Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Front Cell Infect Microbiol ; 12: 974200, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36081774

RESUMO

Giardia lamblia, a protozoan parasite, is a major cause of waterborne infection, worldwide. While the trophozoite form of this parasite induces pathological symptoms in the gut, the cyst form transmits the infection. Since Giardia is a noninvasive parasite, the actual mechanism by which it causes disease remains elusive. We have previously reported that Giardia assembles cholesterol and GM1 glycosphingolipid-enriched lipid rafts (LRs) that participate in encystation and cyst production. To further delineate the role of LRs in pathogenesis, we isolated LRs from Giardia and subjected them to proteomic analysis. Various cellular proteins including potential virulence factors-e.g., giardins, variant surface proteins, arginine deaminases, elongation factors, ornithine carbomyltransferases, and high cysteine-rich membrane proteins-were found to be present in LRs. Since Giardia secretes virulence factors encapsulated in extracellular vesicles (EVs) that induce proinflammatory responses in hosts, EVs released by the parasite were isolated and subjected to nanoparticle tracking and proteomic analysis. Two types of EV-i.e., small vesicles (SVs; <100 nm, exosome-like particles) and large vesicles (LVs; 100-400 nm, microvesicle-like particles)-were identified and found to contain a diverse group of proteins including above potential virulence factors. Although pretreatment of the parasite with two giardial lipid raft (gLR) disruptors, nystatin (27 µM) and oseltamivir (20 µM), altered the expression profiles of virulence factors in LVs and SVs, the effects were more robust in the case of SVs. To examine the potential role of rafts and vesicles in pathogenicity, Giardia-infected mice were treated with oseltamivir (1.5 and 3.0 mg/kg), and the shedding of cysts were monitored. We observed that this drug significantly reduced the parasite load in mice. Taken together, our results suggest that virulence factors partitioning in gLRs, released into the extracellular milieu via SVs and LVs, participate in spread of giardiasis and could be targeted for future drug development.


Assuntos
Cistos , Giardíase , Animais , Giardia/metabolismo , Giardíase/parasitologia , Microdomínios da Membrana/metabolismo , Camundongos , Oseltamivir , Proteômica , Proteínas de Protozoários/metabolismo , Fatores de Virulência/metabolismo
2.
J Mol Biol ; 433(18): 167111, 2021 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-34153286

RESUMO

5-aza-cytidine (5-aza-C) has been shown to be a potent human immunodeficiency virus type 1 (HIV-1) mutagen that induces G-to-C hypermutagenesis by incorporation of the reduced form (i.e., 5-aza-dC, 5-aza-dCTP). Evidence to date suggests that this lethal mutagenesis is the primary antiretroviral mechanism for 5-aza-C. To investigate the breadth of application of 5-aza-C as an antiretroviral mutagen, we have conducted a comparative, parallel analysis of the antiviral mechanism of 5-aza-C between HIV-1 and gammaretroviruses - i.e., murine leukemia virus (MuLV) and feline leukemia virus (FeLV). Intriguingly, in contrast to the hallmark G-to-C hypermutagenesis observed with HIV-1, MuLV and FeLV did not reveal the presence of a significant increase in mutational burden, particularly that of G-to-C transversion mutations. The effect of 5-aza-dCTP on DNA synthesis revealed that while HIV-1 RT was not inhibited by 5-aza-dCTP even at 100 µM, 5-aza-dCTP was incorporated and significantly inhibited MuLV RT, generating pause sites and reducing the fully extended product. 5-aza-dCTP was found to be incorporated into DNA by MuLV RT or HIV-1 RT, but only acted as a non-obligate chain terminator for MuLV RT. This biochemical data provides an independent line of experimental evidence in support of the conclusion that HIV-1 and MuLV have distinct primary mechanisms of antiretroviral action with 5-aza-C. Taken together, our data provides striking evidence that an antiretroviral mutagen can have strong potency via distinct mechanisms of action among closely related viruses, unlinking antiviral activity from antiviral mechanism of action.


Assuntos
Antivirais/farmacologia , Azacitidina/análogos & derivados , Citidina Trifosfato/análogos & derivados , Infecções por HIV/tratamento farmacológico , Leucemia Experimental/tratamento farmacológico , Mutação/efeitos dos fármacos , Infecções por Retroviridae/tratamento farmacológico , Infecções Tumorais por Vírus/tratamento farmacológico , Animais , Azacitidina/farmacologia , Gatos , Citidina Trifosfato/farmacologia , HIV/efeitos dos fármacos , Infecções por HIV/virologia , Humanos , Vírus da Leucemia Felina/efeitos dos fármacos , Vírus da Leucemia Murina/efeitos dos fármacos , Leucemia Experimental/virologia , Camundongos , Mutagênese , Mutagênicos , Infecções por Retroviridae/virologia , Infecções Tumorais por Vírus/virologia , Replicação Viral
3.
Bioorg Med Chem Lett ; 30(12): 127175, 2020 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-32327222

RESUMO

Novel nucleoside analogues named "triazoxins" were synthesized. Of these, two analogues were found to be highly effective against Giardia lamblia, an intestinal parasite and a major cause of waterborne infection, worldwide. While compound 7 reduced the growth of trophozoites in culture (IC50, ~5 µM), compound 21 blocked the in vitro cyst production (IC50 ~5 µM). Compound 21 was also effective against trophozoites (IC50, ~36 µM). A third analogue (compound 8) was effective against both trophozoites (IC50, ~36 µM) and cysts (IC50, ~20 µM) although at higher concentration. Thus triazoxin analogues are unique and exhibit morphology (i.e., trohozoites or cysts) -specific effects against Giardia.


Assuntos
Anti-Infecciosos/síntese química , Giardia lamblia/efeitos dos fármacos , Giardíase/tratamento farmacológico , Nucleosídeos/síntese química , Anti-Infecciosos/farmacologia , Catálise , Desenho de Fármacos , Humanos , Imidazóis/química , Estrutura Molecular , Nucleosídeos/análogos & derivados , Nucleosídeos/farmacologia , Propanóis/química , Relação Estrutura-Atividade , Trofozoítos/efeitos dos fármacos , Uridina/química
4.
Antiviral Res ; 170: 104540, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31247245

RESUMO

Reverse transcriptase (RT) is an essential enzyme for the replication of retroviruses and hepadnaviruses. Current therapies do not eliminate the intracellular viral replication intermediate termed covalently closed circular (ccc) DNA, which has enhanced interest in hepatitis B virus (HBV) reverse transcription and cccDNA formation. The HBV cccDNA is generated as a plasmid-like episome in the host cell nucleus from the protein-linked relaxed circular (rc) DNA genome in incoming virions during HBV replication. The creation of the cccDNA via conversion from rcDNA remains not fully understood. Here, we sought to investigate whether viral mutagens can effect HBV replication. In particular, we investigated whether nucleoside analogs that act as viral mutagens with retroviruses could impact hepadnaviral DNA synthesis. We observed that a viral mutagen (e.g., 5-aza-2'-deoxycytidine, 5-aza-dC or 5-azacytidine, 5-aza-C) severely diminished the ability of a HBV vector to express a reporter gene following virus transfer and infection of target cells. As predicted, the treatment of 5-aza-dC or 5-aza-C elevated the HBV rcDNA mutation frequency, primarily by increasing the frequency of G-to-C transversion mutations. A reduction in rcDNA synthesis was also observed. Intriguingly, the cccDNA nick/gap region transcription was diminished by 5-aza-dC, but did not enhance viral mutagenesis. Taken together, our results demonstrate that viral mutagens can impact HBV reverse transcription, and propose a model in which viral mutagens can induce mutagenesis during rcDNA formation and diminish viral DNA synthesis during both rcDNA formation and the conversion of rcDNA to cccDNA.


Assuntos
Antivirais/farmacologia , Replicação do DNA/efeitos dos fármacos , Vírus da Hepatite B/efeitos dos fármacos , Vírus da Hepatite B/genética , Mutagênese , Nucleosídeos/farmacologia , Linhagem Celular , DNA Circular/genética , DNA Viral/genética , Células Hep G2 , Hepatócitos/virologia , Humanos , Mutagênicos/farmacologia , Transcrição Reversa/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Replicação Viral/genética
5.
Ann Am Thorac Soc ; 14(6): 1060-1072, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28418689

RESUMO

This report is based on the proceedings from the Inhalational Lung Injury Workshop jointly sponsored by the American Thoracic Society (ATS) and the National Institutes of Health (NIH) Countermeasures Against Chemical Threats (CounterACT) program on May 21, 2013, in Philadelphia, Pennsylvania. The CounterACT program facilitates research leading to the development of new and improved medical countermeasures for chemical threat agents. The workshop was initiated by the Terrorism and Inhalational Disasters Section of the Environmental, Occupational, and Population Health Assembly of the ATS. Participants included both domestic and international experts in the field, as well as representatives from U.S. governmental funding agencies. The meeting objectives were to (1) provide a forum to review the evidence supporting current standard medical therapies, (2) present updates on our understanding of the epidemiology and underlying pathophysiology of inhalational lung injuries, (3) discuss innovative investigative approaches to further delineating mechanisms of lung injury and identifying new specific therapeutic targets, (4) present promising novel medical countermeasures, (5) facilitate collaborative research efforts, and (6) identify challenges and future directions in the ongoing development, manufacture, and distribution of effective and specific medical countermeasures. Specific inhalational toxins discussed included irritants/pulmonary toxicants (chlorine gas, bromine, and phosgene), vesicants (sulfur mustard), chemical asphyxiants (cyanide), particulates (World Trade Center dust), and respirable nerve agents.


Assuntos
Acidentes de Trabalho , Planejamento em Desastres , Desastres , Exposição Ambiental/efeitos adversos , Lesão Pulmonar/induzido quimicamente , Pulmão/fisiopatologia , Animais , Terrorismo Químico , Humanos , Modelos Animais , Sociedades Médicas , Estados Unidos
6.
Ann N Y Acad Sci ; 1374(1): 202-9, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27308865

RESUMO

Cyanide is a metabolic poison that inhibits the utilization of oxygen to form ATP. The consequences of acute cyanide exposure are severe; exposure results in loss of consciousness, cardiac and respiratory failure, hypoxic brain injury, and dose-dependent death within minutes to hours. In a mass-casualty scenario, such as an industrial accident or terrorist attack, currently available cyanide antidotes would leave many victims untreated in the short time available for successful administration of a medical countermeasure. This restricted therapeutic window reflects the rate-limiting step of intravenous administration, which requires both time and trained medical personnel. Therefore, there is a need for rapidly acting antidotes that can be quickly administered to large numbers of people. To meet this need, our laboratory is developing sulfanegen, a potential antidote for cyanide poisoning with a novel mechanism based on 3-mercaptopyruvate sulfurtransferase (3-MST) for the detoxification of cyanide. Additionally, sulfanegen can be rapidly administered by intramuscular injection and has shown efficacy in many species of animal models. This article summarizes the journey from concept to clinical leads for this promising cyanide antidote.


Assuntos
Cianetos/toxicidade , Cisteína/análogos & derivados , Incidentes com Feridos em Massa , Animais , Cisteína/química , Cisteína/farmacologia , Humanos , Cinética , Pró-Fármacos/química , Pró-Fármacos/farmacologia
7.
Retrovirology ; 13: 20, 2016 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-27009333

RESUMO

BACKGROUND: HIV-1 replication kinetics inherently depends on the availability of cellular dNTPs for viral DNA synthesis. In activated CD4(+) T cells and other rapidly dividing cells, the concentrations of dNTPs are high and HIV-1 reverse transcription occurs in an efficient manner. In contrast, nondividing cells such as macrophages have lower dNTP pools, which restricts efficient reverse transcription. Clofarabine is an FDA approved ribonucleotide reductase inhibitor, which has shown potent antiretroviral activity in transformed cell lines. Here, we explore the potency, toxicity and mechanism of action of clofarabine in the human primary HIV-1 target cells: activated CD4(+) T cells and macrophages. RESULTS: Clofarabine is a potent HIV-1 inhibitor in both activated CD4(+) T cells and macrophages. Due to its minimal toxicity in macrophages, clofarabine displays a selectivity index over 300 in this nondividing cell type. The anti-HIV-1 activity of clofarabine correlated with a significant decrease in both cellular dNTP levels and viral DNA synthesis. Additionally, we observed that clofarabine triphosphate was directly incorporated into DNA by HIV-1 reverse transcriptase and blocked processive DNA synthesis, particularly at the low dNTP levels found in macrophages. CONCLUSIONS: Taken together, these data provide strong mechanistic evidence that clofarabine is a dual action inhibitor of HIV-1 replication that both limits dNTP substrates for viral DNA synthesis and directly inhibits the DNA polymerase activity of HIV-1 reverse transcriptase.


