Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(24): 9879-84, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23716685

RESUMEN

T-cell costimulation and coinhibition generated by engagement of the B7 family and their receptor CD28 family are of central importance in regulating the T-cell response, making these pathways very attractive therapeutic targets. Here we describe HERV-H LTR-associating protein 2 (HHLA2) as a member of the B7 family that shares 10-18% amino acid identity and 23-33% similarity to other human B7 proteins and phylogenetically forms a subfamily with B7x and B7-H3 within the family. HHLA2 is expressed in humans but not in mice, which is unique within the B7 and CD28 families. HHLA2 protein is constitutively expressed on the surface of human monocytes and is induced on B cells after stimulation with LPS and IFN-γ. HHLA2 does not interact with other known members of the CD28 family or the B7 family, but does bind a putative receptor that is constitutively expressed not only on resting and activated CD4 and CD8 T cells but also on antigen-presenting cells. HHLA2 inhibits proliferation of both CD4 and CD8 T cells in the presence of T-cell receptor signaling. In addition, HHLA2 significantly reduces cytokine production by T cells including IFN-γ, TNF-α, IL-5, IL-10, IL-13, IL-17A, and IL-22. Thus, we have identified a unique B7 pathway that is able to inhibit human CD4 and CD8 T-cell proliferation and cytokine production. This unique human T-cell coinhibitory pathway may afford unique strategies for the treatment of human cancers, autoimmune disorders, infection, and transplant rejection and may help to design better vaccines.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoglobulinas/inmunología , Células 3T3 , Secuencia de Aminoácidos , Animales , Células Presentadoras de Antígenos/metabolismo , Linfocitos B/efectos de los fármacos , Linfocitos B/inmunología , Linfocitos B/metabolismo , Antígenos B7/genética , Antígenos B7/inmunología , Antígenos B7/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Citoplasma/metabolismo , Evolución Molecular , Citometría de Flujo , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos BALB C , Microscopía Confocal , Datos de Secuencia Molecular , Homología de Secuencia de Aminoácido
2.
J Am Chem Soc ; 133(23): 8810-3, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21473587

RESUMEN

The discovery of molecules that bind tightly and selectively to desired proteins continues to drive innovation at the interface of chemistry and biology. This paper describes the binding of human insulin by the synthetic receptor cucurbit[7]uril (Q7) in vitro. Isothermal titration calorimetry and fluorescence spectroscopy experiments show that Q7 binds to insulin with an equilibrium association constant of 1.5 × 10(6) M(-1) and with 50-100-fold selectivity versus proteins that are much larger but lack an N-terminal aromatic residue, and with >1000-fold selectivity versus an insulin variant lacking the N-terminal phenylalanine (Phe) residue. The crystal structure of the Q7·insulin complex shows that binding occurs at the N-terminal Phe residue and that the N-terminus unfolds to enable binding. These findings suggest that site-selective recognition is based on the properties inherent to a protein terminus, including the unique chemical epitope presented by the terminal residue and the greater freedom of the terminus to unfold, like the end of a ball of string, to accommodate binding. Insulin recognition was predicted accurately from studies on short peptides and exemplifies an approach to protein recognition by targeting the terminus.


Asunto(s)
Hidrocarburos Aromáticos con Puentes/metabolismo , Imidazoles/metabolismo , Insulina/metabolismo , Sitios de Unión , Humanos , Insulina/química , Modelos Moleculares , Fenilalanina/metabolismo , Unión Proteica , Conformación Proteica , Especificidad por Sustrato
3.
Sci Immunol ; 6(61)2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244312

RESUMEN

The B7 family ligand HERV-H LTR-associating protein 2 (HHLA2) is an attractive target for cancer immunotherapy because of its coinhibitory function, overexpression in human cancers, and association with poor prognoses. However, the knowledge of the HHLA2 pathway is incomplete. HHLA2 has an established positive receptor transmembrane and immunoglobulin (Ig) domain containing 2 (TMIGD2) but a poorly characterized negative receptor human killer cell Ig-like receptor, three Ig domains, and long cytoplasmic tail (KIR3DL3). Here, KIR3DL3 and TMIGD2 simultaneously bound to different sites of HHLA2. KIR3DL3 was mainly expressed on CD56dim NK and terminally differentiated effector memory CD8+ T (CD8+ TEMRA) cells. KIR3DL3+ CD8+ TEMRA acquired an NK-like phenotype and function. HHLA2 engagement recruited KIR3DL3 to the immunological synapse and coinhibited CD8+ T and NK cell function and killing, inducing immune-evasive HHLA2+ tumors. KIR3DL3 recruited SHP-1 and SHP-2 to attenuate Vav1, ERK1/2, AKT, and NF-κB signaling. HHLA2+ tumors from human kidney, lung, gallbladder, and stomach were infiltrated by KIR3DL3+ immune cells. KIR3DL3 blockade inhibited tumor growth in multiple humanized mouse models. Thus, our findings elucidated the molecular and cellular basis for the inhibitory function of KIR3DL3, demonstrating that the KIR3DL3-HHLA2 pathway is a potential immunotherapeutic target for cancer.


Asunto(s)
Inmunoglobulinas/inmunología , Neoplasias/inmunología , Receptores KIR/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Linfocitos T CD8-positivos/inmunología , Supervivencia Celular , Células Cultivadas , Humanos , Tolerancia Inmunológica , Inmunoglobulinas/genética , Células Asesinas Naturales/inmunología , Ratones Endogámicos BALB C , Neoplasias/tratamiento farmacológico , Receptores KIR/antagonistas & inhibidores
4.
Clin Cancer Res ; 23(3): 825-832, 2017 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-27553831

RESUMEN

PURPOSE: Immunotherapy with antibodies against B7/CD28 family members, including PD-1, PD-L1, and CTLA-4 has shifted the treatment paradigm for non-small cell lung carcinoma (NSCLC) with improved clinical outcome. HHLA2 is a newly discovered member of the family. By regulating T-cell function, HHLA2 could contribute to tumor immune suppression and thus be a novel target for cancer immunotherapy. There is limited information and critical need to characterize its expression profile and clinical significance in NSCLC. EXPERIMENTAL DESIGN: We performed IHC with an HHLA2-specific antibody (clone 566.1) using tissue microarrays constructed from 679 NSCLC tumor tissues, including 392 cases in the discovery set and 287 cases in the validation cohort. We also studied clinicopathologic characteristics of these patients. RESULTS: Overall, HHLA2 was not detected in most of normal lung tissue but expressed in 66% of NSCLC across different subtypes. In particular, EGFR-mutated NSCLC was significantly associated with higher tumor HHLA2 expression in both discovery (EGFR vs. WT: 76% vs. 53%, P = 0.01) and validation cohorts (89% vs. 69%, P = 0.01). In one of the two cohorts, HHLA2 expression was higher in lung adenocarcinoma as compared with squamous and large cell histology, non-Hispanic White versus Hispanics, and tumors with high tumor-infiltrating lymphocyte (TIL) density. In the multivariate analysis, EGFR mutation status and high TIL intensity were independently associated with HHLA2 expression in lung adenocarcinoma. CONCLUSIONS: HHLA2 is widely expressed in NSCLC and is associated with EGFR mutation and high TILs in lung adenocarcinoma. It is potentially a novel target for lung cancer immunotherapy. Clin Cancer Res; 23(3); 825-32. ©2016 AACR.


Asunto(s)
Adenocarcinoma/inmunología , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Genes erbB-1 , Inmunoglobulinas/análisis , Neoplasias Pulmonares/inmunología , Linfocitos Infiltrantes de Tumor/patología , Proteínas de Neoplasias/análisis , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Anciano , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/inmunología , Carcinoma de Células Grandes/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/metabolismo , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Inmunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Metástasis Linfática , Masculino , Persona de Mediana Edad , Terapia Molecular Dirigida , Mutación , Grupos Raciales/genética , Estudios Retrospectivos , Análisis de Matrices Tisulares
5.
Sci Rep ; 6: 30093, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27456063

RESUMEN

Osteosarcoma patient survival has remained stagnant for 30 years. Novel therapeutic approaches are needed to improve outcomes. We examined the expression of Programmed Death Ligand 1 (PD-L1) and defined the tumor immune microenvironment to assess the prognostic utility in osteosarcoma. PD-L1 expression in osteosarcoma was examined in two patient cohorts using immunohistochemistry (IHC) (n = 48, n = 59) and expression was validated using quantitative real time PCR (n = 21) and western blotting (n = 9). IHC was used to determine the presence of tumor infiltrating lymphocytes and antigen-presenting cells (APCs) in the tumor. Expression of PD-L1 was correlated with immune cell infiltration and event-free-survival (EFS). The 25% of primary osteosarcoma tumors that express PD-L1 were more likely to contain cells that express PD-1 than PD-L1 negative tumors (91.7% vs 47.2%, p = 0.002). Expression of PD-L1 was significantly associated with the presence of T cells, dendritic cells, and natural killer cells. Although all immune cell types examined were present in osteosarcoma samples, only infiltration by dendritic cells (28.3% vs. 83.9%, p = 0.001) and macrophages (45.5% vs. 84.4%, p = 0.031) were associated with worse five-year-EFS. PD-L1 expression was significantly associated with poorer five-year-EFS (25.0%. vs. 69.4%, p = 0.014). Further studies in osteosarcoma are needed to determine if targeting the PD-L1:PD-1 axis improves survival.


Asunto(s)
Antígeno B7-H1/biosíntesis , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Osteosarcoma/inmunología , Osteosarcoma/patología , Microambiente Tumoral/inmunología , Biomarcadores de Tumor/inmunología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/patología , Línea Celular Tumoral , Humanos , Células Asesinas Naturales/inmunología , Células MCF-7 , Macrófagos/inmunología , Osteosarcoma/mortalidad , Pronóstico
6.
Sci Rep ; 6: 31154, 2016 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-27531281

RESUMEN

Over the past four decades there have been minimal improvements in outcomes for patients with osteosarcoma. New targets and novel therapies are needed to improve outcomes for these patients. We sought to evaluate the prevalence and clinical significance of the newest immune checkpoint, HHLA2, in osteosarcoma. HHLA2 protein expression was evaluated in primary tumor specimens and metastatic disease using an osteosarcoma tumor microarray (TMA) (n = 62). The association of HHLA2 with the presence of tumor infiltrating lymphocytes (TILs) and five-year-event-free-survival were examined. HHLA2 was expressed in 68% of osteosarcoma tumors. HHLA2 was expressed in almost all metastatic disease specimens and was more prevalent than in primary specimens without known metastases (93% vs 53%, p = 0.02). TILs were present in 75% of all osteosarcoma specimens. Patients whose tumors were ≥25% or ≥50% HHLA2 positive had significantly worse five-year event-free-survival (33% vs 64%, p = 0.03 and 14% vs 59%, p = 0.02). Overall, we have shown that HHLA2 is expressed in the majority of osteosarcoma tumors and its expression is associated with metastatic disease and poorer survival. Along with previously reported findings that HHLA2 is a T cell co-inhibitor, these results suggest that HHLA2 may be a novel immunosuppressive mechanism within the osteosarcoma tumor microenvironment.


Asunto(s)
Neoplasias Óseas/inmunología , Inmunoglobulinas/metabolismo , Osteosarcoma/inmunología , Osteosarcoma/secundario , Adolescente , Adulto , Antígeno B7-H1/metabolismo , Biomarcadores de Tumor/inmunología , Neoplasias Óseas/patología , Niño , Supervivencia sin Enfermedad , Femenino , Humanos , Estimación de Kaplan-Meier , Linfocitos Infiltrantes de Tumor/patología , Masculino , Osteosarcoma/patología , Pronóstico , Análisis por Matrices de Proteínas , Adulto Joven
7.
Oncoimmunology ; 4(8): e1026534, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26405587

RESUMEN

We and others recently discovered HHLA2 as a new B7 family member and transmembrane and immunoglobulin domain containing 2 (TMIGD2) as one of its receptors. Based on a new study we propose that HHLA2 may represent a novel immunosuppressive mechanism within the tumor microenvironment and hence could be a target for cancer therapy. TMIGD2 may be another therapeutic target.

8.
Trends Pharmacol Sci ; 36(9): 587-95, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26162965

RESUMEN

Ligands from the B7 family bind to receptors of the CD28 family, which regulate early T cell activation in lymphoid organs and control inflammation and autoimmunity in peripheral tissues. Programmed death-1 (PD-1), a member of the CD28 family, is an inhibitory receptor on T cells and is responsible for their dysfunction in infectious diseases and cancers. The complex mechanisms controlling the expression and signaling of PD-1 and programmed death ligand 1 (PD-L1) are emerging. Recently completed and ongoing clinical trials that target these molecules have shown remarkable success by generating durable clinical responses in some cancer patients. In chronic viral infections, preclinical data reveal that targeting PD-1 and its ligands can improve T cell responses and virus clearance. There is also promise in stimulating this pathway for the treatment of autoimmune and inflammatory disorders.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Antineoplásicos/inmunología , Antivirales/inmunología , Inmunoterapia/métodos , Neoplasias/terapia , Receptor de Muerte Celular Programada 1/inmunología , Virosis/terapia , Animales , Anticuerpos Monoclonales/uso terapéutico , Antineoplásicos/uso terapéutico , Antivirales/uso terapéutico , Humanos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores
9.
Clin Cancer Res ; 21(10): 2359-66, 2015 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-25549724

RESUMEN

PURPOSE: HHLA2 (B7H7/B7-H5/B7y) is a newly identified B7 family member that regulates human T-cell functions. However, its protein expression in human organs and significance in human diseases are unknown. The objective of this study was to analyze HHLA2 protein expression in normal human tissues and cancers, as well as its prognostic significance, to explore mechanisms regulating HHLA2 expression, and to identify candidate HHLA2 receptors. EXPERIMENTAL DESIGN: An immunohistochemistry protocol and a flow cytometry assay with newly generated monoclonal antibodies were developed to examine HHLA2 protein. HHLA2 gene copy-number variation was analyzed from cancer genomic data. The combination of bioinformatics analysis and immunologic approaches was established to explore HHLA2 receptors. RESULTS: HHLA2 protein was detected in trophoblastic cells of the placenta and the epithelium of gut, kidney, gallbladder, and breast, but not in most other organs. In contrast, HHLA2 protein was widely expressed in human cancers from the breast, lung, thyroid, melanoma, pancreas, ovary, liver, bladder, colon, prostate, kidney, and esophagus. In a cohort of 50 patients with stage I-III triple-negative breast cancer, 56% of patients had aberrant expression of HHLA2 on their tumors, and high HHLA2 expression was significantly associated with regional lymph node metastasis and stage. The Cancer Genome Atlas revealed that HHLA2 copy-number gains were present in 29% of basal breast cancers, providing a potential mechanism for increased HHLA2 protein expression in breast cancer. Finally, Transmembrane and Immunoglobulin Domain Containing 2 (TMIGD2) was identified as one of the receptors for HHLA2. CONCLUSIONS: Wide expression of HHLA2 in human malignancies, together with its association with poor prognostic factors and its T-cell coinhibitory capability, suggests that the HHLA2 pathway represents a novel immunosuppressive mechanism within the tumor microenvironment and an attractive target for human cancer therapy.


Asunto(s)
Inmunoglobulinas/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Células 3T3 , Secuencia de Aminoácidos , Animales , Antígenos CD28/química , Antígenos CD28/metabolismo , Línea Celular Tumoral , Variaciones en el Número de Copia de ADN , Femenino , Expresión Génica , Glicosilación , Humanos , Inmunoglobulinas/genética , Metástasis Linfática , Ratones , Persona de Mediana Edad , Datos de Secuencia Molecular , Procesamiento Proteico-Postraduccional , Neoplasias de la Mama Triple Negativas/patología
10.
Cell Rep ; 9(3): 1089-98, 2014 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-25437562

RESUMEN

B7x (B7-H4 or B7S1) is a member of the B7 family that can inhibit T cell function. B7x protein is absent in most normal human tissues and immune cells, but it is overexpressed in human cancers and often correlates with negative clinical outcome. The expression pattern and function of B7x suggest that it may be a potent immunosuppressive pathway in human cancers. Here, we determined the crystal structure of the human B7x immunoglobulin variable (IgV) domain at 1.59 Å resolution and mapped the epitopes recognized by monoclonal antibodies. We developed an in vivo system to screen therapeutic monoclonal antibodies against B7x and found that the clone 1H3 significantly inhibited growth of B7x-expressing tumors in vivo via multiple mechanisms. Furthermore, the surviving mice given 1H3 treatment were resistant to tumor rechallenge. Our data suggest that targeting B7x on tumors is a promising cancer immunotherapy and humanized 1H3 may be efficacious for immunotherapy of human cancers.


Asunto(s)
Inmunoterapia , Neoplasias/inmunología , Neoplasias/terapia , Inhibidor 1 de la Activación de Células T con Dominio V-Set/química , Inhibidor 1 de la Activación de Células T con Dominio V-Set/inmunología , Animales , Anticuerpos Monoclonales/metabolismo , Anticuerpos Monoclonales/farmacología , Citotoxicidad Celular Dependiente de Anticuerpos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Mapeo Epitopo , Epítopos/química , Epítopos/inmunología , Humanos , Terapia de Inmunosupresión , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Ratones Endogámicos BALB C , Modelos Moleculares , Neoplasias/patología , Unión Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie , Análisis de Supervivencia , Linfocitos T/inmunología , Microambiente Tumoral/efectos de los fármacos , Inhibidor 1 de la Activación de Células T con Dominio V-Set/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA