Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Toxicol Appl Pharmacol ; 390: 114883, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31981640

RESUMO

Human-based in silico models are emerging as important tools to study the effects of integrating inward and outward ion channel currents to predict clinical proarrhythmic risk. The aims of this study were 2-fold: 1) Evaluate the capacity of an in silico model to predict QTc interval prolongation in the in vivo anesthetized cardiovascular guinea pig (CVGP) assay for new chemical entities (NCEs) and; 2) Determine if a translational pharmacokinetic/pharmacodynamic (tPKPD) model can improve the predictive capacity. In silico simulations for NCEs were performed using a population of human ventricular action potential (AP) models. PatchXpress® (PX) or high throughput screening (HTS) ion channel data from respectively n = 73 and n = 51 NCEs were used as inputs for the in silico population. These NCEs were also tested in the CVGP (n = 73). An M5 pruned decision tree-based regression tPKPD model was used to evaluate the concentration at which an NCE is liable to prolong the QTc interval in the CVGP. In silico results successfully predicted the QTc interval prolongation outcome observed in the CVGP with an accuracy/specificity of 85%/73% and 75%/77%, when using PX and HTS ion channel data, respectively. Considering the tPKPD predicted concentration resulting in QTc prolongation (EC5%) increased accuracy/specificity to 97%/95% using PX and 88%/97% when using HTS. Our results support that human-based in silico simulations in combination with tPKPD modeling can provide correlative results with a commonly used early in vivo safety assay, suggesting a path toward more rapid NCE assessment with reduced resources, cycle time, and animal use.


Assuntos
Antiarrítmicos/farmacologia , Arritmias Cardíacas , Simulação por Computador , Técnicas Eletrofisiológicas Cardíacas , Modelos Biológicos , Animais , Cálcio/metabolismo , Canais de Cálcio/metabolismo , Linhagem Celular , Fenômenos Eletrofisiológicos/efeitos dos fármacos , Cobaias , Células HEK293 , Humanos , Potenciais da Membrana/efeitos dos fármacos , Modelos Químicos
2.
Cell Metab ; 30(4): 735-753.e4, 2019 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-31577934

RESUMO

Dietary sugars, fructose and glucose, promote hepatic de novo lipogenesis and modify the effects of a high-fat diet (HFD) on the development of insulin resistance. Here, we show that fructose and glucose supplementation of an HFD exert divergent effects on hepatic mitochondrial function and fatty acid oxidation. This is mediated via three different nodes of regulation, including differential effects on malonyl-CoA levels, effects on mitochondrial size/protein abundance, and acetylation of mitochondrial proteins. HFD- and HFD plus fructose-fed mice have decreased CTP1a activity, the rate-limiting enzyme of fatty acid oxidation, whereas knockdown of fructose metabolism increases CPT1a and its acylcarnitine products. Furthermore, fructose-supplemented HFD leads to increased acetylation of ACADL and CPT1a, which is associated with decreased fat metabolism. In summary, dietary fructose, but not glucose, supplementation of HFD impairs mitochondrial size, function, and protein acetylation, resulting in decreased fatty acid oxidation and development of metabolic dysregulation.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Açúcares da Dieta/efeitos adversos , Ácidos Graxos/metabolismo , Frutose/efeitos adversos , Fígado/metabolismo , Proteínas Mitocondriais , Obesidade/metabolismo , Animais , Linhagem Celular , Glucose/efeitos adversos , Lipogênese , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Processamento de Proteína Pós-Traducional , Transcrição Gênica
3.
Endocr Relat Cancer ; 26(3): 339-353, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30640711

RESUMO

Resistance to endocrine therapy remains a clinical challenge in the treatment of estrogen receptor-positive (ER+) breast cancer. We investigated if adding a traditional Asian herbal mixture consisting of 12 herbs, called Jaeumkanghwa-tang (JEKHT), to tamoxifen (TAM) therapy might prevent resistance and recurrence in the ER+ breast cancer model of 7,12-dimethylbenz[a]anthracene (DMBA)-exposed Sprague-Dawley rats. Rats were divided into four groups treated as follows: 15 mg/kg TAM administered via diet as TAM citrate (TAM only); 500 mg/kg JEKHT administered via drinking water (JEKHT only group); TAM + JEKHT and no treatment control group. The study was replicated using two different batches of JEKHT. In both studies, a significantly higher proportion of ER+ mammary tumors responded to TAM if animals also were treated with JEKHT (experiment 1: 47% vs 65%, P = 0.015; experiment 2: 43% vs 77%, P < 0.001). The risk of local recurrence also was reduced (31% vs 12%, P = 0.002). JEKHT alone was mostly ineffective. In addition, JEKHT prevented the development of premalignant endometrial lesions in TAM-treated rats (20% in TAM only vs 0% in TAM + JEKHT). Co-treatment of antiestrogen-resistant LCC9 human breast cancer cells with 1.6 mg/mL JEKHT reversed their TAM resistance in dose-response studies in vitro. Several traditional herbal medicine preparations can exhibit anti-inflammatory properties and may increase anti-tumor immune activities in the tumor microenvironment. In the tumors of rats treated with both JEKHT and TAM, expression of Il-6 (P = 0.03), Foxp3/T regulatory cell (Treg) marker (P = 0.033) and Tgfß1 that activates Tregs (P < 0.001) were significantly downregulated compared with TAM only group. These findings indicate that JEKHT may prevent TAM-induced evasion of tumor immune responses.


Assuntos
Antineoplásicos Hormonais/administração & dosagem , Neoplasias da Mama/tratamento farmacológico , Antagonistas de Estrogênios/administração & dosagem , Extratos Vegetais/farmacologia , Receptores de Estrogênio/metabolismo , Tamoxifeno/administração & dosagem , 9,10-Dimetil-1,2-benzantraceno/toxicidade , Animais , Neoplasias da Mama/induzido quimicamente , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocinas/sangue , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Endométrio/efeitos dos fármacos , Endométrio/patologia , Feminino , Fatores de Transcrição Forkhead/genética , Humanos , Neoplasias Mamárias Experimentais , Medicina Tradicional do Leste Asiático , Recidiva Local de Neoplasia/prevenção & controle , Extratos Vegetais/administração & dosagem , Ratos , Ratos Sprague-Dawley , Fator de Crescimento Transformador beta1/genética , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
4.
J Clin Invest ; 127(11): 4059-4074, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28972537

RESUMO

Overconsumption of high-fat diet (HFD) and sugar-sweetened beverages are risk factors for developing obesity, insulin resistance, and fatty liver disease. Here we have dissected mechanisms underlying this association using mice fed either chow or HFD with or without fructose- or glucose-supplemented water. In chow-fed mice, there was no major physiological difference between fructose and glucose supplementation. On the other hand, mice on HFD supplemented with fructose developed more pronounced obesity, glucose intolerance, and hepatomegaly as compared to glucose-supplemented HFD mice, despite similar caloric intake. Fructose and glucose supplementation also had distinct effects on expression of the lipogenic transcription factors ChREBP and SREBP1c. While both sugars increased ChREBP-ß, fructose supplementation uniquely increased SREBP1c and downstream fatty acid synthesis genes, resulting in reduced liver insulin signaling. In contrast, glucose enhanced total ChREBP expression and triglyceride synthesis but was associated with improved hepatic insulin signaling. Metabolomic and RNA sequence analysis confirmed dichotomous effects of fructose and glucose supplementation on liver metabolism in spite of inducing similar hepatic lipid accumulation. Ketohexokinase, the first enzyme of fructose metabolism, was increased in fructose-fed mice and in obese humans with steatohepatitis. Knockdown of ketohexokinase in liver improved hepatic steatosis and glucose tolerance in fructose-supplemented mice. Thus, fructose is a component of dietary sugar that is distinctively associated with poor metabolic outcomes, whereas increased glucose intake may be protective.


Assuntos
Frutose/farmacologia , Glucose/farmacologia , Insulina/fisiologia , Lipogênese/efeitos dos fármacos , Fígado/efeitos dos fármacos , Adolescente , Animais , Dieta Hiperlipídica/efeitos adversos , Indução Enzimática , Ácidos Graxos/biossíntese , Frutoquinases/genética , Frutoquinases/metabolismo , Intolerância à Glucose , Humanos , Resistência à Insulina , Fígado/enzimologia , Masculino , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica/enzimologia , Hepatopatia Gordurosa não Alcoólica/etiologia , Obesidade/enzimologia , Transdução de Sinais/efeitos dos fármacos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Ativação Transcricional , Transcriptoma , Regulação para Cima
5.
J Pediatr Surg ; 52(5): 795-801, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28189450

RESUMO

BACKGROUND: Pediatric intestinal failure (PIF) is a life-altering chronic condition with significant morbidity and mortality. Omegaven® therapy has been used to treat children with advanced intestinal failure associated liver disease. Our objective was to determine the evolution of hepatic fibrosis in PIF patients who received Omegaven® and describe their clinical outcome. METHODS: A retrospective review in PIF patients who received Omegaven® was performed. Patients were included if they had liver biopsies completed before Omegaven® therapy and after resolution of hyperbilirubinemia. Biopsy results were evaluated to determine the degree of fibrosis, inflammation, and cholestasis. Clinical and biochemical data was collected. RESULTS: Six patients were identified. Assessment of fibrosis at last follow-up demonstrated improvement in 2 patients and progression or stable fibrosis in 4/6. All patients demonstrated reduction in cholestasis and inflammation. One patient received a liver/intestine transplant and a second is listed, both of them with progressive fibrosis. One patient achieved full enteral nutrition, while the rest remain partially parenteral nutrition dependent. CONCLUSION: Use of Omegaven® is associated with reduced cholestasis and inflammation, but with persistence or worsening of fibrosis in some patients. A subset of patients with progressive fibrosis may develop portal hypertension and progressive liver disease.


Assuntos
Emulsões Gordurosas Intravenosas/uso terapêutico , Óleos de Peixe/uso terapêutico , Enteropatias/complicações , Cirrose Hepática/terapia , Biópsia , Criança , Pré-Escolar , Progressão da Doença , Nutrição Enteral , Feminino , Seguimentos , Humanos , Hiperbilirrubinemia/etiologia , Hiperbilirrubinemia/patologia , Lactente , Recém-Nascido , Enteropatias/terapia , Cirrose Hepática/diagnóstico , Cirrose Hepática/etiologia , Cirrose Hepática/patologia , Transplante de Fígado , Masculino , Nutrição Parenteral Total , Estudos Retrospectivos , Método Simples-Cego , Resultado do Tratamento , Triglicerídeos
6.
Amyloid ; 23(2): 109-18, 2016 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-27033334

RESUMO

ATTR amyloidosis is a systemic, debilitating and fatal disease caused by transthyretin (TTR) amyloid accumulation. RNA interference (RNAi) is a clinically validated technology that may be a promising approach to the treatment of ATTR amyloidosis. The vast majority of TTR, the soluble precursor of TTR amyloid, is expressed and synthesized in the liver. RNAi technology enables robust hepatic gene silencing, the goal of which would be to reduce systemic levels of TTR and mitigate many of the clinical manifestations of ATTR that arise from hepatic TTR expression. To test this hypothesis, TTR-targeting siRNAs were evaluated in a murine model of hereditary ATTR amyloidosis. RNAi-mediated silencing of hepatic TTR expression inhibited TTR deposition and facilitated regression of existing TTR deposits in pathologically relevant tissues. Further, the extent of deposit regression correlated with the level of RNAi-mediated knockdown. In comparison to the TTR stabilizer, tafamidis, RNAi-mediated TTR knockdown led to greater regression of TTR deposits across a broader range of affected tissues. Together, the data presented herein support the therapeutic hypothesis behind TTR lowering and highlight the potential of RNAi in the treatment of patients afflicted with ATTR amyloidosis.


Assuntos
Neuropatias Amiloides Familiares/terapia , Fígado/metabolismo , Pré-Albumina/antagonistas & inibidores , RNA Mensageiro/antagonistas & inibidores , RNA Interferente Pequeno/administração & dosagem , Neuropatias Amiloides Familiares/genética , Neuropatias Amiloides Familiares/metabolismo , Neuropatias Amiloides Familiares/patologia , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Benzoxazóis/farmacologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Feminino , Expressão Gênica , Humanos , Fígado/patologia , Macaca fascicularis , Masculino , Camundongos , Camundongos Transgênicos , Pré-Albumina/genética , Pré-Albumina/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacocinética
7.
Am J Hematol ; 90(4): 310-3, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25557851

RESUMO

ß-thalassemias result from diminished ß-globin synthesis and are associated with ineffective erythropoiesis and secondary iron overload caused by inappropriately low levels of the iron regulatory hormone hepcidin. The serine protease TMPRSS6 attenuates hepcidin production in response to iron stores. Hepcidin induction reduces iron overload and mitigates anemia in murine models of ß-thalassemia intermedia. To further interrogate the efficacy of an RNAi-therapeutic downregulating Tmprss6, ß-thalassemic Hbb(th3/+) animals on an iron replete, an iron deficient, or an iron replete diet also containing the iron chelator deferiprone were treated with Tmprss6 siRNA. We demonstrate that the total body iron burden is markedly improved in Hbb(th3/+) animals treated with siRNA and chelated with oral deferiprone, representing a significant improvement compared to either compound alone. These data indicate that siRNA suppression of Tmprss6, in conjunction with oral iron chelation therapy, may prove superior for treatment of anemia and secondary iron loading seen in ß-thalassemia intermedia.


Assuntos
Quelantes de Ferro/uso terapêutico , Ferro/metabolismo , Proteínas de Membrana/genética , Piridonas/uso terapêutico , Interferência de RNA , Serina Endopeptidases/genética , Talassemia beta/tratamento farmacológico , Administração Oral , Animais , Terapia Combinada , Deferiprona , Modelos Animais de Doenças , Portadores de Fármacos/química , Feminino , Hepcidinas/biossíntese , Hepcidinas/sangue , Ferro/sangue , Quelantes de Ferro/administração & dosagem , Camundongos , Nanopartículas/química , Piridonas/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Talassemia beta/genética , Talassemia beta/metabolismo
8.
Proc Natl Acad Sci U S A ; 111(21): 7777-82, 2014 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-24821812

RESUMO

The acute hepatic porphyrias are inherited disorders of heme biosynthesis characterized by life-threatening acute neurovisceral attacks. Factors that induce the expression of hepatic 5-aminolevulinic acid synthase 1 (ALAS1) result in the accumulation of the neurotoxic porphyrin precursors 5-aminolevulinic acid (ALA) and porphobilinogen (PBG), which recent studies indicate are primarily responsible for the acute attacks. Current treatment of these attacks involves i.v. administration of hemin, but a faster-acting, more effective, and safer therapy is needed. Here, we describe preclinical studies of liver-directed small interfering RNAs (siRNAs) targeting Alas1 (Alas1-siRNAs) in a mouse model of acute intermittent porphyria, the most common acute hepatic porphyria. A single i.v. dose of Alas1-siRNA prevented the phenobarbital-induced biochemical acute attacks for approximately 2 wk. Injection of Alas1-siRNA during an induced acute attack significantly decreased plasma ALA and PBG levels within 8 h, more rapidly and effectively than a single hemin infusion. Alas1-siRNA was well tolerated and a therapeutic dose did not cause hepatic heme deficiency. These studies provide proof-of-concept for the clinical development of RNA interference therapy for the prevention and treatment of the acute attacks of the acute hepatic porphyrias.


Assuntos
5-Aminolevulinato Sintetase/metabolismo , Fígado/metabolismo , Porfiria Aguda Intermitente/prevenção & controle , Interferência de RNA/imunologia , RNA Interferente Pequeno/farmacologia , 5-Aminolevulinato Sintetase/genética , Análise de Variância , Animais , Western Blotting , Avaliação Pré-Clínica de Medicamentos , Eletroforese em Gel de Poliacrilamida , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Tamanho da Partícula , Interferência de RNA/efeitos dos fármacos , Reação em Cadeia da Polimerase em Tempo Real
9.
PLoS Genet ; 2(4): e57, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16683034

RESUMO

We report here on a chemical genetic screen designed to address the mechanism of action of a small molecule. Small molecules that were active in models of urinary incontinence were tested on the nematode Caenorhabditis elegans, and the resulting phenotypes were used as readouts in a genetic screen to identify possible molecular targets. The mutations giving resistance to compound were found to affect members of the RGS protein/G-protein complex. Studies in mammalian systems confirmed that the small molecules inhibit muscarinic G-protein coupled receptor (GPCR) signaling involving G-alphaq (G-protein alpha subunit). Our studies suggest that the small molecules act at the level of the RGS/G-alphaq signaling complex, and define new mutations in both RGS and G-alphaq, including a unique hypo-adapation allele of G-alphaq. These findings suggest that therapeutics targeted to downstream components of GPCR signaling may be effective for treatment of diseases involving inappropriate receptor activation.


Assuntos
Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/genética , Subunidades alfa de Proteínas de Ligação ao GTP/metabolismo , Proteínas RGS/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Triazóis/farmacologia , Animais , Caenorhabditis elegans/efeitos dos fármacos , Proteínas de Caenorhabditis elegans/metabolismo , Cálcio/metabolismo , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Feminino , Subunidades alfa de Proteínas de Ligação ao GTP/genética , Humanos , Proteínas RGS/genética , Ensaio Radioligante , Ratos , Ratos Sprague-Dawley , Receptores Acoplados a Proteínas G/genética , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Bexiga Urinária/efeitos dos fármacos , Bexiga Urinária/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA