Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(2)2023 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-36674959

RESUMEN

The strength of Ca2+ signaling is a hallmark of T cell activation, yet the role of Ca2+ homeostasis in developing T cells before expressing a mature T cell receptor is poorly understood. We aimed to unveil specific functions of the two plasma membrane Ca2+ ATPases expressed in T cells, PMCA1 and PMCA4. On a transcriptional and protein level we found that PMCA4 was expressed at low levels in CD4-CD8- double negative (DN) thymocytes and was even downregulated in subsequent stages while PMCA1 was present throughout development and upregulated in CD4+CD8+ double positive (DP) thymocytes. Mice with a targeted deletion of Pmca1 in DN3 thymocytes had an almost complete block of DP thymocyte development with an accumulation of DN4 thymocytes but severely reduced numbers of CD8+ immature single positive (ISP) thymocytes. The DN4 thymocytes of these mice showed strongly elevated basal cytosolic Ca2+ levels and a pre-mature CD5 expression, but in contrast to the DP thymocytes they were only mildly prone to apoptosis. Surprisingly, mice with a germline deletion of Pmca4 did not show any signs of altered progression through the developmental thymocyte stages, nor altered Ca2+ homeostasis throughout this process. PMCA1 is, therefore, non-redundant in keeping cellular Ca2+ levels low in the early thymocyte development required for the DN to DP transition.


Asunto(s)
Adenosina Trifosfatasas , Timocitos , Ratones , Animales , Timocitos/metabolismo , Antígenos CD8/metabolismo , Adenosina Trifosfatasas/metabolismo , Antígenos CD4/metabolismo , Membrana Celular/metabolismo , Homeostasis , Diferenciación Celular/genética , Timo/metabolismo
2.
Front Cell Dev Biol ; 10: 654181, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35281114

RESUMEN

Btk and Vav proteins are all components of the signalosome that builds upon B cell receptor (BCR) activation. However, the role of Vav proteins within the signalosome is quite complex and not yet fully understood. Until now, studies of these have focused predominantly on a deficiency of Vav proteins alone or in combination with other Vav protein family members. Since a physical association of Btk with Vav was shown previously, we asked whether these molecules lie in the same or independent signaling pathways. By analyzing Vav1 and Vav3 single knock-out mice and generating double-knock-out animals deficient for either Vav1 or Vav3 and Btk, we observed, in line with previous publications, no severe B cell developmental defects when either Vav1 or Vav3 alone are not expressed. However, a simultaneous deficiency of Btk together with either Vav1 or Vav3 leads to a severe reduction of splenic B cells, which exhibit an immature phenotype. B cell developmental defects of Btk/Vav1-double deficient mice in the periphery were more severe than those observed in Btk-single-deficient animals. Additionally, morphological changes in splenic microarchitecture were observed in double- but also in single-knock-out mutants. These observations were accompanied by reduced BCR-induced Ca2+ mobilization, proliferation, germinal center formation and immunoglobulin secretion. Although deletion of Btk alone impaired Ca2+ mobilization upon BCR activation, the defect was even more severe when Vav1 or Vav3 were also mutated, indicating that Btk and the Vav proteins act in separate pathways that converge on Ca2+ signaling. In vitro ASC differentiation suggests that both B and T cells contribute to the observed phenotype of a Btk/Vav-double deficiency. Our results show that Vav proteins and Btk are both components of the BCR-activated signalosome but control separate signaling pathways important for B cell development.

3.
Development ; 149(8)2022 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-34528666

RESUMEN

B cell participation in early embryo/fetal development and the underlying molecular pathways have not been explored. To understand whether maternal B cell absence or impaired signaling interferes with placental and fetal growth, we paired CD19-deficient (CD19-/-) mice, females with B cell-specific MyD88 (BMyD88-/-) or IL10 (BIL10-/-) deficiency as well as wild-type and MyD88-/- controls on C57Bl/6 background with BALB/c males. Pregnancies were followed by ultrasound and Doppler measurements. Implantation number was reduced in BMyD88-/- and MyD88-/- mice. Loss of MyD88 or B cell-specific deletion of MyD88 or IL10 resulted in decreased implantation areas at gestational day (gd) 5, gd8 and gd10, accompanied by reduced placental thickness, diameter and areas at gd10. Uterine artery resistance was enhanced in BIL10-/- dams at gd10. Challenge with 0.4 mg lipopolysaccharide/kg bodyweight at gd16 revealed that BMyD88-/-, BIL10-/- and CD19-/- mothers delivered preterm, whereas controls maintained their pregnancy. B cell-specific MyD88 and IL10 expression is essential for appropriate in utero development. IL10+B cells are involved in uterine blood flow regulation during pregnancy. Finally, B cell-specific CD19, MyD88 and IL10 expression influences susceptibility towards preterm birth.


Asunto(s)
Linfocitos B/metabolismo , Desarrollo Fetal , Feto/embriología , Transducción de Señal , Arteria Uterina/metabolismo , Útero , Resistencia Vascular , Animales , Antígenos CD19/genética , Antígenos CD19/metabolismo , Femenino , Interleucina-10/deficiencia , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/metabolismo , Embarazo , Útero/irrigación sanguínea , Útero/metabolismo
4.
Cells ; 10(10)2021 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-34685673

RESUMEN

Immunological networks balance tolerance towards paternal alloantigens during pregnancy with normal immune response to pathogens. Subclinical infections can impact this balance and lead to preterm birth or even intrauterine fetal death (IUFD). We recently showed that loss of maternal B cells renders murine fetuses susceptible to IUFD after LPS exposure. Since the signaling pathway involved in this B-cell mediated response remains unclear, we aimed to understand the participation of MyD88 in this response using B-cell-specific MyD88-deficient (BMyD88-/-) mice. B cells isolated from wild-type (WT), BMyD88-/-, CD19-/- and MyD88-/- dams on gestational day (gd) 10 responded differently to LPS concerning cytokine secretion. In vivo LPS challenge on gd 10 provoked IUFD in CD19-/- mothers with functional MyD88, while fetuses from BMyD88-/- and MyD88-/- mice were protected. These outcomes were associated with altered cytokine levels in the maternal serum and changes in CD4+ T-cell responses. Overall, the loss of MyD88 signaling in maternal B cells prevents the activation of cytokine release that leads to IUFD. Thus, while MyD88 signaling in maternal B cells protects the mother from infection, it ultimately kills the fetus. Understanding the cellular mechanisms underlying infection-driven pregnancy complications is the first step to designing powerful therapeutic strategies in the future.


Asunto(s)
Linfocitos B/metabolismo , Muerte Fetal/etiología , Factor 88 de Diferenciación Mieloide/metabolismo , Transducción de Señal , Animales , Antígenos CD19/metabolismo , Citocinas/metabolismo , Decidua/patología , Femenino , Inflamación/patología , Lipopolisacáridos , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Factor 88 de Diferenciación Mieloide/deficiencia , Embarazo
5.
J Leukoc Biol ; 110(5): 839-852, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33527537

RESUMEN

Immunity is governed by successful T cell migration, optimized to enable a T cell to fully scan its environment without wasted movement by balancing speed and turning. Here we report that the Arhgef6 RhoGEF (aka alpha-PIX; αPIX; Cool-2), an activator of small GTPases, is required to restrain cell migration speed and cell turning during spontaneous migration on 2D surfaces. In Arhgef6-/- T cells, expression of Arhgef7 (beta-PIX; ßPIX; Cool-1), a homolog of Arhgef6, was increased and correlated with defective activation and localization of Rac1 and CDC42 GTPases, respectively. Downstream of Arhgef6, PAK2 (p21-activated kinase 2) and LIMK1 phosphorylation was reduced, leading to increased activation of Cofilin, the actin-severing factor. Consistent with defects in these signaling pathways, Arhgef6-/- T cells displayed abnormal bilobed lamellipodia and migrated faster, turned more, and arrested less than wild-type (WT) T cells. Using pharmacologic inhibition of LIMK1 (LIM domain kinase 1) to induce Cofilin activation in WT T cells, we observed increased migration speed but not increased cell turning. In contrast, inhibition of Cdc42 increased cell turning but not speed. These results suggested that the increased speed of the Arhgef6-/- T cells is due to hyperactive Cofilin while the increased turning may be due to abnormal GTPase activation and recruitment. Together, these findings reveal that Arhgef6 acts as a repressor of T cell speed and turning by limiting actin polymerization and lamellipodia formation.


Asunto(s)
Factores Despolimerizantes de la Actina/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Quimiotaxis de Leucocito/fisiología , GTP Fosfohidrolasas/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Actinas/metabolismo , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Polimerizacion , Seudópodos/metabolismo , Transducción de Señal/inmunología
6.
Eur J Immunol ; 51(3): 594-602, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33098669

RESUMEN

The amplitude and duration of Ca2+ signaling is crucial for B-cell development and self-tolerance; however, the mechanisms for terminating Ca2+ signals in B cells have not been determined. In lymphocytes, plasma membrane Ca2+ ATPase (PMCA) isoforms 1 and 4 (PMCA1 and PMCA4, aka ATP2B1 and ATP2B4) are the main candidates for expelling Ca2+ from the cell through the plasma membrane. We report here that Pmca4 (Atp2b4) KO mice had normal B-cell development, while mice with a conditional KO of Pmca1 (Atp2b1) had greatly reduced numbers of B cells, particularly splenic follicular B cells, marginal zone B cells, and peritoneal B-1a cells. Mouse and naïve human B cells showed only PMCA1 expression and no PMCA4 by western blot, in contrast to T cells, which did express PMCA4. Calcium handling was normal in Pmca4-/- B cells, but Pmca1 KO B cells had elevated basal levels of Ca2+ , elevated levels in ER stores, and reduced Ca2+ clearance. These findings show that the PMCA1 isoform alone is required to ensure normal B-cell Ca2+ signaling and development, which may have implications for therapeutic targeting of PMCAs and Ca2+ in B cells.


Asunto(s)
Linfocitos B/metabolismo , ATPasas Transportadoras de Calcio/metabolismo , Calcio/metabolismo , Membrana Celular/metabolismo , Homeostasis/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Isoformas de Proteínas/metabolismo , Transducción de Señal/fisiología
7.
J Cell Biol ; 219(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32379884

RESUMEN

Cells navigating through complex tissues face a fundamental challenge: while multiple protrusions explore different paths, the cell needs to avoid entanglement. How a cell surveys and then corrects its own shape is poorly understood. Here, we demonstrate that spatially distinct microtubule dynamics regulate amoeboid cell migration by locally promoting the retraction of protrusions. In migrating dendritic cells, local microtubule depolymerization within protrusions remote from the microtubule organizing center triggers actomyosin contractility controlled by RhoA and its exchange factor Lfc. Depletion of Lfc leads to aberrant myosin localization, thereby causing two effects that rate-limit locomotion: (1) impaired cell edge coordination during path finding and (2) defective adhesion resolution. Compromised shape control is particularly hindering in geometrically complex microenvironments, where it leads to entanglement and ultimately fragmentation of the cell body. We thus demonstrate that microtubules can act as a proprioceptive device: they sense cell shape and control actomyosin retraction to sustain cellular coherence.


Asunto(s)
Actomiosina/metabolismo , Movimiento Celular/fisiología , Células Dendríticas/citología , Centro Organizador de los Microtúbulos/metabolismo , Microtúbulos/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Adhesión Celular/fisiología , Forma de la Célula/fisiología , Células Cultivadas , Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Centro Organizador de los Microtúbulos/efectos de los fármacos , Microtúbulos/efectos de los fármacos , Nocodazol/farmacología , Unión Proteica , Factores de Intercambio de Guanina Nucleótido Rho/deficiencia , Factores de Intercambio de Guanina Nucleótido Rho/genética
8.
Int J Mol Sci ; 21(3)2020 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-32013053

RESUMEN

Vav proteins activate GTPases of the RhoA subfamily that regulate the cytoskeleton and are involved in adhesion, migration, differentiation, polarity and the cell cycle. While the importance of RhoA GTPases for neuronal morphology is undisputed, their regulation is less well understood. In this perspective, we studied the consequences of the deletion of Vav2, Vav3 and Vav2 and 3 (Vav2-/-, Vav3-/-, Vav2-/-/3-/-) for the development of embryonic hippocampal neurons in vitro. Using an indirect co-culture system of hippocampal neurons with primary wild-type (WT) cortical astrocytes, we analysed axonal and dendritic parameters, structural synapse numbers and the spontaneous network activity via immunocytochemistry and multielectrode array analysis (MEA). Here, we observed a higher process complexity in Vav3-/-, but not in Vav2-/- neurons after three and five days in vitro (DIV). Furthermore, an enhanced synapse formation was observed in Vav3-/- after 14 days in culture. Remarkably, Vav2-/-/3-/- double knockout neurons did not display synergistic effects. Interestingly, these differences were transient and compensated after a cultivation period of 21 days. Network analysis revealed a diminished number of spontaneously occurring action potentials in Vav3-/- neurons after 21 DIV. Based on these results, it appears that Vav3 participates in key events of neuronal differentiation.


Asunto(s)
Astrocitos/citología , Hipocampo/embriología , Neuronas/citología , Proteínas Proto-Oncogénicas c-vav/genética , Potenciales de Acción , Animales , Astrocitos/metabolismo , Diferenciación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Técnicas de Inactivación de Genes , Hipocampo/citología , Hipocampo/metabolismo , Masculino , Ratones , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Análisis de Matrices Tisulares
9.
J Vis Exp ; (141)2018 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-30531718

RESUMEN

Marginal zone B cells (MZBs) are a population of B cells that reside in the mouse splenic marginal zones that envelop follicles. To reach the follicles, MZBs must migrate up the shear force of blood flow. We present here a method for analyzing this flow-induced MZB migration in vitro. First, MZBs are isolated from the mouse spleen. Second, MZBs are settled on integrin ligands in flow chamber slides, exposed to shear flow, and imaged under a microscope while migrating. Third, images of the migrating MZBs are processed using the MTrack2 automatic cell tracking plugin for ImageJ, and the resulting cell tracks are quantified using the Ibidi chemotaxis tool. The migration data reveal how fast the cells move, how often they change direction, whether the shear flow vector affects their migration direction, and which integrin ligands are involved. Although we use MZBs, the method can easily be adapted for analyzing migration of any leukocyte that responds to the force of shear flow.


Asunto(s)
Linfocitos B/fisiología , Movimiento Celular/fisiología , Quimiotaxis/fisiología , Imagen de Lapso de Tiempo/métodos , Animales , Linfocitos B/química , Células Cultivadas , Tejido Linfoide/química , Tejido Linfoide/citología , Tejido Linfoide/fisiología , Ratones , Bazo/química , Bazo/citología , Bazo/fisiología
10.
Sci Rep ; 8(1): 8811, 2018 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-29891904

RESUMEN

The characteristic six layers of the mammalian neocortex develop sequentially as neurons are generated by neural progenitors and subsequently migrate past older neurons to their final position in the cortical plate. One of the earliest steps of neuronal differentiation is the formation of an axon. Small GTPases play essential roles during this process by regulating cytoskeletal dynamics and intracellular trafficking. While the function of GTPases has been studied extensively in cultured neurons and in vivo much less is known about their upstream regulators. Here we show that Arhgef7 (also called ßPix or Cool1) is essential for axon formation during cortical development. The loss of Arhgef7 results in an extensive loss of axons in cultured neurons and in the developing cortex. Arhgef7 is a guanine-nucleotide exchange factor (GEF) for Cdc42, a GTPase that has a central role in directing the formation of axons during brain development. However, active Cdc42 was not able to rescue the knockdown of Arhgef7. We show that Arhgef7 interacts with the GTPase TC10 that is closely related to Cdc42. Expression of active TC10 can restore the ability to extend axons in Arhgef7-deficient neurons. Our results identify an essential role of Arhgef7 during neuronal development that promotes axon formation upstream of TC10.


Asunto(s)
Axones/fisiología , Diferenciación Celular , Corteza Cerebral/embriología , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Animales , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Factores de Intercambio de Guanina Nucleótido Rho/deficiencia , Proteína de Unión al GTP cdc42/metabolismo
11.
Nat Commun ; 8(1): 2261, 2017 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-29273735

RESUMEN

Splenic marginal zone B cells (MZB) shuttle between the blood-filled marginal zone for antigen collection and the follicle for antigen delivery. However, it is unclear how MZBs migrate directionally from the marginal zone to the follicle. Here, we show that murine MZBs migrate up shear flow via the LFA-1 (αLß2) integrin ligand ICAM-1, but adhere or migrate down the flow via the VLA-4 integrin (α4ß1) ligand VCAM-1. MZBs lacking Arhgef6 (Pak-interacting exchange factor (αPIX)) or functional LFA-1 are impaired in shuttling due to mislocalization toward the VCAM-1-rich red pulp. Sphingosine-1-phosphate (S1P) signaling through the S1PR3 receptor inhibits MZB migration up the flow, and deletion of S1pr3 in Arhgef6 -/- mice rescues mislocalized MZBs. These findings establish shear flow as a directional cue for MZB migration to the follicle, and define S1PR3 and VCAM-1 as counteracting forces that inhibit this migration.


Asunto(s)
Linfocitos B/fisiología , Movimiento Celular/fisiología , Molécula 1 de Adhesión Intercelular/metabolismo , Lisofosfolípidos/metabolismo , Receptores de Lisoesfingolípidos/genética , Esfingosina/análogos & derivados , Estrés Mecánico , Molécula 1 de Adhesión Celular Vascular/metabolismo , Animales , Linfocitos B/metabolismo , Integrina alfa4beta1/metabolismo , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Ratones , Ratones Noqueados , Receptores de Lisoesfingolípidos/metabolismo , Flujo Sanguíneo Regional , Factores de Intercambio de Guanina Nucleótido Rho/genética , Transducción de Señal , Esfingosina/metabolismo , Receptores de Esfingosina-1-Fosfato , Bazo/citología
12.
Sci Rep ; 7(1): 8358, 2017 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-28827723

RESUMEN

The outcome of T cell activation is determined by mechanisms that balance Ca2+ influx and clearance. Here we report that murine CD4 T cells lacking Neuroplastin (Nptn -/-), an immunoglobulin superfamily protein, display elevated cytosolic Ca2+ and impaired post-stimulation Ca2+ clearance, along with increased nuclear levels of NFAT transcription factor and enhanced T cell receptor-induced cytokine production. On the molecular level, we identified plasma membrane Ca2+ ATPases (PMCAs) as the main interaction partners of Neuroplastin. PMCA levels were reduced by over 70% in Nptn -/- T cells, suggesting an explanation for altered Ca2+ handling. Supporting this, Ca2+ extrusion was impaired while Ca2+ levels in internal stores were increased. T cells heterozygous for PMCA1 mimicked the phenotype of Nptn -/- T cells. Consistent with sustained Ca2+ levels, differentiation of Nptn -/- T helper cells was biased towards the Th1 versus Th2 subset. Our study thus establishes Neuroplastin-PMCA modules as important regulators of T cell activation.


Asunto(s)
Calcio/metabolismo , Membrana Celular/metabolismo , Glicoproteínas de Membrana/fisiología , ATPasas Transportadoras de Calcio de la Membrana Plasmática/fisiología , Linfocitos T/fisiología , Animales , Señalización del Calcio , Diferenciación Celular , Núcleo Celular , Regulación de la Expresión Génica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T/inmunología
13.
Eur J Immunol ; 44(7): 2139-52, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24723371

RESUMEN

Although CD8(+) T cells that produce IL-17 (Tc17 cells) have been linked to host defense, Tc17 cells show reduced cytotoxic activity, which is the characteristic function of CD8(+) T cells. Here, we show that CTLA-4 enhances the frequency of IL-17 in CD8(+) T cells, indicating that CTLA-4 (CD152) specifically promotes Tc17 differentiation. Simultaneous stimulation of CTLA-4(+/+) and CTLA-4(-/-) T cells in cocultures and agonistic CTLA-4 stimulation unambiguously revealed a cell-intrinsic mechanism for IL-17 control by CTLA-4. The quality of CTLA-4-induced Tc17 cells was tested in vivo, utilizing infection with the facultative intracellular bacterium Listeria monocytogenes (LM). Unlike CTLA-4(+/+) Tc17 cells, CTLA-4(-/-) were nearly as efficient as Tc1 CTLA-4(+/+) cells in LM clearance. Additionally, adoptively transferred CTLA-4(-/-) Tc17 cells expressed granzyme B after rechallenge, and produced Tc1 cytokines such as IFN-γ and TNF-α, which strongly correlate with bacterial clearance. CTLA-4(+/+) Tc17 cells demonstrated a high-quality Tc17 differentiation program ex vivo, which was also evident in isolated IL-17-secreting Tc17 cells, with CTLA-4-mediated enhanced upregulation of Tc17-related molecules such as IL-17A, RORγt, and IRF-4. Our results show that CTLA-4 promotes Tc17 differentiation that results in robust Tc17 responses. Its inactivation might therefore represent a central therapeutic target to enhance clearance of infection.


Asunto(s)
Linfocitos T CD8-positivos/citología , Antígeno CTLA-4/fisiología , Diferenciación Celular , Interleucina-17/biosíntesis , Animales , Apoptosis , Linaje de la Célula , Proliferación Celular , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Granzimas/biosíntesis , Ratones , Ratones Endogámicos C57BL
14.
J Immunol ; 192(7): 3228-38, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24591366

RESUMEN

Thymocytes mature in a series of stages by migrating through specific areas of the thymus and interacting with other cells to receive the necessary developmental signals; however, little is known about the molecular mechanisms governing this migration. We report that murine thymocytes with a knockout mutation in α-PAK (p21-activated kinase)-interacting exchange factor (PIX; Arhgef6), an activator of Rho GTPases, showed greatly increased motility and altered morphology in two-dimensional migration on ICAM-1. αPIX was also required for efficient positive selection, but not negative selection, of thymocytes. TCR signaling was normal in αPix(-) thymocytes, indicating that the effects of αPIX on positive selection are largely independent of TCR signaling. αPix(-) thymocytes also paused less during migration in the thymic cortex, interacted less with ICAM-1 coated beads, and could overcome TCR stop signals, consistent with defective scanning behavior. These results identify αPIX as a regulator of thymocyte migration and subsequent arrest that is linked to positive selection.


Asunto(s)
Movimiento Celular/inmunología , Factores de Intercambio de Guanina Nucleótido Rho/inmunología , Timocitos/inmunología , Timo/inmunología , Animales , Movimiento Celular/genética , Células Cultivadas , Citometría de Flujo , Molécula 1 de Adhesión Intercelular/inmunología , Molécula 1 de Adhesión Intercelular/metabolismo , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Unión Proteica/inmunología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Timocitos/citología , Timocitos/metabolismo , Timo/citología , Timo/metabolismo
15.
Mol Cell Proteomics ; 12(9): 2551-67, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23754785

RESUMEN

The formation of the immunological synapse between T cells and antigen-presenting cells (APC) begins within minutes of contact and can take hours for full T-cell activation. Although early phases of the synapse have been extensively studied for a select number of proteins, later phases have not yet been examined in detail. We studied the signaling network in stable synapses by measuring the simultaneous localization of 25 signaling and structural molecules over 2 h at the level of individual synapses using multi-epitope ligand cartography (MELC). Signaling proteins including phospho(p)ZAP70, pSLP76, pCD3ζ, and pLAT, along with proteins that influence synapse structure such as F-actin, tubulin, CD45, and ICAM-1, were localized in images of synapses and revealed the multidimensional construction of a mature synapse. The construction of the stable synapse included intense early TCR signaling, a phase of recruitment of structural proteins, and a sustained increase in signaling molecules and colocalization of TCR and pLAT signaling clusters in the center of the synapse. Consolidation of TCR and associated proteins resulted in formation of a small number of discrete synaptic microclusters. Development of synapses and cSMAC composition was greatly affected by the absence of Vav1, with an associated loss in PLCγ1 recruitment, pSLP76, and increased CXCR4. Together, these data demonstrate the use of multi-epitope ligand cartography to quantitatively analyze synapse formation and reveal successive recruitment of structural and signaling proteins and sustained phosphorylation at the mature synapse.


Asunto(s)
Sinapsis Inmunológicas/metabolismo , Transducción de Señal/inmunología , Animales , Pollos , Análisis por Conglomerados , Mapeo Epitopo , Epítopos/metabolismo , Cinética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Proto-Oncogénicas c-vav/deficiencia , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
16.
J Exp Med ; 200(11): 1491-501, 2004 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-15583019

RESUMEN

Marginal zones (MZs) are microdomains in the spleen that contain various types of immune cells, including MZ B cells, MOMA1(+) metallophilic macrophages, and mucosal addressin cell adhesion molecule 1 (MAdCAM-1)(+) endothelial cells. MAdCAM-1(+) and MOMA1(+) cells line the sinus, that separates MZs from splenic follicles. Here we show that a receptor for the lysophospholipid sphingosine-1-phosphate (S1P), S1P(3), is required for normal numbers of splenic immature and MZ B cells, and for S1P-induced chemotaxis of MZ B cells. S1P(3) is also essential for proper alignment of MOMA1(+) macrophages and MAdCAM-1(+) endothelial cells along the marginal sinus. The lack of cohesion of the marginal sinus in S1P(3)(-/-) mice affects MZ B cell functions, as wild-type (WT) MZ B cells migrate more into S1P(3)(-/-) follicles than into WT follicles after treatment with lipopolysaccharide. Additionally, short-term homing experiments demonstrate that WT MZ B cells home to the S1P(3)(-/-) spleen in increased numbers, suggesting a role for the marginal sinus in regulating MZ B cells numbers. Moreover, S1P(3)(-/-) mice are defective in mounting immune responses to thymus-independent antigen type 2 due to defects in radiation-resistant cells in the spleen. These data identify lysophospholipids and the S1P(3) receptor as essential regulators of the MZ sinus and its role as a barrier to the follicle.


Asunto(s)
Linfocitos B/fisiología , Células Endoteliales/fisiología , Inmunoglobulinas/análisis , Mucoproteínas/análisis , Receptores de Lisoesfingolípidos/fisiología , Bazo/citología , Animales , Moléculas de Adhesión Celular , Movimiento Celular , Lipopolisacáridos/farmacología , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...