Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Clin Toxicol (Phila) ; 61(4): 212-222, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-37010385

RESUMO

CONTEXT: The azide anion (N3-) is highly toxic. It exists most commonly as sodium azide, which is used widely and is readily available, raising the potential for occupational incidents and use as a weapon of mass destruction. Azide-poisoned patients present with vomiting, seizures, hypotension, metabolic acidosis, and coma; death can occur. No specific azide antidote exists, with treatment being solely supportive. Azide inhibits mitochondrial cytochrome c oxidase and is likely oxidized to nitric oxide in vivo. Cytochrome c oxidase inhibition depletes intracellular adenosine triphosphate and increases oxidative stress, while increased nitric oxide causes hypotension and exacerbates oxidative damage. Here, we tested whether the cobalamin (vitamin B12) analog cobinamide, a strong and versatile antioxidant that also neutralizes nitric oxide, can reverse azide toxicity in mammalian cells, Drosophila melanogaster, and mice. RESULTS: We found cobinamide bound azide with a moderate affinity (Ka 2.87 × 105 M-1). Yet, cobinamide improved growth, increased intracellular adenosine triphosphate, and reduced apoptosis and malondialdehyde, a marker of oxidative stress, in azide-exposed cells. Cobinamide rescued Drosophila melanogaster and mice from lethal exposure to azide and was more effective than hydroxocobalamin. Azide likely generated nitric oxide in the mice, as evidenced by increased serum nitrite and nitrate, and reduced blood pressure and peripheral body temperature in the animals; the reduced temperature was likely due to reflex vasoconstriction in response to the hypotension. Cobinamide improved recovery of both blood pressure and body temperature. CONCLUSION: We conclude cobinamide likely acted by neutralizing both oxidative stress and nitric oxide, and that it should be given further consideration as an azide antidote.


Assuntos
Hipotensão , Vitamina B 12 , Camundongos , Animais , Drosophila melanogaster/metabolismo , Azidas/metabolismo , Antídotos/farmacologia , Óxido Nítrico , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Cobamidas , Trifosfato de Adenosina , Vitaminas , Mamíferos/metabolismo
2.
Chem Res Toxicol ; 35(11): 1983-1996, 2022 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-36201358

RESUMO

The development of rapidly acting cyanide countermeasures using intramuscular injection (IM) represents an unmet medical need to mitigate toxicant exposures in mass casualty settings. Previous work established that cisplatin and other platinum(II) or platinum(IV)-based agents effectively mitigate cyanide toxicity in zebrafish. Cyanide's in vivo reaction with platinum-containing materials was proposed to reduce the risk of acute toxicities. However, cyanide antidote activity depended on a formulation of platinum-chloride salts with dimethyl sulfoxide (DMSO) followed by dilution in phosphate-buffered saline (PBS). A working hypothesis to explain the DMSO requirement is that the formation of platinum-sulfoxide complexes activates the cyanide scavenging properties of platinum. Preparations of isolated NaPtCl5-DMSO and Na (NH3)2PtCl-DMSO complexes in the absence of excess DMSO provided agents with enhanced reactivity toward cyanide in vitro and fully recapitulated in vivo cyanide rescue in zebrafish and mouse models. The enhancement of the cyanide scavenging effects of the DMSO ligand could be attributed to the activation of platinum(IV) and (II) with a sulfur ligand. Unfortunately, the efficacy of DMSO complexes was not robust when administered IM. Alternative Pt(II) materials containing sulfide and amine ligands in bidentate complexes show enhanced reactivity toward cyanide addition. The cyanide addition products yielded tetracyanoplatinate(II), translating to a stoichiometry of 1:4 Pt to each cyanide scavenger. These new agents demonstrate a robust and enhanced potency over the DMSO-containing complexes using IM administration in mouse and rabbit models of cyanide toxicity. Using the zebrafish model with these Pt(II) complexes, no acute cardiotoxicity was detected, and dose levels required to reach lethality exceeded 100 times the effective dose. Data are presented to support a general chemical design approach that can expand a new lead candidate series for developing next-generation cyanide countermeasures.


Assuntos
Antineoplásicos , Platina , Camundongos , Coelhos , Animais , Platina/química , Peixe-Zebra , Cianetos , Dimetil Sulfóxido , Ligantes , Sulfetos , Antineoplásicos/farmacologia
3.
Clin Toxicol (Phila) ; 60(5): 615-622, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-34989638

RESUMO

CONTEXT: Methyl mercaptan (CH3SH) is a colorless, toxic gas with potential for occupational exposure and used as a weapon of mass destruction. Inhalation at high concentrations can result in dyspnea, hypoventilation, seizures, and death. No specific methyl mercaptan antidote exists, highlighting a critical need for such an agent. Here, we investigated the mechanism of CH3SH toxicity, and rescue from CH3SH poisoning by the vitamin B12 analog cobinamide, in mammalian cells. We also developed lethal CH3SH inhalation models in mice and rabbits, and tested the efficacy of intramuscular injection of cobinamide as a CH3SH antidote. RESULTS: We found that cobinamide binds to CH3SH (Kd = 84 µM), and improved growth of cells exposed to CH3SH. CH3SH reduced cellular oxygen consumption and intracellular ATP content and activated the stress protein c-Jun N-terminal kinase (JNK); cobinamide reversed these changes. A single intramuscular injection of cobinamide (20 mg/kg) rescued 6 of 6 mice exposed to a lethal dose of CH3SH gas, while all six saline-treated mice died (p = 0.0013). In rabbits exposed to CH3SH gas, 11 of 12 animals (92%) treated with two intramuscular injections of cobinamide (50 mg/kg each) survived, while only 2 of 12 animals (17%) treated with saline survived (p = 0.001). CONCLUSION: We conclude that cobinamide could potentially serve as a CH3SH antidote.


Assuntos
Antídotos , Cobamidas , Animais , Antídotos/uso terapêutico , Chlorocebus aethiops , Humanos , Camundongos , Coelhos , Compostos de Sulfidrila , Vitamina B 12
4.
Sci Rep ; 10(1): 19907, 2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-33199755

RESUMO

Cisplatin is a mainstay of cancer chemotherapy. It forms DNA adducts, thereby activating poly(ADP-ribose) polymerases (PARPs) to initiate DNA repair. The PARP substrate NAD+ is synthesized from 5-phosphoribose-1-pyrophosphate (PRPP), and we found that treating cells for 6 h with cisplatin reduced intracellular PRPP availability. The decrease in PRPP was likely from (1) increased PRPP consumption, because cisplatin increased protein PARylation and PARP1 shRNA knock-down returned PRPP towards normal, and (2) decreased intracellular phosphate, which down-regulated PRPP synthetase activity. Depriving cells of a single essential amino acid decreased PRPP synthetase activity with a half-life of ~ 8 h, and combining cisplatin and amino acid deprivation synergistically reduced intracellular PRPP. PRPP is a rate-limiting substrate for purine nucleotide synthesis, and cisplatin inhibited de novo purine synthesis and DNA synthesis, with amino acid deprivation augmenting cisplatin's effects. Amino acid deprivation enhanced cisplatin's cytotoxicity, increasing cellular apoptosis and DNA strand breaks in vitro, and intermittent deprivation of lysine combined with a sub-therapeutic dose of cisplatin inhibited growth of ectopic hepatomas in mice. Augmentation of cisplatin's biochemical and cytotoxic effects by amino acid deprivation suggest that intermittent deprivation of an essential amino acid could allow dose reduction of cisplatin; this could reduce the drug's side effects, and allow its use in cisplatin-resistant tumors.


Assuntos
Aminoácidos/deficiência , Apoptose , Carcinoma Hepatocelular/patologia , Cisplatino/farmacologia , Neoplasias Hepáticas/patologia , Fosforribosil Pirofosfato/metabolismo , Animais , Antineoplásicos/farmacologia , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/metabolismo , Proliferação de Células , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/metabolismo , Masculino , Camundongos , Camundongos Nus , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Cell Chem Biol ; 24(5): 565-575.e4, 2017 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-28416275

RESUMO

Cisplatin holds an illustrious position in the history of chemistry most notably for its role in the virtual cure of testicular cancer. Here we describe a role for this small molecule in cyanide detoxification in vivo. Cyanide kills organisms as diverse as insects, fish, and humans within seconds to hours. Current antidotes exhibit limited efficacy and are not amenable to mass distribution requiring the development of new classes of antidotes. The binding affinity of the cyanide anion for the positively charged metal platinum is known to create an extremely stable complex in vitro. We therefore screened a panel of diverse cisplatin analogs and identified compounds that conferred protection from cyanide poisoning in zebrafish, mice, and rabbits. Cumulatively, this discovery pipeline begins to establish the characteristics of platinum ligands that influence their solubility, toxicity, and efficacy, and provides proof of concept that platinum-based complexes are effective antidotes for cyanide poisoning.


Assuntos
Antídotos/química , Antídotos/farmacologia , Cisplatino/análogos & derivados , Cisplatino/farmacologia , Cianetos/intoxicação , Animais , Antídotos/metabolismo , Linhagem Celular , Cisplatino/metabolismo , Cianetos/química , Cianetos/metabolismo , Aprovação de Drogas , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Humanos , Dose Letal Mediana , Oxirredução/efeitos dos fármacos , Coelhos , Solubilidade , Enxofre/química , Peixe-Zebra
6.
Nat Commun ; 6: 7389, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-26067754

RESUMO

Mutant p53 (mtp53) is an oncogene that drives cancer cell proliferation. Here we report that mtp53 associates with the promoters of numerous nucleotide metabolism genes (NMG). Mtp53 knockdown reduces NMG expression and substantially depletes nucleotide pools, which attenuates GTP-dependent protein activity and cell invasion. Addition of exogenous guanosine or GTP restores the invasiveness of mtp53 knockdown cells, suggesting that mtp53 promotes invasion by increasing GTP. In addition, mtp53 creates a dependency on the nucleoside salvage pathway enzyme deoxycytidine kinase for the maintenance of a proper balance in dNTP pools required for proliferation. These data indicate that mtp53-harbouring cells have acquired a synthetic sick or lethal phenotype relationship with the nucleoside salvage pathway. Finally, elevated expression of NMG correlates with mutant p53 status and poor prognosis in breast cancer patients. Thus, mtp53's control of nucleotide biosynthesis has both a driving and sustaining role in cancer development.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica/genética , Nucleotídeos/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Western Blotting , Neoplasias Encefálicas/secundário , Neoplasias da Mama/metabolismo , Ciclo Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Desoxicitidina Quinase , Feminino , Técnicas de Silenciamento de Genes , Guanosina Trifosfato , Humanos , Imunoprecipitação , Estimativa de Kaplan-Meier , Camundongos , Mutação , Invasividade Neoplásica/genética , Transplante de Neoplasias , Nucleosídeos/metabolismo , Prognóstico , Regiões Promotoras Genéticas , Modelos de Riscos Proporcionais , Ensaio Tumoral de Célula-Tronco , Proteína Supressora de Tumor p53/metabolismo
7.
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
8.
J Infect Dis ; 197(3): 457-64, 2008 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18199034

RESUMO

Some Pseudomonas aeruginosa strains are cyanogenic, and cyanide may contribute to the bacterium's virulence. Using human isolates of P. aeruginosa, we have shown that Drosophila melanogaster suspended above cyanogenic strains become motionless and develop bradycardia and that flies injected with cyanogenic bacterial strains die more rapidly than those injected with noncyanogenic strains. Flies exposed to cyanogenic strains had high cyanide and low adenosine triphosphate (ATP) concentrations in body extracts, and treatment with a cyanide antidote equalized survival of flies injected with cyanogenic and noncyanogenic strains. P. aeruginosa PAO1 strain with a mutation in the hydrogen cyanide synthase gene cluster was much less toxic to flies than the parental cyanogenic strain or 2 knock-in strains. Transgenic flies overexpressing rhodanese, which detoxifies cyanide by converting it to thiocyanate, were resistant to cyanide and the increased virulence of cyanogenic strains. We conclude that D. melanogaster is a good model for studying cyanide produced by P. aeruginosa.


Assuntos
Cianetos/isolamento & purificação , Cianetos/toxicidade , Drosophila melanogaster/efeitos dos fármacos , Pseudomonas aeruginosa/isolamento & purificação , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Drosophila melanogaster/classificação , Frequência Cardíaca/efeitos dos fármacos , Humanos , Nitratos/farmacologia , Nitritos/farmacologia , Pseudomonas aeruginosa/fisiologia , Tiocianatos/isolamento & purificação , Tiocianatos/metabolismo , Tiocianatos/farmacologia
9.
Exp Biol Med (Maywood) ; 232(6): 789-98, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17526771

RESUMO

Sodium nitroprusside is used to treat hypertensive emergencies and acute heart failure. It acts by releasing nitric oxide (NO), a highly potent vasodilator, but unfortunately, for each NO molecule released, five cyanide ions are released. Thus, nitroprusside therapy is limited by cyanide toxicity. Therefore, a cyanide scavenger could be beneficial when administering nitroprusside. Hydroxocobalamin, which has a relatively high binding affinity for cyanide, has been shown to reduce cyanide levels in nitroprusside-treated patients. Cobinamide, the penultimate precursor in hydroxocobalamin biosynthesis, has a much greater affinity for cyanide than cobalamin, and binds two cyanide ions. We now show that cobinamide is highly effective in neutralizing cyanide ions released by nitroprusside in cultured mammalian cells, Drosophila melanogaster, and mice. Cobinamide also binds NO, but at molar concentrations 2.5-5 times that of nitroprusside, it did not decrease NO concentrations or the physiological effectiveness of nitroprusside. We conclude that cobinamide could be a valuable adjunct to nitroprusside therapy.


Assuntos
Anti-Hipertensivos/farmacocinética , Cobamidas/farmacologia , Cianetos/metabolismo , Óxido Nítrico/metabolismo , Nitroprussiato/farmacocinética , Trifosfato de Adenosina/metabolismo , Animais , Anti-Hipertensivos/farmacologia , Células Cultivadas , Drosophila melanogaster , Frequência Cardíaca/efeitos dos fármacos , Masculino , Túbulos de Malpighi/efeitos dos fármacos , Túbulos de Malpighi/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/metabolismo , Nitratos/metabolismo , Nitritos/metabolismo , Nitroprussiato/farmacologia , Consumo de Oxigênio/efeitos dos fármacos , Artéria Pulmonar/citologia , Ratos , Tiocianatos/sangue , Tiocianatos/urina , Vitamina B 12
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA