Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 82
Filter
1.
ACS Chem Neurosci ; 12(14): 2619-2628, 2021 07 21.
Article in English | MEDLINE | ID: mdl-34184863

ABSTRACT

Neuropathic pain is one of the foremost adverse effects that worsens quality of life for patients undergoing an antiretroviral treatment. Currently, there are no effective analgesics for relieving it; thus, there is an urgent need to develop novel treatments for neuropathic pain. Previously, we described and validated F11 cells as a model of DRG (dorsal root ganglia) neurons. In the current work, we employed F11 cells to identify regulators of antiretroviral-induced neuropathic pain combining functional and transcriptomic analysis. The antiretroviral zalcitabine (ddC) increased the excitability of differentiated F11 cells associated with calcium signaling without morphological changes in the neuronal phenotype, mimicking the observed increase of painful signaling in patients suffering from antiretroviral-induced neuropathic pain. Employing RNA sequencing, we observed that zalcitabine treatment upregulated genes related with oxidative stress and calcium homeostasis. The functional impact of the transcriptomic changes was explored, finding that the exposure to zalcitabine significantly increased intracellular oxidative stress and reduced store-operated calcium entry (SOCE). Because the functional and transcriptomic evidence points toward fundamental changes in calcium signaling and oxidative stress upon zalcitabine exposure, we identified that NAD(P)H quinone dehydrogenase and the sarcoplasmic/endoplasmic reticulum calcium ATPase 3 were involved in zalcitabine-induced hyperexcitability of F11 cells. Overexpression of those genes increases the calcium-elicited hyperexcitability response and reduces SOCE, as well as increases intracellular ROS levels. These data do not only mimic the effects of zalcitabine but also highlight the relevance of oxidative stress and of calcium-mediated signaling in antiretroviral-induced hyperexcitability of sensory neurons, shedding light on new therapeutic targets for antiviral-induced neuropathic pain.


Subject(s)
Neuralgia , Zalcitabine , Animals , Calcium Signaling , Disease Models, Animal , Ganglia, Spinal , Humans , Hyperalgesia , Neuralgia/chemically induced , Neuralgia/drug therapy , Quality of Life , Sensory Receptor Cells , Zalcitabine/toxicity
2.
J Biol Chem ; 296: 100206, 2021.
Article in English | MEDLINE | ID: mdl-33334881

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) were the first drugs used to treat human immunodeficiency virus infection, and their use can cause mitochondrial toxicity, including mitochondrial DNA (mtDNA) depletion in several cases. The first-generation NRTIs, including 2',3'-dideoxycytidine (ddC), were originally and are still pursued as anticancer agents. NRTI-sensitive DNA polymerases localizing to mitochondria allow for the opportunity to poison proliferating cancer cell mtDNA replication as certain cancers rely heavily on mitochondrial functions. However, mtDNA replication is independent of the cell cycle creating a significant concern that toxicants such as ddC impair mtDNA maintenance in both proliferating and nonproliferating cells. To examine this possibility, we tested the utility of the HepaRG cell line to study ddC-induced toxicity in isogenic proliferating (undifferentiated) and nonproliferating (differentiated) cells. Following ddC exposures, we measured cell viability, mtDNA copy number, and mitochondrial bioenergetics utilizing trypan blue, Southern blotting, and extracellular flux analysis, respectively. After 13 days of 1 µM ddC exposure, proliferating and differentiated HepaRG harbored mtDNA levels of 0.9% and 17.9% compared with control cells, respectively. Cells exposed to 12 µM ddC contained even less mtDNA. By day 13, differentiated cell viability was maintained but declined for proliferating cells. Proliferating HepaRG bioenergetic parameters were severely impaired by day 8, with 1 and 12 µM ddC, whereas differentiated cells displayed defects of spare and maximal respiratory capacities (day 8) and proton-leak linked respiration (day 14) with 12 µM ddC. These results indicate HepaRG is a useful model to study proliferating and differentiated cell mitochondrial toxicant exposures.


Subject(s)
DNA Replication/drug effects , Hepatocytes/drug effects , Mitochondria/drug effects , Reverse Transcriptase Inhibitors/toxicity , Zalcitabine/toxicity , Cell Differentiation/drug effects , Cell Line, Transformed , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA Copy Number Variations , DNA, Mitochondrial/antagonists & inhibitors , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Energy Metabolism/drug effects , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Inhibitory Concentration 50 , Mitochondria/genetics , Mitochondria/metabolism
3.
J Mol Neurosci ; 71(3): 651-661, 2021 Mar.
Article in English | MEDLINE | ID: mdl-32812184

ABSTRACT

Highly active antiretroviral therapy (HAART) works effectively in inhibiting HIV replication in patients. However, the use of nucleoside reverse transcriptase inhibitors (NRTIs) often causes side effects of neuropathic pain, and its mechanism remains to be elucidated. Therefore, we aim to explore the mechanism of NRTIs-induced neuropathic pain at the transcriptome level. C57BL/6 J mice were given intraperitoneal injection of zalcitabine (ddC) or saline (control) for 2 weeks, during which the mechanical pain threshold of the mice was detected by von Frey test. Then the L3~L5 spinal segments of the mice were isolated and subsequently used for RNA sequencing (RNA-seq) on the last day of treatment. The mechanical pain threshold of mice given ddC decreased significantly. Compared with the control group, ddC caused significant changes in the expression of 135 genes, of which 66 upregulated and 69 downregulated. Enrichment analysis showed that the functions of these genes are mainly enriched in regulation of transcription, multicellular organism development, and cell differentiation, and the pathway is mainly enriched in the cGMP-PKG signaling pathway and AMPK signaling pathway. Furthermore, key genes such as Gabrd, Kcnd3, Npcd, Insr, Lypd6, Scd2, and Mef2d were also identified. These may serve as drug targets for the prevention or treatment of NRTI-induced neuropathic pain.


Subject(s)
Neuralgia/genetics , Spinal Cord/metabolism , Transcriptome , Animals , Male , Mice , Mice, Inbred C57BL , Neuralgia/etiology , Neuralgia/metabolism , Reverse Transcriptase Inhibitors/toxicity , Signal Transduction , Zalcitabine/toxicity
4.
Purinergic Signal ; 14(1): 47-58, 2018 03.
Article in English | MEDLINE | ID: mdl-29159762

ABSTRACT

The direct neurotoxicity of HIV and neurotoxicity of combination antiretroviral therapy medications both contribute to the development of neuropathic pain. Activation of satellite glial cells (SGCs) in the dorsal root ganglia (DRG) plays a crucial role in mechanical and thermal hyperalgesia. The P2Y12 receptor expressed in SGCs of the DRG is involved in pain transmission. In this study, we explored the role of the P2Y12 receptor in neuropathic pain induced by HIV envelope glycoprotein 120 (gp120) combined with ddC (2',3'-dideoxycytidine). A rat model of gp120+ddC-induced neuropathic pain was used. Peripheral nerve exposure to HIV-gp120+ddC increased mechanical and thermal hyperalgesia in gp120+ddC-treated model rats. The gp120+ddC treatment increased expression of P2Y12 receptor mRNA and protein in DRG SGCs. In primary cultured DRG SGCs treated with gp120+ddC, the levels of [Ca2+]i activated by the P2Y12 receptor agonist 2-(Methylthio) adenosine 5'-diphosphate trisodium salt (2-MeSADP) were significantly increased. P2Y12 receptor shRNA treatment inhibited 2-MeSADP-induced [Ca2+]i in primary cultured DRG SGCs treated with gp120+ddC. Intrathecal treatment with a shRNA against P2Y12 receptor in DRG SGCs reduced the release of pro-inflammatory cytokines, decreased phosphorylation of p38 MAPK in the DRG of gp120+ddC-treated rats. Thus, downregulating the P2Y12 receptor relieved mechanical and thermal hyperalgesia in gp120+ddC-treated rats.


Subject(s)
HIV Envelope Protein gp120 , Neuralgia/metabolism , Neuroglia/metabolism , Receptors, Purinergic P2/metabolism , Zalcitabine/toxicity , Animals , Anti-HIV Agents/toxicity , Ganglia, Spinal/metabolism , HIV Infections/complications , Hyperalgesia/metabolism , Hyperalgesia/virology , Male , Neuralgia/etiology , Rats , Rats, Sprague-Dawley , Receptors, Purinergic P2Y12 , Up-Regulation
5.
Exp Neurol ; 281: 17-27, 2016 07.
Article in English | MEDLINE | ID: mdl-27090160

ABSTRACT

Human immunodeficiency virus (HIV) patients treated with nucleoside reverse transcriptase inhibitors (NRTIs), have been known to develop neuropathic pain. While there has been a major shift away from some neurotoxic NRTIs in current antiretroviral therapy, a large number of HIV patients alive today have previously received them, and many have developed painful peripheral neuropathy. The exact mechanisms by which HIV with NRTIs contribute to the development of neuropathic pain are not known. Previous studies suggest that cytoplasmic polyadenylation element-binding protein (CPEB), reactive oxygen species (ROS), and cAMP-response element-binding protein (CREB)-binding protein (CBP), are involved in the neuroimmunological diseases including inflammatory/neuropathic pain. In this study, we investigated the role of CPEB, mitochondrial ROS (mtROS), or CBP in neuropathic pain induced by HIV envelope protein gp120 combined with antiretroviral drug. The application of recombinant gp120 into the sciatic nerve plus systemic ddC (one of NRTIs) induced mechanical allodynia. Knockdown of CPEB or CBP using intrathecal antisense oligodeoxynucleotide (AS-ODN) reduced mechanical allodynia. Intrathecal mitochondrial superoxide scavenger mito-tempol (Mito-T) increased mechanical withdrawal threshold. Knockdown of CPEB using intrathecal AS-ODN, reduced the up-regulated mitochondrial superoxide in the spinal dorsal horn in rats with gp120 combined with ddC. Intrathecal Mito-T lowered the increased expression of CBP in the spinal dorsal horn. Immunostaining studies showed that neuronal CPEB positive cells were co-localized with MitoSox positive profiles, and that MitoSox positive profiles were co-localized with neuronal CBP. Our studies suggest that neuronal CPEB-mtROS-CBP pathway in the spinal dorsal horn, plays an important role in the gp120/ddC-induced neuropathic pain in rats.


