 12/12/2022] Open
Abstract
CDGSH iron-sulfur domain 2 (Cisd2) is an evolutionarily conserved protein that plays an important regulatory role in aging-related diseases and cancers. Since its discovery, Cisd2 has been identified as a regulatory factor for the aging of the human body and the regulation of mammalian lifespan. Cisd2 is also an oncoprotein that regulates the occurrence and development of cancer. Cisd2 mediates the occurrence of diseases related to human aging and the proliferation, differentiation, metastasis, and invasion of various cancer cells through various mechanisms. Multiple studies have shown that Cisd2 expression is related to the clinical characteristics of aging-related diseases and patients with cancer, and its expression profile is a novel diagnostic and prognostic biomarker for a variety of human diseases. Modulating the expression or function of Cisd2 may be a potential treatment strategy for different diseases. In this review, we summarize the role of Cisd2 in human aging-related diseases and various cancers, as well as the biological functions, underlying mechanisms, and potential clinical significance.
Collapse
Affiliation(s)
- Hai-Yang Liao
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China.
| | - Bei Liao
- Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China; The First Clinical Medical College of Lanzhou University, 1 Donggang Road, Lanzhou 730000, PR China.
| | - Hai-Hong Zhang
- The Second Clinical Medical College of Lanzhou University, 82 Cuiying Men, Lanzhou 730030, PR China; Orthopedics Key Laboratory of Gansu Province, Lanzhou 730000, PR China.
| |
Collapse
|
24
|
Yang M, Yan J, Wu A, Zhao W, Qin J, Pogwizd SM, Wu X, Yuan S, Ai X. Alterations of housekeeping proteins in human aged and diseased hearts. Pflugers Arch 2021; 473:351-362. [PMID: 33638007 PMCID: PMC10468297 DOI: 10.1007/s00424-021-02538-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/20/2021] [Accepted: 02/04/2021] [Indexed: 01/10/2023]
Abstract
Pathological remodeling includes alterations of ion channel function and calcium homeostasis and ultimately cardiac maladaptive function during the process of disease development. Biochemical assays are important approaches for assessing protein abundance and post-translational modification of ion channels. Several housekeeping proteins are commonly used as internal controls to minimize loading variabilities in immunoblotting protein assays. Yet, emerging evidence suggests that some housekeeping proteins may be abnormally altered under certain pathological conditions. However, alterations of housekeeping proteins in aged and diseased human hearts remain unclear. In the current study, immunoblotting was applied to measure three commonly used housekeeping proteins (β-actin, calsequestrin, and GAPDH) in well-procured human right atria (RA) and left ventricles (LV) from diabetic, heart failure, and aged human organ donors. Linear regression analysis suggested that the amounts of linearly loaded total proteins and quantified intensity of total proteins from either Ponceau S (PS) blot-stained or Coomassie Blue (CB) gel-stained images were highly correlated. Thus, all immunoblotting data were normalized with quantitative CB or PS data to calibrate potential loading variabilities. In the human heart, β-actin was reduced in diabetic RA and LV, while GAPDH was altered in aged and diabetic RA but not LV. Calsequestrin, an important Ca2+ regulatory protein, was significantly changed in aged, diabetic, and ischemic failing hearts. Intriguingly, expression levels of all three proteins were unchanged in non-ischemic failing human LV. Overall, alterations of human housekeeping proteins are heart chamber specific and disease context dependent. The choice of immunoblotting loading controls should be carefully evaluated. Usage of CB or PS total protein analysis could be a viable alternative approach for some complicated pathological specimens.
Collapse
Affiliation(s)
- Mei Yang
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 West Harrison St. 1255 Jelke South, Chicago, IL, 60612, USA
| | - Jiajie Yan
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 West Harrison St. 1255 Jelke South, Chicago, IL, 60612, USA
| | - Aimee Wu
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 West Harrison St. 1255 Jelke South, Chicago, IL, 60612, USA
| | - Weiwei Zhao
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 West Harrison St. 1255 Jelke South, Chicago, IL, 60612, USA
| | - Jin Qin
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 West Harrison St. 1255 Jelke South, Chicago, IL, 60612, USA
| | - Steven M Pogwizd
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Xin Wu
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 West Harrison St. 1255 Jelke South, Chicago, IL, 60612, USA
| | - Shengtao Yuan
- Jiangsu Key Laboratory of Drug Screening, China Pharmaceutical University, No. 24, Tongjiaxiang, Nanjing, 210009, China.
| | - Xun Ai
- Department of Physiology and Biophysics, Rush University Medical Center, 1750 West Harrison St. 1255 Jelke South, Chicago, IL, 60612, USA.
| |
Collapse
|
25
|
Salazar-Ramírez F, Ramos-Mondragón R, García-Rivas G. Mitochondrial and Sarcoplasmic Reticulum Interconnection in Cardiac Arrhythmia. Front Cell Dev Biol 2021; 8:623381. [PMID: 33585462 PMCID: PMC7876262 DOI: 10.3389/fcell.2020.623381] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/30/2020] [Indexed: 12/31/2022] Open
Abstract
Ca2+ plays a pivotal role in mitochondrial energy production, contraction, and apoptosis. Mitochondrial Ca2+-targeted fluorescent probes have demonstrated that mitochondria Ca2+ transients are synchronized with Ca2+ fluxes occurring in the sarcoplasmic reticulum (SR). The presence of specialized proteins tethering SR to mitochondria ensures the local Ca2+ flux between these organelles. Furthermore, communication between SR and mitochondria impacts their functionality in a bidirectional manner. Mitochondrial Ca2+ uptake through the mitochondrial Ca2+ uniplex is essential for ATP production and controlled reactive oxygen species levels for proper cellular signaling. Conversely, mitochondrial ATP ensures the proper functioning of SR Ca2+-handling proteins, which ensures that mitochondria receive an adequate supply of Ca2+. Recent evidence suggests that altered SR Ca2+ proteins, such as ryanodine receptors and the sarco/endoplasmic reticulum Ca2+ ATPase pump, play an important role in maintaining proper cardiac membrane excitability, which may be initiated and potentiated when mitochondria are dysfunctional. This recognized mitochondrial role offers the opportunity to develop new therapeutic approaches aimed at preventing cardiac arrhythmias in cardiac disease.
Collapse
Affiliation(s)
- Felipe Salazar-Ramírez
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico
| | - Roberto Ramos-Mondragón
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Gerardo García-Rivas
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Cátedra de Cardiología y Medicina Cardiovascular, Monterrey, Mexico.,TecSalud, Centro de Investigación Biomédica, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico.,TecSalud, Centro de Medicina Funcional, Hospital Zambrano-Hellion, San Pedro Garza García, Mexico
| |
Collapse
|
26
|
Ezeani M, Prabhu S. Pathophysiology and therapeutic relevance of PI3K(p110α) protein in atrial fibrillation: A non-interventional molecular therapy strategy. Pharmacol Res 2021; 165:105415. [PMID: 33412279 DOI: 10.1016/j.phrs.2020.105415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/04/2020] [Accepted: 12/23/2020] [Indexed: 10/22/2022]
Abstract
Genetically modified animal studies have revealed specific expression patterns and unequivocal roles of class I PI3K isoenzymes. PI3K(p110α), a catalytic subunit of class I PI3Ks is ubiquitously expressed and is well characterised in the cardiovascular system. Given that genetic inhibition of PI3K(p110α) causes lethal phenotype embryonically, the catalytic subunit is critically important in housekeeping and biological processes. A growing number of studies underpin crucial roles of PI3K(p110α) in cell survival, proliferation, hypertrophy and arrhythmogenesis. While the studies provide great insights, the precise mechanisms involved in PI3K(p110α) hypofunction and atrial fibrillation (AF) are not fully known. AF is a well recognised clinical problem with significant management limitations. In this translational review, we attempted a narration of PI3K(p110α) hypofunction in the molecular basis of AF pathophysiology. We sought to cautiously highlight the relevance of this molecule in the therapeutic approaches for AF management per se (i.e without conditions associate with cell proliferation, like cancer), and in mitigating effects of clinical risk factors in atrial substrate formation leading to AF progression. We also considered PI3K(p110α) in AF gene association, with the aim of identifying mechanistic links between the ever increasingly well-defined genetic loci (regions and genes) and AF. Such mechanisms will aid in identifying new drug targets for arrhythmogenic substrate and AF.
Collapse
Affiliation(s)
- Martin Ezeani
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, 3004, Australia.
| | - Sandeep Prabhu
- The Alfred, and Baker Heart and Diabetes Institute, Melbourne, Australia; University of Melbourne, Melbourne, Australia
| |
Collapse
|
27
|
Jin G, Manninger M, Adelsmayr G, Schwarzl M, Alogna A, Schönleitner P, Zweiker D, Blaschke F, Sherif M, Radulovic S, Wakula P, Schauer S, Höfler G, Reiter U, Reiter G, Post H, Scherr D, Acsai K, Antoons G, Pieske B, Heinzel FR. Cellular contribution to left and right atrial dysfunction in chronic arterial hypertension in pigs. ESC Heart Fail 2020; 8:151-161. [PMID: 33251761 PMCID: PMC7835565 DOI: 10.1002/ehf2.13087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 09/02/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Aims Atrial contractile dysfunction contributes to worse prognosis in hypertensive heart disease (HHD), but the role of cardiomyocyte dysfunction in atrial remodelling in HHD is not well understood. We investigated and compared cellular mechanisms of left (LA) and right atrial (RA) contractile dysfunction in pigs with HHD. Methods and results In vivo electrophysiological and magnetic resonance imaging studies were performed in control and pigs treated with 11‐deoxycorticosterone acetate (DOCA)/high‐salt/glucose diet (12 weeks) to induce HHD. HHD leads to significant atrial remodelling and loss of contractile function in LA and a similar trend in RA (magnetic resonance imaging). Atrial remodelling was associated with a higher inducibility of atrial fibrillation but unrelated to changes in atrial refractory period or fibrosis (histology). Reduced atrial function in DOCA pigs was related to reduced contraction amplitude of isolated LA (already at baseline) and RA myocytes (at higher frequencies) due to reduced intracellular Ca release (Fura 2‐AM, field stimulation). However, Ca regulation differed in LA and RA cardiomyocytes: LA cardiomyocytes showed reduced sarcoplasmic reticulum (SR) [Ca], whereas in RA, SR [Ca] was unchanged and SR Ca2+‐ATPase activity was increased. Sodium–calcium exchanger (NCX) activity was not significantly altered. We used ORM‐10103 (3 μM), a specific NCX inhibitor to improve Ca availability in LA and RA cardiomyocytes from DOCA pigs. Partial inhibition of NCX increased Ca2+ transient amplitude and SR Ca in LA, but not RA cells. Conclusions In this large animal model of HHD, atrial remodelling in sinus rhythm in vivo was related to differential LA and RA cardiomyocyte dysfunction and Ca signalling. Selective acute inhibition of NCX improved Ca release in diseased LA cardiomyocytes, suggesting a potential therapeutic approach to improve atrial inotropy in HHD.
Collapse
Affiliation(s)
- Ge Jin
- Division of Cardiology, Medical University of Graz, Graz, Austria.,The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Martin Manninger
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | | | - Michael Schwarzl
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Alessio Alogna
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | | | - David Zweiker
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Florian Blaschke
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Mohammad Sherif
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany
| | | | - Paulina Wakula
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Sylvia Schauer
- Department of Pathology, Medical University of Graz, Graz, Austria
| | - Gerald Höfler
- Department of Pathology, Medical University of Graz, Graz, Austria
| | - Ursula Reiter
- Department of Radiology, Medical University of Graz, Graz, Austria
| | - Gert Reiter
- Research & Development, Siemens AG Healthcare, Vienna, Austria
| | - Heiner Post
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| | - Daniel Scherr
- Division of Cardiology, Medical University of Graz, Graz, Austria
| | - Karoly Acsai
- Division of Pharmacology and Pharmacotherapy, University of Szeged, Szeged, Hungary
| | - Gudrun Antoons
- Faculty of Sciences, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Burkert Pieske
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany.,Department of Cardiology, German Heart Center Berlin (DHZB), Berlin, Germany
| | - Frank R Heinzel
- Department of Internal Medicine and Cardiology, Charité-Universitätsmedizin Berlin, Campus Virchow-Klinikum, Augustenburgerplatz 1, Berlin, 13353, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin, Berlin, Germany
| |
Collapse
|
28
|
Madreiter-Sokolowski CT, Thomas C, Ristow M. Interrelation between ROS and Ca 2+ in aging and age-related diseases. Redox Biol 2020; 36:101678. [PMID: 32810740 PMCID: PMC7451758 DOI: 10.1016/j.redox.2020.101678] [Citation(s) in RCA: 219] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 02/07/2023] Open
Abstract
Calcium (Ca2+) and reactive oxygen species (ROS) are versatile signaling molecules coordinating physiological and pathophysiological processes. While channels and pumps shuttle Ca2+ ions between extracellular space, cytosol and cellular compartments, short-lived and highly reactive ROS are constantly generated by various production sites within the cell. Ca2+ controls membrane potential, modulates mitochondrial adenosine triphosphate (ATP) production and affects proteins like calcineurin (CaN) or calmodulin (CaM), which, in turn, have a wide area of action. Overwhelming Ca2+ levels within mitochondria efficiently induce and trigger cell death. In contrast, ROS comprise a diverse group of relatively unstable molecules with an odd number of electrons that abstract electrons from other molecules to gain stability. Depending on the type and produced amount, ROS act either as signaling molecules by affecting target proteins or as harmful oxidative stressors by damaging cellular components. Due to their wide range of actions, it is little wonder that Ca2+ and ROS signaling pathways overlap and impact one another. Growing evidence suggests a crucial implication of this mutual interplay on the development and enhancement of age-related disorders, including cardiovascular and neurodegenerative diseases as well as cancer.
Collapse
Affiliation(s)
- Corina T Madreiter-Sokolowski
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland; Holder of an Erwin Schroedinger Abroad Fellowship, Austrian Science Fund (FWF), Austria.
| | - Carolin Thomas
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| | - Michael Ristow
- Energy Metabolism Laboratory, Institute of Translational Medicine, Department of Health Sciences and Technology, Swiss Federal Institute of Technology (ETH), Zurich, Switzerland
| |
Collapse
|
29
|
Weiss AKH, Albertini E, Holzknecht M, Cappuccio E, Dorigatti I, Krahbichler A, Damisch E, Gstach H, Jansen-Dürr P. Regulation of cellular senescence by eukaryotic members of the FAH superfamily - A role in calcium homeostasis? Mech Ageing Dev 2020; 190:111284. [PMID: 32574647 PMCID: PMC7116474 DOI: 10.1016/j.mad.2020.111284] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 01/04/2023]
Abstract
Fumarylacetoacetate hydrolase (FAH) superfamily members are commonly expressed in the prokaryotic kingdom, where they take part in the committing steps of degradation pathways of complex carbon sources. Besides FAH itself, the only described FAH superfamily members in the eukaryotic kingdom are fumarylacetoacetate hydrolase domain containing proteins (FAHD) 1 and 2, that have been a focus of recent work in aging research. Here, we provide a review of current knowledge on FAHD proteins. Of those, FAHD1 has recently been described as a regulator of mitochondrial function and senescence, in the context of mitochondrial dysfunction associated senescence (MiDAS). This work further describes data based on bioinformatics analysis, 3D structure comparison and sequence alignment, that suggests a putative role of FAHD proteins as calcium binding proteins.
Collapse
Affiliation(s)
- Alexander K H Weiss
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria.
| | - Eva Albertini
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria
| | - Max Holzknecht
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria
| | - Elia Cappuccio
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria
| | - Ilaria Dorigatti
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria
| | - Anna Krahbichler
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria
| | - Elisabeth Damisch
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria
| | - Hubert Gstach
- University of Vienna, UZ2 E349, Department of Pharmaceutical Chemistry, Faculty of Life Sciences, Althanstrasse 14, 1090, Vienna, Austria
| | - Pidder Jansen-Dürr
- University of Innsbruck, Research Institute for Biomedical Aging Research, Rennweg 10, A-6020, Innsbruck, Austria; University of Innsbruck, Center for Molecular Biosciences Innsbruck (CMBI), Austria
| |
Collapse
|
30
|
Jansen HJ, Bohne LJ, Gillis AM, Rose RA. Atrial remodeling and atrial fibrillation in acquired forms of cardiovascular disease. Heart Rhythm O2 2020; 1:147-159. [PMID: 34113869 PMCID: PMC8183954 DOI: 10.1016/j.hroo.2020.05.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Atrial fibrillation (AF) is prevalent in common conditions and acquired forms of heart disease, including diabetes mellitus (DM), hypertension, cardiac hypertrophy, and heart failure. AF is also prevalent in aging. Although acquired heart disease is common in aging individuals, age is also an independent risk factor for AF. Importantly, not all individuals age at the same rate. Rather, individuals of the same chronological age can vary in health status from fit to frail. Frailty can be quantified using a frailty index, which can be used to assess heterogeneity in individuals of the same chronological age. AF is thought to occur in association with electrical remodeling due to changes in ion channel expression or function as well as structural remodeling due to fibrosis, myocyte hypertrophy, or adiposity. These forms of remodeling can lead to triggered activity and electrical re-entry, which are fundamental mechanisms of AF initiation and maintenance. Nevertheless, the underlying determinants of electrical and structural remodeling are distinct in different conditions and disease states. In this focused review, we consider the factors leading to atrial electrical and structural remodeling in human patients and animal models of acquired cardiovascular disease or associated risk factors. Our goal is to identify similarities and differences in the cellular and molecular bases for atrial electrical and structural remodeling in conditions including DM, hypertension, hypertrophy, heart failure, aging, and frailty.
Collapse
Affiliation(s)
- Hailey J Jansen
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Loryn J Bohne
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Anne M Gillis
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Robert A Rose
- Department of Cardiac Sciences, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Physiology and Pharmacology, Libin Cardiovascular Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
31
|
Abstract
A progressive decline in maximum heart rate (mHR) is a fundamental aspect of aging in humans and other mammals. This decrease in mHR is independent of gender, fitness, and lifestyle, affecting in equal measure women and men, athletes and couch potatoes, spinach eaters and fast food enthusiasts. Importantly, the decline in mHR is the major determinant of the age-dependent decline in aerobic capacity that ultimately limits functional independence for many older individuals. The gradual reduction in mHR with age reflects a slowing of the intrinsic pacemaker activity of the sinoatrial node of the heart, which results from electrical remodeling of individual pacemaker cells along with structural remodeling and a blunted β-adrenergic response. In this review, we summarize current evidence about the tissue, cellular, and molecular mechanisms that underlie the reduction in pacemaker activity with age and highlight key areas for future work.
Collapse
Affiliation(s)
- Colin H Peters
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA; , ,
| | - Emily J Sharpe
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA; , ,
| | - Catherine Proenza
- Department of Physiology and Biophysics, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA; , ,
- Department of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, USA
| |
Collapse
|
32
|
Nesterova T, Shmarko D, Ushenin K, Solovyova O. In-silico analysis of aging mechanisms of action potential remodeling in human atrial cardiomyocites. BIO WEB OF CONFERENCES 2020. [DOI: 10.1051/bioconf/20202201025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Electrophysiology of cardiomyocytes changes with aging. Agerelated ionic remodeling in cardiomyocytes may increase the incidence and prevalence of atrial fibrillation (AF) in the elderly and affect the efficiency of antiarrhythmic drugs. There is the deep lack of experimental data on an action potential and transmembrane currents recorded in the healthy human cardiomyocytes of different age. Experimental data in mammals is also incomplete and often contradicting depending on the experimental conditions. In this in-silico study, we used a population of ionic models of human atrial cardiomyocytes to transfer data on the age- related ionic remodeling in atrial cardiomyocytes from canines and mice to predict possible consequences for human cardiomyocyte activity. Based on experimental data, we analyzes two hypotheses on the aging effect on the ionic currents using two age-related sets of varied model parameters and evaluated corresponding changes in action potential morphology with aging. Using the two populations of aging models, we analyzed the agedependent sensitivity of atrial cardiomyocytes to Dofetilide which is one of the antiarrhythmic drugs widely used in patients with atrial fibrillation.
Collapse
|
33
|
Abi-Gerges N, Miller PE, Ghetti A. Human Heart Cardiomyocytes in Drug Discovery and Research: New Opportunities in Translational Sciences. Curr Pharm Biotechnol 2019; 21:787-806. [PMID: 31820682 DOI: 10.2174/1389201021666191210142023] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/14/2019] [Accepted: 10/28/2019] [Indexed: 12/28/2022]
Abstract
In preclinical drug development, accurate prediction of drug effects on the human heart is critically important, whether in the context of cardiovascular safety or for the purpose of modulating cardiac function to treat heart disease. Current strategies have significant limitations, whereby, cardiotoxic drugs can escape detection or potential life-saving therapies are abandoned due to false positive toxicity signals. Thus, new and more reliable translational approaches are urgently needed to help accelerate the rate of new therapy development. Renewed efforts in the recovery of human donor hearts for research and in cardiomyocyte isolation methods, are providing new opportunities for preclinical studies in adult primary cardiomyocytes. These cells exhibit the native physiological and pharmacological properties, overcoming the limitations presented by artificial cellular models, animal models and have great potential for providing an excellent tool for preclinical drug testing. Adult human primary cardiomyocytes have already shown utility in assessing drug-induced cardiotoxicity risk and helping in the identification of new treatments for cardiac diseases, such as heart failure and atrial fibrillation. Finally, strategies with actionable decision-making trees that rely on data derived from adult human primary cardiomyocytes will provide the holistic insights necessary to accurately predict human heart effects of drugs.
Collapse
Affiliation(s)
- Najah Abi-Gerges
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Paul E Miller
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| | - Andre Ghetti
- AnaBios Corporation, 3030 Bunker Hill St., Suite 312, San Diego, CA 92109, United States
| |
Collapse
|
34
|
Yeh CH, Shen ZQ, Hsiung SY, Wu PC, Teng YC, Chou YJ, Fang SW, Chen CF, Yan YT, Kao LS, Kao CH, Tsai TF. Cisd2 is essential to delaying cardiac aging and to maintaining heart functions. PLoS Biol 2019; 17:e3000508. [PMID: 31593566 PMCID: PMC6799937 DOI: 10.1371/journal.pbio.3000508] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 10/18/2019] [Accepted: 09/24/2019] [Indexed: 11/18/2022] Open
Abstract
CDGSH iron-sulfur domain-containing protein 2 (Cisd2) is pivotal to mitochondrial integrity and intracellular Ca2+ homeostasis. In the heart of Cisd2 knockout mice, Cisd2 deficiency causes intercalated disc defects and leads to degeneration of the mitochondria and sarcomeres, thereby impairing its electromechanical functioning. Furthermore, Cisd2 deficiency disrupts Ca2+ homeostasis via dysregulation of sarco/endoplasmic reticulum Ca2+-ATPase (Serca2a) activity, resulting in an increased level of basal cytosolic Ca2+ and mitochondrial Ca2+ overload in cardiomyocytes. Most strikingly, in Cisd2 transgenic mice, a persistently high level of Cisd2 is sufficient to delay cardiac aging and attenuate age-related structural defects and functional decline. In addition, it results in a younger cardiac transcriptome pattern during old age. Our findings indicate that Cisd2 plays an essential role in cardiac aging and in the heart's electromechanical functioning. They highlight Cisd2 as a novel drug target when developing therapies to delay cardiac aging and ameliorate age-related cardiac dysfunction.
Collapse
Affiliation(s)
- Chi-Hsiao Yeh
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- * E-mail: (C-HY); (T-FT)
| | - Zhao-Qing Shen
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
| | - Shao-Yu Hsiung
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Pei-Chun Wu
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yuan-Chi Teng
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Yi-Ju Chou
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
| | - Su-Wen Fang
- Department of Thoracic and Cardiovascular Surgery, Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Chian-Feng Chen
- Genome Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Yu-Ting Yan
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Lung-Sen Kao
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
- Brain Research Center, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Heng Kao
- Center of General Education, Chang Gung University, Taoyuan, Taiwan
| | - Ting-Fen Tsai
- Department of Life Sciences and Institute of Genome Sciences, National Yang-Ming University, Taipei, Taiwan
- Program in Molecular Medicine, School of Life Sciences, National Yang-Ming University and Academia Sinica, Taipei, Taiwan
- Aging and Health Research Center, National Yang-Ming University, Taipei, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
- * E-mail: (C-HY); (T-FT)
| |
Collapse
|
35
|
Abstract
Cardiovascular diseases are the most prominent maladies in aging societies. Indeed, aging promotes the structural and functional declines of both the heart and the blood circulation system. In this review, we revise the contribution of known longevity pathways to cardiovascular health and delineate the possibilities to interfere with them. In particular, we evaluate autophagy, the intracellular catabolic recycling system associated with life- and health-span extension. We present genetic models, pharmacological interventions, and dietary strategies that block, reduce, or enhance autophagy upon age-related cardiovascular deterioration. Caloric restriction or caloric restriction mimetics like metformin, spermidine, and rapamycin (all of which trigger autophagy) are among the most promising cardioprotective interventions during aging. We conclude that autophagy is a fundamental process to ensure cardiac and vascular health during aging and outline its putative therapeutic importance.
Collapse
Affiliation(s)
- Mahmoud Abdellatif
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.)
| | - Simon Sedej
- From the Department of Cardiology, Medical University of Graz, Austria (M.A., S.S.).,BioTechMed Graz, Austria (S.S., D.C.-G., F.M.)
| | - Didac Carmona-Gutierrez
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Frank Madeo
- BioTechMed Graz, Austria (S.S., D.C.-G., F.M.).,Institute of Molecular Biosciences, NAWI Graz, University of Graz, Austria (D.C.-G., F.M.)
| | - Guido Kroemer
- Equipe 11 Labellisée Ligue Contre le Cancer, Centre de Recherche des Cordeliers, Paris, France (G.K.).,Cell Biology and Metabolomics Platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France (G.K.).,INSERM, U1138, Paris, France (G.K.).,Université Paris Descartes, Sorbonne Paris Cité, France (G.K.).,Université Pierre et Marie Curie, Paris, France (G.K.).,Pôle de Biologie, Hôpital Européen Georges Pompidou, Paris, France (G.K.).,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden (G.K.)
| |
Collapse
|
36
|
Yao Y, Jiang C, Wang F, Yan H, Long D, Zhao J, Wang J, Zhang C, Li Y, Tian X, Wang QK, Wu G, Zhang Z. Integrative Analysis of miRNA and mRNA Expression Profiles Associated With Human Atrial Aging. Front Physiol 2019; 10:1226. [PMID: 31607954 PMCID: PMC6761282 DOI: 10.3389/fphys.2019.01226] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2019] [Accepted: 09/09/2019] [Indexed: 11/28/2022] Open
Abstract
Background Limited findings have been reported to systematically study miRNA and mRNA expression profiles in aged human atria. In this study, we aimed to identify miRNAs, genes, and miRNA-mRNA interaction networks for human atrial aging (AA). Methods Right atrial appendages from twelve patients who received aortic valve replacement were subjected to miRNA-seq and RNA-seq. All the patients were in sinus rhythm (SR) and stratified by age into four groups. Differential expression analysis was carried out to identify miRNAs and genes for human AA. The miRNA-mRNA interactions for human AA were identified by Pearson correlation analysis and miRNA target prediction programs. Results Seven miRNAs (4 upregulation and 3 downregulation) and 42 genes (23 upregulation and 19 downregulation) were differentially expressed in human right atrial tissues between older samples and younger samples. Bioinformatic analysis identified 114 pairs of putative miRNA-mRNA interactions on AA and four types of correlation. Pathway enrichment analysis identified over 40 significant pathways and the top three pathways included rhythmic process (P = 7.5 × 10–5, Q = 0.034), senescence and autophagy in cancer (P = 9.0 × 10–5, Q = 0.034), and positive regulation of cytokine biosynthetic process (P = 1.1 × 10–4, Q = 0.034). Conclusion Our study revealed novel miRNA-mRNA interaction networks and signaling pathways for AA, providing novel insights into the development of human AA. Future studies are needed to investigate the potential significance of these miRNA-mRNA interactions in human AA or AA-related cardiovascular diseases.
Collapse
Affiliation(s)
- Yan Yao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chenxi Jiang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Fan Wang
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Case Western Reserve University, Cleveland, OH, United States
| | - Han Yan
- Department of Human Population Genetics and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Deyong Long
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jinghua Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Jiangang Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chunxiao Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Li
- Department of Human Population Genetics and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xiaoli Tian
- Department of Human Population Genetics and Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Qing K Wang
- Center for Cardiovascular Genetics, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, Case Western Reserve University, Cleveland, OH, United States
| | - Gang Wu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihui Zhang
- Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, China
| |
Collapse
|
37
|
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: 55] [Impact Index Per Article: 9.2] [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
|
38
|
Alterations of protein expression of phospholamban, ZASP and plakoglobin in human atria in subgroups of seniors. Sci Rep 2019; 9:5610. [PMID: 30948763 PMCID: PMC6449388 DOI: 10.1038/s41598-019-42141-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 03/22/2019] [Indexed: 01/20/2023] Open
Abstract
The mature mammalian myocardium contains composite junctions (areae compositae) that comprise proteins of adherens junctions as well as desmosomes. Mutations or deficiency of many of these proteins are linked to heart failure and/or arrhythmogenic cardiomyopathy in patients. We firstly wanted to address the question whether the expression of these proteins shows an age-dependent alteration in the atrium of the human heart. Right atrial biopsies, obtained from patients undergoing routine bypass surgery for coronary heart disease were subjected to immunohistology and/or western blotting for the plaque proteins plakoglobin (γ-catenin) and plakophilin 2. Moreover, the Z-band protein cypher 1 (Cypher/ZASP) and calcium handling proteins of the sarcoplasmic reticulum (SR) like phospholamban, SERCA and calsequestrin were analyzed. We noted expression of plakoglobin, plakophilin 2 and Cypher/ZASP in these atrial preparations on western blotting and/or immunohistochemistry. There was an increase of Cypher/ZASP expression with age. The present data extend our knowledge on the expression of anchoring proteins and SR regulatory proteins in the atrium of the human heart and indicate an age-dependent variation in protein expression. It is tempting to speculate that increased expression of Cypher/ZASP may contribute to mechanical changes in the aging human myocardium.
Collapse
|
39
|
ROCK2 promotes ryanodine receptor phosphorylation and arrhythmic calcium release in diabetic cardiomyocytes. Int J Cardiol 2019; 281:90-98. [DOI: 10.1016/j.ijcard.2019.01.075] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 01/08/2019] [Accepted: 01/18/2019] [Indexed: 11/16/2022]
|
40
|
Age-Dependent Protein Expression of Serine/Threonine Phosphatases and Their Inhibitors in the Human Cardiac Atrium. Adv Med 2019; 2019:2675972. [PMID: 30719459 PMCID: PMC6334353 DOI: 10.1155/2019/2675972] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 11/22/2018] [Accepted: 11/29/2018] [Indexed: 12/20/2022] Open
Abstract
Heart failure and aging of the heart show many similarities regarding hemodynamic and biochemical parameters. There is evidence that heart failure in experimental animals and humans is accompanied and possibly exacerbated by increased activity of protein phosphatase (PP) 1 and/or 2A. Here, we wanted to study the age-dependent protein expression of major members of the protein phosphatase family in human hearts. Right atrial samples were obtained during bypass surgery. Patients (n=60) were suffering from chronic coronary artery disease (CCS 2-3; New York Heart Association (NYHA) stage 1-3). Age ranged from 48 to 84 years (median 69). All patients included in the study were given β-adrenoceptor blockers. Other medications included angiotensin-converting enzyme (ACE) or angiotensin-receptor-1 (AT1) inhibitors, statins, nitrates, and acetylsalicylic acid (ASS). 100 µg of right atrial homogenates was used for western blotting. Antibodies against catalytic subunits (and their major regulatory proteins) of all presently known cardiac serine/threonine phosphatases were used for antigen detection. In detail, we studied the expression of the catalytic subunit of PP1 (PP1c); I1 PP1 and I2 PP1, proteins that can inhibit the activity of PP1c; the catalytic subunit of PP2A (PP2Ac); regulatory A-subunit of PP2A (PP2AA); regulatory B56α-subunit of PP2A (PP2AB); I1 PP2A and I2 PP2A, inhibitory subunits of PP2A; catalytic and regulatory subunits of calcineurin: PP2BA and PP2BB; PP2C; PP5; and PP6. All data were obtained within the linear range of the assay. There was a significant decline in PP2Ac and I2 PP2A expression in older patients, whereas all other parameters remained unchanged with age. It remains to be elucidated whether the decrease in the protein expression of I2 PP2A might elevate cardiac PP2A activity in a detrimental way or is overcome by a reduced protein expression and thus a reduced activity of PP2Ac.
Collapse
|
41
|
Chadda KR, Ajijola OA, Vaseghi M, Shivkumar K, Huang CLH, Jeevaratnam K. Ageing, the autonomic nervous system and arrhythmia: From brain to heart. Ageing Res Rev 2018; 48:40-50. [PMID: 30300712 DOI: 10.1016/j.arr.2018.09.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 08/21/2018] [Accepted: 09/30/2018] [Indexed: 02/08/2023]
Abstract
An ageing myocardium possesses significant electrophysiological alterations that predisposes the elderly patient to arrhythmic risk. Whilst these alterations are intrinsic to the cardiac myocytes, they are modulated by the cardiac autonomic nervous system (ANS) and consequently, ageing of the cardiac ANS is fundamental to the development of arrhythmias. A systems-based approach that incorporates the influence of the cardiac ANS could lead to better mechanistic understanding of how arrhythmogenic triggers and substrates interact spatially and temporally to produce sustained arrhythmia and why its incidence increases with age. Despite the existence of physiological oscillations of ANS activity on the heart, pathological oscillations can lead to defective activation and recovery properties of the myocardium. Such changes can be attributable to the decrease in functionality and structural alterations to ANS specific receptors in the myocardium with age. These altered ANS adaptive responses can occur either as a normal ageing process or accelerated in the presence of specific cardiac pathologies, such as genetic mutations or neurodegenerative conditions. Targeted intervention that seek to manipulate the ageing ANS influence on the myocardium may prove to be an efficacious approach for the management of arrhythmia in the ageing population.
Collapse
Affiliation(s)
- Karan R Chadda
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom
| | - Olujimi A Ajijola
- UCLA Cardiac Arrhythmia Center, UCLA Health System/David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Marmar Vaseghi
- UCLA Cardiac Arrhythmia Center, UCLA Health System/David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kalyanam Shivkumar
- UCLA Cardiac Arrhythmia Center, UCLA Health System/David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Christopher L-H Huang
- Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom; Department of Biochemistry, Hopkins Building, University of Cambridge, Cambridge, CB2 1QW, United Kingdom
| | - Kamalan Jeevaratnam
- Faculty of Health and Medical Sciences, University of Surrey, Guildford, GU2 7AL, United Kingdom; Physiological Laboratory, University of Cambridge, Downing Street, Cambridge, CB2 3EG, United Kingdom.
| |
Collapse
|
42
|
Zhang JC, Wu HL, Chen Q, Xie XT, Zou T, Zhu C, Dong Y, Xiang GJ, Ye L, Li Y, Zhu PL. Calcium-Mediated Oscillation in Membrane Potentials and Atrial-Triggered Activity in Atrial Cells of Casq2 R33Q/R33Q Mutation Mice. Front Physiol 2018; 9:1447. [PMID: 30450052 PMCID: PMC6224359 DOI: 10.3389/fphys.2018.01447] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 09/24/2018] [Indexed: 12/11/2022] Open
Abstract
Aim: We investigated the underlying mechanisms in atrial fibrillation (AF) associated with R33Q mutation and Ca2+-triggered activity. Methods and Results: We examined AF susceptibility with intraesophageal burst pacing in the sarcoplasmic reticulum (SR) Ca2+ leak model calsequestrin 2 R33Q (Casq2R33Q/R33Q) mice. Atrial trigger appeared in R33Q mice but not WT mice (17.24%, 5/29 vs. 0.00%, 0/32, P < 0.05). AF was induced by 25 Hz pacing in R33Q mice (48.27%, 14/29 vs. 6.25%, 2/32, P < 0.01). The mice were given 1.5 mg/kg isoproterenol (Iso), and the incidences of AF increased (65.51%, 19/29 vs. 9.21%, 3/32, P < 0.01). Electrophysiology experiments and the recording of intracellular Ca2+ indicated significant increases in the Ca2+ sparks (5.24 ± 0.75 100 μM-1.s-1 vs. 0.29 ± 0.04 100 μM-1.s-1, n = 20, P < 0.05), intracellular free Ca2+ (0.238 ± 0.009 μM vs. 0.172 ± 0.006 μM, n = 20, P < 0.05), Ca2+ wave (11.74% vs. 2.24%, n = 20, P < 0.05), transient inward current (ITi) (-0.56 ± 0.02 pA/pF vs. -0.42 ± 0.01 pA/pF, n = 10, P < 0.05), and oscillation in membrane potentials (10.71%, 3/28 vs. 4.16%, 1/24, P < 0.05) in the R33Q group, but there was no significant difference in the L-type calcium current. These effects were enhanced by Iso, and the inhibition of calmodulin-dependent protein kinase II (CaMKII) by 1 μM KN93 reversed the effects of Iso on Ca2+ sparks (5.01 ± 0.66 100 μm-1.s-1 vs. 11.33 ± 1.63 100 μm-1.s-1, P < 0.05), intracellular Ca2+ (0.245 ± 0.005 μM vs. 0.324 ± 0.008 μM, P < 0.05), Ca2+ wave (12.35% vs. 17.83%, P < 0.05), ITi (-0.61 ± 0.02 pA/pF vs. -0.78 ± 0.03 pA/pF, n = 10, P < 0.05), and oscillation in membrane potential (17.85% 5/28 vs. 32.17% 9/28, P < 0.05). The reduction of ryanodine receptor 2 (RyR2) stable subunits (Casq2, triadin, and junctin) rather than RYR2 and the increase in CaMKII, phosphor-CaMKII, phosphor-RyR2 (Ser 2814), SERCA, and NCX1.1 was reflected in the R33Q group. Conclusion: This study demonstrates that the increase in spontaneous calcium elevations corresponding to ITi that may trigger the oscillation in membrane potentials in the R33Q group, thereby increasing the risk of AF. The occurrence of spontaneous calcium elevations in R33Q atrial myocytes is due to the dysfunction of RyR2 stable subunits, CaMKII hyperactivity, and CaMKII-mediated RyR phosphorylation. An effective therapeutic strategy to intervene in Ca2+-induced AF associated with the R33Q mutation may be through CaMKII inhibition.
Collapse
Affiliation(s)
- Jian-Cheng Zhang
- Department of Cardiology, Fujian Provincial Hospital, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Hong-Lin Wu
- Department of Cardiology, Fujian Provincial Hospital, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Qian Chen
- Department of Critical Care Medicine Division Four, Fujian Provincial Hospital, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Xiao-Ting Xie
- Department of Cardiology, Fujian Provincial Hospital, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Tian Zou
- Department of Cardiology, Fujian Provincial Hospital, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Chao Zhu
- Department of Cardiology, General Hospital of People's Liberation Army, Beijing, China
| | - Ying Dong
- Department of Cardiology, General Hospital of People's Liberation Army, Beijing, China
| | - Guo-Jian Xiang
- Department of Cardiology, Fujian Provincial Hospital, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Yang Li
- Department of Cardiology, General Hospital of People's Liberation Army, Beijing, China
| | - Peng-Li Zhu
- Department of Geriatric Medicine, Fujian Provincial Center for Geriatrics, Fujian Provincial Hospital, Provincial Clinical Medicine College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
43
|
Panel M, Ghaleh B, Morin D. Mitochondria and aging: A role for the mitochondrial transition pore? Aging Cell 2018; 17:e12793. [PMID: 29888494 PMCID: PMC6052406 DOI: 10.1111/acel.12793] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/15/2018] [Indexed: 12/15/2022] Open
Abstract
The cellular mechanisms responsible for aging are poorly understood. Aging is considered as a degenerative process induced by the accumulation of cellular lesions leading progressively to organ dysfunction and death. The free radical theory of aging has long been considered the most relevant to explain the mechanisms of aging. As the mitochondrion is an important source of reactive oxygen species (ROS), this organelle is regarded as a key intracellular player in this process and a large amount of data supports the role of mitochondrial ROS production during aging. Thus, mitochondrial ROS, oxidative damage, aging, and aging-dependent diseases are strongly connected. However, other features of mitochondrial physiology and dysfunction have been recently implicated in the development of the aging process. Here, we examine the potential role of the mitochondrial permeability transition pore (mPTP) in normal aging and in aging-associated diseases.
Collapse
Affiliation(s)
- Mathieu Panel
- INSERM U955, équipe 3; Créteil France
- Université Paris-Est, UMR_S955, DHU A-TVB, UPEC; Créteil France
| | - Bijan Ghaleh
- INSERM U955, équipe 3; Créteil France
- Université Paris-Est, UMR_S955, DHU A-TVB, UPEC; Créteil France
| | - Didier Morin
- INSERM U955, équipe 3; Créteil France
- Université Paris-Est, UMR_S955, DHU A-TVB, UPEC; Créteil France
| |
Collapse
|
44
|
Cannata' A, Merlo M, Artico J, Gentile P, Camparini L, Cristallini J, Porcari A, Loffredo F, Sinagra G. Cardiovascular aging: the unveiled enigma from bench to bedside. J Cardiovasc Med (Hagerstown) 2018; 19:517-526. [PMID: 30024423 DOI: 10.2459/jcm.0000000000000694] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
: The rapid increase in the median age of the world's population requires particular attention towards older and more fragile people. Cardiovascular risk factors, time and comorbidities play a vicious role in the development of heart failure, both with reduced and preserved ejection fraction, in the elderly. Understanding the mechanisms underlying the pathophysiological processes observed with aging is pivotal to target those patients and their therapeutic needs properly. This review aims to investigate and to dissect the main pathways leading to the aging cardiomyopathy, helping to understand the relationship from bench to bedside of the clinical phenotype.
Collapse
Affiliation(s)
- Antonio Cannata'
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Marco Merlo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Jessica Artico
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Piero Gentile
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Luca Camparini
- International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Jacopo Cristallini
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Aldostefano Porcari
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| | - Francesco Loffredo
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste.,International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Gianfranco Sinagra
- Cardiovascular and Thoracic Department, Azienda Sanitaria Universitaria Integrata di Trieste and University of Trieste
| |
Collapse
|
45
|
Chen Q, Yi Z, Cheng J. Atrial fibrillation in aging population. Aging Med (Milton) 2018; 1:67-74. [PMID: 31942483 PMCID: PMC6880740 DOI: 10.1002/agm2.12015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 04/02/2018] [Indexed: 12/19/2022] Open
Abstract
With aging, the pathogenesis processes of atrial fibrillation (AF) are heightened. In this article, we review the mechanisms that predispose elderly patients to AF. We also highlight the unique features in diagnosis, stroke prevention, and treatment strategies for the elderly patient with AF.
Collapse
Affiliation(s)
- Qi Chen
- Texas Heart InstituteHoustonTXUSA
| | - Zhong Yi
- Department of GeriatricsAerospace Center HospitalBeijingChina
| | | |
Collapse
|
46
|
Guo W, Pencina KM, Gagliano-Jucá T, Jasuja R, Morris N, O'Connell KE, Westmoreland S, Bhasin S. Effects of an ActRIIB.Fc Ligand Trap on Cardiac Function in Simian Immunodeficiency Virus-Infected Male Rhesus Macaques. J Endocr Soc 2018; 2:817-831. [PMID: 30019021 PMCID: PMC6041778 DOI: 10.1210/js.2018-00099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022] Open
Abstract
An important safety consideration in the use of antagonists of myostatin and activins is whether these drugs induce myocardial hypertrophy and impair cardiac function. The current study evaluated the effects of a soluble ActRIIB receptor Fc fusion protein (ActRIIB.Fc), a ligand trap for TGF-β/activin family members including myostatin, on myocardial mass and function in simian immunodeficiency virus (SIV)-infected juvenile rhesus macaques (Macaca mulatta). Fourteen pair-housed, juvenile male rhesus macaques were inoculated with SIVmac239; 4 weeks postinoculation, they were treated with weekly injections of 10 mg/kg ActRIIB.Fc or saline for 12 weeks. Myocardial mass and function were evaluated using two-dimensional echocardiography at baseline and after 12 weeks. The administration of ActRIIB.Fc was associated with a significantly greater increase in thickness of left ventricular posterior wall and interventricular septum both in diastole and systole. Cardiac output and cardiac index increased with time, more in animals treated with ActRIIB.Fc than in those treated with saline, but the difference was not statistically significant. The changes in ejection fraction, fractional shortening, and stroke volume did not differ significantly between groups. The changes in end-diastolic and end-systolic volumes did not differ between groups. In addition to a large reduction in IGF1 mRNA expression in the ActRIIB.Fc-treated animals, complex changes were detected in the myocardial expression of proteins related to calcium transport and storage. In conclusion, ActRIIB.Fc administration for 12 weeks was associated with increased myocardial mass but did not adversely affect myocardial function in juvenile SIV-infected rhesus macaques. Further studies are necessary to establish long-term cardiac safety.
Collapse
Affiliation(s)
- Wen Guo
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Karol M Pencina
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Thiago Gagliano-Jucá
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Ravi Jasuja
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Nancy Morris
- Division of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts
| | - Karyn E O'Connell
- Division of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts
| | - Susan Westmoreland
- Division of Comparative Pathology, New England Primate Research Center, Southborough, Massachusetts
| | - Shalender Bhasin
- Research Program in Men's Health: Aging and Metabolism, Boston Claude D. Pepper Older Americans Independence Center, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
47
|
Argenziano M, Lambers E, Hong L, Sridhar A, Zhang M, Chalazan B, Menon A, Savio-Galimberti E, Wu JC, Rehman J, Darbar D. Electrophysiologic Characterization of Calcium Handling in Human Induced Pluripotent Stem Cell-Derived Atrial Cardiomyocytes. Stem Cell Reports 2018; 10:1867-1878. [PMID: 29731429 PMCID: PMC5989733 DOI: 10.1016/j.stemcr.2018.04.005] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 04/05/2018] [Accepted: 04/06/2018] [Indexed: 11/20/2022] Open
Abstract
Human induced pluripotent stem cell (hiPSC)-derived atrial cardiomyocytes (CMs) hold great promise for elucidating underlying cellular mechanisms that cause atrial fibrillation (AF). In order to use atrial-like hiPSC-CMs for arrhythmia modeling, it is essential to better understand the molecular and electrophysiological phenotype of these cells. We performed comprehensive molecular, transcriptomic, and electrophysiologic analyses of retinoic acid (RA)-guided hiPSC atrial-like CMs and demonstrate that RA results in differential expression of genes involved in calcium ion homeostasis that directly interact with an RA receptor, chicken ovalbumin upstream promoter-transcription factor 2 (COUP-TFII). We report a mechanism by which RA generates an atrial-like electrophysiologic signature through the downstream regulation of calcium channel gene expression by COUP-TFII and modulation of calcium handling. Collectively, our results provide important insights into the underlying molecular mechanisms that regulate atrial-like hiPSC-CM electrophysiology and support the use of atrial-like CMs derived from hiPSCs to model AF. RA-guided differentiation generates atrial-like cardiomyocytes from hiPSCs RA upregulates calcium handling genes associated with COUP-TFII RA modulates calcium handling of atrial-like hiPSC-CMs Atrial-like hiPSC-CMs show reduced calcium currents and channel availability
Collapse
Affiliation(s)
- Mariana Argenziano
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA; Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | - Erin Lambers
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA
| | - Liang Hong
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA
| | - Arvind Sridhar
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA
| | - Meihong Zhang
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA
| | - Brandon Chalazan
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA
| | - Ambili Menon
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA
| | - Eleonora Savio-Galimberti
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA; Lankenau Institute for Medical Research, Wynnewood, PA, USA
| | - Joseph C Wu
- Division of Cardiology, Department of Medicine, Stanford Cardiovascular Institute, Stanford University Medical Center, Stanford, CA, USA
| | - Jalees Rehman
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA
| | - Dawood Darbar
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, 840 S. Wood Street, 920S (MC 715), Chicago, IL 60612, USA; Department of Pharmacology, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
48
|
Pluteanu F, Nikonova Y, Holzapfel A, Herzog B, Scherer A, Preisenberger J, Plačkić J, Scheer K, Ivanova T, Bukowska A, Goette A, Kockskämper J. Progressive impairment of atrial myocyte function during left ventricular hypertrophy and heart failure. J Mol Cell Cardiol 2018; 114:253-263. [DOI: 10.1016/j.yjmcc.2017.11.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 10/22/2017] [Accepted: 11/27/2017] [Indexed: 12/29/2022]
|
49
|
Uzhachenko R, Boyd K, Olivares-Villagomez D, Zhu Y, Goodwin JS, Rana T, Shanker A, Tan WJT, Bondar T, Medzhitov R, Ivanova AV. Mitochondrial protein Fus1/Tusc2 in premature aging and age-related pathologies: critical roles of calcium and energy homeostasis. Aging (Albany NY) 2017; 9:627-649. [PMID: 28351997 PMCID: PMC5391223 DOI: 10.18632/aging.101213] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 03/18/2017] [Indexed: 12/20/2022]
Abstract
Decreased energy production and increased oxidative stress are considered to be major contributors to aging and aging-associated pathologies. The role of mitochondrial calcium homeostasis has also been highlighted as an important factor affecting different pathological conditions. Here, we present evidence that loss of a small mitochondrial protein Fus1 that maintains mitochondrial homeostasis results in premature aging, aging-associated pathologies, and decreased survival. We showed that Fus1KO mice develop multiple early aging signs including lordokyphosis, lack of vigor, inability to accumulate fat, reduced ability to tolerate stress, and premature death. Other prominent pathological changes included low sperm counts, compromised ability of adult stem cells to repopulate tissues, and chronic inflammation. At the molecular level, we demonstrated that mitochondria of Fus1 KO cells have low reserve respiratory capacity (the ability to produce extra energy during sudden energy demanding situations), and show significantly altered dynamics of cellular calcium response. Our recent studies on early hearing and memory loss in Fus1 KO mice combined with the new data presented here suggest that calcium and energy homeostasis controlled by Fus1 may be at the core of its aging-regulating activities. Thus, Fus1 protein and Fus1-dependent pathways and processes may represent new tools and targets for anti-aging strategies.
Collapse
Affiliation(s)
- Roman Uzhachenko
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Kelli Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Danyvid Olivares-Villagomez
- Department of Pathology, Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - Yueming Zhu
- Department of Radiation Oncology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - J Shawn Goodwin
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA
| | - Tanu Rana
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA.,Present address: Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37235, USA
| | - Anil Shanker
- Department of Biochemistry and Cancer Biology, School of Medicine, Meharry Medical College, Nashville, TN 37208, USA.,Department of Surgery, Section of Otolaryngology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Winston J T Tan
- Department of Surgery, Section of Otolaryngology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Tanya Bondar
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 0651, USA
| | - Alla V Ivanova
- Department of Surgery, Section of Otolaryngology, Yale University School of Medicine, New Haven, CT 0651, USA
| |
Collapse
|
50
|
Berridge MJ. Vitamin D deficiency accelerates ageing and age-related diseases: a novel hypothesis. J Physiol 2017; 595:6825-6836. [PMID: 28949008 DOI: 10.1113/jp274887] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/11/2017] [Indexed: 12/24/2022] Open
Abstract
Ageing can occur at different rates, but what controls this variable rate is unknown. Here I have developed a hypothesis that vitamin D may act to control the rate of ageing. The basis of this hypothesis emerged from studyng the various cellular processes that control ageing. These processes such as autophagy, mitochondrial dysfunction, inflammation, oxidative stress, epigenetic changes, DNA disorders and alterations in Ca2+ and reactive oxygen species (ROS) signalling are all known to be regulated by vitamin D. The activity of these processes will be enhanced in individuals that are deficient in vitamin D. Not only will this increase the rate of ageing, but it will also increase the probability of developing age-related diseases such as Alzheimer's disease, Parkinson's disease, multiple sclerosis and cardiovascular disease. In individual with normal vitamin D levels, these ageing-related processes will occur at lower rates resulting in a reduced rate of ageing and enhanced protection against these age-related diseases.
Collapse
|