Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.166
Filter
1.
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
2.
Molecules ; 27(13)2022 Jun 21.
Article in English | MEDLINE | ID: mdl-35807233

ABSTRACT

An improved protocol for the transformation of ribonucleosides into 2',3'-dideoxynucleoside and 2',3'-didehydro-2',3'-dideoxynucleoside derivatives, including the anti-HIV drugs stavudine (d4T), zalcitabine (ddC) and didanosine (ddI), was established. The process involves radical deoxygenation of xanthate using environmentally friendly and low-cost reagents. Bromoethane or 3-bromopropanenitrile was the alkylating agent of choice to prepare the ribonucleoside 2',3'-bisxanthates. In the subsequent radical deoxygenation reaction, tris(trimethylsilyl)silane and 1,1'-azobis(cyclohexanecarbonitrile) were used to replace hazardous Bu3SnH and AIBN, respectively. In addition, TBAF was substituted for camphorsulfonic acid in the deprotection step of the 5'-O-silyl ether group, and an enzyme (adenosine deaminase) was used to transform 2',3'-dideoxyadenosine into 2',3'-dideoxyinosine (ddI) in excellent yield.


Subject(s)
Anti-HIV Agents , Zidovudine , Didanosine , Dideoxynucleosides , Stavudine , Zalcitabine
3.
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
4.
Blood ; 129(19): 2657-2666, 2017 05 11.
Article in English | MEDLINE | ID: mdl-28283480

ABSTRACT

Mitochondrial DNA (mtDNA) biosynthesis requires replication factors and adequate nucleotide pools from the mitochondria and cytoplasm. We performed gene expression profiling analysis of 542 human acute myeloid leukemia (AML) samples and identified 55% with upregulated mtDNA biosynthesis pathway expression compared with normal hematopoietic cells. Genes that support mitochondrial nucleotide pools, including mitochondrial nucleotide transporters and a subset of cytoplasmic nucleoside kinases, were also increased in AML compared with normal hematopoietic samples. Knockdown of cytoplasmic nucleoside kinases reduced mtDNA levels in AML cells, demonstrating their contribution in maintaining mtDNA. To assess cytoplasmic nucleoside kinase pathway activity, we used a nucleoside analog 2'3'-dideoxycytidine (ddC), which is phosphorylated to the activated antimetabolite, 2'3'-dideoxycytidine triphosphate by cytoplasmic nucleoside kinases. ddC is a selective inhibitor of the mitochondrial DNA polymerase γ. ddC was preferentially activated in AML cells compared with normal hematopoietic progenitor cells. ddC treatment inhibited mtDNA replication, oxidative phosphorylation, and induced cytotoxicity in a panel of AML cell lines. Furthermore, ddC preferentially inhibited mtDNA replication in a subset of primary human leukemia cells and selectively targeted leukemia cells while sparing normal progenitor cells. In animal models of human AML, treatment with ddC decreased mtDNA, electron transport chain proteins, and induced tumor regression without toxicity. ddC also targeted leukemic stem cells in secondary AML xenotransplantation assays. Thus, AML cells have increased cytidine nucleoside kinase activity that regulates mtDNA biogenesis and can be leveraged to selectively target oxidative phosphorylation in AML.


Subject(s)
DNA, Mitochondrial/genetics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Oxidative Phosphorylation , Phosphotransferases/metabolism , Animals , Cell Line, Tumor , Cells, Cultured , DNA Replication , Humans , Mice, SCID , NM23 Nucleoside Diphosphate Kinases/metabolism , Nucleoside-Phosphate Kinase/metabolism , Signal Transduction , Tumor Cells, Cultured , Zalcitabine/metabolism
5.
J Pharmacol Sci ; 139(4): 325-332, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30871874

ABSTRACT

Neuropathic pain sometimes occurs during chemotherapy with paclitaxel or HIV/AIDS antiretroviral therapy with nucleoside reverse transcriptase inhibitors (NRTIs). We previously reported that coadministration of indomethacin plus minocycline (IPM) was antihyperalgesic in a cannabinoid type 1 (CB1) receptor-dependent manner in a mouse model of paclitaxel-induced neuropathic pain. We evaluated if IPM combination has antihyperalgesic and antiallodynic activities in animal models of paclitaxel or NRTI (ddC, zalcitabine)-induced neuropathic pain, and whether antagonists of CB1, CB2 receptors or G protein-coupled receptor 55 (GPR55) can inhibit these activities of IPM. IPM produced antihyperalgesic and antiallodynic effects against paclitaxel and ddC-induced thermal hyperalgesia and mechanical allodynia. WIN 55,212-2, a cannabinoid receptor agonist, also had antihyperalgesic activity. The antihyperalgesic and antiallodynic activities of IPM were antagonized by a CB1 receptor antagonist AM251 and a CB2 receptor antagonist AM630, but not a GPR55 antagonist ML193. IPM had no effects on the mean time spent on the rotarod, whereas WIN 55,212-2 reduced it in a dose-dependent manner. These results show that IPM at a fixed ratio produces antihyperalgesic and antiallodynic effects in mice models of both paclitaxel and NRTI-induced neuropathic pain which is dependent on both CB1 and CB2 receptors, without causing the typical cannabinoid receptor agonist-induced motor impairment.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Retroviral Agents/adverse effects , Antineoplastic Agents, Phytogenic/adverse effects , Indomethacin/administration & dosage , Minocycline/administration & dosage , Neuralgia/chemically induced , Neuralgia/drug therapy , Paclitaxel/adverse effects , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/metabolism , Zalcitabine/adverse effects , Animals , Anti-Bacterial Agents/pharmacology , Disease Models, Animal , Female , Indomethacin/pharmacology , Mice, Inbred BALB C , Minocycline/pharmacology
6.
Molecules ; 25(1)2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31892132

ABSTRACT

Neuropathic pain associated with nucleoside reverse transcriptase inhibitors (NRTIs), therapeutic agents for human immunodeficiency virus (HIV), responds poorly to available drugs. Smoked cannabis was reported to relieve HIV-associated neuropathic pain in clinical trials. Some constituents of cannabis (Cannabis sativa) activate cannabinoid type 1 (CB1) and cannabinoid type 2 (CB2) receptors. However, activation of the CB1 receptor is associated with side effects such as psychosis and physical dependence. Therefore, we investigated the effect of ß-caryophyllene (BCP), a CB2-selective phytocannabinoid, in a model of NRTI-induced neuropathic pain. Female BALB/c mice treated with 2'-3'-dideoxycytidine (ddC, zalcitabine), a NRTI, for 5 days developed mechanical allodynia, which was prevented by cotreatment with BCP, minocycline or pentoxifylline. A CB2 receptor antagonist (AM 630), but not a CB1 receptor antagonist (AM 251), antagonized BCP attenuation of established ddC-induced mechanical allodynia. ß-Caryophyllene prevented the ddC-induced increase in cytokine (interleukin 1 beta, tumor necrosis factor alpha and interferon gamma) transcripts in the paw skin and brain, as well as the phosphorylation level of Erk1/2 in the brain. In conclusion, BCP prevents NRTI-induced mechanical allodynia, possibly via reducing the inflammatory response, and attenuates mechanical allodynia through CB2 receptor activation. Therefore, BCP could be useful for prevention and treatment of antiretroviral-induced neuropathic pain.


Subject(s)
Hyperalgesia/drug therapy , MAP Kinase Signaling System/drug effects , Neuralgia/metabolism , Polycyclic Sesquiterpenes/pharmacology , Receptor, Cannabinoid, CB2/agonists , Reverse Transcriptase Inhibitors/adverse effects , Zalcitabine/adverse effects , Animals , Brain/metabolism , Brain/pathology , Disease Models, Animal , Female , Humans , Hyperalgesia/chemically induced , Hyperalgesia/metabolism , Hyperalgesia/pathology , Mice , Mice, Inbred BALB C , Neuralgia/chemically induced , Neuralgia/pathology , Polycyclic Sesquiterpenes/chemistry , Receptor, Cannabinoid, CB2/metabolism , Reverse Transcriptase Inhibitors/pharmacology , Skin/metabolism , Skin/pathology , Zalcitabine/pharmacology
7.
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
8.
Biochemistry ; 56(1): 33-46, 2017 Jan 10.
Article in English | MEDLINE | ID: mdl-27936595

ABSTRACT

Reverse transcriptases (RTs) are typically assayed in vitro with 5-10 mM Mg2+, whereas the free Mg2+ concentration in cells is much lower. Artificially high Mg2+ concentrations used in vitro can misrepresent different properties of human immunodeficiency virus (HIV) RT, including fidelity, catalysis, pausing, and RNase H activity. Here, we analyzed nucleoside (NRTIs) and non-nucleoside RT inhibitors (NNRTIs) in primer extension assays at different concentrations of free Mg2+. At low concentrations of Mg2+, NRTIs and dideoxynucleotides (AZTTP, ddCTP, ddGTP, and 3TCTP) inhibited HIV-1 and HIV-2 RT synthesis less efficiently than they did with large amounts of Mg2+, whereas inhibition by the "translocation-defective RT inhibitor" EFdA (4'-ethynyl-2-fluoro-2'-deoxyadenosine) was unaffected by Mg2+ concentrations. Steady-state kinetic analyses revealed that the reduced level of inhibition at low Mg2+ concentrations resulted from a 3-9-fold (depending on the particular nucleotide and inhibitor) less efficient incorporation (based on kcat/Km) of these NRTIs under this condition compared to incorporation of natural dNTPs. In contrast, EFdATP was incorporated with an efficiency similar to that of its analogue dATP at low Mg2+ concentrations. Unlike NRTIs, NNRTIs (nevirapine, efavirenz, and rilviripine), were approximately 4-fold (based on IC50 values) more effective at low than at high Mg2+ concentrations. Drug-resistant HIV-1 RT mutants also displayed the Mg2+-dependent difference in susceptibility to NRTIs and NNRTIs. In summary, analyzing the efficiency of inhibitors under more physiologically relevant low-Mg2+ conditions yielded results dramatically different from those from measurements using commonly employed high-Mg2+ in vitro conditions. These results also emphasize differences in Mg2+ sensitivity between the translocation inhibitor EFdATP and other NRTIs.


Subject(s)
Dideoxynucleotides/pharmacology , HIV Reverse Transcriptase/antagonists & inhibitors , Magnesium/pharmacology , Nucleosides/pharmacology , Reverse Transcriptase Inhibitors/pharmacology , Deoxycytosine Nucleotides/pharmacology , Deoxyguanine Nucleotides/pharmacology , Dose-Response Relationship, Drug , Drug Interactions , Electrophoresis, Polyacrylamide Gel , HIV Reverse Transcriptase/genetics , HIV Reverse Transcriptase/metabolism , Humans , Kinetics , Mutation , Thymine Nucleotides/pharmacology , Zalcitabine/pharmacology , Zidovudine/analogs & derivatives , Zidovudine/pharmacology
10.
Article in English | MEDLINE | ID: mdl-28396546

ABSTRACT

HIV-1 reverse transcriptase (RT) is targeted by multiple drugs. RT mutations that confer resistance to nucleoside RT inhibitors (NRTIs) emerge during clinical use. Q151M and four associated mutations, A62V, V75I, F77L, and F116Y, were detected in patients failing therapies with dideoxynucleosides (didanosine [ddI], zalcitabine [ddC]) and/or zidovudine (AZT). The cluster of the five mutations is referred to as the Q151M complex (Q151Mc), and an RT or virus containing Q151Mc exhibits resistance to multiple NRTIs. To understand the structural basis for Q151M and Q151Mc resistance, we systematically determined the crystal structures of the wild-type RT/double-stranded DNA (dsDNA)/dATP (complex I), wild-type RT/dsDNA/ddATP (complex II), Q151M RT/dsDNA/dATP (complex III), Q151Mc RT/dsDNA/dATP (complex IV), and Q151Mc RT/dsDNA/ddATP (complex V) ternary complexes. The structures revealed that the deoxyribose rings of dATP and ddATP have 3'-endo and 3'-exo conformations, respectively. The single mutation Q151M introduces conformational perturbation at the deoxynucleoside triphosphate (dNTP)-binding pocket, and the mutated pocket may exist in multiple conformations. The compensatory set of mutations in Q151Mc, particularly F116Y, restricts the side chain flexibility of M151 and helps restore the DNA polymerization efficiency of the enzyme. The altered dNTP-binding pocket in Q151Mc RT has the Q151-R72 hydrogen bond removed and has a switched conformation for the key conserved residue R72 compared to that in wild-type RT. On the basis of a modeled structure of hepatitis B virus (HBV) polymerase, the residues R72, Y116, M151, and M184 in Q151Mc HIV-1 RT are conserved in wild-type HBV polymerase as residues R41, Y89, M171, and M204, respectively; functionally, both Q151Mc HIV-1 and wild-type HBV are resistant to dideoxynucleoside analogs.


Subject(s)
Anti-HIV Agents/therapeutic use , Didanosine/therapeutic use , Gene Products, pol/antagonists & inhibitors , HIV Reverse Transcriptase/antagonists & inhibitors , HIV-1/drug effects , Hepatitis B virus/drug effects , Reverse Transcriptase Inhibitors/therapeutic use , Zalcitabine/therapeutic use , Zidovudine/therapeutic use , Crystallography, X-Ray , DNA-Binding Proteins/genetics , Drug Resistance, Viral/genetics , HIV Reverse Transcriptase/genetics , Hepatitis B virus/genetics , Humans , Mutation/genetics , Protein Conformation , Protein Structure, Quaternary
11.
Article in English | MEDLINE | ID: mdl-28559274

ABSTRACT

We explored if baseline CD4/CD8 T-cell ratio is associated with immunodiscordant response to antiretroviral therapy in HIV-infected subjects. Comparing immunodiscordant and immunoconcordant subjects matched by pretreatment CD4 counts, we observed a lower pretreatment CD4/CD8 T-cell ratio in immunodiscordant subjects. Furthermore, pretreatment CD4/CD8 T-cell ratio, but not CD4 counts, correlated with the main immunological alterations observed in immunodiscordants, including increased regulatory T-cell (Treg) frequency and T-cell turnover-related markers. Then, in a larger cohort, only baseline CD4/CD8 T-cell ratio was independently associated with immunodiscordance, after adjusting by the viral CXCR4-tropic HIV variants. Our results suggest that the CD4/CD8 T-cell ratio could be an accurate biomarker of the subjacent immunological damage triggering immunodiscordance.


Subject(s)
Antiretroviral Therapy, Highly Active/methods , CD4-CD8 Ratio , CD8-Positive T-Lymphocytes/immunology , HIV Infections/drug therapy , HIV Infections/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Anti-HIV Agents/therapeutic use , Biomarkers/metabolism , Cell Survival/drug effects , Didanosine/therapeutic use , Female , Humans , Male , Middle Aged , Receptors, CXCR4/immunology , Stavudine/therapeutic use , Viral Load , Zalcitabine/therapeutic use , Zidovudine/therapeutic use
12.
Neurochem Res ; 42(10): 2996-3004, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28631231

ABSTRACT

DNA polymerase-ß (DNA pol-ß) plays a crucial role in the pathogenesis of Parkinson's disease (PD). The aim of this study was to investigate the neuroprotective effects of a DNA polymerase-ß inhibitor 2',3'-dideoxycytidine (DDC) in PD models. In the in vitro studies, primary cultured neurons were challenged with 1-methyl-4-phenylpyridinium ion (MPP+). The expression of DNA pol-ß was assessed using western blot. The neuroprotective effect of DNA pol-ß knockdown and DNA pol-ß inhibitor DDC was determined using cell viability assay and caspase-3 activity assay. We found that MPP+ induced neuronal death and the activation of caspase-3 in a dose-dependent manner. The expression of DNA pol-ß increased after the neurons were exposed to MPP+. DNA pol-ß siRNA or DNA pol-ß inhibitor DDC attenuated neuronal death induced by MPP+. In the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model of PD, MPTP treatment triggered behavioral deficits and nigrostriatal lesions. Pretreatment with DDC attenuated MPTP-induced behavioral deficits, dopaminergic neuronal death and striatal dopamine depletion in the MPTP mouse model. These results indicate that DNA pol-ß inhibitors may present a novel promising therapeutic option for the neuroprotective treatment of PD.


Subject(s)
Cell Death/drug effects , Dopamine/metabolism , Dopaminergic Neurons/drug effects , Substantia Nigra/drug effects , Zalcitabine/pharmacology , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Cells, Cultured , Disease Models, Animal , Mice , Neuroprotective Agents/pharmacology , Substantia Nigra/metabolism
13.
Bioorg Med Chem ; 25(5): 1696-1702, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28169081

ABSTRACT

Conjugates of phosphorylated dideoxynucleoside antiviral drugs dideoxycytidine (zalcitabine) and lamivudine with SiO2 nanoparticles were obtained via the copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) click chemistry between a nucleoside triphosphate containing an alkynyl group at the γ-phosphate or azidothymidine triphosphate and SiO2 nanoparticles containing alkyl azide or alkynyl groups, respectively. 4-(Prop-2-yn-1-yloxy)butylamino group has been attached to the γ-phosphate group of dideoxycytidine (zalcitabine) and lamivudine 5'-triphosphates via the phosphoramidate linkage. New compounds were shown to be potent killers of human colon carcinoma cells. Anti-HIV activity of the conjugates was demonstrated as well. The conjugates of phosphorylated lamivudine and dideoxycytidine (zalcitabine) showed higher potency than the parent nucleosides. The conjugate of phosphorylated azidothymidine was less active against HIV-1 than the parent nucleoside probably because of the replacement of its 3'-azido group by 1,2,3-triazole ring. These results show an opportunity for using SiO2 nanoparticles as a transport for delivering phosphorylated nucleosides to cells in order to increase their efficiency as antiviral and anticancer drugs.


Subject(s)
Anti-HIV Agents/pharmacology , Cell Proliferation/drug effects , Click Chemistry , Lamivudine/chemistry , Nanoparticles/chemistry , Silicon Dioxide/chemistry , Zalcitabine/chemistry , Cell Line, Transformed , HIV-1/drug effects , Humans , Lamivudine/pharmacology , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Phosphorylation , Spectrometry, Mass, Electrospray Ionization , Zalcitabine/pharmacology
14.
Pharmacol Res ; 111: 23-33, 2016 09.
Article in English | MEDLINE | ID: mdl-27238228

ABSTRACT

Nucleoside reverse transcriptase inhibitors (NRTIs) are key components of HIV/AIDS treatment to reduce viral load. However, antiretroviral toxic neuropathy has become a common peripheral neuropathy among HIV/AIDS patients leading to discontinuation of antiretroviral therapy, for which the underlying pathogenesis is uncertain. This study examines the role of neurofilament (NF) proteins in the spinal dorsal horn, DRG and sciatic nerve after NRTI neurotoxicity in mice treated with zalcitabine (2',3'-dideoxycitidine; ddC). ddC administration up-regulated NF-M and pNF-H proteins with no effect on NF-L. The increase of pNF-H levels was counteracted by the silencing of HuD, an RNA binding protein involved in neuronal development and differentiation. Sciatic nerve sections of ddC exposed mice showed an increased axonal caliber, concomitantly to a pNF-H up-regulation. Both events were prevented by HuD silencing. pNF-H and HuD colocalize in DRG and spinal dorsal horn axons. However, the capability of HuD to bind NF mRNA was not demonstrated, indicating the presence of an indirect mechanism of control of NF expression by HuD. RNA immunoprecipitation experiments showed the capability of HuD to bind the BDNF mRNA and the administration of an anti-BDNF antibody prevented pNF-H increase. These data indicate the presence of a HuD - BDNF - NF-H pathway activated as a regenerative response to the axonal damage induced by ddC treatment to counteract the antiretroviral neurotoxicity. Since analgesics clinically used to treat neuropathic pain are ineffective on antiretroviral neuropathy, a neuroregenerative strategy might represent a new therapeutic opportunity to counteract neurotoxicity and avoid discontinuation or abandon of NRTI therapy.


Subject(s)
Anti-Retroviral Agents , ELAV-Like Protein 4/metabolism , Neurofilament Proteins/metabolism , Sensory Receptor Cells/metabolism , Zalcitabine , Animals , Antibodies/pharmacology , Brain-Derived Neurotrophic Factor/antagonists & inhibitors , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Disease Models, Animal , ELAV-Like Protein 4/genetics , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Gene Silencing , Male , Mice , Neuroprotective Agents/pharmacology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Protein Kinase C/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sciatic Neuropathy/chemically induced , Sciatic Neuropathy/genetics , Sciatic Neuropathy/metabolism , Sciatic Neuropathy/prevention & control , Sensory Receptor Cells/pathology , Signal Transduction , Spinal Cord/metabolism , Spinal Cord/pathology , Up-Regulation
15.
J Biol Chem ; 289(14): 9961-9, 2014 Apr 04.
Article in English | MEDLINE | ID: mdl-24526687

ABSTRACT

The RNA synthesis machinery of non-segmented negative-sense RNA viruses comprises a ribonucleoprotein complex of the genomic RNA coated by a nucleocapsid protein (N) and associated with polymerase. Work with vesicular stomatitis virus (VSV), a prototype, supports a model of RNA synthesis whereby N is displaced from the template to allow the catalytic subunit of the polymerase, the large protein (L) to gain access to the RNA. Consistent with that model, purified L can copy synthetic RNA that contains requisite promoter sequences. Full processivity of L requires its phosphoprotein cofactor and the template-associated N. Here we demonstrate the importance of the 2' position of the RNA template and the substrate nucleotide triphosphates during initiation and elongation by L. The VSV polymerase can initiate on both DNA and RNA and can incorporate dNTPs. During elongation, the polymerase is sensitive to 2' modifications, although dNTPs can be incorporated, and mixed DNA-RNA templates can function. Modifications to the 2' position of the NTP, including 2',3'-ddCTP, arabinose-CTP, and 2'-O-methyl-CTP, inhibit polymerase, whereas 2'-amino-CTP is incorporated. The inhibitory effects of the NTPs were more pronounced on authentic N-RNA with the exception of dGTP, which is incorporated. This work underscores the sensitivity of the VSV polymerase to nucleotide modifications during initiation and elongation and highlights the importance of the 2'-hydroxyl of both template and substrate NTP. Moreover, this study demonstrates a critical role of the template-associated N protein in the architecture of the RNA-dependent RNA polymerase domain of L.


Subject(s)
Cytarabine/chemistry , Promoter Regions, Genetic , RNA, Viral/biosynthesis , RNA-Dependent RNA Polymerase/chemistry , Transcription Elongation, Genetic , Transcription Initiation, Genetic , Vesiculovirus/enzymology , Viral Proteins/chemistry , Zalcitabine/chemistry , Animals , Antimetabolites, Antineoplastic/chemistry , Antimetabolites, Antineoplastic/pharmacology , Cytarabine/pharmacology , RNA, Viral/chemistry , RNA, Viral/genetics , RNA-Dependent RNA Polymerase/genetics , RNA-Dependent RNA Polymerase/metabolism , Sf9 Cells , Spodoptera , Vesiculovirus/genetics , Viral Proteins/genetics , Viral Proteins/metabolism , Zalcitabine/pharmacology
16.
J Immunol ; 191(12): 6060-70, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-24227774

ABSTRACT

The programmed cell death-1 (PD-1)/programmed cell death ligand-1 pathway has been shown to limit cell-mediated effector functions during chronic viral infections impeding clearance of pathogens. As a strategy to reverse this exhaustion and increase T cell polyfunctionality, PD-1 ligands were blocked in vivo using a recombinant macaque PD-1 fused to a macaque Ig-Fc (rPD-1-Fc) in SIVmac239-infected rhesus macaques during the early chronic phase of infection, either alone or in combination with antiretroviral therapy. In vitro blockade showed improvement of Ag-specific CD4(+) and CD8(+) T cells from monkeys chronically infected with SIV. Of note, a prolonged 5-d blockade in culture was beneficial for both gag-specific CD4(+) and CD8(+) T cells based on proliferation and dual cytokine production. Although the in vivo administration of rPD-1-Fc induced enhanced SIV-specific CD4 and CD8 T cell proliferation both in the blood and gut, it failed to alter plasma viremia. However, rPD-1-Fc administration in the context of antiretroviral therapy interruption induced a significant delay of viral load rebound. In addition, rPD-1-Fc administration in MamuA*001(+) monkeys led to both an increase in the frequencies and Ki67 expression of GagCM9(+) CD8(+) T cells in the blood and rectal mucosa and polyfunctionality of GagCM9(+) CD8(+) T cells in blood. In conclusion, however, our data suggest that PD-1/programmed cell death ligand-1 blockade using soluble rPD-1-Fc instead of anti-PD-1 mAb, although effective in rescuing the effector function of SIV-specific CD4(+) and CD8(+) T cells during the early chronic phase of infection, has limited clinical benefit.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunoglobulin Fc Fragments/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/therapeutic use , Simian Acquired Immunodeficiency Syndrome/drug therapy , Viremia/drug therapy , Adenine/analogs & derivatives , Adenine/therapeutic use , Animals , Anti-Retroviral Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Apoptosis , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Combined Modality Therapy , Drug Evaluation, Preclinical , Emtricitabine/analogs & derivatives , Histocompatibility Antigens Class I/immunology , Immunity, Cellular , Immunoglobulin Fc Fragments/pharmacology , Immunotherapy , Lymphokines/metabolism , Macaca mulatta , Organophosphonates/therapeutic use , RNA, Viral/blood , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Simian Acquired Immunodeficiency Syndrome/blood , Simian Acquired Immunodeficiency Syndrome/immunology , Simian Immunodeficiency Virus/genetics , Solubility , Tenofovir , Viremia/blood , Viremia/immunology , Zalcitabine/analogs & derivatives , Zalcitabine/therapeutic use
17.
Anesth Analg ; 120(6): 1394-404, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25851180

ABSTRACT

BACKGROUND: Human immunodeficiency virus (HIV)-related painful sensory neuropathies primarily consist of the HIV infection-related distal sensory polyneuropathy and antiretroviral toxic neuropathies. Pharmacotherapy provides only partial relief of pain in patients with HIV/acquired immune deficiency syndrome because little is known about the exact neuropathological mechanisms for HIV-associated neuropathic pain (NP). Hypofunction of γ-aminobutyric acid (GABA) GABAergic inhibitory mechanisms has been reported after peripheral nerve injury. In this study, we tested the hypothesis that HIV gp120 combined with antiretroviral therapy reduces spinal GABAergic inhibitory tone and that restoration of GABAergic inhibitory tone will reduce HIV-related NP in a rat model. METHODS: The application of recombinant HIV-1 envelope protein gp120 into the sciatic nerve plus systemic ddC (one antiretroviral drug) induced mechanical allodynia. The hind paws of rats were inoculated with replication-defective herpes simplex virus (HSV) vectors genetically encoding gad1 gene to express glutamic acid decarboxylase 67 (GAD67), an enzyme that catalyzes the decarboxylation of glutamate to GABA. Mechanical threshold was tested using von Frey filaments before and after treatments with the vectors. The expression of GAD67 in both the lumbar spinal cord and the L4-5 dorsal root ganglia was examined using western blots. The expression of mitochondrial superoxide in the spinal dorsal horn was examined using MitoSox imaging. The immunoreactivity of spinal GABA, pCREB, and pC/EBPß was tested using immunohistochemistry. RESULTS: In the gp120 with ddC-induced neuropathic pain model, GAD67 expression mediated by the HSV vector caused an elevation of mechanical threshold that was apparent on day 3 after vector inoculation. The antiallodynic effect of the single HSV vector inoculation expressing GAD67 lasted >28 days. The area under the time-effect curves in the HSV vector expressing GAD67 was increased compared with that in the control vectors (P = 0.0005). Intrathecal GABA-A/B agonists elevated mechanical threshold in the pain model. The HSV vectors expressing GAD67 reversed the lowered GABA immunoreactivity in the spinal dorsal horn in the neuropathic rats. HSV vectors expressing GAD67 in the neuropathic rats reversed the increased signals of mitochondrial superoxide in the spinal dorsal horn. The vectors expressing GAD67 reversed the upregulated immunoreactivity expression of pCREB and pC/EBPß in the spinal dorsal horn in rats exhibiting NP. CONCLUSIONS: Based on our results, we suggest that GAD67 mediated by HSV vectors acting through the suppression of mitochondrial reactive oxygen species and transcriptional factors in the spinal cord decreases pain in the HIV-related neuropathic pain model, providing preclinical evidence for gene therapy applications in patients with HIV-related pain states.


Subject(s)
Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Glutamate Decarboxylase/genetics , HIV Envelope Protein gp120/metabolism , HIV Infections/complications , Sciatic Nerve/enzymology , Sciatica/therapy , Simplexvirus/genetics , Zalcitabine , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Decarboxylation , Disease Models, Animal , Glutamate Decarboxylase/biosynthesis , Glutamic Acid/metabolism , HIV Envelope Protein gp120/genetics , HIV Infections/genetics , HIV Infections/metabolism , HIV Infections/virology , Humans , Male , Mitochondria/metabolism , Pain Threshold , Phosphorylation , Rats, Sprague-Dawley , Sciatic Nerve/physiopathology , Sciatic Nerve/virology , Sciatica/enzymology , Sciatica/genetics , Sciatica/physiopathology , Sciatica/virology , Simplexvirus/enzymology , Spinal Cord Dorsal Horn/metabolism , Superoxides/metabolism , Time Factors , gamma-Aminobutyric Acid/metabolism
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL