Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 157
Filtrar
1.
Front Immunol ; 15: 1392933, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38779683

RESUMEN

Introduction: Antigen binding to the T cell antigen receptor (TCR) leads to the phosphorylation of the immunoreceptor tyrosine-based activation motifs (ITAMs) of the CD3 complex, and thereby to T cell activation. The CD3ε subunit plays a unique role in TCR activation by recruiting the kinase LCK and the adaptor protein NCK prior to ITAM phosphorylation. Here, we aimed to investigate how phosphorylation of the individual CD3ε ITAM tyrosines impacts the CD3ε signalosome. Methods: We mimicked irreversible tyrosine phosphorylation by substituting glutamic acid for the tyrosine residues in the CD3ε ITAM. Results: Integrating CD3ε phospho-mimetic variants into the complete TCR-CD3 complex resulted in reduced TCR signal transduction, which was partially compensated by the involvement of the other TCR-CD3 ITAMs. By using novel CD3ε phospho-mimetic Chimeric Antigen Receptor (CAR) variants, we avoided any compensatory effects of other ITAMs in the TCR-CD3 complex. We demonstrated that irreversible CD3ε phosphorylation prevented signal transduction upon CAR engagement. Mechanistically, we demonstrated that glutamic acid substitution at the N-terminal tyrosine residue of the CD3ε ITAM (Y39E) significantly reduces NCK binding to the TCR. In contrast, mutation at the C-terminal tyrosine of the CD3ε ITAM (Y50E) abolished LCK recruitment to the TCR, while increasing NCK binding. Double mutation at the C- and N-terminal tyrosines (Y39/50E) allowed ZAP70 to bind, but reduced the interaction with LCK and NCK. Conclusions: The data demonstrate that the dynamic phosphorylation of the CD3ε ITAM tyrosines is essential for CD3ε to orchestrate optimal TCR and CAR signaling and highlights the key role of CD3ε signalosome to tune signal transduction.


Asunto(s)
Complejo CD3 , Receptores de Antígenos de Linfocitos T , Receptores Quiméricos de Antígenos , Transducción de Señal , Complejo CD3/metabolismo , Complejo CD3/inmunología , Fosforilación , Humanos , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/genética , Activación de Linfocitos/inmunología , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Células HEK293 , Proteína Tirosina Quinasa ZAP-70/metabolismo , Proteína Tirosina Quinasa ZAP-70/genética , Motivo de Activación del Inmunorreceptor Basado en Tirosina , Unión Proteica , Células Jurkat , Proteínas Oncogénicas
2.
Cancer Immunol Res ; 11(1): 93-108, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36265009

RESUMEN

The engagement of the T-cell receptor (TCR) by a specific peptide-MHC ligand initiates transmembrane signaling to induce T-cell activation, a key step in most adaptive immune responses. Previous studies have indicated that TCR signaling is tightly regulated by cholesterol and its sulfate metabolite, cholesterol sulfate (CS), on the membrane. Here, we report a novel mechanism by which CS modulates TCR signaling through a conformational change of CD3 subunits. We found that the negatively charged CS interacted with the positively charged cytoplasmic domain of CD3ε (CD3εCD) to enhance its binding to the cell membrane and induce a stable secondary structure. This secondary structure suppressed the release of CD3εCD from the membrane in the presence of Ca2+, which in turn inhibited TCR phosphorylation and signaling. When a point mutation (I/A) was introduced to the intracellular immunoreceptor tyrosine-based activation motifs (YxxI-x6-8-YxxL) of CD3ε subunit, it reduced the stability of the secondary structure and regained sensitivity to Ca2+, which abolished CS-mediated inhibition and enhanced the signaling of the TCR complex. Notably, the I/A mutation could be applied to both murine and human TCR-T cell therapy to improve the antitumor efficacy. Our study reveals insights into the regulatory mechanism of TCR signaling and provides a strategy to functionally engineer the TCR/CD3 complex for T cell-based cancer immunotherapy.


Asunto(s)
Calcio , Linfocitos T , Animales , Humanos , Ratones , Calcio/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Complejo CD3/genética , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Fosforilación , Lípidos/análisis
3.
Mol Cell ; 82(7): 1278-1287.e5, 2022 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-35271814

RESUMEN

Cholesterol molecules specifically bind to the resting αßTCR to inhibit cytoplasmic CD3ζ ITAM phosphorylation through sequestering the TCR-CD3 complex in an inactive conformation. The mechanisms of cholesterol-mediated inhibition of TCR-CD3 and its activation remain unclear. Here, we present cryoelectron microscopy structures of cholesterol- and cholesterol sulfate (CS)-inhibited TCR-CD3 complexes and an auto-active TCR-CD3 variant. The structures reveal that cholesterol molecules act like a latch to lock CD3ζ into an inactive conformation in the membrane. Mutations impairing binding of cholesterol molecules to the tunnel result in the movement of the proximal C terminus of the CD3ζ transmembrane helix, thereby activating the TCR-CD3 complex in human cells. Together, our data reveal the structural basis of TCR inhibition by cholesterol, illustrate how the cholesterol-binding tunnel is allosterically coupled to TCR triggering, and lay a foundation for the development of immunotherapies through directly targeting the TCR-CD3 complex.


Asunto(s)
Complejo Receptor-CD3 del Antígeno de Linfocito T , Linfocitos T , Complejo CD3/genética , Complejo CD3/metabolismo , Colesterol/metabolismo , Microscopía por Crioelectrón , Humanos , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Linfocitos T/metabolismo
4.
Nat Commun ; 12(1): 7296, 2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34911975

RESUMEN

CD137 (4-1BB; TNFSR9) is an activation-induced surface receptor that through costimulation effects provide antigen-primed T cells with augmented survival, proliferation and effector functions as well as metabolic advantages. These immunobiological mechanisms are being utilised for cancer immunotherapy with agonist CD137-binding and crosslinking-inducing agents that elicit CD137 intracellular signaling. In this study, side-by-side comparisons show that provision of CD137 costimulation in-cis with regard to the TCR-CD3-ligating cell is superior to that provided in-trans in terms of T cell activation, proliferation, survival, cytokine secretion and mitochondrial fitness in mouse and human. Cis ligation of CD137 relative to the TCR-CD3 complex results in more intense canonical and non-canonical NF-κB signaling and provides a more robust induction of cell cycle and DNA damage repair gene expression programs. Here we report that the superiority of cis versus trans CD137-costimulation is readily observed in vivo and is relevant for understanding the immunotherapeutic effects of CAR T cells and CD137 agonistic therapies currently undergoing clinical trials, which may provide costimulation either in cis or in trans.


Asunto(s)
Complejo CD3/inmunología , Linfocitos T CD8-positivos/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología , Animales , Complejo CD3/genética , Proliferación Celular , Citocinas/genética , Citocinas/inmunología , Humanos , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
5.
Mol Syst Biol ; 16(8): e9416, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32779888

RESUMEN

It has recently become possible to simultaneously assay T-cell specificity with respect to large sets of antigens and the T-cell receptor sequence in high-throughput single-cell experiments. Leveraging this new type of data, we propose and benchmark a collection of deep learning architectures to model T-cell specificity in single cells. In agreement with previous results, we found that models that treat antigens as categorical outcome variables outperform those that model the TCR and antigen sequence jointly. Moreover, we show that variability in single-cell immune repertoire screens can be mitigated by modeling cell-specific covariates. Lastly, we demonstrate that the number of bound pMHC complexes can be predicted in a continuous fashion providing a gateway to disentangle cell-to-dextramer binding strength and receptor-to-pMHC affinity. We provide these models in the Python package TcellMatch to allow imputation of antigen specificities in single-cell RNA-seq studies on T cells without the need for MHC staining.


Asunto(s)
Biología Computacional/métodos , Antígenos de Histocompatibilidad/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Análisis de la Célula Individual/métodos , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Aprendizaje Profundo , Antígenos de Histocompatibilidad/genética , Humanos , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Análisis de Secuencia de ARN , Aprendizaje Automático Supervisado
6.
Proc Natl Acad Sci U S A ; 117(27): 15809-15817, 2020 07 07.
Artículo en Inglés | MEDLINE | ID: mdl-32571924

RESUMEN

Src family kinase Lck plays critical roles during T cell development and activation, as it phosphorylates the TCR/CD3 complex to initiate TCR signaling. Lck is present either in coreceptor-bound or coreceptor-unbound (free) forms, and we here present evidence that the two pools of Lck have different molecular properties. We discovered that the free Lck fraction exhibited higher mobility than CD8α-bound Lck in OT-I T hybridoma cells. The free Lck pool showed more activating Y394 phosphorylation than the coreceptor-bound Lck pool. Consistent with this, free Lck also had higher kinase activity, and free Lck mediated higher T cell activation as compared to coreceptor-bound Lck. Furthermore, the coreceptor-Lck coupling was independent of TCR activation. These findings give insights into the initiation of TCR signaling, suggesting that changes in coreceptor-Lck coupling constitute a mechanism for regulation of T cell sensitivity.


Asunto(s)
Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/metabolismo , Familia-src Quinasas/genética , Animales , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/genética , Hibridomas/inmunología , Activación de Linfocitos/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/inmunología , Ratones , Fosforilación/genética , Unión Proteica/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Transducción de Señal , Linfocitos T/inmunología
7.
Immunology ; 159(3): 298-308, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31674657

RESUMEN

Following T-cell antigen receptor (TCR) engagement, rearrangement of the actin cytoskeleton supports intracellular signal transduction and T-cell activation. The non-catalytic region of the tyrosine kinase (Nck) molecule is an adapter protein implicated in TCR-induced actin polymerization. Further, Nck is recruited to the CD3ε subunit of the TCR upon TCR triggering. Here we examine the role of actin polymerization in the recruitment of Nck to the TCR. To this end, Nck binding to CD3ε was quantified in Jurkat cells using the proximity ligation assay. We show that inhibition of actin polymerization using cytochalasin D delayed the recruitment of Nck1 to the TCR upon TCR triggering. Interestingly, CD3ε phosphorylation was also delayed. These findings suggest that actin polymerization promotes the recruitment of Nck to the TCR, enhancing downstream signaling, such as phosphorylation of CD3ε.


Asunto(s)
Citoesqueleto de Actina/metabolismo , Actinas/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Complejo CD3/metabolismo , Activación de Linfocitos , Proteínas Oncogénicas/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Linfocitos T/enzimología , Citoesqueleto de Actina/inmunología , Actinas/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Complejo CD3/inmunología , Citocalasina D/farmacología , Humanos , Células Jurkat , Activación de Linfocitos/efectos de los fármacos , Proteínas Oncogénicas/genética , Fosforilación , Polimerizacion , Unión Proteica , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Transducción de Señal , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Factores de Tiempo , Proteína Tirosina Quinasa ZAP-70/metabolismo
8.
Immunology ; 159(4): 373-383, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31821535

RESUMEN

The anti-tumor immune response is considered to be due to the T-cell receptor (TCR) binding to tumor antigens, which can be either wild-type, early stem cell proteins, presumably foreign to a developed immune system; or mutant peptides, foreign to the immune system because of a mutant amino acid (aa) or otherwise somatically altered aa sequence. Recently, very large numbers of TCR complementarity-determining region-3 (CDR3) aa sequences obtained from tumor specimens have become available. We developed a novel algorithm for assessing the complementarity of tumor mutant peptides and TCR CDR3s, based on the retrieval of TCR CDR3 aa sequences from both tumor specimen and patient blood exomes and by using an automated process of assessing CDR3 and mutant aa electrical charges. Results indicated many instances where high electrostatic complementarity was associated with a higher survival rate. In particular, our approach led to the identification of specific genes contributing significantly to the complementary, TCR CDR3-mutant aa. These results suggest a novel approach to tumor immunoscoring and may lead to the identification of high-priority neo-antigen, peptide vaccines; or to the identification of ex vivo stimulants of tumor-infiltrating lymphocytes.


Asunto(s)
Algoritmos , Antígenos de Neoplasias/química , Neoplasias de la Mama/genética , Regiones Determinantes de Complementariedad/química , Neoplasias Pulmonares/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Neoplasias Cutáneas/genética , Secuencia de Aminoácidos , Aminoácidos , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Sitios de Unión , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/mortalidad , Regiones Determinantes de Complementariedad/genética , Regiones Determinantes de Complementariedad/inmunología , Exoma , Femenino , Expresión Génica , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Masculino , Mutación , Pronóstico , Unión Proteica , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Proyectos de Investigación , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/mortalidad , Electricidad Estática , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/patología
9.
Proc Natl Acad Sci U S A ; 117(1): 285-291, 2020 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-31871161

RESUMEN

The impact of ultrasmall nanoparticles (<10-nm diameter) on the immune system is poorly understood. Recently, ultrasmall silica nanoparticles (USSN), which have gained increasing attention for therapeutic applications, were shown to stimulate T lymphocytes directly and at relatively low-exposure doses. Delineating underlying mechanisms and associated cell signaling will hasten therapeutic translation and is reported herein. Using competitive binding assays and molecular modeling, we established that the T cell receptor (TCR):CD3 complex is required for USSN-induced T cell activation, and that direct receptor complex-particle interactions are permitted both sterically and electrostatically. Activation is not limited to αß TCR-bearing T cells since those with γδ TCR showed similar responses, implying that USSN mediate their effect by binding to extracellular domains of the flanking CD3 regions of the TCR complex. We confirmed that USSN initiated the signaling pathway immediately downstream of the TCR with rapid phosphorylation of both ζ-chain-associated protein 70 and linker for activation of T cells protein. However, T cell proliferation or IL-2 secretion were only triggered by USSN when costimulatory anti-CD28 or phorbate esters were present, demonstrating that the specific impact of USSN is in initiation of the primary, nuclear factor of activated T cells-pathway signaling from the TCR complex. Hence, we have established that USSN are partial agonists for the TCR complex because of induction of the primary T cell activation signal. Their ability to bind the TCR complex rapidly, and then to dissolve into benign orthosilicic acid, makes them an appealing option for therapies targeted at transient TCR:CD3 receptor binding.


Asunto(s)
Activación de Linfocitos/efectos de los fármacos , Nanopartículas/química , Complejo Receptor-CD3 del Antígeno de Linfocito T/efectos de los fármacos , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Dióxido de Silicio/química , Dióxido de Silicio/farmacología , Antígenos CD28/metabolismo , Complejo CD3/química , Complejo CD3/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Interleucina-2/metabolismo , Modelos Moleculares , Fosforilación , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Transducción de Señal/inmunología , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo
10.
Sci Rep ; 8(1): 5926, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29651132

RESUMEN

The diversity of the T cell receptor (TCR) complementarity determining region 3 (CDR3) repertoire is the result of random combinations, insertions and deletions during recombination of the germline V, D and J gene fragments. During evolution, some human TCR beta chain variable (TRBV) pseudogenes have been retained. Many previous studies have focused on functional TRBV genes, while little attention has been given to TRBV pseudogenes. To describe the compositional characteristics of TRBV pseudogene rearrangements, we compared and analysed TRBV pseudogenes, TRBV open reading frames (ORFs) and functional TRBV genes via high-throughput sequencing of DNA obtained from the peripheral blood of 4 healthy volunteers and 4 patients. Our results revealed several differences in J and D gene usage. The V deletion distribution profile of the pseudogenes was significantly different from that of the ORFs and functional genes. In addition, arginine, lysine and cysteine were more frequently used in putative CDR3 pseudogene rearrangements, while functional rearrangements used more leucine. This study presents a comprehensive description of the compositional characteristics of peripheral TRBV pseudogene rearrangements, which will provide a reference for further research on TRBV pseudogenes.


Asunto(s)
Regiones Determinantes de Complementariedad/genética , Seudogenes/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Receptores de Antígenos de Linfocitos T/genética , Regiones Determinantes de Complementariedad/sangre , Genes Codificadores de la Cadena beta de los Receptores de Linfocito T/genética , Voluntarios Sanos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Complejo Receptor-CD3 del Antígeno de Linfocito T/sangre , Complejo Receptor-CD3 del Antígeno de Linfocito T/química
11.
Mol Ther ; 25(2): 456-464, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-28109957

RESUMEN

Chimeric major histocompatibility complex (MHC) molecules supplemented with T cell receptor (TCR) signaling motifs function as activation receptors and can redirect gene-modified T cells against pathogenic CD8 T cells. We have shown that ß2 microglobulin (ß2m) operates as a universal signaling component of MHC-I molecules when fused with the CD3-ζ chain. Linking the H-2Kd-binding insulin B chain peptide insulin B chain, amino acids 15-23 (InsB15-23) to the N terminus of ß2m/CD3-ζ, redirected polyclonal CD8 T cells against pathogenic CD8 T cells in a peptide-specific manner in the non-obese diabetic (NOD) mouse. Here, we describe mRNA electroporation for delivering peptide/ß2m/CD3-ζ genes to a reporter T cell line and purified primary mouse CD8 T cells. The peptide/ß2m/CD3-ζ products paired with endogenous MHC-I chains and transmitted strong activation signals upon MHC-I cross-linking. The reporter T cell line transfected with InsB15-23/ß2m/CD3-ζ mRNA was activated by an InsB15-23-H-2Kd-specific CD8 T cell hybrid only when the transfected T cells expressed H-2Kd. Primary NOD CD8 T cells expressing either InsB15-23/ß2m/CD3-ζ or islet-specific glucose-6-phosphatase catalytic subunit-related protein, amino acids 206-214 (IGRP206-214)/ß2m/CD3-ζ killed their respective autoreactive CD8 T cell targets in vitro. Furthermore, transfer of primary CD8 T cells transfected with InsB15-23/ß2m/CD3-ζ mRNA significantly reduced insulitis and protected NOD mice from diabetes. Our results demonstrate that mRNA encoding chimeric MHC-I receptors can redirect effector CD8 against diabetogenic CD8 T cells, offering a new approach for the treatment of type 1 diabetes.


Asunto(s)
Traslado Adoptivo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Inmunomodulación , ARN Mensajero/genética , Linfocitos T/inmunología , Linfocitos T/metabolismo , Animales , Citotoxicidad Inmunológica , Diabetes Mellitus Tipo 1/prevención & control , Diabetes Mellitus Tipo 1/terapia , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Orden Génico , Vectores Genéticos/genética , Insulina/inmunología , Ratones , Ratones Endogámicos NOD , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Proteínas Recombinantes de Fusión/genética , Transfección , Microglobulina beta-2/genética
12.
Mol Ther ; 25(1): 285-295, 2017 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-28129122

RESUMEN

Outcomes for patients with refractory diffuse large B cell lymphoma (DLBCL) are poor. In the multicenter ZUMA-1 phase 1 study, we evaluated KTE-C19, an autologous CD3ζ/CD28-based chimeric antigen receptor (CAR) T cell therapy, in patients with refractory DLBCL. Patients received low-dose conditioning chemotherapy with concurrent cyclophosphamide (500 mg/m2) and fludarabine (30 mg/m2) for 3 days followed by KTE-C19 at a target dose of 2 × 106 CAR T cells/kg. The incidence of dose-limiting toxicity (DLT) was the primary endpoint. Seven patients were treated with KTE-C19 and one patient experienced a DLT of grade 4 cytokine release syndrome (CRS) and neurotoxicity. Grade ≥3 CRS and neurotoxicity were observed in 14% (n = 1/7) and 57% (n = 4/7) of patients, respectively. All other KTE-C19-related grade ≥3 events resolved within 1 month. The overall response rate was 71% (n = 5/7) and complete response (CR) rate was 57% (n = 4/7). Three patients have ongoing CR (all at 12+ months). CAR T cells demonstrated peak expansion within 2 weeks and continued to be detectable at 12+ months in patients with ongoing CR. This regimen of KTE-C19 was safe for further study in phase 2 and induced durable remissions in patients with refractory DLBCL.


Asunto(s)
Antígenos CD19/inmunología , Antígenos CD28/metabolismo , Linfoma no Hodgkin/inmunología , Linfoma no Hodgkin/terapia , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Proteínas Recombinantes de Fusión , Linfocitos T/inmunología , Linfocitos T/metabolismo , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Biomarcadores , Antígenos CD28/genética , Terapia Combinada , Progresión de la Enfermedad , Resistencia a Antineoplásicos , Femenino , Estudios de Seguimiento , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/efectos adversos , Inmunoterapia Adoptiva/métodos , Linfoma de Células B Grandes Difuso/diagnóstico , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/terapia , Linfoma no Hodgkin/diagnóstico , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Resultado del Tratamiento
13.
Proc Natl Acad Sci U S A ; 113(43): E6649-E6658, 2016 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-27791034

RESUMEN

The T-cell antigen receptor (TCR) is an assembly of eight type I single-pass membrane proteins that occupies a central position in adaptive immunity. Many TCR-triggering models invoke an alteration in receptor complex structure as the initiating event, but both the precise subunit organization and the pathway by which ligand-induced alterations are transferred to the cytoplasmic signaling domains are unknown. Here, we show that the receptor complex transmembrane (TM) domains form an intimately associated eight-helix bundle organized by a specific interhelical TCR TM interface. The salient features of this core structure are absolutely conserved between αß and γδ TCR sequences and throughout vertebrate evolution, and mutations at key interface residues caused defects in the formation of stable TCRαß:CD3δε:CD3γε:ζζ complexes. These findings demonstrate that the eight TCR-CD3 subunits form a compact and precisely organized structure within the membrane and provide a structural basis for further investigation of conformationally regulated models of transbilayer TCR signaling.


Asunto(s)
Complejo CD3/química , Membrana Celular/ultraestructura , Subunidades de Proteína/química , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Receptores de Antígenos de Linfocitos T gamma-delta/química , Secuencia de Aminoácidos , Sitios de Unión , Complejo CD3/genética , Complejo CD3/inmunología , Membrana Celular/química , Membrana Celular/inmunología , Cristalografía por Rayos X , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Simulación de Dinámica Molecular , Unión Proteica , Conformación Proteica en Hélice alfa , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Linfocitos T/química , Linfocitos T/inmunología
14.
Diabetologia ; 58(12): 2800-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26363782

RESUMEN

AIMS/HYPOTHESIS: The LEW.1AR1-iddm rat, an animal model of human type 1 diabetes, arose through a spontaneous mutation within the inbred strain LEW.1AR1. A susceptibility locus (Iddm8) on rat chromosome 1 (RNO1) has been identified previously, which is accompanied by autoimmune diabetes and the additional phenotype of a variable CD3(+) T cell frequency. METHODS: In the present study we characterised the Iddm8 region on RNO1 in backcross strains using the genetically divergent Brown Norway (BN) and Paris (PAR) rats. Candidate genes of the Iddm8 region were sequenced for mutation analysis. RESULTS: The Iddm8 region could be subdivided by single nucleotide polymorphism (SNP) analyses. In the first region, a mutation in exon 44 of the Dock8 gene was identified resulting in an amino acid exchange in the protein from glutamine to glutamate. This exchange is unique for the LEW.1AR1-iddm rat. In the second region, a SNP was detected in exon 11 of the Vwa2 gene with an exchange from arginine to tryptophan. This SNP is also present in other rat strains. CONCLUSIONS/INTERPRETATION: The Dock8 mutation gave rise to a new type 1 diabetes rat model with very close similarity to type 1 diabetes in humans, providing a deepened insight into the impact of genes involved in diabetes development.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Factores de Intercambio de Guanina Nucleótido/genética , Mutación , Alelos , Sustitución de Aminoácidos , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Exones/genética , Humanos , Células Asesinas Naturales , Modelos Moleculares , Polimorfismo de Nucleótido Simple , Ratas , Ratas Endogámicas Lew , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Factor de von Willebrand/genética
15.
Clin Immunol ; 161(2): 190-6, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26360253

RESUMEN

To examine the T cell receptor structure in the absence of B cells, the TCR ß CDR3 was sequenced from DNA of 15 X-linked agammaglobulinemia (XLA) subjects and 18 male controls, using the Illumina HiSeq platform and the ImmunoSEQ analyzer. V gene usage and the V-J combinations, derived from both productive and non-productive sequences, were significantly different between XLA samples and controls. Although the CDR3 length was similar for XLA and control samples, the CDR3 region of the XLA T cell receptor contained significantly fewer deletions and insertions in V, D, and J gene segments, differences intrinsic to the V(D)J recombination process and not due to peripheral T cell selection. XLA CDR3s demonstrated fewer charged amino acid residues, more sharing of CDR3 sequences, and almost completely lacked a population of highly modified Vß gene segments found in control DNA, suggesting both a skewed and contracted T cell repertoire in XLA.


Asunto(s)
Agammaglobulinemia/genética , Enfermedades Genéticas Ligadas al Cromosoma X/genética , Receptores de Antígenos de Linfocitos T/genética , Adolescente , Adulto , Linfocitos B/metabolismo , Estudios de Casos y Controles , Niño , Preescolar , ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Región Variable de Inmunoglobulina/genética , Lactante , Masculino , Persona de Mediana Edad , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Linfocitos T/metabolismo , Adulto Joven
16.
J Biol Chem ; 290(32): 19796-805, 2015 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-26109064

RESUMEN

The T cell receptor (TCR)-CD3 complex is composed of a genetically diverse αß TCR heterodimer associated noncovalently with the invariant CD3 dimers CD3ϵγ, CD3ϵδ, and CD3ζζ. The TCR mediates peptide-MHC recognition, whereas the CD3 molecules transduce activation signals to the T cell. Although much is known about downstream T cell signaling pathways, the mechanism whereby TCR engagement by peptide-MHC initiates signaling is poorly understood. A key to solving this problem is defining the spatial organization of the TCR-CD3 complex and the interactions between its subunits. We have applied solution NMR methods to identify the docking site for CD3 on the ß chain of a human autoimmune TCR. We demonstrate a low affinity but highly specific interaction between the extracellular domains of CD3 and the TCR constant ß (Cß) domain that requires both CD3ϵγ and CD3ϵδ subunits. The mainly hydrophilic docking site, comprising 9-11 solvent-accessible Cß residues, is relatively small (∼400 Å(2)), consistent with the weak interaction between TCR and CD3 extracellular domains, and devoid of glycosylation sites. The docking site is centered on the αA and αB helices of Cß, which are located at the base of the TCR. This positions CD3ϵγ and CD3ϵδ between the TCR and the T cell membrane, permitting us to distinguish among several possible models of TCR-CD3 association. We further correlate structural results from NMR with mutational data on TCR-CD3 interactions from cell-based assays.


Asunto(s)
Complejo CD3/química , Subunidades de Proteína/química , Complejo Receptor-CD3 del Antígeno de Linfocito T/química , Receptores de Antígenos de Linfocitos T alfa-beta/química , Secuencia de Aminoácidos , Complejo CD3/genética , Complejo CD3/inmunología , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Humanos , Espectroscopía de Resonancia Magnética , Simulación del Acoplamiento Molecular , Datos de Secuencia Molecular , Mutación , Pliegue de Proteína , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/genética , Subunidades de Proteína/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Linfocitos T/química , Linfocitos T/inmunología
17.
Mol Immunol ; 58(1): 10-6, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24252355

RESUMEN

Celiac disease (CD) patients who fail to respond to a gluten-free diet suffer from refractory celiac disease (RCD). A marked expansion of intraepithelial lymphocytes (IEL) lacking surface TCR/CD3 expression characterizes the RCD subtype II. In up to 50% of RCDII patients these so-called aberrant IEL (a-IEL) develop into lymphoma and can disseminate into other tissues. Elevated levels of Interleukin-15 (IL-15) in the intestine of CD and RCD patients likely contribute to the expansion of a-IEL. Here, we investigated if interactions with other cells might also influence a-IEL expansion. Similar to IL-15, cells from the monocyte lineage, particularly mature dendritic cells (DCs), promoted proliferation, prevented apoptosis and induced IFNγ secretion of a-IEL derived from RCDII biopsies (RCDII cell lines), which in turn induced CXCL10. In contrast to IL-15, mature DCs did not induce proliferation of regular TCR(+)IEL lines, generated from CD biopsies and IL-15-blocking antibodies did not inhibit DC-induced proliferation of RCDII cell lines. Furthermore, proliferation was dependent on cell-cell contact, but independent of the HLA-genotype of the stimulating cells. Our results suggest that contact with DC, either in the epithelium or upon dissemination, contributes to uncontrolled expansion of a-IEL in RCDII, independent of HLA-genotype and IL-15.


Asunto(s)
Enfermedad Celíaca/inmunología , Células Dendríticas/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/deficiencia , Linfocitos T/inmunología , Anticuerpos Bloqueadores/inmunología , Apoptosis/inmunología , Comunicación Celular/inmunología , Línea Celular , Proliferación Celular , Supervivencia Celular , Quimiocina CXCL10/biosíntesis , Células HT29 , Humanos , Interferón gamma/biosíntesis , Interferón gamma/metabolismo , Interleucina-15/biosíntesis , Interleucina-15/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Monocitos/inmunología , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología
18.
J Immunol ; 192(2): 771-81, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337748

RESUMEN

GTPases act as important switches in many signaling events in cells. Although small and heterotrimeric G proteins are subjects of intensive studies, little is known about the large IFN-inducible GTPases. In this article, we show that the IFN-γ-inducible guanylate binding protein 1 (GBP-1) is a regulator of T cell activation. Silencing of GBP-1 leads to enhanced activation of early T cell Ag receptor/CD3 signaling molecules, including Lck, that is translated to higher IL-2 production. Mass spectrometry analyses showed that regulatory cytoskeletal proteins, like plastin-2 that bundles actin fibers and spectrin ß-chain, brain 1 that links the plasma membrane to the actin cytoskeleton, are binding partners of GBP-1. The spectrin cytoskeleton influences cell spreading and surface expression of TCR/CD3 and the leukocyte phosphatase CD45. We found higher cell spreading and enhanced surface expression of TCR/CD3 and CD45 in GBP-1 silenced T cells that explain their enhanced TCR/CD3 signaling. We conclude that GBP-1 is a downstream processor of IFN-γ via which T cells regulate cytoskeleton-dependent cell functions.


Asunto(s)
Citoesqueleto/metabolismo , Proteínas de Unión al GTP/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Complejo CD3/genética , Complejo CD3/metabolismo , Línea Celular , Línea Celular Tumoral , Citoesqueleto/genética , Citosol/metabolismo , Proteínas de Unión al GTP/genética , Células HEK293 , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-2/genética , Interleucina-2/metabolismo , Células Jurkat , Antígenos Comunes de Leucocito , Leucocitos/metabolismo , Activación de Linfocitos/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/genética , Proteína Tirosina Quinasa p56(lck) Específica de Linfocito/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Receptores de Antígenos de Linfocitos T/genética , Transducción de Señal/genética , Linfocitos T/metabolismo , Regulación hacia Arriba/genética
19.
Gene Ther ; 20(2): 177-86, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22378346

RESUMEN

A majority of cancer deaths are because of an uncontrolled relapse of the disease despite initial remission after therapy, asking for strategies to control tumour cells in the long term. Adoptive therapy with chimeric antigen receptor (CAR)-redirected T cells showed promising success in primary tumour elimination; the capacity of such engineered T cells to establish enduring tumour protection is currently a matter of discussion, in particular as most targeted 'tumour-associated antigens' are self-antigens. To address the issue in a clinically relevant model that closely mimics the human situation, we recorded rejection of carcinoembryonic antigen (CEA)-positive pancreatic tumours in the CEA transgenic mouse that expressed CEA as self-antigen in healthy cells of the gastrointestinal tract. Adoptive therapy with CD8(+) T cells, which were redirected by a CEA-specific, low-affinity CAR with CD3ζ endodomain, eliminated CEA(+) tumours in a primary response; cured mice produced an efficient recall response in the long term towards CEA(+) tumour cells upon rechallenge. Secondary tumour rejection was CEA specific, mediated by engineered T cells and did not require host T cells. No toxicity towards healthy tissues with CEA expression was recorded. Data indicate that adoptive therapy with engineered T cells can establish self-antigen-specific tumour protection in the long term without autoimmunity.


Asunto(s)
Carcinoma/prevención & control , Neoplasias Pancreáticas/prevención & control , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Linfocitos T/inmunología , Animales , Complejo CD3/química , Complejo CD3/genética , Antígeno Carcinoembrionario/genética , Antígeno Carcinoembrionario/metabolismo , Carcinoma/terapia , Línea Celular Tumoral , Terapia Genética , Células HEK293 , Humanos , Inmunoterapia Adoptiva , Ratones , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/terapia , Estructura Terciaria de Proteína , Linfocitos T/trasplante
20.
J Biol Chem ; 287(45): 38168-77, 2012 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-22989874

RESUMEN

Th17 lymphocytes play a key role during immune responses against bacteria and fungi and are involved in the pathophysiology of multiple autoimmune disorders. The co-stimulatory molecules SLAMF3 and SLAMF6 have been implicated in the formation of Th17 phenotypes and IL-17A expression. Increased surface expression of SLAMF3 and SLAMF6 has been linked with disease activity in systemic lupus erythematosus. Here we demonstrate that in human total T lymphocytes the canonical CD28 and the non-canonical SLAMF3/SLAMF6 co-stimulatory pathways cooperate in the recruitment of the transcription factor NFAT1 to the IL17A promoter. Furthermore, the dominance of the SLAMF3/SLAMF6 pathway in inducing IL-17A production can be attributed to an increased nuclear abundance and recruitment of RORγt to the IL17A promoter. Thus, we have identified an additional mechanism that may be central for the specific control of IL17A gene regulation in systemic lupus erythematosus T lymphocytes.


Asunto(s)
Antígenos CD/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Regiones Promotoras Genéticas/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/inmunología , Receptores de Superficie Celular/inmunología , Linfocitos T/inmunología , Anticuerpos/inmunología , Anticuerpos/farmacología , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos CD28/genética , Antígenos CD28/inmunología , Antígenos CD28/metabolismo , Células Cultivadas , Inmunoprecipitación de Cromatina , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Immunoblotting , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-17/metabolismo , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Lupus Eritematoso Sistémico/metabolismo , Mutación , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/inmunología , Factores de Transcripción NFATC/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Unión Proteica , Complejo Receptor-CD3 del Antígeno de Linfocito T/genética , Complejo Receptor-CD3 del Antígeno de Linfocito T/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Miembro 1 de la Familia de Moléculas Señalizadoras de la Activación Linfocitaria , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...