Assuntos
Nucleotídeos de Adenina/farmacologia , Fármacos Anti-HIV/farmacologia , Antimetabólitos/farmacologia , Arabinonucleosídeos/farmacologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , HIV-1/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Nucleotídeos de Adenina/toxicidade , Fármacos Anti-HIV/toxicidade , Antimetabólitos/toxicidade , Arabinonucleosídeos/toxicidade , Linfócitos T CD4-Positivos/virologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Clofarabina , HIV-1/fisiologia , Humanos , Macrófagos/virologia , Replicação Viral/efeitos dos fármacos
8.
Artigo em Inglês | MEDLINE | ID: mdl-26655110

RESUMO

The current suite of Food and Drug Administration (FDA) approved antidotes (i.e., sodium nitrite, sodium thiosulfate, and hydroxocobalamin) are effective for treating cyanide poisoning, but individually, each antidote has major limitations (e.g., large effective dosage or delayed onset of action). To mitigate these limitations, next-generation cyanide antidotes are being investigated, including 3-mercaptopyruvate (3-MP) and cobinamide (Cbi). Analytical methods capable of detecting these therapeutics individually and simultaneously (for combination therapy) are essential for the development of 3-MP and Cbi as potential cyanide antidotes. Therefore, a liquid chromatography-tandem mass-spectrometry method for the simultaneous analysis of 3-MP and Cbi was developed. Sample preparation of 3-MP consisted of spiking plasma with an internal standard ((13)C3-3-MP), precipitation of plasma proteins, and derivatizing 3-MP with monobromobimane to produce 3-mercaptopyruvate-bimane. Preparation of Cbi involved denaturing plasma proteins with simultaneous addition of excess cyanide to convert each Cbi species to dicyanocobinamide (Cbi(CN)2). The limits of detection for 3-MP and Cbi were 0.5µM and 0.2µM, respectively. The linear ranges were 2-500µM for 3-MP and 0.5-50µM for Cbi. The accuracy and precision for 3-MP were 100±9% and <8.3% relative standard deviation (RSD), respectively. For Cbi(CN)2, the accuracy was 100±13% and the precision was <9.5% RSD. The method presented here was used to determine 3-MP and Cbi from treated animals and may ultimately facilitate FDA approval of these antidotes for treatment of cyanide poisoning.


Assuntos
Cromatografia Líquida/métodos , Cobamidas/sangue , Cisteína/análogos & derivados , Espectrometria de Massas em Tandem/métodos , Animais , Cisteína/sangue , Limite de Detecção , Suínos
9.
Artigo em Inglês | MEDLINE | ID: mdl-24480329

RESUMO

Accidental or intentional cyanide poisoning is a serious health risk. The current suite of FDA approved antidotes, including hydroxocobalamin, sodium nitrite, and sodium thiosulfate is effective, but each antidote has specific major limitations, such as large effective dosage or delayed onset of action. Therefore, next generation cyanide antidotes are being investigated to mitigate these limitations. One such antidote, 3-mercaptopyruvate (3-MP), detoxifies cyanide by acting as a sulfur donor to convert cyanide into thiocyanate, a relatively nontoxic cyanide metabolite. An analytical method capable of detecting 3-MP in biological fluids is essential for the development of 3-MP as a potential antidote. Therefore, a high performance liquid chromatography tandem mass spectrometry (HPLC-MS-MS) method was established to analyze 3-MP from rabbit plasma. Sample preparation consisted of spiking the plasma with an internal standard ((13)C3-3-MP), precipitation of plasma proteins, and reaction with monobromobimane to inhibit the characteristic dimerization of 3-MP. The method produced a limit of detection of 0.1µM, a linear dynamic range of 0.5-100µM, along with excellent linearity (R(2)≥0.999), accuracy (±9% of the nominal concentration) and precision (<7% relative standard deviation). The optimized HPLC-MS-MS method was capable of detecting 3-MP in rabbits that were administered sulfanegen, a prodrug of 3-MP, following cyanide exposure. Considering the excellent performance of this method, it will be utilized for further investigations of this promising cyanide antidote.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Cisteína/análogos & derivados , Animais , Cisteína/sangue , Cisteína/química , Estabilidade de Medicamentos , Limite de Detecção , Modelos Lineares , Coelhos , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem/métodos
10.
Bioorg Med Chem ; 21(22): 7222-8, 2013 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-24120088

RESUMO

The nucleoside analog 5,6-dihydro-5-aza-2'-deoxycytidine (KP-1212) has been investigated as a first-in-class lethal mutagen of human immunodeficiency virus type-1 (HIV-1). Since a prodrug monotherapy did not reduce viral loads in Phase II clinical trials, we tested if ribonucleotide reductase inhibitors (RNRIs) combined with KP-1212 would improve antiviral activity. KP-1212 potentiated the activity of gemcitabine and resveratrol and simultaneously increased the viral mutant frequency. G-to-C mutations predominated with the KP-1212-resveratrol combination. These observations represent the first demonstration of a mild anti-HIV-1 mutagen potentiating the antiretroviral activity of RNRIs and encourage the clinical translation of enhanced viral mutagenesis in treating HIV-1 infection.


Assuntos
Fármacos Anti-HIV/química , Fármacos Anti-HIV/farmacologia , Desoxicitidina/análogos & derivados , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , HIV-1/efeitos dos fármacos , Ribonucleotídeo Redutases/antagonistas & inibidores , Proteínas Virais/antagonistas & inibidores , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Desoxicitidina/química , Desoxicitidina/farmacologia , Genes Reporter/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , HIV-1/enzimologia , HIV-1/genética , Humanos , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Mutação , Resveratrol , Ribonucleotídeo Redutases/metabolismo , Estilbenos/química , Estilbenos/farmacologia , Proteínas Virais/metabolismo , Proteína Vermelha Fluorescente
11.
J Med Chem ; 56(23): 9403-14, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23914828

RESUMO

While mutation is the driving force behind evolution, most mutations are detrimental; therefore, elevating the mutation rate of a virus should diminish fitness. Because riboviruses and retroviruses have high mutation rates, a small increase in their mutation rates could exceed their threshold of viability. This approach, elevation of the viral mutation rate beyond the threshold of viability, extinction catastrophe or lethal mutagenesis, was proposed over a decade ago as a novel chemotherapeutic strategy. Extinction catastrophe induced by promutagenic nucleosides has been demonstrated in cell culture models, but most mutagens are carcinogenic and are poorly tolerated. Thus, clinical translation of viral mutagens has been difficult, casting doubt on the clinical viability of this strategy. This Perspective covers recent advances in the use of promutagenic nucleosides and the Vif-APOBEC interaction as chemotherapeutic strategies for targeting viral mutation rates.


Assuntos
Antivirais/farmacologia , Mutagênese , Replicação Viral/efeitos dos fármacos , Vírus/genética , Desaminase APOBEC-1 , Citidina Desaminase/antagonistas & inibidores , HIV/efeitos dos fármacos , Conformação Molecular/efeitos dos fármacos , Mutagênese/efeitos dos fármacos , Mutagênicos/farmacologia , Nucleosídeos/química , Nucleosídeos/farmacologia , Relação Estrutura-Atividade
12.
J Med Chem ; 56(3): 1346-9, 2013 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-23301495

RESUMO

Current cyanide antidotes are administered by IV infusion, which is suboptimal for mass casualties. Therefore, in a cyanide disaster, intramuscular (IM) injectable antidotes would be more appropriate. We report the discovery of the highly water-soluble sulfanegen triethanolamine as a promising lead for development as an IM injectable cyanide antidote.


Assuntos
Antídotos/uso terapêutico , Cianetos/intoxicação , Cisteína/análogos & derivados , Incidentes com Feridos em Massa , Quinolizinas/uso terapêutico , Compostos de Enxofre/uso terapêutico , Cisteína/química , Humanos , Injeções Intramusculares , Intoxicação/tratamento farmacológico , Sais
13.
Anesth Analg ; 114(5): 956-61, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22392971

RESUMO

BACKGROUND: Cyanide (CN) toxicity is a serious clinical problem and can occur with sodium nitroprusside (SNP) administration, accidental smoke inhalation, industrial mishaps, and bio-terrorism. In this study, we induced severe CN toxicity independently with SNP or sodium cyanide (NaCN) in a juvenile pig model to demonstrate reversal of severe CN toxicity with a new antidote, sulfanegen sodium, a prodrug of 3-mercaptopyruvate. METHODS: SNP study: A pilot study in 11 anesthetized, mechanically ventilated juvenile pigs allowed us to determine the dose of SNP to induce CN toxicity. Blood CN, serum lactates, and blood gases were monitored. CN toxicity was defined as the occurrence of severe lactic acidosis accompanied by significant elevation in blood CN levels. Based on this pilot study, 8 anesthetized pigs received a high-dose i.v. infusion of SNP (100 mg/h) for 2 hours to induce CN toxicity. They were then randomized to receive either sulfanegen sodium or placebo. Four pigs received 3 doses of sulfanegen sodium (2.5 g i.v.) every hour after induction of severe CN toxicity, and 4 pigs received placebo. NaCN study: A pilot study was conducted in 4 spontaneously ventilating pigs sedated with propofol plus ketamine to demonstrate hemodynamic and metabolic stability for several hours. After this, 6 pigs were similarly sedated and given NaCN in bolus aliquots to produce CN toxicity ultimately resulting in death. Hemodynamics and metabolic variables were followed to define peak CN toxicity. In another group of 6 pigs, severe CN toxicity was induced by this method, and at peak toxicity, the animals were given sulfanegen sodium (2.5 g i.v.) followed by a repeat dose 60 minutes later in surviving animals. RESULTS: SNP study: The pilot study demonstrated the occurrence of a significant increase in blood CN levels (P < 0.05) accompanied by severe lactic acidemia (P < 0.05) in all pigs receiving a high dose of SNP. Administration of the sulfanegen antidote resulted in progressive significant reduction in blood lactate and CN levels with 100% survival (P < 0.05), whereas the placebo-treated pigs deteriorated and did not survive (P < 0.05). NaCN study: NaCN injection resulted in CN toxicity accompanied by severe lactic acidosis and mortality in all the pigs. Sulfanegen sodium reversed this toxicity and prevented mortality in all the pigs treated with this antidote. CONCLUSIONS: CN toxicity can be successfully induced in a juvenile pig model with SNP or NaCN. The prodrug, sulfanegen sodium, is effective in reversing CN toxicity induced by SNP or NaCN.


Assuntos
Cianetos/antagonistas & inibidores , Cianetos/toxicidade , Cisteína/análogos & derivados , Compostos Heterocíclicos com 1 Anel/farmacologia , Pró-Fármacos/farmacologia , Animais , Gasometria , Pressão Sanguínea/efeitos dos fármacos , Pressão Venosa Central/efeitos dos fármacos , Cianetos/sangue , Cisteína/farmacologia , Frequência Cardíaca/efeitos dos fármacos , Concentração de Íons de Hidrogênio , Ácido Láctico/sangue , Nitroprussiato/efeitos adversos , Projetos Piloto , Artéria Pulmonar/efeitos dos fármacos , Suínos , Vasodilatadores/efeitos adversos
14.
Antimicrob Agents Chemother ; 56(4): 1942-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22271861

RESUMO

The emergence of drug resistance threatens to limit the use of current anti-HIV-1 drugs and highlights the need to expand the number of treatment options available for HIV-1-infected individuals. Our previous studies demonstrated that two clinically approved drugs, decitabine and gemcitabine, potently inhibited HIV-1 replication in cell culture through a mechanism that is distinct from the mechanisms for the drugs currently used to treat HIV-1 infection. We further demonstrated that gemcitabine inhibited replication of a related retrovirus, murine leukemia virus (MuLV), in vivo using the MuLV-based LP-BM5/murine AIDS (MAIDS) mouse model at doses that were not toxic. Since decitabine and gemcitabine inhibited MuLV and HIV-1 replication with similar potency in cell culture, the current study examined the efficacy and toxicity of the drug combination using the MAIDS model. The data demonstrate that the drug combination inhibited disease progression, as detected by histopathology, viral loads, and spleen weights, at doses lower than those that would be required if the drugs were used individually. The combination of decitabine and gemcitabine exerted antiviral activity at doses that were not toxic. These findings indicate that the combination of decitabine and gemcitabine shows potent antiretroviral activity at nontoxic doses and should be further investigated for clinical relevance.


Assuntos
Fármacos Anti-HIV/uso terapêutico , Azacitidina/análogos & derivados , Desoxicitidina/análogos & derivados , HIV-1/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Animais , Fármacos Anti-HIV/efeitos adversos , Azacitidina/uso terapêutico , Peso Corporal/efeitos dos fármacos , Células Cultivadas , Doença Hepática Induzida por Substâncias e Drogas/patologia , Decitabina , Desoxicitidina/efeitos adversos , Desoxicitidina/uso terapêutico , Combinação de Medicamentos , Sinergismo Farmacológico , Feminino , Citometria de Fluxo , Humanos , Fígado/patologia , Linfonodos/patologia , Linfonodos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Síndrome de Imunodeficiência Adquirida Murina/patologia , Síndrome de Imunodeficiência Adquirida Murina/virologia , Provírus/efeitos dos fármacos , Baço/patologia , Baço/virologia , Linfócitos T/efeitos dos fármacos , Transfecção , Gencitabina
15.
J Gen Virol ; 93(Pt 4): 900-905, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22258856

RESUMO

Feline leukemia virus (FeLV) is a gammaretrovirus that is a significant cause of neoplastic-related disorders affecting cats worldwide. Treatment options for FeLV are limited, associated with serious side effects, and can be cost-prohibitive. The development of drugs used to treat a related retrovirus, human immunodeficiency virus type 1 (HIV-1), has been rapid, leading to the approval of five drug classes. Although structural differences affect the susceptibility of gammaretroviruses to anti-HIV drugs, the similarities in mechanism of replication suggest that some anti-HIV-1 drugs may also inhibit FeLV. This study demonstrates the anti-FeLV activity of four drugs approved by the US FDA (Food and Drug Administration) at non-toxic concentrations. Of these, tenofovir and raltegravir are anti-HIV-1 drugs, while decitabine and gemcitabine are approved to treat myelodysplastic syndromes and pancreatic cancer, respectively, but also have anti-HIV-1 activity in cell culture. Our results indicate that these drugs may be useful for FeLV treatment and should be investigated for mechanism of action and suitability for veterinary use.


Assuntos
Antivirais/uso terapêutico , Doenças do Gato/virologia , Vírus da Leucemia Felina/efeitos dos fármacos , Infecções por Retroviridae/veterinária , Infecções Tumorais por Vírus/veterinária , Adenina/análogos & derivados , Adenina/uso terapêutico , Animais , Azacitidina/análogos & derivados , Azacitidina/uso terapêutico , Doenças do Gato/tratamento farmacológico , Gatos , Decitabina , Desoxicitidina/análogos & derivados , Desoxicitidina/uso terapêutico , Relação Dose-Resposta a Droga , Leucemia Felina/tratamento farmacológico , Organofosfonatos/uso terapêutico , Pirrolidinonas/uso terapêutico , Raltegravir Potássico , Infecções por Retroviridae/tratamento farmacológico , Tenofovir , Infecções Tumorais por Vírus/tratamento farmacológico , Gencitabina
16.
Clin Toxicol (Phila) ; 49(5): 366-73, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21740135

RESUMO

CONTEXT: Cyanide is a component of smoke in residential and industrial fires, and accidental exposure to cyanide occurs in a variety of industries. Moreover, cyanide has the potential to be used by terrorists, particularly in a closed space such as an airport or train station. Current therapies for cyanide poisoning must be given by intravenous administration, limiting their use in treating mass casualties. OBJECTIVE: We are developing two new cyanide antidotes--cobinamide, a vitamin B(12) analog, and sulfanegen, a 3-mercaptopyruvate prodrug. Both drugs can be given by intramuscular administration, and therefore could be used to treat a large number of people quickly. We now asked if the two drugs would have an augmented effect when combined. MATERIALS AND METHODS: We used a non-lethal and two different lethal models of cyanide poisoning in mice. The non-lethal model assesses neurologic recovery by quantitatively evaluating the innate righting reflex time of a mouse. The two lethal models are a cyanide injection and a cyanide inhalation model. RESULTS: We found that the two drugs are at least additive when used together in both the non-lethal and lethal models: at doses where all animals died with either drug alone, the combination yielded 80 and 40% survival in the injection and inhalation models, respectively. Similarly, drug doses that yielded 40% survival with either drug alone, yielded 80 and 100% survival in the injection and inhalation models, respectively. As part of the inhalation model, we developed a new paradigm in which animals are exposed to cyanide gas, injected intramuscularly with an antidote, and then re-exposed to cyanide gas. This simulates cyanide exposure of a large number of people in a closed space, because people would remain exposed to cyanide, even after receiving an antidote. CONCLUSION: The combination of cobinamide and sulfanegen shows great promise as a new approach to treating cyanide poisoning.


Assuntos
Antídotos/administração & dosagem , Cobamidas/administração & dosagem , Cianetos/intoxicação , Cisteína/análogos & derivados , Pró-Fármacos/administração & dosagem , Animais , Cisteína/administração & dosagem , Modelos Animais de Doenças , Quimioterapia Combinada , Masculino , Camundongos , Camundongos Endogâmicos C57BL
17.
Ther Clin Risk Manag ; 7: 115-22, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21479142

RESUMO

Antiretroviral drugs have saved and extended the lives of millions of individuals infected with HIV. The major classes of anti-HIV drugs include reverse transcriptase inhibitors, protease inhibitors, integrase inhibitors, and entry/fusion inhibitors. While antiretroviral drug regimens are not commonly used to treat other types of retroviral infections, there are instances where there is a perceived need for re-evaluation of the benefits of antiretroviral therapy. One case in point is that of feline leukemia virus (FeLV), an infection of companion felines. While vaccines exist to prevent FeLV infection and spread, they have not eliminated FeLV infection. For FeLV-infected felines and their human companions, antiretroviral therapy would be desirable and of practical importance if good options were available. Here, we discuss FeLV biology and current treatment options, and propose that there is a need for antiretroviral treatment options for FeLV infection. The comparative use and analysis of antiretroviral therapy can provide new insights into the mechanism of antiretroviral drug action.

18.
PLoS One ; 6(1): e15840, 2011 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-21264291

RESUMO

Replication of retroviral and host genomes requires ribonucleotide reductase to convert rNTPs to dNTPs, which are then used as substrates for DNA synthesis. Inhibition of ribonucleotide reductase by hydroxyurea (HU) has been previously used to treat cancers as well as HIV. However, the use of HU as an antiretroviral is limited by its associated toxicities such as myelosuppression and hepatotoxicity. In this study, we examined the ribonucleotide reductase inhibitor, gemcitabine, both in cell culture and in C57Bl/6 mice infected with LP-BM5 murine leukemia virus (LP-BM5 MuLV, a murine AIDS model). Gemcitabine decreased infectivity of MuLV in cell culture with an EC50 in the low nanomolar range with no detectable cytotoxicity. Similarly, gemcitabine significantly decreased disease progression in mice infected with LP-BM5. Specifically, gemcitabine treatment decreased spleen size, plasma IgM, and provirus levels compared to LP-BM5 MuLV infected, untreated mice. Gemcitabine efficacy was observed at doses as low as 1 mg/kg/day in the absence of toxicity. Higher doses of gemcitabine (3 mg/kg/day and higher) were associated with toxicity as determined by a loss in body mass. In summary, our findings demonstrate that gemcitabine has antiretroviral activity ex vivo and in vivo in the LP-BM5 MuLV model. These observations together with a recent ex vivo study with HIV-1, suggest that gemcitabine has broad antiretroviral activity and could be particularly useful in vivo when used in combination drug therapy.


Assuntos
Antirretrovirais/farmacologia , Desoxicitidina/análogos & derivados , Vírus da Leucemia Murina/efeitos dos fármacos , Síndrome de Imunodeficiência Adquirida Murina/tratamento farmacológico , Animais , Células Cultivadas , Desoxicitidina/farmacologia , Desoxicitidina/uso terapêutico , Progressão da Doença , Imunoglobulina M/sangue , Camundongos , Camundongos Endogâmicos C57BL , Ribonucleotídeo Redutases/antagonistas & inibidores , Baço/patologia , Carga Viral/efeitos dos fármacos , Gencitabina
19.
J Virol ; 84(18): 9301-9, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20610712

RESUMO

The development of HIV drugs is an expensive and a lengthy process. In this study, we used drug repositioning, a process whereby a drug approved to treat one condition is used to treat a different condition, to identify clinically approved drugs that have anti-HIV activity. The data presented here show that a combination of two clinically approved drugs, decitabine and gemcitabine, reduced HIV infectivity by 73% at concentrations that had minimal antiviral activity when used individually. Decreased infectivity coincided with a significant increase in mutation frequency and a shift in the HIV mutation spectrum. These results indicate that an increased mutational load is the primary antiviral mechanism for inhibiting the generation of infectious progeny virus from provirus. Similar results were seen when decitabine was used in combination with another ribonucleotide reductase inhibitor. Our results suggest that HIV infectivity can be decreased by combining a nucleoside analog that forms noncanonical base pairs with certain ribonucleotide reductase inhibitors. Such drug combinations are relevant since members of these drug classes are used clinically. Our observations support a model in which increased mutation frequency decreases infectivity through lethal mutagenesis.


Assuntos
Fármacos Anti-HIV/farmacologia , Azacitidina/análogos & derivados , Desoxicitidina/análogos & derivados , HIV/efeitos dos fármacos , Azacitidina/farmacologia , Decitabina , Desoxicitidina/farmacologia , Interações Medicamentosas , Quimioterapia Combinada/métodos , Infecções por HIV/tratamento farmacológico , Humanos , Mutação , Gencitabina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA