Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Toxicol Sci ; 191(2): 400-413, 2023 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-36515490

RESUMO

Administration of a novel and selective small molecule integrin αvß6 inhibitor, MORF-627, to young cynomolgus monkeys for 28 days resulted in the rapid induction of epithelial proliferative changes in the urinary bladder of 2 animals, in the absence of test agent genotoxicity. Microscopic findings included suburothelial infiltration by irregular nests and/or trabeculae of epithelial cells, variable cytologic atypia, and high mitotic rate, without invasion into the tunica muscularis. Morphologic features and patterns of tumor growth were consistent with a diagnosis of early-stage invasive urothelial carcinoma. Ki67 immunohistochemistry demonstrated diffusely increased epithelial proliferation in the urinary bladder of several monkeys, including those with tumors, and αvß6 was expressed in some epithelial tissues, including urinary bladder, in monkeys and humans. Spontaneous urothelial carcinomas are extremely unusual in young healthy monkeys, suggesting a direct link of the finding to the test agent. Inhibition of integrin αvß6 is intended to locally and selectively block transforming growth factor beta (TGF-ß) signaling, which is implicated in epithelial proliferative disorders. Subsequent in vitro studies using a panel of integrin αvß6 inhibitors in human bladder epithelial cells replicated the increased urothelial proliferation observed in monkeys and was reversed through exogenous application of TGF-ß. Moreover, analysis of in vivo models of liver and lung fibrosis revealed evidence of epithelial hyperplasia and cell cycle dysregulation in mice treated with integrin αvß6 or TGF-ß receptor I inhibitors. The cumulative evidence suggests a direct link between integrin αvß6 inhibition and decreased TGF-ß signaling in the local bladder environment, with implications for epithelial proliferation and carcinogenesis.


Assuntos
Carcinoma de Células de Transição , Integrinas , Neoplasias da Bexiga Urinária , Animais , Humanos , Camundongos , Carcinoma de Células de Transição/induzido quimicamente , Integrinas/antagonistas & inibidores , Integrinas/metabolismo , Macaca fascicularis , Fator de Crescimento Transformador beta/metabolismo , Neoplasias da Bexiga Urinária/induzido quimicamente
2.
Nat Commun ; 11(1): 1920, 2020 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-32317643

RESUMO

Collagen-producing cells maintain the complex architecture of the lung and drive pathologic scarring in pulmonary fibrosis. Here we perform single-cell RNA-sequencing to identify all collagen-producing cells in normal and fibrotic lungs. We characterize multiple collagen-producing subpopulations with distinct anatomical localizations in different compartments of murine lungs. One subpopulation, characterized by expression of Cthrc1 (collagen triple helix repeat containing 1), emerges in fibrotic lungs and expresses the highest levels of collagens. Single-cell RNA-sequencing of human lungs, including those from idiopathic pulmonary fibrosis and scleroderma patients, demonstrate similar heterogeneity and CTHRC1-expressing fibroblasts present uniquely in fibrotic lungs. Immunostaining and in situ hybridization show that these cells are concentrated within fibroblastic foci. We purify collagen-producing subpopulations and find disease-relevant phenotypes of Cthrc1-expressing fibroblasts in in vitro and adoptive transfer experiments. Our atlas of collagen-producing cells provides a roadmap for studying the roles of these unique populations in homeostasis and pathologic fibrosis.


Assuntos
Colágeno/química , Pulmão/metabolismo , Fibrose Pulmonar/metabolismo , Animais , Separação Celular , Proteínas da Matriz Extracelular/metabolismo , Feminino , Fibroblastos/metabolismo , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Fibrose Pulmonar Idiopática/patologia , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Fibrose Pulmonar/patologia , Transtornos Respiratórios/metabolismo , Análise de Célula Única
3.
Mol Pharmacol ; 94(4): 1197-1209, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30068735

RESUMO

The D1 dopamine receptor is linked to a variety of neuropsychiatric disorders and represents an attractive drug target for the enhancement of cognition in schizophrenia, Alzheimer disease, and other disorders. Positive allosteric modulators (PAMs), with their potential for greater selectivity and larger therapeutic windows, may represent a viable drug development strategy, as orthosteric D1 receptor agonists possess known clinical liabilities. We discovered two structurally distinct D1 receptor PAMs, MLS6585 and MLS1082, via a high-throughput screen of the NIH Molecular Libraries program small-molecule library. Both compounds potentiate dopamine-stimulated G protein- and ß-arrestin-mediated signaling and increase the affinity of dopamine for the D1 receptor with low micromolar potencies. Neither compound displayed any intrinsic agonist activity. Both compounds were also found to potentiate the efficacy of partial agonists. We tested maximally effective concentrations of each PAM in combination to determine if the compounds might act at separate or similar sites. In combination, MLS1082 + MLS6585 produced an additive potentiation of dopamine potency beyond that caused by either PAM alone for both ß-arrestin recruitment and cAMP accumulation, suggesting diverse sites of action. In addition, MLS6585, but not MLS1082, had additive activity with the previously described D1 receptor PAM "Compound B," suggesting that MLS1082 and Compound B may share a common binding site. A point mutation (R130Q) in the D1 receptor was found to abrogate MLS1082 activity without affecting that of MLS6585, suggesting this residue may be involved in the binding/activity of MLS1082 but not that of MLS6585. Together, MLS1082 and MLS6585 may serve as important tool compounds for the characterization of diverse allosteric sites on the D1 receptor as well as the development of optimized lead compounds for therapeutic use.


Assuntos
Regulação Alostérica/fisiologia , Sítio Alostérico/fisiologia , Receptores Dopaminérgicos/metabolismo , Animais , Células CHO , Cricetulus , AMP Cíclico/metabolismo , Dopamina/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Transdução de Sinais/fisiologia , beta-Arrestinas/metabolismo
4.
J Neurosci ; 31(1): 126-32, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21209197

RESUMO

Bacteria artificial chromosome (BAC) transgenic mice expressing the reporter protein enhanced green fluorescent protein (EGFP) under the control of the D1 and D2 dopamine receptor promoters (Drd1-EGFP and Drd2-EGFP) have been widely used to study striatal function and have contributed to our understanding of the physiological and pathological functions of the basal ganglia. These tools were produced and promptly made available to address questions in a cell-specific manner that has transformed the way we frame hypotheses in neuroscience. However, these mice have not been fully characterized until now. We found that Drd2-EGFP mice display an ∼40% increase in membrane expression of the dopamine D2 receptor (D2R) and a twofold increase in D2R mRNA levels in the striatum when compared with wild-type and Drd1-EGFP mice. D2R overexpression was accompanied by behavioral hypersensitivity to D2R-like agonists, as well as enhanced electrophysiological responses to D2R activation in midbrain dopaminergic neurons. Dopamine (DA) transients evoked by stimulation in the nucleus accumbens showed slower clearance in Drd2-EGFP mice, and cocaine actions on DA clearance were impaired in these mice. Thus, it was not surprising to find that Drd2-EGFP mice were hyperactive when exposed to a novel environment and locomotion was suppressed by acute cocaine administration. All together, this study demonstrates that Drd2-EGFP mice overexpress D2R and have altered dopaminergic signaling that fundamentally differentiates them from wild-type and Drd1-EGFP mice.


Assuntos
Regulação da Expressão Gênica/genética , Receptores de Dopamina D2/genética , Receptores de Dopamina D2/metabolismo , Animais , Repetição de Anquirina/genética , Comportamento Animal/fisiologia , Cocaína/farmacologia , Corpo Estriado/efeitos dos fármacos , Corpo Estriado/metabolismo , Agonistas de Dopamina/farmacologia , Inibidores da Captação de Dopamina/farmacologia , Relação Dose-Resposta a Droga , Eletroquímica/métodos , Proteínas de Fluorescência Verde/genética , Técnicas In Vitro , Locomoção/efeitos dos fármacos , Camundongos , Camundongos Transgênicos , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Ligação Proteica/efeitos dos fármacos , Quimpirol/farmacologia , RNA Mensageiro/metabolismo , Ensaio Radioligante/métodos , Receptores de Dopamina D1/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Estatísticas não Paramétricas , Fatores de Tempo , Área Tegmentar Ventral/citologia
5.
Curr Protoc Neurosci ; Chapter 5: Unit 5.28, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19170023

RESUMO

Proteomics has evolved from genomic science due to the convergence of advances in protein chemistry, separations, mass spectroscopy, and peptide and protein databases. Where identifying protein-protein interactions was once limited to yeast two-hybrid analyses or empirical data, protein-protein interactions can now be examined in both cells and native tissues by precipitation of the protein complex of interest. Coupling this field to receptor pharmacology has recently allowed for the identification of proteins that differentially and selectively interact with receptors and are integral to their biological effects. It is becoming increasingly apparent that receptors in neurons do not exist as singular independent units, but rather are part of large macromolecular complexes of interacting proteins. It is a primary quest of neuroscience to piece together these interactions and to characterize the regulatory signalplexes of all proteins. This unit presents co-immunoprecipitation-coupled mass spectroscopy as one way of identifying signalplex partners.


Assuntos
Imunoprecipitação/métodos , Espectrometria de Massas/métodos , Complexos Multiproteicos/metabolismo , Mapeamento de Interação de Proteínas/métodos , Proteômica/métodos , Animais , Células Cultivadas , Complexos Multiproteicos/química , Fenômenos Fisiológicos do Sistema Nervoso , Neuroquímica/métodos , Neurônios/química , Neurônios/metabolismo , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Transdução de Sinais/fisiologia
6.
J Biol Chem ; 283(52): 36441-53, 2008 Dec 26.
Artigo em Inglês | MEDLINE | ID: mdl-18984584

RESUMO

It is well documented that dopamine can increase or decrease the activity of the Na+,K+-ATPase (NKA, sodium pump) in an organ-specific fashion. This regulation can occur, at least partially, via receptor-mediated second messenger activation and can promote NKA insertion or removal from the plasma membrane. Using co-immunoprecipitation and mass spectrometry, we now show that, in both brain and HEK293T cells, D1 and D2 dopamine receptors (DARs) can exist in a complex with the sodium pump. To determine the impact of NKA on DAR function, biological assays were conducted with NKA and DARs co-expressed in HEK293T cells. In this system, expression of NKA dramatically decreased D1 and D2 DAR densities with a concomitant functional decrease in DAR-mediated regulation of cAMP levels. Interestingly, pharmacological inhibition of endogenous or overexpressed NKA enhanced DAR function without altering receptor number or localization. Similarly, DAR function was also augmented by small interfering RNA reduction of the endogenous NKA. These data suggest that, under basal conditions, NKA negatively regulates DAR function via protein-protein interactions. In reciprocal fashion, expression of DARs decreases endogenous NKA function in the absence of dopamine, implicating DAR proteins as regulators of NKA activity. Notably, dopamine stimulation or pertussis toxin inhibition of D2 receptor signaling did not alter NKA activity, indicating that the D2-mediated decrease in NKA function is dependent upon protein-protein interactions rather than signaling molecules. This evidence for reciprocal regulation between DARs and NKA provides a novel control mechanism for both DAR signaling and cellular ion balance.


Assuntos
Receptores de Dopamina D1/metabolismo , Receptores de Dopamina D2/metabolismo , ATPase Trocadora de Sódio-Potássio/metabolismo , Sequência de Aminoácidos , Encéfalo/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Dopamina/metabolismo , Humanos , Íons , Espectrometria de Massas/métodos , Modelos Biológicos , Dados de Sequência Molecular , Toxina Pertussis/farmacologia , Transdução de Sinais
7.
J Biol Chem ; 282(29): 21285-300, 2007 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-17395585

RESUMO

As for all proteins, G protein-coupled receptors (GPCRs) undergo synthesis and maturation within the endoplasmic reticulum (ER). The mechanisms involved in the biogenesis and trafficking of GPCRs from the ER to the cell surface are poorly understood, but they may involve interactions with other proteins. We have now identified the ER chaperone protein calnexin as an interacting protein for both D(1) and D(2) dopamine receptors. These protein-protein interactions were confirmed using Western blot analysis and co-immunoprecipitation experiments. To determine the influence of calnexin on receptor expression, we conducted assays in HEK293T cells using a variety of calnexin-modifying conditions. Inhibition of glycosylation either through receptor mutations or treatments with glycosylation inhibitors partially blocks the interactions with calnexin with a resulting decrease in cell surface receptor expression. Confocal fluorescence microscopy reveals the accumulation of D(1)-green fluorescent protein and D(2)-yellow fluorescent protein receptors within internal stores following treatment with calnexin inhibitors. Overexpression of calnexin also results in a marked decrease in both D(1) and D(2) receptor expression. This is likely because of an increase in ER retention because confocal microscopy revealed intracellular clustering of dopamine receptors that were co-localized with an ER marker protein. Additionally, we show that calnexin interacts with the receptors via two distinct mechanisms, glycan-dependent and glycan-independent, which may underlie the multiple effects (ER retention and surface trafficking) of calnexin on receptor expression. Our data suggest that optimal receptor-calnexin interactions critically regulate D(1) and D(2) receptor trafficking and expression at the cell surface, a mechanism likely to be of importance for many GPCRs.


Assuntos
Calnexina/química , Regulação da Expressão Gênica , Receptores de Dopamina D1/química , Receptores de Dopamina D2/química , Calnexina/metabolismo , Linhagem Celular , AMP Cíclico/metabolismo , Retículo Endoplasmático/metabolismo , Humanos , Imunoprecipitação , Cinética , Espectrometria de Massas , Microscopia Confocal , Peptídeos/química , Polissacarídeos/metabolismo , Ligação Proteica
8.
Mol Pharmacol ; 66(5): 1293-300, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15496511

RESUMO

Naturally occurring variation within the human 5-HT(2A) receptor results in an amino acid substitution in the carboxyl terminus of the receptor. This single nucleotide polymorphism (SNP), encoding a His452Tyr substitution, occurs at a frequency of 9% in the general population. It is noteworthy that this SNP has been linked to attention deficit hyperactivity disorder and has been associated with schizophrenic patients that do not respond to treatment with clozapine. To evaluate functional consequences of this SNP, agonist-stimulated signaling was investigated in NIH3T3 cells stably expressing either wild-type or 452Tyr variant receptors. The 452Tyr variant of the 5-HT(2a) receptor had reduced ability to activate phospholipases C and D, suggesting that signaling through both G(q) and G(13) pathways is hindered. This conclusion was supported by assays of G protein coupling, which documented a loss of agonist-induced high affinity binding and a decreased turnover of guanosine 5'-O-(3-[(35)S]thio)triphosphate after agonist stimulation. Kinetic analysis of time-course data revealed an altered desensitization phenotype, resulting in a blunted signal downstream of receptor activation. This diminished signaling implies that the His452Tyr variant receptor alters physiological responses, possibly contributing to psychiatric disease.


Assuntos
Histidina/química , Receptor 5-HT2A de Serotonina/química , Tirosina/química , Substituição de Aminoácidos , Células Cultivadas , Proteínas de Ligação ao GTP/metabolismo , Histidina/genética , Humanos , Polimorfismo Genético , Conformação Proteica , Receptor 5-HT2A de Serotonina/genética , Receptor 5-HT2A de Serotonina/metabolismo , Agonistas do Receptor de Serotonina/farmacologia , Transdução de Sinais , Tirosina/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...