Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
J Immunol ; 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39302113

RESUMEN

Expression of IL-15 on the surface of human graft endothelial cells (ECs) bound to the IL-15Rα subunit can increase the activation of CTLs, potentiating allograft rejection. Our previous work showed that surface expression of this protein complex could be induced by alloantibody-mediated complement activation through increased IL-1ß synthesis, secretion, and autocrine/paracrine IL-1-mediated activation of NF-κB. In this article, we report that cultured human ECs express eight differently spliced IL-15 transcripts. Remarkably, IL-1ß does not alter the expression level of any IL-15 transcript but induces surface expression independently of RNA polymerase II-mediated transcription while requiring new protein translation. Mechanistically, IL-1ß causes an NF-κB-mediated reduction in the level of microRNA Let-7c-3p, thereby relieving a block of translation of IL-15 surface protein. Let7c-3p anti-miR can induce EC surface expression of IL-15/IL-15Rα in the absence of complement activation or of IL-1, enabling IL-15 transpresentation to boost CD8 T cell activation. Because of the complexity we have uncovered in IL-15 regulation, we recommend caution in interpreting increased total IL-15 mRNA or protein levels as a surrogate for transpresentation.

2.
J Clin Invest ; 133(21)2023 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-37676733

RESUMEN

Donor-recipient (D-R) mismatches outside of human leukocyte antigens (HLAs) contribute to kidney allograft loss, but the mechanisms remain unclear, specifically for intronic mismatches. We quantified non-HLA mismatches at variant-, gene-, and genome-wide scales from single nucleotide polymorphism (SNP) data of D-Rs from 2 well-phenotyped transplant cohorts: Genomics of Chronic Allograft Rejection (GoCAR; n = 385) and Clinical Trials in Organ Transplantation-01/17 (CTOT-01/17; n = 146). Unbiased gene-level screening in GoCAR uncovered the LIMS1 locus as the top-ranked gene where D-R mismatches associated with death-censored graft loss (DCGL). A previously unreported, intronic, LIMS1 haplotype of 30 SNPs independently associated with DCGL in both cohorts. Haplotype mismatches showed a dosage effect, and minor-allele introduction to major-allele-carrying recipients showed greater hazard of DCGL. The LIMS1 haplotype and the previously reported LIMS1 SNP rs893403 are expression quantitative trait loci (eQTL) in immune cells for GCC2 (not LIMS1), which encodes a protein involved in mannose-6-phosphase receptor (M6PR) recycling. Peripheral blood and T cell transcriptome analyses associated the GCC2 gene and LIMS1 SNPs with the TGF-ß1/SMAD pathway, suggesting a regulatory effect. In vitro GCC2 modulation impacted M6PR-dependent regulation of active TGF-ß1 and downstream signaling in T cells. Together, our data link LIMS1 locus D-R mismatches to DCGL via GCC2 eQTLs that modulate TGF-ß1-dependent effects on T cells.


Asunto(s)
Trasplante de Riñón , Humanos , Factor de Crecimiento Transformador beta1/genética , Rechazo de Injerto/genética , Riñón , Donantes de Tejidos , Antígenos HLA , Supervivencia de Injerto/genética , Proteínas de la Membrana , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas con Dominio LIM/genética
3.
J Immunol ; 211(6): 923-931, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37530585

RESUMEN

B cells, like T cells, can infiltrate sites of inflammation, but the processes and B cell subsets involved are poorly understood. Using human cells and in vitro assays, we find only a very small number of B cells will adhere to TNF-activated (but not to resting) human microvascular endothelial cells (ECs) under conditions of venular flow and do so by binding to ICAM-1 and VCAM-1. CXCL13 and, to a lesser extent, CXCL10 bound to the ECs can increase adhesion and induce transendothelial migration (TEM) of adherent naive and memory B cells in 10-15 min through a process involving cell spreading, translocation of the microtubule organizing center (MTOC) into a trailing uropod, and interacting with EC activated leukocyte cell adhesion molecule. Engagement of the BCR by EC-bound anti-κ L chain Ab also increases adhesion and TEM of κ+ but not λ+ B cells. BCR-induced TEM takes 30-60 min, requires Syk activation, is initiated by B cell rounding up and translocation of the microtubule organizing center to the region of the B cell adjacent to the EC, and also uses EC activated leukocyte cell adhesion molecule for TEM. BCR engagement reduces the number of B cells responding to chemokines and preferentially stimulates TEM of CD27+ B cells that coexpress IgD, with or without IgM, as well as CD43. RNA-sequencing analysis suggests that peripheral blood CD19+CD27+CD43+IgD+ cells have increased expression of genes that support BCR activation as well as innate immune properties in comparison with total peripheral blood CD19+ cells.


Asunto(s)
Molécula de Adhesión Celular del Leucocito Activado , Migración Transendotelial y Transepitelial , Humanos , Molécula de Adhesión Celular del Leucocito Activado/metabolismo , Células Endoteliales , Movimiento Celular , Endotelio Vascular/metabolismo , Quimiocinas/metabolismo , Antígenos CD/metabolismo , Células Cultivadas
4.
Sci Signal ; 16(777): eabo3406, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36943921

RESUMEN

The zinc finger protein ZFYVE21 is involved in immune signaling. Using humanized mouse models, primary human cells, and patient samples, we identified a T cell-autonomous role for ZFYVE21 in promoting chronic vascular inflammation associated with allograft vasculopathy. Ischemia-reperfusion injury (IRI) stimulated endothelial cells to produce Hedgehog (Hh) ligands, which in turn induced the production of ZFYVE21 in a population of T memory cells with high amounts of the Hh receptor PTCH1 (PTCHhi cells, CD3+CD4+CD45RO+PTCH1hiPD-1hi), vigorous recruitment to injured endothelia, and increased effector responses in vivo. After priming by interferon-γ (IFN-γ), Hh-induced ZFYVE21 activated NLRP3 inflammasome activity in T cells, which potentiated IFN-γ responses. Hh-induced NLRP3 inflammasomes and T cell-specific ZFYVE21 augmented the vascular sequelae of chronic inflammation in mice engrafted with human endothelial cells or coronary arteries that had been subjected to IRI before engraftment. Moreover, the population of PTCHhi T cells producing high amounts of ZFYVE21 was expanded in patients with renal transplant-associated IRI, and sera from these patients expanded this population in control T cells in a manner that depended on Hh signaling. We conclude that Hh-induced ZFYVE21 activates NLRP3 inflammasomes in T cells, thereby promoting chronic inflammation.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Animales , Humanos , Ratones , Células Endoteliales/metabolismo , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Inflamasomas/genética , Inflamasomas/metabolismo , Inflamación/genética , Inflamación/metabolismo , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Linfocitos T/metabolismo , Proteínas de la Membrana/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo
5.
Front Immunol ; 13: 1016361, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36275645

RESUMEN

Endothelial cells (ECs) can present antigens to circulating effector memory T cells (TEM) and to regulatory T cells (T regs), triggering antigen-specific extravasation at specific sites where foreign antigens are introduced, e.g. by infection or transplantation. We model human antigen-induced transendothelial migration (TEM) using presentation of superantigen by cultured human dermal microvascular (HDM)ECs to isolated resting human peripheral blood T cell subpopulations or to T effector cells activated in vitro. T cell receptor (TCR)-mediated cytokine synthesis, a common assay of T cell activation by antigen, is modulated by antigen-independent signals provided by various positive or negative costimulator proteins (the latter known as checkpoint inhibitors) expressed by antigen presenting cells, including ECs. We report here that some EC-expressed costimulators also modulate TCR-TEM, but effects differ between TEM and cytokine production and among some T cell types. Blocking EC LFA-3 interactions with TEM CD2 boosts TEM but reduces cytokine production. Blocking EC ICOS-L interactions with TEM CD28 (but not ICOS) reduces both responses but these involve distinct CD28-induced signals. Activated CD4+ T effector cells no longer undergo TCR-TEM. Engagement of T cell CD28 by EC ICOS-L increases TCR-TEM by activated CD8 effectors while engagement of OX40 promotes TCR-TEM by activated CD4 T regs. B7-H3 mostly affects TEM of resting TEM and some checkpoint inhibitors affect cytokine synthesis or TEM depending upon subtype. Our data suggest that blockade or mimicry of costimulators/checkpoint inhibitors in vivo, clinically used to modulate immune responses, may act in part by modulating T cell homing.


Asunto(s)
Antígenos CD28 , Células Endoteliales , Humanos , Células Endoteliales/metabolismo , Memoria Inmunológica , Receptores de Antígenos de Linfocitos T/metabolismo , Superantígenos , Interleucina-2 , Antígenos CD58
6.
JCI Insight ; 4(20)2019 10 17.
Artículo en Inglés | MEDLINE | ID: mdl-31527312

RESUMEN

Tissue engineering may address organ shortages currently limiting clinical transplantation. Off-the-shelf engineered vascularized organs will likely use allogeneic endothelial cells (ECs) to construct microvessels required for graft perfusion. Vasculogenic ECs can be differentiated from committed progenitors (human endothelial colony-forming cells or HECFCs) without risk of mutation or teratoma formation associated with reprogrammed stem cells. Like other ECs, these cells can express both class I and class II major histocompatibility complex (MHC) molecules, bind donor-specific antibody (DSA), activate alloreactive T effector memory cells, and initiate rejection in the absence of donor leukocytes. CRISPR/Cas9-mediated dual ablation of ß2-microglobulin and class II transactivator (CIITA) in HECFC-derived ECs eliminates both class I and II MHC expression while retaining EC functions and vasculogenic potential. Importantly, dually ablated ECs no longer bind human DSA or activate allogeneic CD4+ effector memory T cells and are resistant to killing by CD8+ alloreactive cytotoxic T lymphocytes in vitro and in vivo. Despite absent class I MHC molecules, these ECs do not activate or elicit cytotoxic activity from allogeneic natural killer cells. These data suggest that HECFC-derived ECs lacking MHC molecule expression can be utilized for engineering vascularized grafts that evade allorejection.


Asunto(s)
Aloinjertos/inmunología , Células Endoteliales/inmunología , Rechazo de Injerto/prevención & control , Proteínas Nucleares/genética , Ingeniería de Tejidos/métodos , Transactivadores/genética , Microglobulina beta-2/genética , Aloinjertos/irrigación sanguínea , Aloinjertos/provisión & distribución , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Sistemas CRISPR-Cas/genética , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Progenitoras Endoteliales , Femenino , Sangre Fetal/citología , Técnicas de Inactivación de Genes , Rechazo de Injerto/sangre , Rechazo de Injerto/inmunología , Voluntarios Sanos , Humanos , Isoanticuerpos/inmunología , Células Asesinas Naturales/inmunología , Activación de Linfocitos/genética , Ratones , Microvasos/citología , Microvasos/inmunología , Microvasos/trasplante , Proteínas Nucleares/inmunología , Trasplante de Órganos/efectos adversos , Trasplante de Órganos/métodos , Cultivo Primario de Células , Transactivadores/inmunología , Microglobulina beta-2/inmunología
7.
Nat Commun ; 10(1): 2247, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31113953

RESUMEN

Complement promotes vascular inflammation in transplant organ rejection and connective tissue diseases. Here we identify ZFYVE21 as a complement-induced Rab5 effector that induces non-canonical NF-κB in endothelial cells (EC). In response to membrane attack complexes (MAC), ZFYVE21 is post-translationally stabilized on MAC+Rab5+ endosomes in a Rab5- and PI(3)P-dependent manner. ZFYVE21 promotes SMURF2-mediated polyubiquitinylation and proteasome-dependent degradation of endosome-associated PTEN to induce vesicular enrichment of PI(3,4,5)P3 and sequential recruitment of activated Akt and NF-κB-inducing kinase (NIK). Pharmacologic alteration of cellular phosphoinositide content with miltefosine reduces ZFYVE21 induction, EC activation, and allograft vasculopathy in a humanized mouse model. ZFYVE21 induction distinctly occurs in response to MAC and is detected in human renal and synovial tissues. Our data identifies ZFYVE21 as a Rab5 effector, defines a Rab5-ZFYVE21-SMURF2-pAkt axis by which it mediates EC activation, and demonstrates a role for this pathway in complement-mediated conditions.


Asunto(s)
Proteínas Portadoras/metabolismo , Endosomas/metabolismo , Rechazo de Injerto/patología , FN-kappa B/metabolismo , Vasculitis/patología , Aloinjertos/patología , Animales , Línea Celular , Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Vasos Coronarios/patología , Vasos Coronarios/trasplante , Modelos Animales de Enfermedad , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Péptidos y Proteínas de Señalización Intracelular , Proteínas de la Membrana , Ratones , Ratones SCID , Fosfatos de Fosfatidilinositol/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas de Unión al GTP rab5/metabolismo
8.
J Immunol ; 201(11): 3167-3174, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30341183

RESUMEN

Early human allograft rejection can be initiated when circulating human host versus graft Ag-specific CD8 and CD4 effector memory T cells directly recognize MHC class I and II, respectively, expressed on the luminal surface by endothelium lining graft blood vessels. TCR engagement triggers both graft entry (TCR-driven transendothelial migration or TEM) and production of proinflammatory cytokines. Both TCR-driven TEM and cytokine expression are known to depend on T cell enzymes, myosin L chain kinase, and calcineurin, respectively, that are activated by cytoplasmic calcium and calmodulin, but whether the sources of calcium that control these enzymes are the same or different is unknown. Using superantigen or anti-CD3 Ab presented by cultured human dermal microvascular cells to freshly isolated peripheral blood human effector memory T cells under conditions of flow (models of alloantigen recognition in a vascularized graft), we tested the effects of pharmacological inhibitors of TCR-activated calcium signaling pathways on TCR-driven TEM and cytokine expression. We report that extracellular calcium entry via CRAC channels is the dominant contributor to cytokine expression, but paradoxically these same inhibitors potentiate TEM. Instead, calcium entry via TRPV1, L-Type Cav, and pannexin-1/P2X receptors appear to control TCR-driven TEM. These data reveal new therapeutic targets for immunosuppression.


Asunto(s)
Canales de Calcio Activados por la Liberación de Calcio/metabolismo , Señalización del Calcio , Células Endoteliales/inmunología , Enfermedad Injerto contra Huésped/inmunología , Linfocitos T/inmunología , Movimiento Celular , Células Cultivadas , Citocinas/metabolismo , Humanos , Memoria Inmunológica , Terapia de Inmunosupresión , Mediadores de Inflamación/metabolismo , Isoantígenos/inmunología , Activación de Linfocitos , Terapia Molecular Dirigida , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Migración Transendotelial y Transepitelial
9.
JCI Insight ; 3(5)2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29515027

RESUMEN

Early acute rejection of human allografts is mediated by circulating alloreactive host effector memory T cells (TEM). TEM infiltration typically occurs across graft postcapillary venules and involves sequential interactions with graft-derived endothelial cells (ECs) and pericytes (PCs). While the role of ECs in allograft rejection has been extensively studied, contributions of PCs to this process are largely unknown. This study aimed to characterize the effects and mechanisms of interactions between human PCs and allogeneic TEM. We report that unstimulated PCs, like ECs, can directly present alloantigen to TEM, but while IFN-γ-activated ECs (γ-ECs) show increased ability to stimulate alloreactive T cells, IFN-γ-activated PCs (γ-PCs) instead suppress TEM proliferation but not cytokine production or signaling. RNA sequencing analysis of PCs, γ-PCs, ECs, and γ-ECs reveal induction of indoleamine 2,3-dioxygenase 1 (IDO1) in γ-PCs to significantly higher levels than in γ-ECs that correlates with tryptophan depletion in vitro. Consistently, shRNA knockdown of IDO1 markedly reduces γ-PC-mediated immunoregulatory effects. Furthermore, human PCs express IDO1 in a skin allograft rejection humanized mouse model and in human renal allografts with acute T cell-mediated rejection. We conclude that immunosuppressive properties of human PCs are not intrinsic but instead result from IFN-γ-induced IDO1-mediated tryptophan depletion.


Asunto(s)
Aloinjertos/inmunología , Rechazo de Injerto/inmunología , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón gamma/metabolismo , Pericitos/inmunología , Aloinjertos/irrigación sanguínea , Aloinjertos/citología , Animales , Presentación de Antígeno/inmunología , Comunicación Celular/inmunología , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/inmunología , Endotelio Vascular/citología , Femenino , Voluntarios Sanos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interferón gamma/inmunología , Isoantígenos/inmunología , Ratones SCID , Microvasos/citología , Microvasos/inmunología , Pericitos/metabolismo , Cultivo Primario de Células , ARN Interferente Pequeño/metabolismo , Piel/irrigación sanguínea , Piel/citología , Piel/inmunología , Trasplante de Piel/efectos adversos , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo , Quimera por Trasplante , Trasplante Homólogo/efectos adversos , Triptófano/metabolismo
10.
Front Immunol ; 8: 1907, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29312357

RESUMEN

Antigen presentation by cells of the vessel wall may initiate rapid and localized memory immune responses in peripheral tissues. Peptide antigens displayed on major histocompatibility complex (MHC) molecules on the surface of endothelial cells (ECs) can be recognized by T cell receptors on circulating effector memory T cells (TEM), triggering both transendothelial migration and activation. The array of co-stimulatory receptors, adhesion molecules, and cytokines expressed by ECs serves to modulate T cell activation responses. While the effects of these interactions vary among species, vascular beds, and vascular segments within the same tissue, they are capable of triggering allograft rejection without direct involvement of professional antigen-presenting cells and may play a similar role in host defense against infections and in autoimmunity. Once across the endothelium, extravasating TEM then contact mural cells of the vessel wall, including pericytes or vascular smooth muscle cells, which may also present antigens and provide signals that further regulate T cell responses. Collectively, these interactions provide an unexplored opportunity in which targeting of vascular cells can be used to modulate immune responses. In organ transplantation, targeting ECs with siRNA to reduce expression of MHC molecules may additionally mitigate perioperative injuries by preformed alloantibodies, further reducing the risk of graft rejection. Similarly, genetic manipulation of vascular cells to minimize antigen-dependent responses can be used to increase perfusion of tissue engineered organs without triggering rejection.

11.
Arterioscler Thromb Vasc Biol ; 36(9): 1910-8, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27444200

RESUMEN

OBJECTIVE: Circulating human T effector memory cell (TEM) recognition of nonself MHC (major histocompatibility complex) molecules on allograft endothelial cells can initiate graft rejection despite elimination of professional antigen-presenting cells necessary for naive T-cell activation. Our previous studies of CD4 TEM have established that engagement of the T-cell receptor not only activates T cells but also triggers transendothelial migration (TEM) by a process that is distinct from that induced by activating chemokine receptors on T cells, being slower, requiring microtubule-organizing center-directed cytolytic granule polarization to and release from the leading edge of the T cell, and requiring engagement of proteins of the endothelial cell lateral border recycling compartment. Although CD4 TEM may contribute to acute allograft rejection, the primary effectors are alloreactive CD8 TEM. Whether and how T-cell receptor engagement affects TEM of human CD8 TEM is unknown. APPROACH AND RESULTS: We modeled TEM of CD8 TEM across cultured human microvascular endothelial cells engineered to present superantigen under conditions of venular shear stress in vitro in a flow chamber. Here, we report that T-cell receptor engagement can also induce TEM of this population that similarly differs from chemokine receptor-driven TEM with regard to kinetics, morphological manifestations, and microtubule-organizing center dynamics as with CD4 TEM. However, CD8 TEM do not require either cytolytic granule release or interactions with proteins of the lateral border recycling compartment. CONCLUSIONS: These results imply that therapeutic strategies designed to inhibit T-cell receptor-driven recruitment based on targeting granule release or components of the lateral border recycling compartment will not affect CD8 TEM and are unlikely to block acute rejection in the clinic.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Quimiotaxis de Leucocito , Células Endoteliales/inmunología , Rechazo de Injerto/inmunología , Memoria Inmunológica , Proteínas Nucleares/inmunología , Superantígenos/inmunología , Transactivadores/inmunología , Migración Transendotelial y Transepitelial , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Degranulación de la Célula , Membrana Celular/inmunología , Membrana Celular/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Gránulos Citoplasmáticos/inmunología , Gránulos Citoplasmáticos/metabolismo , Células Endoteliales/metabolismo , Rechazo de Injerto/metabolismo , Rechazo de Injerto/prevención & control , Humanos , Cinética , Centro Organizador de los Microtúbulos/inmunología , Centro Organizador de los Microtúbulos/metabolismo , Proteínas Nucleares/biosíntesis , Proteínas Nucleares/genética , Fenotipo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal , Superantígenos/biosíntesis , Superantígenos/genética , Transactivadores/biosíntesis , Transactivadores/genética , Transfección , Trasplante Homólogo
12.
Proc Natl Acad Sci U S A ; 112(31): 9686-91, 2015 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-26195760

RESUMEN

Complement membrane attack complexes (MACs) promote inflammatory functions in endothelial cells (ECs) by stabilizing NF-κB-inducing kinase (NIK) and activating noncanonical NF-κB signaling. Here we report a novel endosome-based signaling complex induced by MACs to stabilize NIK. We found that, in contrast to cytokine-mediated activation, NIK stabilization by MACs did not involve cIAP2 or TRAF3. Informed by a genome-wide siRNA screen, instead this response required internalization of MACs in a clathrin-, AP2-, and dynamin-dependent manner into Rab5(+)endosomes, which recruited activated Akt, stabilized NIK, and led to phosphorylation of IκB kinase (IKK)-α. Active Rab5 was required for recruitment of activated Akt to MAC(+) endosomes, but not for MAC internalization or for Akt activation. Consistent with these in vitro observations, MAC internalization occurred in human coronary ECs in vivo and was similarly required for NIK stabilization and EC activation. We conclude that MACs activate noncanonical NF-κB by forming a novel Akt(+)NIK(+) signalosome on Rab5(+) endosomes.


Asunto(s)
Complejo de Ataque a Membrana del Sistema Complemento/metabolismo , Endosomas/metabolismo , FN-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Proteínas de Unión al GTP rab5/metabolismo , Animales , Proteína 3 que Contiene Repeticiones IAP de Baculovirus , Clatrina/metabolismo , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Endocitosis/efectos de los fármacos , Endosomas/efectos de los fármacos , Estabilidad de Enzimas/efectos de los fármacos , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana/efectos de los fármacos , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Hidrazonas/farmacología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Ratones SCID , Biosíntesis de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Vesículas Secretoras/efectos de los fármacos , Vesículas Secretoras/metabolismo , Transducción de Señal/efectos de los fármacos , Factor 3 Asociado a Receptor de TNF/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Quinasa de Factor Nuclear kappa B
13.
Biochem J ; 466(3): 525-36, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25558779

RESUMEN

Transforming growth factor (TGF) ß1 activity depends on a complex signalling cascade that controls expression of several genes. Among others, TGFß1 regulates expression of matrix metalloproteinases (MMPs) through activation of Smads. In the present study, we demonstrate for the first time that the αvß6 integrin interacts with TGFß receptor II (TßRII) through the ß6 cytoplasmic domain and promotes Smad3 activation in prostate cancer (PrCa) cells. Another related αv integrin, αvß5, as well as the αvß6/3 integrin, which contains a chimeric form of ß6 with a ß3 cytoplasmic domain, do not associate with TßRII and fail to show similar responses. We provide evidence that αvß6 is required for up-regulation of MMP2 by TGFß1 through a Smad3-mediated transcriptional programme in PrCa cells. The functional relevance of these results is underscored by the finding that αvß6 modulates cell migration in an MMP2-dependent manner on an αvß6-specific ligand, latency-associated peptide (LAP)-TGFß. Overall, these mechanistic studies establish that expression of a single integrin, αvß6, is sufficient to promote activation of Smad3, regulation of MMP2 levels and consequent catalytic activity, as well as cell migration. Our study describes a new TGFß1-αvß6-MMP2 signalling pathway that, given TGFß1 pro-metastatic activity, may have profound implications for PrCa therapy.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Regulación Enzimológica de la Expresión Génica , Integrinas/metabolismo , Metaloproteinasa 2 de la Matriz/biosíntesis , Factor de Crecimiento Transformador beta1/farmacología , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/fisiología , Humanos , Masculino
14.
J Immunol ; 193(12): 5809-15, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25367116

RESUMEN

Human effector memory CD4 T cells may transmigrate across endothelial cell (EC) monolayers either in response to inflammatory chemokines or in response to TCR recognition of Ag presented on the surface of the EC. The kinetics, morphologic manifestations, and molecular requirements of chemokine- and TCR-driven transendothelial migration (TEM) differ significantly. In this study, we report that, whereas the microtubule organizing center (MTOC) and cytosolic granules follow the nucleus across the endothelium in a uropod during chemokine-driven TEM, MTOC reorientation to the contact region between the T cell and the EC, accompanied by dynein-driven transport of granzyme-containing granules to and exocytosis at the contact region, are early events in TCR-driven, but not chemokine-driven TEM. Inhibitors of either granule function or granzyme proteolytic activity can arrest TCR-driven TEM, implying a requirement for granule discharge in the process. In the final stages of TCR-driven TEM, the MTOC precedes, rather than follows, the nucleus across the endothelium. Thus, TCR-driven TEM of effector memory CD4 T cells appears to be a novel process that more closely resembles immune synapse formation than it does conventional chemotaxis.


Asunto(s)
Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Granzimas/metabolismo , Migración Transendotelial y Transepitelial/inmunología , Quimiocinas/metabolismo , Gránulos Citoplasmáticos/metabolismo , Exocitosis/inmunología , Humanos , Microtúbulos/metabolismo , Transporte de Proteínas , Receptores de Antígenos de Linfocitos T/metabolismo
15.
J Exp Med ; 211(3): 395-404, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24516119

RESUMEN

Recruitment of circulating leukocytes into inflamed tissues depends on adhesion molecules expressed by endothelial cells (ECs). Here we report that rapamycin pretreatment reduced the ability of TNF-treated ECs to capture T cells under conditions of venular flow. This functional change was caused by inhibition of TNF-induced expression of vascular cell adhesion molecule-1 (VCAM-1) and could be mimicked by knockdown of mammalian target of rapamycin (mTOR) or rictor, but not raptor, implicating mTORC2 as the target of rapamycin for this effect. Mechanistically, mTORC2 acts through Akt to repress Raf1-MEK1/2-ERK1/2 signaling, and inhibition of mTORC2 consequently results in hyperactivation of ERK1/2. Increased ERK1/2 activity antagonizes VCAM-1 expression by repressing TNF induction of the transcription factor IRF-1. Preventing activation of ERK1/2 reduced the ability of rapamycin to inhibit TNF-induced VCAM-1 expression. In vivo, rapamycin inhibited mTORC2 activity and potentiated activation of ERK1/2. These changes correlated with reduced endothelial expression of TNF-induced VCAM-1, which was restored via pharmacological inhibition of ERK1/2. Functionally, rapamycin reduced infiltration of leukocytes into renal glomeruli, an effect which was partially reversed by inhibition of ERK1/2. These data demonstrate a novel mechanism by which rapamycin modulates the ability of vascular endothelium to mediate inflammation and identifies endothelial mTORC2 as a potential therapeutic target.


Asunto(s)
Células Endoteliales/metabolismo , Complejos Multiproteicos/antagonistas & inhibidores , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Molécula 1 de Adhesión Celular Vascular/metabolismo , Análisis de Varianza , Western Blotting , Adhesión Celular/inmunología , Inmunoprecipitación de Cromatina , Cartilla de ADN/genética , Citometría de Flujo , Humanos , Immunoblotting , Diana Mecanicista del Complejo 2 de la Rapamicina , Microscopía Fluorescente , Complejos Multiproteicos/metabolismo , Proteína Oncogénica v-akt/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Linfocitos T/inmunología , Serina-Treonina Quinasas TOR/metabolismo
16.
Circulation ; 128(23): 2504-16, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24045046

RESUMEN

BACKGROUND: Cardiac allograft vasculopathy is the major cause of late allograft loss after heart transplantation. Cardiac allograft vasculopathy lesions contain alloreactive T cells that secrete interferon-γ, a vasculopathic cytokine, and occur more frequently in patients with donor-specific antibody. Pathological interactions between these immune effectors, representing cellular and humoral immunity, respectively, remain largely unexplored. METHODS AND RESULTS: We used human panel reactive antibody to form membrane attack complexes on allogeneic endothelial cells in vitro and in vivo. Rather than inducing cytolysis, membrane attack complexes upregulated inflammatory genes, enhancing the capacity of endothelial cells to recruit and activate allogeneic interferon-γ--producing CD4(+) T cells in a manner dependent on the activation of noncanonical nuclear factor-κB signaling. Noncanonical nuclear factor-κB signaling was detected in situ within endothelial cells both in renal biopsies from transplantation patients with chronic antibody-mediated rejection and in panel-reactive antibody--treated human coronary artery xenografts in immunodeficient mice. On retransplantation into immunodeficient hosts engrafted with human T cells, panel-reactive antibody--treated grafts recruited more interferon-γ--producing T cells and enhanced cardiac allograft vasculopathy lesion formation. CONCLUSIONS: Alloantibody and complement deposition on graft endothelial cells activates noncanonical nuclear factor-κB signaling, initiating a proinflammatory gene program that enhances alloreactive T cell activation and development of cardiac allograft vasculopathy. Noncanonical nuclear factor-κB signaling in endothelial cells, observed in human allograft specimens and implicated in lesion pathogenesis, may represent a target for new pharmacotherapies to halt the progression of cardiac allograft vasculopathy.


Asunto(s)
Proteínas del Sistema Complemento/fisiología , Vasos Coronarios/inmunología , Células Endoteliales/metabolismo , Isoanticuerpos/fisiología , FN-kappa B/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Aloinjertos/inmunología , Aloinjertos/patología , Aloinjertos/fisiopatología , Animales , Células Cultivadas , Vasos Coronarios/patología , Vasos Coronarios/trasplante , Células Endoteliales/inmunología , Células Endoteliales/patología , Femenino , Xenoinjertos/inmunología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Isoanticuerpos/sangre , Ratones , Ratones SCID , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/patología
17.
J Immunol ; 190(7): 3079-88, 2013 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-23420881

RESUMEN

Human effector memory (EM) CD4 T cells may be recruited from the blood into a site of inflammation in response either to inflammatory chemokines displayed on or specific Ag presented by venular endothelial cells (ECs), designated as chemokine-driven or TCR-driven transendothelial migration (TEM), respectively. We have previously described differences in the morphological appearance of transmigrating T cells as well as in the molecules that mediate T cell-EC interactions distinguishing these two pathways. In this study, we report that TCR-driven TEM requires ZAP-70-dependent activation of a pathway involving Vav, Rac, and myosin IIA. Chemokine-driven TEM also uses ZAP-70, albeit in a quantitatively and spatially different manner of activation, and is independent of Vav, Rac, and mysosin IIA, depending instead on an as-yet unidentified GTP exchange factor that activates Cdc42. The differential use of small Rho family GTPases to activate the cytoskeleton is consistent with the morphological differences observed in T cells that undergo TEM in response to these distinct recruitment signals.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Miosina Tipo IIA no Muscular/metabolismo , Proteínas Proto-Oncogénicas c-vav/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Migración Transendotelial y Transepitelial/inmunología , Proteínas de Unión al GTP rac/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Proteínas de Unión al GTP/metabolismo , Humanos , Memoria Inmunológica , Proteínas de Neoplasias/metabolismo , Receptores de Cinasa C Activada , Receptores de Superficie Celular/metabolismo , Proteína Tirosina Quinasa ZAP-70/metabolismo
18.
Circulation ; 124(2): 196-205, 2011 Jul 12.
Artículo en Inglés | MEDLINE | ID: mdl-21690493

RESUMEN

BACKGROUND: Ligands activating the transcription factor peroxisome proliferator-activated receptor-γ (PPARγ) have antiinflammatory effects. Vascular rejection induced by allogeneic T cells can be responsible for acute and chronic graft loss. Studies in rodents suggest that PPARγ agonists may inhibit graft vascular rejection, but human T-cell responses to allogeneic vascular cells differ from those in rodents, and the effects of PPARγ in human transplantation are unknown. METHODS AND RESULTS: We tested the effects of PPARγ agonists on human vascular graft rejection using a model in which human artery is interposed into the abdominal aorta of immunodeficient mice, followed by adoptive transfer of allogeneic (to the artery donor) human peripheral blood mononuclear cells. Interferon-γ-dependent rejection ensues within 4 weeks, characterized by intimal thickening, T-cell infiltrates, and vascular cell activation, a response resembling clinical intimal arteritis. The PPARγ agonists 15-deoxy-prostaglandin-J(2), ciglitazone, and pioglitazone reduced intimal expansion, intimal infiltration of CD45RO(+) memory T cells, and plasma levels of inflammatory cytokines. The PPARγ antagonist GW9662 reversed the protective effects of PPARγ agonists, confirming the involvement of PPARγ-mediated pathways. In vitro, pioglitazone inhibited both alloantigen-induced proliferation and superantigen-induced transendothelial migration of memory T cells, indicating the potential mechanisms of PPARγ effects. CONCLUSION: Our results suggest that PPARγ agonists inhibit allogeneic human memory T cell responses and may be useful for the treatment of vascular graft rejection.


Asunto(s)
Arterias/inmunología , Arterias/trasplante , Rechazo de Injerto/inmunología , Hipoglucemiantes/farmacología , PPAR gamma/agonistas , Prostaglandina D2/análogos & derivados , Linfocitos T/inmunología , Tiazolidinedionas/farmacología , Traslado Adoptivo , Anilidas/farmacología , Animales , Arterias/patología , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Proliferación Celular/efectos de los fármacos , Citocinas/inmunología , Rechazo de Injerto/tratamiento farmacológico , Rechazo de Injerto/patología , Humanos , Memoria Inmunológica/efectos de los fármacos , Memoria Inmunológica/inmunología , Isoantígenos/inmunología , Ratones , Ratones SCID , PPAR gamma/antagonistas & inhibidores , PPAR gamma/inmunología , Pioglitazona , Prostaglandina D2/farmacología , Superantígenos/farmacología , Linfocitos T/patología , Linfocitos T/trasplante , Trasplante Heterólogo , Trasplante Homólogo
19.
Blood ; 117(7): 2284-95, 2011 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-21183689

RESUMEN

The reticulon (Rtn) family of proteins are localized primarily to the endoplasmic reticulum (ER) of most cells. The Rtn-4 family, (aka Nogo) consists of 3 splice variants of a common gene called Rtn-4A, Rtn-4B, and Rtn-4C. Recently, we identified the Rtn-4B (Nogo-B) protein in endothelial and smooth muscle cells of the vessel wall, and showed that Nogo-B is a regulator of cell migration in vitro and vascular remodeling and angiogenesis in vivo. However, the role of Nogo-B in inflammation is still largely unknown. In the present study, we use 2 models of inflammation to show that endothelial Nogo-B regulates leukocyte transmigration and intercellular adhesion molecule-1 (ICAM-1)-dependent signaling. Mice lacking Nogo-A/B have a marked reduction in neutrophil and monocyte recruitment to sites of inflammation, while Nogo-A/B(-/-) mice engrafted with wild-type (WT) bone marrow still exhibit impaired inflammation compared with WT mice engrafted with Nogo-A/B(-/-) bone marrow, arguing for a critical role of host Nogo in this response. Using human leukocytes and endothelial cells, we show mechanistically that the silencing of Nogo-B with small interfering RNA (siRNA) impairs the transmigration of neutrophils and reduces ICAM-1-stimulated phosphorylation of vascular endothelial-cell cadherin (VE-cadherin). Our results reveal a novel role of endothelial Nogo-B in basic immune functions and provide a key link in the molecular network governing endothelial-cell regulation of diapedesis.


Asunto(s)
Inflamación/etiología , Molécula 1 de Adhesión Intercelular/fisiología , Leucocitos/fisiología , Proteínas de la Mielina/fisiología , Animales , Antígenos CD/fisiología , Cadherinas/fisiología , Carragenina/toxicidad , Movimiento Celular/fisiología , Células Endoteliales/patología , Células Endoteliales/fisiología , Quinasa 2 de Adhesión Focal/metabolismo , Humanos , Técnicas In Vitro , Inflamación/patología , Inflamación/fisiopatología , Leucocitos/patología , Macrófagos/patología , Macrófagos/fisiología , Masculino , Ratones , Ratones Congénicos , Ratones Noqueados , Monocitos/patología , Monocitos/fisiología , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas de la Mielina/deficiencia , Proteínas de la Mielina/genética , Neutrófilos/patología , Neutrófilos/fisiología , Proteínas Nogo , Fosforilación , ARN Interferente Pequeño/genética , Transducción de Señal , Familia-src Quinasas/metabolismo
20.
J Immunol ; 186(3): 1763-8, 2011 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21191062

RESUMEN

Human effector memory (EM) CD4(+) T cells can rapidly transmigrate across an endothelial cell (EC) monolayer in response either to chemokine or to TCR-activating signals displayed by human dermal microvascular EC under conditions of venular shear stress. We previously reported that the TCR-stimulated transendothelial migration (TEM) depends on fractalkine (CX3CL1), PECAM-1 (CD31), and ICAM-1 (CD54) expression by the EC, whereas chemokine-stimulated TEM does not. In this study, we further analyze these responses using blocking mAb and small interfering RNA knockdown to show that TCR-stimulated TEM depends on CD99 on EC as well as on PECAM-1 and depends on nectin-2 (CD112) and poliovirus receptor (CD155) as well as EC ICAM-1. ICAM-1 is engaged by EM CD4(+) T cell LFA-1 (CD11a/CD18) but not Mac-1 (CD11b/CD18); nectin-2 and poliovirus receptor are engaged by both DNAX accessory molecule-1 (CD226) and Tactile (CD96). EC junctional adhesion molecule-1 (JAM-1), an alternative ligand for LFA-1, contributes exclusively to chemokine-stimulated TEM and ICAM-2 appears to be uninvolved in either pathway. These data further define and further highlight the differences in the two pathways of EM CD4(+) T cell recruitment into sites of peripheral inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Moléculas de Adhesión Celular/fisiología , Comunicación Celular/inmunología , Movimiento Celular/inmunología , Endotelio Vascular/inmunología , Endotelio Vascular/metabolismo , Memoria Inmunológica , Receptores de Antígenos de Linfocitos T/fisiología , Antígeno 12E7 , Anticuerpos Bloqueadores/farmacología , Antígenos CD/biosíntesis , Antígenos CD/metabolismo , Antígenos CD/fisiología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Moléculas de Adhesión Celular/antagonistas & inhibidores , Moléculas de Adhesión Celular/biosíntesis , Moléculas de Adhesión Celular/inmunología , Moléculas de Adhesión Celular/metabolismo , Células Cultivadas , Quimiocinas/metabolismo , Quimiocinas/fisiología , Endotelio Vascular/citología , Humanos , Moléculas de Adhesión de Unión , Antígeno-1 Asociado a Función de Linfocito/biosíntesis , Antígeno-1 Asociado a Función de Linfocito/metabolismo , Antígeno-1 Asociado a Función de Linfocito/fisiología , Nectinas , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores Virales/fisiología , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA