Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
2.
Biochemistry ; 62(4): 893-898, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36757899

ABSTRACT

Post-translational modification of arginine to citrulline is catalyzed by members of the peptidylarginine deiminase (PAD) family. Dysregulation of this catalysis is a significant driver of the pathogenesis of numerous inflammatory diseases, including cancer. However, dysregulation of PAD activity has not been examined in breast cancer with respect to hormone receptor status. In this study, we measured PAD enzyme levels using Western blotting and investigated protein citrullination using a mass spectrometry-based proteomics approach in primary estrogen receptor negative (ER-) or positive (ER+) breast tumor and matched adjacent normal tissue. Our findings reveal 72 and 41 citrullinated proteins in ER- tumor and adjacent healthy tissue, respectively, where 20 of these proteins are common between the two groups. We detected 64 and 49 citrullinated proteins in ER+ tumor and adjacent healthy tissue, respectively, where 32 proteins are common. Interestingly, upon comparison of ER- and ER+ tumor tissue, only 32 citrullinated proteins are shared between the two and the rest are unique to the tumor's receptor status. Using the STRING database for protein-protein interaction network analysis, these proteins are involved in protein-folding events (i.e., heat shock proteins) in ER- samples and blood-clotting events (i.e., fibulin) in ER+ samples. Constituents of the extracellular matrix structure (i.e., collagen and fibrinogen) were found in both. Herein, we establish evidence that supports the role of this unique post-translational modification in breast cancer biology. Finally, to aid drug discovery against citrullination, we developed a liquid chromatography-ultraviolet method to measure PAD enzymatic activity and optimized glucagon-like peptide II to quantitatively measure the ability of PADs to citrullinate its substrate.


Subject(s)
Breast Neoplasms , Citrullination , Humans , Female , Proteins/metabolism , Protein-Arginine Deiminases/metabolism , Protein Processing, Post-Translational , Citrulline/chemistry , Hydrolases/chemistry
3.
J Biol Chem ; 298(11): 102579, 2022 11.
Article in English | MEDLINE | ID: mdl-36220393

ABSTRACT

Calcium/calmodulin-dependent protein kinase II δ (CaMKIIδ) has a pivotal role in cardiac signaling. Constitutive and deleterious CaMKII "autonomous" activation is induced by oxidative stress, and the previously reported mechanism involves oxidation of methionine residues in the regulatory domain. Here, we demonstrate that covalent oxidation leads to a disulfide bond with Cys273 in the regulatory domain causing autonomous activity. Autonomous activation was induced by treating CaMKII with diamide or histamine chloramine, two thiol-oxidizing agents. Autonomy was reversed when the protein was incubated with DTT or thioredoxin to reduce disulfide bonds. Tryptic mapping of the activated CaMKII revealed formation of a disulfide between Cys273 and Cys290 in the regulatory domain. We determined the apparent pKa of those Cys and found that Cys273 had a low pKa while that of Cys290 was elevated. The low pKa of Cys273 facilitates oxidation of its thiol to the sulfenic acid at physiological pH. The reactive sulfenic acid then attacks the thiol of Cys290 to form the disulfide. The previously reported CaMKII mutant in which methionine residues 281 and 282 were mutated to valine (MMVV) protects mice and flies from cardiac decompensation induced by oxidative stress. Our initial hypothesis was that the MMVV mutant underwent a conformational change that prevented disulfide formation and autonomous activation. However, we found that the thiol-oxidizing agents induced autonomy in the MMVV mutant and that the mutant undergoes rapid degradation by the cell, potentially preventing accumulation of the injurious autonomous form. Together, our results highlight additional mechanistic details of CaMKII autonomous activation.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Calcium , Mice , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Calcium/metabolism , Disulfides/metabolism , Calmodulin/metabolism , Sulfenic Acids , Oxidation-Reduction , Sulfhydryl Compounds , Methionine/metabolism , Oxidants , Oxidative Stress
4.
Hypertension ; 79(8): 1777-1788, 2022 08.
Article in English | MEDLINE | ID: mdl-35766034

ABSTRACT

BACKGROUND: Aortic stiffening is strongly associated with both aging and hypertension, but the underlying mechanisms remain unclear. We hypothesized that aging-induced aortic stiffness is mediated by a mechanism differing from hypertension. METHODS: We conducted comprehensive in vivo and in vitro experiments using multiple rat models to dissect the different mechanisms of aortic stiffening mediated by aging and hypertension. RESULTS: A time-course study in spontaneously hypertensive rats (SHR) and Wistar-Kyoto (WKY) normotensive rats showed more pronounced aging-associated aortic stiffening in SHR versus WKY. Angiotensin II-induced hypertension was associated with more significant aortic stiffening in older versus young WKY rats. Hypertension aggravated aging effects on aortic wall thickness and extracellular matrix content, indicating combinational effects of aging and hypertension on aortic stiffening. Intrinsic stiffness of isolated aortic vascular smooth muscle cells (VSMCs) increased with age in WKY rats, although no significant difference between older SHR and older WKY VSMCs was observed in 2-dimensional culture, reconstituted 3-dimensional tissues were stiffer for older SHR versus older WKY. A selective inhibitor that reduced hypertension-mediated aortic stiffening did not decrease age-related stiffening in aortic VSMCs and aortic wall. Integrin ß1 and SM22 (smooth muscle-specific SM22 protein) expression were negligibly changed in WKY VSMCs during aging but were markedly increased by hypertension in older versus young WKY VSMCs. A notable shift of filamin isoforms from B to A was detected in older WKY VSMCs. CONCLUSIONS: Our results indicate distinct mechanisms mediating aging-associated aortic VSMC and vessel stiffness, providing new insights into aortic stiffening and the pathogenesis of hypertension in the elderly.


Subject(s)
Hypertension , Muscle, Smooth, Vascular , Animals , Blood Pressure/physiology , Cells, Cultured , Muscle, Smooth, Vascular/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY
5.
Cells ; 10(11)2021 10 26.
Article in English | MEDLINE | ID: mdl-34831118

ABSTRACT

Valosin-containing protein (VCP) was found to play a vital protective role against cardiac stresses. Genetic mutations of VCP are associated with human dilated cardiomyopathy. However, the essential role of VCP in the heart during the physiological condition remains unknown since the VCP knockout in mice is embryonically lethal. We generated a cardiac-specific dominant-negative VCP transgenic (DN-VCP TG) mouse to determine the effects of impaired VCP activity on the heart. Using echocardiography, we showed that cardiac-specific overexpression of DN-VCP induced a remarkable cardiac dilation and progressively declined cardiac function during the aging transition. Mechanistically, DN-VCP did not affect the endogenous VCP (EN-VCP) expression but significantly reduced cardiac ATPase activity in the DN-VCP TG mouse hearts, indicating a functional inhibition. DN-VCP significantly impaired the aging-related cytoplasmic/nuclear shuffling of EN-VCP and its co-factors in the heart tissues and interrupted the balance of the VCP-cofactors interaction between the activating co-factors, ubiquitin fusion degradation protein 1 (UFD-1)/nuclear protein localization protein 4 (NPL-4) complex, and its inhibiting co-factor P47, leading to the binding preference with the inhibitory co-factor, resulting in functional repression of VCP. This DN-VCP TG mouse provides a unique functional-inactivation model for investigating VCP in the heart in physiological and pathological conditions.


Subject(s)
Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Dilated/physiopathology , Valosin Containing Protein/antagonists & inhibitors , Aging/pathology , Animals , Cell Nucleus/metabolism , Disease Models, Animal , Mice, Transgenic , Myocardium/pathology , Repressor Proteins/metabolism , Valosin Containing Protein/metabolism
6.
Antioxidants (Basel) ; 10(10)2021 Sep 29.
Article in English | MEDLINE | ID: mdl-34679684

ABSTRACT

Heat shock protein 22 (Hsp22) is a small heat shock protein predominantly expressed in skeletal and cardiac muscle. Previous studies indicate that Hsp22 plays a vital role in protecting the heart against cardiac stress. However, the essential role of Hsp22 in the heart under physiological conditions remains largely unknown. In this study, we used an Hsp22 knockout (KO) mouse model to determine whether loss of Hsp22 impairs cardiac growth and function with increasing age under physiological conditions. Cardiac structural and functional alterations at baseline were measured using echocardiography and invasive catheterization in Hsp22 KO mice during aging transition compared to their age-matched wild-type (WT) littermates. Our results showed that Hsp22 deletion induced progressive cardiac dilation along with declined function during the aging transition. Mechanistically, the loss of Hsp22 impaired BCL-2-associated athanogene 3 (BAG3) expression and its associated cardiac autophagy, undermined cardiac energy metabolism homeostasis and increased oxidative damage. This study showed that Hsp22 played an essential role in the non-stressed heart during the early stage of aging, which may bring new insight into understanding the pathogenesis of age-related dilated cardiomyopathy.

7.
J Cardiovasc Transl Res ; 14(2): 371-376, 2021 04.
Article in English | MEDLINE | ID: mdl-32748205

ABSTRACT

Using electronic cigarette (e-cig) among youth is becoming a critical public health crisis in the USA. However, the biological impacts of the e-cig on multiple organ systems, especially in the cardiovascular system, are largely unknown. Unlike conventional tobacco, e-cig combines various chemical ingredients including nicotine and other add-on non-nicotine chemicals, such as the solvents (propylene glycol and/or vegetable glycerin) and flavoring chemicals, which dramatically increases the diversity of the potential implications. The recent outbreak of e-cig vaping-related tragic deaths in youth and multiple hospitalized patients raised a question on the safety of e-cig use and led to an urgent need for the knowledge of the health risk of the e-cig compositions. Therefore, in the review, we summarized the latest findings from both human and animal studies on the potential cardiovascular toxicological effects of e-cig on the cardiovascular system in terms of the systemic physiological implications and the cellular and molecular mechanisms involved.


Subject(s)
Cardiovascular Diseases/chemically induced , Cardiovascular System/drug effects , E-Cigarette Vapor/adverse effects , Electronic Nicotine Delivery Systems , Flavoring Agents/adverse effects , Nicotine/adverse effects , Nicotinic Agonists/adverse effects , Solvents/adverse effects , Vaping/adverse effects , Animals , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cardiovascular Diseases/physiopathology , Cardiovascular System/metabolism , Cardiovascular System/pathology , Cardiovascular System/physiopathology , Humans , Risk Assessment
8.
Int J Mol Sci ; 21(22)2020 Nov 12.
Article in English | MEDLINE | ID: mdl-33198081

ABSTRACT

The mammalian cell cycle is important in controlling normal cell proliferation and the development of various diseases. Cell cycle checkpoints are well regulated by both activators and inhibitors to avoid cell growth disorder and cancerogenesis. Cyclin dependent kinase 20 (CDK20) and p21Cip1/Waf1 are widely recognized as key regulators of cell cycle checkpoints controlling cell proliferation/growth and involving in developing multiple cancers. Emerging evidence demonstrates that these two cell cycle regulators also play an essential role in promoting cell survival independent of the cell cycle, particularly in those cells with a limited capability of proliferation, such as cardiomyocytes. These findings bring new insights into understanding cytoprotection in these tissues. Here, we summarize the new progress of the studies on these two molecules in regulating cell cycle/growth, and their new roles in cell survival by inhibiting various cell death mechanisms. We also outline their potential implications in cancerogenesis and protection in heart diseases. This information renews the knowledge in molecular natures and cellular functions of these regulators, leading to a better understanding of the pathogenesis of the associated diseases and the discovery of new therapeutic strategies.


Subject(s)
Cell Cycle/physiology , Cell Survival/physiology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinases/metabolism , Animals , Cell Death/physiology , Cell Proliferation/physiology , Humans
9.
Int J Mol Sci ; 21(20)2020 Oct 17.
Article in English | MEDLINE | ID: mdl-33080805

ABSTRACT

Calcium ion (Ca2+) plays a critical role in the cardiac mitochondria function. Ca2+ entering the mitochondria is necessary for ATP production and the contractile activity of cardiomyocytes. However, excessive Ca2+ in the mitochondria results in mitochondrial dysfunction and cell death. Mitochondria maintain Ca2+ homeostasis in normal cardiomyocytes through a comprehensive regulatory mechanism by controlling the uptake and release of Ca2+ in response to the cellular demand. Understanding the mechanism of modulating mitochondrial Ca2+ homeostasis in the cardiomyocyte could bring new insights into the pathogenesis of cardiac disease and help developing the strategy to prevent the heart from damage at an early stage. In this review, we summarized the latest findings in the studies on the cardiac mitochondrial Ca2+ homeostasis, focusing on the regulation of mitochondrial calcium uptake, which acts as a double-edged sword in the cardiac function. Specifically, we discussed the dual roles of mitochondrial Ca2+ in mitochondrial activity and the impact on cardiac function, the molecular basis and regulatory mechanisms, and the potential future research interest.


Subject(s)
Calcium Signaling , Heart/physiology , Mitochondria, Heart/metabolism , Myocardium/metabolism , Animals , Calcium/metabolism , Heart/physiopathology , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology
10.
J Clin Invest ; 130(9): 4663-4678, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32749237

ABSTRACT

Oxidant stress can contribute to health and disease. Here we show that invertebrates and vertebrates share a common stereospecific redox pathway that protects against pathological responses to stress, at the cost of reduced physiological performance, by constraining Ca2+/calmodulin-dependent protein kinase II (CaMKII) activity. MICAL1, a methionine monooxygenase thought to exclusively target actin, and MSRB, a methionine reductase, control the stereospecific redox status of M308, a highly conserved residue in the calmodulin-binding (CaM-binding) domain of CaMKII. Oxidized or mutant M308 (M308V) decreased CaM binding and CaMKII activity, while absence of MICAL1 in mice caused cardiac arrhythmias and premature death due to CaMKII hyperactivation. Mimicking the effects of M308 oxidation decreased fight-or-flight responses in mice, strikingly impaired heart function in Drosophila melanogaster, and caused disease protection in human induced pluripotent stem cell-derived cardiomyocytes with catecholaminergic polymorphic ventricular tachycardia, a CaMKII-sensitive genetic arrhythmia syndrome. Our studies identify a stereospecific redox pathway that regulates cardiac physiological and pathological responses to stress across species.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Drosophila Proteins/metabolism , Microfilament Proteins/metabolism , Mixed Function Oxygenases/metabolism , Mutation, Missense , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Tachycardia, Ventricular/enzymology , Amino Acid Substitution , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster , Humans , Mice , Mice, Knockout , Microfilament Proteins/genetics , Mixed Function Oxygenases/genetics , Myocardium/pathology , Myocytes, Cardiac/pathology , Oxidation-Reduction , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/pathology
11.
Redox Biol ; 34: 101555, 2020 07.
Article in English | MEDLINE | ID: mdl-32388268

ABSTRACT

the acute and chronic myocardial ischemia results in oxidative stress that impairs myocardial contractility and eventually leads to heart failure. However, the underlying regulatory molecular mechanisms are not fully understood. The heat shock protein 22 (Hsp22), a small-molecular-weight protein preferentially expressed in the heart, was found to be dramatically increased in the cardiac oxidative stress conditions in both human and animal models after the acute and chronic ischemia. Overexpression of Hsp22 largely protects the heart against ischemic damage. Mechanistically, overexpression of Hsp22 attenuates hypoxia-induced oxidative phosphorylation in mitochondrial and the high rate of superoxide production. Short term gene delivery of Hsp22 reduces the infarct size caused by the ischemia/reperfusion, providing a clinical therapeutic potential. This review discusses the new progress of the studies on Hsp22 by focusing on its protective effect against the excessive cardiac oxidative stress, including its adaptive induction in myocardium upon the oxidative stress, its protective role in myocardial ischemia/reperfusion, its regulation in mitochondrial oxidative phosphorylation and the underlying molecular signaling pathways promoting cell survival. This information will increase our understanding of the molecular regulation of cardiac adaption under the oxidative stress and the potential therapeutic relevance.


Subject(s)
Heat-Shock Proteins , Oxidative Stress , Animals , Heart , Heat-Shock Proteins/metabolism , Humans , Ischemia/metabolism , Molecular Chaperones , Myocardium/metabolism
12.
Free Radic Biol Med ; 145: 374-384, 2019 12.
Article in English | MEDLINE | ID: mdl-31606431

ABSTRACT

Oxidation of methionine residues to methionine sulfoxide scavenges reactive species, thus protecting against oxidative stress. Reduction of the sulfoxide back to methionine by methionine sulfoxide reductases creates a cycle with catalytic efficiency. Protection by the methionine sulfoxide reductases is well documented in cultured cells, from microorganisms to mammals. However, knocking out one or two of the 4 mammalian reductases had little effect in mice that were not stressed. We hypothesized that the minimal effect is due to redundancy provided by the 4 reductases. We tested the hypothesis by creating a transgenic mouse line lacking all 4 reductases and predicted that this mouse would be exceptionally sensitive to oxidative stress. The mutant mice were phenotypically normal at birth, exhibited normal post-natal growth, and were fertile. Surprisingly, rather than being more sensitive to oxidative stress, they were more resistant to both cardiac ischemia-reperfusion injury and to parenteral paraquat, a redox-cycling agent. Resistance was not a result of hormetic induction of the antioxidant transcription factor Nrf2 nor activation of Akt. The mechanism of protection may be novel.


Subject(s)
Methionine Sulfoxide Reductases/genetics , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Reperfusion Injury/drug therapy , Animals , Catalysis , Methionine/analogs & derivatives , Methionine/genetics , Methionine/metabolism , Methionine Sulfoxide Reductases/metabolism , Mice, Transgenic/genetics , Oxidation-Reduction/drug effects , Paraquat/pharmacology , Reperfusion Injury/genetics , Reperfusion Injury/pathology , Stress, Physiological/genetics
13.
Pharm Res ; 36(7): 103, 2019 May 17.
Article in English | MEDLINE | ID: mdl-31101998

ABSTRACT

PURPOSE: A rapid and broadly applicable method to assess relevant oxidative damage in biopharmaceuticals is important for lifecycle management of product quality. Multiple methods are currently employed as stress tests to induce oxidative damage for assessment of stability, safety, and efficacy. We compared two common methods for inducing oxidative damage to assess differences in impact on bioactivity and structure of the biopharmaceuticals. METHODS: Biopharmaceuticals were treated with either metal-catalyzed oxidation (MCO) conditions or the reactive-oxygen species (ROS) inducer 2,2'-Azobis(2-amidinopropane) dihydrochloride (AAPH), then analyzed for changes in structure and bioactivity. RESULTS: We demonstrate that commonly used chemical methods for assessing oxidation yield distinct oxidation profiles for each of the biotechnology products analyzed, including monoclonal antibodies. We further report oxidant- and product-specific changes in bioactivity under oxidizing conditions, along with differential oxidation on the molecular subunits of monoclonal antibodies. CONCLUSION: Our results highlight the need for product-specific optimization and selection of orthogonal, relevant oxidizers when characterizing stress responses in biopharmaceuticals.


Subject(s)
Biological Products/chemistry , Oxidative Stress , Antibodies, Monoclonal, Humanized/chemistry , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/chemistry , Antineoplastic Agents, Immunological/pharmacology , Biological Products/pharmacology , Cell Line, Tumor , Drug Stability , Humans , Indicators and Reagents/chemistry , Methionine/chemistry , Oxidants/chemistry , Oxidation-Reduction , Reactive Oxygen Species/chemistry , Rituximab/chemistry , Rituximab/pharmacology , Trastuzumab/chemistry , Trastuzumab/pharmacology
14.
Aging Cell ; 14(6): 1075-84, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26424149

ABSTRACT

The most important physiological mechanism mediating enhanced exercise performance is increased sympathetic, beta adrenergic receptor (ß-AR), and adenylyl cyclase (AC) activity. This is the first report of decreased AC activity mediating increased exercise performance. We demonstrated that AC5 disruption, that is, knock out (KO) mice, a longevity model, increases exercise performance. Importantly for its relation to longevity, exercise was also improved in old AC5 KO. The mechanism resided in skeletal muscle rather than in the heart, as confirmed by cardiac- and skeletal muscle-specific AC5 KO's, where exercise performance was no longer improved by the cardiac-specific AC5 KO, but was by the skeletal muscle-specific AC5 KO, and there was no difference in cardiac output during exercise in AC5 KO vs. WT. Mitochondrial biogenesis was a major mechanism mediating the enhanced exercise. SIRT1, FoxO3a, MEK, and the anti-oxidant, MnSOD were upregulated in AC5 KO mice. The improved exercise in the AC5 KO was blocked with either a SIRT1 inhibitor, MEK inhibitor, or by mating the AC5 KO with MnSOD hetero KO mice, confirming the role of SIRT1, MEK, and oxidative stress mechanisms. The Caenorhabditis elegans worm AC5 ortholog, acy-3 by RNAi, also improved fitness, mitochondrial function, antioxidant defense, and lifespan, attesting to the evolutionary conservation of this pathway. Thus, decreasing sympathetic signaling through loss of AC5 is not only a mechanism to improve exercise performance, but is also a mechanism to improve healthful aging, as exercise also protects against diabetes, obesity, and cardiovascular disease, which all limit healthful aging.


Subject(s)
Adenylyl Cyclases/genetics , Caenorhabditis elegans/metabolism , Cardiac Output/genetics , Mitochondria/metabolism , Muscle, Skeletal/metabolism , Physical Conditioning, Animal , Adenylyl Cyclases/metabolism , Animals , Caenorhabditis elegans/genetics , Cell Respiration , Forkhead Box Protein O3 , Forkhead Transcription Factors/biosynthesis , Heart/physiology , Longevity/genetics , MAP Kinase Signaling System , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Mitogen-Activated Protein Kinase Kinases/biosynthesis , RNA Interference , RNA, Small Interfering/genetics , Sirtuin 1/antagonists & inhibitors , Sirtuin 1/biosynthesis , Superoxide Dismutase/biosynthesis , Up-Regulation
15.
Am J Physiol Heart Circ Physiol ; 305(1): H1-8, 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23624627

ABSTRACT

G protein-coupled receptor/adenylyl cyclase (AC)/cAMP signaling is crucial for all cellular responses to physiological and pathophysiological stimuli. There are nine isoforms of membrane-bound AC, with type 5 being one of the two major isoforms in the heart. Since the role of AC in the heart in regulating cAMP and acute changes in inotropic and chronotropic state are well known, this review will address our current understanding of the distinct regulatory role of the AC5 isoform in response to chronic stress. Transgenic overexpression of AC5 in cardiomyocytes of the heart (AC5-Tg) improves baseline cardiac function but impairs the ability of the heart to withstand stress. For example, chronic catecholamine stimulation induces cardiomyopathy, which is more severe in AC5-Tg mice, mediated through the AC5/sirtuin 1/forkhead box O3a pathway. Conversely, disrupting AC5, i.e., AC5 knockout, protects the heart from chronic catecholamine cardiomyopathy as well as the cardiomyopathies resulting from chronic pressure overload or aging. Moreover, AC5 knockout results in a 30% increase in a healthy life span, resembling the most widely studied model of longevity, i.e., calorie restriction. These two models of longevity share similar gene regulation in the heart, muscle, liver, and brain in that they are both protected against diabetes, obesity, and diabetic and aging cardiomyopathy. A pharmacological inhibitor of AC5 also provides protection against cardiac stress, diabetes, and obesity. Thus AC5 inhibition has novel, potential therapeutic applicability to several diseases not only in the heart but also in aging, diabetes, and obesity.


Subject(s)
Adenylyl Cyclases/metabolism , Cardiomyopathies/enzymology , Longevity/genetics , Adenylyl Cyclase Inhibitors , Adenylyl Cyclases/genetics , Animals , Cardiomyopathies/drug therapy , Cardiomyopathies/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Longevity/drug effects
16.
Circulation ; 127(16): 1692-701, 2013 Apr 23.
Article in English | MEDLINE | ID: mdl-23536361

ABSTRACT

BACKGROUND: For reasons that remain unclear, whether type 5 adenylyl cyclase (AC5), 1 of 2 major AC isoforms in heart, is protective or deleterious in response to cardiac stress is controversial. To reconcile this controversy we examined the cardiomyopathy induced by chronic isoproterenol in AC5 transgenic (Tg) mice and the signaling mechanisms involved. METHODS AND RESULTS: Chronic isoproterenol increased oxidative stress and induced more severe cardiomyopathy in AC5 Tg, as left ventricular ejection fraction fell 1.9-fold more than wild type, along with greater left ventricular dilation and increased fibrosis, apoptosis, and hypertrophy. Oxidative stress induced by chronic isoproterenol, detected by 8-OhDG was 15% greater, P=0.007, in AC5 Tg hearts, whereas protein expression of manganese superoxide dismutase (MnSOD) was reduced by 38%, indicating that the susceptibility of AC5 Tg to cardiomyopathy may be attributable to decreased MnSOD expression. Consistent with this, susceptibility of the AC5 Tg to cardiomyopathy was suppressed by overexpression of MnSOD, whereas protection afforded by the AC5 knockout (KO) was lost in AC5 KO×MnSOD heterozyous KO mice. Elevation of MnSOD was eliminated by both sirtuin and MEK inhibitors, suggesting both the SIRT1/FoxO3a and MEK/ERK pathway are involved in MnSOD regulation by AC5. CONCLUSIONS: Overexpression of AC5 exacerbates the cardiomyopathy induced by chronic catecholamine stress by altering regulation of SIRT1/FoxO3a, MEK/ERK, and MnSOD, resulting in oxidative stress intolerance, thereby shedding light on new approaches for treatment of heart failure.


Subject(s)
Adenylyl Cyclases/physiology , Cardiomyopathies/physiopathology , Forkhead Transcription Factors/physiology , MAP Kinase Signaling System/physiology , Oxidative Stress/physiology , Sirtuin 1/physiology , Superoxide Dismutase/physiology , Adenylyl Cyclases/deficiency , Adenylyl Cyclases/genetics , Animals , Cardiomyopathies/chemically induced , Crosses, Genetic , Cyclic N-Oxides/therapeutic use , Enzyme Induction/physiology , Forkhead Box Protein O3 , Isoproterenol/toxicity , MAP Kinase Signaling System/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Oxidative Stress/genetics , Protein Kinase Inhibitors/pharmacology , Sirtuin 1/antagonists & inhibitors , Spin Labels , Superoxide Dismutase/biosynthesis , Superoxide Dismutase/genetics , Transcription, Genetic
17.
Age (Dordr) ; 35(6): 2177-82, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23334601

ABSTRACT

Calorie restriction (CR) is the most widely studied intervention protecting from the adverse effects of aging. Almost all prior studies have examined the effects of CR initiated in young animals. Studies examining the effects of CR on development of aging cardiomyopathy found only partial prevention. The major goal of this study was to determine whether CR initiated after aging cardiomyopathy developed could reverse the cardiomyopathy. Aging cardiomyopathy in 2-year-old mice was characterized by reduced left ventricular (LV) function, cardiac hypertrophy, and increased cardiac apoptosis and fibrosis. When short-term (2 months) CR was initiated after aging cardiomyopathy developed in 20-month-old mice, the decrease in cardiac function, and increases in LV weight, myocardial fibrosis and apoptosis were reversed, such that the aging hearts in these mice were indistinguishable from those of young mice or mice where CR was initiated in young mice. If apoptosis was the mechanism for protecting against aging cardiomyopathy, then total myocyte numbers should have reverted to normal with CR, but did not. However, the alterations in cytoskeletal proteins, which contribute to aging cardiomyopathy, were no longer observed with CR. This is the first study to demonstrate complete prevention of aging cardiomyopathy by CR and, more importantly, that instituting this intervention even later in life can rapidly correct aging cardiomyopathy, which could have important therapeutic implications.


Subject(s)
Aging/physiology , Caloric Restriction , Cardiomyopathies/prevention & control , Recovery of Function , Animals , Cardiomyopathies/physiopathology , Mice , Prognosis
18.
Aging Cell ; 11(6): 1110-20, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23020244

ABSTRACT

Adenylyl cyclase type 5 knockout mice (AC5 KO) live longer and are stress resistant, similar to calorie restriction (CR). AC5 KO mice eat more, but actually weigh less and accumulate less fat compared with WT mice. CR applied to AC5 KO results in rapid decrease in body weight, metabolic deterioration, and death. These data suggest that despite restricted food intake in CR, but augmented food intake in AC5 KO, the two models affect longevity and metabolism similarly. To determine shared molecular mechanisms, mRNA expression was examined genome-wide for brain, heart, skeletal muscle, and liver. Significantly more genes were regulated commonly rather than oppositely in all the tissues in both models, indicating commonality between AC5 KO and CR. Gene ontology analysis identified many significantly regulated, tissue-specific pathways shared by the two models, including sensory perception in heart and brain, muscle function in skeletal muscle, and lipid metabolism in liver. Moreover, when comparing gene expression changes in the heart under stress, the glutathione regulatory pathway was consistently upregulated in the longevity models but downregulated with stress. In addition, AC5 and CR shared changes in genes and proteins involved in the regulation of longevity and stress resistance, including Sirt1, ApoD, and olfactory receptors in both young- and intermediate-age mice. Thus, the similarly regulated genes and pathways in AC5 KO and CR mice, particularly related to the metabolic phenotype, suggest a unified theory for longevity and stress resistance.


Subject(s)
Adenylyl Cyclases/genetics , Caloric Restriction , Gene Expression Regulation , Longevity/genetics , Stress, Physiological/genetics , Adenylyl Cyclases/deficiency , Animals , Apolipoproteins D/genetics , Apolipoproteins D/metabolism , Body Weight , Brain/metabolism , Gene Expression Profiling , Insulin Resistance/genetics , Liver/metabolism , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , Myocardium/metabolism , Receptors, Odorant/genetics , Receptors, Odorant/metabolism , Sirtuin 1/genetics , Sirtuin 1/metabolism
19.
Am J Physiol Heart Circ Physiol ; 302(12): H2622-8, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22505646

ABSTRACT

Despite numerous discoveries from genetically engineered mice, relatively few have been translated to the bedside, mainly because it is difficult to translate from genes to drugs. This investigation examines an antiviral drug, which also has an action to selectively inhibit type 5 adenylyl cyclase (AC5), a pharmaceutical correlate of the AC5 knockout (KO) model, which exhibits longevity and stress resistance. Our objective was to examine the extent to which pretreatment with this drug, adenine 9-ß-d-arabinofuranoside (Ara-A), favorably ameliorates the development of heart failure (HF). Ara-A exhibited selective inhibition for AC5 compared with the other major cardiac AC isoform, AC6, i.e., it reduced AC activity significantly in AC5 transgenic (Tg) mice, but not in AC5KO mice and had little effect in either wild-type or AC6Tg mice. Permanent coronary artery occlusion for 3 wk in C57Bl/6 mice increased mortality and induced HF in survivors, as reflected by reduced cardiac function, while increasing cardiac fibrosis. The AC5 inhibitor Ara-A significantly improved all of these end points and also ameliorated chronic isoproterenol-induced cardiomyopathy. As with the AC5KO mice, Ara-A increased mitogen/extracellular signal-regulated kinase (MEK)/extracellular signal-regulated kinase (ERK) phosphorylation. A MEK inhibitor abolished the beneficial effects of the AC5 inhibitor in the HF model, indicating the involvement of the downstream MEK-ERK pathway of AC5. Our data suggest that pharmacological AC5 inhibition may serve as a new therapeutic approach for HF.


Subject(s)
Adenylyl Cyclase Inhibitors , Antiviral Agents/therapeutic use , Heart Failure/prevention & control , Heart/drug effects , Vidarabine/therapeutic use , Adenylyl Cyclases/metabolism , Animals , Antiviral Agents/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/physiopathology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Transgenic , Myocardium/metabolism , Vidarabine/pharmacology
20.
Dalton Trans ; (11): 1412-4, 2008 Mar 21.
Article in English | MEDLINE | ID: mdl-18322617

ABSTRACT

Novel phosphorescent conjugated oligo(phenylene-ethynylene)s featuring a central tunable platinum(II) Schiff base signalling unit with promising photophysical properties have been investigated to pave their development towards polymeric congeners for sensing applications.


Subject(s)
Luminescent Agents/chemistry , Organoplatinum Compounds/chemistry , Polymers/chemistry , Schiff Bases/chemistry , Luminescent Agents/chemical synthesis , Luminescent Measurements , Molecular Structure , Organoplatinum Compounds/chemical synthesis , Polymers/chemical synthesis , Schiff Bases/chemical synthesis , Spectrophotometry , Spectrophotometry, Ultraviolet
SELECTION OF CITATIONS
SEARCH DETAIL
...