Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 15(1): 4271, 2024 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-38769289

RESUMEN

T Cell Receptor (TCR) antigen binding underlies a key mechanism of the adaptive immune response yet the vast diversity of TCRs and the complexity of protein interactions limits our ability to build useful low dimensional representations of TCRs. To address the current limitations in TCR analysis we develop a capacity-controlled disentangling variational autoencoder trained using a dataset of approximately 100 million TCR sequences, that we name TCR-VALID. We design TCR-VALID such that the model representations are low-dimensional, continuous, disentangled, and sufficiently informative to provide high-quality TCR sequence de novo generation. We thoroughly quantify these properties of the representations, providing a framework for future protein representation learning in low dimensions. The continuity of TCR-VALID representations allows fast and accurate TCR clustering and is benchmarked against other state-of-the-art TCR clustering tools and pre-trained language models.


Asunto(s)
Receptores de Antígenos de Linfocitos T , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Humanos , Aprendizaje Profundo , Algoritmos , Análisis por Conglomerados , Biología Computacional/métodos , Secuencia de Aminoácidos
2.
Oncoimmunology ; 9(1): 1758602, 2020 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-32923116

RESUMEN

Clinical observations suggest that responses to cancer immunotherapy are correlated with intra-tumoral T cell receptor (TCR) clonality, tumor mutation burden (TMB) and host HLA genotype, highlighting the importance of host T cell recognition of tumor antigens. However, the dynamic interplay between T cell activation state and changes in TCR repertoire in driving the identification of potential immunodominant antigen(s) remains largely unexplored. Here, we performed single-cell RNA-sequencing on CD8+ tumor-infiltrating T cells (TILs) using the murine colorectal tumor model MC38 to identify unique TCR sequences and validate their tumor reactivity. We found that the majority of clonally expanded TILs are tumor-reactive and their TCR repertoire is unique amongst individual MC38 tumor-bearing mice. Our query identified that multiple expanded TCR clones recognized the retroviral epitope p15E as an immunodominant antigen. In addition, we found that the endogenous retroviral genome encoding for p15E is highly expressed in MC38 tumors, but not in normal tissues, due to epigenetic derepression. Further, we demonstrated that the p15E-specific TILs exhibit an activated phenotype and an increase in frequency upon treatment with anti-41BB and anti-PD-1 combination immunotherapy. Importantly, we showed that although p15E-specific TILs are not required to mount a primary anti-tumor response, they contributed to the development of strong immune memory. Overall our results revealed that endogenous retroviral antigens expressed by tumor cells may represent an important and underappreciated category of tumor antigens that could be readily targeted in the clinic.


Asunto(s)
Retrovirus Endógenos , Neoplasias , Animales , Inmunoterapia , Activación de Linfocitos , Ratones , Neoplasias/terapia , Receptores de Antígenos de Linfocitos T/genética
3.
Sci Transl Med ; 12(549)2020 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-32581132

RESUMEN

Monoclonal antibodies that block the programmed cell death 1 (PD-1) checkpoint have revolutionized cancer immunotherapy. However, many major tumor types remain unresponsive to anti-PD-1 therapy, and even among responsive tumor types, most of the patients do not develop durable antitumor immunity. It has been shown that bispecific antibodies activate T cells by cross-linking the TCR/CD3 complex with a tumor-specific antigen (TSA). The class of TSAxCD3 bispecific antibodies have generated exciting results in early clinical trials. We have recently described another class of "costimulatory bispecifics" that cross-link a TSA to CD28 (TSAxCD28) and cooperate with TSAxCD3 bispecifics. Here, we demonstrate that these TSAxCD28 bispecifics (one specific for prostate cancer and the other for epithelial tumors) can also synergize with the broader anti-PD-1 approach and endow responsiveness-as well as long-term immune memory-against tumors that otherwise do not respond to anti-PD-1 alone. Unlike CD28 superagonists, which broadly activate T cells and induce cytokine storm, TSAxCD28 bispecifics display little or no toxicity when used alone or in combination with a PD-1 blocker in genetically humanized immunocompetent mouse models or in primates and thus may provide a well-tolerated and "off the shelf" combination approach with PD-1 immunotherapy that can markedly enhance antitumor efficacy.


Asunto(s)
Anticuerpos Biespecíficos , Neoplasias , Animales , Anticuerpos Biespecíficos/uso terapéutico , Antígenos CD28 , Humanos , Inmunoterapia , Ratones , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1
4.
Sci Transl Med ; 12(525)2020 01 08.
Artículo en Inglés | MEDLINE | ID: mdl-31915305

RESUMEN

T cell activation is initiated upon binding of the T cell receptor (TCR)/CD3 complex to peptide-major histocompatibility complexes ("signal 1"); activation is enhanced by engagement of a second "costimulatory" receptor, such as the CD28 receptor on T cells binding to its cognate ligand(s) on the target cell ("signal 2"). CD3-based bispecific antibodies act by replacing conventional signal 1, linking T cells to tumor cells by binding a tumor-specific antigen (TSA) with one arm of the bispecific and bridging to TCR/CD3 with the other. Although some of these so-called TSAxCD3 bispecifics have demonstrated promising antitumor efficacy in patients with cancer, their activity remains to be optimized. Here, we introduce a class of bispecific antibodies that mimic signal 2 by bridging TSA to the costimulatory CD28 receptor on T cells. We term these TSAxCD28 bispecifics and describe two such bispecific antibodies: one specific for ovarian and the other for prostate cancer antigens. Unlike CD28 superagonists, which broadly activate T cells and resulted in profound toxicity in early clinical trials, these TSAxCD28 bispecifics show limited activity and no toxicity when used alone in genetically humanized immunocompetent mouse models or in primates. However, when combined with TSAxCD3 bispecifics, they enhance the artificial synapse between a T cell and its target cell, potentiate T cell activation, and markedly improve antitumor activity of CD3 bispecifics in a variety of xenogeneic and syngeneic tumor models. Combining this class of CD28-costimulatory bispecific antibodies with the emerging class of TSAxCD3 bispecifics may provide well-tolerated, off-the-shelf antibody therapies with robust antitumor efficacy.


Asunto(s)
Anticuerpos Biespecíficos/inmunología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Neoplasias/inmunología , Animales , Antígenos de Neoplasias/inmunología , Línea Celular Tumoral , Proliferación Celular , Citocinas/metabolismo , Citotoxicidad Inmunológica , Femenino , Células HEK293 , Humanos , Sinapsis Inmunológicas/metabolismo , Activación de Linfocitos/inmunología , Macaca fascicularis , Ratones , Neoplasias/patología , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Nat Commun ; 6: 6840, 2015 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-25924227

RESUMEN

A central mechanism of tumour progression and metastasis involves the generation of an immunosuppressive 'macroenvironment' mediated in part through tumour-secreted factors. Here we demonstrate that upregulation of the Inhibitor of Differentiation 1 (Id1), in response to tumour-derived factors, such as TGFß, is responsible for the switch from dendritic cell (DC) differentiation to myeloid-derived suppressor cell expansion during tumour progression. Genetic inactivation of Id1 largely corrects the myeloid imbalance, whereas Id1 overexpression in the absence of tumour-derived factors re-creates it. Id1 overexpression leads to systemic immunosuppression by downregulation of key molecules involved in DC differentiation and suppression of CD8 T-cell proliferation, thus promoting primary tumour growth and metastatic progression. Furthermore, advanced melanoma patients have increased plasma TGFß levels and express higher levels of ID1 in myeloid peripheral blood cells. This study reveals a critical role for Id1 in suppressing the anti-tumour immune response during tumour progression and metastasis.


Asunto(s)
Proteína 1 Inhibidora de la Diferenciación/fisiología , Melanoma Experimental/inmunología , Células Mieloides/fisiología , Animales , Diferenciación Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Factores Reguladores del Interferón/metabolismo , Leucocitos Mononucleares/metabolismo , Melanoma Experimental/metabolismo , Ratones Endogámicos C57BL , Metástasis de la Neoplasia , Factor de Crecimiento Transformador beta/metabolismo
6.
Eur J Immunol ; 43(12): 3343-54, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23939929

RESUMEN

Entry of lymphocytes into secondary lymphoid organs (SLOs) involves intravascular arrest and intracellular calcium ion ([Ca(2+)]i) elevation. TCR activation triggers increased [Ca(2+)]i and can arrest T-cell motility in vitro. However, the requirement for [Ca(2+)]i elevation in arresting T cells in vivo has not been tested. Here, we have manipulated the Ca(2+) release-activated Ca(2+) (CRAC) channel pathway required for [Ca(2+)]i elevation in T cells through genetic deletion of stromal interaction molecule (STIM) 1 or by expression of a dominant-negative ORAI1 channel subunit (ORAI1-DN). Interestingly, the absence of CRAC did not interfere with homing of naïve CD4(+) T cells to SLOs and only moderately reduced crawling speeds in vivo. T cells expressing ORAI1-DN lacked TCR activation induced [Ca(2+)]i elevation, yet arrested motility similar to control T cells in vitro. In contrast, antigen-specific ORAI1-DN T cells had a twofold delayed onset of arrest following injection of OVA peptide in vivo. CRAC channel function is not required for homing to SLOs, but enhances spatiotemporal coordination of TCR signaling and motility arrest.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Canales de Calcio/inmunología , Calcio/inmunología , Glicoproteínas de Membrana/inmunología , Animales , Canales de Calcio/genética , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Proteína ORAI1 , Péptidos/inmunología , Péptidos/farmacología , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/inmunología , Molécula de Interacción Estromal 1
7.
Int J Inflam ; 2012: 819467, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22229105

RESUMEN

The proinflammatory activity of T helper 17 (Th17) cells can be beneficial to the host during infection. However, uncontrolled or inappropriate Th17 activation has been linked to several autoimmune and autoinflammatory pathologies. Indeed, preclinical and clinical data show that Th17 cells are associated with several autoimmune diseases such as arthritis, multiple sclerosis, psoriasis, and lupus. Furthermore, targeting the interleukin-17 (IL-17) pathway has attenuated disease severity in preclinical models of autoimmune diseases. Interestingly, a recent report brings to light a potential role for Th17 cells in the autoinflammatory disorder adult-onset Still's disease (AOSD). Whether Th17 cells are the cause or are directly involved in AOSD remains to be shown. In this paper, we discuss the biology of Th17 cells, their role in autoimmune disease development, and in AOSD in particular, as well as the growing interest of the pharmaceutical industry in their use as therapeutic targets.

8.
Eur J Immunol ; 41(8): 2207-16, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21598246

RESUMEN

The essential role of the Delta-like ligand 4 (Dll4)-Notch signaling pathway in T-lymphocyte development is well established. It has been shown that specific inactivation of Dll4 on thymic stromal cells during early post-natal development leads to a deregulation in T-cell differentiation. However, whether ongoing Dll4-Notch signaling is required for T-cell development in the adult thymus is unknown. The use of anti-Dll4 Abs allowed us to confirm and expand previous studies by examining the kinetics and the reversibility of Dll4-Notch signaling blockade in T-cell development in adult mice. We found that anti-Dll4 treatment reduced thymic cellularity after 7 days, as a consequence of a developmental delay in T-cell maturation at the pro-T-cell double negative 1 (CD4(-) CD8(-) c-kit(+) CD44(+) CD25(-) ) stage, leading to decreased numbers of immature double-positive (CD4(+) CD8(+) ) T cells without affecting the frequency of mature single positive CD4(+) and CD8(+) thymocytes, while promoting alternative thymic B-cell expansion. This cellular phenotype was similarly observed in both young adult and aged mice (>1.5 years), extending our understanding of the ongoing role for Dll4-Notch signaling during T-cell development in the adult thymus. Finally, after cessation of Dll4 Ab treatment, thymic cellularity and thymocyte subset ratios returned to normal levels, indicating reversibility of this phenotype in both adult and aged mice, which has important implications for potential clinical use of Dll4-Notch inhibitors.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/inmunología , Proteínas de la Membrana/inmunología , Receptores Notch/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Timo/inmunología , Proteínas Adaptadoras Transductoras de Señales , Factores de Edad , Animales , Anticuerpos/inmunología , Anticuerpos/farmacología , Atrofia/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Proteínas de Unión al Calcio , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Citometría de Flujo , Perfilación de la Expresión Génica , Homeostasis/inmunología , Humanos , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores Notch/genética , Receptores Notch/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Linfocitos T/metabolismo , Timo/metabolismo , Timo/patología , Factores de Tiempo
9.
PLoS Pathog ; 7(3): e1001326, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21455492

RESUMEN

Host defense against the intracellular pathogen Listeria monocytogenes (Lm) requires innate and adaptive immunity. Here, we directly imaged immune cell dynamics at Lm foci established by dendritic cells in the subcapsular red pulp (scDC) using intravital microscopy. Blood borne Lm rapidly associated with scDC. Myelomonocytic cells (MMC) swarmed around non-motile scDC forming foci from which blood flow was excluded. The depletion of scDC after foci were established resulted in a 10-fold reduction in viable Lm, while graded depletion of MMC resulted in 30-1000 fold increase in viable Lm in foci with enhanced blood flow. Effector CD8+ T cells at sites of infection displayed a two-tiered reduction in motility with antigen independent and antigen dependent components, including stable interactions with infected and non-infected scDC. Thus, swarming MMC contribute to control of Lm prior to development of T cell immunity by direct killing and sequestration from blood flow, while scDC appear to promote Lm survival while preferentially interacting with CD8+ T cells in effector sites.


Asunto(s)
Inmunidad Adaptativa , Inmunidad Innata , Listeria monocytogenes/patogenicidad , Listeriosis/patología , Linfocitos T Citotóxicos/patología , Animales , Citotoxicidad Inmunológica , Células Dendríticas/inmunología , Células Dendríticas/patología , Femenino , Técnicas de Sustitución del Gen , Interacciones Huésped-Patógeno , Listeriosis/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Monocitos/inmunología , Monocitos/patología , Linfocitos T Citotóxicos/inmunología
10.
J Immunol ; 181(7): 4852-63, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18802089

RESUMEN

Short-lived TCR microclusters and a longer-lived protein kinase Ctheta-focusing central supramolecular activation cluster (cSMAC) have been defined in model immunological synapses (IS). In different model systems, CD28-mediated costimulatory interactions have been detected in microclusters, the cSMAC, or segregated from the TCR forming multiple distinct foci. The relationship between TCR and costimulatory molecules in the physiological IS of T cell-dendritic cell (DC) is obscure. To study the dynamic relationship of CD28-CD80 and TCR interactions in the T cell-DC IS during Ag-specific T cell activation, we generated CD80-eCFP mice using bacterial artificial chromosome transgenic technology. In splenic DCs, endogenous CD80 and CD80-eCFP localized to plasma membrane and Golgi apparatus, and CD80-eCFP was functional in vivo. In the OT-II T cell-DC IS, multiple segregated TCR, CD80, and LFA-1 clusters were detected. In the T cell-DC synapse CD80 clusters were colocalized with CD28 and PKCtheta, a characteristic of the cSMAC. Acute blockade of TCR signaling with anti-MHC Ab resulted in a rapid reduction in Ca(2+) signaling and the number and size of the CD80 clusters, a characteristic of TCR microclusters. Thus, the T cell-DC interface contains dynamic costimulatory foci that share characteristics of microclusters and cSMACs.


Asunto(s)
Antígeno B7-1/metabolismo , Antígenos CD28/metabolismo , Células Dendríticas/inmunología , Sinapsis Inmunológicas/metabolismo , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Receptores de Antígenos de Linfocitos T/fisiología , Subgrupos de Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Antígeno B7-1/genética , Antígeno B7-1/fisiología , Antígenos CD28/genética , Antígenos CD28/fisiología , Células CHO , Cromosomas Artificiales Bacterianos/genética , Cricetinae , Cricetulus , Células Dendríticas/enzimología , Células Dendríticas/metabolismo , Proteínas Fluorescentes Verdes/genética , Sinapsis Inmunológicas/enzimología , Sinapsis Inmunológicas/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Datos de Secuencia Molecular , Proteína Quinasa C-theta , Transporte de Proteínas/genética , Transporte de Proteínas/inmunología , Receptores de Antígenos de Linfocitos T/genética , Subgrupos de Linfocitos T/enzimología , Subgrupos de Linfocitos T/metabolismo
12.
Nat Immunol ; 8(8): 835-44, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17632517

RESUMEN

T cells survey antigen-presenting dendritic cells (DCs) by migrating through DC networks, arresting and maintaining contact with DCs for several hours after encountering high-potency complexes of peptide and major histocompatibility complex (pMHC), leading to T cell activation. The effects of low-potency pMHC complexes on T cells in vivo, however, are unknown, as is the mechanism controlling T cell arrest. Here we evaluated T cell responses in vivo to high-, medium- and low-potency pMHC complexes and found that regardless of potency, pMHC complexes induced upregulation of CD69, anergy and retention of T cells in lymph nodes. However, only high-potency pMHC complexes expressed by DCs induced calcium-dependent T cell deceleration and calcineurin-dependent anergy. The pMHC complexes of lower potency instead induced T cell anergy by a biochemically distinct process that did not affect T cell dynamics.


Asunto(s)
Presentación de Antígeno/inmunología , Comunicación Celular/inmunología , Células Dendríticas/inmunología , Ganglios Linfáticos/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Linfocitos T/inmunología , Animales , Anergia Clonal , Ganglios Linfáticos/citología , Activación de Linfocitos/inmunología , Ratones , Ratones Transgénicos , Péptidos/inmunología
13.
Cell ; 129(4): 773-85, 2007 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-17512410

RESUMEN

The immunological synapse (IS) is a junction between the T cell and antigen-presenting cell and is composed of supramolecular activation clusters (SMACs). No studies have been published on naive T cell IS dynamics. Here, we find that IS formation during antigen recognition comprises cycles of stable IS formation and autonomous naive T cell migration. The migration phase is driven by PKCtheta, which is localized to the F-actin-dependent peripheral (p)SMAC. PKCtheta(-/-) T cells formed hyperstable IS in vitro and in vivo and, like WT cells, displayed fast oscillations in the distal SMAC, but they showed reduced slow oscillations in pSMAC integrity. IS reformation is driven by the Wiscott Aldrich Syndrome protein (WASp). WASp(-/-) T cells displayed normal IS formation but were unable to reform IS after migration unless PKCtheta was inhibited. Thus, opposing effects of PKCtheta and WASp control IS stability through pSMAC symmetry breaking and reformation.


Asunto(s)
Presentación de Antígeno/fisiología , Células Presentadoras de Antígenos/metabolismo , Uniones Intercelulares/metabolismo , Isoenzimas/metabolismo , Proteína Quinasa C/metabolismo , Linfocitos T/metabolismo , Proteína del Síndrome de Wiskott-Aldrich/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Comunicación Celular/fisiología , Movimiento Celular/fisiología , Activación Enzimática/fisiología , Inhibidores Enzimáticos/farmacología , Represión Enzimática/efectos de los fármacos , Represión Enzimática/fisiología , Uniones Intercelulares/genética , Uniones Intercelulares/inmunología , Isoenzimas/genética , Activación de Linfocitos/fisiología , Lípidos de la Membrana/metabolismo , Ratones , Ratones Noqueados , Proteína Quinasa C/genética , Proteína Quinasa C-theta , Linfocitos T/inmunología , Proteína del Síndrome de Wiskott-Aldrich/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA