Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters

Publication year range
1.
Physiol Genomics ; 54(1): 22-35, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34766515

ABSTRACT

Broad cellular functions and diseases including muscular dystrophy, arrhythmogenic right ventricular cardiomyopathy (ARVC5) and cancer are associated with transmembrane protein43 (TMEM43/LUMA). The study aimed to investigate biological roles of TMEM43 through genetic regulation, gene pathways and gene networks, candidate interacting genes, and up- or downstream regulators. Cardiac transcriptomes from 40 strains of recombinant inbred BXD mice and two parental strains representing murine genetic reference population (GRP) were applied for genetic correlation, functional enrichment, and coexpression network analysis using systems genetics approach. The results were validated in a newly created knock-in Tmem43-S358L mutation mouse model (Tmem43S358L) that displayed signs of cardiac dysfunction, resembling ARVC5 phenotype seen in humans. We found high Tmem43 levels among BXDs with broad variability in expression. Expression of Tmem43 highly negatively correlated with heart mass and heart rate among BXDs, whereas levels of Tmem43 highly positively correlated with plasma high-density lipoproteins (HDL). Through finding differentially expressed genes (DEGs) between Tmem43S358L mutant and wild-type (Tmem43WT) lines, 18 pathways (out of 42 found in BXDs GRP) that are involved in ARVC, hypertrophic cardiomyopathy, dilated cardiomyopathy, nonalcoholic fatty liver disease, Alzheimer's disease, Parkinson's disease, and Huntington's disease were verified. We further constructed Tmem43-mediated gene network, in which Ctnna1, Adcy6, Gnas, Ndufs6, and Uqcrc2 were significantly altered in Tmem43S358L mice versus Tmem43WT controls. Our study defined the importance of Tmem43 for cardiac- and metabolism-related pathways, suggesting that cardiovascular disease-relevant risk factors may also increase risk of metabolic and neurodegenerative diseases via TMEM43-mediated pathways.


Subject(s)
Arrhythmogenic Right Ventricular Dysplasia , Membrane Proteins , Animals , Arrhythmogenic Right Ventricular Dysplasia/diagnosis , Arrhythmogenic Right Ventricular Dysplasia/genetics , Heart , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mutation/genetics , Phenotype
2.
Am J Physiol Heart Circ Physiol ; 320(5): H2130-H2146, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33861145

ABSTRACT

The actin-binding sarcomeric nebulette (NEBL) protein provides efficient contractile flexibility via interaction with desmin intermediate filaments. NEBL gene mutations affecting the nebulin repeat (NR) domain are known to induce cardiomyopathy. The study aimed to explore the roles of NEBL in exercise and biomechanical stress response. We ablated exon3 encoding the first NR of Nebl and created global Neblex3-/ex3- knockout mice. Cardiac function, structure, and transcriptome were assessed before and after a 4-wk treadmill regimen. A Nebl-based exercise signaling network was constructed using systems genetics methods. H9C2 and neonatal rat cardiomyocytes (NRCs) expressing wild-type or mutant NEBL underwent cyclic mechanical strain. Neblex3-/ex3- mice demonstrated diastolic dysfunction with preserved systolic function at 6 mo of age. After treadmill running, 4-mo-old Neblex3-/ex3- mice developed concentric cardiac hypertrophy and left ventricular dilation compared with running Nebl+/+ and sedentary Neblex3-/ex3- mice. Disturbance of sarcomeric Z-disks and thin filaments architecture and disruption of intercalated disks and mitochondria were found in exercised Neblex3-/ex3- mice. A Nebl-based exercise signaling network included Csrp3, Des, Fbox32, Jup, Myh6, and Myh7. Disturbed expression of TM1, DES, JUP, ß-catenin, MLP, α-actinin2, and vinculin proteins was demonstrated. In H9C2 cells, NEBL was recruited into focal adhesions at 24-h poststrain and redistributed along with F-actin at 72-h poststrain, suggesting time-dependent redistribution of NEBL in response to strain. NEBL mutations cause desmin disorganization in NRCs upon stretch. We conclude that Nebl's NR ablation causes disturbed sarcomere, Z-disks, and desmin organization, and prevents NEBL redistribution to focal adhesions in cardiomyocytes, weakening cardiac tolerance to biomechanical stress.NEW & NOTEWORTHY We demonstrate that ablation of first nebulin-repeats of sarcomeric nebulette (Nebl) causes diastolic dysfunction in Neblex3-/ex3- mice. Exercise-induced development of diastolic dysfunction, cardiac hypertrophy and ventricular dilation in knockouts. This was associated with sarcomere disturbance, intercalated disks disruption, and mitochondrial distortion upon stress and altered expression of genes involved in Nebl-based stress network. We demonstrate that G202R and A592 mutations alter actin and desmin expression causing disorganization of desmin filaments upon cyclic strain.


Subject(s)
Cytoskeletal Proteins/metabolism , Exercise Tolerance/physiology , LIM Domain Proteins/metabolism , Myocytes, Cardiac/metabolism , Physical Conditioning, Animal/physiology , Sarcomeres/metabolism , Actin Cytoskeleton/metabolism , Animals , Cardiomegaly/metabolism , Cytoskeletal Proteins/genetics , LIM Domain Proteins/genetics , Mice , Mice, Knockout , Myocardium/metabolism , Rats , Stress, Mechanical
3.
Hum Mol Genet ; 26(19): 3776-3791, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28934388

ABSTRACT

Recently, we identified biallelic mutations of SLC25A46 in patients with multiple neuropathies. Functional studies revealed that SLC25A46 may play an important role in mitochondrial dynamics by mediating mitochondrial fission. However, the cellular basis and pathogenic mechanism of the SLC25A46-related neuropathies are not fully understood. Thus, we generated a Slc25a46 knock-out mouse model. Mice lacking SLC25A46 displayed severe ataxia, mainly caused by degeneration of Purkinje cells. Increased numbers of small, unmyelinated and degenerated optic nerves as well as loss of retinal ganglion cells indicated optic atrophy. Compound muscle action potentials in peripheral nerves showed peripheral neuropathy associated with degeneration and demyelination in axons. Mutant cerebellar neurons have large mitochondria, which exhibit abnormal distribution and transport. Biochemically mutant mice showed impaired electron transport chain activity and accumulated autophagy markers. Our results suggest that loss of SLC25A46 causes degeneration in neurons by affecting mitochondrial dynamics and energy production.


Subject(s)
Mitochondria/genetics , Mitochondria/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Phosphate Transport Proteins/genetics , Phosphate Transport Proteins/metabolism , Animals , Ataxia/pathology , Female , Humans , Male , Mice , Mice, Knockout , Mitochondrial Dynamics/physiology , Mutation , Retinal Ganglion Cells/pathology
4.
Cell Mol Life Sci ; 74(15): 2795-2813, 2017 08.
Article in English | MEDLINE | ID: mdl-28378042

ABSTRACT

Growing number of studies provide strong evidence that the mitochondrial permeability transition pore (PTP), a non-selective channel in the inner mitochondrial membrane, is involved in the pathogenesis of cardiac ischemia-reperfusion and can be targeted to attenuate reperfusion-induced damage to the myocardium. The molecular identity of the PTP remains unknown and cyclophilin D is the only protein commonly accepted as a major regulator of the PTP opening. Therefore, cyclophilin D is an attractive target for pharmacological or genetic therapies to reduce ischemia-reperfusion injury in various animal models and humans. Most animal studies demonstrated cardioprotective effects of PTP inhibition; however, a recent large clinical trial conducted by international groups demonstrated that cyclosporine A, a cyclophilin D inhibitor, failed to protect the heart in patients with myocardial infarction. These studies, among others, raise the question of whether cyclophilin D, which plays an important physiological role in the regulation of cell metabolism and mitochondrial bioenergetics, is a viable target for cardioprotection. This review discusses previous studies to provide comprehensive information on the physiological role of cyclophilin D as well as PTP opening in the cell that can be taken into consideration for the development of new PTP inhibitors.


Subject(s)
Cardiotonic Agents/pharmacology , Cyclophilins/metabolism , Drug Discovery , Mitochondrial Membrane Transport Proteins/metabolism , Myocardial Ischemia/drug therapy , Myocardial Ischemia/metabolism , Animals , Calcium/metabolism , Peptidyl-Prolyl Isomerase F , Cyclophilins/antagonists & inhibitors , Energy Metabolism/drug effects , Heart/drug effects , Humans , Mitochondrial Membrane Transport Proteins/antagonists & inhibitors , Mitochondrial Permeability Transition Pore , Molecular Targeted Therapy , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Oxidative Stress/drug effects , Protein Interaction Maps/drug effects , Protein Processing, Post-Translational/drug effects
5.
Int J Mol Sci ; 19(11)2018 Nov 04.
Article in English | MEDLINE | ID: mdl-30400386

ABSTRACT

Peroxisome proliferator-activated receptors (PPARs) are nuclear hormone receptors that bind to DNA and regulate transcription of genes involved in lipid and glucose metabolism. A growing number of studies provide strong evidence that PPARs are the promising pharmacological targets for therapeutic intervention in various diseases including cardiovascular disorders caused by compromised energy metabolism. PPAR agonists have been widely used for decades as lipid-lowering and anti-inflammatory drugs. Existing studies are mainly focused on the anti-atherosclerotic effects of PPAR agonists; however, their role in the maintenance of cellular bioenergetics remains unclear. Recent studies on animal models and patients suggest that PPAR agonists can normalize lipid metabolism by stimulating fatty acid oxidation. These studies indicate the importance of elucidation of PPAR agonists as potential pharmacological agents for protection of the heart from energy deprivation. Here, we summarize and provide a comprehensive analysis of previous studies on the role of PPARs in the heart under normal and pathological conditions. In addition, the review discusses the PPARs as a therapeutic target and the beneficial effects of PPAR agonists, particularly bezafibrate, to attenuate cardiomyopathy and heart failure in patients and animal models.


Subject(s)
Atherosclerosis/drug therapy , Cardiomyopathies/drug therapy , Cardiotonic Agents/therapeutic use , Energy Metabolism/drug effects , Heart Failure/prevention & control , Peroxisome Proliferator-Activated Receptors/agonists , Animals , Anti-Inflammatory Agents/therapeutic use , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bezafibrate/therapeutic use , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Energy Metabolism/genetics , Fatty Acids/metabolism , Gene Expression Regulation , Heart Failure/genetics , Heart Failure/metabolism , Heart Failure/pathology , Humans , Hypolipidemic Agents/therapeutic use , Lipid Metabolism/drug effects , Lipid Metabolism/genetics , Oxidation-Reduction , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferator-Activated Receptors/metabolism , Signal Transduction
6.
Amino Acids ; 48(8): 2057-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27401086

ABSTRACT

Creatine (Cr) is a guanidino compound required for rapid replenishment of ATP in cells with a high-energy demand. In humans, mutations in the Cr transporter (CRT;SLC6A8) prevent Cr entry into tissue and result in a significant intellectual impairment, epilepsy, and aphasia. The lack of Cr on both the whole body and cellular metabolism was evaluated in Crt knockout (Crt (-/y) ) mice, a high-fidelity model of human CRT deficiency. Crt (-/y) mice have reduced body mass and, however, show a twofold increase in body fat. There was increased energy expenditure in a home cage environment and during treadmill running in Crt (-/y) mice. Consistent with the increases in the whole-body metabolic function, Crt (-/y) mice show increased cellular metabolism as well. Mitochondrial respiration increased in skeletal muscle fibers and hippocampal lysates from Crt (-/y) mice. In addition, Crt (-/y) mice had increased citrate synthase activity, suggesting a higher number of mitochondria instead of an increase in mitochondrial activity. To determine if the increase in respiration was due to increased mitochondrial numbers, we measured oxygen consumption in an equal number of mitochondria from Crt (+/y) and Crt (-/y) mice. There were no changes in mitochondrial respiration when normalized to mitochondrial number, suggesting that the increase in respiration observed could be to higher mitochondrial content in Crt (-/y) mice.


Subject(s)
Adiposity , Hippocampus/metabolism , Membrane Transport Proteins/metabolism , Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Animals , Citrate (si)-Synthase/genetics , Citrate (si)-Synthase/metabolism , Female , Humans , Male , Membrane Transport Proteins/genetics , Mice , Mice, Mutant Strains , Mitochondria, Muscle/genetics , Oxygen Consumption/genetics
7.
J Cell Mol Med ; 18(4): 709-20, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24444314

ABSTRACT

AMP-kinase (AMPK) activation reduces cardiac hypertrophy, although underlying molecular mechanisms remain unclear. In this study, we elucidated the anti-hypertrophic action of metformin, specifically, the role of the AMPK/eNOS/p53 pathway. H9c2 rat cardiomyocytes were treated with angiotensin II (AngII) for 24 hrs in the presence or absence of metformin (AMPK agonist), losartan [AngII type 1 receptor (AT1R) blocker], Nω-nitro-L-arginine methyl ester (L-NAME, pan-NOS inhibitor), splitomicin (SIRT1 inhibitor) or pifithrin-α (p53 inhibitor). Results showed that treatment with metformin significantly attenuated AngII-induced cell hypertrophy and death. Metformin attenuated AngII-induced activation (cleavage) of caspase 3, Bcl-2 down-regulation and p53 up-regulation. It also reduced AngII-induced AT1R up-regulation by 30% (P < 0.05) and enhanced AMPK phosphorylation by 99% (P < 0.01) and P-eNOS levels by 3.3-fold (P < 0.01). Likewise, losartan reduced AT1R up-regulation and enhanced AMPK phosphorylation by 54% (P < 0.05). The AMPK inhibitor, compound C, prevented AT1R down-regulation, indicating that metformin mediated its effects via AMPK activation. Beneficial effects of metformin and losartan converged on mitochondria that demonstrated high membrane potential (Δψm ) and low permeability transition pore opening. Thus, this study demonstrates that the anti-hypertrophic effects of metformin are associated with AMPK-induced AT1R down-regulation and prevention of mitochondrial dysfunction through the SIRT1/eNOS/p53 pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Angiotensin II/administration & dosage , Cardiomegaly/metabolism , Mitochondria/metabolism , Myocytes, Cardiac/metabolism , AMP-Activated Protein Kinases/biosynthesis , Angiotensin II Type 1 Receptor Blockers/administration & dosage , Animals , Cardiomegaly/drug therapy , Cardiomegaly/pathology , Gene Expression Regulation/drug effects , Losartan/administration & dosage , Metformin/administration & dosage , Mitochondria/drug effects , Mitochondria/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Nitric Oxide Synthase Type III/metabolism , Rats , Receptor, Angiotensin, Type 1/biosynthesis , Signal Transduction , Sirtuin 1/metabolism , Tumor Suppressor Protein p53/metabolism
8.
Am J Physiol Heart Circ Physiol ; 306(3): H326-38, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24285112

ABSTRACT

The very long-chain acyl-CoA dehydrogenase (VLCAD) enzyme catalyzes the first step of mitochondrial ß-oxidation. Patients with VLCAD deficiency present with hypoketotic hypoglycemia and cardiomyopathy, which can be exacerbated by fasting and/or cold stress. Global VLCAD knockout mice recapitulate these phenotypes: mice develop cardiomyopathy, and cold exposure leads to rapid hypothermia and death. However, the contribution of different tissues to development of these phenotypes has not been studied. We generated cardiac-specific VLCAD-deficient (cVLCAD(-/-)) mice by Cre-mediated ablation of the VLCAD in cardiomyocytes. By 6 mo of age, cVLCAD(-/-) mice demonstrated increased end-diastolic and end-systolic left ventricular dimensions and decreased fractional shortening. Surprisingly, selective VLCAD gene ablation in cardiomyocytes was sufficient to evoke severe cold intolerance in mice who rapidly developed severe hypothermia, bradycardia, and markedly depressed cardiac function in response to fasting and cold exposure (+5°C). We conclude that cardiac-specific VLCAD deficiency is sufficient to induce cold intolerance and cardiomyopathy and is associated with reduced ATP production. These results provide strong evidence that fatty acid oxidation in myocardium is essential for maintaining normal cardiac function under these stress conditions.


Subject(s)
Acyl-CoA Dehydrogenase, Long-Chain/deficiency , Cardiomyopathy, Dilated/enzymology , Hypothermia/enzymology , Adenosine Triphosphate/metabolism , Animals , Cardiomyopathy, Dilated/etiology , Cardiomyopathy, Dilated/metabolism , Cold Temperature , Congenital Bone Marrow Failure Syndromes , Disease Models, Animal , Hypothermia/etiology , Hypothermia/metabolism , Lipid Metabolism, Inborn Errors , Mice , Mice, Inbred C57BL , Mice, Knockout , Mitochondrial Diseases , Muscular Diseases , Oxidation-Reduction , Stress, Physiological
9.
PLoS One ; 19(1): e0297166, 2024.
Article in English | MEDLINE | ID: mdl-38285689

ABSTRACT

Src is a non-receptor tyrosine kinase participating in a range of neuronal processes, including synaptic plasticity. We have recently shown that the amounts of total Src and its two phosphorylated forms, at tyrosine-416 (activated) and tyrosine-527 (inhibited), undergoes time-dependent, region-specific learning-related changes in the domestic chick forebrain after visual imprinting. These changes occur in the intermediate medial mesopallium (IMM), a site of memory formation for visual imprinting, but not the posterior pole of the nidopallium (PPN), a control brain region not involved in imprinting. Src interacts with mitochondrial genome-coded NADH dehydrogenase subunit 2 (NADH2), a component of mitochondrial respiratory complex I. This interaction occurs at brain excitatory synapses bearing NMDA glutamate receptors. The involvement of Src-NADH2 complexes in learning and memory is not yet explored. We show for the first time that, independently of changes in total Src or total NADH2, NADH2 bound to Src immunoprecipitated from the P2 plasma membrane-mitochondrial fraction: (i) is increased in a learning-related manner in the left IMM 1 h after the end of training; (ii), is decreased in the right IMM in a learning-related way 24 h after training. These changes occurred in the IMM but not the PPN. They are attributable to learning occurring during training rather than a predisposition to learn. Learning-related changes in Src-bound NADH2 are thus time- and region-dependent.


Subject(s)
Imprinting, Psychological , NADH Dehydrogenase , src-Family Kinases , Animals , Chickens , Imprinting, Psychological/physiology , Learning/physiology , Prosencephalon/physiology , Tyrosine , src-Family Kinases/metabolism
10.
Br J Haematol ; 161(3): 330-8, 2013 May.
Article in English | MEDLINE | ID: mdl-23432031

ABSTRACT

Barth syndrome (BTHS) is an X-linked autosomal recessive disorder characterized by neutropenia, cardiomyopathy and growth retardation. BTHS was first described as mitochondrial disease affecting neutrophils as well as cardiac and skeletal muscles. Patients with neutropenia may have extremely low levels of circulating neutrophils and suffer from recurring sometimes life-threatening bacterial infections. Sepsis is not infrequent, may occur unexpectedly in a patient with no history for pronounced bacterial infections and may lead to death. The reduced level of circulating neutrophils suggests either a reduced production of myeloid cells in the bone marrow and premature apoptosis or aberrant clearance of neutrophils in peripheral blood. The underlying molecular defects are truncation, deletion or substitution mutations in the TAZ gene that appear to result in loss-of-function of the gene product tafazzin. Molecular mechanisms triggering neutropenia and cardiomyopathy in BTHS remain largely unclear. The current review focusses on recent advances in the understanding of molecular and cellular bases of neutropenia in Barth syndrome and covers the functional implications of the TAZ mutations, experimental models for neutropenia, the specific cellular abnormalities triggered by loss of TAZ function and potential novel therapeutic strategies for restoring the normal phenotype.


Subject(s)
Barth Syndrome , Acylation , Acyltransferases , Animals , Bacterial Infections/etiology , Barth Syndrome/genetics , Barth Syndrome/immunology , Barth Syndrome/pathology , Barth Syndrome/therapy , Bone Marrow/pathology , Cardiolipins/metabolism , Chromosomes, Human, X/genetics , Disease Models, Animal , Drosophila Proteins/deficiency , Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Fetal Heart/pathology , Gene Knockdown Techniques , Growth Disorders/genetics , Humans , Immunocompromised Host , Male , Mice , Mice, Transgenic , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mutation , Neutropenia/genetics , Neutropenia/physiopathology , RNA Interference , Sequence Deletion , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription Factors/physiology , Zebrafish/genetics , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
11.
J Biol Chem ; 286(2): 899-908, 2011 Jan 14.
Article in English | MEDLINE | ID: mdl-21068380

ABSTRACT

Barth syndrome is an X-linked genetic disorder caused by mutations in the tafazzin (taz) gene and characterized by dilated cardiomyopathy, exercise intolerance, chronic fatigue, delayed growth, and neutropenia. Tafazzin is a mitochondrial transacylase required for cardiolipin remodeling. Although tafazzin function has been studied in non-mammalian model organisms, mammalian genetic loss of function approaches have not been used. We examined the consequences of tafazzin knockdown on sarcomeric mitochondria and cardiac function in mice. Tafazzin knockdown resulted in a dramatic decrease of tetralinoleoyl cardiolipin in cardiac and skeletal muscles and accumulation of monolysocardiolipins and cardiolipin molecular species with aberrant acyl groups. Electron microscopy revealed pathological changes in mitochondria, myofibrils, and mitochondrion-associated membranes in skeletal and cardiac muscles. Echocardiography and magnetic resonance imaging revealed severe cardiac abnormalities, including left ventricular dilation, left ventricular mass reduction, and depression of fractional shortening and ejection fraction in tafazzin-deficient mice. Tafazzin knockdown mice provide the first mammalian model system for Barth syndrome in which the pathophysiological relationships between altered content of mitochondrial phospholipids, ultrastructural abnormalities, myocardial and mitochondrial dysfunction, and clinical outcome can be completely investigated.


Subject(s)
Barth Syndrome , Cardiomyopathy, Dilated , Muscle, Skeletal/metabolism , Myocardium/metabolism , Transcription Factors/genetics , Acyltransferases , Animals , Barth Syndrome/genetics , Barth Syndrome/pathology , Barth Syndrome/physiopathology , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/cytology , Female , Humans , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron , Mitochondria/metabolism , Mitochondria/pathology , Mitochondria/ultrastructure , Muscle, Skeletal/pathology , Muscle, Skeletal/ultrastructure , Myocardium/pathology , Myocardium/ultrastructure , Phospholipids/metabolism , RNA, Small Interfering
12.
J Mol Med (Berl) ; 99(1): 57-73, 2021 01.
Article in English | MEDLINE | ID: mdl-33201259

ABSTRACT

Mitochondria are recognized as the main source of ATP to meet the energy demands of the cell. ATP production occurs by oxidative phosphorylation when electrons are transported through the electron transport chain (ETC) complexes and develop the proton motive force across the inner mitochondrial membrane that is used for ATP synthesis. Studies since the 1960s have been concentrated on the two models of structural organization of ETC complexes known as "solid-state" and "fluid-state" models. However, advanced new techniques such as blue-native gel electrophoresis, mass spectroscopy, and cryogenic electron microscopy for analysis of macromolecular protein complexes provided new data in favor of the solid-state model. According to this model, individual ETC complexes are assembled into macromolecular structures known as respiratory supercomplexes (SCs). A large number of studies over the last 20 years proposed the potential role of SCs to facilitate substrate channeling, maintain the integrity of individual ETC complexes, reduce electron leakage and production of reactive oxygen species, and prevent excessive and random aggregation of proteins in the inner mitochondrial membrane. However, many other studies have challenged the proposed functional role of SCs. Recently, a third model known as the "plasticity" model was proposed that partly reconciles both "solid-state" and "fluid-state" models. According to the "plasticity" model, respiratory SCs can co-exist with the individual ETC complexes. To date, the physiological role of SCs remains unknown, although several studies using tissue samples of patients or animal/cell models of human diseases revealed an associative link between functional changes and the disintegration of SC assembly. This review summarizes and discusses previous studies on the mechanisms and regulation of SC assembly under physiological and pathological conditions.


Subject(s)
Electron Transport Chain Complex Proteins/metabolism , Mitochondria/metabolism , Animals , Cell Respiration , Humans
13.
Mitochondrion ; 50: 71-81, 2020 01.
Article in English | MEDLINE | ID: mdl-31669621

ABSTRACT

Mitochondria have been widely accepted as the main source of ATP in the cell. The inner mitochondrial membrane (IMM) is important for the maintenance of ATP production and other functions of mitochondria. The electron transport chain (ETC) generates an electrochemical gradient of protons known as the proton-motive force across the IMM and thus produces the mitochondrial membrane potential that is critical to ATP synthesis. One of the main factors regulating the structural and functional integrity of the IMM is the changes in the matrix volume. Mild (reversible) swelling regulates mitochondrial metabolism and function; however, excessive (irreversible) swelling causes mitochondrial dysfunction and cell death. The central mechanism of mitochondrial swelling includes the opening of non-selective channels known as permeability transition pores (PTPs) in the IMM by high mitochondrial Ca2+ and reactive oxygen species (ROS). The mechanisms of reversible and irreversible mitochondrial swelling and transition between these two states are still unknown. The present study elucidates an upgraded biophysical model of reversible and irreversible mitochondrial swelling dynamics. The model provides a description of the PTP regulation dynamics using an additional differential equation. The rigidity tensor was used in numerical simulations of the mitochondrial parameter dynamics with different initial conditions defined by Ca2+ concentration in the sarco/endoplasmic reticulum. We were able to estimate the values of the IMM rigidity tensor components by fitting the model to the previously reported experimental data. Overall, the model provides a better description of the reversible and irreversible mitochondrial swelling dynamics.


Subject(s)
Cell Death/physiology , Computer Simulation , Mitochondria/physiology , Mitochondrial Membranes/physiology , Mitochondrial Swelling/physiology , Animals , Biophysical Phenomena , Membrane Potential, Mitochondrial , Models, Biological
14.
Cells ; 9(2)2020 02 22.
Article in English | MEDLINE | ID: mdl-32098394

ABSTRACT

The main purpose of the review article is to assess the contributions of telomere length and telomerase activity to the cardiac function at different stages of development and clarify their role in cardiac disorders. It has been shown that the telomerase complex and telomeres are of great importance in many periods of ontogenesis due to the regulation of the proliferative capacity of heart cells. The review article also discusses the problems of heart regeneration and the identification of possible causes of dysfunction of telomeres and telomerase.


Subject(s)
Aging/metabolism , Myocytes, Cardiac/metabolism , Organogenesis/physiology , Regeneration/physiology , Telomerase/metabolism , Telomere Homeostasis/physiology , Telomere/metabolism , Adult , Animals , Animals, Newborn , Cell Proliferation/physiology , Humans
15.
J Am Soc Nephrol ; 19(10): 1955-64, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18701606

ABSTRACT

A screen of recessive mutations generated by the chemical mutagen n-ethyl-n-nitrosourea (ENU) mapped a new mutant locus (5772SB) termed sudden juvenile death syndrome (sjds) to chromosome 7 in mice. These mutant mice, which exhibit severe proximal tubule injury and formation of giant vacuoles in the renal cortex, die from renal failure, a phenotype that resembles aquaporin 11 (Aqp11) knockout mice. In this report, the ENU-induced single-nucleotide variant (sjds mutation) is identified. To determine whether this variant, which causes an amino acid substitution (Cys227Ser) in the predicted E-loop region of aquaporin 11, is responsible for the sjds lethal renal phenotype, Aqp11-/sjds compound heterozygous mice were generated from Aqp11 +/sjds and Aqp11 +/- intercrosses. The compound heterozygous Aqp11 -/sjds offspring exhibited a lethal renal phenotype (renal failure by 2 wk), similar to the Aqp11 sjds/sjds and Aqp11-/- phenotypes. These results demonstrate that the identified mutation causes renal failure in Aqp11 sjds/sjds mutant mice, providing a model for better understanding of the structure and function of aquaporin 11 in renal physiology.


Subject(s)
Amino Acid Substitution/genetics , Aquaporins/genetics , Ethylnitrosourea , Point Mutation/genetics , Renal Insufficiency/genetics , Animals , Disease Models, Animal , Female , Male , Mice , Mice, Mutant Strains , Polymorphism, Single Nucleotide/genetics , Syndrome
16.
Cell Stem Cell ; 24(4): 621-636.e16, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30930145

ABSTRACT

Tafazzin (TAZ) is a mitochondrial transacylase that remodels the mitochondrial cardiolipin into its mature form. Through a CRISPR screen, we identified TAZ as necessary for the growth and viability of acute myeloid leukemia (AML) cells. Genetic inhibition of TAZ reduced stemness and increased differentiation of AML cells both in vitro and in vivo. In contrast, knockdown of TAZ did not impair normal hematopoiesis under basal conditions. Mechanistically, inhibition of TAZ decreased levels of cardiolipin but also altered global levels of intracellular phospholipids, including phosphatidylserine, which controlled AML stemness and differentiation by modulating toll-like receptor (TLR) signaling.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Mitochondria/enzymology , Phospholipids/metabolism , Transcription Factors/metabolism , Acyltransferases , Animals , Cell Line, Tumor , Doxorubicin/pharmacology , Female , Humans , Leukemia, Myeloid, Acute/pathology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Signal Transduction/drug effects , Toll-Like Receptors/metabolism , Transcription Factors/antagonists & inhibitors , Transcription Factors/deficiency
17.
Circ Res ; 99(2): 201-8, 2006 Jul 21.
Article in English | MEDLINE | ID: mdl-16794186

ABSTRACT

Barth syndrome is an X-linked disorder characterized by cardiomyopathy, skeletal myopathy, neutropenia, organic aciduria, and growth retardation caused by mutations in tafazzin. The sequence similarity of tafazzin to acyltransferases suggests a role in mitochondrial phospholipid metabolism. To study the role of tafazzin in heart function and development, we created a knockdown zebrafish model. Zebrafish tafazzin mRNA is first evident at 7 hours post-fertilization (hpf). At 10 and 24 hpf, tafazzin mRNA is ubiquitous, with highest levels in the head. By 51 hpf, expression becomes cardiac restricted. The tafazzin knockdown created by antisense morpholino yolk injection resulted in dose-dependent lethality, severe developmental and growth retardation, marked bradycardia and pericardial effusions, and generalized edema, signs that resemble human Barth syndrome heart failure. This knockdown phenotype was rescued by concomitant injection of normal tafazzin mRNA. Abnormal cardiac development, with a linear, nonlooped heart, and hypomorphic tail and eye development proves that tafazzin is essential for overall zebrafish development, especially of the heart. The tafazzin knockdown zebrafish provides an animal model similar to Barth syndrome to analyze the severity of human mutants and to test potential treatments.


Subject(s)
Cardiomyopathy, Dilated/congenital , Cardiomyopathy, Dilated/etiology , Heart/growth & development , Transcription Factors/deficiency , Zebrafish Proteins/deficiency , Acyltransferases , Animals , Cardiomyopathy, Dilated/genetics , Child Development , Disease Models, Animal , Gene Expression Regulation, Developmental , Heart/physiology , Humans , Infant , RNA, Antisense/pharmacology , RNA, Messenger/antagonists & inhibitors , Syndrome , Transcription Factors/genetics , Transcription Factors/physiology , Zebrafish , Zebrafish Proteins/genetics , Zebrafish Proteins/physiology
18.
Front Pharmacol ; 9: 318, 2018.
Article in English | MEDLINE | ID: mdl-29695963

ABSTRACT

Aim: Tafazzin knockdown (TazKD) in mice is widely used to create an experimental model of Barth syndrome (BTHS) that exhibits dilated cardiomyopathy and impaired exercise capacity. Peroxisome proliferator-activated receptors (PPARs) are a group of nuclear receptor proteins that play essential roles as transcription factors in the regulation of carbohydrate, lipid, and protein metabolism. We hypothesized that the activation of PPAR signaling with PPAR agonist bezafibrate (BF) may ameliorate impaired cardiac and skeletal muscle function in TazKD mice. This study examined the effects of BF on cardiac function, exercise capacity, and metabolic status in the heart of TazKD mice. Additionally, we elucidated the impact of PPAR activation on molecular pathways in TazKD hearts. Methods: BF (0.05% w/w) was given to TazKD mice with rodent chow. Cardiac function in wild type-, TazKD-, and BF-treated TazKD mice was evaluated by echocardiography. Exercise capacity was evaluated by exercising mice on the treadmill until exhaustion. The impact of BF on metabolic pathways was evaluated by analyzing the total transcriptome of the heart by RNA sequencing. Results: The uptake of BF during a 4-month period at a clinically relevant dose effectively protected the cardiac left ventricular systolic function in TazKD mice. BF alone did not improve the exercise capacity however, in combination with everyday voluntary running on the running wheel BF significantly ameliorated the impaired exercise capacity in TazKD mice. Analysis of cardiac transcriptome revealed that BF upregulated PPAR downstream target genes involved in a wide spectrum of metabolic (energy and protein) pathways as well as chromatin modification and RNA processing. In addition, the Ostn gene, which encodes the metabolic hormone musclin, is highly induced in TazKD myocardium and human failing hearts, likely as a compensatory response to diminished bioenergetic homeostasis in cardiomyocytes. Conclusion: The PPAR agonist BF at a clinically relevant dose has the therapeutic potential to attenuate cardiac dysfunction, and possibly exercise intolerance in BTHS. The role of musclin in the failing heart should be further investigated.

19.
JCI Insight ; 3(22)2018 11 15.
Article in English | MEDLINE | ID: mdl-30429366

ABSTRACT

The mitochondrial Ca2+ uniporter (MCU) complex mediates acute mitochondrial Ca2+ influx. In skeletal muscle, MCU links Ca2+ signaling to energy production by directly enhancing the activity of key metabolic enzymes in the mitochondria. Here, we examined the role of MCU in skeletal muscle development and metabolic function by generating mouse models for the targeted deletion of Mcu in embryonic, postnatal, and adult skeletal muscle. Loss of Mcu did not affect muscle growth and maturation or otherwise cause pathology. Skeletal muscle-specific deletion of Mcu in mice also did not affect myofiber intracellular Ca2+ handling, but it did inhibit acute mitochondrial Ca2+ influx and mitochondrial respiration stimulated by Ca2+, resulting in reduced acute exercise performance in mice. However, loss of Mcu also resulted in enhanced muscle performance under conditions of fatigue, with a preferential shift toward fatty acid metabolism, resulting in reduced body fat with aging. Together, these results demonstrate that MCU-mediated mitochondrial Ca2+ regulation underlies skeletal muscle fuel selection at baseline and under enhanced physiological demands, which affects total homeostatic metabolism.


Subject(s)
Calcium Channels/metabolism , Calcium/metabolism , Muscle, Skeletal/metabolism , Animals , Calcium Channels/genetics , Calcium Signaling , Energy Metabolism , Female , Gene Targeting , Male , Mice , Mice, Transgenic , Muscle, Skeletal/growth & development
20.
Nat Cell Biol ; 20(10): 1228, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30089841

ABSTRACT

In the version of this Article originally published, the competing interests statement was missing. The authors declare no competing interests; this statement has now been added in all online versions of the Article.

SELECTION OF CITATIONS
SEARCH DETAIL