Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 136
Filter
1.
Annu Rev Physiol ; 83: 107-126, 2021 02 10.
Article in English | MEDLINE | ID: mdl-33141632

ABSTRACT

Mitochondria are responsible for ATP production but are also known as regulators of cell death, and mitochondrial matrix Ca2+ is a key modulator of both ATP production and cell death. Although mitochondrial Ca2+ uptake and efflux have been studied for over 50 years, it is only in the past decade that the proteins responsible for mitochondrial Ca2+ uptake and efflux have been identified. The identification of the mitochondrial Ca2+ uniporter (MCU) led to an explosion of studies identifying regulators of the MCU. The levels of these regulators vary in a tissue- and disease-specific manner, providing new insight into how mitochondrial Ca2+ is regulated. This review focuses on the proteins responsible for mitochondrial transport and what we have learned from mouse studies with genetic alterations in these proteins.


Subject(s)
Biological Transport/physiology , Calcium Channels/metabolism , Calcium/metabolism , Mitochondria/metabolism , Animals , Humans
2.
J Biol Chem ; 299(1): 102780, 2023 01.
Article in English | MEDLINE | ID: mdl-36496071

ABSTRACT

Ischemia and reperfusion affect multiple elements of cardiomyocyte electrophysiology, especially within the mitochondria. We previously showed that in cardiac monolayers, upon reperfusion after coverslip-induced ischemia, mitochondrial inner membrane potential (ΔΨ) unstably oscillates between polarized and depolarized states, and ΔΨ instability corresponds with arrhythmias. Here, through confocal microscopy of compartment-specific molecular probes, we investigate the mechanisms underlying the postischemic ΔΨ oscillations, focusing on the role of Ca2+ and oxidative stress. During reperfusion, transient ΔΨ depolarizations occurred concurrently with periods of increased mitochondrial oxidative stress (5.07 ± 1.71 oscillations/15 min, N = 100). Supplementing the antioxidant system with GSH monoethyl ester suppressed ΔΨ oscillations (1.84 ± 1.07 oscillations/15 min, N = 119, t test p = 0.027) with 37% of mitochondrial clusters showing no ΔΨ oscillations (versus 4% in control, odds ratio = 14.08, Fisher's exact test p < 0.001). We found that limiting the production of reactive oxygen species using cyanide inhibited postischemic ΔΨ oscillations (N = 15, t test p < 10-5). Furthermore, ΔΨ oscillations were not associated with any discernable pattern in cell-wide oxidative stress or with the changes in cytosolic or mitochondrial Ca2+. Sustained ΔΨ depolarization followed cytosolic and mitochondrial Ca2+ increase and was associated with increased cell-wide oxidative stress. Collectively, these findings suggest that transient bouts of increased mitochondrial oxidative stress underlie postischemic ΔΨ oscillations, regardless of Ca2+ dynamics.


Subject(s)
Mitochondria, Heart , Oxidative Stress , Humans , Calcium/metabolism , Ischemia/metabolism , Membrane Potential, Mitochondrial , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Reactive Oxygen Species/metabolism , Reperfusion
3.
Circ Res ; 130(5): 741-759, 2022 03 04.
Article in English | MEDLINE | ID: mdl-35109669

ABSTRACT

BACKGROUND: Abnormalities in cardiac energy metabolism occur in heart failure (HF) and contribute to contractile dysfunction, but their role, if any, in HF-related pathologic remodeling is much less established. CK (creatine kinase), the primary muscle energy reserve reaction which rapidly provides ATP at the myofibrils and regenerates mitochondrial ADP, is down-regulated in experimental and human HF. We tested the hypotheses that pathologic remodeling in human HF is related to impaired cardiac CK energy metabolism and that rescuing CK attenuates maladaptive hypertrophy in experimental HF. METHODS: First, in 27 HF patients and 14 healthy subjects, we measured cardiac energetics and left ventricular remodeling using noninvasive magnetic resonance 31P spectroscopy and magnetic resonance imaging, respectively. Second, we tested the impact of metabolic rescue with cardiac-specific overexpression of either Ckmyofib (myofibrillar CK) or Ckmito (mitochondrial CK) on HF-related maladaptive hypertrophy in mice. RESULTS: In people, pathologic left ventricular hypertrophy and dilatation correlate closely with reduced myocardial ATP levels and rates of ATP synthesis through CK. In mice, transverse aortic constriction-induced left ventricular hypertrophy and dilatation are attenuated by overexpression of CKmito, but not by overexpression of CKmyofib. CKmito overexpression also attenuates hypertrophy after chronic isoproterenol stimulation. CKmito lowers mitochondrial reactive oxygen species, tissue reactive oxygen species levels, and upregulates antioxidants and their promoters. When the CK capacity of CKmito-overexpressing mice is limited by creatine substrate depletion, the protection against pathologic remodeling is lost, suggesting the ADP regenerating capacity of the CKmito reaction rather than CK protein per se is critical in limiting adverse HF remodeling. CONCLUSIONS: In the failing human heart, pathologic hypertrophy and adverse remodeling are closely related to deficits in ATP levels and in the CK energy reserve reaction. CKmito, sitting at the intersection of cardiac energetics and redox balance, plays a crucial role in attenuating pathologic remodeling in HF. Registration: URL: https://www.clinicaltrials.gov; Unique identifier: NCT00181259.


Subject(s)
Creatine Kinase, Mitochondrial Form , Heart Failure , Adenosine Diphosphate , Adenosine Triphosphate/metabolism , Animals , Creatine Kinase/metabolism , Creatine Kinase, Mitochondrial Form/metabolism , Energy Metabolism , Heart Failure/metabolism , Humans , Hypertrophy, Left Ventricular/metabolism , Mice , Myocardium/metabolism , Reactive Oxygen Species/metabolism , Ventricular Remodeling
4.
Am J Physiol Heart Circ Physiol ; 324(5): H598-H609, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36827227

ABSTRACT

Insulin resistance (IR) is one of the hallmarks of heart failure (HF). Abnormalities in skeletal muscle (SM) metabolism have been identified in patients with HF. However, the underlying mechanisms of IR development in SM in HF are poorly understood. Herein, we hypothesize that HF upregulates miR-133b in SM and in turn alters glucose metabolism and the propensity toward IR. Mitochondria isolated from SM of mice with HF induced by transverse aortic constriction (TAC) showed lower respiration and downregulation of muscle-specific components of the tricarboxylic acid (TCA) cycle, AMP deaminase 1 (AMPD1), and fumarate compared with those from control animals. RNA-Seq and subsequent qPCR validation confirmed upregulation of SM-specific microRNA (miRNA), miR-133b, in TAC versus sham animals. miR-133b overexpression alone resulted in significantly lower mitochondrial respiration, cellular glucose uptake, and glycolysis along with lower ATP production and cellular energy reserve compared with the scramble (Scr) in C2C12 cells. miR-133b binds to the 3'-untranslated region (UTR) of KLF15, the transcription factor for the insulin-sensitive glucose transporter, GLUT4. Overexpression of miR-133b lowers GLUT4 and lowers pAkt in presence of insulin in C2C12 cells. Finally, lowering miR-133b in primary skeletal myocytes isolated from TAC mice using antagomir-133b reversed the changes in KLF15, GLUT4, and AMPD1 compared with the scramble-transfected myocytes. Taken together, these data demonstrate a role for SM miR-133b in altered glucose metabolism in HF and suggest the therapeutic potential in HF to improve glucose uptake and glycolysis by restoring GLUT4 abundance. The data uncover a novel mechanism for IR and ultimately SM metabolic abnormalities in patients with HF.NEW & NOTEWORTHY Heart failure is associated with systemic insulin resistance and abnormalities in glucose metabolism but the underlying mechanisms are poorly understood. In the skeletal muscle, the major peripheral site of glucose utilization, we observe an increase in miR-133b in heart failure mice, which reduces the insulin-sensitive glucose transporter (GLUT4), glucose uptake, and metabolism in C2C12 and in myocytes. The antagomir for miR-133b restores GLUT4 protein and markers of metabolism in skeletal myocytes from heart failure mice demonstrating that miR-133b is an exciting target for systemic insulin resistance in heart failure and an important player in the cross talk between the heart and the periphery in the heart failure syndrome.


Subject(s)
Heart Failure , Insulin Resistance , MicroRNAs , Mice , Animals , Insulin Resistance/genetics , Antagomirs/metabolism , Muscle, Skeletal/metabolism , Glucose/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Insulin/metabolism , Heart Failure/genetics , Heart Failure/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Glucose Transporter Type 4/genetics , Glucose Transporter Type 4/metabolism
5.
J Mol Cell Cardiol ; 144: 87-96, 2020 07.
Article in English | MEDLINE | ID: mdl-32442661

ABSTRACT

AIMS: In cardiomyocytes, there is microRNA (miR) in the mitochondria that originates from the nuclear genome and matures in the cytoplasm before translocating into the mitochondria. Overexpression of one such miR, miR-181c, can lead to heart failure by stimulating reactive oxygen species (ROS) production and increasing mitochondrial calcium level ([Ca2+]m). Mitochondrial calcium uptake 1 protein (MICU1), a regulatory protein in the mitochondrial calcium uniporter complex, plays an important role in regulating [Ca2+]m. Obesity results in miR-181c overexpression and a decrease in MICU1. We hypothesize that lowering miR-181c would protect against obesity-induced cardiac dysfunction. METHODS AND RESULTS: We used an in vivo mouse model of high-fat diet (HFD) for 18 weeks and induced high lipid load in H9c2 cells with oleate-conjugated bovine serum albumin in vitro. We tested the cardioprotective role of lowering miR-181c by using miR-181c/d-/- mice (in vivo) and AntagomiR against miR-181c (in vitro). HFD significantly upregulated heart levels of miR-181c and led to cardiac hypertrophy in wild-type mice, but not in miR-181c/d-/- mice. HFD also increased ROS production and pyruvate dehydrogenase activity (a surrogate for [Ca2+]m), but the increases were alleviated in miR-181c/d-/- mice. Moreover, miR-181c/d-/- mice fed a HFD had higher levels of MICU1 than did wild-type mice fed a HFD, attenuating the rise in [Ca2+]m. Overexpression of miR-181c in neonatal ventricular cardiomyocytes (NMVM) caused increased ROS production, which oxidized transcription factor Sp1 and led to a loss of Sp1, thereby slowing MICU1 transcription. Hence, miR-181c increases [Ca2+]m through Sp1 oxidation and downregulation of MICU1, suggesting that the cardioprotective effect of miR-181c/d-/- results from inhibition of Sp1 oxidation. CONCLUSION: This study has identified a unique nuclear-mitochondrial communication mechanism in the heart orchestrated by miR-181c. Obesity-induced overexpression of miR-181c increases [Ca2+]m via downregulation of MICU1 and leads to cardiac injury. A strategy to inhibit miR-181c in cardiomyocytes can preserve cardiac function during obesity by improving mitochondrial function. Altering miR-181c expression may provide a pharmacologic approach to improve cardiomyopathy in individuals with obesity/type 2 diabetes.


Subject(s)
Cell Nucleus/metabolism , MicroRNAs/genetics , Mitochondria, Heart/metabolism , Obesity/genetics , Obesity/metabolism , Ventricular Dysfunction/etiology , Ventricular Dysfunction/metabolism , Animals , Biomarkers , Calcium/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/physiopathology , Diet, High-Fat , Disease Models, Animal , Disease Susceptibility , Gene Expression Regulation , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/physiopathology , Mice , Mice, Knockout , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Myocytes, Cardiac/metabolism , Obesity/complications , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Sp1 Transcription Factor/metabolism , Ventricular Dysfunction/physiopathology
6.
Am J Physiol Heart Circ Physiol ; 317(5): H1116-H1124, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31625778

ABSTRACT

Vascular stiffness plays a key role in the pathogenesis of hypertension. Recent studies indicate that the age-associated reduction in miR-181b levels in vascular smooth muscle cells (VSMCs) contributes to increased vascular stiffness. As these findings suggest that inhibiting degradation of miR-181b might prevent vascular stiffening, we have assessed whether the microRNA-degrading translin/trax (TN/TX) complex mediates degradation of miR-181b in the aorta.We found that TN-/- mice display elevated levels of miR-181b expression in the aorta. Therefore, we tested whether TN deletion prevents vascular stiffening in a mouse model of hypertension, induced by chronic high-salt intake (4%NaCl in drinking water for 3 wk; HSW). TN-/- mice subjected to HSW stress do not show increased vascular stiffness, as monitored by pulse wave velocity and tensile testing. The protective effect of TN deletion in the HSW paradigm appears to be mediated by its ability to increase miR-181b in the aorta since HSW decreases levels of miR-181b in WT mice, but not in TN KO mice. We demonstrate for the first time that interfering with microRNA degradation can have a beneficial impact on the vascular system and identify the microRNA-degrading TN/TX RNase complex as a potential therapeutic target in combatting vascular stiffness.NEW & NOTEWORTHY While the biogenesis and mechanism of action of mature microRNA are well understood, much less is known about the regulation of microRNA via degradation. Recent studies have identified the protein complex, translin(TN)/trax(TX), as a microRNA-degrading enzyme. Here, we demonstrate that TN/TX is expressed in vascular smooth muscle cells. Additionally, deletion of the TN/TX complex selectively increases aortic miR-181b and prevents increased vascular stiffness caused by ingestion of high-salt water. To our knowledge, this is first report describing the role of a microRNA RNAse in cardiovascular biology or pathobiology.


Subject(s)
Aorta/enzymology , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/metabolism , Hypertension/enzymology , MicroRNAs/metabolism , Vascular Stiffness , Animals , Aorta/physiopathology , DNA-Binding Proteins/genetics , Disease Models, Animal , Gene Deletion , Hypertension/genetics , Hypertension/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , RNA Stability , RNA-Binding Proteins/genetics , Sodium Chloride, Dietary , Up-Regulation
7.
J Card Fail ; 23(4): 340-344, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28011000

ABSTRACT

BACKGROUND: Recent studies have suggested a high prevalence of cardiac amyloidosis (CAm) in heart failure (HF) patients. CAm might be underdiagnosed owing to low clinical suspicion. METHODS AND RESULTS: We performed retrospective analysis of 259 patients with HF and ejection fraction (EF) ≥50% referred for endomyocardial biopsy. Seventy-three (28%) had CAm. Multivariable independent predictors of CAm were identified. Over a mean follow-up of 2.6 ± 3.3 years, CAm patients had worse survival than those without (1.5 y vs 6.3 y; log rank P < .0001). CONCLUSIONS: Clinicians should be suspicious of CAm in patients with EF 50%-75%, >50 years of age, BMI <30 kg/m2, peripheral neuropathy, Sokolow-Lyon index ≤15 mm, and septal wall thickness ≥1.4 cm.


Subject(s)
Amyloid , Amyloidosis/diagnosis , Endocardium , Endomyocardial Fibrosis/diagnosis , Heart Failure , Myocardium , Aged , Amyloid/analysis , Amyloid/metabolism , Amyloidosis/pathology , Biopsy/methods , Electrocardiography/methods , Endocardium/metabolism , Endocardium/pathology , Endomyocardial Fibrosis/pathology , Female , Heart Failure/metabolism , Heart Failure/mortality , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Maryland , Middle Aged , Myocardium/metabolism , Myocardium/pathology , Prognosis , Retrospective Studies , Statistics as Topic , Stroke Volume
8.
Circ Res ; 116(4): 572-86, 2015 Feb 13.
Article in English | MEDLINE | ID: mdl-25499773

ABSTRACT

RATIONALE: Despite 4 decades of intense effort and substantial financial investment, the cardioprotection field has failed to deliver a single drug that effectively reduces myocardial infarct size in patients. A major reason is insufficient rigor and reproducibility in preclinical studies. OBJECTIVE: To develop a multicenter, randomized, controlled, clinical trial-like infrastructure to conduct rigorous and reproducible preclinical evaluation of cardioprotective therapies. METHODS AND RESULTS: With support from the National Heart, Lung, and Blood Institute, we established the Consortium for preclinicAl assESsment of cARdioprotective therapies (CAESAR), based on the principles of randomization, investigator blinding, a priori sample size determination and exclusion criteria, appropriate statistical analyses, and assessment of reproducibility. To validate CAESAR, we tested the ability of ischemic preconditioning to reduce infarct size in 3 species (at 2 sites/species): mice (n=22-25 per group), rabbits (n=11-12 per group), and pigs (n=13 per group). During this validation phase, (1) we established protocols that gave similar results between centers and confirmed that ischemic preconditioning significantly reduced infarct size in all species and (2) we successfully established a multicenter structure to support CAESAR's operations, including 2 surgical centers for each species, a Pathology Core (to assess infarct size), a Biomarker Core (to measure plasma cardiac troponin levels), and a Data Coordinating Center-all with the oversight of an external Protocol Review and Monitoring Committee. CONCLUSIONS: CAESAR is operational, generates reproducible results, can detect cardioprotection, and provides a mechanism for assessing potential infarct-sparing therapies with a level of rigor analogous to multicenter, randomized, controlled clinical trials. This is a revolutionary new approach to cardioprotection. Importantly, we provide state-of-the-art, detailed protocols ("CAESAR protocols") for measuring infarct size in mice, rabbits, and pigs in a manner that is rigorous, accurate, and reproducible.


Subject(s)
Cardiovascular Agents/pharmacology , Drug Evaluation, Preclinical , Ischemic Preconditioning, Myocardial/methods , Myocardial Infarction/prevention & control , National Heart, Lung, and Blood Institute (U.S.) , Research Design , Animals , Biomarkers/blood , Cooperative Behavior , Disease Models, Animal , Drug Evaluation, Preclinical/standards , Female , Guidelines as Topic , Humans , Ischemic Preconditioning, Myocardial/standards , Male , Mice , Myocardial Infarction/blood , Myocardial Infarction/pathology , Myocardium/pathology , Predictive Value of Tests , Rabbits , Reproducibility of Results , Research Design/standards , Species Specificity , Swine , Time Factors , Troponin I/blood , United States
9.
Am J Physiol Heart Circ Physiol ; 310(4): H505-15, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26702143

ABSTRACT

Premenopausal women exhibit endogenous cardioprotective signaling mechanisms that are thought to result from the beneficial effects of estrogen, which we have shown to increase protein S-nitrosylation in the heart. S-nitrosylation is a labile protein modification that increases with a number of different forms of cardioprotection, including ischemic preconditioning. Herein, we sought to identify a potential role for protein S-nitrosylation in sex-dependent cardioprotection. We utilized a Langendorff-perfused mouse heart model of ischemia-reperfusion injury with male and female hearts, and S-nitrosylation-resin-assisted capture with liquid chromatography tandem mass spectrometry to identify S-nitrosylated proteins and modification sites. Consistent with previous studies, female hearts exhibited resilience to injury with a significant increase in functional recovery compared with male hearts. In a separate set of hearts, we identified a total of 177 S-nitrosylated proteins in female hearts at baseline compared with 109 S-nitrosylated proteins in male hearts. Unique S-nitrosylated proteins in the female group included the F1FO-ATPase and cyclophilin D. We also utilized label-free peptide analysis to quantify levels of common S-nitrosylated identifications and noted that the S-nitrosylation of sarcoplasmic/endoplasmic reticulum Ca(2+)-ATPase 2a was nearly 70% lower in male hearts compared with female, with no difference in expression. Furthermore, we found a significant increase in endothelial nitric oxide synthase expression, phosphorylation, and total nitric oxide production in female hearts compared with males, likely accounting for the enhanced S-nitrosylation protein levels in female hearts. In conclusion, we identified a number of novel S-nitrosylated proteins in female hearts that are likely to contribute to sex-dependent cardioprotection.


Subject(s)
Coronary Circulation/drug effects , Heart/drug effects , Proteome/drug effects , S-Nitrosothiols/metabolism , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/metabolism , Endoplasmic Reticulum/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Myocardial Reperfusion Injury/metabolism , Myocardium/metabolism , Nitric Oxide/biosynthesis , Nitric Oxide Synthase Type III/biosynthesis , Phosphorylation , Sex Characteristics
10.
Circ Res ; 114(8): 1268-80, 2014 Apr 11.
Article in English | MEDLINE | ID: mdl-24557881

ABSTRACT

RATIONALE: There is tight coupling between Akt activation and suppression of cell death. Full Akt activation requires mammalian target of rapamycin complex 2 (mTORC2), but the regulation of mTORC2 is unclear. OBJECTIVE: To gain new insights into mechanisms of mTORC2/Akt signaling. METHODS AND RESULTS: The role of mTORC2 in cardioprotection was examined. In perfused mouse hearts, ischemic preconditioning increased mTORC2 activity, leading to phosphorylation of Akt on Ser473. The protective effect of ischemic preconditioning was lost by pretreatment with dual mTORC inhibitors but not with rapamycin, an mTORC1 inhibitor, which indicates the fundamental role of mTORC2 activation in cardioprotection. Next, the regulation and downstream targets of mTORC2/Akt signaling were explored. We have found that ischemic preconditioning and other Akt activators (insulin and opioids) result in phosphorylation of ribosomal protein S6 (Rps6) at Ser235/236 in mouse hearts and neonatal rat ventricular myocytes. Rps6 interacts with components of mTORC2, and siRNA-mediated knockdown of Rps6 attenuates insulin-induced mTORC2 activation and Akt-Ser473 phosphorylation. On the other hand, Rps6 overexpression enhanced Akt-Ser473 phosphorylation, indicating that Rps6 activation amplifies mTORC2/Akt signaling. Disruption of the Rps6/mTORC2 pathway by knockdown of Rps6 or rictor abrogated insulin-induced cytoprotection against oxidative stress. Although rapamycin blocks Rps6-dependent mTORC2 activation, mTORC2 is still activated by an alternative signaling pathway, demonstrating the redundancy in cardioprotective signaling. CONCLUSIONS: Activation of mTORC2 plays a pivotal role in cardioprotection, and Rps6 is a convergence point of cardioprotective signaling, providing positive feedback regulation of mTORC2/Akt signaling.


Subject(s)
Multiprotein Complexes/physiology , Myocardial Ischemia/prevention & control , Proto-Oncogene Proteins c-akt/physiology , Ribosomal Protein S6/physiology , Signal Transduction/physiology , TOR Serine-Threonine Kinases/physiology , Animals , Cells, Cultured , Disease Models, Animal , Feedback, Physiological/physiology , Ischemic Preconditioning, Myocardial , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Mice, Inbred C57BL , Myocardial Ischemia/physiopathology , Phosphorylation/physiology
11.
J Immunol ; 192(4): 1806-14, 2014 Feb 15.
Article in English | MEDLINE | ID: mdl-24403532

ABSTRACT

Sepsis is a major cause of mortality, and dysregulation of the immune response plays a central role in this syndrome. H2S, a recently discovered gaso-transmitter, is endogenously generated by many cell types, regulating a number of physiologic processes and pathophysiologic conditions. We report that H2S increased survival after experimental sepsis induced by cecal ligation and puncture (CLP) in mice. Exogenous H2S decreased the systemic inflammatory response, reduced apoptosis in the spleen, and accelerated bacterial eradication. We found that C/EBP homologous protein 10 (CHOP), a mediator of the endoplasmic reticulum stress response, was elevated in several organs after CLP, and its expression was inhibited by H2S treatment. Using CHOP-knockout (KO) mice, we demonstrated for the first time, to our knowledge, that genetic deletion of Chop increased survival after LPS injection or CLP. CHOP-KO mice displayed diminished splenic caspase-3 activation and apoptosis, decreased cytokine production, and augmented bacterial clearance. Furthermore, septic CHOP-KO mice treated with H2S showed no additive survival benefit compared with septic CHOP-KO mice. Finally, we showed that H2S inhibited CHOP expression in macrophages by a mechanism involving Nrf2 activation. In conclusion, our findings show a protective effect of H2S treatment afforded, at least partially, by inhibition of CHOP expression. The data reveal a major negative role for the transcription factor CHOP in overall survival during sepsis and suggest a new target for clinical intervention, as well potential strategies for treatment.


Subject(s)
Bacteria/immunology , Hydrogen Sulfide/metabolism , Sepsis/metabolism , Transcription Factor CHOP/antagonists & inhibitors , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Cecum/surgery , Cytokines/biosynthesis , Endoplasmic Reticulum Stress/drug effects , Enzyme Activation , Lipopolysaccharides , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Sepsis/drug therapy , Spleen/drug effects , Survival , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics
12.
Am J Physiol Heart Circ Physiol ; 309(8): H1271-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26254336

ABSTRACT

Levels of the HER2/ErbB2 protein in the heart are upregulated in some women during breast cancer therapy, and these women are at high risk for developing heart dysfunction after sequential treatment with anti-ErbB2/trastuzumab or doxorubicin. Doxorubicin is known to increase oxidative stress in the heart, and thus we considered the possibility that ErbB2 protein influences the status of cardiac antioxidant defenses in cardiomyocytes. In this study, we measured reactive oxygen species (ROS) in cardiac mitochondria and whole hearts from mice with cardiac-specific overexpression of ErbB2 (ErbB2(tg)) and found that, compared with control mice, high levels of ErbB2 in myocardium result in lower levels of ROS in mitochondria (P = 0.0075) and whole hearts (P = 0.0381). Neonatal cardiomyocytes isolated from ErbB2(tg) hearts have lower ROS levels and less cellular death (P < 0.0001) following doxorubicin treatment. Analyzing antioxidant enzyme levels and activities, we found that ErbB2(tg) hearts have increased levels of glutathione peroxidase 1 (GPx1) protein (P < 0.0001) and GPx activity (P = 0.0031) in addition to increased levels of two known GPx activators, c-Abl (P = 0.0284) and Arg (P < 0.0001). Interestingly, although mitochondrial ROS emission is reduced in the ErbB2(tg) hearts, oxygen consumption rates and complex I activity are similar to control littermates. Compared with these in vivo studies, H9c2 cells transfected with ErbB2 showed less cellular toxicity and produced less ROS (P < 0.0001) after doxorubicin treatment but upregulated GR activity (P = 0.0237) instead of GPx. Our study shows that ErbB2-dependent signaling contributes to antioxidant defenses and suggests a novel mechanism by which anticancer therapies involving ErbB2 antagonists can harm myocardial structure and function.


Subject(s)
Antineoplastic Agents/toxicity , Antioxidants/metabolism , Doxorubicin/toxicity , Glutathione Peroxidase/metabolism , Heart Diseases/prevention & control , Mitochondria, Heart/drug effects , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Receptor, ErbB-2/metabolism , Animals , Animals, Newborn , Cell Death/drug effects , Cell Line , Dose-Response Relationship, Drug , Glutathione Reductase/metabolism , Heart Diseases/chemically induced , Heart Diseases/enzymology , Heart Diseases/genetics , Heart Diseases/pathology , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/enzymology , Mitochondria, Heart/pathology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Rats , Receptor, ErbB-2/genetics , Glutathione Peroxidase GPX1
14.
Circ Res ; 113(4): 464-77, 2013 Aug 02.
Article in English | MEDLINE | ID: mdl-23908333

ABSTRACT

With the advent of thrombolytic therapy and angioplasty, it has become possible to reduce myocardial infarct size through early reperfusion. Enormous effort has been expended to find therapies that can further reduce infarct size after early intervention. Animal studies have identified many cardioprotective pathways that have the potential to reduce infarct size if activated before the onset of ischemia. More recently, interventions effective at the onset of reperfusion have been described. Although basic research has identified many targets, most has been conducted in rodent models which may not be directly applicable to human disease and even promising agents have been disappointing in large-scale clinical trials. There are many potential explanations for this failure which is the subject of this review. Potential factors include (1) the variability inherent in the patient population, whereas animal studies usually use single sex homogeneous groups maintained on standard diets in carefully controlled environments; (2) the duration of ischemia is generally shorter in animal studies, resulting in potentially more salvageable myocardium than is often the case in patients; (3) that the animals are usually young without comorbidities, whereas the patient population is generally older and has significant comorbidities; (4) animals are not treated with medications a priori, whereas the patient population is often taking medications that may affect ischemic injury; and (5) animal studies may not involve thorough assessment of effects on organs other than the heart, whereas patients can experience adverse effects of treatment in other organs that can preclude clinical use.


Subject(s)
Cardiotonic Agents/therapeutic use , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Translational Research, Biomedical , Animals , Cardiotonic Agents/adverse effects , Clinical Trials as Topic , Disease Models, Animal , Humans , Myocardial Infarction/pathology , Myocardial Reperfusion Injury/pathology , Severity of Illness Index , Time Factors
15.
Circ Res ; 113(12): 1308-19, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24062335

ABSTRACT

RATIONALE: Mice lacking cyclophilin D (CypD(-/-)), a mitochondrial chaperone protein, have altered cardiac metabolism. As acetylation has been shown to regulate metabolism, we tested whether changes in protein acetylation might play a role in these metabolic changes in CypD(-/-) hearts. OBJECTIVE: Our aim was to test the hypothesis that loss of CypD alters the cardiac mitochondrial acetylome. METHODS AND RESULTS: To identify changes in lysine-acetylated proteins and to map acetylation sites after ablation of CypD, we subjected tryptic digests of isolated cardiac mitochondria from wild-type and CypD(-/-) mice to immunoprecipitation using agarose beads coupled to antiacetyl lysine antibodies followed by mass spectrometry. We used label-free analysis for the relative quantification of the 875 common peptides that were acetylated in wild-type and CypD(-/-) samples and found 11 peptides (10 proteins) decreased and 96 peptides (48 proteins) increased in CypD(-/-) samples. We found increased acetylation of proteins in fatty acid oxidation and branched-chain amino acid metabolism. To evaluate whether this increase in acetylation might play a role in the inhibition of fatty acid oxidation that was previously reported in CypD(-/-) hearts, we measured the activity of l-3-hydroxyacyl-CoA dehydrogenase, which was acetylated in the CypD(-/-) hearts. Consistent with the hypothesis, l-3-hydroxyacyl-CoA dehydrogenase activity was inhibited by ≈50% compared with the wild-type mitochondria. CONCLUSIONS: These results implicate a role for CypD in modulating protein acetylation. Taken together, these results suggest that ablation of CypD leads to changes in the mitochondrial acetylome, which may contribute to altered mitochondrial metabolism in CypD(-/-) mice.


Subject(s)
Cyclophilins/physiology , Mitochondria, Heart/metabolism , Acetylation , Animals , Peptidyl-Prolyl Isomerase F , Cyclophilins/antagonists & inhibitors , Cyclophilins/deficiency , Male , Mice , Mice, Knockout , Organ Culture Techniques , Proteome/genetics
16.
Mol Ther ; 22(4): 752-61, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24434934

ABSTRACT

Fragile skin, susceptible to decubitus ulcers and incidental trauma, is a problem particularly for the elderly and for those with spinal cord injury. Here, we present a simple approach to strengthen the skin by the topical delivery of keratinocyte growth factor-1 (KGF-1) DNA. In initial feasibility studies with the novel minimalized, antibiotic-free DNA expression vector, NTC8385-VA1, the reporter genes luciferase and enhanced green fluorescent protein were delivered. Transfection was documented when luciferase expression significantly increased after transfection. Microscopic imaging of enhanced green fluorescent protein-transfected skin showed green fluorescence in hair follicles, hair shafts, and dermal and superficial epithelial cells. With KGF-1 transfection, KGF-1 mRNA level and protein production were documented with quantitative reverse transcriptase-polymerase chain reaction and immunohistochemistry, respectively. Epithelial thickness of the transfected skin in the KGF group was significantly increased compared with the control vector group (26 ± 2 versus 16 ± 4 µm) at 48 hours (P = 0.045). Dermal thickness tended to be increased in the KGF group (255 ± 36 versus 162 ± 16 µm) at 120 hours (P = 0.057). Biomechanical assessment showed that the KGF-1-treated skin was significantly stronger than control vector-transfected skin. These findings indicate that topically delivered KGF-1 DNA plasmid can increase epithelial thickness and strength, demonstrating the potential of this approach to restore compromised skin.


Subject(s)
Fibroblast Growth Factor 7/genetics , Gene Transfer Techniques , Genetic Therapy , Skin Abnormalities/genetics , Administration, Topical , Animals , DNA/administration & dosage , DNA/genetics , Fibroblast Growth Factor 7/administration & dosage , Humans , Mice , Plasmids/administration & dosage , Skin Abnormalities/therapy , Wound Healing/genetics
17.
Proc Natl Acad Sci U S A ; 109(14): E841-50, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22403061

ABSTRACT

Chronic systemic hypertension causes cardiac pressure overload leading to increased myocardial O(2) consumption. Hypoxia-inducible factor 1 (HIF-1) is a master regulator of O(2) homeostasis. Mouse embryos lacking expression of the O(2)-regulated HIF-1α subunit die at midgestation with severe cardiac malformations and vascular regression. Here we report that Hif1a(f/f);Tie2-Cre conditional knockout mice, which lack HIF-1α expression only in Tie2(+) lineage cells, develop normally, but when subjected to pressure overload induced by transaortic constriction (TAC), they manifest rapid cardiac decompensation, which is accompanied by excess cardiac fibrosis and myocardial hypertrophy, decreased myocardial capillary density, increased myocardial hypoxia and apoptosis, and increased TGF-ß signaling through both canonical and noncanonical pathways that activate SMAD2/3 and ERK1/2, respectively, within endothelial cells of cardiac blood vessels. TAC also induces dilatation of the proximal aorta through enhanced TGF-ß signaling in Hif1a(f/f);Tie2-Cre mice. Inhibition of TGF-ß signaling by treatment with neutralizing antibody or pharmacologic inhibition of MEK-ERK signaling prevented TAC-induced contractile dysfunction and pathological remodeling. Thus, HIF-1 plays a critical protective role in the adaptation of the heart and aorta to pressure overload by negatively regulating TGF-ß signaling in endothelial cells. Treatment of wild-type mice with digoxin, which inhibits HIF-1α synthesis, resulted in rapid cardiac failure after TAC. Although digoxin has been used for decades as an inotropic agent to treat heart failure, it does not improve survival, suggesting that the countertherapeutic effects of digoxin observed in the TAC mouse model may have clinical relevance.


