Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Mol Ther ; 32(6): 1895-1916, 2024 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-38549376

RESUMO

Malignant tumors are often associated with an immunosuppressive tumor microenvironment (TME), rendering most of them resistant to standard-of-care immune checkpoint inhibitors (CPIs). Signal transducer and activator of transcription 3 (STAT3), a ubiquitously expressed transcription factor, has well-defined immunosuppressive functions in several leukocyte populations within the TME. Since the STAT3 protein has been challenging to target using conventional pharmaceutical modalities, we investigated the feasibility of applying systemically delivered RNA interference (RNAi) agents to silence its mRNA directly in tumor-associated immune cells. In preclinical rodent tumor models, chemically stabilized acylated small interfering RNAs (siRNAs) selectively silenced Stat3 mRNA in multiple relevant cell types, reduced STAT3 protein levels, and increased cytotoxic T cell infiltration. In a murine model of CPI-resistant pancreatic cancer, RNAi-mediated Stat3 silencing resulted in tumor growth inhibition, which was further enhanced in combination with CPIs. To further exemplify the utility of RNAi for cancer immunotherapy, this technology was used to silence Cd274, the gene encoding the immune checkpoint protein programmed death-ligand 1 (PD-L1). Interestingly, silencing of Cd274 was effective in tumor models that are resistant to PD-L1 antibody therapy. These data represent the first demonstration of systemic delivery of RNAi agents to the TME and suggest applying this technology for immuno-oncology applications.


Assuntos
Antígeno B7-H1 , Interferência de RNA , RNA Interferente Pequeno , Fator de Transcrição STAT3 , Microambiente Tumoral , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT3/genética , Animais , Camundongos , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Humanos , Microambiente Tumoral/imunologia , RNA Interferente Pequeno/genética , Imunoterapia/métodos , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores de Checkpoint Imunológico/farmacologia , Modelos Animais de Doenças , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/patologia , Neoplasias/terapia , Neoplasias/imunologia , Neoplasias/genética
2.
Cancer Prev Res (Phila) ; 8(9): 786-95, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26069204

RESUMO

Despite widespread use as well as epidemiologic indications, there have been no investigations into the effect of St. John's wort (SJW) extract on colorectal carcinogenesis in vivo. This study reports a systematic evaluation of the impact of dietary supplementation of SJW extract on azoxymethane-induced colorectal carcinogenesis in mice. Mice were fed with either AIN-93G (control) diet or SJW extract-supplemented diet (SJW diet) prior to azoxymethane treatment. SJW diet was found to significantly improve the overall survival of azoxymethane-treated mice. Pretreatment with the SJW diet significantly reduced body weight loss as well as decrease of serum albumin and cholesterol levels associated with azoxymethane-induced colorectal tumorigenesis. SJW diet-fed mice showed a significant decrease in tumor multiplicity along with a decrease in incidence of large tumors and a trend toward decreased total tumor volume in a dose-dependent manner. A short-term study, which examined the effect of SJW prior to rectal bleeding, also showed decrease in colorectal polyps in SJW diet-fed mice. Nuclear factor kappa B (NF-κB) and extracellular signal-regulated kinase (ERK1/2) pathways were attenuated by SJW administration. SJW extract resulted in early and continuous attenuation of these pathways in the colon epithelium of SJW diet-fed mice under both short-term and long-term treatment regimens. In conclusion, this study demonstrated the chemopreventive potential of SJW extract against colorectal cancer through attenuation of proinflammatory processes.


Assuntos
Anticarcinógenos/uso terapêutico , Carcinogênese/efeitos dos fármacos , Neoplasias Colorretais/prevenção & controle , Hypericum/química , Inflamação/tratamento farmacológico , Extratos Vegetais/uso terapêutico , Animais , Azoximetano/química , Transformação Celular Neoplásica/efeitos dos fármacos , Colo/patologia , Neoplasias Colorretais/tratamento farmacológico , Dieta , Suplementos Nutricionais , Modelos Animais de Doenças , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Masculino , Camundongos , NF-kappa B/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Oligonucleotídeos/química , Transdução de Sinais/efeitos dos fármacos
3.
J Med Chem ; 56(5): 2110-24, 2013 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-23445220

RESUMO

Protein arginine methyltransferases (PRMTs) play an important role in diverse biological processes. Among the nine known human PRMTs, PRMT3 has been implicated in ribosomal biosynthesis via asymmetric dimethylation of the 40S ribosomal protein S2 and in cancer via interaction with the DAL-1 tumor suppressor protein. However, few selective inhibitors of PRMTs have been discovered. We recently disclosed the first selective PRMT3 inhibitor, which occupies a novel allosteric binding site and is noncompetitive with both the peptide substrate and cofactor. Here we report comprehensive structure-activity relationship studies of this series, which resulted in the discovery of multiple PRMT3 inhibitors with submicromolar potencies. An X-ray crystal structure of compound 14u in complex with PRMT3 confirmed that this inhibitor occupied the same allosteric binding site as our initial lead compound. These studies provide the first experimental evidence that potent and selective inhibitors can be created by exploiting the allosteric binding site of PRMT3.


Assuntos
Inibidores Enzimáticos/farmacologia , Proteína-Arginina N-Metiltransferases/antagonistas & inibidores , Sítio Alostérico/efeitos dos fármacos , Inibidores Enzimáticos/síntese química , Humanos , Concentração Inibidora 50 , Proteína-Arginina N-Metiltransferases/química , Proteínas Ribossômicas/metabolismo , Relação Estrutura-Atividade , Tiadiazóis/química , Tiadiazóis/farmacologia , Difração de Raios X
4.
J Med Chem ; 55(14): 6467-77, 2012 Jul 26.
Artigo em Inglês | MEDLINE | ID: mdl-22738238

RESUMO

Adenosine A(1) receptor (A(1)AR) agonists have antinociceptive effects in multiple preclinical models of acute and chronic pain. Although numerous A(1)AR agonists have been developed, clinical applications of these agents have been hampered by their cardiovascular side effects. Herein we report a series of novel A(1)AR agonists, some of which are structurally related to adenosine 5'-monophosphate (5'-AMP), a naturally occurring nucleotide that itself activates A(1)AR. These novel compounds potently activate A(1)AR in several orthogonal in vitro assays and are subtype selective for A(1)AR over A(2A)AR, A(2B)AR, and A(3)AR. Among them, UNC32A (3a) is orally active and has dose-dependent antinociceptive effects in wild-type mice. The antinociceptive effects of 3a were completely abolished in A(1)AR knockout mice, revealing a strict dependence on A(1)AR for activity. The apparent lack of cardiovascular side effects when administered orally and high affinity (K(i) of 36 nM for the human A(1)AR) make this compound potentially suitable as a therapeutic.


Assuntos
Agonistas do Receptor A1 de Adenosina/administração & dosagem , Agonistas do Receptor A1 de Adenosina/farmacologia , Monofosfato de Adenosina/administração & dosagem , Monofosfato de Adenosina/farmacologia , Analgésicos/administração & dosagem , Analgésicos/farmacologia , Receptor A1 de Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina/química , Agonistas do Receptor A1 de Adenosina/metabolismo , Monofosfato de Adenosina/química , Monofosfato de Adenosina/metabolismo , Administração Oral , Analgésicos/química , Analgésicos/metabolismo , Animais , Comportamento Animal/efeitos dos fármacos , Células HEK293 , Humanos , Masculino , Camundongos , Nociceptividade/efeitos dos fármacos , Especificidade por Substrato , Temperatura
5.
J Biol Chem ; 287(8): 5301-9, 2012 Feb 17.
Artigo em Inglês | MEDLINE | ID: mdl-22215671

RESUMO

Numerous receptors for ATP, ADP, and adenosine exist; however, it is currently unknown whether a receptor for the related nucleotide adenosine 5'-monophosphate (AMP) exists. Using a novel cell-based assay to visualize adenosine receptor activation in real time, we found that AMP and a non-hydrolyzable AMP analog (deoxyadenosine 5'-monophosphonate, ACP) directly activated the adenosine A(1) receptor (A(1)R). In contrast, AMP only activated the adenosine A(2B) receptor (A(2B)R) after hydrolysis to adenosine by ecto-5'-nucleotidase (NT5E, CD73) or prostatic acid phosphatase (PAP, ACPP). Adenosine and AMP were equipotent human A(1)R agonists in our real-time assay and in a cAMP accumulation assay. ACP also depressed cAMP levels in mouse cortical neurons through activation of endogenous A(1)R. Non-selective purinergic receptor antagonists (pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid and suramin) did not block adenosine- or AMP-evoked activation. Moreover, mutation of His-251 in the human A(1)R ligand binding pocket reduced AMP potency without affecting adenosine potency. In contrast, mutation of a different binding pocket residue (His-278) eliminated responses to AMP and to adenosine. Taken together, our study indicates that the physiologically relevant nucleotide AMP is a full agonist of A(1)R. In addition, our study suggests that some of the physiological effects of AMP may be direct, and not indirect through ectonucleotidases that hydrolyze this nucleotide to adenosine.


Assuntos
Agonistas do Receptor A1 de Adenosina/farmacologia , Monofosfato de Adenosina/farmacologia , Receptor A1 de Adenosina/metabolismo , 5'-Nucleotidase/metabolismo , Adenosina/metabolismo , Agonistas do Receptor A1 de Adenosina/química , Agonistas do Receptor A1 de Adenosina/metabolismo , Monofosfato de Adenosina/análogos & derivados , Monofosfato de Adenosina/metabolismo , Animais , Córtex Cerebral/citologia , Colforsina/farmacologia , Células HEK293 , Histidina , Humanos , Hidrólise/efeitos dos fármacos , Ligantes , Camundongos , Imagem Molecular , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Receptor A1 de Adenosina/química , Receptor A2B de Adenosina/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Proteínas Recombinantes de Fusão/agonistas , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Análise de Célula Única
6.
Acta Biochim Biophys Sin (Shanghai) ; 44(1): 70-9, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22194015

RESUMO

During the last decade, we saw an explosion of studies investigating the role of lysine methylation/demethylation of histones and non-histone proteins, such as p53, NF-kappaB, and E2F1. These 'Ying-Yang' post-translational modifications are important to fine-tuning the activity of these proteins. Lysine methylation and demethylation are catalyzed by protein lysine methyltransferases (PKMTs) and protein lysine demethylases (PKDMs). PKMTs, PKDMs, and their substrates have been shown to play important roles in cancers. Although the underlying mechanisms of tumorigenesis are still largely unknown, growing evidence is starting to link aberrant regulation of methylation to tumorigenesis. This review focuses on summarizing the recent progress in understanding of the function of protein lysine methylation, and in the discovery of small molecule inhibitors for PKMTs and PKDMs. We also discuss the potential and the caveats of targeting protein lysine methylation for the treatment of cancer.


Assuntos
Inibidores Enzimáticos/uso terapêutico , Histona Desmetilases/metabolismo , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Histona-Lisina N-Metiltransferase/metabolismo , Neoplasias/tratamento farmacológico , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteína Potenciadora do Homólogo 2 de Zeste , Epigênese Genética , Antígenos de Histocompatibilidade , Histona Desmetilases/antagonistas & inibidores , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Metiltransferases/antagonistas & inibidores , Neoplasias/genética , Complexo Repressor Polycomb 2 , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Repressoras/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo
7.
Curr Chem Genomics ; 5(Suppl 1): 72-84, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21966347

RESUMO

Growing evidence suggests that protein lysine methyltransferases (PKMTs) and protein arginine methyltransferases (PRMTs) are associated with the development of various human diseases, including cancer, inflammation, and psychiatric disorders. Given the significant role of these proteins in human disease, efforts to discover selective small-molecule inhibitors of these enzymes are quickly gaining momentum. In this review, we focus on the recent progress in the discovery of selective PKMT and PRMT inhibitors. A future perspective on developing methyltransferase inhibitors is also offered.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA