Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Cell ; 165(5): 1092-1105, 2016 May 19.
Artículo en Inglés | MEDLINE | ID: mdl-27133165

RESUMEN

Effector T cells and fibroblasts are major components in the tumor microenvironment. The means through which these cellular interactions affect chemoresistance is unclear. Here, we show that fibroblasts diminish nuclear accumulation of platinum in ovarian cancer cells, resulting in resistance to platinum-based chemotherapy. We demonstrate that glutathione and cysteine released by fibroblasts contribute to this resistance. CD8(+) T cells abolish the resistance by altering glutathione and cystine metabolism in fibroblasts. CD8(+) T-cell-derived interferon (IFN)γ controls fibroblast glutathione and cysteine through upregulation of gamma-glutamyltransferases and transcriptional repression of system xc(-) cystine and glutamate antiporter via the JAK/STAT1 pathway. The presence of stromal fibroblasts and CD8(+) T cells is negatively and positively associated with ovarian cancer patient survival, respectively. Thus, our work uncovers a mode of action for effector T cells: they abrogate stromal-mediated chemoresistance. Capitalizing upon the interplay between chemotherapy and immunotherapy holds high potential for cancer treatment.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Resistencia a Antineoplásicos , Neoplasias Ováricas/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Cisplatino/uso terapéutico , Femenino , Fibroblastos/metabolismo , Glutatión/metabolismo , Humanos , Interferón gamma/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones Desnudos
2.
Immunity ; 51(4): 724-734.e4, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31586542

RESUMEN

HIV- and SIV-envelope (Env) trimers are both extensively glycosylated, and antibodies identified to date have been unable to fully neutralize SIVmac239. Here, we report the isolation, structure, and glycan interactions of antibody ITS90.03, a monoclonal antibody that completely neutralized the highly neutralization-resistant isolate, SIVmac239. The co-crystal structure of a fully glycosylated SIVmac239-gp120 core in complex with rhesus CD4 and the antigen-binding fragment of ITS90.03 at 2.5-Å resolution revealed that ITS90 recognized an epitope comprised of 45% glycan. SIV-gp120 core, rhesus CD4, and their complex could each be aligned structurally to their human counterparts. The structure revealed that glycans masked most of the SIV Env protein surface, with ITS90 targeting a glycan hole, which is occupied in ∼83% of SIV strains by glycan N238. Overall, the SIV glycan shield appears to functionally resemble its HIV counterpart in coverage of spike, shielding from antibody, and modulation of receptor accessibility.


Asunto(s)
Anticuerpos Monoclonales/química , Anticuerpos Neutralizantes/química , Infecciones por VIH/inmunología , VIH/fisiología , Polisacáridos/química , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/fisiología , Animales , Anticuerpos Monoclonales/aislamiento & purificación , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/aislamiento & purificación , Anticuerpos Neutralizantes/metabolismo , Antígenos CD4/metabolismo , Células Cultivadas , Cristalización , Cristalografía por Rayos X , Modelos Animales de Enfermedad , Glicosilación , Anticuerpos Anti-VIH/inmunología , Anticuerpos Anti-VIH/metabolismo , Proteína gp120 de Envoltorio del VIH/metabolismo , Humanos , Macaca mulatta , Glicoproteínas de Membrana/metabolismo , Polisacáridos/metabolismo , Unión Proteica , Relación Estructura-Actividad , Proteínas del Envoltorio Viral/metabolismo
3.
Immunity ; 50(3): 677-691.e13, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30876875

RESUMEN

Lineage-based vaccine design is an attractive approach for eliciting broadly neutralizing antibodies (bNAbs) against HIV-1. However, most bNAb lineages studied to date have features indicative of unusual recombination and/or development. From an individual in the prospective RV217 cohort, we identified three lineages of bNAbs targeting the membrane-proximal external region (MPER) of the HIV-1 envelope. Antibodies RV217-VRC42.01, -VRC43.01, and -VRC46.01 used distinct modes of recognition and neutralized 96%, 62%, and 30%, respectively, of a 208-strain virus panel. All three lineages had modest levels of somatic hypermutation and normal antibody-loop lengths and were initiated by the founder virus MPER. The broadest lineage, VRC42, was similar to the known bNAb 4E10. A multimeric immunogen based on the founder MPER activated B cells bearing the unmutated common ancestor of VRC42, with modest maturation of early VRC42 intermediates imparting neutralization breadth. These features suggest that VRC42 may be a promising template for lineage-based vaccine design.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Vacunas contra el SIDA/inmunología , Secuencia de Aminoácidos , Linfocitos B/inmunología , Línea Celular , Células HEK293 , Infecciones por VIH/inmunología , Humanos , Leucocitos Mononucleares , Estudios Longitudinales
4.
Nature ; 568(7752): 415-419, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30971821

RESUMEN

The HIV-1 envelope glycoprotein (Env) trimer mediates cell entry and is conformationally dynamic1-8. Imaging by single-molecule fluorescence resonance energy transfer (smFRET) has revealed that, on the surface of intact virions, mature pre-fusion Env transitions from a pre-triggered conformation (state 1) through a default intermediate conformation (state 2) to a conformation in which it is bound to three CD4 receptor molecules (state 3)8-10. It is currently unclear how these states relate to known structures. Breakthroughs in the structural characterization of the HIV-1 Env trimer have previously been achieved by generating soluble and proteolytically cleaved trimers of gp140 Env that are stabilized by a disulfide bond, an isoleucine-to-proline substitution at residue 559 and a truncation at residue 664 (SOSIP.664 trimers)5,11-18. Cryo-electron microscopy studies have been performed with C-terminally truncated Env of the HIV-1JR-FL strain in complex with the antibody PGT15119. Both approaches have revealed similar structures for Env. Although these structures have been presumed to represent the pre-triggered state 1 of HIV-1 Env, this hypothesis has never directly been tested. Here we use smFRET to compare the conformational states of Env trimers used for structural studies with native Env on intact virus. We find that the constructs upon which extant high-resolution structures are based predominantly occupy downstream conformations that represent states 2 and 3. Therefore, the structure of the pre-triggered state-1 conformation of viral Env that has been identified by smFRET and that is preferentially stabilized by many broadly neutralizing antibodies-and thus of interest for the design of immunogens-remains unknown.


Asunto(s)
Transferencia Resonante de Energía de Fluorescencia , VIH-1/química , Imagen Individual de Molécula , Productos del Gen env del Virus de la Inmunodeficiencia Humana/química , Animales , Anticuerpos Neutralizantes/inmunología , Bovinos , Disulfuros/química , Células HEK293 , VIH-1/genética , VIH-1/inmunología , Humanos , Modelos Moleculares , Mutación , Conformación Proteica , Multimerización de Proteína , Estabilidad Proteica , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología
5.
Immunity ; 40(5): 772-784, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24816405

RESUMEN

Little is known about how the immune system impacts human colorectal cancer invasiveness and stemness. Here we detected interleukin-22 (IL-22) in patient colorectal cancer tissues that was produced predominantly by CD4(+) T cells. In a mouse model, migration of these cells into the colon cancer microenvironment required the chemokine receptor CCR6 and its ligand CCL20. IL-22 acted on cancer cells to promote activation of the transcription factor STAT3 and expression of the histone 3 lysine 79 (H3K79) methytransferase DOT1L. The DOT1L complex induced the core stem cell genes NANOG, SOX2, and Pou5F1, resulting in increased cancer stemness and tumorigenic potential. Furthermore, high DOT1L expression and H3K79me2 in colorectal cancer tissues was a predictor of poor patient survival. Thus, IL-22(+) cells promote colon cancer stemness via regulation of stemness genes that negatively affects patient outcome. Efforts to target this network might be a strategy in treating colorectal cancer patients.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Neoplasias Colorrectales/inmunología , Interleucinas/inmunología , Metiltransferasas/inmunología , Células Madre Neoplásicas/inmunología , Factor de Transcripción STAT3/inmunología , Animales , Línea Celular Tumoral , Proliferación Celular , Quimiocina CCL20/inmunología , Quimiocina CCL20/metabolismo , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Activación Enzimática/inmunología , Células HT29 , N-Metiltransferasa de Histona-Lisina , Proteínas de Homeodominio/inmunología , Proteínas de Homeodominio/metabolismo , Humanos , Metiltransferasas/metabolismo , Ratones , Proteína Homeótica Nanog , Trasplante de Neoplasias , Células Madre Neoplásicas/patología , Factor 3 de Transcripción de Unión a Octámeros/inmunología , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Receptores CCR6/inmunología , Receptores CCR6/metabolismo , Factores de Transcripción SOXB1/inmunología , Factores de Transcripción SOXB1/metabolismo , Factor de Transcripción STAT3/metabolismo , Interleucina-22
6.
Nature ; 527(7577): 249-53, 2015 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-26503055

RESUMEN

Epigenetic silencing including histone modifications and DNA methylation is an important tumorigenic mechanism. However, its role in cancer immunopathology and immunotherapy is poorly understood. Using human ovarian cancers as our model, here we show that enhancer of zeste homologue 2 (EZH2)-mediated histone H3 lysine 27 trimethylation (H3K27me3) and DNA methyltransferase 1 (DNMT1)-mediated DNA methylation repress the tumour production of T helper 1 (TH1)-type chemokines CXCL9 and CXCL10, and subsequently determine effector T-cell trafficking to the tumour microenvironment. Treatment with epigenetic modulators removes the repression and increases effector T-cell tumour infiltration, slows down tumour progression, and improves the therapeutic efficacy of programmed death-ligand 1 (PD-L1; also known as B7-H1) checkpoint blockade and adoptive T-cell transfusion in tumour-bearing mice. Moreover, tumour EZH2 and DNMT1 are negatively associated with tumour-infiltrating CD8(+) T cells and patient outcome. Thus, epigenetic silencing of TH1-type chemokines is a novel immune-evasion mechanism of tumours. Selective epigenetic reprogramming alters the T-cell landscape in cancer and may enhance the clinical efficacy of cancer therapy.


Asunto(s)
Quimiocinas/genética , Epigénesis Genética , Silenciador del Gen , Inmunoterapia , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/terapia , Células TH1/metabolismo , Animales , Antígeno B7-H1/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Quimiocina CXCL10/biosíntesis , Quimiocina CXCL10/genética , Quimiocina CXCL10/inmunología , Quimiocina CXCL9/biosíntesis , Quimiocina CXCL9/genética , Quimiocina CXCL9/inmunología , Quimiocinas/biosíntesis , Quimiocinas/inmunología , ADN (Citosina-5-)-Metiltransferasa 1 , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Metilación de ADN/efectos de los fármacos , Proteína Potenciadora del Homólogo Zeste 2 , Epigénesis Genética/efectos de los fármacos , Femenino , Histonas/química , Histonas/metabolismo , Humanos , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Lisina/metabolismo , Ratones , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/patología , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/metabolismo , Pronóstico , Células TH1/inmunología , Células Tumorales Cultivadas , Escape del Tumor/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
J Immunol ; 186(10): 5638-47, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21471442

RESUMEN

Earlier, we demonstrated the essential role of Kruppel-like transcription factor, TIEG1, in TGF-ß-induced regulatory T cell (Treg) development. In this article, we demonstrate that IL-6, which promotes Th17 development, abrogated TIEG1 nuclear translocation and inhibited TGF-ß-induced Treg development. Tyrosine kinase Tyk2-mediated phosphorylation of TIEG1 at Tyr179 promoted noncanonical K-27-linked polyubiquitination, which inhibited TIEG1 nuclear translocation. To test the role of TIEG1-regulated Treg/Th17 development in antitumor immunity, we analyzed TRAMP-C2 tumor growth in TIEG1(-/-) mice. The defective Treg development and elevated Th17 response resulted in enhanced immune reactivity in the tumor and inhibition of TRAMP-C2 tumor growth in TIEG1(-/-) mice. Thus, our results uncovered a novel regulatory mechanism that modulates Tregs and may regulate tumor progression.


Asunto(s)
Proliferación Celular , Proteínas de Unión al ADN/metabolismo , Factores de Transcripción Forkhead/genética , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/patología , Factores de Transcripción/metabolismo , Transporte Activo de Núcleo Celular , Traslado Adoptivo , Animales , Western Blotting , Núcleo Celular/metabolismo , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Inmunoprecipitación , Interleucina-6/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación , Reacción en Cadena de la Polimerasa , Receptores de Antígenos de Linfocitos T/inmunología , Transducción de Señal , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , TYK2 Quinasa/metabolismo , Células Th17/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Ubiquitinación
8.
Biochem Biophys Res Commun ; 394(3): 600-5, 2010 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-20214883

RESUMEN

The mechanism of cisplatin resistance in cancer cells is not fully understood. Here, we show that the Akt/mTOR survival pathway plays an important role in cisplatin resistance in human ovarian cancer cells. Specifically, we found that cisplatin treatment activates the Akt/mTOR survival pathway and that inhibition of this pathway by the PI3K inhibitor LY294002 or knockdown of Akt sensitizes ovarian cancer cells to cisplatin. Furthermore, we generated cisplatin-resistant cells and found that resistant cells express a higher level of activated Akt as compared to their cisplatin sensitive counterparts. Importantly, inhibition of Akt or mTOR sensitized resistant cells to cisplatin-induced apoptosis. Taken together, our data indicate that activation of the Akt/mTOR pathway prevents cisplatin-induced apoptosis, leading to cisplatin resistance. Therefore, our study suggests that cisplatin resistance can be overcome by targeting the Akt/mTOR survival pathway in human ovarian cancer cells.


Asunto(s)
Antineoplásicos/farmacología , Cisplatino/farmacología , Resistencia a Antineoplásicos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Apoptosis , Línea Celular Tumoral , Supervivencia Celular , Cromonas/farmacología , Técnicas de Silenciamiento del Gen , Humanos , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Péptidos y Proteínas de Señalización Intracelular/genética , Morfolinas/farmacología , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Serina-Treonina Quinasas TOR
9.
Int J Cancer ; 124(12): 2973-81, 2009 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-19326452

RESUMEN

Apoptin protein harbors tumor-selective cell death activity, which makes it a potential anticancer therapy candidate. This study reports an apoptin therapy approach based on protein transduction domain 4 (PTD4)-mediated transduction of recombinant apoptin protein. In vitro, the PTD4-apoptin fusion protein is located in the nucleus and induces cell death in, e.g., human hepatocarcinoma HepG2 cells. In normal human L-02 hepatocytes, PTD4-apoptin protein retained mainly cytoplasmic and did not induce detectable levels of cell death, illustrating that the PTD4 domain does not affect apoptin's tumor-selective characteristics. In vivo, liver, cervix and gastric carcinoma xenografts treated with PTD4-apoptin protein for 6 days via the tumor epidermis exhibited a significant tumor growth inhibition because of apoptin-mediated cell death. In addition, treatment of human hepatocarcinoma xenografts during 3 weeks showed that PTD4-apoptin protein has significant anticancer activity, whereas control treatment with PTD4-enhanced green fluorescence protein or saline did not. Cell death and disruption of the tumor integrity were apparent in the PTD4-apoptin transduced xenografted tumors. As important, although PTD4-apoptin protein could be detected in the epidermal tissue covering the subcutaneous tumor tissue and in several organs, such as liver and brain, of the treated mice, no tissue disruption or signs of cell death could be detected. Our in vivo data reveal that apoptin protein delivery constitutes a novel powerful and safe anticancer therapy.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Proteínas de la Cápside/metabolismo , Carcinoma Hepatocelular/terapia , Proteínas Portadoras/metabolismo , Neoplasias Hepáticas/terapia , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Proteínas de la Cápside/genética , Carcinoma Hepatocelular/patología , Proteínas Portadoras/genética , Núcleo Celular/metabolismo , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Femenino , Citometría de Flujo , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Etiquetado Corte-Fin in Situ , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Señales de Localización Nuclear , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología , Neoplasias Gástricas/terapia , Fracciones Subcelulares , Neoplasias del Cuello Uterino/metabolismo , Neoplasias del Cuello Uterino/patología , Neoplasias del Cuello Uterino/terapia
10.
Pathog Immun ; 4(2): 294-323, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31893251

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) subtypes, A and B, co-circulate in annual epidemics and alternate in dominance. We have shown that a subtype A RSV fusion (F) glycoprotein, stabilized in its prefusion conformation by DS-Cav1 mutations, is a promising RSV-vaccine immunogen, capable of boosting RSV-neutralizing titers in healthy adults. In both humans and vaccine-tested animals, neutralizing titers elicited by this subtype A DS-Cav1 immunogen were ~ 2- to 3-fold higher against the homologous subtype A virus than against the heterologous subtype B virus. METHODS: To understand the molecular basis for this subtype difference, we introduced DS-Cav1 mutations into RSV strain B18537 F, determined the trimeric crystal structure, and carried out immunogenicity studies. RESULTS: The B18537 DS-Cav1 F structure at 2-Å resolution afforded a precise delineation of prefusion F characteristics, including those of antigenic site Ø, a key trimer-apex site. Structural comparison with the subtype A prefusion F indicated 11% of surface residues to be different, with an alpha-carbon root-mean-square deviation (RMSD) of 1.2 Å; antigenic site Ø, however, differed in 23% of its surface residues and had an alpha-carbon RMSD of 2.2 Å. Immunization of vaccine-tested animals with DS-Cav1-stabilized B18537 F induced neutralizing responses ~100-fold higher than with postfusion B18537 F. Notably, elicited responses neutralized RSV subtypes A and B at similar levels and were directed towards both conserved equatorial and diverse apical regions. CONCLUSION: We propose that structural differences in apical and equatorial sites-coupled to differently focused immune responses-provide a molecular explanation for observed differences in elicited subtype A and B neutralizing responses.

11.
Nat Commun ; 10(1): 47, 2019 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-30604750

RESUMEN

Diverse entry inhibitors targeting the gp120 subunit of the HIV-1 envelope (Env) trimer have been developed including BMS-626529, also called temsavir, a prodrug version of which is currently in phase III clinical trials. Here we report the characterization of a panel of small-molecule inhibitors including BMS-818251, which we show to be >10-fold more potent than temsavir on a cross-clade panel of 208-HIV-1 strains, as well as the engineering of a crystal lattice to enable structure determination of the interaction between these inhibitors and the HIV-1 Env trimer at higher resolution. By altering crystallization lattice chaperones, we identify a lattice with both improved diffraction and robust co-crystallization of HIV-1 Env trimers from different clades complexed to entry inhibitors with a range of binding affinities. The improved diffraction reveals BMS-818251 to utilize functional groups that interact with gp120 residues from the conserved ß20-ß21 hairpin to improve potency.


Asunto(s)
Ingeniería Química/métodos , Proteína gp120 de Envoltorio del VIH/antagonistas & inhibidores , Inhibidores de Fusión de VIH/farmacología , VIH-1/efectos de los fármacos , Internalización del Virus/efectos de los fármacos , Cristalografía por Rayos X , Diseño de Fármacos , Proteína gp120 de Envoltorio del VIH/metabolismo , Inhibidores de Fusión de VIH/química , VIH-1/fisiología , Células HeLa , Humanos , Simulación del Acoplamiento Molecular , Nanopartículas/química , Piperazinas/química , Piperazinas/farmacología , Unión Proteica , Conformación Proteica , Multimerización de Proteína , Triazoles/química , Triazoles/farmacología
12.
Nat Med ; 24(6): 857-867, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29867235

RESUMEN

A central goal of HIV-1 vaccine research is the elicitation of antibodies capable of neutralizing diverse primary isolates of HIV-1. Here we show that focusing the immune response to exposed N-terminal residues of the fusion peptide, a critical component of the viral entry machinery and the epitope of antibodies elicited by HIV-1 infection, through immunization with fusion peptide-coupled carriers and prefusion stabilized envelope trimers, induces cross-clade neutralizing responses. In mice, these immunogens elicited monoclonal antibodies capable of neutralizing up to 31% of a cross-clade panel of 208 HIV-1 strains. Crystal and cryoelectron microscopy structures of these antibodies revealed fusion peptide conformational diversity as a molecular explanation for the cross-clade neutralization. Immunization of guinea pigs and rhesus macaques induced similarly broad fusion peptide-directed neutralizing responses, suggesting translatability. The N terminus of the HIV-1 fusion peptide is thus a promising target of vaccine efforts aimed at eliciting broadly neutralizing antibodies.


Asunto(s)
Vacunas contra el SIDA/inmunología , Anticuerpos Neutralizantes/inmunología , Epítopos/inmunología , Anticuerpos Anti-VIH/inmunología , VIH-1/inmunología , Péptidos/farmacología , Proteínas Recombinantes de Fusión/farmacología , Secuencia de Aminoácidos , Animales , Femenino , Cobayas , VIH-1/efectos de los fármacos , Inmunización , Macaca mulatta , Ratones Endogámicos C57BL , Modelos Moleculares , Pruebas de Neutralización , Productos del Gen env del Virus de la Inmunodeficiencia Humana/química , Productos del Gen env del Virus de la Inmunodeficiencia Humana/metabolismo
13.
Oncoimmunology ; 5(8): e1082704, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27622053

RESUMEN

Th22 cells traffic to and retain in the colon cancer microenvironment, and target core stem cell genes and promote colon cancer stemness via STAT3 and H3K79me2 signaling pathway and contribute to colon carcinogenesis. However, whether Th22 cells affect colon cancer cell proliferation and apoptosis remains unknown. We studied the interaction between Th22 cells and colon cancer cells in the colon cancer microenvironment. Colon cancer proliferation was examined by flow cytometry analysis and H(3) thymidine incorporation. Cell cycle related genes were quantified by real-time PCR and Western blotting. We transfected colon cancer cells with lentiviral vector encoding specific gene shRNAs and used chromatin immunoprecipitation (ChIP) assay to determine the genetic signaling involved in interleukin (IL)-22-mediated colon cancer cell proliferation. We showed that Th22 cells released IL-22 and stimulated colon cancer proliferation. Mechanistically, IL-22 activated STAT3, and subsequently STAT3 bound to the promoter areas of the Polycomb Repression complex 2 (PRC2) components SUZ12 and EED, and stimulated the expression of PRC2. Consequently, the activated PRC2 catalyzed the promoters of the cell cycle check-point genes p16 and p21, and inhibited their expression through H3K27me3-mediated histone methylation, and ultimately caused colon cancer cell proliferation. Bioinformatics analysis revealed that the levels of IL-22 expression positively correlated with the levels of genes controlling cancer proliferation and cell cycling in colon cancer. In addition to controlling colon cancer stemness, Th22 cells support colon carcinogenesis via affecting colon cancer cell proliferation through a distinct histone modification.

14.
Cancer Res ; 76(2): 275-82, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26567139

RESUMEN

Infiltration of tumors with effector T cells is positively associated with therapeutic efficacy and patient survival. However, the mechanisms underlying effector T-cell trafficking to the tumor microenvironment remain poorly understood in patients with colon cancer. The polycomb repressive complex 2 (PRC2) is involved in cancer progression, but the regulation of tumor immunity by epigenetic mechanisms has yet to be investigated. In this study, we examined the relationship between the repressive PRC2 machinery and effector T-cell trafficking. We found that PRC2 components and demethylase JMJD3-mediated histone H3 lysine 27 trimethylation (H3K27me3) repress the expression and subsequent production of Th1-type chemokines CXCL9 and CXCL10, mediators of effector T-cell trafficking. Moreover, the expression levels of PRC2 components, including EZH2, SUZ12, and EED, were inversely associated with those of CD4, CD8, and Th1-type chemokines in human colon cancer tissue, and this expression pattern was significantly associated with patient survival. Collectively, our findings reveal that PRC2-mediated epigenetic silencing is not only a crucial oncogenic mechanism, but also a key circuit controlling tumor immunosuppression. Therefore, targeting epigenetic programs may have significant implications for improving the efficacy of current cancer immunotherapies relying on effective T-cell-mediated immunity at the tumor site.


Asunto(s)
Quimiocinas/inmunología , Neoplasias del Colon/genética , Complejo Represivo Polycomb 2/genética , Linfocitos T/inmunología , Células TH1/inmunología , Movimiento Celular/genética , Movimiento Celular/inmunología , Proliferación Celular/genética , Neoplasias del Colon/inmunología , Neoplasias del Colon/patología , Epigénesis Genética , Humanos , Complejo Represivo Polycomb 2/inmunología , Linfocitos T/metabolismo , Células TH1/metabolismo , Transfección
15.
Cancer Res ; 76(11): 3156-65, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27197152

RESUMEN

Myeloid-derived suppressor cells (MDSC) contribute to immune suppression in cancer, but the mechanisms through which they drive metastatic progression are not fully understood. In this study, we show how MDSC convey stem-like qualities to breast cancer cells that coordinately help enable immune suppression and escape. We found that MDSC promoted tumor formation by enhancing breast cancer cell stem-like properties as well as by suppressing T-cell activation. Mechanistic investigations indicated that these effects relied upon cross-talk between the STAT3 and NOTCH pathways in cancer cells, with MDSC inducing IL6-dependent phosphorylation of STAT3 and activating NOTCH through nitric oxide leading to prolonged STAT3 activation. In clinical specimens of breast cancer, the presence of MDSC correlated with the presence of cancer stem-like cells (CSC) and independently predicted poor survival outcomes. Collectively, our work revealed an immune-associated mechanism that extrinsically confers cancer cell stemness properties and affects patient outcome. We suggest that targeting STAT3-NOTCH cross-talk between MDSC and CSC could offer a unique locus to improve cancer treatment, by coordinately targeting a coupled mechanism that enables cancer stemness and immune escape. Cancer Res; 76(11); 3156-65. ©2016 AACR.


Asunto(s)
Neoplasias de la Mama/patología , Interleucina-6/metabolismo , Células Supresoras de Origen Mieloide/patología , Células Madre Neoplásicas/patología , Óxido Nítrico/metabolismo , Receptores Notch/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Apoptosis , Western Blotting , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Proliferación Celular , Estudios de Cohortes , Femenino , Citometría de Flujo , Estudios de Seguimiento , Humanos , Técnicas para Inmunoenzimas , Interleucina-6/genética , Activación de Linfocitos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Células Supresoras de Origen Mieloide/metabolismo , Estadificación de Neoplasias , Células Madre Neoplásicas/metabolismo , Fosforilación , Pronóstico , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Cross-Talk , Receptores Notch/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Transducción de Señal , Tasa de Supervivencia , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
16.
J Oncol ; 2012: 345164, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22481922

RESUMEN

Multiple layers of suppressive components including regulatory T (T(Reg)) cells, suppressive antigen-presenting cells, and inhibitory cytokines form suppressive networks in the ovarian cancer microenvironment. It has been demonstrated that as a major suppressive element, T(Reg) cells infiltrate tumor, interact with several types of immune cells, and mediate immune suppression through different molecular and cellular mechanisms. In this paper, we focus on human ovarian cancer and will discuss the nature of T(Reg) cells including their subsets, trafficking, expansion, and function. We will briefly review the development of manipulation of T(Reg) cells in preclinical and clinical settings.

17.
Cell Cycle ; 9(23): 4650-5, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-21084841

RESUMEN

MKP-2 is a member of the dual-specificity phosphatase family that can dephosphorylate and inactivate mitogen-activated protein kinases (MAPKs). Although MKP-2 can be induced by ERK signaling, little is known about the regulation of MKP-2 at the post-translational level. Here we show that MKP-2 is phosphorylated by ERK and that such phosphorylation leads to stabilization of MKP-2 protein. Importantly, we find that MKP-2 can be phosphorylated on Ser386 and Ser391 at its C-terminus. Blockage of ERK activation results in enhanced proteasomal degradation of MKP-2 protein. Moreover, we find that phosphorylation has no effect on MKP-2 phosphatase activity. Taken together, these results illustrate an important post-translational regulation of MKP-2 protein as a feedback mechanism to control ERK activity.


Asunto(s)
Fosfatasas de Especificidad Dual/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/metabolismo , Dominio Catalítico , Línea Celular Tumoral , Fosfatasas de Especificidad Dual/genética , Humanos , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Mitógenos/farmacología , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA