Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 73
Filter
1.
Environ Mol Mutagen ; 60(5): 404-409, 2019 06.
Article in English | MEDLINE | ID: mdl-29206312

ABSTRACT

All nucleoside analogues for treating HIV infection, due to their capacity to integrate into and alter human DNA, are experimentally genotoxic to some extent. The long-term oncogenic risk after in utero exposure remains to be determined. Cancer incidence in uninfected children exposed to nucleos(t)ide reverse transcriptase inhibitors (NRTIs) was evaluated, by cross-checking against the National Cancer Registry, in the French perinatal study of children born to HIV+ mothers. Twenty-one cancers were identified in 15,163 children (median age: 9.9 years [interquartile range (IQR): 5.8-14.2]) exposed to at least one NRTI in utero between 1990 and 2014. Five of these children were exposed to zidovudine monotherapy, and 15 to various combinations, seven of which included didanosine. Overall, the total number of cases was not significantly different from that expected for the general population (SIR = 0.8[0.47-1.24]), but the number of cases after didanosine exposure was twice that expected (SIR = 2.5 [1.01-5.19]). Didanosine accounted for only 10% of prescriptions but was associated with one-third of cancers. In multivariate analysis, didanosine exposure was significantly associated with higher risk (HR = 3.0 [0.9-9.8]). This risk was specifically linked to first-trimester exposure (HR = 5.5 [2.1-14.4]). Three cases of pineoblastoma, a very rare cancer, were observed, whereas 0.03 were expected. Two were associated with didanosine exposure. Despite reassuring data overall, there is strong evidence to suggest that didanosine displays transplacental oncogenicity. These findings cannot be extrapolated to other NRTIs, but they highlight the need for comprehensive evaluations of the transplacental genotoxicity of this antiretroviral class. Environ. Mol. Mutagen., 60:404-409, 2019. © 2017 Wiley Periodicals, Inc.


Subject(s)
Anti-HIV Agents/toxicity , Maternal Exposure , Maternal-Fetal Exchange/physiology , Neoplasms/epidemiology , Nucleosides/toxicity , Prenatal Exposure Delayed Effects/pathology , Adolescent , Anti-HIV Agents/therapeutic use , Child , Child, Preschool , Didanosine/therapeutic use , Didanosine/toxicity , Female , HIV Infections/drug therapy , Humans , Incidence , Infant , Infant, Newborn , Male , Neoplasms/genetics , Nucleosides/therapeutic use , Pregnancy , Prospective Studies , Reverse Transcriptase Inhibitors/therapeutic use , Reverse Transcriptase Inhibitors/toxicity , Risk , Surveys and Questionnaires , Zidovudine/therapeutic use , Zidovudine/toxicity
2.
J Antimicrob Chemother ; 71(4): 916-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26747094

ABSTRACT

BACKGROUND: NRTIs are essential components of HIV therapy with well-documented, long-term mitochondrial toxicity in hepatic cells, but whose acute effects on mitochondria are unclear. As acetaminophen-induced hepatotoxicity also involves mitochondrial interference, we hypothesized that it would be exacerbated in the context of ART. METHODS: We evaluated the acute effects of clinically relevant concentrations of the most widely used NRTIs, alone or combined with acetaminophen, on mitochondrial function and cellular viability. RESULTS: The purine analogues abacavir and didanosine produced an immediate and concentration-dependent inhibition of oxygen consumption and complex I and III activity. This inhibition was accompanied by an undermining of mitochondrial function, with increased production of reactive oxygen species and reduction of mitochondrial membrane potential and intracellular ATP levels. However, this interference did not compromise cell survival. Co-administration with concentrations of acetaminophen below those considered hepatotoxic exacerbated the deleterious effects of both compounds on mitochondrial function and compromised cellular viability, showing a clear correlation with diminished glutathione levels. CONCLUSIONS: The simultaneous presence of purine analogues and low concentrations of acetaminophen significantly potentiates mitochondrial dysfunction, increasing the risk of liver injury. This new mechanism is relevant given the liver's susceptibility to mitochondrial dysfunction-related toxicity and the tendency of the HIV infection to increase oxidative stress.


Subject(s)
Acetaminophen/toxicity , Analgesics, Non-Narcotic/toxicity , Anti-HIV Agents/toxicity , Chemical and Drug Induced Liver Injury/pathology , Didanosine/toxicity , Dideoxynucleosides/toxicity , Mitochondria, Liver/drug effects , Mitochondrial Diseases/chemically induced , Cell Line , Electron Transport Chain Complex Proteins/drug effects , Glutathione/metabolism , Humans , Oxygen Consumption/drug effects , Reactive Nitrogen Species/metabolism
3.
PLoS One ; 10(5): e0126220, 2015.
Article in English | MEDLINE | ID: mdl-25970180

ABSTRACT

Today HIV-1 infection is recognized as a chronic disease with obligatory lifelong treatment to keep viral titers below detectable levels. The continuous intake of antiretroviral drugs however, leads to severe and even life-threatening side effects, supposedly by the deleterious impact of nucleoside-analogue type compounds on the functioning of the mitochondrial DNA polymerase. For detailed investigation of the yet partially understood underlying mechanisms, the availability of a versatile model system is crucial. We therefore set out to develop the use of Caenorhabditis elegans to study drug induced mitochondrial toxicity. Using a combination of molecular-biological and functional assays, combined with a quantitative analysis of mitochondrial network morphology, we conclude that anti-retroviral drugs with similar working mechanisms can be classified into distinct groups based on their effects on mitochondrial morphology and biochemistry. Additionally we show that mitochondrial toxicity of antiretroviral drugs cannot be exclusively attributed to interference with the mitochondrial DNA polymerase.


Subject(s)
Anti-HIV Agents/toxicity , Caenorhabditis elegans/drug effects , DNA, Mitochondrial/antagonists & inhibitors , Drug Evaluation/methods , Mitochondria/drug effects , Reverse Transcriptase Inhibitors/toxicity , Animals , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/genetics , DNA-Directed DNA Polymerase/metabolism , Didanosine/toxicity , Dideoxynucleosides/toxicity , Humans , Mitochondria/genetics , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Models, Biological , Oxygen Consumption/drug effects , Stavudine/toxicity , Ubiquinone/antagonists & inhibitors , Ubiquinone/metabolism , Zalcitabine/toxicity , Zidovudine/toxicity
4.
Biochem Biophys Res Commun ; 450(2): 1021-6, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24976398

ABSTRACT

Mitochondrial thymidine kinase 2 (TK2) and deoxyguanosine kinase (dGK) catalyze the initial rate limiting phosphorylation of deoxynucleosides and are essential enzymes for mitochondrial function. Chemotherapy using nucleoside analogs is often associated with mitochondrial toxicities. Here we showed that incubation of U2OS cells with didanosine (ddI, 2',3'-dideoxyinosine), a purine nucleoside analog used in the highly active antiretroviral therapy (HAART), led to selective degradation of both mitochondrial TK2 and dGK while the cytosolic deoxycytidine kinase (dCK) and thymidine kinase 1 (TK1) were not affected. Addition of guanosine to the ddI-treated cells prevented the degradation of mitochondrial TK2 and dGK. The levels of intracellular reactive oxygen species and protein oxidation in ddI-treated and control cells were also measured. The results suggest that down-regulation of mitochondrial TK2 and dGK may be a mechanism of mitochondrial toxicity caused by antiviral and anticancer nucleoside analogs.


Subject(s)
Anti-HIV Agents/toxicity , Didanosine/toxicity , Mitochondria/drug effects , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Thymidine Kinase/metabolism , Cell Line, Tumor , Deoxycytidine Kinase/metabolism , Drug Interactions , Guanosine/pharmacology , Humans , Mitochondria/enzymology , Protein Carbonylation , Reactive Oxygen Species/metabolism
5.
Toxicol Sci ; 139(1): 133-41, 2014 May.
Article in English | MEDLINE | ID: mdl-24591154

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs), essential components of combinational therapies used for treatment of human immunodeficiency virus-1, damage heart mitochondria. Here, we have shown mitochondrial compromise in H9c2 rat cardiomyocytes exposed for 16 passages (P) to the NRTIs zidovudine (AZT, 50µM) and didanosine (ddI, 50µM), and we have demonstrated protection from mitochondrial compromise in cells treated with 200µM 1-oxyl-2,2,6,6-tetramethyl-4-hydroxypiperidine (Tempol) or 200µM 1-hydroxy-4-[2-triphenylphosphonio)-acetamido]-2,2,6,6-tetramethylpiperidine (Tempol-H), along with AZT/ddI, for 16P. Exposure to AZT/ddI caused a moderate growth inhibition at P3, P5, P7, and P13, which was not altered by addition of Tempol or Tempol-H. Mitochondrial oxidative phosphorylation capacity was determined as uncoupled oxygen consumption rate (OCR) by Seahorse XF24 Analyzer. At P5, P7, and P13, AZT/ddI-exposed cells showed an OCR reduction of 8.8-57.2% in AZT/ddI-exposed cells, compared with unexposed cells. Addition of Tempol or Tempol-H, along with AZT/ddI, resulted in OCR levels increased by about 300% above the values seen with AZT/ddI alone. The Seahorse data were further supported by electron microscopy (EM) studies in which P16 cells exposed to AZT/ddI/Tempol had less mitochondrial pathology than P16 cells exposed to AZT/ddI. Western blots of P5 cells showed that Tempol and Tempol-H upregulated expression of mitochondrial uncoupling protein-2 (UCP-2). However, Complex I activity that was reduced by AZT/ddI, was not restored in the presence of AZT/ddI/Tempol. Superoxide levels were increased in the presence of AZT/ddI and significantly decreased in cells exposed to AZT/3TC/Tempol at P3, P7, and P10. In conclusion, Tempol protects against NRTI-induced mitochondrial compromise, and UCP-2 plays a role through mild uncoupling.


Subject(s)
Cyclic N-Oxides/pharmacology , Mitochondria/drug effects , Myocytes, Cardiac/drug effects , Reverse Transcriptase Inhibitors/toxicity , Animals , Cell Line , Didanosine/toxicity , Microscopy, Electron , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Rats , Spin Labels , Superoxides/metabolism , Zidovudine/toxicity
6.
Am J Pathol ; 180(6): 2276-83, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22469844

ABSTRACT

The roles and actions of the tumor suppressor protein p53 have been extensively studied with regard to nuclear events, including transcription and DNA damage repair. However, the direct roles of p53 in mitochondrial DNA (mtDNA) replication and function are less well understood. Studies herein used a mitochondrial-targeted p53 (MTS-p53) to determine its effects on both mtDNA abundance and mitochondrial function. MTS-p53 decreased cellular proliferation and mtDNA abundance in HepG2 cells transfected with wild-type (WT) human p53. When MTS-p53 cells were treated with the nucleoside reverse transcriptase inhibitor (NRTI), 2',3'-dideoxycytidine or 2',3'-dideoxyinosine, mtDNA depletion that resembled untransfected controls was observed in both instances. p53-Overexpressing cells showed reduced mitochondrial function by oximetry, including a reduction in maximal respiratory capacity and reserve capacity. A truncated p53 (MTS-p53-290) was generated for localization exclusively to the mitochondria. MTS-p53-290 cells proliferated at control levels but displayed decreased mtDNA abundance and mitochondrial function with NRTI treatment. The MTS-p53-290 cells demonstrated that only the nuclear fraction of p53 controlled cellular proliferation, which was supported by the MTS-p53 results. Data herein indicate that overexpression of p53 in the mitochondria reduces mtDNA abundance and increases the sensitivity of mammalian cells to NRTI exposure by reducing mitochondrial function.


Subject(s)
DNA, Mitochondrial/biosynthesis , Mitochondria/physiology , Reverse Transcriptase Inhibitors/toxicity , Tumor Suppressor Protein p53/physiology , Cell Proliferation , DNA Replication/physiology , DNA, Mitochondrial/drug effects , Didanosine/toxicity , Hep G2 Cells , Homeostasis/genetics , Homeostasis/physiology , Humans , Mitochondria/drug effects , Mitochondria/metabolism , Transfection , Tumor Suppressor Protein p53/metabolism , Zalcitabine/toxicity
7.
Toxicol Sci ; 118(1): 191-201, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20702595

ABSTRACT

Mitochondrial compromise has been documented in infants born to women infected with the human immunodeficiency virus (HIV-1) who received nucleoside reverse transcriptase inhibitor (NRTI) therapy during pregnancy. To model these human exposures, we examined mitochondrial integrity at birth and 1 year in brain cortex and liver from offspring of retroviral-free Erythrocebus patas dams-administered human-equivalent NRTI doses for the last half (10 weeks) of gestation. Additional infants, followed for 1 year, were given the same drugs as their mothers for the first 6 weeks of life. Exposures included: no drug, Zidovudine (AZT), Lamivudine (3TC), AZT/3TC, AZT/Didanosine (ddI), and Stavudine (d4T)/3TC. In brain and liver, oxidative phosphorylation (OXPHOS) enzyme activities (complexes I, II, and IV) showed minimal differences between unexposed and NRTI-exposed offspring at both times. Brain and liver mitochondria from most NRTI-exposed patas, both at birth and 1 year of age, contained significant (p < 0.05) morphological damage observed by electron microscopy (EM), based on scoring of coded photomicrographs. Brain and liver mitochondrial DNA (mtDNA) levels in NRTI-exposed patas were depleted significantly in the 3TC and d4T/3TC groups at birth and were depleted significantly (p < 0.05) at 1 year in all NRTI-exposed groups. In 1-year-old infants exposed in utero to NRTIs, mtDNA depletion was 28.8-51.8% in brain and 37.4-56.5% in liver. These investigations suggest that some NRTI-exposed human infants may sustain similar mitochondrial compromise in brain and liver and should be followed long term for cognitive integrity and liver function.


Subject(s)
Cerebral Cortex/drug effects , Erythrocebus patas , Mitochondria, Liver/drug effects , Reverse Transcriptase Inhibitors/toxicity , Animals , Animals, Newborn , Cerebral Cortex/metabolism , Cerebral Cortex/ultrastructure , DNA, Mitochondrial/analysis , DNA, Mitochondrial/metabolism , Didanosine/toxicity , Disease Models, Animal , Drug Therapy, Combination , Female , Lamivudine/toxicity , Maternal Exposure , Mitochondria, Liver/metabolism , Mitochondria, Liver/ultrastructure , Oxidative Phosphorylation/drug effects , Pregnancy , Stavudine/toxicity , Zidovudine/toxicity
9.
Curr HIV Res ; 8(3): 232-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20158454

ABSTRACT

BACKGROUND: Peripheral neuropathy is the dose-limiting toxicity of stavudine and didanosine (nucleoside analogs used in HIV treatment) and is attributed to mitochondrial toxicity from these drugs. Acetyl L-carnitine (ALC) and co-enzyme Q(10) are proposed as neuropathy treatments, but evidence to support these is limited. METHODS: We examined ALC and a water-soluble formulation of co-enzyme Q(10) (H(Q)O) for the prevention of d4T and ddI neurotoxicity using cultured fetal rat DRG as an in vitro model. RESULTS: DdI (33microM) and d4T (50microM) caused clear toxicity (impaired neurite growth) by day 8 of DRG culture. H(Q)O at concentrations 1-100microM completely prevented the toxicity of 33microM ddI in vitro and ALC at concentrations 1-100 microM substantially (but incompletely) prevented ddI toxicity in this model. In contrast, ALC was ineffective at all concentrations tested for preventing the toxicity of 50microM d4T. H(Q)O showed dose-dependent efficacy for preventing d4T toxicity. H(Q)O (1microM) partially prevented d4T toxicity while 10 and 100microM H(Q)O completely prevented d4T toxicity in this model. CONCLUSIONS: We find H(Q)O is superior to ALC for preventing the neurotoxicity of d4T (the HIV treatment most associated with neuropathy) and ddI in vitro. Further study is needed to clarify any clinical role for co-enzyme Q(10) co-administration with d4T and ddI and to assess whether this compound may have a role in treating established cases of neuropathy.


Subject(s)
Anti-Retroviral Agents/toxicity , Ubiquinone/analogs & derivatives , Vitamins/pharmacology , AIDS-Associated Nephropathy/drug therapy , AIDS-Associated Nephropathy/prevention & control , Acetylcarnitine/pharmacology , Animals , Cell Survival , Didanosine/toxicity , Ganglia, Spinal/drug effects , Neurons/drug effects , Organ Culture Techniques , Rats , Stavudine/toxicity , Ubiquinone/pharmacology
10.
J Acquir Immune Defic Syndr ; 51(3): 258-63, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19582894

ABSTRACT

OBJECTIVES: Tenofovir disoproxil fumarate (tenofovir DF) use has been associated with renal dysfunction and Fanconi syndrome. Tenofovir is taken up into renal tubules by anion transporters where high intracellular drug concentration may induce a functionally relevant depletion of mitochondrial DNA (mtDNA). We investigated if tenofovir may induce renal mtDNA depletion and respiratory chain dysfunction. METHODS: Rats (n = 8) were gavaged daily with 100 mg x kg(-1) x d(-1) of tenofovir DF or didanosine. Kidneys and livers were examined after 8 weeks of treatment. RESULTS: The tenofovir group had significantly lower body and kidney weights than rats exposed to water or didanosine. Proximal but not distal tubules were of increased diameter and contained small lipid droplets. Tubular mitochondria were enlarged, and their crystal architecture was disrupted. Tenofovir-exposed kidneys contained low mtDNA copy numbers and impaired expression of mtDNA-encoded cytochrome c oxidase (COX) I but not nucleus-encoded COX IV subunits. Histochemistry demonstrated low tubular COX and nicotinamide adenine dinucleotide dehydrogenase (NADH-DH) activities, whereas succinate dehydrogenase activity was preserved. COX activity was preserved in the glomeruli of tenofovir-exposed rats. Didanosine did not elicit renal effects but, unlike tenofovir, depleted mtDNA in liver (by 52%). CONCLUSIONS: Tenofovir DF induces an organ-specific nephrotoxicity with mtDNA depletion and dysfunction of mtDNA-encoded respiratory chain subunits. The data do not support nephrotoxicity of didanosine.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/toxicity , DNA, Mitochondrial/drug effects , Kidney Tubules, Proximal/drug effects , Mitochondria/drug effects , Mitochondrial Diseases/chemically induced , Organophosphonates/toxicity , Reverse Transcriptase Inhibitors/toxicity , Adenine/toxicity , Animals , Didanosine/toxicity , Electron Transport/drug effects , Electron Transport Complex IV/drug effects , Electron Transport Complex IV/metabolism , Kidney Tubules, Proximal/ultrastructure , Male , Microscopy, Electron, Transmission , Mitochondria/ultrastructure , Mitochondrial Diseases/pathology , NADH Dehydrogenase/drug effects , NADH Dehydrogenase/metabolism , Rats , Rats, Sprague-Dawley , Tenofovir
11.
Environ Mol Mutagen ; 50(6): 460-72, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19334055

ABSTRACT

The success of nucleoside reverse transcriptase inhibitors (NRTIs) in treating HIV-1 infection and reducing mother-to-child transmission of the virus during pregnancy is accompanied by evidence that NRTIs cause long-term health risks for cancer and mitochondrial disease. Thus, agents that mitigate toxicities of the current combination drug therapies are needed. Previous work had shown that the NRTI-drug pair zidovudine (AZT)-didanosine (ddI) was highly cytotoxic and mutagenic; thus, we conducted preliminary studies to investigate the ability of the active moiety of amifostine, WR1065, to protect against the deleterious effects of this NRTI-drug pair. In TK6 cells exposed to 100 muM AZT-ddI (equimolar) for 3 days with or without 150 muM WR1065, WR1065 enhanced long-term cell survival and significantly reduced AZT-ddI-induced mutations. Follow-up studies were conducted to determine if coexposure to AZT and WR1065 abrogated the antiretroviral efficacy of AZT. In human T-cell blasts infected with HIV-1 in culture, inhibition of p24 protein production was observed in cells treated with 10 muM AZT in the absence or presence of 5-1,000 muM WR1065. Surprisingly, WR1065 alone exhibited dose-related inhibition of HIV-1 p24 protein production. WR1065 also had antiviral efficacy against three species of adenovirus and influenza A and B. Intracellular levels of unbound WR1065 were measured following in vitro/in vivo drug exposure. These pilot study results indicate that WR1065, at low intracellular levels, has cytoprotective and antimutagenic activities against the most mutagenic pair of NRTIs and has broad spectrum antiviral effects. These findings suggest that the activities have a possible common mode of action that merits further investigation.


Subject(s)
Didanosine/analogs & derivatives , Dideoxynucleotides/toxicity , Mercaptoethylamines/pharmacology , Mutagenesis/drug effects , Virus Replication/drug effects , Zidovudine/analogs & derivatives , Adenoviridae/drug effects , Adenoviridae/physiology , Cell Line , Cytoplasm/drug effects , Cytoplasm/metabolism , Didanosine/toxicity , Dose-Response Relationship, Drug , HIV Core Protein p24/metabolism , HIV-1/drug effects , HIV-1/physiology , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Influenza A virus/drug effects , Influenza A virus/physiology , Influenza B virus/drug effects , Influenza B virus/physiology , Intracellular Space/drug effects , Intracellular Space/metabolism , Lymphocytes/drug effects , Lymphocytes/virology , Mutation/genetics , Phytohemagglutinins/pharmacology , Serotyping , Time Factors , Zidovudine/toxicity
12.
Environ Mol Mutagen ; 49(4): 312-7, 2008 May.
Article in English | MEDLINE | ID: mdl-18366095

ABSTRACT

Antiretroviral therapies based on nucleoside reverse transcriptase inhibitors, like zidovudine (3'-azido-3'-deoxythymidine; AZT) and didanosine (2',3'-dideoxyinosine; ddI), markedly reduce human immunodeficiency virus loads. The Somatic Mutation And Recombination Test in Drosophila melanogaster (wing SMART), in its standard version, was applied to compare AZT and ddI genetic toxicity expressed as point and chromosomal mutation as well as homologous mitotic recombination. The present findings provide evidence that the mechanistic basis underlying the genetic toxicity of these antiretrovirals is mainly related to mitotic recombination. However, a genotoxic pattern can correspondingly be discerned: AZT is able to induce recombination ( approximately 85%) and mutation ( approximately 15%), and ddI causes only homologous recombination (100%) in the wing SMART assay. Another point to be considered is the fact that ddI is 3.8 times less active to induce mutant clones per mg/ml unit as compared to AZT. The clinical significance of these observations has to be interpreted in the light of data obtained from long-term toxicity in patients treated with the above mentioned agents.


Subject(s)
Anti-HIV Agents/toxicity , Didanosine/toxicity , Mutagens/toxicity , Reverse Transcriptase Inhibitors/toxicity , Wings, Animal/drug effects , Zidovudine/toxicity , Animals , Drosophila melanogaster/anatomy & histology , Drosophila melanogaster/genetics , Female , Male , Mitosis , Mutagenicity Tests , Mutation , Recombination, Genetic , Wings, Animal/abnormalities , Wings, Animal/cytology
13.
J Antimicrob Chemother ; 61(6): 1340-3, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18339636

ABSTRACT

OBJECTIVES: Stavudine is widely used in Thailand and is associated with mitochondrial toxicity. Here, we evaluated the effect of switching from stavudine/didanosine to tenofovir/lamivudine on measures of metabolic and mitochondrial toxicity in Thai patients. METHODS: Thirty-five Thai patients with full HIV RNA suppression were switched from stavudine/didanosine to tenofovir/lamivudine while receiving saquinavir/ritonavir 1600/100 mg once daily. Patients were assessed at the time of switch and 24 and 48 weeks after for lipids, liver enzymes, lactate, mitochondrial DNA content and limb/total fat mass by dual energy X-ray absorptiometry (DEXA) scanning. RESULTS: Forty-eight weeks after the switch, there were significant reductions in lipids and lactate, but no change in liver enzymes. There was reversal of lipoatrophy, as shown by rises in limb fat mass (+0.38 kg, P = 0.006) and total fat mass (+0.69 kg, P = 0.02) on DEXA scan. Patients perceived weight improvement, but did not report reversal of lipoatrophy of individual body parts. The mitochondrial DNA/nuclear DNA ratio rose (+1.06, P < 0.0001). CONCLUSIONS: After the nucleoside reverse transcriptase inhibitor switch, reversal of mitochondrial toxicity was consistent with switch studies of mainly Caucasian patients, although the peripheral mononuclear cell mitochondrial DNA rise exceeded previous reports.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/toxicity , Didanosine/toxicity , Lamivudine/toxicity , Organophosphonates/toxicity , Stavudine/toxicity , Absorptiometry, Photon , Adenine/toxicity , Adult , Anti-HIV Agents/administration & dosage , Body Fat Distribution , DNA, Mitochondrial/analysis , HIV Infections/drug therapy , Humans , Lipids/blood , Liver Function Tests , RNA, Viral/blood , Ritonavir/administration & dosage , Saquinavir/administration & dosage , Tenofovir , Thailand
14.
Cell Biol Toxicol ; 24(1): 113-21, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17619152

ABSTRACT

The purpose of the present study was to investigate whether didanosine (ddI) directly causes morphological and ultrastructural abnormalities of dorsal root ganglion (DRG) neurons in vitro. Dissociated DRG cells and organotypic DRG explants from embryonic 15-day-old Wistar rats were cultured for 3 days and then exposed to ddI (1 microg/ml, 5 microg/ml, 10 microg/ml, and 20 microg/ml) for another 3 days and 6 days, respectively. Neurons cultured continuously in medium served as normal controls. The diameter of the neuronal cell body and neurite length were measured in dissociated DRG cell cultures. Neuronal ultrastructural changes were observed in both culture models. ddI induced dose-dependent decreases in neurite number, length of the longest neurite in each neuron, and total neurite length per neuron in dissociated DRG cell cultures with 3 days treatment. There were no morphological changes seen in organotypic DRG cultures even with longer exposure time (6 days). But ddI induced ultrastructural changes in both culture models. Ultrastructural abnormalities included loss of cristae in mitochondria, clustering of microtubules and neurofilaments, accumulation of glycogen-like granules, and emergence of large dense particles between neurites or microtubules. Lysosome-like large particles emerged inconstantly in neurites. ddI induced a neurite retraction or neurite loss in a dose-dependent manner in dissociated DRG neurons, suggesting that ddI may partially contribute to developing peripheral neuropathy. Cytoskeletal rearrangement and ultrastructural abnormalities caused by ddI in both culture models may have a key role in neurite degeneration.


Subject(s)
Didanosine/toxicity , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Neurons/drug effects , Toxicity Tests , Animals , Cell Shape/drug effects , Cells, Cultured , Ganglia, Spinal/ultrastructure , Neurites/drug effects , Neurites/ultrastructure , Neurons/cytology , Rats , Rats, Wistar
15.
Brain ; 130(Pt 8): 2011-23, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17616550

ABSTRACT

Antiretroviral toxic neuropathy (ATN) has become a common peripheral neuropathy among HIV/AIDS patients, for which the underlying pathogenesis is uncertain. Indeed, no models exist for ATN that assess the interaction between retroviral infection and antiretroviral therapy. Herein, we developed ex vivo and in vivo models of ATN induced by didanosine (ddI) following infection by the lentivirus, feline immunodeficiency virus (FIV), permitting us to address the working hypothesis that ddI mediates ATN through mitochondrial injury in neurons. We investigated neuronal morphology, neurobehavioural testing, viral load, mitochondrial and neurotrophic factor gene expression after ddI treatment of FIV-infected and uninfected animals or dorsal root ganglia (DRG) cultures. ddI caused concentration-dependent neuronal injury in cultured feline DRGs (P < 0.05), together with reduced viral replication and diminished expression of mitochondrial cytochrome C oxidase subunit I gene (mtCOX I) and the neurotrophin, brain-derived neurotrophic factor (BDNF). Indeed, BDNF treatment reversed neuronal injury caused by FIV infection in the presence or absence of ddI exposure (P < 0.05). In vivo FIV infection revealed delays in withdrawal latency to a noxious stimulus, which were exacerbated by ddI treatment. Epidermal density of nerve endings was reduced after FIV infection (P < 0.05), especially with ddI treatment. Although viral replication in blood was suppressed in ddI-treated animals (P < 0.05), ddI had a limited effect on viral abundance in DRGs of the same animals. ddI decreased mtCOX I expression in DRG neurons of FIV-infected animals (P < 0.05). BDNF expression was downregulated by ddI in DRG Schwann cells following FIV infection. Thus, ddI treatment during FIV infection resulted in additive pathogenic effects contributing to the development of ATN, which was associated with mitochondrial injury on neurons and reduced BDNF production by Schwann cells in DRGs, highlighting the convergent pathogenic effects that antiretroviral drugs might have in patients with HIV infection.


Subject(s)
Anti-HIV Agents/toxicity , Brain-Derived Neurotrophic Factor/metabolism , Didanosine/toxicity , Feline Acquired Immunodeficiency Syndrome/drug therapy , Mitochondria/drug effects , Peripheral Nervous System Diseases/chemically induced , Animals , Blotting, Western , Cats , Cells, Cultured , Disease Models, Animal , Feline Acquired Immunodeficiency Syndrome/metabolism , Feline Acquired Immunodeficiency Syndrome/virology , Ganglia, Spinal/drug effects , Mitochondria/pathology , Peripheral Nervous System Diseases/metabolism , Polymerase Chain Reaction/methods , Viral Load
16.
Antimicrob Agents Chemother ; 51(7): 2531-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17470651

ABSTRACT

Nucleoside analogs are associated with various mitochondrial toxicities, and it is becoming increasingly difficult to accommodate these differences solely in the context of DNA polymerase gamma inhibition. Therefore, we examined the toxicities of zidovudine (AZT) (10 and 50 microM; 2.7 and 13.4 microg/ml), didanosine (ddI) (10 and 50 microM; 2.4 and 11.8 microg/ml), and zalcitabine (ddC) (1 and 5 microM; 0.21 and 1.1 microg/ml) in HepG2 and H9c2 cells without the presumption of mitochondrial DNA (mtDNA) depletion. Ethidium bromide (EtBr) (0.5 microg/ml; 1.3 microM) was used as a positive control. AZT treatment resulted in metabolic disruption (increased lactate and superoxide) and increased cell mortality with decreased proliferation, while mtDNA remained unchanged or increased (HepG2 cells; 50 microM AZT). ddC caused pronounced mtDNA depletion in HepG2 cells but not in H9c2 cells and increased mortality in HepG2 cells, but no significant metabolic disruption in either cell type. ddI caused a moderate depletion of mtDNA in both cell types but showed no other effects. EtBr exposure resulted in metabolic disruption, increased cell mortality with decreased cell proliferation, and mtDNA depletion in both cell types. We conclude that nucleoside analogs display unique toxicities within and between culture models, and therefore, care should be taken when generalizing about the mechanisms of nucleoside reverse transcriptase inhibitor toxicity. Additionally, mtDNA abundance does not necessarily correlate with metabolic disruption, especially in cell culture; careful discernment is recommended in this regard.


Subject(s)
Antimetabolites/toxicity , Didanosine/toxicity , Mitochondria/drug effects , Zalcitabine/toxicity , Zidovudine/toxicity , Animals , Carcinoma, Hepatocellular/metabolism , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA, Mitochondrial/metabolism , Dose-Response Relationship, Drug , Humans , Lactates/metabolism , Liver Neoplasms/metabolism , Myocytes, Cardiac/metabolism , Rats , Superoxides/metabolism
17.
Environ Mol Mutagen ; 48(3-4): 239-47, 2007.
Article in English | MEDLINE | ID: mdl-17358029

ABSTRACT

Experiments were performed to investigate the impact of didanosine (ddI), lamivudine (3TC), and stavudine (d4T) on cell survival and mutagenicity in two reporter genes, hypoxanthine-guanine phosphoribosyltransferase (HPRT) and thymidine kinase (TK), using a cell cloning assay for assessing the effects of individual nucleoside analogs (NRTIs)/drug combinations in human TK6 B-lymphoblastoid cells. Three-day treatments with 0, 33, 100, or 300 microM ddI, 3TC, or ddI-3TC produced positive trends for increased HPRT and TK mutant frequencies. While dose-related trends were too small to reach significance after treatments with d4T or d4T-3TC, pairwise comparisons with control cells indicated that exposure to 100 microM d4T or d4T-3TC caused significant elevations in HPRT mutants. Measurements of mutagenicity in cells exposed to d4T (or d4T-3TC) were complicated by the cytotoxicity of this NRTI. Enhanced increases in mutagenic responses to combined NRTI treatments, compared with single drug treatments, occurred as additive to synergistic effects in the HPRT gene of cells exposed to 100 microM ddI-3TC or 100 microM d4T-3TC, and in the TK gene of cells exposed to 100 or 300 microM ddI-3TC. Comparisons of these data to mutagenicity studies of other NRTIs in the same system (Meng Q et al. [2000c]: Proc Natl Acad Sci USA 97:12667-126671; Torres SM et al. [2007]: Environ Mol Mutagen) indicate that the relative mutagenic potencies for all drugs tested to date are: AZT-ddI > ddI-3TC > AZT-3TC congruent with AZT-3TC-ABC (abacavir) > AZT >/=ddI > d4T-3TC > 3TC > d4T >/= ABC. These collective data suggest that all NRTIs with antiviral activity against HIV-1 may cause host cell DNA damage and mutations, and impose a cancer risk.


Subject(s)
Didanosine/toxicity , Lamivudine/toxicity , Mutagens/toxicity , Reverse Transcriptase Inhibitors/toxicity , Stavudine/toxicity , Anti-HIV Agents/toxicity , Cell Line , Drug Interactions , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Mutagenicity Tests , Mutation , Thymidine Kinase/genetics
18.
Antimicrob Agents Chemother ; 50(11): 3824-32, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16940060

ABSTRACT

We assessed the in vitro toxicity of tenofovir (TFV) and compared it with those of zidovudine (AZT), didanosine (ddI), ritonavir (RTV), and lopinavir (LPV) alone and in combination in human renal proximal tubule epithelial cells (RPTECs). The cells were treated with various concentrations and combinations of the tested antiretrovirals for up to 22 days, and cytotoxicity was determined. In addition, we assessed the levels of mitochondrial DNA (mtDNA) and cytochrome oxidase II (COII) mRNA in RPTECs treated with reverse transcriptase inhibitors. TFV alone was not associated with significant cytotoxicity. ddI showed pronounced cytotoxicity that was greater than those of AZT (P = 0.002) and TFV (P = 0.0001). The combination of 10 muM RTV and 40 muM LPV significantly reduced RPTEC viability (P < 0.0001), and TFV tended to partially reduce this effect. TFV alone affected neither mtDNA nor COII mRNA levels, whereas ddI caused a profound depletion of mtDNA and a parallel reduction in COII mRNA expression. The effects of ddI, but not those of AZT, on mtDNA and COII mRNA were further enhanced in the presence of TFV, a finding consistent with the inhibition of ddI clearance by TFV. The addition of TFV to ddI or AZT appeared to slightly increase the COII mRNA/mtDNA ratio relative to that in cells treated with ddI or AZT alone. Together, these in vitro results indicate that combination with other antiretrovirals does not significantly increase the toxic potential of TFV in RPTECs.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/toxicity , Kidney Tubules, Proximal/cytology , Kidney Tubules, Proximal/drug effects , Mitochondria/drug effects , Organophosphonates/toxicity , Adenine/toxicity , Cell Survival/drug effects , Cells, Cultured , DNA, Mitochondrial/drug effects , Didanosine/toxicity , Drug Interactions , Electron Transport Complex IV/biosynthesis , Humans , Lopinavir , Pyrimidinones/toxicity , RNA/biosynthesis , Ritonavir/toxicity , Tenofovir , Zidovudine/toxicity
19.
Birth Defects Res B Dev Reprod Toxicol ; 77(3): 207-15, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16767756

ABSTRACT

BACKGROUND: In pregnant women, antiretroviral drugs improve maternal health and reduce vertical transmission of human immunodeficiency virus to the infant. However, few nonclinical studies have examined the potential for adverse drug interactions. METHODS: On gestational days (GD) 6-16, mice were dosed with vehicle, ddI (360, 1440, or 2,880 mg/kg/day, p.o.), d4T (60, 240, or 480), or ddI/d4T combinations (360/60, 1,440/240, or 2,880/480). Daily doses were divided into two equal parts that were administered >or=6-hr apart. Body weight, clinical signs, and feed consumption were monitored. Pregnancies (22-24/group) were confirmed at necropsy. Maternal liver and gravid uterine weights (GUW), uterine implants (resorption, live or dead fetus), fetal body weight, gender, and morphologic anomalies (external, visceral, skeletal) were recorded. RESULTS: Maternal body weight, clinical signs, and GUW were unaffected. Maternal weight change corrected for GUW was greater than controls at 60 and 480 d4T. Relative feed consumption during treatment was increased relative to controls at 1,440 and 2,880 ddI and 2,880/480 ddI/d4T. Relative maternal liver weight was elevated above controls at 240 and 480 d4T and 2,880/480 ddI/d4T, and above the constituent dose of ddI at 1,440/240 and 2,880/480 ddI/d4T. Liver weight was not affected by ddI and there was no significant drug interaction. Prenatal mortality and morphologic anomalies were not increased. Fetal body weight showed only a decreasing trend for ddI/d4T, no effect for ddI or d4T, and no statistically significant drug interaction. CONCLUSIONS: In pregnant mice, ddI/d4T combinations were not associated with well-defined developmental toxicity or adverse drug interactions.


Subject(s)
Didanosine/administration & dosage , Didanosine/toxicity , Fetus/drug effects , Fetus/embryology , Stavudine/administration & dosage , Stavudine/toxicity , Animals , Animals, Outbred Strains , Embryo Implantation/drug effects , Feeding Behavior/drug effects , Female , Fetal Death , Fetal Weight/drug effects , Fetus/abnormalities , Litter Size/drug effects , Mice , Organ Size/drug effects , Pregnancy
20.
Antivir Ther ; 10(2): 327-34, 2005.
Article in English | MEDLINE | ID: mdl-15865227

ABSTRACT

OBJECTIVE: To evaluate if nucleoside analogue reverse transcriptase inhibitors (NRTIs) and polymerase-gamma inhibitors deplete mitochondrial DNA (mtDNA) in cultured primary lymphocytes and if such depletion might be associated with functional defects. METHODS: Primary peripheral blood CD4 and CD8 lymphocytes were purified from six healthy humans (three male and three female), stimulated mitotically (CD3/CD28) and cultured for 10 days in the presence or absence of NRTIs. Lymphocyte proliferation, mtDNA content, the expression of mtDNA-encoded cytochrome c-oxidase II (COXII) and lactate production were assessed. RESULTS: In CD4 lymphocytes, 10-day exposure to zalcitabine (1.77 microM), didanosine (118 microM) and stavudine (361 microM) induced a time-dependent decline of mtDNA. Compared with controls, residual mtDNA levels were 25%, 21% and 40%, respectively. COXII was reduced to 55%, 35% and 70% of control values. Lactic acid production was increased (by 214%, 294% and 175%, respectively). At day 10, lymphocyte counts were reduced (to 60%, 51%, and 41%, respectively). Zidovudine (71 microM) also reduced lymphocyte counts to 34% and increased lactic acid production by 170%, but did not induce mtDNA and COXII depletion. All these changes were highly significant. Lower NRTI concentrations (0.177 microM of zalcitabine, 11.8 microM of didanosine, 3.6 microM of stavudine and 7.1 microM of zidovudine) had effects at the border of significance. Similar observations were made in CD8 lymphocytes. CONCLUSIONS: In human lymphocytes, zalcitabine, didanosine and stavudine induce dose- and time-dependent mtDNA depletion, which is associated with decreased cell proliferation and increased lactate production. Zidovudine impairs lymphocyte division without inducing mtDNA depletion.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , DNA, Mitochondrial/drug effects , Didanosine/toxicity , Reverse Transcriptase Inhibitors/toxicity , Stavudine/toxicity , Zalcitabine/toxicity , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , DNA, Mitochondrial/metabolism , Female , Humans , Lactic Acid/biosynthesis , Lactic Acid/metabolism , Lymphocyte Activation , Male , Mitochondrial Diseases/chemically induced
SELECTION OF CITATIONS
SEARCH DETAIL