Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Immunol ; 41(5): 1016-1030, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33651234

RESUMO

Acute graft-versus-host disease (aGVHD) is a significant complication of allogeneic hematopoietic stem cell transplant (HSCT) and negatively affects T cell reconstitution. Extracorporeal photopheresis (ECP) reduces aGVHD, but the mechanisms remain incompletely understood. Our objective was to examine the impact of ECP on thymopoiesis in pediatric aGVHD and the mechanisms at a cellular and transcriptional level. Sixteen pediatric HSCT patients were recruited: 6 with ECP-treated aGVHD, 5 without aGVHD, and 5 with aGVHD treated with corticosteroids only. Thymopoiesis was evaluated by measuring naive T cells, TRECs, IL-7, and T cell receptor repertoire diversity. Regulatory T cell (Treg) enumeration and function and dendritic cell (DC) enumeration and phenotype were analyzed using flow cytometry. T cell transcriptome analysis was performed on ECP patients after treatment and responders pre- and post-treatment. Four ECP responders demonstrated thymic-dependent T cell recovery, and superior median naïve T cell numbers at 8 and 12 months post-HSCT compared to the aGVHD corticosteroid group. Increased Tregs and Treg suppressive function, reduced cDC/pDC and DC co-stimulatory marker expression in ECP responders suggest upregulated peripheral tolerance; these findings were not observed in partial responders. Responder post-ECP CD3+ T cell transcriptional profile demonstrated 3333 downregulated and 364 upregulated genes, with significant downregulation of ERRα and GαS pathways, and reduced expression of pro-inflammatory and adhesion proteins.Thymic function improves with successful ECP treatment. ECP reduces T cell activation and impacts peripheral tolerance via DCs and Tregs. Differences in thymic recovery, DC, and Treg cellular patterns and the T cell transcriptome were observed between ECP responders and partial responders and require further validation and investigation in additional patients.


Assuntos
Células Dendríticas/imunologia , Doença Enxerto-Hospedeiro/terapia , Fotoferese , Linfócitos T/imunologia , Adolescente , Criança , Pré-Escolar , Feminino , Doença Enxerto-Hospedeiro/sangue , Doença Enxerto-Hospedeiro/imunologia , Humanos , Lactente , Interleucina-7/sangue , Masculino , Receptores de Antígenos de Linfócitos T/imunologia , Timo/citologia , Transcriptoma
3.
PLoS One ; 9(6): e98328, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24896823

RESUMO

Chemokine CXCL12 promotes growth and metastasis of more than 20 different human cancers, as well as pathogenesis of other common diseases. CXCL12 binds two different receptors, CXCR4 and CXCR7, both of which recruit and signal through the cytosolic adapter protein ß-arrestin 2. Differences in CXCL12-dependent recruitment of ß-arrestin 2 in cells expressing one or both receptors remain poorly defined. To quantitatively investigate parameters controlling association of ß-arrestin 2 with CXCR4 or CXCR7 in cells co-expressing both receptors, we used a systems biology approach combining real-time, multi-spectral luciferase complementation imaging with computational modeling. Cells expressing only CXCR4 maintain low basal association with ß-arrestin 2, and CXCL12 induces a rapid, transient increase in this interaction. In contrast, cells expressing only CXCR7 have higher basal association with ß-arrestin 2 and exhibit more gradual, prolonged recruitment of ß-arrestin 2 in response to CXCL12. We developed and fit a data-driven computational model for association of either CXCR4 or CXCR7 with ß-arrestin 2 in cells expressing only one type of receptor. We then experimentally validated model predictions that co-expression of CXCR4 and CXCR7 on the same cell substantially decreases both the magnitude and duration of CXCL12-regulated recruitment of ß-arrestin 2 to CXCR4. Co-expression of both receptors on the same cell only minimally alters recruitment of ß-arrestin 2 to CXCR7. In silico experiments also identified ß-arrestin 2 as a limiting factor in cells expressing both receptors, establishing that CXCR7 wins the "competition" with CXCR4 for CXCL12 and recruitment of ß-arrestin 2. These results reveal how competition for ß-arrestin 2 controls integrated responses to CXCL12 in cells expressing both CXCR4 and CXCR7. These results advance understanding of normal and pathologic functions of CXCL12, which is critical for developing effective strategies to target these pathways therapeutically.


Assuntos
Arrestinas/metabolismo , Quimiocina CXCL12/metabolismo , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Transdução de Sinais/fisiologia , Linhagem Celular Tumoral , Humanos , beta-Arrestina 2 , beta-Arrestinas
4.
Biochem Pharmacol ; 86(9): 1263-71, 2013 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-23973527

RESUMO

The chemokine CXCL12 and its shared seven-transmembrane receptors CXCR4 and CXCR7 regulate diseases including cancer, atherosclerosis, autoimmunity, and HIV infection, making these molecules promising drug targets. These molecules also control key processes in normal development and physiology, suggesting the need to selectively modulate CXCR4 and/or CXCR7 functions and signaling to reduce potential complications of long-term therapy. We previously identified two peptides that functioned as allosteric agonists driving CXCR4-dependent chemotaxis, providing key structural information to design a small number of additional peptides to investigate determinants of CXCL12 interactions and signaling through CXCR4 and CXCR7. In the current study, we show that the previously identified peptides only minimally activated CXCR4 signaling through the cytosolic adapter protein ß-arrestin 2 and do not initiate signaling to ERK1/2. By comparison, peptides with diverse N-terminal amino acid sequences effectively activated CXCR7 signaling to ß-arrestin 2. One peptide, designated as GSLW based on its N-terminal amino acids, activated CXCR7 signaling and potentiated CXCL12-CXCR7 signaling without blocking the scavenger function of CXCR7 to internalize CXCL12. These results advance our understanding of CXCR7 ligand recognition and signaling, and provide structural information to target allosteric binding sites on this receptor as chemical probes and potential therapeutic agents.


Assuntos
Peptídeos/farmacologia , Receptores CXCR4/metabolismo , Receptores CXCR/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Arrestinas/metabolismo , Ligação Competitiva , Linhagem Celular Tumoral , Quimiocina CXCL12/metabolismo , Relação Dose-Resposta a Droga , Humanos , Ligantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Dados de Sequência Molecular , Terapia de Alvo Molecular/métodos , Peptídeos/química , Peptídeos/metabolismo , Receptores CXCR/genética , Receptores CXCR4/genética , Transdução de Sinais/efeitos dos fármacos , beta-Arrestina 2 , beta-Arrestinas
5.
PLoS One ; 8(1): e51500, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23372646

RESUMO

Chemokine CXCL12 and receptor CXCR4 have emerged as promising therapeutic targets for ovarian cancer, a disease that continues to have a dismal prognosis. CXCL12-CXCR4 signaling drives proliferation, survival, and invasion of ovarian cancer cells, leading to tumor growth and metastasis. Pleiotropic effects of CXCR4 in multiple key steps in ovarian cancer suggest that blocking this pathway will improve outcomes for patients with this disease. To quantify CXCL12-CXCR4 signaling in cell-based assays and living mouse models of ovarian cancer, we developed a click beetle red luciferase complementation reporter that detects activation of CXCR4 based on recruitment of the cytosolic adapter protein ß-arrestin 2. Both in two-dimensional and three-dimensional cell cultures, we established that bioluminescence from this reporter measures CXCL12-dependent activation of CXCR4 and inhibition of this pathway with AMD3100, a clinically-approved small molecule that blocks CXCL12-CXCR4 binding. We used this imaging system to quantify CXCL12-CXCR4 signaling in a mouse model of metastatic ovarian cancer and showed that treatment with AMD3100 interrupted this pathway in vivo. Combination therapy with AMD3100 and cisplatin significantly decreased tumor burden in mice, although differences in overall survival were not significantly greater than treatment with either agent as monotherapy. These studies establish a molecular imaging reporter system for analyzing CXCL12-CXCR4 signaling in ovarian cancer, which can be used to investigate biology and therapeutic targeting of this pathway in cell-based assays and living mice.


Assuntos
Antineoplásicos/farmacologia , Quimiocina CXCL12/genética , Compostos Heterocíclicos/farmacologia , Imagem Molecular/métodos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Receptores CXCR4/genética , Animais , Arrestinas/genética , Arrestinas/metabolismo , Benzilaminas , Linhagem Celular Tumoral , Proliferação de Células , Quimiocina CXCL12/metabolismo , Cisplatino/farmacologia , Ciclamos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Humanos , Luciferases , Medições Luminescentes , Camundongos , Camundongos SCID , Transplante de Neoplasias , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Receptores CXCR4/antagonistas & inibidores , Receptores CXCR4/metabolismo , Transdução de Sinais/efeitos dos fármacos , beta-Arrestina 2 , beta-Arrestinas
6.
Int J Biochem Cell Biol ; 44(4): 669-78, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22300987

RESUMO

Chemokine receptor CXCR7 is essential for normal development, and this receptor promotes initiation and progression of diseases including cancer and autoimmunity. To understand normal and pathologic functions of CXCR7 and advance development of therapeutic agents, there is a need to define structural domains that regulate this receptor. We generated mutants of CXCR7 with deletion of different lengths of the predicted intracellular tail and analyzed effects on CXCR7 signaling and function in cell-based assays. While wild-type CXCR7 predominantly localized to intracellular vesicles, progressive deletion of the carboxy terminus redistributed the receptor to the plasma membrane. Truncating the intracellular tail of CXCR7 did not alter binding to CXCL12, but mutant receptors had reduced scavenging of this chemokine. Using a firefly luciferase complementation system, we established that deletions of the carboxy terminus decreased basal interactions and eliminated ligand-dependent recruitment of the scaffolding protein ß-arrestin-2 to receptors. Deleting the carboxy terminus of CXCR7 impaired constitutive internalization of the receptor and reduced activation of ERK1/2 by CXCL12-CXCR7. Inhibiting dynamin, a molecule required for internalization of CXCR7, increased ligand-dependent association of the receptor with ß-arrestin-2 and enhanced activation of ERK1/2. These studies establish mechanisms of action for CXCR7 and establish the intracellular tail of CXCR7 as a critical determinant of receptor trafficking, chemokine scavenging, and signaling.


Assuntos
Receptores CXCR/química , Receptores CXCR/metabolismo , Arrestinas/metabolismo , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Quimiocinas/metabolismo , Ativação Enzimática , Humanos , Espaço Intracelular/metabolismo , Ligantes , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Estrutura Terciária de Proteína , Transporte Proteico , Receptores CXCR/genética , Deleção de Sequência , beta-Arrestina 2 , beta-Arrestinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...