Subject(s)
Neuralgia/metabolism , Signal Transduction/physiology , Spinal Cord/metabolism , Transcription Factors/metabolism , mRNA Cleavage and Polyadenylation Factors/metabolism , Animals , Anti-HIV Agents/toxicity , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , HIV Envelope Protein gp120/toxicity , Humans , Hyperalgesia/physiopathology , Male , Membrane Proteins/metabolism , Neuralgia/chemically induced , Neuralgia/drug therapy , Oligodeoxyribonucleotides, Antisense/therapeutic use , Pain Management , Pain Threshold/drug effects , Pain Threshold/physiology , Phosphoproteins/metabolism , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Spinal Cord/pathology , Succinate Dehydrogenase/metabolism , Transcription Factors/chemistry , Zalcitabine/toxicity , mRNA Cleavage and Polyadenylation Factors/chemistry
6.
EMBO J ; 34(14): 1959-70, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26056153

ABSTRACT

The human DNA polymerase gamma (Pol γ) is responsible for DNA replication in mitochondria. Pol γ is particularly susceptible to inhibition by dideoxynucleoside-based inhibitors designed to fight viral infection. Here, we report crystal structures of the replicating Pol γ-DNA complex bound to either substrate or zalcitabine, an inhibitor used for HIV reverse transcriptase. The structures reveal that zalcitabine binds to the Pol γ active site almost identically to the substrate dCTP, providing a structural basis for Pol γ-mediated drug toxicity. When compared to the apo form, Pol γ undergoes intra- and inter-subunit conformational changes upon formation of the ternary complex with primer/template DNA and substrate. We also find that the accessory subunit Pol γB, which lacks intrinsic enzymatic activity and does not contact the primer/template DNA directly, serves as an allosteric regulator of holoenzyme activities. The structures presented here suggest a mechanism for processivity of the holoenzyme and provide a model for understanding the deleterious effects of Pol γ mutations in human disease. Crystal structures of the mitochondrial DNA polymerase, Pol γ, in complex with substrate or antiviral inhibitor zalcitabine provide a basis for understanding Pol γ-mediated drug toxicity.


Subject(s)
DNA-Directed DNA Polymerase/chemistry , DNA-Directed DNA Polymerase/metabolism , Zalcitabine/toxicity , Amino Acid Sequence , Base Sequence , Binding Sites , Catalytic Domain , Crystallography, X-Ray , DNA Polymerase gamma , DNA, Mitochondrial/metabolism , DNA-Directed DNA Polymerase/genetics , Humans , Models, Molecular , Molecular Sequence Data , Mutation , Protein Conformation , Reverse Transcriptase Inhibitors/chemistry , Reverse Transcriptase Inhibitors/metabolism , Reverse Transcriptase Inhibitors/toxicity , Zalcitabine/chemistry , Zalcitabine/metabolism
7.
Pain ; 156(9): 1729-1736, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25932691

ABSTRACT

Painful peripheral neuropathy due to the antiretroviral therapy used to treat HIV is one of the most prevalent side effects occurring in at least 30% of patients living with this infection. We have evaluated the electrophysiological and behavioral effects of d4T and ddC on peripheral large and small nerve fibers in male rats treated with d4T (Sprague-Dawley, 50 mg/kg, twice within 1 week), ddC (Wistar, 50 mg/kg, 3 times per week for 3 weeks), or vehicle. The effect of the interventions was assessed using behavioral measures of mechanical sensitivity, conventional nerve conduction studies, and microneurographic single nerve C-fiber recordings. To mimic as much as possible the human clinical condition, all treated animals were included in the study. No statistically significant differences were observed in behavioral parameters of mechanical sensitivity. Nerve conduction studies did not reveal any significant change in the ddC-treated group. In contrast, we observed electrophysiological evidence of significant demyelinating neuropathy 1 week after the start of d4T treatment. Additionally, spontaneous activity in mechanoinsensitive C-nociceptors was observed in both drug-treated groups. No relationship could be established between measures of spontaneous activity in C-nociceptors and the results of the behavioral tests. Our results show that both models of antiretroviral-induced neuropathy differ in their effects on peripheral nerves. However, both groups present abnormal spontaneous activity in mechanoinsensitive C-nociceptors that can be used as a model for pharmacological intervention.


Subject(s)
Anti-HIV Agents/toxicity , Disease Models, Animal , Neural Conduction/drug effects , Pain Threshold/drug effects , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/complications , Action Potentials/drug effects , Animals , Male , Nerve Fibers, Unmyelinated/physiology , Neurophysiology , Pain Measurement , Physical Stimulation , Rats , Rats, Sprague-Dawley , Rats, Wistar , Stavudine/toxicity , Zalcitabine/toxicity
8.
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
9.
Exp Neurol ; 267: 53-63, 2015 May.
Article in English | MEDLINE | ID: mdl-25765490

ABSTRACT

The antiretroviral toxic neuropathy, a distal sensory polyneuropathy associated with antiretroviral treatment, is a frequently occurring neurological complication during treatment of patients with AIDS and often leads to discontinuation of antiretroviral therapy. The mechanisms by which antiretroviral drugs contribute to the development of neuropathic pain are not known. Using drugs that reduce intracellular calcium ions (Ca(2+)), we investigated the hypothesis that altered cytosolic Ca(2+) concentration contributes to the 2',3'-dideoxycytidine (ddC)-evoked painful neuropathy. Administration of ddC induced mechanical and cold allodynia, which were abolished by intrathecal administration of TMB-8, a blocker of Ca(2+) release from intracellular stores, and by ryanodine, a RyR antagonist. Treatment with the IP3R antagonist heparin prevented mechanical allodynia with no effect on thermal response. To further clarify the pathway involved, we investigated the role of HuD, a RNA binding protein involved in neuronal function. HuD silencing reverted both mechanical and cold allodynia inducing, a phenotype comparable to that of ryanodine-exposed mice. HuD binding to the RyR2 mRNA, the most abundant RyR isoform in the spinal cord, was demonstrated and RyR2 silencing prevented the ddC-induced neuropathic pain. A positive regulation of gene expression on CaMKIIα by HuD was also observed, but sequestration of CaMKIIα had no effect on ddC-induced allodynia. The present findings identify a spinal RyR2 pathway activated in response to ddC administration, involving the binding activity on RyR2 mRNA by HuD. We propose the modulation of the RyR2 pathway as a therapeutic perspective in the management of antiretroviral painful neuropathy.


Subject(s)
ELAV Proteins/metabolism , Pain Threshold/drug effects , Pain/chemically induced , Pain/pathology , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/pathology , Ryanodine Receptor Calcium Release Channel/metabolism , Spinal Cord/physiology , Zalcitabine/toxicity , Analgesics, Non-Narcotic/pharmacology , Animals , Anti-HIV Agents/toxicity , Apomorphine/therapeutic use , Calcium Channel Blockers/therapeutic use , Cell Line, Tumor , Disease Models, Animal , Dopamine Agonists/pharmacology , Dopamine Uptake Inhibitors/pharmacology , ELAV Proteins/genetics , ELAV-Like Protein 4 , Exploratory Behavior/drug effects , Gene Expression Regulation/physiology , Hyperalgesia/etiology , Hyperalgesia/metabolism , Hyperalgesia/therapy , Male , Mice , Motor Activity/drug effects , Neuroblastoma/pathology , Pain/complications , Pain/drug therapy , Peripheral Nervous System Diseases/complications , Peripheral Nervous System Diseases/drug therapy
10.
Mol Pain ; 10: 49, 2014 Jul 30.
Article in English | MEDLINE | ID: mdl-25078297

ABSTRACT

BACKGROUND: HIV-associated sensory neuropathy affects over 50% of HIV patients and is a common peripheral nerve complication of HIV infection and highly active antiretroviral therapy (HAART). Evidence shows that painful HIV sensory neuropathy is influenced by neuroinflammatory events that include the proinflammatory molecules, MAP Kinase, tumor necrosis factor-α (TNFα), stromal cell-derived factor 1-α (SDF1α), and C-X-C chemokine receptor type 4 (CXCR4). However, the exact mechanisms of painful HIV sensory neuropathy are not known, which hinders our ability to develop effective treatments. In this study, we investigated whether inhibition of proinflammatory factors reduces the HIV-associated neuropathic pain state. RESULTS: Neuropathic pain was induced by peripheral HIV coat protein gp120 combined with 2',3'-dideoxycytidine (ddC, one of the nucleoside reverse transcriptase inhibitors (NRTIs)). Mechanical threshold was tested using von Frey filament fibers. Non-replicating herpes simplex virus (HSV) vectors expressing interleukin 10 (IL10) were inoculated into the hindpaws of rats. The expression of TNFα, SDF1α, and CXCR4 in the lumbar spinal cord and L4/5 dorsal root ganglia (DRG) was examined using western blots. IL-10 expression mediated by the HSV vectors resulted in a significant elevation of mechanical threshold. The anti-allodynic effect of IL-10 expression mediated by the HSV vectors lasted more than 3 weeks. The area under the effect-time curves (AUC) in mechanical threshold in rats inoculated with the HSV vectors expressing IL-10, was increased compared with the control vectors, indicating antinociceptive effect of the IL-10 vectors. The HSV vectors expressing IL-10 also concomitantly reversed the upregulation of p-p38, TNFα, SDF1α, and CXCR4 induced by gp120 in the lumbar spinal dorsal horn and/or the DRG at 2 and/or 4 weeks. CONCLUSION: The blocking of the signaling of these proinflammatory molecules is able to reduce HIV-related neuropathic pain, which provide a novel mechanism-based approach to treating HIV-associated neuropathic pain using gene therapy.


Subject(s)
Antiviral Agents/toxicity , HIV Envelope Protein gp120/toxicity , Interleukin-10/metabolism , Interleukin-10/therapeutic use , Neuralgia/chemically induced , Neuralgia/therapy , Zalcitabine/toxicity , Animals , Chemokine CXCL12/metabolism , Disease Models, Animal , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Genetic Vectors/physiology , Interleukin-10/genetics , Male , Neuralgia/pathology , Pain Threshold/drug effects , Physical Stimulation , Rats , Rats, Sprague-Dawley , Simplexvirus/genetics , Spinal Cord Dorsal Horn/metabolism , Spinal Cord Dorsal Horn/pathology , Time Factors , Tumor Necrosis Factor-alpha/metabolism
11.
Exp Neurol ; 261: 343-53, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24861443

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) are known to produce painful neuropathies and to enhance states of pain hypersensitivity produced by HIV-1 infection in patients with AIDS leading to discontinuation of antiretroviral therapy, thus limiting viral suppression strategies. The mechanisms by which NRTIs contribute to the development of neuropathic pain are not known. In the current study, we tested the hypothesis that HuD, an RNA binding protein known to be an essential promoter of neuronal differentiation and survival, might be involved in the response to NRTI-induced neuropathy. Antiretroviral neuropathy was induced by a single intraperitoneal administration of 2',3'-dideoxycytidine (ddC) in mice. HuD was physiologically expressed in the cytoplasm of the soma and in axons of neurons within DRG and spinal cord and was considerably overexpressed following ddC treatment. ddC up-regulated spinal GAP43 protein, a marker of neuroregeneration, and this increase was counteracted by HuD silencing. GAP43 and HuD colocalize in DRG and spinal dorsal horn (SDH) axons and administration of an anti-GAP43 antibody aggravated the ddC-induced axonal damage. The administration of a protein kinase C (PKC) inhibitor or the PKCγ silencing prevented both HuD and GAP43 increased expression. Conversely, treatment with the PKC activator PDBu potentiated HuD and GAP43 overexpression, demonstrating the presence of a spinal PKC-dependent HuD-GAP43 pathway activated by ddC. These results indicated that HuD recruitment and GAP43 protein increase are mechanistically linked events involved in the response to antiretroviral-induced neurodegenerative processes.


Subject(s)
Anti-Retroviral Agents/toxicity , ELAV Proteins/metabolism , GAP-43 Protein/metabolism , Pain/chemically induced , Peripheral Nervous System Diseases/chemically induced , Spinal Cord/metabolism , Zalcitabine/toxicity , Animals , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation/drug effects , Hyperalgesia/metabolism , Hyperalgesia/physiopathology , Male , Mice , Neurons/drug effects , Neurons/metabolism , Oligodeoxyribonucleotides, Antisense/pharmacology , Pain/physiopathology , Peripheral Nervous System Diseases/physiopathology , Phosphopyruvate Hydratase/metabolism , Protein Kinase C/metabolism , Spinal Cord/drug effects , Time Factors
12.
Behav Pharmacol ; 25(2): 137-46, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24525712

ABSTRACT

Individuals infected with the HIV and taking certain antiretroviral drugs to suppress viral replication have a high prevalence of neuropathic pain that is not alleviated by analgesic/adjuvant drugs that are often efficacious for the relief of other types of neuropathic pain. There is therefore a great need for new analgesics to alleviate the pain of antiretroviral toxic neuropathy (ATN). Small-molecule angiotensin II type 2 receptor (AT2R) antagonists, with ≥1000-fold selectivity over the angiotensin II type 1 receptor, produced analgesia in the chronic constriction injury of the sciatic nerve rat model of peripheral nerve trauma. Hence, the present study was designed to assess their analgesic efficacy in a rat model of ATN. The analgesic efficacy of small-molecule AT2R antagonists (EMA200 and EMA300) was assessed in a rat model of dideoxycytidine (ddC)-induced ATN. Single intraperitoneal bolus doses of EMA200 (0.3-10 mg/kg) induced dose-dependent analgesia in ddC-rats; the mean ED50 was 3.2 mg/kg. Twice-daily intraperitoneal administration of EMA300 at 30 mg/kg to ddC-rats for 3 days produced significant analgesia on days 2 and 3 of the treatment period. Therefore, small-molecule AT2R antagonists should be investigated further as novel analgesics for the relief of ATN.


Subject(s)
Analgesics/pharmacology , Angiotensin II Type 2 Receptor Blockers/pharmacology , Anti-HIV Agents/toxicity , Neuralgia/drug therapy , Polyneuropathies/drug therapy , Zalcitabine/toxicity , Angiotensin II Type 2 Receptor Blockers/chemistry , Animals , Disease Models, Animal , Dose-Response Relationship, Drug , Exploratory Behavior/drug effects , Hot Temperature , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Imidazoles/chemistry , Imidazoles/pharmacology , Male , Neuralgia/chemically induced , Polyneuropathies/chemically induced , Polyneuropathies/complications , Pyridines/chemistry , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley , Rats, Wistar , Sciatic Neuropathy/drug therapy , Time Factors , Touch
13.
Anesth Analg ; 118(3): 671-80, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24557113

ABSTRACT

BACKGROUND: In the human immunodeficiency virus (HIV)-associated sensory neuropathy, neuropathic pain associated with the use of nucleoside reverse transcriptase inhibitors (NRTIs) in patients with HIV/acquired immunodeficiency syndrome is clinically common. While evidence demonstrates that neuropathic pain is influenced by neuroinflammatory events that include the proinflammatory molecules, tumor necrosis factor-α (TNF-α), stromal cell-derived factor 1-α (SDF1-α), and C-X-C chemokine receptor type 4 (CXCR4), the detailed mechanisms by which NRTIs contribute to the development of neuropathic pain are not known. In this study, we investigated the role of these proinflammatory molecules in the dorsal root ganglion (DRG) and the spinal dorsal horn in NRTIs-mediated neuropathic pain state. METHODS: Neuropathic pain was induced by intraperitoneal administration of 2',3'-dideoxycytidine (ddC, one of the NRTIs). Mechanical threshold was tested using von Frey filament fibers. Nonreplicating herpes simplex virus (HSV) vectors expressing p55 TNF soluble receptor (p55TNFSR) were inoculated into hindpaw of rats. The expression of TNF-α, SDF1-α, and CXCR4 in both the lumbar spinal cord and the L4/5 DRG was examined using Western blots. Intrathecal CXCR4 antagonist was administered. RESULTS: The present study demonstrated that (1) systemic ddC induced upregulation of TNF-α, SDF1-α, and CXCR4 in both the lumbar spinal cord and the L4/5 DRG; (2) p55TNFSR mediated by a nonreplicating HSV vector reversed mechanical allodynia induced by systemic ddC; (3) intrathecal administration of the CXCR4 antagonist AMD3100 increased mechanical threshold; and (4) HSV vector expressing p55TNFSR reversed upregulation of TNF-α, SDF1-α, and CXCR4 induced by ddC in the lumbar spinal dorsal horn and the DRG. CONCLUSIONS: Our studies demonstrate that TNF-α through the SDF1/CXCR4 system is involved in the NRTIs-related neuropathic pain state and that blocking the signaling of these proinflammatory molecules is able to reduce NRTIs-related neuropathic pain. These results provide a novel mechanism-based approach (gene therapy) to treating HIV-associated neuropathic pain.


Subject(s)
Chemokine CXCL12/physiology , Hyperalgesia/metabolism , Receptors, CXCR4/physiology , Receptors, Tumor Necrosis Factor/biosynthesis , Reverse Transcriptase Inhibitors/toxicity , Simplexvirus/physiology , Animals , Chemokine CXCL12/antagonists & inhibitors , Chemokine CXCL12/biosynthesis , Hyperalgesia/chemically induced , Hyperalgesia/prevention & control , Male , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/biosynthesis , Spinal Cord/drug effects , Spinal Cord/metabolism , Up-Regulation/physiology , Zalcitabine/toxicity
14.
Cell Mol Neurobiol ; 34(2): 183-94, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24198040

ABSTRACT

Dideoxycytidine (zalcitabine, ddC) produces neurotoxic effects. It is particularly important to understand the toxic effects of ddC on different subpopulations of dorsal root ganglion (DRG) neurons which express distinct tyrosine kinase receptor (Trk) and to find therapeutic factors for prevention and therapy for ddC-induced peripheral sensory neuropathy. Insulin-like growth factor-1 (IGF-1) has been shown to have neurotrophic effects on DRG sensory neurons. However, little is known about the effects of ddC on distinct Trk (TrkA, TrkB, and TrkC) expression in DRG neurons and the neuroprotective effects of IGF-1 on ddC-induced neurotoxicity. Here, we have tested the extent to which the expression of TrkA, TrkB, and TrkC receptors in primary cultured DRG neurons is affected by ddC in the presence or absence of IGF-1. In this experiment, we found that exposure of 5, 25, and 50 µmol/L ddC caused a dose-dependent decrease of the mRNA, protein, and the proportion of TrkA-, TrkB-, and TrkC-expressing neurons. IGF-1 (20 nmol/L) could partially reverse the decrease of TrkA and TrkB, but not TrkC, expression with ddC exposure. The phosphatidylinositol 3-kinase (PI3K) inhibitor LY294002 (10 µmol/L) blocked the effects of IGF-1. These results suggested that the subpopulations of DRG neurons which express distinct TrkA, TrkB, and TrkC receptors were affected by ddC exposure. IGF-1 might relieve the ddC-induced toxicity of TrkA- and TrkB-, but not TrkC-expressing DRG neurons. These data offer new clues for a better understanding of the association of ddC with distinct Trk receptor expression and provide new evidence of the potential therapeutic role of IGF-1 on ddC-induced neurotoxicity.


Subject(s)
Ganglia, Spinal/cytology , Insulin-Like Growth Factor I/pharmacology , Neurons/enzymology , Receptor Protein-Tyrosine Kinases/metabolism , Zalcitabine/toxicity , Animals , Blotting, Western , Gene Expression Regulation/drug effects , Microtubule-Associated Proteins/metabolism , Neurons/drug effects , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics , Receptor, trkA/genetics , Receptor, trkA/metabolism , Receptor, trkB/genetics , Receptor, trkB/metabolism , Receptor, trkC/genetics , Receptor, trkC/metabolism
15.
Nucleic Acids Res ; 41(16): 7793-803, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23775789

ABSTRACT

Chain-terminating nucleoside analogs (CTNAs) that cause stalling or premature termination of DNA replication forks are widely used as anticancer and antiviral drugs. However, it is not well understood how cells repair the DNA damage induced by these drugs. Here, we reveal the importance of tyrosyl-DNA phosphodiesterase 1 (TDP1) in the repair of nuclear and mitochondrial DNA damage induced by CTNAs. On investigating the effects of four CTNAs-acyclovir (ACV), cytarabine (Ara-C), zidovudine (AZT) and zalcitabine (ddC)-we show that TDP1 is capable of removing the covalently linked corresponding CTNAs from DNA 3'-ends. We also show that Tdp1-/- cells are hypersensitive and accumulate more DNA damage when treated with ACV and Ara-C, implicating TDP1 in repairing CTNA-induced DNA damage. As AZT and ddC are known to cause mitochondrial dysfunction, we examined whether TDP1 repairs the mitochondrial DNA damage they induced. We find that AZT and ddC treatment leads to greater depletion of mitochondrial DNA in Tdp1-/- cells. Thus, TDP1 seems to be critical for repairing nuclear and mitochondrial DNA damage caused by CTNAs.


Subject(s)
Antimetabolites, Antineoplastic/toxicity , Antiviral Agents/toxicity , DNA Damage , DNA Repair , Phosphoric Diester Hydrolases/metabolism , Acyclovir/metabolism , Acyclovir/toxicity , Animals , Anti-HIV Agents/metabolism , Anti-HIV Agents/toxicity , Antimetabolites, Antineoplastic/metabolism , Antiviral Agents/metabolism , Cell Line , Cell Nucleus/drug effects , Cells, Cultured , Chickens , Cytarabine/metabolism , Cytarabine/toxicity , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Gene Deletion , Mice , Phosphoric Diester Hydrolases/genetics , Zalcitabine/metabolism , Zalcitabine/toxicity , Zidovudine/metabolism , Zidovudine/toxicity
16.
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
17.
J Neurosci ; 31(31): 11404-10, 2011 Aug 03.
Article in English | MEDLINE | ID: mdl-21813700

ABSTRACT

While oxidative stress has been implicated in small-fiber painful peripheral neuropathies, antioxidants are only partially effective to treat patients. We have tested the hypothesis that Drp1 (dynamin-related protein 1), a GTPase that catalyzes the process of mitochondrial fission, which is a mechanism central for the effect and production of reactive oxygen species (ROS), plays a central role in these neuropathic pain syndromes. Intrathecal administration of oligodeoxynucleotide antisense against Drp1 produced a decrease in its expression in peripheral nerve and markedly attenuated neuropathic mechanical hyperalgesia caused by HIV/AIDS antiretroviral [ddC (2',3'-dideoxycytidine)] and anticancer (oxaliplatin) chemotherapy in male Sprague Dawley rats. To confirm the role of Drp1 in these models of neuropathic pain, as well as to demonstrate its contribution at the site of sensory transduction, we injected a highly selective Drp1 inhibitor, mdivi-1, at the site of nociceptive testing on the dorsum of the rat's hindpaw. mdivi-1 attenuated both forms of neuropathic pain. To evaluate the role of Drp1 in hyperalgesia induced by ROS, we demonstrated that intradermal hydrogen peroxide produced dose-dependent hyperalgesia that was inhibited by mdivi-1. Finally, mechanical hyperalgesia induced by diverse pronociceptive mediators involved in inflammatory and neuropathic pain-tumor necrosis factor α, glial-derived neurotrophic factor, and nitric oxide-was also inhibited by mdivi-1. These studies provide support for a substantial role of mitochondrial fission in preclinical models of inflammatory and neuropathic pain.


Subject(s)
Dynamins/metabolism , Neuralgia/metabolism , Analysis of Variance , Animals , Anti-HIV Agents/toxicity , Antineoplastic Agents/toxicity , Disease Models, Animal , Dose-Response Relationship, Drug , Dynamins/genetics , Epinephrine/therapeutic use , Gene Expression Regulation/drug effects , Glial Cell Line-Derived Neurotrophic Factor/therapeutic use , Hydrogen Peroxide , Hyperalgesia/chemically induced , Hyperalgesia/drug therapy , Hyperalgesia/metabolism , Male , Nerve Growth Factor/therapeutic use , Neuralgia/chemically induced , Neuralgia/drug therapy , Nitric Oxide Donors/toxicity , Nitro Compounds/toxicity , Oligodeoxyribonucleotides, Antisense/therapeutic use , Organoplatinum Compounds/toxicity , Oxaliplatin , Pain Measurement/methods , Quinazolinones/therapeutic use , RNA, Messenger , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/metabolism , Time Factors , Tumor Necrosis Factor-alpha/therapeutic use , Zalcitabine/toxicity
18.
J Acquir Immune Defic Syndr ; 55(5): 550-7, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-20827217

ABSTRACT

OBJECTIVE: Long-term antiretroviral treatment with nucleoside analogue reverse transcriptase inhibitors (NRTI) may result in a cardiomyopathy due to mitochondrial DNA (mtDNA) depletion. An intact mitochondrial function is required for the synthesis of intramyocardial pyrimidine nucleotides, which in turn are building blocks of mtDNA. We investigated if NRTI-related cardiomyopathy can be prevented with pyrimidine precursors. METHODS: Mice were fed with zidovudine or zalcitabine with or without simultaneous Mitocnol, a dietary supplement with high uridine bioavailability. Myocardia were examined after 9 weeks. RESULTS: Both NRTI induced a cardiomyopathy with mitochondrial enlargement, a disrupted cristal architecture on electron microscopy and diminished myocardial mtDNA copy numbers. The myocardial mtDNA-encoded cytochrome c-oxidase I subunit was impaired more profoundly than the nucleus-encoded cytochrome c-oxidase IV subunit. The myocardial formation of reactive oxygen species and mtDNA mutations was enhanced in zidovudine and zalcitabine treated animals. Mitocnol attenuated or normalized all myocardial pathology when given with both NRTI, but by itself had no intrinsic effects and no apparent adverse effects. CONCLUSIONS: Zidovudine and zalcitabine induce a mitochondrial cardiomyopathy, which is antagonized with uridine supplementation, implicating pyrimidine pool depletion in its pathogenesis. Pyrimidine pool replenishment may be exploited clinically because uridine is well tolerated.


Subject(s)
Cardiomyopathies/chemically induced , Cardiotoxins/toxicity , Heart/drug effects , Mitochondria, Heart/drug effects , Pyrimidines/metabolism , Reverse Transcriptase Inhibitors/toxicity , Animals , Anti-HIV Agents/administration & dosage , Anti-HIV Agents/pharmacology , Anti-HIV Agents/toxicity , DNA Copy Number Variations/drug effects , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/genetics , Dietary Supplements , Electron Transport Complex IV/metabolism , HIV Infections/drug therapy , HIV Infections/genetics , Mice , Mice, Inbred BALB C , Mitochondria, Heart/ultrastructure , Mutation , Nucleosides , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Reactive Oxygen Species , Reverse Transcriptase Inhibitors/administration & dosage , Reverse Transcriptase Inhibitors/pharmacology , Uridine/administration & dosage , Uridine/pharmacology , Uridine/therapeutic use , Zalcitabine/administration & dosage , Zalcitabine/pharmacology , Zalcitabine/toxicity , Zidovudine/administration & dosage , Zidovudine/pharmacology , Zidovudine/toxicity
19.
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
20.
Cytometry B Clin Cytom ; 76(3): 181-90, 2009 May.
Article in English | MEDLINE | ID: mdl-18823003

ABSTRACT

BACKGROUND: Real-time PCR has quantified decreased mitochondrial DNA levels in association with nucleoside reverse transcriptase inhibitor (NRTI) therapy of HIV-infected populations. However, real-time PCR is best suited to distinguish log differences in an analyte. In an effort to monitor individuals in more detail, we developed a flow cytometric assay to gauge mitochondrial function. METHODS: Flow cytometric quantification of a mitochondrial DNA-encoded mitochondrial protein (cytochrome c oxidase subunit I (COX-I)) and a nuclear DNA-encoded mitochondrial protein [ATP synthase subunit D (Sub-D)] was optimized and validated. RESULTS: Intra-assay and interassay variability was low using peripheral blood mononuclear cells (PBMCs) (CV of 6.15% for COX-I and 7.11% Sub-D, and 9.38% and 9.83% for COX-I and Sub-D, respectively). Mitochondrial protein depletion was evident with in vitro treatment of cells with ethidium bromide (EtBr) and zalcitabine (ddC). Mitochondrial protein expression in 40 healthy adults clustered tightly. Depletion of mitochondrial protein, however, was neither detected in cryopreserved PBMC from NRTI-treated children (n = 9) nor in adults with a history of symptoms consistent with mitochondrial toxicity or ongoing treatment with didanosine (ddI) or stavudine (d4T) (n = 51). CONCLUSIONS: A validated flow cytometric assay allows simultaneous detection of mitochondrial DNA and nuclear DNA encoded proteins at the single cell level, offering a method to monitor for mitochondrial function. Prospective studies are required to evaluate whether mitochondrial protein loss is observed in at-risk patients prior to the onset of symptoms from mitochondrial dysfunction.


Subject(s)
Ethidium/toxicity , Leukocytes, Mononuclear/cytology , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Zalcitabine/toxicity , Adenosine Triphosphatases/metabolism , Adult , Child , Child, Preschool , DNA, Mitochondrial/genetics , Dose-Response Relationship, Drug , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Flow Cytometry , Humans , Infant , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/enzymology , Middle Aged , Mitochondria/enzymology , Mitochondrial Proteins/genetics , Reproducibility of Results , Sensitivity and Specificity , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...