Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Más filtros

Banco de datos
Tipo del documento
Publication year range
1.
Immunity ; 47(5): 862-874.e3, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29166587

RESUMEN

Chemoattractant-mediated recruitment of hematopoietic cells to sites of pathogen growth or tissue damage is critical to host defense and organ homeostasis. Chemotaxis is typically considered to rely on spatial sensing, with cells following concentration gradients as long as these are present. Utilizing a microfluidic approach, we found that stable gradients of intermediate chemokines (CCL19 and CXCL12) failed to promote persistent directional migration of dendritic cells or neutrophils. Instead, rising chemokine concentrations were needed, implying that temporal sensing mechanisms controlled prolonged responses to these ligands. This behavior was found to depend on G-coupled receptor kinase-mediated negative regulation of receptor signaling and contrasted with responses to an end agonist chemoattractant (C5a), for which a stable gradient led to persistent migration. These findings identify temporal sensing as a key requirement for long-range myeloid cell migration to intermediate chemokines and provide insights into the mechanisms controlling immune cell motility in complex tissue environments.


Asunto(s)
Movimiento Celular , Factores Quimiotácticos/fisiología , Células Mieloides/fisiología , Animales , Quimiocina CCL19/fisiología , Quimiocina CXCL12/fisiología , Células Dendríticas/fisiología , Quinasa 3 del Receptor Acoplado a Proteína-G/fisiología , Quinasas de Receptores Acoplados a Proteína-G/fisiología , Ratones , Ratones Endogámicos C57BL , Microfluídica
2.
PLoS Comput Biol ; 9(9): e1003222, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24068905

RESUMEN

Natural killer (NK) cells are innate lymphocytes that provide early host defense against intracellular pathogens, such as viruses. Although NK cell development, homeostasis, and proliferation are regulated by IL-15, the influence of IL-15 receptor (IL-15R)-mediated signaling at the cellular level has not been quantitatively characterized. We developed a mathematical model to analyze the kinetic interactions that control the formation and localization of IL-15/IL-15R complexes. Our computational results demonstrated that IL-15/IL-15R complexes on the cell surface were a key determinant of the magnitude of the IL-15 proliferative signal and that IL-15R occupancy functioned as an effective surrogate measure of receptor signaling. Ligand binding and receptor internalization modulated IL-15R occupancy. Our work supports the hypothesis that the total number and duration of IL-15/IL-15R complexes on the cell surface crosses a quantitative threshold prior to the initiation of NK cell division. Furthermore, our model predicted that the upregulation of IL-15Rα on NK cells substantially increased IL-15R complex formation and accelerated the expansion of dividing NK cells with the greatest impact at low IL-15 concentrations. Model predictions of the threshold requirement for NK cell recruitment to the cell cycle and the subsequent exponential proliferation correlated well with experimental data. In summary, our modeling analysis provides quantitative insight into the regulation of NK cell proliferation at the receptor level and provides a framework for the development of IL-15 based immunotherapies to modulate NK cell proliferation.


Asunto(s)
Proliferación Celular , Células Asesinas Naturales/citología , Modelos Biológicos , Receptores de Interleucina-15/agonistas , Humanos , Células Asesinas Naturales/inmunología
3.
J Immunol ; 188(7): 2981-90, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22379026

RESUMEN

NK cells are innate lymphocytes that mediate early host defense against viruses, such as cytomegalovirus. IL-15 is upregulated during viral infections and drives the expansion of NK cells. However, the influence of IL-15 on murine NK cell division and death rates has not been quantitatively studied. Therefore, we developed a series of two-compartment (representing quiescent and dividing NK cell subpopulations) mathematical models, incorporating different assumptions about the kinetic parameters regulating NK cell expansion. Using experimentally derived division and death rates, we tested each model's assumptions by comparing predictions of NK cell numbers with independent experimental results and demonstrated that the kinetic parameters are distinct for nondividing and dividing NK cell subpopulations. IL-15 influenced NK cell expansion by modulating recruitment and division rates to a greater extent than death rates. The observed time delay to first division could be accounted for by differences in the kinetic parameters of nondividing and dividing subsets of NK cells. Although the duration of the time delay to first division was not significantly influenced by IL-15, the recruitment of nondividing NK cells into the replicating subpopulation increased with greater IL-15 concentrations. Our model quantitatively predicted changes in NK cell accumulation when IL-15 stimulation was reduced, demonstrating that NK cell divisional commitment was interrupted when cytokine stimulation was removed. In summary, this quantitative analysis reveals novel insights into the in vitro regulation of NK cell proliferation and provides a foundation for modeling in vivo NK cell responses to viral infections.


Asunto(s)
Interleucina-15/farmacología , Células Asesinas Naturales/citología , Modelos Inmunológicos , Animales , División Celular/efectos de los fármacos , Quimiotaxis de Leucocito , Relación Dosis-Respuesta a Droga , Femenino , Inmunidad Innata , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Organismos Libres de Patógenos Específicos
4.
J Immunol ; 183(9): 5830-6, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19828630

RESUMEN

NK cells vigorously proliferate during viral infections, resulting in an expanded pool of innate lymphocytes that are able to participate in early host defense. The relative contributions of cytokines and activation receptors in stimulating NK cell proliferation during viral infections are not well characterized. In this study, we demonstrated that signaling through the NK cell activation receptor Ly49H was able to compensate for the absence of cytokine stimulation in the preferential phase of viral-induced proliferation during murine cytomegalovirus infection. In the absence of type I IFN stimulation, NK cell proliferation was strongly biased toward cells expressing the Ly49H receptor, even at early time points when minimal preferential Ly49H-mediated proliferation was observed in wild-type mice. In the absence of effective Ly49H signaling or following infection with virus that did not express the ligand for Ly49H, no difference was observed in the proliferation of subsets of NK cells that either express or lack expression of Ly49H, although the overall proliferation of NK cells in IFNalphabetaR(-/-) mice was substantially reduced. These results highlight the contribution of NK cell activation receptors in stimulating proliferation and subsequent expansion of NK cells that are able to recognize virally infected cells.


Asunto(s)
Proliferación Celular , Infecciones por Herpesviridae/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/inmunología , Muromegalovirus/inmunología , Subfamilia A de Receptores Similares a Lectina de Células NK/metabolismo , Receptor de Interferón alfa y beta/deficiencia , Transducción de Señal/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Técnicas de Sustitución del Gen , Infecciones por Herpesviridae/genética , Células Asesinas Naturales/citología , Células Asesinas Naturales/metabolismo , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Células 3T3 NIH , Subfamilia A de Receptores Similares a Lectina de Células NK/fisiología , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/fisiología , Transducción de Señal/genética
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda