Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Physiol Genomics ; 48(4): 274-80, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26757797

ABSTRACT

Cardiomyopathy (CM) is an intrinsic weakening of myocardium with contractile dysfunction and congestive heart failure (CHF). CHF has been postulated to result from decreased mitochondrial energy production and oxidative stress. Effects of decreased mitochondrial oxygen consumption also can accelerate with aging. We previously showed DNA methylation changes in human hearts with CM. This was associated with mitochondrial DNA depletion, being another molecular marker of CM. We examined the relationship between mitochondrial dysfunction and cardiac epigenetic DNA methylation changes in both young and old mice. We used genetically engineered C57Bl/6 mice transgenic for a cardiac-specific mutant of the mitochondrial polymerase-γ (termed Y955C). Y955C mice undergo left ventricular hypertrophy (LVH) at a young age (∼ 94 days old), and LVH decompensated to CHF at old age (∼ 255 days old). Results found 95 genes differentially expressed as a result of Y955C expression, while 4,452 genes were differentially expressed as a result of aging hearts. Moreover, cardiac DNA methylation patterns differed between Y955C (4,506 peaks with 68.5% hypomethylation) and aged hearts (73,286 peaks with 80.2% hypomethylated). Correlatively, of the 95 Y955C-dependent differentially expressed genes, 30 genes (31.6%) also displayed differential DNA methylation; in the 4,452 age-dependent differentially expressed genes, 342 genes (7.7%) displayed associated DNA methylation changes. Both Y955C and aging demonstrated significant enrichment of CACGTG-associated E-box motifs in differentially methylated regions. Cardiac mitochondrial polymerase dysfunction alters nuclear DNA methylation. Furthermore, aging causes a robust change in cardiac DNA methylation that is partially associated with mitochondrial polymerase dysfunction.


Subject(s)
Aging/genetics , DNA Methylation , DNA-Directed DNA Polymerase/metabolism , Heart/physiopathology , Animals , DNA Polymerase gamma , DNA, Mitochondrial , DNA-Directed DNA Polymerase/genetics , Electrocardiography , Female , Gene Expression Profiling , Heart Failure/genetics , Hypertrophy, Left Ventricular/genetics , Male , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria, Heart/genetics , Mitochondria, Heart/physiology
2.
Toxicol Appl Pharmacol ; 288(3): 409-19, 2015 Nov 01.
Article in English | MEDLINE | ID: mdl-26307267

ABSTRACT

This study addresses the individual and combined effects of HIV-1 and methamphetamine (N-methyl-1-phenylpropan-2-amine, METH) on cardiac dysfunction in a transgenic mouse model of HIV/AIDS. METH is abused epidemically and is frequently associated with acquisition of HIV-1 infection or AIDS. We employed microarrays to identify mRNA differences in cardiac left ventricle (LV) gene expression following METH administration (10d, 3mg/kg/d, subcutaneously) in C57Bl/6 wild-type littermates (WT) and Tat-expressing transgenic (TG) mice. Arrays identified 880 differentially expressed genes (expression fold change>1.5, p<0.05) following METH exposure, Tat expression, or both. Using pathway enrichment analysis, mRNAs encoding polypeptides for calcium signaling and contractility were altered in the LV samples. Correlative DNA methylation analysis revealed significant LV DNA methylation changes following METH exposure and Tat expression. By combining these data sets, 38 gene promoters (27 related to METH, 11 related to Tat) exhibited differences by both methods of analysis. Among those, only the promoter for CACNA1C that encodes L-type calcium channel Cav1.2 displayed DNA methylation changes concordant with its gene expression change. Quantitative PCR verified that Cav1.2 LV mRNA abundance doubled following METH. Correlative immunoblots specific for Cav1.2 revealed a 3.5-fold increase in protein abundance in METH LVs. Data implicate Cav1.2 in calcium dysregulation and hypercontractility in the murine LV exposed to METH. They suggest a pathogenetic role for METH exposure to promote LV dysfunction that outweighs Tat-induced effects.


Subject(s)
DNA Methylation/drug effects , HIV Infections/physiopathology , Heart/drug effects , Methamphetamine/pharmacology , tat Gene Products, Human Immunodeficiency Virus/genetics , Animals , Calcium Channels, L-Type/genetics , Calcium Channels, L-Type/metabolism , Dose-Response Relationship, Drug , Electrocardiography , Epigenesis, Genetic , Gene Expression/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondria/drug effects , Promoter Regions, Genetic , Up-Regulation , tat Gene Products, Human Immunodeficiency Virus/metabolism
3.
Toxicol Sci ; 148(1): 183-91, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26251327

ABSTRACT

MDMA (ecstasy) is an illicit drug that stimulates monoamine neurotransmitter release and inhibits reuptake. MDMA's acute cardiotoxicity includes tachycardia and arrhythmia which are associated with cardiomyopathy. MDMA acute cardiotoxicity has been explored, but neither long-term MDMA cardiac pathological changes nor epigenetic changes have been evaluated. Microarray analyses were employed to identify cardiac gene expression changes and epigenetic DNA methylation changes. To identify permanent MDMA-induced pathogenetic changes, mice received daily 10- or 35-day MDMA, or daily 10-day MDMA followed by 25-day saline washout (10 + 25 days). MDMA treatment caused differential gene expression (p < .05, fold change >1.5) in 752 genes following 10 days, 558 genes following 35 days, and 113 genes following 10-day MDMA + 25-day saline washout. Changes in MAPK and circadian rhythm gene expression were identified as early as 10 days. After 35 days, circadian rhythm genes (Per3, CLOCK, ARNTL, and NPAS2) persisted to be differentially expressed. MDMA caused DNA hypermethylation and hypomethylation that was independent of gene expression; hypermethylation of genes was found to be 71% at 10 days, 68% at 35 days, and 91% at 10 + 25 days washout. Differential gene expression paralleled DNA methylation in 22% of genes at 10-day treatment, 17% at 35 days, and 48% at 10 + 25 days washout. We show here that MDMA induced cardiac epigenetic changes in DNA methylation where hypermethylation predominated. Moreover, MDMA induced gene expression of key elements of circadian rhythm regulatory genes. This suggests a fundamental organism-level event to explain some of the etiologies of MDMA dysfunction in the heart.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiomyopathies/chemically induced , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Hallucinogens/toxicity , Heart/drug effects , N-Methyl-3,4-methylenedioxyamphetamine/toxicity , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cardiotoxins/toxicity , Circadian Rhythm Signaling Peptides and Proteins/agonists , Circadian Rhythm Signaling Peptides and Proteins/antagonists & inhibitors , Circadian Rhythm Signaling Peptides and Proteins/genetics , Circadian Rhythm Signaling Peptides and Proteins/metabolism , Dose-Response Relationship, Drug , Epigenesis, Genetic/drug effects , Female , Gene Expression Profiling , Heart/physiopathology , MAP Kinase Signaling System/drug effects , Male , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Survival Analysis , Tachycardia/etiology
4.
Physiol Genomics ; 47(10): 447-54, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26199398

ABSTRACT

Mitochondrial dysfunction causes oxidative stress and cardiomyopathy. Oxidative stress also is a side effect of dideoxynucleoside antiretrovirals (NRTI) and is observed in NRTI-induced cardiomyopathy. We show here that treatment with the NRTI AZT {1-[(2R,4S,5S)-4-azido-5-(hydroxymethyl)oxolan-2-yl]-5-methylpyrimidine-2,4-dione} modulates cardiac gene expression epigenetically through production of mitochondrially derived reactive oxygen species. Transgenic mice with ubiquitous expression of mitochondrially targeted catalase (MCAT) and C57Bl/6 wild-type mice littermates (WT) were administered AZT (0.22 mg/day po, 35 days), and cardiac DNA and mRNA were isolated. In AZT-treated WT, 95 cardiac genes were differentially expressed compared with vehicle-treated WTs. When MCAT mice were treated with AZT, each of those 95 genes reverted toward the expression of vehicle-treated WTs. In AZT-treated WT hearts, Mthfr [5,10-methylenetetrahydrofolate reductase; a critical enzyme in synthesis of methionine cycle intermediates including S-adenosylmethionine (SAM)], was overexpressed. Steady-state abundance of SAM in cardiac extracts from AZT-treated MCAT mice increased 60% above that of vehicle-treated MCAT. No such change occurred in WT. AZT caused hypermethylation (47%) and hypomethylation (53%) of differentially methylated DNA regions in WT cardiac DNA. AZT-treated MCAT heart DNA exhibited greater hypermethylation (91%) and less hypomethylation (9%) compared with vehicle-treated MCAT controls. The gene encoding protein kinase C-α displayed multifocal epigenetic regulation caused by oxidative stress. Results show that mitochondrially derived oxidative stress in the heart hinders cardiac DNA methylation, alters steady-state abundance of SAM, alters cardiac gene expression, and promotes characteristic pathophysiological changes of cardiomyopathy. This mechanism for NRTI toxicity offers insight into long-term side effects from these commonly used antiviral agents.


Subject(s)
Epigenesis, Genetic/drug effects , Mitochondria/metabolism , Oxidative Stress/drug effects , Zidovudine/pharmacology , Animals , Catalase/metabolism , Cluster Analysis , DNA Methylation/drug effects , DNA Methylation/genetics , Gene Expression Profiling , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Mice, Inbred C57BL , Mitochondria/drug effects , Oligonucleotide Array Sequence Analysis , Oxidative Stress/genetics , S-Adenosylmethionine/pharmacology
5.
Lab Invest ; 93(2): 151-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23090637

ABSTRACT

Mitochondria produce the energy required for proper cardiac contractile function, and cardiomyocytes that exhibit reduced mitochondrial electron transport will have reduced energy production and decreased contractility. Mitochondrial DNA (mtDNA) encodes the core subunits for the protein complexes of the electron transport chain (ETC). Reduced mtDNA abundance has been linked to reduced ETC and the development of heart failure in genetically engineered mice and in human diseases. Nucleoside reverse-transcriptase inhibitors for HIV/AIDS are used in antiretroviral regimens, which cause decreased mtDNA abundance by inhibiting the mitochondrial polymerase, pol-γ, as a limiting side effect. We explored consequences of AZT (1-[(2R,4S,5S)-4-azido-5-(hydroxymethyl)oxolan-2-yl]-5-methylpyrimidine-2,4-dione) exposure on mtDNA abundance in an established transgenic mouse model (TG) in which a cardiac-targeted mutant form of pol-γ displays a dilated cardiomyopathy (DCM) phenotype with increased left ventricle (LV)-mass and increased LV-end diastolic dimension. TG and wild-type littermate mice received 0.22 mg per day AZT or vehicle for 35 days, and were subsequently analyzed for physiological, histological, and molecular changes. After 35 days, Y955C TGs exhibited cardiac fibrosis independent of AZT. Reduced mtDNA abundance was observed in the Y955C mouse; AZT treatment had no effect on the depletion, suggesting that Y955C was sufficient to reduce mtDNA abundance maximally. Isolated mitochondria from AZT-treated Y955C hearts displayed reduced mitochondrial energetic function by oximetric measurement. AZT treatment of the Y955C mutation further reduced basal mitochondrial respiration and state IV(0) respiration. Together, these results demonstrate that defective pol-γ function promotes cardiomyopathy, cardiac fibrosis, mtDNA depletion, and reduced mitochondrial energy production.


Subject(s)
DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/genetics , Disease Models, Animal , Endomyocardial Fibrosis/complications , Mitochondrial Diseases/complications , Mitochondrial Diseases/physiopathology , Myocardial Contraction/physiology , Respiratory Physiological Phenomena , Analysis of Variance , Animals , Crosses, Genetic , DNA Polymerase gamma , DNA Primers/genetics , DNA, Mitochondrial/genetics , DNA-Directed DNA Polymerase/metabolism , Echocardiography , Genotype , Histological Techniques , Hydroxyproline/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial Diseases/genetics , Mutation, Missense/genetics , Myocardial Contraction/drug effects , Oximetry , Zidovudine/pharmacology
6.
J Assist Reprod Genet ; 29(7): 637-42, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22527902

ABSTRACT

PURPOSE: To establish the phenotype of reproductive aging in our mouse model. To test the hypotheses that reproductive aging is associated with a decrease in mitochondrial abundance that could ultimately reflect dysfunction in oocytes. METHODS: Breeding studies were performed in young and aged female virgin wild type C57BL6J mice to establish their reproductive phenotype by measuring time to conception, litter size, and live birth per dam. Individual oocytes were analyzed for mtDNA content. Transmission electron microscopy was used to study ultrastructure of mitochondria in oocytes. RESULTS: Old females were found to have significantly prolonged time to conception and fewer surviving pups in their litters. Oocytes from old mice had 2.7-fold less mtDNA compared to younger controls (p < 0.001; 95 % CI 2.1-3.5). Decrease in mitochondrial organelle abundance in old animal's oocytes was confirmed with transmission electron microscopy. Distinct morphological changes were noted in mitochondria, suggesting altered mitochondrial biogenesis in the old animals' oocytes. CONCLUSIONS: Reproductive aging in mice is associated with reduced reproductive competence. Aging is associated with a significant decrease in number of mitochondria in oocytes. Our data support mitochondrial organelle loss and dysfunction in oocytes as a potential etiology for reproductive senescence.


Subject(s)
Aging/genetics , Aging/physiology , DNA, Mitochondrial/analysis , Mitochondria/genetics , Mitochondria/ultrastructure , Oocytes/ultrastructure , Animals , Female , Mice , Mice, Inbred C57BL , Oocytes/physiology , Reproduction
7.
Lab Invest ; 91(6): 852-8, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21403643

ABSTRACT

Tenofovir disoproxil fumarate (TDF) is an oral prodrug and acyclic nucleotide analog of adenosine monophosphate that inhibits HIV-1 (HIV) reverse transcriptase. A growing subset of TDF-treated HIV(+) individuals presented with acute renal failure, suggesting tenofovir-associated kidney-specific toxicity. Our previous studies using an HIV transgenic mouse model (TG) demonstrated specific changes in renal proximal tubular mitochondrial DNA (mtDNA) abundance. Nucleosides are regulated in biological systems via transport and metabolism in cellular compartments. In this study, the role(s) of organic anion transporter type 1 (OAT1) and multidrug-resistant protein type 4 (MRP4) in transport and regulation of tenofovir in proximal tubules were assessed. Renal toxicity was assessed in kidney tissues from OAT1 knockout (KO) or MRP4 KO compared with wild-type (WT, C57BL/6) mice following treatment with TDF (0.11 mg/day), didanosine (ddI, a related adenosine analog, 0.14 mg/day) or vehicle (0.1 M NaOH) daily gavage for 5 weeks. Laser-capture microdissection (LCM) was used to isolate renal proximal tubules for molecular analyses. mtDNA abundance and ultrastructural pathology were analyzed. mtDNA abundance in whole kidneys from both KO and WT was unchanged regardless of treatment. Renal proximal tubular mtDNA abundance from OAT1 KO also remained unchanged, suggesting prevention of TDF toxicity due to loss of tenofovir transport into proximal tubules. In contrast, renal proximal tubules from MRP4 KO exhibited increased mtDNA abundance following TDF treatment compared with WT littermates, suggesting compensation. Renal proximal tubules from TDF-treated WT and MRP4 KO exhibited increased numbers of irregular mitochondria with sparse, fragmented cristae compared with OAT1 KO. Treatment with ddI had a compensatory effect on mtDNA abundance in OAT1 KO but not in MRP4 KO. Both OAT1 and MRP4 have a direct role in transport and efflux of tenofovir, regulating levels of tenofovir in proximal tubules. Disruption of OAT1 activity prevents tenofovir toxicity but loss of MRP4 can lead to increased renal proximal tubular toxicity. These data help to explain mechanisms of human TDF renal toxicity.


Subject(s)
Adenine/analogs & derivatives , Kidney Tubules, Proximal/drug effects , Kidney Tubules, Proximal/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Organic Anion Transport Protein 1/metabolism , Organophosphonates/toxicity , Adenine/administration & dosage , Adenine/toxicity , Analysis of Variance , Animals , DNA, Mitochondrial/metabolism , Kidney Tubules, Proximal/pathology , Lasers , Mice , Mice, Inbred C57BL , Mice, Knockout , Microdissection , Mitochondria/pathology , Multidrug Resistance-Associated Proteins/genetics , Organic Anion Transport Protein 1/genetics , Organophosphonates/administration & dosage , Reverse Transcriptase Polymerase Chain Reaction , Tenofovir
8.
Cardiovasc Toxicol ; 10(2): 146-51, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20379802

ABSTRACT

Abacavir (ABC) is a guanosine nucleoside reverse transcriptase inhibitor (NRTI) with potent antiretroviral activity. Since NRTIs exhibit tissue-specific inhibition of mitochondrial DNA (mtDNA) synthesis, the ability of ABC to inhibit mtDNA synthesis in vivo was evaluated. Inbred wild-type (WT) and transgenic mice (TG) treated with ABC (3.125 mg/d p. o., 35 days) were used to define mitochondrial oxidative stress and cardiac function. Chosen TGs exhibited overexpression of HIV-1 viral proteins (NL4-3Deltagag/pol, non-replication competent), hemizygous depletion or overexpression of mitochondrial superoxide dismutase (SOD2(+/-) knock-out (KO) or MnSOD OX, respectively), overexpression of mitochondrially targeted catalase (MCAT), or double "knockout" deletion of aldehyde dehydrogenase activity (ALDH2 KO). Impact on mtDNA synthesis was assessed by comparing changes in mtDNA abundance between ABC-treated and vehicle-treated WTs and TGs. No changes in mtDNA abundance occurred from ABC treatment in any mice, suggesting no inhibition of mtDNA synthesis. Left ventricle (LV) mass and LV end-diastolic dimension (LVEDD) were defined echocardiographically and remained unchanged as well. These results indicate that treatment with ABC has no visible cardiotoxicity in these adult mice exposed for 5 weeks compared to findings with other antiretroviral NRTI studies and support some claims for its relative safety.


Subject(s)
Anti-HIV Agents/toxicity , Dideoxynucleosides/toxicity , Heart Diseases/chemically induced , Heart/drug effects , Mitochondria, Heart/drug effects , Animals , DNA, Mitochondrial/biosynthesis , Echocardiography , Heart Diseases/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Mitochondria, Heart/enzymology , Mitochondria, Heart/genetics , Oxidative Stress/drug effects , Superoxide Dismutase
9.
Lab Invest ; 90(3): 383-90, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20065942

ABSTRACT

Thymidylate kinase (TMPK) is a nucleoside monophosphate kinase that catalyzes phosphorylation of thymidine monophosphate to thymidine diphosphate. TMPK also mediates phosphorylation of monophosphates of thymidine nucleoside analog (NA) prodrugs on the pathway to their active triphosphate antiviral or antitumor moieties. Novel transgenic mice (TG) expressing human (h) TMPK were genetically engineered using the alpha-myosin heavy chain promoter to drive its cardiac-targeted overexpression. In '2 by 2' protocols, TMPK TGs and wild-type (WT) littermates were treated with the NA zidovudine (a deoxythymidine analog, 3'-azido-3'deoxythymidine (AZT)) or vehicle for 35 days. Alternatively, TGs and WTs were treated with a deoxycytidine NA (racivir, RCV) or vehicle. Changes in mitochondrial DNA (mtDNA) abundance and mitochondrial ultrastructure were defined quantitatively by real-time PCR and transmission electron microscopy, respectively. Cardiac performance was determined echocardiographically. Results showed TMPK TGs treated with either AZT or RCV exhibited decreased cardiac mtDNA abundance. Cardiac ultrastructural changes were seen only with AZT. AZT-treated TGs exhibited increased left ventricle (LV) mass. In contrast, LV mass in RCV-treated TGs and WTs remained unchanged. In all cohorts, LV end-diastolic dimension remained unchanged. This novel cardiac-targeted overexpression of hTMPK helps define the role of TMPK in mitochondrial toxicity of antiretrovirals.


Subject(s)
Anti-HIV Agents/toxicity , DNA, Mitochondrial/metabolism , Myocardium/metabolism , Nucleoside-Phosphate Kinase/metabolism , Nucleosides/metabolism , Zalcitabine/analogs & derivatives , Zidovudine/toxicity , Animals , Anti-HIV Agents/metabolism , DNA Replication/drug effects , DNA, Mitochondrial/drug effects , Echocardiography , Emtricitabine/analogs & derivatives , Female , Humans , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/diagnostic imaging , Male , Mice , Mice, Transgenic , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Myocardium/pathology , Myocardium/ultrastructure , Nucleoside-Phosphate Kinase/genetics , Phosphorylation , Ventricular Function, Left , Zalcitabine/metabolism , Zalcitabine/toxicity , Zidovudine/metabolism
10.
Lab Invest ; 89(7): 782-90, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19398959

ABSTRACT

Transgenic mice (TG) were used to define mitochondrial oxidative stress and cardiomyopathy (CM) induced by zidovudine (AZT), an antiretroviral used to treat HIV/AIDS. Genetically engineered mice either depleted or overexpressed mitochondrial superoxide dismutase (SOD2(+/-) KOs and SOD2-OX, respectively) or expressed mitochondrially targeted catalase (mCAT). TGs and wild-type (WT) littermates were treated (oral AZT, 35 days). Cardiac mitochondrial H(2)O(2), aconitase activity, histology and ultrastructure were analyzed. Left ventricle (LV) mass and LV end-diastolic dimension were determined echocardiographically. AZT induced cardiac oxidative stress and LV dysfunction in WTs. Cardiac mitochondrial H(2)O(2) increased and aconitase was inactivated in SOD2(+/-) KOs, and cardiac dysfunction was worsened by AZT. Conversely, the cardiac function in SOD2-OX and mCAT hearts was protected. In SOD2-OX and mCAT TG hearts, mitochondrial H(2)O(2), LV mass and LV cavity volume resembled corresponding values from vehicle-treated WTs. AZT worsens cardiac dysfunction and increases mitochondrial H(2)O(2) in SOD2(+/-) KO. Conversely, both SOD2-OX and mCAT TGs prevent or attenuate AZT-induced cardiac oxidative stress and LV dysfunction. As dysfunctional changes are ameliorated by decreasing and worsened by increasing H(2)O(2) abundance, oxidative stress from H(2)O(2) is crucial pathogenetically in AZT-induced mitochondrial CM.


Subject(s)
Anti-HIV Agents/toxicity , Cardiomyopathies/chemically induced , Cardiomyopathies/prevention & control , Catalase/metabolism , Mitochondria, Heart/drug effects , Mitochondria, Heart/enzymology , Superoxide Dismutase/metabolism , Zidovudine/toxicity , Aconitate Hydratase/metabolism , Animals , Cardiomyopathies/genetics , Cardiomyopathies/metabolism , Catalase/genetics , Female , Gene Expression , Hydrogen Peroxide/metabolism , Male , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria, Heart/ultrastructure , Models, Cardiovascular , Myocardium/pathology , Oxidative Stress/drug effects , Phenotype , Superoxide Dismutase/deficiency , Superoxide Dismutase/genetics
11.
Lab Invest ; 89(5): 513-9, 2009 May.
Article in English | MEDLINE | ID: mdl-19274046

ABSTRACT

Tenofovir disoproxil fumarate (TDF) is an analog of adenosine monophosphate that inhibits HIV reverse transcriptase in HIV/AIDS. Despite its therapeutic success, renal tubular side effects are reported. The mechanisms and targets of tenofovir toxicity were determined using '2 x 2' factorial protocols, and HIV transgenic (TG) and wild-type (WT) littermate mice with or without TDF (5 weeks). A parallel study used didanosine (ddI) instead of TDF. At termination, heart, kidney, and liver samples were retrieved. Mitochondrial DNA (mtDNA) abundance, and histo- and ultrastructural pathology were analyzed. Laser-capture microdissection (LCM) was used to isolate renal proximal tubules for molecular analyses. Tenofovir increased mtDNA abundance in TG whole kidneys, but not in their hearts or livers. In contrast, ddI decreased mtDNA abundance in the livers of WTs and TGs, but had no effect on their hearts or kidneys. Histological analyses of kidneys showed no disruption of glomeruli or proximal tubules with TDF or ddI treatments. Ultrastructural changes in renal proximal tubules from TDF-treated TGs included an increased number and irregular shape of mitochondria with sparse fragmented cristae. LCM-captured renal proximal tubules from TGs showed decreased mtDNA abundance with tenofovir. The results indicate that tenofovir targets mitochondrial toxicity on the renal proximal tubule in an AIDS model.


Subject(s)
AIDS-Associated Nephropathy/chemically induced , Adenine/analogs & derivatives , Anti-HIV Agents/adverse effects , Kidney Tubules, Proximal/drug effects , Mitochondria/drug effects , Organophosphonates/adverse effects , AIDS-Associated Nephropathy/pathology , Adenine/adverse effects , Animals , DNA, Mitochondrial/metabolism , Didanosine/adverse effects , Female , HIV-1 , Kidney Tubules, Proximal/metabolism , Kidney Tubules, Proximal/ultrastructure , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microdissection , Mitochondria/ultrastructure , Tenofovir , Urothelium/ultrastructure
12.
Lab Invest ; 89(2): 122-30, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19079325

ABSTRACT

Mitochondrial toxicity results from pyrimidine nucleoside reverse transcriptase inhibitors (NRTIs) for HIV/AIDS. In the heart, this can deplete mitochondrial (mt) DNA and cause cardiac dysfunction (eg, left ventricle hypertrophy, LVH). Four unique transgenic, cardiac-targeted overexpressors (TGs) were generated to determine their individual impact on native mitochondrial biogenesis and effects of NRTI administration on development of mitochondrial toxicity. TGs included cardiac-specific overexpression of native thymidine kinase 2 (TK2), two pathogenic TK2 mutants (H121N and I212N), and a mutant of mtDNA polymerase, pol-gamma (Y955C). Each was treated with antiretrovirals (AZT-HAART, 3 or 10 weeks, zidovudine (AZT) + lamivudine (3TC) + indinavir, or vehicle control). Parameters included left ventricle (LV) performance (echocardiography), LV mtDNA abundance (real-time PCR), and mitochondrial fine structure (electron microscopy, EM) as a function of duration of treatment and presence of TG. mtDNA abundance significantly decreased in Y955C TG, increased in TK2 native and I212N TGs, and was unchanged in H121N TGs at 10 weeks regardless of treatment. Y955C and I212N TGs exhibited LVH during growth irrespective of treatment. Y955C TGs exhibited cardiomyopathy (CM) at 3 and 10 weeks irrespective of treatment, whereas H121N and I212N TGs exhibited CM only after 10 weeks AZT-HAART. EM features were consistent with cardiac dysfunction. mtDNA abundance and cardiac functional changes were related to TG expression of mitochondrially related genes, mutations thereof, and NRTIs.


Subject(s)
Anti-HIV Agents/toxicity , DNA, Mitochondrial/metabolism , Mitochondria, Heart/metabolism , Myocytes, Cardiac/metabolism , Reverse Transcriptase Inhibitors/toxicity , Thymidine Kinase/metabolism , Animals , Antiretroviral Therapy, Highly Active , Cardiomyopathy, Dilated/chemically induced , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cell Line , DNA, Mitochondrial/analysis , Echocardiography , Female , Heart Ventricles/chemistry , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Hypertrophy, Left Ventricular/chemically induced , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/metabolism , Indinavir/toxicity , Lamivudine/toxicity , Male , Mice , Mice, Transgenic , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Phosphorylation , Thymidine Kinase/genetics , Zidovudine/toxicity
13.
Cardiovasc Toxicol ; 8(2): 57-69, 2008.
Article in English | MEDLINE | ID: mdl-18446447

ABSTRACT

Mitochondrial (mt) DNA biogenesis is critical to cardiac contractility. DNA polymerase gamma (Pol gamma) replicates mtDNA, whereas thymidine kinase 2 (TK2) monophosphorylates pyrimidines intramitochondrially. Point mutations in POLG and TK2 result in clinical diseases associated with mtDNA depletion and organ dysfunction. Pyrimidine analogs (NRTIs) inhibit Pol gamma and mtDNA replication. Cardiac "dominant negative" murine transgenes (TGs; Pol gamma Y955C, and TK2 H121N or I212N) defined the role of each in the heart. mtDNA abundance, histopathological features, histochemistry, mitochondrial protein abundance, morphometry, and echocardiography were determined for TGs in "2 x 2" studies with or without pyrimidine analogs. Cardiac mtDNA abundance decreased in Y955C TGs ( approximately 50%) but increased in H121N and I212N TGs (20-70%). Succinate dehydrogenase (SDH) increased in hearts of all mutants. Ultrastructural changes occurred in Y955C and H121N TGs. Histopathology demonstrated hypertrophy in H121N, LV dilation in I212N, and both hypertrophy and dilation in Y955C TGs. Antiretrovirals increased LV mass ( approximately 50%) for all three TGs which combined with dilation indicates cardiomyopathy. Taken together, these studies demonstrate three manifestations of cardiac dysfunction that depend on the nature of the specific mutation and antiretroviral treatment. Mutations in genes for mtDNA biogenesis increase risk for defective mtDNA replication, leading to LV hypertrophy.


Subject(s)
Anti-Retroviral Agents/toxicity , Cardiomyopathies/enzymology , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/metabolism , Mitochondria, Heart/enzymology , Myocytes, Cardiac/enzymology , Thymidine Kinase/metabolism , Animals , Cardiomyopathies/diagnostic imaging , Cardiomyopathies/etiology , Cardiomyopathy, Dilated/enzymology , Cardiomyopathy, Dilated/etiology , DNA Polymerase gamma , DNA-Directed DNA Polymerase/genetics , Electron Transport Complex I/metabolism , Humans , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/etiology , Mice , Mice, Transgenic , Mitochondria, Heart/drug effects , Mitochondria, Heart/ultrastructure , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/ultrastructure , Point Mutation , Succinate Dehydrogenase/metabolism , Thymidine Kinase/genetics , Ultrasonography
14.
Lab Invest ; 87(4): 326-35, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17310215

ABSTRACT

POLG is the human gene that encodes the catalytic subunit of DNA polymerase gamma (Pol gamma), the replicase for human mitochondrial DNA (mtDNA). A POLG Y955C point mutation causes human chronic progressive external ophthalmoplegia (CPEO), a mitochondrial disease with eye muscle weakness and mtDNA defects. Y955C POLG was targeted transgenically (TG) to the murine heart. Survival was determined in four TG (+/-) lines and wild-type (WT) littermates (-/-). Left ventricle (LV) performance (echocardiography and MRI), heart rate (electrocardiography), mtDNA abundance (real time PCR), oxidation of mtDNA (8-OHdG), histopathology and electron microscopy defined the phenotype. Cardiac targeted Y955C POLG yielded a molecular signature of CPEO in the heart with cardiomyopathy (CM), mitochondrial oxidative stress, and premature death. Increased LV cavity size and LV mass, bradycardia, decreased mtDNA, increased 8-OHdG, and cardiac histopathological and mitochondrial EM defects supported and defined the phenotype. This study underscores the pathogenetic role of human mutant POLG and its gene product in mtDNA depletion, mitochondrial oxidative stress, and CM as it relates to the genetic defect in CPEO. The transgenic model pathophysiologically links human mutant Pol gamma, mtDNA depletion, and mitochondrial oxidative stress to the mtDNA replication apparatus and to CM.


Subject(s)
Cardiomyopathies/pathology , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/physiology , Oxidative Stress , 8-Hydroxy-2'-Deoxyguanosine , Animals , Cardiomyopathies/genetics , Cardiomyopathies/mortality , DNA Polymerase gamma , DNA-Directed DNA Polymerase/genetics , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Heart Ventricles/pathology , Humans , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Mitochondria, Heart/metabolism , Mitochondria, Heart/ultrastructure , Mutation , Myocardium/metabolism , Myocardium/pathology
15.
Am J Pathol ; 170(3): 865-74, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17322372

ABSTRACT

Mitochondrial toxicity limits nucleoside reverse transcriptase inhibitors (NRTIs) for acquired immune deficiency syndrome. NRTI triphosphates, the active moieties, inhibit human immunodeficiency virus reverse transcriptase and eukaryotic mitochondrial DNA polymerase pol-gamma. NRTI phosphorylation seems to correlate with mitochondrial toxicity, but experimental evidence is lacking. Transgenic mice (TGs) with cardiac overexpression of thymidine kinase isoforms (mitochondrial TK2 and cytoplasmic TK1) were used to study NRTI mitochondrial toxicity. Echocardiography and nuclear magnetic resonance imaging defined cardiac performance and structure. TK gene copy and enzyme activity, mitochondrial (mt) DNA and polypeptide abundance, succinate dehydrogenase and cytochrome oxidase histochemistry, and electron microscopy correlated with transgenesis, mitochondrial structure, and biogenesis. Antiretroviral combinations simulated therapy. Untreated hTK1 or TK2 TGs exhibited normal left ventricle mass. In TK2 TGs, cardiac TK2 gene copy doubled, activity increased 300-fold, and mtDNA abundance doubled. Abundance of the 17-kd subunit of complex I, succinate dehydrogenase histochemical activity, and cristae density increased. NRTIs increased left ventricle mass 20% in TK2 TGs. TK activity increased 3 logs in hTK1 TGs, but no cardiac phenotype resulted. NRTIs abrogated functional effects of transgenically increased TK2 activity but had no effect on TK2 mtDNA abundance. Thus, NRTI mitochondrial phosphorylation by TK2 is integral to clinical NRTI mitochondrial toxicity.


Subject(s)
Mitochondria/drug effects , Reverse Transcriptase Inhibitors/toxicity , Thymidine Kinase/drug effects , Thymidine Kinase/metabolism , Transgenes , Animals , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Echocardiography , Humans , Immunoblotting , Immunohistochemistry , Isoenzymes/drug effects , Isoenzymes/metabolism , Magnetic Resonance Spectroscopy , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria/metabolism , Myocardium/enzymology , Peptides/drug effects , Phosphorylation/drug effects , Reverse Transcriptase Polymerase Chain Reaction , Succinate Dehydrogenase/drug effects , Up-Regulation
16.
AIDS ; 20(5): 675-84, 2006 Mar 21.
Article in English | MEDLINE | ID: mdl-16514297

ABSTRACT

DESIGN: Nucleoside reverse transcriptase inhibitors (NRTIs) exhibit mitochondrial toxicity. The mitochondrial deoxynucleotide carrier (DNC) transports nucleotide precursors (or phosphorylated NRTIs) into mitochondria for mitochondrial (mt)DNA replication or inhibition of mtDNA replication by NRTIs. Transgenic mice (TG) expressing human DNC targeted to murine myocardium served to define mitochondrial events from NRTIs in vivo and findings were corroborated by biochemical events in vitro. METHODS: Zidovudine (3'-azido-2',3'-deoxythymidine; ZDV), stavudine (2', 3'-didehydro-2', 3'-deoxythymidine; d4T), or lamivudine ((-)-2'-deoxy-3'-thiacytidine; 3TC) were administered individually to TGs and wild-type (WT) littermates (35 days) at human doses with drug-free vehicle as control. Left ventricle (LV) mass was defined echocardiographically, mitochondrial ultrastructural defects were identified by electron microscopy, the abundance of cardiac mtDNA was quantified by real time polymerase chain reaction, and mtDNA-encoded polypeptides were quantified. RESULTS: Untreated TGs exhibited normal LV mass with minor mitochondrial damage. NRTI monotherapy (either d4T or ZDV) increased LV mass in TGs and caused significant mitochondrial destruction. Cardiac mtDNA was depleted in ZDV and d4T-treated TG hearts and mtDNA-encoded polypeptides decreased. Changes were absent in 3TC-treated cohorts. In supportive structural observations from molecular modeling, ZDV demonstrated close contacts with K947 and Y951 in the DNA pol gamma active site that were absent in the HIV reverse transcriptase active site. CONCLUSIONS: NRTIs deplete mtDNA and polypeptides, cause mitochondrial structural and functional defects in vivo, follow inhibition kinetics with DNA pol gamma in vitro, and are corroborated by molecular models. Disrupted pools of nucleotide precursors and inhibition of DNA pol gamma by specific NRTIs are mechanistically important in mitochondrial toxicity.


Subject(s)
DNA, Mitochondrial/drug effects , Genes, pol , HIV-1/genetics , Membrane Transport Proteins , Reverse Transcriptase Inhibitors/pharmacology , Zidovudine/pharmacology , Animals , DNA/analysis , DNA, Mitochondrial/analysis , Echocardiography , HIV Infections/drug therapy , HIV Infections/pathology , Humans , Immunoblotting , Lamivudine/pharmacology , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria, Heart/ultrastructure , Mitochondrial Membrane Transport Proteins , Models, Molecular , Myocardium/ultrastructure , Reverse Transcriptase Polymerase Chain Reaction , Stavudine/pharmacology
17.
Lab Invest ; 85(8): 972-81, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15951836

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) are antiretrovirals for AIDS with limiting mitochondrial side effects. The mitochondrial deoxynucleotide carrier (DNC) transports phosphorylated nucleosides for mitochondrial DNA replication and can transport phosphorylated NRTIs into mitochondria. Transgenic mice (TG) that exclusively overexpress DNC in the heart tested DNC's role in mitochondrial dysfunction from NRTIs. Two TG lines were created that overexpressed the human DNC gene in murine myocardium. Cardiac and mitochondrial structure and function were examined by magnetic resonance imaging, echocardiography, electrocardiography, transmission electron microscopy, and plasma lactate. Antiretroviral combinations (HAART) that contained NRTIs (stavudine (2', 3'-didehydro-2', 3'-deoxythymidine or d4T)/lamivudine/indinavir; or zidovudine (3' azido-3'-deoxythymidine or AZT)/lamivudine/indinavir; 35 days) were administered to simulate AIDS therapy. In parallel, a HAART combination without NRTIs (nevirapine/efavirenz/indinavir; 35 days) served as an NRTI-sparing, control regimen. Untreated DNC TGs exhibited normal cardiac function but abnormal mitochondrial ultrastructure. HAART that contained NRTIs caused cardiomyopathy in TGs with increased left ventricle mass and volume, heart rate variability, and worse mitochondrial ultrastructural defects. In contrast, treatment with an NRTI-sparing HAART regimen caused no cardiac changes. Data suggest the DNC is integral to mitochondrial homeostasis in vivo and may relate mechanistically to mitochondrial dysfunction in patients treated with HAART regimens that contain NRTIs.


Subject(s)
Acquired Immunodeficiency Syndrome/drug therapy , Membrane Transport Proteins/physiology , Mitochondria, Heart/drug effects , Reverse Transcriptase Inhibitors/adverse effects , Animals , Antiretroviral Therapy, Highly Active/adverse effects , Electrocardiography , Lactic Acid/blood , Membrane Transport Proteins/genetics , Mice , Mice, Transgenic , Microscopy, Electron, Transmission , Mitochondria, Heart/ultrastructure , Mitochondrial Membrane Transport Proteins
18.
Lab Invest ; 85(2): 182-92, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15608661

ABSTRACT

HIV viral protein R (Vpr) affects the immunocyte cell cycle and circulates as free polypeptide in plasma of AIDS patients. Effects of Vpr on cardiomyocytes were explored using transgenic mice (TG) with Vpr targeted to cardiomyocytes by the alpha-myosin heavy-chain promoter. TG and WT littermate hearts were evaluated histopathologically, ultrastructurally, molecularly via RNA microarray analysis and quantitative RT-PCR, and functionally by cardiac magnetic resonance imaging (MRI) and electrocardiograms (ECG). Six hemizygous lines were created (Vpr(a,b,c,d,e,h)). Vpr RNA was expressed exclusively in myocardium and Vpr mRNA expression correlated with phenotypic changes. Vpr(b) exhibited the highest expression and mortality. TGs developed congestive heart failure ( approximately 8 weeks), abnormal cardiomyocyte nuclei and mitoses ( approximately 12 weeks), and became moribund ( approximately 20 weeks) with atrial mesenchymal tumors. MRI revealed four-chamber dilation, defective contraction, and atrial masses. Pathologically, cardiomegaly and atrial mesenchymal tumors occurred ( approximately 16-20 weeks). ECGs showed prolonged R-R, Q-T, and P-R intervals ( approximately 12 weeks). RNA encoding collagen and bone morphogenic protein 4, 6, and 7 were increased. Vpr targeted to cardiomyocytes caused defective contractility and atrial tumors. Since some Vpr cardiomyocytic effects resemble those found in terminally differentiated immunocytes, some pathogenetic mechanisms may be shared at the subcellular level.


Subject(s)
Arrhythmias, Cardiac/genetics , Gene Products, vpr/genetics , Heart Failure/genetics , Heart Neoplasms/pathology , Mitosis , Myocytes, Cardiac/pathology , Animals , Biomarkers , Cardiomegaly/genetics , Electrocardiography , Gene Expression Regulation, Viral , HIV-1/genetics , Heart Atria/pathology , Magnetic Resonance Imaging , Mice , Mice, Transgenic , Myocardium/metabolism , Myocardium/pathology , Myocardium/ultrastructure , Myocytes, Cardiac/ultrastructure , Myosin Heavy Chains/genetics , Oligonucleotide Array Sequence Analysis , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , vpr Gene Products, Human Immunodeficiency Virus
19.
Lab Invest ; 83(7): 983-9, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12861038

ABSTRACT

Cardiac dysfunction in AIDS is an important problem. Cocaine is an epidemic associated with sudden death, cardiac dysfunction, and congestive heart failure. Cocaine use and HIV infection frequently coexist in the same patient, yet the combined impact of both is poorly understood. The present study uses cocaine treatment of an established murine AIDS transgenic model (NL4-3Delta gag/pol; TG) to define the combined effects of AIDS and cocaine on cardiac pathophysiology. To determine the effects of cocaine and HIV-1 proteins on mortality, wild-type and NL4-3Delta gag/pol mice received saline or cocaine via continuous infusion by Alzet osmotic pumps for 28 days (chronic). Acute cocaine administration (10 days; 40 mg/kg/day) was used to study the nonlethal effects of cocaine in TGs. Echocardiograms and single time point electrocardiograms were performed at the termination of each experiment. Hearts were removed and examined histopathologically. Chronic cocaine treatment (80 mg/kg/day; 28 days) markedly decreased median survival in both wild-type and TG; however, TG survival was significantly more decreased. In acute studies, TG echocardiographic changes included increased left ventricular mass and increased left ventricular fractional shortening compared with all cohorts. Electrocardiographic changes were absent among the groups. Histopathologically, perivascular fibrosis and interstitial fibrosis were evident in cocaine-treated TG. Data suggest that additive cardiac insults (from AIDS and cocaine) result in combined deleterious effects.


Subject(s)
Acquired Immunodeficiency Syndrome/pathology , Cardiomyopathies/pathology , Cocaine/pharmacology , Heart Ventricles/drug effects , Heart Ventricles/pathology , Acquired Immunodeficiency Syndrome/complications , Acquired Immunodeficiency Syndrome/mortality , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/mortality , Disease Models, Animal , Electrocardiography , Female , Heart Ventricles/physiopathology , Infusion Pumps, Implantable , Male , Mice , Mice, Inbred Strains , Mice, Transgenic , Myocardium/pathology , Organ Size/drug effects , Survival Rate , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/virology
20.
Am J Physiol Heart Circ Physiol ; 283(6): H2363-70, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12388299

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) have been used successfully to reduce acquired immunodeficiency syndrome mortality. However, the use of these compounds is associated with numerous tissue toxicities, including cardiomyopathy. These studies address the effects of NRTIs on vascular function. Functional assays of contraction and relaxation were performed on isolated mouse aorta segments obtained from FVB/n mice exposed to zidovudine (AZT), stavudine, or water for 35 days. AZT and stavudine treatment impaired sensitivity to endothelium-dependent relaxation by acetylcholine. Dihydroethidium staining revealed that AZT treatment was associated with an increase in superoxide levels. Pretreatment of AZT-treated vessels with tiron (1 mM), a free radical scavenger, restored endothelium-dependent relaxation in mice. In cellular preparations, electron spin resonance measurements revealed elevated superoxide in cultured endothelial cells exposed to AZT; elevation was dependent on the length of exposure. These results indicate that NRTIs impair endothelium-dependent relaxation by increasing superoxide levels and suggest that NRTI therapy contributes to cardiovascular complications in acquired immunodeficiency syndrome.


Subject(s)
Endothelium, Vascular/drug effects , Nucleosides/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Superoxides/metabolism , Vasodilation/drug effects , 1,2-Dihydroxybenzene-3,5-Disulfonic Acid Disodium Salt/pharmacology , Animals , Aorta/cytology , Aorta/drug effects , Aorta/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Electron Spin Resonance Spectroscopy , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Free Radical Scavengers/pharmacology , In Vitro Techniques , Male , Mice , Mice, Inbred Strains , Stavudine/pharmacology , Vasoconstriction/drug effects , Vasoconstriction/physiology , Vasoconstrictor Agents/pharmacology , Vasodilation/physiology , Vasodilator Agents/pharmacology , Zidovudine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...