Subject(s)
Aorta/physiopathology , Endothelium, Vascular/metabolism , Heart/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Signal Transduction , Transforming Growth Factor beta/antagonists & inhibitors , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Mice, Knockout , Pressure , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
18.
J Mol Cell Cardiol ; 69: 67-74, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24487118

ABSTRACT

Oxidative stress and membrane damage following myocardial ischemia/reperfusion injury are important contributors to cardiomyocyte death and the loss of myocardial function. Our previous study identified cysteine 144 (C144) of tripartite motif-containing protein 72 (TRIM72) as a potential site for S-nitrosylation (SNO). TRIM72 is a cardioprotective membrane repair protein that can be both activated and targeted for degradation by different oxidative modifications. Consistent with the potential regulation of TRIM72 by various oxidative modifications, we found that SNO levels increased at C144 of TRIM72 with ischemic preconditioning. Therefore, to investigate the role of C144 in the regulation of TRIM72 function, we mutated C144 of TRIM72 to a serine residue (TRIM72(C144S)), and expressed either TRIM72(WT) or TRIM72(C144S) in HEK-293 cells, which lack endogenous TRIM72, in order to examine the effect of this mutation on the functional stability of TRIM72 and on cell survival. We hypothesized that SNO of TRIM72 stabilizes the protein, thus allowing for membrane repair and enhanced cell survival. Upon treatment with hydrogen peroxide (H2O2), we found that TRIM72(WT) levels were decreased, but not TRIM72(C144S) and this correlated with increased H2O2-induced cell death in TRIM72(WT) cells. Additionally, we found that treatment with the cardioprotective S-nitrosylating agent S-nitrosoglutathione (GSNO), was able to preserve TRIM72(WT) protein levels and enhance TRIM72(WT)-mediated cell survival, but had no effect on TRIM72(C144S) levels. Consistent with our hypothesis, GSNO was also found to increase SNO levels and inhibit H2O2-induced irreversible oxidation for TRIM72(WT) without affecting TRIM72(C144S). In further support of our hypothesis, GSNO blocked the ischemia/reperfusion-induced decrease in TRIM72 levels and reduced infarct size in a Langendorff-perfused heart model. The results of these studies have important implications for cardioprotection and suggest that SNO of TRIM72 at C144 prevents the oxidation-induced degradation of TRIM72 following oxidative insult, therefore enhancing cardiomyocyte survival.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cysteine/chemistry , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/pathology , Nitric Oxide/metabolism , Animals , Blotting, Western , Carrier Proteins/genetics , Cell Death , Cell Survival , Cysteine/metabolism , Female , Ischemic Preconditioning , Male , Membrane Proteins , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Reperfusion Injury/metabolism , Oxidation-Reduction , Proteolysis
19.
J Mol Cell Cardiol ; 73: 18-25, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24440455

ABSTRACT

Nitric oxide is a gaseous signaling molecule that is well-known for the Nobel prize-winning research that defined nitric oxide as a physiological regulator of blood pressure in the cardiovascular system. Nitric oxide can signal via the classical pathway involving activation of guanylyl cyclase or by a post-translational modification, referred to as S-nitrosylation (SNO) that can occur on cysteine residues of proteins. As proteins with cysteine residues are common, this allows for amplification of the nitric oxide signaling. This review will focus on the possible mechanisms through which SNO can alter protein function in cardiac cells, and the role of SNO occupancy in these mechanisms. The specific mechanisms that regulate protein SNO, including redox-dependent processes, will also be discussed. This article is part of a Special Issue entitled "Redox Signalling in the Cardiovascular System".


Subject(s)
Heart , S-Nitrosothiols/metabolism , Signal Transduction/physiology , Animals , Humans , Nitric Oxide/metabolism , Oxidation-Reduction , Protein Processing, Post-Translational
20.
Am J Physiol Heart Circ Physiol ; 306(6): H825-32, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24441547

ABSTRACT

Previous studies have shown a role for nitric oxide and S-nitrosylation (SNO) in postconditioning (PostC), but specific SNO proteins and sites have not been identified in the myocardium after PostC. In this study, we examined SNO signaling in PostC using a Langendorff-perfused mouse heart model. After 20 min of equilibrium perfusion and 25 min of global ischemia, PostC was applied at the beginning of reperfusion with six cycles of 10 s of reperfusion and 10 s of ischemia. The total period of reperfusion was 90 min. Compared with the ischemia-reperfusion (I/R) control, PostC significantly reduced postischemic contractile dysfunction and infarct size. PostC-induced protection was blocked by treatment with N(G)-nitro-l-arginine methyl ester (l-NAME) (10 µmol/l; a constitutive NO synthase inhibitor), but not by either ODQ (10 µmol/l, a highly selective soluble guanylyl cyclase inhibitor) or KT5823 (1 µmol/l, a specific protein kinase G inhibitor). Two biotin switch based methods, two dimensional CyDye-maleimide difference gel electrophoresis (2D CyDye-maleimide DIGE) and SNO-resin-assisted capture (SNO-RAC), were utilized to identify SNO-modified proteins and sites. Using 2D CyDye-maleimide DIGE analysis, PostC was found to cause a 25% or greater increase in SNO of a number of proteins, which was blocked by treatment with l-NAME in parallel with the loss of protection. Using SNO-RAC, we identified 77 unique proteins with SNO sites after PostC. These results suggest that NO-mediated SNO signaling is involved in PostC-induced cardioprotection and these data provide the first set of candidate SNO proteins in PostC hearts.


Subject(s)
Nitric Oxide/metabolism , Protein S/metabolism , Reperfusion Injury/metabolism , Animals , Carbazoles/pharmacology , Ischemic Postconditioning , Male , Mice , Mice, Inbred C57BL , Models, Animal , NG-Nitroarginine Methyl Ester/pharmacology , Signal Transduction/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL