1
|
Banerjee K, Mandal S, Nath A, Chakraborty SB, Mitra A, Gupta S. Thyroxine (T3)-mediated regulation of early cardiac repair in a chemical-induced hypoxia/reoxygenation model of adult zebrafish (Danio rerio). Wound Repair Regen 2025; 33:e13244. [PMID: 39727215 DOI: 10.1111/wrr.13244] [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: 08/23/2024] [Revised: 11/24/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024]
Abstract
Hypoxia-mediated cardiac tissue injury and its repair or regeneration are one of the major health management challenges globally. Unlike mammals, lower vertebrate species such as zebrafish (Danio rerio) represent a natural model to study cardiac injury, repair and regeneration. Thyroxine (T3) has been hypothesised to be one of the endocrine factors responsible for the evolutionary trade-off for acquiring endothermy and regenerative capability in higher vertebrates. However, the specific targets of T3 during cardiac repair are still obscure. In this study, cardiac injury was generated in adult zebrafish by acute anaemia-induced hypoxia/reoxygenation (H/R) in the presence or absence of exogenous T3 alone or along with 1-850 (inhibitor of T3 receptor) and iopanoic acid (IOA, blocker of T3 release), respectively. A microarray analysis showed that 10,226 gene expression changes in expression across all experimental groups, providing a comprehensive understanding of the cardiac transcriptome. Analysis of 11 candidate genes was conducted using qRT-PCR and the findings aligned with the microarray data. Histological assessment by Masson's trichrome staining and immunofluorescence studies also corroborated the microarray data. GO enrichment analysis showed noteworthy involvement of T3 in the modulation of genes involved in oxidative stress, cardiac fibrosis, energy metabolism, autophagy, apoptosis and regeneration during the initial repair phase (7 days) of H/R-damaged cardiac tissue. Overall, this is the first study that presents a holistic picture of cardiac repair and regeneration post H/R injury in zebrafish and the effect of T3 pre-treatment on it.
Collapse
Affiliation(s)
- Kalyan Banerjee
- Department of Zoology, Trivenidevi Bhalotia College, Raniganj, West Bengal, India
| | - Subhadeep Mandal
- Department of Zoology, Trivenidevi Bhalotia College, Raniganj, West Bengal, India
| | - Arghya Nath
- Department of Zoology, University of Burdwan, Bardhaman, West Bengal, India
| | | | - Arkadeep Mitra
- Department of Zoology, City College, Kolkata, West Bengal, India
| | - Shreyasi Gupta
- Department of Zoology, Trivenidevi Bhalotia College, Raniganj, West Bengal, India
| |
Collapse
|
2
|
Sakamoto T, Kelly DP. Cardiac maturation. J Mol Cell Cardiol 2024; 187:38-50. [PMID: 38160640 PMCID: PMC10923079 DOI: 10.1016/j.yjmcc.2023.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024]
Abstract
The heart undergoes a dynamic maturation process following birth, in response to a wide range of stimuli, including both physiological and pathological cues. This process entails substantial re-programming of mitochondrial energy metabolism coincident with the emergence of specialized structural and contractile machinery to meet the demands of the adult heart. Many components of this program revert to a more "fetal" format during development of pathological cardiac hypertrophy and heart failure. In this review, emphasis is placed on recent progress in our understanding of the transcriptional control of cardiac maturation, encompassing the results of studies spanning from in vivo models to cardiomyocytes derived from human stem cells. The potential applications of this current state of knowledge to new translational avenues aimed at the treatment of heart failure is also addressed.
Collapse
Affiliation(s)
- Tomoya Sakamoto
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Daniel P Kelly
- Cardiovascular Institute, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
3
|
Galow AM, Brenmoehl J, Hoeflich A. Synergistic effects of hormones on structural and functional maturation of cardiomyocytes and implications for heart regeneration. Cell Mol Life Sci 2023; 80:240. [PMID: 37541969 PMCID: PMC10403476 DOI: 10.1007/s00018-023-04894-6] [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: 04/04/2023] [Revised: 07/18/2023] [Accepted: 07/22/2023] [Indexed: 08/06/2023]
Abstract
The limited endogenous regenerative capacity of the human heart renders cardiovascular diseases a major health threat, thus motivating intense research on in vitro heart cell generation and cell replacement therapies. However, so far, in vitro-generated cardiomyocytes share a rather fetal phenotype, limiting their utility for drug testing and cell-based heart repair. Various strategies to foster cellular maturation provide some success, but fully matured cardiomyocytes are still to be achieved. Today, several hormones are recognized for their effects on cardiomyocyte proliferation, differentiation, and function. Here, we will discuss how the endocrine system impacts cardiomyocyte maturation. After detailing which features characterize a mature phenotype, we will contemplate hormones most promising to induce such a phenotype, the routes of their action, and experimental evidence for their significance in this process. Due to their pleiotropic effects, hormones might be not only valuable to improve in vitro heart cell generation but also beneficial for in vivo heart regeneration. Accordingly, we will also contemplate how the presented hormones might be exploited for hormone-based regenerative therapies.
Collapse
Affiliation(s)
- Anne-Marie Galow
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany.
| | - Julia Brenmoehl
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| | - Andreas Hoeflich
- Institute of Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196, Dummerstorf, Germany
| |
Collapse
|
4
|
Tugrul Ersak D, Oluklu D, Uyan Hendem D, Turgut E, Göncü Ayhan Ş, Kara Ö, Şahin D. The assessment of fetal cardiac output in maternal hypothyroidism under levothyroxine treatment. Echocardiography 2022; 39:1434-1438. [PMID: 36266738 DOI: 10.1111/echo.15474] [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: 07/20/2022] [Revised: 09/13/2022] [Accepted: 10/02/2022] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVE In this study, we investigated whether maternal hypothyroidism has a role in the cardiac output (CO) of the fetus or not. METHODS Pregnant women between 33 and 37 gestational weeks known to have hypothyroidism and using levothyroxine were accepted as the case group. Gestational age-matched healthy euthyroid pregnant women constituted the control group. Fetal echocardiography was performed. Diameters and the velocity waveform of the pulmonary artery (PA) and aortic valves were measured. Velocity time integral (VTI) was also measured from the ventricular outflow tract. CO was calculated using VTI × π (Aortic Valve or Pulmonary Valve diameter/2) 2 × heart rate formula. RESULTS The aortic and PA annulus were measured larger in the control group. (p = .003, p = .005, respectively). Furthermore, the right and left CO of the case group were lower than the control group. Whereas the mean combined CO (ml/min) of the case group was 674.8 ± 146.2, it was 827.8 ± 167.9 in the control group (p < .001). Additionally, a negative correlation was observed between thyroid-stimulating hormone and aortic VTI (r:-.480; p:.006). CONCLUSION The findings of our study suggest that the CO of the fetus may be affected by maternal hypothyroidism.
Collapse
Affiliation(s)
- Duygu Tugrul Ersak
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Deniz Oluklu
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Derya Uyan Hendem
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Ezgi Turgut
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Şule Göncü Ayhan
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Özgür Kara
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Dilek Şahin
- Department of Obstetrics and Gynecology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
5
|
Emamnejad R, Dass M, Mahlis M, Bozkurt S, Ye S, Pagnin M, Theotokis P, Grigoriadis N, Petratos S. Thyroid hormone-dependent oligodendroglial cell lineage genomic and non-genomic signaling through integrin receptors. Front Pharmacol 2022; 13:934971. [PMID: 36133808 PMCID: PMC9483185 DOI: 10.3389/fphar.2022.934971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 07/18/2022] [Indexed: 11/13/2022] Open
Abstract
Multiple sclerosis (MS) is a heterogeneous autoimmune disease whereby the pathological sequelae evolve from oligodendrocytes (OLs) within the central nervous system and are targeted by the immune system, which causes widespread white matter pathology and results in neuronal dysfunction and neurological impairment. The progression of this disease is facilitated by a failure in remyelination following chronic demyelination. One mediator of remyelination is thyroid hormone (TH), whose reliance on monocarboxylate transporter 8 (MCT8) was recently defined. MCT8 facilitates the entry of THs into oligodendrocyte progenitor cell (OPC) and pre-myelinating oligodendrocytes (pre-OLs). Patients with MS may exhibit downregulated MCT8 near inflammatory lesions, which emphasizes an inhibition of TH signaling and subsequent downstream targeted pathways such as phosphoinositide 3-kinase (PI3K)-Akt. However, the role of the closely related mammalian target of rapamycin (mTOR) in pre-OLs during neuroinflammation may also be central to the remyelination process and is governed by various growth promoting signals. Recent research indicates that this may be reliant on TH-dependent signaling through β1-integrins. This review identifies genomic and non-genomic signaling that is regulated through mTOR in TH-responsive pre-OLs and mature OLs in mouse models of MS. This review critiques data that implicates non-genomic Akt and mTOR signaling in response to TH-dependent integrin receptor activation in pre-OLs. We have also examined whether this can drive remyelination in the context of neuroinflammation and associated sequelae. Importantly, we outline how novel therapeutic small molecules are being designed to target integrin receptors on oligodendroglial lineage cells and whether these are viable therapeutic options for future use in clinical trials for MS.
Collapse
Affiliation(s)
- Rahimeh Emamnejad
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Mary Dass
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Michael Mahlis
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Salome Bozkurt
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Sining Ye
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Maurice Pagnin
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
| | - Paschalis Theotokis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Nikolaos Grigoriadis
- B’, Department of Neurology, Laboratory of Experimental Neurology and Neuroimmunology, AHEPA University Hospital, Thessaloniki, Greece
| | - Steven Petratos
- Department of Neuroscience, Central Clinical School, Monash University, Prahran, VIC, Australia
- *Correspondence: Steven Petratos,
| |
Collapse
|
6
|
Role of thyroid hormones-induced oxidative stress on cardiovascular physiology. Biochim Biophys Acta Gen Subj 2022; 1866:130239. [PMID: 36064072 DOI: 10.1016/j.bbagen.2022.130239] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 12/21/2021] [Accepted: 08/09/2022] [Indexed: 11/21/2022]
Abstract
Thyroid hormones (THs) play an essential role in the maintenance of cardiovascular homeostasis and are involved in the modulation of cardiac contractility, heart rate, diastolic function, systemic vascular resistance, and vasodilation. THs have actions on cardiovascular physiology through the activation or repression of target genes or the activation of intracellular signals through non-genomic mechanisms. Hyperthyroidism alters certain intracellular pathways involved in the preservation of the structure and functionality of the heart, causing relevant cardiovascular disorders. Reactive oxygen species (ROS) play an important role in the cardiovascular system, but the exacerbated increase in ROS caused by chronic hyperthyroidism together with regulation on the antioxidant system have been associated with the development of cardiovascular dysfunction. In this review, we analyze the role of THs-induced oxidative stress in the cellular and molecular changes that lead to cardiac dysfunction, as well as the effectiveness of antioxidant treatments in attenuating cardiac abnormalities developed during hyperthyroidism.
Collapse
|
7
|
Rosenstock TR, Sun C, Hughes GW, Winter K, Sarkar S. Analysis of Mitochondrial Dysfunction by Microplate Reader in hiPSC-Derived Neuronal Cell Models of Neurodegenerative Disorders. Methods Mol Biol 2022; 2549:1-21. [PMID: 35347693 DOI: 10.1007/7651_2021_451] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mitochondria are responsible for many vital pathways governing cellular homeostasis, including cellular energy management, heme biosynthesis, lipid metabolism, cellular proliferation and differentiation, cell cycle regulation, and cellular viability. Electron transport and ADP phosphorylation coupled with proton pumping through the mitochondrial complexes contribute to the preservation of mitochondrial membrane potential (ΔΨm). Importantly, mitochondrial polarization is essential for reactive oxygen species (ROS) production and cytosolic calcium (Ca2+) handling. Thus, changes in mitochondrial oxidative phosphorylation (OXPHOS), ΔΨm, and ATP/ADP may occur in parallel or stimulate each other. Brain cells like neurons are heavily reliant on mitochondrial OXPHOS for its high-energy demands, and hence improper mitochondrial function is detrimental for neuronal survival. Indeed, several neurodegenerative disorders are associated with mitochondrial dysfunction. Modeling this disease-relevant phenotype in neuronal cells differentiated from patient-derived human induced pluripotent stem cells (hiPSCs) provide an appropriate cellular platform for studying the disease pathology and drug discovery. In this review, we describe high-throughput analysis of crucial parameters related to mitochondrial function in hiPSC-derived neurons. These methodologies include measurement of ΔΨm, intracellular Ca2+, oxidative stress, and ATP/ADP levels using fluorescence probes via a microplate reader. Benefits of such an approach include analysis of mitochondrial parameters on a large population of cells, simultaneous analysis of different cell lines and experimental conditions, and for drug screening to identify compounds restoring mitochondrial function.
Collapse
Affiliation(s)
- Tatiana R Rosenstock
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
- Department of Pharmacology, University of São Paulo, São Paulo, Brazil
- Department of Bioscience, Sygnature Discovery, BioCity, Nottingham, United Kingdom
| | - Congxin Sun
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Georgina Wynne Hughes
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Katherine Winter
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK
| | - Sovan Sarkar
- Institute of Cancer and Genomic Sciences, Institute of Biomedical Research, College of Medical and Dental Sciences, University of Birmingham, Edgbaston, Birmingham, UK.
| |
Collapse
|
8
|
Functional State of Rat Heart Mitochondria in Experimental Hyperthyroidism. Int J Mol Sci 2021; 22:ijms222111744. [PMID: 34769177 PMCID: PMC8583836 DOI: 10.3390/ijms222111744] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 11/16/2022] Open
Abstract
In this work, the effect of thyroxine on energy and oxidative metabolism in the mitochondria of the rat heart was studied. Hyperthyroidism was observed in experimental animals after chronic administration of T4, which was accompanied by an increase in serum concentrations of free triiodothyronine (T3) and thyroxine (T4) by 1.8 and 3.4 times, respectively. The hyperthyroid rats (HR) had hypertrophy of the heart. In HR, there was a change in the oxygen consumption in the mitochondria of the heart, especially when using palmitoylcarnitine. The assay of respiratory chain enzymes revealed that the activities of complexes I, I + III, III, IV increased, whereas the activities of complexes II, II + III decreased in heart mitochondria of the experimental animals. It was shown that the level of respiratory complexes of the electron transport chain in hyperthyroid rats increased, except for complex V, the quantity of which was reduced. The development of oxidative stress in HR was observed: an increase in the hydrogen peroxide production rate, increase in lipid peroxidation and reduced glutathione. The activity of superoxide dismutase in the heart of HR was higher than in the control. At the same time, the activity of glutathione peroxidase decreased. The obtained data indicate that increased concentrations of thyroid hormones lead to changes in energy metabolism and the development of oxidative stress in the heart of rats, which in turn contributes to heart dysfunction.
Collapse
|
9
|
Abstract
Mammalian cardiomyocytes mostly utilize oxidation of fatty acids to generate ATP. The fetal heart, in stark contrast, mostly uses anaerobic glycolysis. During perinatal development, thyroid hormone drives extensive metabolic remodeling in the heart for adaptation to extrauterine life. These changes coincide with critical functional maturation and exit of the cell cycle, making the heart a post-mitotic organ. Here, we review the current understanding on the perinatal shift in metabolism, hormonal status, and proliferative potential in cardiomyocytes. Thyroid hormone and glucocorticoids have roles in adult cardiac metabolism, and both pathways have been implicated as regulators of myocardial regeneration. We discuss the evidence that suggests these processes could be interrelated and how this can help explain variation in cardiac regeneration across ontogeny and phylogeny, and we note what breakthroughs are still to be made.
Collapse
Affiliation(s)
- Niall Graham
- Cardiovascular Research Institute and Department of Physiology, University of California San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - Guo N Huang
- Cardiovascular Research Institute and Department of Physiology, University of California San Francisco, San Francisco, CA 94158, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Correspondence: Guo N Huang, Ph.D., University of California San Francisco, 555 Mission Bay Blvd South, Room 352V, San Francisco, CA 94158, USA.
| |
Collapse
|
10
|
Waldman M, Arad M, Abraham NG, Hochhauser E. The Peroxisome Proliferator-Activated Receptor-Gamma Coactivator-1α-Heme Oxygenase 1 Axis, a Powerful Antioxidative Pathway with Potential to Attenuate Diabetic Cardiomyopathy. Antioxid Redox Signal 2020; 32:1273-1290. [PMID: 32027164 PMCID: PMC7232636 DOI: 10.1089/ars.2019.7989] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023]
Abstract
Significance: From studies of diabetic animal models, the downregulation of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α)-heme oxygenase 1 (HO-1) axis appears to be a crucial event in the development of obesity and diabetic cardiomyopathy (DCM). In this review, we discuss the role of metabolic and biochemical stressors in the rodent and human pathophysiology of DCM. A crucial contributor for many cardiac pathologies is excessive production of reactive oxygen species (ROS) pathologies, which lead to extensive cellular damage by impairing mitochondrial function and directly oxidizing DNA, proteins, and lipid membranes. We discuss the role of ROS production and inflammatory pathways with multiple contributing and confounding factors leading to DCM. Recent Advances: The relevant biochemical pathways that are critical to a therapeutic approach to treat DCM, specifically caloric restriction and its relation to the PGC-1α-HO-1 axis in the attenuation of DCM, are elucidated. Critical Issues: The increased prevalence of diabetes mellitus type 2, a major contributor to unique cardiomyopathy characterized by cardiomyocyte hypertrophy with no effective clinical treatment. This review highlights the role of mitochondrial dysfunction in the development of DCM and potential oxidative targets to attenuate oxidative stress and attenuate DCM. Future Directions: Targeting the PGC-1α-HO-1 axis is a promising approach to ameliorate DCM through improvement in mitochondrial function and antioxidant defenses. A pharmacological inducer to activate PGC-1α and HO-1 described in this review may be a promising therapeutic approach in the clinical setting.
Collapse
Affiliation(s)
- Maayan Waldman
- Cardiac Research Laboratory, Felsenstein Medical Research Institute at Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
- Cardiac Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Ramat Gan, Israel
| | - Michael Arad
- Cardiac Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Ramat Gan, Israel
| | - Nader G. Abraham
- Department of Pharmacology, New York Medical College, Valhalla, New York, USA
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute at Rabin Medical Center, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
11
|
Arad M, Waldman M, Abraham NG, Hochhauser E. Therapeutic approaches to diabetic cardiomyopathy: Targeting the antioxidant pathway. Prostaglandins Other Lipid Mediat 2020; 150:106454. [PMID: 32413571 DOI: 10.1016/j.prostaglandins.2020.106454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 03/23/2020] [Accepted: 05/06/2020] [Indexed: 12/25/2022]
Abstract
The global epidemic of cardiovascular disease continues unabated and remains the leading cause of death both in the US and worldwide. We hereby summarize the available therapies for diabetes and cardiovascular disease in diabetics. Clearly, the current approaches to diabetic heart disease often target the manifestations and certain mediators but not the specific pathways leading to myocardial injury, remodeling and dysfunction. Better understanding of the molecular events determining the evolution of diabetic cardiomyopathy will provide insight into the development of specific and targeted therapies. Recent studies largely increased our understanding of the role of enhanced inflammatory response, ROS production, as well as the contribution of Cyp-P450-epoxygenase-derived epoxyeicosatrienoic acid (EET), Peroxisome Proliferator-Activated Receptor Gamma Coactivator-1α (PGC-1α), Heme Oxygenase (HO)-1 and 20-HETE in pathophysiology and therapy of cardiovascular disease. PGC-1α increases production of the HO-1 which has a major role in protecting the heart against oxidative stress, microcirculation and mitochondrial dysfunction. This review describes the potential drugs and their downstream targets, PGC-1α and HO-1, as major loci for developing therapeutic approaches beside diet and lifestyle modification for the treatment and prevention of heart disease associated with obesity and diabetes.
Collapse
Affiliation(s)
- Michael Arad
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| | - Maayan Waldman
- Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel; Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Tel Aviv, Israel
| | - Nader G Abraham
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595, USA
| | - Edith Hochhauser
- Cardiac Research Laboratory, Felsenstein Medical Research Institute, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
12
|
Wang X, Mao J, Zhou X, Li Q, Gao L, Zhao J. Thyroid Stimulating Hormone Triggers Hepatic Mitochondrial Stress through Cyclophilin D Acetylation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1249630. [PMID: 31998431 PMCID: PMC6970002 DOI: 10.1155/2020/1249630] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 09/01/2019] [Accepted: 09/14/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND & AIMS Oxidative stress-related liver diseases were shown to be associated with elevated serum thyroid stimulating hormone (TSH) levels. Mitochondria are the main source of cellular reactive oxygen species. However, the relationship between TSH and hepatic mitochondrial stress/dysfunction and the underlying mechanisms are largely unknown. Here, we focused on exploring the effects and mechanism of TSH on hepatic mitochondrial stress. METHODS As the function of TSH is mediated through the TSH receptor (TSHR), Tshr -/- mice and liver-specific Tshr -/- mice and liver-specific Tshr -/- mice and liver-specific. RESULTS A relatively lower degree of mitochondrial stress was observed in the livers of Tshr -/- mice and liver-specific in vitro. Microarray and RT-PCR analyses showed that Tshr -/- mice and liver-specific. CONCLUSIONS TSH stimulates hepatic CypD acetylation through the lncRNA-AK044604/SIRT1/SIRT3 signaling pathway, indicating an essential role for TSH in mitochondrial stress in the liver.
Collapse
Affiliation(s)
- Xiaolei Wang
- Shandong Institute of Endocrine & Metabolic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250014, China
| | - Jinbao Mao
- Department of Anesthesiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Xinli Zhou
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Qiu Li
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Ling Gao
- Scientific Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan 250021, China
| | - Jiajun Zhao
- Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
- Shandong Provincial Key Laboratory of Endocrinology and Lipid Metabolism, Jinan 250021, China
| |
Collapse
|
13
|
Protective Effects of Euthyroidism Restoration on Mitochondria Function and Quality Control in Cardiac Pathophysiology. Int J Mol Sci 2019; 20:ijms20143377. [PMID: 31295805 PMCID: PMC6678270 DOI: 10.3390/ijms20143377] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/05/2019] [Accepted: 07/08/2019] [Indexed: 12/18/2022] Open
Abstract
Mitochondrial dysfunctions are major contributors to heart disease onset and progression. Under ischemic injuries or cardiac overload, mitochondrial-derived oxidative stress, Ca2+ dis-homeostasis, and inflammation initiate cross-talking vicious cycles leading to defects of mitochondrial DNA, lipids, and proteins, concurrently resulting in fatal energy crisis and cell loss. Blunting such noxious stimuli and preserving mitochondrial homeostasis are essential to cell survival. In this context, mitochondrial quality control (MQC) represents an expanding research topic and therapeutic target in the field of cardiac physiology. MQC is a multi-tier surveillance system operating at the protein, organelle, and cell level to repair or eliminate damaged mitochondrial components and replace them by biogenesis. Novel evidence highlights the critical role of thyroid hormones (TH) in regulating multiple aspects of MQC, resulting in increased organelle turnover, improved mitochondrial bioenergetics, and the retention of cell function. In the present review, these emerging protective effects are discussed in the context of cardiac ischemia-reperfusion (IR) and heart failure, focusing on MQC as a strategy to blunt the propagation of connected dangerous signaling cascades and limit adverse remodeling. A better understanding of such TH-dependent signaling could provide insights into the development of mitochondria-targeted treatments in patients with cardiac disease.
Collapse
|
14
|
Decreased PGC1-α levels and increased apoptotic protein signaling are associated with the maladaptive cardiac hypertrophy in hyperthyroidism. J Biosci 2018. [DOI: 10.1007/s12038-018-9816-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
15
|
Integrative analysis of differentially expressed genes and miRNAs predicts complex T3-mediated protective circuits in a rat model of cardiac ischemia reperfusion. Sci Rep 2018; 8:13870. [PMID: 30218079 PMCID: PMC6138681 DOI: 10.1038/s41598-018-32237-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 07/12/2018] [Indexed: 12/22/2022] Open
Abstract
Thyroid hormone (T3) dyshomeostasis in the cardiac ischemia-reperfusion (IR) setting negatively impacts on mitochondria function and extracellular matrix remodeling. The modulation of cardiac miRNAs may represent the underlying molecular mechanisms, but a systems biology perspective investigating this critical issue in depth is still lacking. A rat model of myocardial IR, with or without an early short-term T3-replacement, was used to predict putative T3-dependent miRNA-gene interactions targeted to mitochondria quality control and wound healing repair. As evidenced by mRNA and miRNA expression profiling, the T3 supplementation reverted the expression of 87 genes and 11 miRNAs that were dysregulated in the untreated group. In silico crossing and functional analysis of the T3-associated differentially expressed transcripts, identified a signature of interconnected miRNA-gene regulatory circuits that confer resistance to noxious cascades of acute stress. In this network the T3-down-regulated Tp53, Jun and Sp1 transcription factors emerge as critical nodes linking intrinsic cell death and oxidative stress pathways to adverse remodeling cascades. The data presented here provide a novel insight into the molecular basis of T3 cardioprotection in the early post-IR phase and highlight the contribution of a previously unappreciated complex T3-regulatory network that may be helpful in translating T3 replacement into clinical practice.
Collapse
|
16
|
Underlying mechanism of the contractile dysfunction in atrophied ventricular myocytes from a murine model of hypothyroidism. Cell Calcium 2018; 72:26-38. [DOI: 10.1016/j.ceca.2018.01.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Revised: 01/18/2018] [Accepted: 01/31/2018] [Indexed: 11/20/2022]
|
17
|
Applying a systems approach to thyroid physiology: Looking at the whole with a mitochondrial perspective instead of judging single TSH values or why we should know more about mitochondria to understand metabolism. BBA CLINICAL 2017; 7:127-140. [PMID: 28417080 PMCID: PMC5390562 DOI: 10.1016/j.bbacli.2017.03.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 03/20/2017] [Accepted: 03/28/2017] [Indexed: 12/17/2022]
Abstract
Classical thinking in endocrine physiology squeezes our diagnostic handling into a simple negative feedback mechanism with a controller and a controlled variable. In the case of the thyroid this is reduced to TSH and fT3 and fT4, respectively. The setting of this tight notion has no free space for any additions. In this paper we want to challenge this model of limited application by proposing a construct based on a systems approach departing from two basic considerations. In first place since the majority of cases of thyroid disease develop and appear during life it has to be considered as an acquired condition. In the second place, our experience with the reversibility of morphological changes makes the autoimmune theory inconsistent. While medical complexity can expand into the era of OMICS as well as into one where manipulations with the use of knock-outs and -ins are common in science, we have preferred to maintain a simple and practical approach. We will describe the interactions of iron, magnesium, zinc, selenium and coenzyme Q10 with the thyroid axis. The discourse will be then brought into the context of ovarian function, i.e. steroid hormone production. Finally the same elemental players will be presented in relation to the basic mitochondrial machinery that supports the endocrine. We propose that an intact mitochondrial function can guard the normal endocrine function of both the thyroid as well as of the ovarian axis. The basic elements required for this function appear to be magnesium and iron. In the case of the thyroid, magnesium-ATP acts in iodine uptake and the heme protein peroxidase in thyroid hormone synthesis. A similar biochemical process is found in steroid synthesis with cholesterol uptake being the initial energy-dependent step and later the heme protein ferredoxin 1 which is required for steroid synthesis. Magnesium plays a central role in determining the clinical picture associated with thyroid disease and is also involved in maintaining fertility. With the aid of 3D sonography patients needing selenium and/or coenzyme Q10 can be easily identified. By this we firmly believe that physicians should know more about basic biochemistry and the way it fits into mitochondrial function in order to understand metabolism. Contemplating only TSH is highly reductionistic. Outline Author's profiles and motivation for this analysis The philosophical alternatives in science and medicine Reductionism vs. systems approach in clinical thyroid disease guidelines The entry into complexity: the involvement of the musculoskeletal system Integrating East and West: teachings from Chinese Medicine and from evidence based medicine (EBM) Can a mathematical model represent complexity in the daily thyroid practice? How effective is thyroxine treatment? Resolving the situation of residual symptoms in treated patients with thyroid disease Importance of iron, zinc and magnesium in relation to thyroid function Putting together new concepts related to thyroid function for a systems approach Expanding our model into general aspects of medicine
Collapse
|
18
|
Kadlec AO, Chabowski DS, Ait-Aissa K, Gutterman DD. Role of PGC-1α in Vascular Regulation: Implications for Atherosclerosis. Arterioscler Thromb Vasc Biol 2016; 36:1467-74. [PMID: 27312223 DOI: 10.1161/atvbaha.116.307123] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 06/02/2016] [Indexed: 11/16/2022]
Abstract
Mitochondrial dysfunction results in high levels of oxidative stress and mitochondrial damage, leading to disruption of endothelial homeostasis. Recent discoveries have clarified several pathways, whereby mitochondrial dysregulation contributes to endothelial dysfunction and vascular disease burden. One such pathway centers around peroxisome proliferator receptor-γ coactivator 1α (PGC-1α), a transcriptional coactivator linked to mitochondrial biogenesis and antioxidant defense, among other functions. Although primarily investigated for its therapeutic potential in obesity and skeletal muscle differentiation, the ability of PGC-1α to alter a multitude of cellular functions has sparked interest in its role in the vasculature. Within this context, recent studies demonstrate that PGC-1α plays a key role in endothelial cell and smooth muscle cell regulation through effects on oxidative stress, apoptosis, inflammation, and cell proliferation. The ability of PGC-1α to affect these parameters is relevant to vascular disease progression, particularly in relation to atherosclerosis. Upregulation of PGC-1α can prevent the development of, and even encourage regression of, atherosclerotic lesions. Therefore, PGC-1α is poised to serve as a promising target in vascular disease. This review details recent findings related to PGC-1α in vascular regulation, regulation of PGC-1α itself, the role of PGC-1α in atherosclerosis, and therapies that target this key protein.
Collapse
Affiliation(s)
- Andrew O Kadlec
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.)
| | - Dawid S Chabowski
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.)
| | - Karima Ait-Aissa
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.)
| | - David D Gutterman
- From the Department of Physiology (A.O.K., D.D.G.), Division of Cardiology, Department of Medicine (D.S.C., K.A.-A., D.D.G.), and Cardiovascular Center (A.O.K., D.S.C., K.A.-A., D.D.G.), Medical College of Wisconsin, Milwaukee; and Department of Veterans Administration Medical Center, Milwaukee, WI (D.D.G.).
| |
Collapse
|
19
|
Low T3 State Is Correlated with Cardiac Mitochondrial Impairments after Ischemia Reperfusion Injury: Evidence from a Proteomic Approach. Int J Mol Sci 2015; 16:26687-705. [PMID: 26561807 PMCID: PMC4661832 DOI: 10.3390/ijms161125973] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Revised: 10/13/2015] [Accepted: 10/26/2015] [Indexed: 11/22/2022] Open
Abstract
Mitochondria are major determinants of cell fate in ischemia/reperfusion injury (IR) and common effectors of cardio-protective strategies in cardiac ischemic disease. Thyroid hormone homeostasis critically affects mitochondrial function and energy production. Since a low T3 state (LT3S) is frequently observed in the post infarction setting, the study was aimed to investigate the relationship between 72 h post IR T3 levels and both the cardiac function and the mitochondrial proteome in a rat model of IR. The low T3 group exhibits the most compromised cardiac performance along with the worst mitochondrial activity. Accordingly, our results show a different remodeling of the mitochondrial proteome in the presence or absence of a LT3S, with alterations in groups of proteins that play a key role in energy metabolism, quality control and regulation of cell death pathways. Overall, our findings highlight a relationship between LT3S in the early post IR and poor cardiac and mitochondrial outcomes, and suggest a potential implication of thyroid hormone in the cardio-protection and tissue remodeling in ischemic disease.
Collapse
|
20
|
Ghanbari M, Jeddi S, Bagheripuor F, Ghasemi A. The effect of maternal hypothyroidism on cardiac function and tolerance to ischemia-reperfusion injury in offspring male and female rats. J Endocrinol Invest 2015; 38:915-22. [PMID: 25823371 DOI: 10.1007/s40618-015-0267-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 03/03/2015] [Indexed: 01/13/2023]
Abstract
PURPOSE Accumulating evidence indicates that intrauterine evolution disturbance can contribute to myocardial ischemia reperfusion (IR) injury; in addition, thyroid hormones (THs) have a crucial role in the development of different systems during fetal life. The aim of this study was to determine the effect of TH deficiency during fetal life on tolerance of isolated heart to ischemia during adulthood in both genders. METHODS Hypothyroidism was induced in pregnant Wistar rats by administrating 0.025 % 6-propyl-2-thiouracil in drinking water throughout pregnancy. Offspring of rats with maternal hypothyroidism (MH) and control groups were tested in adulthood. Isolated hearts were perfused with Langendorff setup and exposed to 30 min of ischemia, followed by 45 min of reperfusion. Baseline values of the left ventricular end-diastolic pressure (LVEDP), left ventricular developed pressure (LVDP), heart rate (HR), and peak rates of positive and negative changes in left ventricular pressure (±dp/dt) were recorded. RESULTS In the MH groups the baseline levels of LVDP (male: 23 %, female: 33 %), HR (male: 31 %, female: 26 %), and ±dp/dt were significantly (p < 0.01) lower, compared to controls. After ischemia, hearts from male rats with MH had less tolerance to IR injury as assessed in terms of reductions in recovery of hemodynamic parameters compared to controls, while in female rats there were no significant differences between MH and controls. CONCLUSIONS MH decreases hemodynamic parameters in the heart of both male and female offspring in adulthood; in addition, hearts of male rats with MH show less tolerance to ischemia, compared to those of females.
Collapse
Affiliation(s)
- M Ghanbari
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, No. 24, Parvaneh Street, Velenjak, 1985717413, Tehran, Iran
| | | | | | | |
Collapse
|
21
|
Negro A, Boehm M. Cardiomyocyte maturation: It takes a village to raise a kid. J Mol Cell Cardiol 2014; 74:193-5. [PMID: 24874422 DOI: 10.1016/j.yjmcc.2014.05.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 11/25/2022]
Affiliation(s)
- Alejandra Negro
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, NIH, 10 Center Drive, MSC 1454, Building 10-CRC, Room 5E-3232, Bethesda, MD 20892-1454, USA.
| | - Manfred Boehm
- Center for Molecular Medicine, NHLBI-NIH, 10 Center Drive, MSC 1454, Building 10-CRC, RM 5 East 3132, Bethesda, MD 20892, USA.
| |
Collapse
|
22
|
New insights into mechanisms of cardioprotection mediated by thyroid hormones. J Thyroid Res 2013; 2013:264387. [PMID: 23555069 PMCID: PMC3608184 DOI: 10.1155/2013/264387] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 02/14/2013] [Indexed: 01/19/2023] Open
Abstract
Heart failure represents the final common outcome in cardiovascular diseases. Despite significant therapeutic advances, morbidity and mortality of heart failure remain unacceptably high. Heart failure is preceded and sustained by a process of structural remodeling of the entire cardiac tissue architecture. Prevention or limitation of cardiac remodeling in the early stages of the process is a crucial step in order to ameliorate patient prognosis. Acquisition of novel pathophysiological mechanisms of cardiac remodeling is therefore required to develop more efficacious therapeutic strategies. Among all neuroendocrine systems, thyroid hormone seems to play a major homeostatic role in cardiovascular system. In these years, accumulating evidence shows that the “low triiodothyronine” syndrome is a strong prognostic, independent predictor of death in patients affected by both acute and chronic heart disease. In experimental models of cardiac hypertrophy or myocardial infarction, alterations in the thyroid hormone signaling, concerning cardiac mitochondrion, cardiac interstitium, and vasculature, have been suggested to be related to heart dysfunction. The aim of this brief paper is to highlight new developments in understanding the cardioprotective role of thyroid hormone in reverting regulatory networks involved in adverse cardiac remodeling. Furthermore, new recent advances on the role of specific miRNAs in thyroid hormone regulation at mitochondrion and interstitial level are also discussed.
Collapse
|
23
|
Thyroid hormones are involved in 5'-nucleotidase modulation in soluble fraction of cardiac tissue. Life Sci 2012; 91:137-42. [PMID: 22771697 DOI: 10.1016/j.lfs.2012.06.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2012] [Revised: 06/20/2012] [Accepted: 06/22/2012] [Indexed: 11/23/2022]
Abstract
AIMS To investigate the role of TH (thyroid hormones) in 5'-nucleotidase activity and expression in cardiac soluble fraction (SF). MAIN METHODS Male Wistar rats received daily injections of T4 (10, 25 or 50 μg T4/100g body weight) for 14 days to develop a hyperthyroidism condition. Thyroidectomy was performed in other animals to mimic hypothyroidism, and 14 days after surgery they were submitted to TH replacement therapy. KEY FINDINGS T4 reduced the 5'-nucleotidase activity (T4-25, P<0.05 and T4-50, P<0.01) in the SF. Conversely, hypothyroidism significantly increased the 5'-nucleotidase activity in this fraction (P<0.001) and TH replacement therapy reversed the latter result (P<0.001 compared to hypothyroid group). The analysis of protein expression in the SF showed that 5'-nucleotidase was more expressed in hypothyroid than in the control group and that the phosphorylated state of PKC observed in this condition may contribute to a possible mechanism of 5'-nucleotidase modulation by thyroid status. SIGNIFICANCE Taken together, these data reveal that TH can influence adenosine production by modulating 5'-nucleotidase activity and expression, which may contribute to the cardioprotective effect and the maintenance of cardiac function under TH privation.
Collapse
|
24
|
Xu W, Hou D, Jiang X, Lu Z, Guo T, Liu Y, Wang D, Zen K, Yu B, Zhang CY. The protective role of peroxisome proliferator-activated receptor γ coactivator-1α in hyperthyroid cardiac hypertrophy. J Cell Physiol 2012; 227:3243-53. [DOI: 10.1002/jcp.24015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
25
|
Forini F, Lionetti V, Ardehali H, Pucci A, Cecchetti F, Ghanefar M, Nicolini G, Ichikawa Y, Nannipieri M, Recchia FA, Iervasi G. Early long-term L-T3 replacement rescues mitochondria and prevents ischemic cardiac remodelling in rats. J Cell Mol Med 2011; 15:514-24. [PMID: 20100314 PMCID: PMC3922373 DOI: 10.1111/j.1582-4934.2010.01014.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
3,5,3′-Levo-triiodothyronine (L-T3) is essential for DNA transcription, mitochondrial biogenesis and respiration, but its circulating levels rapidly decrease after myocardial infarction (MI). The main aim of our study was to test whether an early and sustained normalization of L-T3 serum levels after MI exerts myocardial protective effects through a mitochondrial preservation. Seventy-two hours after MI induced by anterior interventricular artery ligation, rats were infused with synthetic L-T3 (1.2 μg/kg/day) or saline over 4 weeks. Compared to saline, L-T3 infusion restored FT3 serum levels at euthyroid state (3.0 ± 0.2 versus 4.2 ± 0.3 pg/ml), improved left ventricular (LV) ejection fraction (39.5 ± 2.5 versus 65.5 ± 6.9%), preserved LV end-systolic wall thickening in the peri-infarct zone (6.34 ± 3.1 versus 33.7 ± 6.21%) and reduced LV infarct-scar size by approximately 50% (all P < 0.05). Moreover, L-T3 significantly increased angiogenesis and cell survival and enhanced the expression of nuclear-encoded transcription factors involved in these processes. Finally, L-T3 significantly increased the expression of factors involved in mitochondrial DNA transcription and biogenesis, such as hypoxic inducible factor-1α, mitochondrial transcription factor A and peroxisome proliferator activated receptor γ coactivator-1α, in the LV peri-infarct zone. To further explore mechanisms of L-T3 protective effects, we exposed isolated neonatal cardiomyocytes to H2O2 and found that L-T3 rescued mitochondrial biogenesis and function and protected against cell death via a mitoKATP dependent pathway. Early and sustained physiological restoration of circulating L-T3 levels after MI halves infarct scar size and prevents the progression towards heart failure. This beneficial effect is likely due to enhanced capillary formation and mitochondrial protection.
Collapse
|
26
|
Lanza IR, Sreekumaran Nair K. Regulation of skeletal muscle mitochondrial function: genes to proteins. Acta Physiol (Oxf) 2010; 199:529-47. [PMID: 20345409 DOI: 10.1111/j.1748-1716.2010.02124.x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The impact of ageing on mitochondrial function and the deterministic role of mitochondria on senescence continue to be topics of vigorous debate. Many studies report that skeletal muscle mitochondrial content and function are reduced with ageing and metabolic diseases associated with insulin resistance. However, an accumulating body of literature suggests that physical inactivity typical of ageing may be a more important determinant of mitochondrial function than chronological age, per se. Reports of age-related declines in mitochondrial function have spawned a vast body of literature devoted to understanding the underlying mechanisms. These mechanisms include decreased abundance of mtDNA, reduced mRNA levels, as well as decreased synthesis and expression of mitochondrial proteins, ultimately resulting in decreased function of the whole organelle. Effective therapies to prevent, reverse or delay the onset of the aforementioned mitochondrial changes, regardless of their inevitability or precise underlying causes, require an intimate understanding of the processes that regulate mitochondrial biogenesis, which necessitates the coordinated regulation of nuclear and mitochondrial genomes. Herein we review the current thinking on regulation of mitochondrial biogenesis by transcription factors and transcriptional co-activators and the role of hormones and exercise in initiating this process. We review how exercise may help preserve mitochondrial content and functionality across the lifespan, and how physical inactivity is emerging as a major determinant of many age-associated changes at the level of the mitochondrion. We also review evidence that some mitochondrial changes with ageing are independent of exercise or physical activity and appear to be inevitable consequences of old age.
Collapse
Affiliation(s)
- I R Lanza
- Endocrinology Research Unit, Division of Endocrinology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA
| | | |
Collapse
|
27
|
Terman A, Kurz T, Navratil M, Arriaga EA, Brunk UT. Mitochondrial turnover and aging of long-lived postmitotic cells: the mitochondrial-lysosomal axis theory of aging. Antioxid Redox Signal 2010; 12:503-35. [PMID: 19650712 PMCID: PMC2861545 DOI: 10.1089/ars.2009.2598] [Citation(s) in RCA: 362] [Impact Index Per Article: 24.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2009] [Revised: 07/22/2009] [Accepted: 08/02/2009] [Indexed: 12/19/2022]
Abstract
It is now generally accepted that aging and eventual death of multicellular organisms is to a large extent related to macromolecular damage by mitochondrially produced reactive oxygen species, mostly affecting long-lived postmitotic cells, such as neurons and cardiac myocytes. These cells are rarely or not at all replaced during life and can be as old as the whole organism. The inherent inability of autophagy and other cellular-degradation mechanisms to remove damaged structures completely results in the progressive accumulation of garbage, including cytosolic protein aggregates, defective mitochondria, and lipofuscin, an intralysosomal indigestible material. In this review, we stress the importance of crosstalk between mitochondria and lysosomes in aging. The slow accumulation of lipofuscin within lysosomes seems to depress autophagy, resulting in reduced turnover of effective mitochondria. The latter not only are functionally deficient but also produce increased amounts of reactive oxygen species, prompting lipofuscinogenesis. Moreover, defective and enlarged mitochondria are poorly autophagocytosed and constitute a growing population of badly functioning organelles that do not fuse and exchange their contents with normal mitochondria. The progress of these changes seems to result in enhanced oxidative stress, decreased ATP production, and collapse of the cellular catabolic machinery, which eventually is incompatible with survival.
Collapse
Affiliation(s)
- Alexei Terman
- Department of Clinical Pathology and Cytology, Karolinska University Hospital, Huddinge, Stockholm, Sweden.
| | | | | | | | | |
Collapse
|
28
|
Attia RR, Connnaughton S, Boone LR, Wang F, Elam MB, Ness GC, Cook GA, Park EA. Regulation of pyruvate dehydrogenase kinase 4 (PDK4) by thyroid hormone: role of the peroxisome proliferator-activated receptor gamma coactivator (PGC-1 alpha). J Biol Chem 2009; 285:2375-85. [PMID: 19948729 DOI: 10.1074/jbc.m109.039081] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
PDK4 (pyruvate dehydrogenase kinase 4) regulates pyruvate oxidation through the phosphorylation and inhibition of the pyruvate dehydrogenase complex (PDC). PDC catalyzes the conversion of pyruvate to acetyl-CoA and is an important control point in glucose and pyruvate metabolism. PDK4 gene expression is stimulated by thyroid hormone (T(3)), glucocorticoids, and long chain fatty acids. The effects of T(3) on gene expression in the liver are mediated via the thyroid hormone receptor. Here, we have identified two binding sites for thyroid hormone receptor beta in the promoter of the rat PDK4 (rPDK4) gene. In addition, we have investigated the role of transcriptional coactivators and found that the PGC-1 alpha (peroxisome proliferator-activated receptor gamma coactivator) enhances the T(3) induction of rPDK4. Following T(3) administration, there is an increase in the association of PGC-1 alpha with the rPDK4 promoter. Interestingly, this increased association is with the proximal rPDK4 promoter rather than the distal region of the gene that contains the T(3) response elements. Administration of T(3) to hypothyroid rats elevated the abundance of PGC-1 alpha mRNA and protein in the liver. In addition, we observed greater association of PGC-1 alpha not only with the rPDK4 gene but also with phosphoenolpyruvate carboxykinase and CPT-1a (carnitine palmitoyltransferase 1a) genes. Knockdown of PGC-1 alpha in rat hepatocytes reduced the T(3) induction of PDK4, PEPCK, and CPT-1a genes. Our results indicate that T(3) regulates PGC-1 alpha abundance and association with hepatic genes, and in turn PGC-1 alpha is an important participant in the T(3) induction of selected genes.
Collapse
Affiliation(s)
- Ramy R Attia
- Department of Pharmacology, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, USA
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Marín-García J. Thyroid hormone and myocardial mitochondrial biogenesis. Vascul Pharmacol 2009; 52:120-30. [PMID: 19857604 DOI: 10.1016/j.vph.2009.10.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2009] [Accepted: 10/18/2009] [Indexed: 10/20/2022]
Abstract
Mitochondria have been central in the development of some of the most important ideas in modern biology. Since the discovery that mitochondria have its own DNA and specific mutations and deletions were found in association with neuromuscular and heart diseases, as well as in aging, an extraordinary number of publications have followed, and the term mitochondrial medicine was coined. Recently, it has been found that thyroid hormone (TH) stimulates cardiac mitochondrial biogenesis increasing myocardial mitochondrial mass, mitochondrial respiration, oxidative phosphorylation (OXPHOS), enzyme activities, mitochondrial protein synthesis (by stimulation in a T3-dependent manner), cytochrome, phospholipid and mtDNA content. Also, TH therapy may modulate cardiac mitochondrial protein-import apparatus. To identify the sequence of events, molecules and signaling pathways that is activated by TH affecting mitochondrial structure, biogenesis and function further research is warranted.
Collapse
Affiliation(s)
- José Marín-García
- The Molecular Cardiology and Neuromuscular Institute, 75 Raritan Avenue, Highland Park, NJ 08904, USA.
| |
Collapse
|
30
|
López-Lluch G, Irusta PM, Navas P, de Cabo R. Mitochondrial biogenesis and healthy aging. Exp Gerontol 2008; 43:813-9. [PMID: 18662766 DOI: 10.1016/j.exger.2008.06.014] [Citation(s) in RCA: 267] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2008] [Accepted: 06/26/2008] [Indexed: 02/09/2023]
Abstract
Aging is associated with an overall loss of function at the level of the whole organism that has origins in cellular deterioration. Most cellular components, including mitochondria, require continuous recycling and regeneration throughout the lifespan. Mitochondria are particularly susceptive to damage over time as they are the major bioenergetic machinery and source of oxidative stress in cells. Effective control of mitochondrial biogenesis and turnover, therefore, becomes critical for the maintenance of energy production, the prevention of endogenous oxidative stress and the promotion of healthy aging. Multiple endogenous and exogenous factors regulate mitochondrial biogenesis through the peroxisome proliferator-activated receptor gamma coactivator-1alpha (PGC-1alpha). Activators of PGC-1alpha include nitric oxide, CREB and AMPK. Calorie restriction (CR) and resveratrol, a proposed CR mimetic, also increase mitochondrial biogenesis through activation of PGC-1alpha. Moderate exercise also mimics CR by inducing mitochondrial biogenesis. Negative regulators of PGC-1alpha such as RIP140 and 160MBP suppress mitochondrial biogenesis. Another mechanism involved in mitochondrial maintenance is mitochondrial fission/fusion and this process also involves an increasing number of regulatory proteins. Dysfunction of either biogenesis or fission/fusion of mitochondria is associated with diseases of the neuromuscular system and aging, and a greater understanding of the regulation of these processes should help us to ultimately control the aging process.
Collapse
Affiliation(s)
- Guillermo López-Lluch
- Centro Andaluz de Biología del Desarrollo, Universidad Pablo de Olavide-CSIC, Carretera de Utrera Km 1, 41013 Sevilla, Spain
| | | | | | | |
Collapse
|
31
|
Fernández-Vizarra E, Enriquez JA, Pérez-Martos A, Montoya J, Fernández-Silva P. Mitochondrial gene expression is regulated at multiple levels and differentially in the heart and liver by thyroid hormones. Curr Genet 2008; 54:13-22. [PMID: 18481068 DOI: 10.1007/s00294-008-0194-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Revised: 04/24/2008] [Accepted: 04/27/2008] [Indexed: 01/17/2023]
Abstract
Biogenesis of the oxidative phosphorylation system (OXPHOS) requires the coordinated expression of the nuclear and the mitochondrial genomes. Thyroid hormones play an important role in cell growth and differentiation and are one of the main effectors in mitochondrial biogenesis. To determine how mtDNA expression is regulated, we have investigated the response of two different tissues, the heart and liver, to the thyroid hormone status in vivo and in vitro. We show here that mtDNA expression is a tightly regulated process and that several levels of control can take place simultaneously. In addition, we show that the mechanisms operating in the control of mtDNA expression and their relevance differ between the two tissues, being gene dosage important only in heart while transcription rate and translation efficiency have more weight in liver cells. Another interesting difference is the lack of a direct effect of thyroid hormones on heart mitochondrial transcription.
Collapse
Affiliation(s)
- Erika Fernández-Vizarra
- Departamento de Bioquímica y Biología Molecular y Celular, Universidad de Zaragoza, Miguel Servet 177, Zaragoza, Spain
| | | | | | | | | |
Collapse
|
32
|
Pantos C, Mourouzis I, Xinaris C, Papadopoulou-Daifoti Z, Cokkinos D. Thyroid hormone and “cardiac metamorphosis”: Potential therapeutic implications. Pharmacol Ther 2008; 118:277-94. [DOI: 10.1016/j.pharmthera.2008.02.011] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Accepted: 02/29/2008] [Indexed: 10/22/2022]
|
33
|
Ventura-Clapier R, Garnier A, Veksler V. Transcriptional control of mitochondrial biogenesis: the central role of PGC-1alpha. Cardiovasc Res 2008; 79:208-17. [PMID: 18430751 DOI: 10.1093/cvr/cvn098] [Citation(s) in RCA: 689] [Impact Index Per Article: 40.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although the concept of energy starvation in the failing heart was proposed decades ago, still very little is known about the origin of energetic failure. Recent advances in molecular biology have started to elucidate the transcriptional events governing mitochondrial biogenesis. In particular, a great step was taken with the discovery that peroxisome proliferator-activated receptor gamma co-activator (PGC-1alpha) is the master regulator of mitochondrial biogenesis. The molecular mechanisms underlying the downregulation of PGC-1alpha and the consequent decrease in mitochondrial function in heart failure are, however, still poorly understood. Indeed, the main pathways involved in mitochondrial biogenesis are thought to be up- rather than down-regulated in pathological hypertrophy and heart failure. The current review summarizes recent advances in this field and is restricted to the heart when cardiac data are available.
Collapse
|
34
|
Abstract
Thyroid hormone has various effects on the cardiovascular system and its effects on cardiac contractility, heart rhythm and vascular function has long been recognized. However, new evidence is emerged on the importance of thyroid hormone in the response of the myocardium to ischaemic stress and cardiac remodelling following myocardial infarction. Based on this new information, this review highlights the role of thyroid hormone in myocardial ischaemia and cardiac remodelling, the possible underlying mechanisms and the potential therapeutic implications. Thyroid hormone or analogs may prove new therapeutic agents for treating ischaemic heart disease.
Collapse
Affiliation(s)
- Constantinos Pantos
- Department of Pharmacology, University of Athens, 75 Mikras Asias Avenue, 11527 Goudi, Athens, Greece.
| | | | | | | |
Collapse
|
35
|
Abstract
Thyroid hormone regulates cardiac metabolism through multiple mechanisms. Traditionally, most cardiac metabolic studies have focused on presumed transcriptional actions by defining thyroid hormone-induced changes in mRNA or protein levels. Recent studies have established metabolic pathways in heart that rapidly respond to thyroid hormone. Functions have also been implicated for thyroid hormone receptors, which are separate from their transcriptional actions. Finally, thyroid through ligand binding may play a direct role in transactivation of mitochondrial DNA. This review will explore these newly identified modes of thyroid action on metabolism in heart.
Collapse
Affiliation(s)
- Michael A Portman
- Department of Cardiology, Children's Hospital and Regional Medical Center, Department of Pediatrics, University of Washington, Seattle, Washington 98105, USA.
| |
Collapse
|
36
|
Abstract
Myocardial protection aims at preventing myocardial tissue loss: (a) In the acute stage, i.e., during primary angioplasty in acute myocardial infarction. In this setup, the attenuation of reperfusion injury is the main target. As a "mechanical" means, post-conditioning has already been tried in man with encouraging results. Pharmacologic interventions that could be of promise are statins, insulin, peptide hormones, including erythropoietin, fibroblast growth factor, and many others. (b) The patient with chronic coronary artery disease offers another paradigm, with the target of avoidance of further myocyte loss through apoptosis and inflammation. Various pharmacologic agents may prove useful in this context, together with exercise and "mechanical" improvement of cardiac function with attenuation of myocardial stretch, which by itself is a noxious influence. A continuous effort toward acute and chronically preserving myocardial integrity is a concept concerning both the researcher and the clinician.
Collapse
Affiliation(s)
- Dennis V Cokkinos
- 1st Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece.
| | | |
Collapse
|
37
|
Kokkinos A, Mourouzis I, Kyriaki D, Pantos C, Katsilambros N, Cokkinos DV. Possible implications of leptin, adiponectin and ghrelin in the regulation of energy homeostasis by thyroid hormone. Endocrine 2007; 32:30-2. [PMID: 17992599 DOI: 10.1007/s12020-007-9002-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2007] [Revised: 08/30/2007] [Accepted: 09/06/2007] [Indexed: 10/22/2022]
Abstract
Thyroid hormone plays a critical role in energy homeostasis through mechanisms, which are not fully understood. In the present study, we investigated possible alterations of important energy regulators such as leptin, adiponectin, and ghrelin in relation to changes in thyroid hormones. Thyroid hormone (250 microg/kg) was administered in male Wistar rats for 2 weeks (THYR), while hypothyroidism (HYPO) was induced by propylthiouracil administration (0.05% in drinking water) for 3 weeks. Untreated animals served as controls (NORM). Leptin and adiponectin were measured in plasma by ELISA, while total ghrelin was measured with RIA. Body weight was significantly reduced both in THYR and HYPO rats, while food intake was significantly increased in THYR and decreased in HYPO. This response was associated with various changes in leptin, adiponectin, and ghrelin in plasma. In fact, in THYR rats, leptin levels (mean +/- SEM) were 240 +/- 55 pg/ml as compared to 819 +/- 70 pg/ml in untreated rats (P < 0.05), while no changes were observed in ghrelin and adiponectin. In HYPO rats, leptin levels were 1400 +/- 200 pg/ml vs. 819 +/- 70 pg/ml in untreated rats (P < 0.05), while ghrelin and adiponectin were significantly increased in HYPO rats as compared to untreated rats (P < 0.05). Furthermore, T(3) and T(4) levels were inversely correlated to leptin (P = 0.014), while ghrelin and adiponectin were inversely correlated to weight changes (P = 0.05 and P = 0.03, respectively). In conclusion, leptin seems mainly to be involved in the thyroid hormone effects on energy homeostasis. Ghrelin and adiponectin may serve a compensatory physiological role in hypothyroidism.
Collapse
Affiliation(s)
- Alexander Kokkinos
- 1st Department of Propaedeutic Medicine, Laiko General Hospital, University of Athens Medical School, 11527, Goudi, Athens, Greece
| | | | | | | | | | | |
Collapse
|
38
|
Athéa Y, Garnier A, Fortin D, Bahi L, Veksler V, Ventura-Clapier R. Mitochondrial and energetic cardiac phenotype in hypothyroid rat. Relevance to heart failure. Pflugers Arch 2007; 455:431-42. [PMID: 17638011 PMCID: PMC4710782 DOI: 10.1007/s00424-007-0307-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2007] [Revised: 05/16/2007] [Accepted: 06/11/2007] [Indexed: 12/24/2022]
Abstract
Changes in thyroid status are associated with profound alterations in biochemical and physiological functioning of cardiac muscle, although its impact on cardiac energy metabolism is still debated. Similarities between the changes in cardiac gene expression in pathological hypertrophy leading to heart failure and hypothyroidism prompted scientists to suggest a role for thyroid hormone status in the development of metabolic and functional alterations in this disease. We thus investigated the effects of hypothyroidism on cardiac energy metabolism. Hypothyroid state (HYPO) was induced by thyroidectomy and propyl-thio-uracyl in male rats for 3 weeks. We examined the effects of hypothyroid state on oxidative capacity and mitochondrial substrate utilization by measuring oxygen consumption of saponin permeabilized cardiac fibers, mitochondrial biogenesis by reverse transcription polymerase chain reaction and energy metabolism, and energy transfer enzymes by spectrophotometry. The results show that maximal oxidative capacity of the myocardium was decreased from 24.9 +/- 0.9 in control (CT) to 19.3 +/- 0.7 micromol O(2) min(-1) g dry weight(-1) in HYPO. However, protein content and messenger RNA (mRNA) of PGC-1alpha and mRNA of its transcription cascade that is thought to control mitochondrial content in normal myocardium and heart failure, were unchanged in HYPO. Mitochondrial utilization of glycerol-3P (-70%), malate (-45%), and octanoate (-24%) but not pyruvate was decreased in HYPO. Moreover, the creatine kinase system and energy transfer were hardly affected in HYPO. Besides, hypothyroidism decreased the activation of other signaling pathways like p38 mitogen-activated protein kinases, AMP-activated protein kinase, and calcineurin. These results show that cellular hypothyroidism can hardly account for the specific energetic alterations of heart failure.
Collapse
Affiliation(s)
- Yoni Athéa
- Signalisation et Physiopathologie Cardiaque
INSERMUniversité Paris-Sud - Paris 11IFR141Faculté de Pharmacie 5 Rue Jean-Baptiste Clément 92296 Chatenay Malabry Cedex
| | - Anne Garnier
- Signalisation et Physiopathologie Cardiaque
INSERMUniversité Paris-Sud - Paris 11IFR141Faculté de Pharmacie 5 Rue Jean-Baptiste Clément 92296 Chatenay Malabry Cedex
| | - Dominique Fortin
- Signalisation et Physiopathologie Cardiaque
INSERMUniversité Paris-Sud - Paris 11IFR141Faculté de Pharmacie 5 Rue Jean-Baptiste Clément 92296 Chatenay Malabry Cedex
| | - Lahoucine Bahi
- Signalisation et Physiopathologie Cardiaque
INSERMUniversité Paris-Sud - Paris 11IFR141Faculté de Pharmacie 5 Rue Jean-Baptiste Clément 92296 Chatenay Malabry Cedex
| | - Vladimir Veksler
- Signalisation et Physiopathologie Cardiaque
INSERMUniversité Paris-Sud - Paris 11IFR141Faculté de Pharmacie 5 Rue Jean-Baptiste Clément 92296 Chatenay Malabry Cedex
| | - Renée Ventura-Clapier
- Signalisation et Physiopathologie Cardiaque
INSERMUniversité Paris-Sud - Paris 11IFR141Faculté de Pharmacie 5 Rue Jean-Baptiste Clément 92296 Chatenay Malabry Cedex
- * Correspondence should be addressed to Renée Ventura-Clapier
| |
Collapse
|
39
|
Abstract
Myocardial ischemia and reperfusion injury have been extensively investigated in the laboratory mainly in healthy tissues. However, in clinical settings, ischemic heart disease coexists with certain illnesses, which could potentially influence the response of the myocardium to ischemia and reperfusion. Recent research has revealed that the abnormal heart may not be always vulnerable to ischemic injury. Furthermore, the effect of powerful means of protection, such as ischemic preconditioning, may not be in operation under certain pathological conditions. With this evidence in mind, the present review will focus on the response of the abnormal heart to ischemia and reperfusion, the possible underlying mechanisms, and potential cardioprotective strategies.
Collapse
Affiliation(s)
- Constantinos Pantos
- Department of Pharmacology, University of Athens, 75 Mikras Asias Avenue, 11527 Goudi, Athens, Greece.
| | | | | |
Collapse
|
40
|
Finck BN, Kelly DP. Peroxisome proliferator-activated receptor gamma coactivator-1 (PGC-1) regulatory cascade in cardiac physiology and disease. Circulation 2007; 115:2540-8. [PMID: 17502589 DOI: 10.1161/circulationaha.107.670588] [Citation(s) in RCA: 207] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Brian N Finck
- Center for Human Nutrition, Washington University School of Medicine, St. Louis, MO 63110, USA
| | | |
Collapse
|
41
|
Katz E, Zhang Q, Weiss HR, Scholz PM. T4-induced cardiac hypertrophy disrupts cyclic GMP mediated responses to brain natriuretic peptide in rabbit myocardium. Peptides 2006; 27:2276-83. [PMID: 16762459 DOI: 10.1016/j.peptides.2006.04.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2006] [Revised: 04/12/2006] [Accepted: 04/13/2006] [Indexed: 10/24/2022]
Abstract
Brain natriuretic peptide (BNP) affects the regulation of myocardial metabolism through the production of cGMP and these effects may be altered by cardiac hypertrophy. We tested the hypothesis that BNP would cause decreased metabolism and function in the heart and cardiac myocytes by increasing cGMP and that these effects would be disrupted after thyroxine-induced cardiac hypertrophy (T4). Open-chest control and T4 rabbits were instrumented to determine local effects of epicardial BNP (10(-3) M). Function of isolated cardiac myocytes was examined with BNP (10(-8)-10(-7) M) with or without KT5823 (10(-6) M, cGMP protein kinase inhibitor). Cyclic GMP levels were measured in myocytes. In open-chest controls, O2 consumption was reduced in the BNP area of the subepicardium (6.6+/-1.3 ml O2/min/100 g versus 8.9+/-1.4 ml O2/min/100 g) and subendocardium (9.4+/-1.3 versus 11.3+/-0.99). In T4 animals, functional and metabolic rates were higher than controls, but there was no difference between BNP-treated and untreated areas. In isolated control myocytes, BNP (10(-7) M) reduced percent shortening (PSH) from 6.5+/-0.6 to 4.3+/-0.4%. With KT5823 there was no effect of BNP on PSH. In T4 myocytes, BNP had no effect on PSH. In control myocytes, BNP caused cGMP levels to rise from 279+/-8 to 584+/-14 fmol/10(5) cells. In T4 myocytes, baseline cGMP levels were lower (117+/-2 l) and were not significantly increased by BNP. Thus, BNP caused decreased metabolism and function while increasing cGMP in control. These effects were lost after T4 due to lack of cGMP production. These data indicated that the effects of BNP on heart function operated through a cGMP-dependent mechanism, and that this mechanism was disrupted in T4-induced cardiac hypertrophy.
Collapse
Affiliation(s)
- Elizabeth Katz
- Heart and Brain Circulation Laboratory, Department of Physiology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ 08854-5635, USA
| | | | | | | |
Collapse
|
42
|
Morrish F, Buroker NE, Ge M, Ning XH, Lopez-Guisa J, Hockenbery D, Portman MA. Thyroid hormone receptor isoforms localize to cardiac mitochondrial matrix with potential for binding to receptor elements on mtDNA. Mitochondrion 2006; 6:143-8. [PMID: 16730242 DOI: 10.1016/j.mito.2006.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 04/13/2006] [Accepted: 04/21/2006] [Indexed: 11/29/2022]
Abstract
Thyroid hormone (T(3)) rapidly promotes both nuclear and mitochondrial DNA transcription in cardiomyocytes, suggesting that T3 directly binds and activates mitochondrial genes. We showed for the first time mitochondrial localization for multiple TRalpha isoforms in heart, including truncated versions. Additionally, we demonstrated novel mitochondrial localization for versions of TRalpha(2), the dominant negative isoform lacking a functional ligand-binding domain. We also confirmed by electromobility shift assays, that TRalpha(2) in mitochondrial extracts binds to thyroid receptor response elements present in the 12S rRNA (DRO) and D-loop region (DR2) of mitochondrial DNA. Thus, TRalpha isoforms may directly regulate T(3) responses at mtDNA in the heart.
Collapse
Affiliation(s)
- Fionnuala Morrish
- Children's Hospital and Regional Medical Center, Seattle, WA 98105, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
McClure TD, Young ME, Taegtmeyer H, Ning XH, Buroker NE, López-Guisa J, Portman MA. Thyroid hormone interacts with PPARalpha and PGC-1 during mitochondrial maturation in sheep heart. Am J Physiol Heart Circ Physiol 2005; 289:H2258-64. [PMID: 16024569 DOI: 10.1152/ajpheart.00473.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Thyroid hormone (TH) promotes cardiac mitochondrial maturation and substrate metabolism after birth. This regulation involves ligand-dependent binding of nuclear TH receptors to target gene elements. TH also putatively controls genes indirectly by modulating transcription and/or translation of other nuclear steroid receptors and coactivators, such as peroxisome proliferator-activated receptor-alpha (PPARalpha) and peroxisome proliferator-activated receptor-gamma coactivator-1 (PGC-1). We tested the hypothesis that TH influences PPARalpha and PGC-1 regulation of metabolic genes during postnatal maturation in sheep heart in vivo. We measured their mRNAs and/or protein levels and downstream targets in left ventricle from lambs: fetal (F), 30-day-old after postnatal thyroidectomy (THY), and 30-day-old euthyroid (Con). Both PPARalpha and PGC-1 mRNA expression decreased from F to Con, while PGC-1 protein increased substantially and PPARalpha did not change. THY limited this mRNA response and attenuated the paradoxical postnatal PGC-1 protein elevation but did not alter mRNA levels for PPARalpha, nuclear respiratory factor-1 and hypoxia-inducible factor-1alpha. THY promotion in PPARalpha mRNA did not change PPARalpha protein or mRNA for PPARalpha target genes, pyruvate-dehydrogenase kinase 4 (PDK4) and muscle type carnitine palmitoyltransferase I (mCPTI). THY reduction in PGC-1 protein occurred, while reducing cytochrome c oxidase and cytochrome c content and decreasing cardiac maximal inherent respiratory capacity. These data imply that TH modulates mitochondrial maturation partly through posttranscriptional control of PGC-1, while any important regulation of PDK4 and mCPTI by change in PPARalpha protein expression remains doubtful. Also, the paradoxical expression pattern between mRNA and protein, particularly for PGC-1, suggests a feedback control mechanism.
Collapse
Affiliation(s)
- Timothy D McClure
- University of Washington, School of Medicine, and Children's Hospital and Regional Medical Center W4841, 4800 Sand Point Way NE, Seattle, WA 98105, USA
| | | | | | | | | | | | | |
Collapse
|