Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
ACS Med Chem Lett ; 14(8): 1088-1094, 2023 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-37583812

RESUMO

Glutamate plays a key role in cognition and mood, and it has been shown that inhibiting ionotropic glutamate receptors disrupts cognition, while enhancing ionotropic receptor activity is pro-cognitive. One approach to elevating glutamatergic tone has been to antagonize presynaptic metabotropic glutamate receptor 2 (mGluR2). A desire for selectivity over the largely homologous mGluR3 motivated a strategy to achieve selectivity through the identification of mGluR2 negative allosteric modulators (NAMs). Extensive screening and optimization efforts led to the identification of a novel series of 4-arylquinoline-2-carboxamides. This series was optimized for mGluR2 NAM potency, clean off-target activity, and desirable physical properties, which resulted in the identification of improved C4 and C7 substituents. The initial lead compound from this series was Ames-positive in a single strain with metabolic activation, indicating that a reactive metabolite was likely responsible for the genetic toxicity. Metabolic profiling and Ames assessment across multiple analogs identified key structure-activity relationships associated with Ames positivity. Further optimization led to the Ames-negative mGluR2 negative allosteric modulator MK-8768.

2.
Channels (Austin) ; 16(1): 230-243, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36239534

RESUMO

As part of a drug discovery effort to identify potent inhibitors of NaV1.7 for the treatment of pain, we observed that inhibitors produced unexpected cardiovascular and respiratory effects in vivo. Specifically, inhibitors administered to rodents produced changes in cardiovascular parameters and respiratory cessation. We sought to determine the mechanism of the in vivo adverse effects by studying the selectivity of the compounds on NaV1.5, NaV1.4, and NaV1.6 in in vitro and ex vivo assays. Inhibitors lacking sufficient NaV1.7 selectivity over NaV1.6 were associated with respiratory cessation after in vivo administration to rodents. Effects on respiratory rate in rats were consistent with effects in an ex vivo hemisected rat diaphragm model and in vitro NaV1.6 potency. Furthermore, direct blockade of the phrenic nerve signaling was observed at exposures known to cause respiratory cessation in rats. Collectively, these results support a significant role for NaV1.6 in phrenic nerve signaling and respiratory function.


Assuntos
Canal de Sódio Disparado por Voltagem NAV1.7 , Insuficiência Respiratória , Animais , Dor , Nervo Frênico , Ratos , Insuficiência Respiratória/tratamento farmacológico
3.
ACS Chem Neurosci ; 3(2): 129-40, 2012 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-22860182

RESUMO

Reduced dopamine neurotransmission in the prefrontal cortex has been implicated as causal for the negative symptoms and cognitive deficit associated with schizophrenia; thus, a compound which selectively enhances dopamine neurotransmission in the prefrontal cortex may have therapeutic potential. Inhibition of catechol-O-methyltransferase (COMT, EC 2.1.1.6) offers a unique advantage, since this enzyme is the primary mechanism for the elimination of dopamine in cortical areas. Since membrane bound COMT (MB-COMT) is the predominant isoform in human brain, a high throughput screen (HTS) to identify novel MB-COMT specific inhibitors was completed. Subsequent optimization led to the identification of novel, non-nitrocatechol COMT inhibitors, some of which interact specifically with MB-COMT. Compounds were characterized for in vitro efficacy versus human and rat MB and soluble (S)-COMT. Select compounds were administered to male Wistar rats, and ex vivo COMT activity, compound levels in plasma and cerebrospinal fluid (CSF), and CSF dopamine metabolite levels were determined as measures of preclinical efficacy. Finally, novel non-nitrocatechol COMT inhibitors displayed less potent uncoupling of the mitochondrial membrane potential (MMP) compared to tolcapone as well as nonhepatotoxic entacapone, thus mitigating the risk of hepatotoxicity.


Assuntos
Antipsicóticos/farmacocinética , Inibidores de Catecol O-Metiltransferase , Catecol O-Metiltransferase/metabolismo , Inibidores Enzimáticos/farmacologia , Animais , Antipsicóticos/síntese química , Benzofenonas/química , Benzofenonas/farmacologia , Biomarcadores , Western Blotting , Catecol O-Metiltransferase/isolamento & purificação , Membrana Celular/enzimologia , Membrana Celular/metabolismo , Dopamina/metabolismo , Inibidores Enzimáticos/química , Humanos , Isoenzimas/antagonistas & inibidores , Isoenzimas/isolamento & purificação , Isoenzimas/metabolismo , Masculino , Metaloproteinases da Matriz/metabolismo , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Nitrofenóis/química , Nitrofenóis/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Proteínas Recombinantes/química , Esquizofrenia/tratamento farmacológico , Especificidade por Substrato , Tolcapona
4.
Assay Drug Dev Technol ; 6(3): 407-19, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18537465

RESUMO

Phospholipidosis (PLD) is an accumulation of phospholipids in lysosome-derived multilamellar vesicles. More than 50 commercial drugs are known to cause PLD. In vitro screening assays were developed in HepG2 cells, rat primary hepatocytes, and rhesus monkey hepatocytes using the fluorescent-labeled phospholipid probe N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)-1,2-dihexadecanoyl-sn-glycero-3-phosphoethanolamine (NBD-PE) or Nile Red lipid stain. The assays were qualified using amiodarone and fluoxetine as positive controls and precocene and valproic acid as negative controls. NBD-PE accumulation and Nile Red staining were first measured using fluorescence microscopy with morphometric analysis, and the throughput of the NBD-PE assay in HepG2 cells was increased by measuring fluorescence with a multiwell spectrofluorometer. The PLD potential values obtained for the tested compounds from the morphometric analysis were similar to the values obtained from the spectrofluorometer, suggesting the plate reader assay was effective at measuring the induction of NBD-PE accumulation. Fifteen commercial compounds were evaluated using the NBD-PE assay in HepG2 cells, rat primary hepatocytes, and rhesus monkey hepatocytes. The relative NBD-PE accumulation and PLD potentials of the evaluated compounds were similar and comparable to the values observed from other in vitro PLD assays referenced in the literature using different probes and cell lines. NBD-PE accumulations observed in rat hepatocytes after drug treatments were similar to those in HepG2 cells. NBD-PE accumulation potential observed in rhesus monkey hepatocytes after drug treatment was different for tamoxifen, perhexiline, clomipramine, and haloperidol. These agents caused potent NBD-PE accumulation in HepG2 cells, but minimal or no activity was observed in rhesus monkey hepatocytes. These data suggest that the NBD-PE spectrofluorometer assay in HepG2 cells has the speed and throughput to sensitively and quantitatively determine the PLD potential of various drug candidates. In addition, these data demonstrate the species differences in PLD potential between rat and monkey hepatocytes.


Assuntos
Hepatócitos/metabolismo , Lipidoses/induzido quimicamente , Fosfatidiletanolaminas/metabolismo , Fosfolipídeos/metabolismo , Amiodarona/farmacologia , Animais , Células Cultivadas , Humanos , Macaca mulatta , Masculino , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
5.
Life Sci ; 71(17): 2015-23, 2002 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-12175895

RESUMO

Fumonisin B(1) (FB(1)), produced by Fusarium verticillioides, is a common contaminant in foods and feeds. Increase in tissue free sphingoid bases resulting from the inhibition of ceramide synthase is a biomarker of fumonisin exposure. Tumor necrosis factor alpha (TNFalpha) is induced in liver in response to FB(1) treatment. This study determined whether fumonisin B(1) caused increases in free sphingoid bases and altered the expression of TNFalpha in heart and lung, organs that are not targets of FB(1) toxicity, of male and female mice treated with 5-daily subcutaneous injection of 2.25 mg/kg FB(1). A significant increase in free sphingoid bases was observed in both heart and lung of FB(1)-exposed mice. The magnitude of increases in free sphingoid bases in both organs of female mice was much higher than that in males. The expression of TNFalpha was increased by FB(1) treatment in the lung of male mice and in the heart of female mice, whereas the expression of interferon gamma was unaltered. Results suggest that both sphingolipid accumulation and TNFalpha induction are observed in the tissues of mice that are not associated with FB(1) toxicity.


Assuntos
Ácidos Carboxílicos/toxicidade , Carcinógenos Ambientais/toxicidade , Fumonisinas , Coração/efeitos dos fármacos , Pulmão/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Ácidos Carboxílicos/administração & dosagem , Carcinógenos Ambientais/administração & dosagem , Feminino , Injeções Subcutâneas , Interferon gama/metabolismo , Pulmão/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Miocárdio/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores Sexuais , Fator de Necrose Tumoral alfa/genética
6.
Toxicology ; 172(2): 81-92, 2002 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-11882348

RESUMO

Fumonisin B(1) (FB(1)), a carcinogenic mycotoxin produced primarily by fungus Fusarium verticillioides in corn, causes several fatal animal diseases. In mice, liver is the primary site of its toxicity. Our previous study showed that maximum induction of interferon gamma (IFNgamma) and tumor necrosis factor alpha (TNFalpha) was observed at 4 and 8 h, respectively, after an acute po FB(1) treatment. To further investigate the time-related induction of other cytokines and genes involved in apoptosis signaling, male BALB/c mice were administered orally with either saline or 25 mg/kg of FB(1) and sampled 4 or 8 h after treatment. Expression of various genes was analyzed by ribonuclease protection assay. FB(1) treatment caused increased expression of TNFalpha and interleukin (IL)-1beta in both liver and kidney, whereas IL-1alpha and IL-1 receptor antagonist (IL-1Ra) expression was induced only in the liver. Expression of TNFalpha signaling molecules, TNF receptor 55 and receptor interacting protein, was increased in liver and kidney after FB(1) treatment. Caspase 8 expression was increased only in liver with no changes in kidney with FB(1). FB(1) treatment induced expression of Fas in liver and kidney with no alterations in Fas signaling molecules, Fas ligand, Fas-associated death domain and Fas-associated protein factor. Treatment of mice with FB(1) increased the expression of B-Myc, c-Myc and Max, oncogenic transcription factors in the kidney. FB(1) toxicity caused induction of cytokine network in liver with involvement of TNFalpha signaling pathway. Increased expression of caspase 8 involved in the TNFalpha signaling pathway may contribute to the apoptosis, whereas IL-1Ra induction could contribute to the proliferating effects observed in FB(1) toxicity.


Assuntos
Apoptose/genética , Ácidos Carboxílicos/toxicidade , Carcinógenos Ambientais/toxicidade , Citocinas/biossíntese , Fumonisinas , Rim/metabolismo , Fígado/metabolismo , Transdução de Sinais/genética , Animais , Apoptose/efeitos dos fármacos , Comportamento Animal/efeitos dos fármacos , Genes myc/genética , Rim/efeitos dos fármacos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Ensaios de Proteção de Nucleases , Oncogenes/genética , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Transdução de Sinais/efeitos dos fármacos
7.
Comp Biochem Physiol C Toxicol Pharmacol ; 131(2): 113-22, 2002 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-11879779

RESUMO

Fumonisin B(1) (FB(1)), a toxic metabolite of Fusarium verticillioides, is a carcinogen and causative agent of various animal diseases. Our previous studies indicated the involvement of tumor necrosis factor-alpha (TNF alpha) in FB(1)-induced toxic responses. To further investigate the time-course of TNF alpha production and signaling, mice (four/group) were treated subcutaneously (s.c.) or per os (p.o.) with either vehicle or 25 mg/kg of FB(1) as a single dose and sacrificed at 0, 2, 4, 8, 12 and 24 h after treatment. The TNF alpha expression was increased in liver and kidney after both routes of FB(1) exposure without any alterations in spleen. The p.o.-route FB(1) treatment caused greater hepatotoxicity compared to the s.c. route, as depicted by increased alanine aminotransferase and aspartate aminotransferase level in plasma, observed only after p.o. FB(1) treatment. The increase in enzymes at 8 h after p.o. treatment correlated with the highest TNF alpha expression, also noted at 8 h after p.o. treatment, thus further confirming the involvement of TNF alpha in FB(1) toxicity. The interferon (IFN)-gamma expression was increased in liver at 4 h after p.o. FB(1) treatment, suggesting a possible combined role of TNF alpha and IFN gamma in their induction and hepatotoxicity.


Assuntos
Ácidos Carboxílicos/farmacologia , Fumonisinas , Interferon gama/metabolismo , Micotoxinas/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Administração Oral , Alanina Transaminase/sangue , Animais , Aspartato Aminotransferases/sangue , Ácidos Carboxílicos/administração & dosagem , Contagem de Eritrócitos , Injeções Subcutâneas , Interferon gama/efeitos dos fármacos , Rim/efeitos dos fármacos , Contagem de Leucócitos , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Neutrófilos/efeitos dos fármacos , Neutrófilos/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fator de Necrose Tumoral alfa/efeitos dos fármacos
8.
Chem Biol Interact ; 139(3): 317-31, 2002 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-11879819

RESUMO

Fumonisin B1 (FB1) is a naturally occurring mycotoxin produced primarily by Fusarium verticillioides and related fungi, common contaminants of corn throughout the world. FB1 is a carcinogen and causative agent of several lethal animal diseases, including equine leukoencephalomalacia and porcine pulmonary edema. Liver is the primary target organ in mice. In vivo and vitro, cells exposed to FB1 undergo a mixture of necrotic and apoptotic cell death. Our previous studies showed gender differences in hepatotoxicity caused after 5 day FB1 treatment. Gene alterations in cytokine network and apoptosis signaling molecules were also observed after an acute single dose of FB1. To further investigate the gene alterations after a subchronic FB1 exposure and its correlation to observed gender differences, male and female BALB/c mice (five per group) were injected subcutaneously with either saline or 2.25 mg/kg per day of FB1 for 5 days. FB1 caused increased expression of tumor necrosis factor alpha (TNFalpha), interleukin (IL)-1alpha, IL-1beta, IL-1 receptor antagonist (IL-1Ra), IL-6, IL-10, IL-12 p40, IL-18 and interferon gamma (IFNgamma) in male liver, with a similar increase in females except for IL-1beta and IL-18. Control females showed higher basal levels of IL-1alpha, IL-1Ra, IL-10, IL-12 p40 and IFNgamma compared with males. Expression of TNF receptor 55 and TNF receptor associated death domain (TRADD) was increased, with no changes in Fas signaling molecules, Fas, Fas ligand (FasL), Fas associated death domain (FADD) and Fas-associated protein factor (FAF). Expression of oncogenic transcription factors, c-Myc, B-Myc, Max and Mad, and apoptotic genes, namely Bcl-2, Bax and Bad, was increased after FB1 treatment. FB1 caused an activation of cytokine network in liver, particularly the TNFalpha signaling pathway, suggesting its involvement in hepatotoxic mechanisms. Induction of IL-1Ra and oncogenes is a likely mechanism for the cancer promoting properties of FB1, through a mechanism involving apoptotic necrosis, oncotic necrosis and consequent regeneration.


Assuntos
Ácidos Carboxílicos/toxicidade , Fumonisinas , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Micotoxinas/toxicidade , Transdução de Sinais/genética , Animais , Citocinas/genética , Feminino , Fusarium , Fígado/patologia , Fígado/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA/genética , RNA/isolamento & purificação , Transdução de Sinais/efeitos dos fármacos , Zea mays/microbiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA