Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.304
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 20(11): 1494-1505, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31611701

RESUMEN

Immunotherapy has transformed cancer treatment. However, current immunotherapy modalities face various limitations. In the present study, we developed multiplexed activation of endogenous genes as an immunotherapy (MAEGI), a new form of immunotherapy that elicits antitumor immunity through multiplexed activation of endogenous genes in tumors. We leveraged CRISPR activation (CRISPRa) to directly augment the in situ expression of endogenous genes, and thereby the presentation of tumor antigens, leading to dramatic antitumor immune responses. Deploying this as a cell-based vaccination strategy showed efficacy in both prophylactic and therapeutic settings. Intratumoral adeno-associated virus delivery of CRISPRa libraries elicited strong antitumor immunity across multiple cancer types. Precision targeting of mutated gene sets eradicated a large fraction of established tumors at both local and distant sites. This treatment modality led to alterations in the tumor microenvironment, marked by enhanced T cell infiltration and antitumor immune signatures. Multiplexed endogenous gene activation is a versatile and highly scalable strategy to elicit potent immune responses against cancer, distinct from all existing cancer therapies.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Regulación Neoplásica de la Expresión Génica/inmunología , Terapia Genética/métodos , Inmunoterapia/métodos , Neoplasias/tratamiento farmacológico , Animales , Presentación de Antígeno/genética , Antígenos de Neoplasias/inmunología , Vacunas contra el Cáncer/administración & dosificación , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Técnicas de Cocultivo , Terapia Combinada/métodos , Dependovirus/genética , Modelos Animales de Enfermedad , Femenino , Vectores Genéticos/administración & dosificación , Vectores Genéticos/genética , Células HEK293 , Humanos , Inyecciones Intralesiones , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Neoplasias/genética , Neoplasias/inmunología , Linfocitos T Citotóxicos/inmunología , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
2.
Nat Immunol ; 19(3): 255-266, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29476183

RESUMEN

Key events in T cell-dependent antibody responses, including affinity maturation, are dependent on the B cell's presentation of antigen to helper T cells at critical checkpoints in germinal-center formation in secondary lymphoid organs. Here we found that signaling via Toll-like receptor 9 (TLR9) blocked the ability of antigen-specific B cells to capture, process and present antigen and to activate antigen-specific helper T cells in vitro. In a mouse model in vivo and in a human clinical trial, the TLR9 agonist CpG enhanced the magnitude of the antibody response to a protein vaccine but failed to promote affinity maturation. Thus, TLR9 signaling might enhance antibody titers at the expense of the ability of B cells to engage in germinal-center events that are highly dependent on B cells' capture and presentation of antigen.


Asunto(s)
Formación de Anticuerpos/inmunología , Presentación de Antígeno/genética , Activación de Linfocitos/inmunología , Receptor Toll-Like 9/inmunología , Animales , Afinidad de Anticuerpos , Centro Germinal/inmunología , Humanos , Vacunas contra la Malaria , Ratones , Receptor Toll-Like 9/agonistas
3.
Immunity ; 54(4): 753-768.e5, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33765435

RESUMEN

Viral infections induce a conserved host response distinct from bacterial infections. We hypothesized that the conserved response is associated with disease severity and is distinct between patients with different outcomes. To test this, we integrated 4,780 blood transcriptome profiles from patients aged 0 to 90 years infected with one of 16 viruses, including SARS-CoV-2, Ebola, chikungunya, and influenza, across 34 cohorts from 18 countries, and single-cell RNA sequencing profiles of 702,970 immune cells from 289 samples across three cohorts. Severe viral infection was associated with increased hematopoiesis, myelopoiesis, and myeloid-derived suppressor cells. We identified protective and detrimental gene modules that defined distinct trajectories associated with mild versus severe outcomes. The interferon response was decoupled from the protective host response in patients with severe outcomes. These findings were consistent, irrespective of age and virus, and provide insights to accelerate the development of diagnostics and host-directed therapies to improve global pandemic preparedness.


Asunto(s)
Inmunidad/genética , Virosis/inmunología , Presentación de Antígeno/genética , Estudios de Cohortes , Hematopoyesis/genética , Humanos , Interferones/sangre , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/patología , Células Mieloides/inmunología , Células Mieloides/patología , Pronóstico , Índice de Severidad de la Enfermedad , Biología de Sistemas , Transcriptoma , Virosis/sangre , Virosis/clasificación , Virosis/genética , Virus/clasificación , Virus/patogenicidad
4.
Nature ; 629(8010): 193-200, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38600383

RESUMEN

Sex differences in mammalian complex traits are prevalent and are intimately associated with androgens1-7. However, a molecular and cellular profile of sex differences and their modulation by androgens is still lacking. Here we constructed a high-dimensional single-cell transcriptomic atlas comprising over 2.3 million cells from 17 tissues in Mus musculus and explored the effects of sex and androgens on the molecular programs and cellular populations. In particular, we found that sex-biased immune gene expression and immune cell populations, such as group 2 innate lymphoid cells, were modulated by androgens. Integration with the UK Biobank dataset revealed potential cellular targets and risk gene enrichment in antigen presentation for sex-biased diseases. This study lays the groundwork for understanding the sex differences orchestrated by androgens and provides important evidence for targeting the androgen pathway as a broad therapeutic strategy for sex-biased diseases.


Asunto(s)
Andrógenos , Células , Caracteres Sexuales , Análisis de la Célula Individual , Transcriptoma , Animales , Femenino , Humanos , Masculino , Ratones , Andrógenos/metabolismo , Andrógenos/farmacología , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/genética , Inmunidad Innata , Linfocitos/metabolismo , Linfocitos/citología , Linfocitos/inmunología , Linfocitos/efectos de los fármacos , Ratones Endogámicos C57BL , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Biobanco del Reino Unido , Células/efectos de los fármacos , Células/inmunología , Células/metabolismo
5.
Nat Immunol ; 18(2): 161-172, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27941786

RESUMEN

Aire is a transcriptional regulator that induces promiscuous expression of thousands of genes encoding tissue-restricted antigens (TRAs) in medullary thymic epithelial cells (mTECs). While the target genes of Aire are well characterized, the transcriptional programs that regulate its own expression have remained elusive. Here we comprehensively analyzed both cis-acting and trans-acting regulatory mechanisms and found that the Aire locus was insulated by the global chromatin organizer CTCF and was hypermethylated in cells and tissues that did not express Aire. In mTECs, however, Aire expression was facilitated by concurrent eviction of CTCF, specific demethylation of exon 2 and the proximal promoter, and the coordinated action of several transcription activators, including Irf4, Irf8, Tbx21, Tcf7 and Ctcfl, which acted on mTEC-specific accessible regions in the Aire locus.


Asunto(s)
Células Epiteliales/inmunología , Redes Reguladoras de Genes , Linfocitos T/fisiología , Timo/inmunología , Factores de Transcripción/metabolismo , Animales , Presentación de Antígeno/genética , Autoantígenos/metabolismo , Factor de Unión a CCCTC , Diferenciación Celular , Células Cultivadas , Selección Clonal Mediada por Antígenos , Metilación de ADN , Regulación de la Expresión Génica , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Timo/citología , Factores de Transcripción/genética , Proteína AIRE
6.
Nat Immunol ; 17(8): 938-45, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27294792

RESUMEN

The cells that stimulate positive selection express specialized proteasome ß-subunits different from those expressed by all other cells, including those involved in negative selection. Mice that lack all four specialized proteasome ß-subunits, and therefore express only constitutive proteasomes in all cells, had a profound defect in the generation of CD8(+) T cells. While a defect in positive selection would reflect an inability to generate the appropriate positively selecting peptides, a block at negative selection would point to the potential need to switch peptides between positive selection and negative selection to avoid the two processes' often cancelling each other out. We found that the block in T cell development occurred around the checkpoints of positive selection and, unexpectedly, negative selection as well.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Selección Clonal Mediada por Antígenos , Cisteína Endopeptidasas/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Timo/inmunología , Animales , Presentación de Antígeno/genética , Diferenciación Celular , Células Cultivadas , Cisteína Endopeptidasas/genética , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Péptidos/metabolismo , Complejo de la Endopetidasa Proteasomal/genética
7.
Nat Immunol ; 17(10): 1206-1215, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27548434

RESUMEN

Thymic epithelial cell differentiation, growth and function depend on the expression of the transcription factor Foxn1; however, its target genes have never been physically identified. Using static and inducible genetic model systems and chromatin studies, we developed a genome-wide map of direct Foxn1 target genes for postnatal thymic epithelia and defined the Foxn1 binding motif. We determined the function of Foxn1 in these cells and found that, in addition to the transcriptional control of genes involved in the attraction and lineage commitment of T cell precursors, Foxn1 regulates the expression of genes involved in antigen processing and thymocyte selection. Thus, critical events in thymic lympho-stromal cross-talk and T cell selection are indispensably choreographed by Foxn1.


Asunto(s)
Células Epiteliales/fisiología , Factores de Transcripción Forkhead/metabolismo , Células Precursoras de Linfocitos T/fisiología , Linfocitos T/fisiología , Timo/fisiología , Animales , Presentación de Antígeno/genética , Comunicación Celular , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Selección Clonal Mediada por Antígenos/genética , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Genoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos
8.
PLoS Genet ; 20(6): e1011309, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38885192

RESUMEN

PLK1 (Polo-like kinase 1) plays a critical role in the progression of lung adenocarcinoma (LUAD). Recent studies have unveiled that targeting PLK1 improves the efficacy of immunotherapy, highlighting its important role in the regulation of tumor immunity. Nevertheless, our understanding of the intricate interplay between PLK1 and the tumor microenvironment (TME) remains incomplete. Here, using genetically engineered mouse model and single-cell RNA-seq analysis, we report that PLK1 promotes an immunosuppressive TME in LUAD, characterized with enhanced M2 polarization of tumor associated macrophages (TAM) and dampened antigen presentation process. Mechanistically, elevated PLK1 coincides with increased secretion of CXCL2 cytokine, which promotes M2 polarization of TAM and diminishes expression of class II major histocompatibility complex (MHC-II) in professional antigen-presenting cells. Furthermore, PLK1 negatively regulates MHC-II expression in cancer cells, which has been shown to be associated with compromised tumor immunity and unfavorable patient outcomes. Taken together, our results reveal PLK1 as a novel modulator of TME in LUAD and provide possible therapeutic interventions.


Asunto(s)
Adenocarcinoma del Pulmón , Proteínas de Ciclo Celular , Neoplasias Pulmonares , Quinasa Tipo Polo 1 , Proteínas Serina-Treonina Quinasas , Proteínas Proto-Oncogénicas , Análisis de la Célula Individual , Microambiente Tumoral , Animales , Humanos , Ratones , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/patología , Presentación de Antígeno/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Macrófagos Asociados a Tumores/inmunología , Macrófagos Asociados a Tumores/metabolismo
9.
J Cell Sci ; 137(10)2024 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-38682259

RESUMEN

SARS-CoV-2 interferes with antigen presentation by downregulating major histocompatibility complex (MHC) II on antigen-presenting cells, but the mechanism mediating this process is unelucidated. Herein, analysis of protein and gene expression in human antigen-presenting cells reveals that MHC II is downregulated by the SARS-CoV-2 main protease, NSP5. This suppression of MHC II expression occurs via decreased expression of the MHC II regulatory protein CIITA. CIITA downregulation is independent of the proteolytic activity of NSP5, and rather, NSP5 delivers HDAC2 to the transcription factor IRF3 at an IRF-binding site within the CIITA promoter. Here, HDAC2 deacetylates and inactivates the CIITA promoter. This loss of CIITA expression prevents further expression of MHC II, with this suppression alleviated by ectopic expression of CIITA or knockdown of HDAC2. These results identify a mechanism by which SARS-CoV-2 limits MHC II expression, thereby delaying or weakening the subsequent adaptive immune response.


Asunto(s)
Antígenos de Histocompatibilidad Clase II , Histona Desacetilasa 2 , Proteínas Nucleares , Regiones Promotoras Genéticas , SARS-CoV-2 , Transactivadores , Humanos , Presentación de Antígeno/genética , Células Presentadoras de Antígenos/metabolismo , Células Presentadoras de Antígenos/inmunología , COVID-19/virología , COVID-19/inmunología , COVID-19/genética , COVID-19/metabolismo , Cisteína Endopeptidasas/metabolismo , Cisteína Endopeptidasas/genética , Regulación hacia Abajo/genética , Células HEK293 , Antígenos de Histocompatibilidad Clase II/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Histona Desacetilasa 2/metabolismo , Histona Desacetilasa 2/genética , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Proteínas Nucleares/metabolismo , Proteínas Nucleares/genética , Regiones Promotoras Genéticas/genética , SARS-CoV-2/genética , SARS-CoV-2/metabolismo , SARS-CoV-2/inmunología , Transactivadores/metabolismo , Transactivadores/genética , Proteínas no Estructurales Virales/metabolismo , Proteínas no Estructurales Virales/genética
10.
Nat Immunol ; 15(2): 161-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24362890

RESUMEN

CD11b(+) dendritic cells (DCs) seem to be specialized for presenting antigens via major histocompatibility (MHC) class II complexes to stimulate helper T cells, but the genetic and regulatory basis for this is not established. Conditional deletion of Irf4 resulted in loss of CD11b(+) DCs, impaired formation of peptide-MHC class II complexes and defective priming of helper T cells but not of cytotoxic T lymphocyte (CTL) responses. Gene expression and chromatin immunoprecipitation followed by deep sequencing (ChIP-Seq) analyses delineated an IRF4-dependent regulatory module that programs enhanced MHC class II antigen presentation. Expression of the transcription factor IRF4 but not of IRF8 restored the ability of IRF4-deficient DCs to efficiently process and present antigen to MHC class II-restricted T cells and promote helper T cell responses. We propose that the evolutionary divergence of IRF4 and IRF8 facilitated the specialization of DC subsets for distinct modes of antigen presentation and priming of helper T cell versus CTL responses.


Asunto(s)
Presentación de Antígeno/genética , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Factores Reguladores del Interferón/metabolismo , Linfocitos T Citotóxicos/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Células Cultivadas , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Antígenos de Histocompatibilidad Clase II/genética , Factores Reguladores del Interferón/genética , Activación de Linfocitos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Unión Proteica/genética , Transgenes/genética
11.
Immunity ; 47(2): 310-322.e7, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28813660

RESUMEN

Select humans and animals control persistent viral infections via adaptive immune responses that include production of neutralizing antibodies. The precise genetic basis for the control remains enigmatic. Here, we report positional cloning of the gene responsible for production of retrovirus-neutralizing antibodies in mice of the I/LnJ strain. It encodes the beta subunit of the non-classical major histocompatibility complex class II (MHC-II)-like molecule H2-O, a negative regulator of antigen presentation. The recessive and functionally null I/LnJ H2-Ob allele supported the production of virus-neutralizing antibodies independently of the classical MHC haplotype. Subsequent bioinformatics and functional analyses of the human H2-Ob homolog, HLA-DOB, revealed both loss- and gain-of-function alleles, which could affect the ability of their carriers to control infections with human hepatitis B (HBV) and C (HCV) viruses. Thus, understanding of the previously unappreciated role of H2-O (HLA-DO) in immunity to infections may suggest new approaches in achieving neutralizing immunity to viruses.


Asunto(s)
Anticuerpos Neutralizantes , Antígenos HLA-D/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Inmunidad Humoral , Virus del Tumor Mamario del Ratón/inmunología , Virus Rauscher/inmunología , Infecciones por Retroviridae/inmunología , Animales , Anticuerpos Neutralizantes/metabolismo , Anticuerpos Antivirales/metabolismo , Presentación de Antígeno/genética , Biología Computacional , Femenino , Predisposición Genética a la Enfermedad , Antígenos HLA-D/genética , Células HeLa , Hepatitis B/inmunología , Hepatitis B/transmisión , Hepatitis C/inmunología , Hepatitis C/transmisión , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Inmunidad Humoral/genética , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Mutación/genética , Polimorfismo Genético , Infecciones por Retroviridae/transmisión
12.
Proc Natl Acad Sci U S A ; 120(39): e2305756120, 2023 09 26.
Artículo en Inglés | MEDLINE | ID: mdl-37722062

RESUMEN

Mutations in RNA/DNA-binding proteins cause amyotrophic lateral sclerosis (ALS), but the underlying disease mechanisms remain unclear. Here, we report that a set of ALS-associated proteins, namely FUS, EWSR1, TAF15, and MATR3, impact the expression of genes encoding the major histocompatibility complex II (MHC II) antigen presentation pathway. Both subunits of the MHC II heterodimer, HLA-DR, are down-regulated in ALS gene knockouts/knockdown in HeLa and human microglial cells, due to loss of the MHC II transcription factor CIITA. Importantly, hematopoietic progenitor cells (HPCs) derived from human embryonic stem cells bearing the FUSR495X mutation and HPCs derived from C9ORF72 ALS patient induced pluripotent stem cells also exhibit disrupted MHC II expression. Given that HPCs give rise to numerous immune cells, our data raise the possibility that loss of the MHC II pathway results in global failure of the immune system to protect motor neurons from damage that leads to ALS.


Asunto(s)
Esclerosis Amiotrófica Lateral , Humanos , Esclerosis Amiotrófica Lateral/genética , Presentación de Antígeno/genética , Genes MHC Clase II , Complejo Mayor de Histocompatibilidad , Neuronas Motoras , Proteínas de Unión al ARN/genética , Proteínas Asociadas a Matriz Nuclear
13.
Nat Immunol ; 14(5): 446-53, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23502855

RESUMEN

Spleen-resident dendritic cell (DC) populations occupy sentinel positions for the capture and presentation of blood-borne antigens. Here we found a difference in expression of the chemotactic receptor EBI2 (GPR183) on splenic DC subsets and that EBI2 regulated the positioning and homeostasis of DCs in the spleen. EBI2 and its main ligand, 7α,25-OHC, were required for the generation of the splenic CD4(+) DC subset and the localization of DCs in bridging channels. Absence of EBI2 from DCs resulted in defects in both the activation of CD4(+) T cells and the induction of antibody responses. Regulated expression of EBI2 on DC populations is therefore critical for the generation and correct positioning of splenic DCs and the initiation of immune responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Quimiotaxis/inmunología , Células Dendríticas/inmunología , Activación de Linfocitos , Receptores Acoplados a Proteínas G/metabolismo , Bazo/inmunología , Animales , Formación de Anticuerpos/genética , Presentación de Antígeno/genética , Antígenos CD4/metabolismo , Movimiento Celular/genética , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/patología , Homeostasis/genética , Homeostasis/inmunología , Activación de Linfocitos/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptores Acoplados a Proteínas G/genética , Bazo/patología
14.
Nat Immunol ; 14(9): 908-16, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23872678

RESUMEN

Human T cells that express a T cell antigen receptor (TCR) containing γ-chain variable region 9 and δ-chain variable region 2 (Vγ9Vδ2) recognize phosphorylated prenyl metabolites as antigens in the presence of antigen-presenting cells but independently of major histocompatibility complex (MHC), the MHC class I-related molecule MR1 and antigen-presenting CD1 molecules. Here we used genetic approaches to identify the molecule that binds and presents phosphorylated antigens. We found that the butyrophilin BTN3A1 bound phosphorylated antigens with low affinity, at a stoichiometry of 1:1, and stimulated mouse T cells with transgenic expression of a human Vγ9Vδ2 TCR. The structures of the BTN3A1 distal domain in complex with host- or microbe-derived phosphorylated antigens had an immunoglobulin-like fold in which the antigens bound in a shallow pocket. Soluble Vγ9Vδ2 TCR interacted specifically with BTN3A1-antigen complexes. Accordingly, BTN3A1 represents an antigen-presenting molecule required for the activation of Vγ9Vδ2 T cells.


Asunto(s)
Antígenos CD/metabolismo , Antígenos/inmunología , Antígenos/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Células Presentadoras de Antígenos/metabolismo , Antígenos CD/química , Antígenos CD/genética , Butirofilinas , Cromosomas Humanos Par 6 , Humanos , Ratones , Ratones Transgénicos , Modelos Moleculares , Organofosfatos/química , Organofosfatos/metabolismo , Fosforilación , Unión Proteica , Conformación Proteica , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología
15.
Nature ; 576(7787): 465-470, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31827286

RESUMEN

Tumour-infiltrating lymphocytes are associated with a survival benefit in several tumour types and with the response to immunotherapy1-8. However, the reason some tumours have high CD8 T cell infiltration while others do not remains unclear. Here we investigate the requirements for maintaining a CD8 T cell response against human cancer. We find that CD8 T cells within tumours consist of distinct populations of terminally differentiated and stem-like cells. On proliferation, stem-like CD8 T cells give rise to more terminally differentiated, effector-molecule-expressing daughter cells. For many T cells to infiltrate the tumour, it is critical that this effector differentiation process occur. In addition, we show that these stem-like T cells reside in dense antigen-presenting-cell niches within the tumour, and that tumours that fail to form these structures are not extensively infiltrated by T cells. Patients with progressive disease lack these immune niches, suggesting that niche breakdown may be a key mechanism of immune escape.


Asunto(s)
Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias/inmunología , Células Madre/citología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Linfocitos T CD8-positivos/metabolismo , Progresión de la Enfermedad , Epigénesis Genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Neoplasias/patología , Nicho de Células Madre/inmunología , Transcripción Genética , Escape del Tumor/genética , Escape del Tumor/inmunología
16.
J Cell Mol Med ; 28(8): e18309, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38613345

RESUMEN

There are hundreds of prognostic models for ovarian cancer. These genes are based on different gene classes, and there are many ways to construct the models. Therefore, this paper aims to build the most stable prognostic evaluation system known to date through 101 machine learning strategies. We combined 101 algorithm combinations with 10 machine learning algorithms to create antigen presentation-associated genetic markers (AIDPS) with outstanding precision and steady performance. The inclusive set of algorithms comprises the elastic network (Enet), Ridge, stepwise Cox, Lasso, generalized enhanced regression model (GBM), random survival forest (RSF), supervised principal component (SuperPC), Cox partial least squares regression (plsRcox), survival support vector machine (Survival-SVM). Then, in the train cohort, the prediction model was fitted using a leave-one cross-validation (LOOCV) technique, which involved 101 different possible combinations of prognostic genes. Seven validation data sets (GSE26193, GSE26712, GSE30161, GSE63885, GSE9891, GSE140082 and ICGC_OV_AU) were compared and analysed, and the C-index was calculated. Finally, we collected 32 published ovarian cancer prognostic models (including mRNA and lncRNA). All data sets and prognostic models were subjected to a univariate Cox regression analysis, and the C-index was calculated to demonstrate that the antigen presentation process should be the core criterion for evaluating ovarian cancer prognosis. In a univariate Cox regression analysis, 22 prognostic genes were identified based on the expression profiles of 283 genes involved in antigen presentation and the intersection of genes (p < 0.05). AIDPS were developed by our machine learning-based integration method, which was applied to these 22 genes. One hundred and one prediction models are fitted using the LOOCV framework, and the C-index is calculated for each model across all validation sets. Interestingly, RSF + Lasso was the best model overall since it had the greatest average C-index and the highest C-index of any combination of models tested on the validated data sets. In comparing external cohorts, we found that the C-index correlated AIDPS method using the RSF + Lasso method in 101 prediction models was in contrast to other features. Notably, AIDPS outperformed the vast majority of models across all data sets. Antigen-presenting anti-tumour immune pathways can be used as a representative gene set of ovarian cancer to track the prognosis of patients with cancer. The antigen-presenting model obtained by the RSF + Lasso method has the best C-INDEX, which plays a key role in developing antigen-presenting targeted drugs in ovarian cancer and improving the treatment outcome of patients.


Asunto(s)
Presentación de Antígeno , Neoplasias Ováricas , Humanos , Femenino , Presentación de Antígeno/genética , Neoplasias Ováricas/genética , Algoritmos , Sistemas de Liberación de Medicamentos
17.
J Transl Med ; 22(1): 57, 2024 01 14.
Artículo en Inglés | MEDLINE | ID: mdl-38221616

RESUMEN

BACKGROUND: Cancer-associated fibroblasts (CAFs) are heterogeneous and can influence the progression of prostate cancer in multiple ways; however, their capacity to present and process antigens in PRAD has not been investigated. In this study, antigen presentation and process-related CAFs (APPCAFs) were identified using bioinformatics, and the clinical implications of APPCAF-related signatures in PRAD were investigated. METHODS: SMART technology was used to sequence the transcriptome of primary CAFs isolated from patients undergoing different treatments. Differential expression gene (DEG) screening was conducted. A CD4 + T-cell early activation assay was used to assess the activation degree of CD4 + T cells. The datasets of PRAD were obtained from The Cancer Genome Atlas (TCGA) database and NCBI Gene Expression Omnibus (GEO), and the list of 431 antigen presentation and process-related genes was obtained from the InnateDB database. Subsequently, APP-related CAFs were identified by nonnegative matrix factorization (NMF) based on a single-cell seq (scRNA) matrix. GSVA functional enrichment analyses were performed to depict the biological functions. A risk signature based on APPCAF-related genes (APPCAFRS) was developed by least absolute shrinkage and selection operator (LASSO) regression analysis, and the independence of the risk score as a prognostic factor was evaluated by univariate and multivariate Cox regression analyses. Furthermore, a biochemical recurrence-free survival (BCRFS)-related nomogram was established, and immune-related characteristics were assessed using the ssGSEA function. The immune treatment response in PRAD was further analyzed by the Tumor Immune Dysfunction and Exclusion (TIDE) tool. The expression levels of hub genes in APPCAFRS were verified in cell models. RESULTS: There were 134 upregulated and 147 downregulated genes, totaling 281 differentially expressed genes among the primary CAFs. The functions and pathways of 147 downregulated DEGs were significantly enriched in antigen processing and presentation processes, MHC class II protein complex and transport vesicle, MHC class II protein complex binding, and intestinal immune network for IgA production. Androgen withdrawal diminished the activation effect of CAFs on T cells. NMF clustering of CAFs was performed by APPRGs, and pseudotime analysis yielded the antigen presentation and process-related CAF subtype CTSK + MRC2 + CAF-C1. CTSK + MRC2 + CAF-C1 cells exhibited ligand‒receptor connections with epithelial cells and T cells. Additionally, we found a strong association between CTSK + MRC2 + CAF-C1 cells and inflammatory CAFs. Through differential gene expression analysis of the CTSK + MRC2 + CAF-C1 and NoneAPP-CAF-C2 subgroups, 55 significant DEGs were identified, namely, APPCAFRGs. Based on the expression profiles of APPCAFRGs, we divided the TCGA-PRAD cohort into two clusters using NMF consistent cluster analysis, with the genetic coefficient serving as the evaluation index. Four APPCAFRGs, THBS2, DPT, COL5A1, and MARCKS, were used to develop a prognostic signature capable of predicting BCR occurrence in PRAD patients. Subsequently, a nomogram with stability and accuracy in predicting BCR was constructed based on Gleason grade (p = n.s.), PSA (p < 0.001), T stage (p < 0.05), and risk score (p < 0.01). The analysis of immune infiltration showed a positive correlation between the abundance of resting memory CD4 + T cells, M1 macrophages, resting dendritic cells, and the risk score. In addition, the mRNA expression levels of THBS2, DPT, COL5A1, and MARCKS in the cell models were consistent with the results of the bioinformatics analysis. CONCLUSIONS: APPCAFRS based on four potential APPCAFRGs was developed, and their interaction with the immune microenvironment may play a crucial role in the progression to castration resistance of PRAD. This novel approach provides valuable insights into the pathogenesis of PRAD and offers unexplored targets for future research.


Asunto(s)
Fibroblastos Asociados al Cáncer , Neoplasias de la Próstata , Masculino , Humanos , Presentación de Antígeno/genética , Análisis de Secuencia de ARN , Algoritmos , Pronóstico , Microambiente Tumoral
18.
Immunity ; 42(6): 1048-61, 2015 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-26070482

RESUMEN

Thymic antigen-presenting cells (APCs) such as dendritic cells and medullary thymic epithelial cells (mTECs) use distinct strategies of self-antigen expression and presentation to mediate central tolerance. The thymus also harbors B cells; whether they also display unique tolerogenic features and how they genealogically relate to peripheral B cells is unclear. Here, we found that Aire is expressed in thymic but not peripheral B cells. Aire expression in thymic B cells coincided with major histocompatibility class II (MHCII) and CD80 upregulation and immunoglobulin class-switching. These features were recapitulated upon immigration of naive peripheral B cells into the thymus, whereby this intrathymic licensing required CD40 signaling in the context of cognate interactions with autoreactive CD4(+) thymocytes. Moreover, a licensing-dependent neo-antigen selectively upregulated in immigrating B cells mediated negative selection through direct presentation. Thus, autoreactivity within the nascent T cell repertoire fuels a feed forward loop that endows thymic B cells with tolerogenic features.


Asunto(s)
Linfocitos B/fisiología , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/metabolismo , Timo/inmunología , Factores de Transcripción/metabolismo , Animales , Presentación de Antígeno/genética , Autoantígenos/inmunología , Antígeno B7-1/genética , Antígeno B7-1/metabolismo , Antígenos CD40/genética , Diferenciación Celular/genética , Células Cultivadas , Tolerancia Central/genética , Selección Clonal Mediada por Antígenos/genética , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Cambio de Clase de Inmunoglobulina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Transducción de Señal , Factores de Transcripción/genética , Proteína AIRE
19.
PLoS Biol ; 19(4): e3001057, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33901176

RESUMEN

Viral diseases pose major threats to humans and other animals, including the billions of chickens that are an important food source as well as a public health concern due to zoonotic pathogens. Unlike humans and other typical mammals, the major histocompatibility complex (MHC) of chickens can confer decisive resistance or susceptibility to many viral diseases. An iconic example is Marek's disease, caused by an oncogenic herpesvirus with over 100 genes. Classical MHC class I and class II molecules present antigenic peptides to T lymphocytes, and it has been hard to understand how such MHC molecules could be involved in susceptibility to Marek's disease, given the potential number of peptides from over 100 genes. We used a new in vitro infection system and immunopeptidomics to determine peptide motifs for the 2 class II molecules expressed by the MHC haplotype B2, which is known to confer resistance to Marek's disease. Surprisingly, we found that the vast majority of viral peptide epitopes presented by chicken class II molecules arise from only 4 viral genes, nearly all having the peptide motif for BL2*02, the dominantly expressed class II molecule in chickens. We expressed BL2*02 linked to several Marek's disease virus (MDV) peptides and determined one X-ray crystal structure, showing how a single small amino acid in the binding site causes a crinkle in the peptide, leading to a core binding peptide of 10 amino acids, compared to the 9 amino acids in all other reported class II molecules. The limited number of potential T cell epitopes from such a complex virus can explain the differential MHC-determined resistance to MDV, but raises questions of mechanism and opportunities for vaccine targets in this important food species, as well as providing a basis for understanding class II molecules in other species including humans.


Asunto(s)
Pollos/inmunología , Herpesvirus Gallináceo 2/inmunología , Antígenos de Histocompatibilidad Clase II , Enfermedad de Marek/inmunología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Bolsa de Fabricio/inmunología , Células Cultivadas , Pollos/genética , Pollos/virología , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Haplotipos , Herpesvirus Gallináceo 2/química , Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Epítopos Inmunodominantes/química , Epítopos Inmunodominantes/genética , Epítopos Inmunodominantes/inmunología , Epítopos Inmunodominantes/metabolismo , Enfermedad de Marek/genética , Enfermedad de Marek/virología , Modelos Moleculares , Péptidos/química , Péptidos/genética , Péptidos/inmunología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/virología , Proteínas Virales/química , Proteínas Virales/genética , Proteínas Virales/inmunología
20.
Int J Mol Sci ; 25(13)2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-39000461

RESUMEN

Antigen presentation is a crucial mechanism that drives the T cell-mediated immune response and the development of Multiple Sclerosis (MS). Genetic alterations within the highly variable Major Histocompatibility Complex Class II (MHC II) have been proven to result in significant changes in the molecular basis of antigen presentation and the clinical course of patients with both Adult-Onset MS (AOMS) and Pediatric-Onset MS (POMS). Among the numerous polymorphisms of the Human Leucocyte Antigens (HLA), within MHC II complex, HLA-DRB1*15:01 has been labeled, in Caucasian ethnic groups, as a high-risk allele for MS due to the ability of its structure to increase affinity to Myelin Basic Protein (MBP) epitopes. This characteristic, among others, in the context of the trimolecular complex or immunological synapsis, provides the foundation for autoimmunity triggered by environmental or endogenous factors. As with all professional antigen presenting cells, macrophages are characterized by the expression of MHC II and are often implicated in the formation of MS lesions. Increased presence of M1 macrophages in MS patients has been associated both with progression and onset of the disease, each involving separate but similar mechanisms. In this critical narrative review, we focus on macrophages, discussing how HLA genetic alterations can promote dysregulation of this population's homeostasis in the periphery and the Central Nervous System (CNS). We also explore the potential interconnection in observed pathological macrophage mechanisms and the function of the diverse structure of HLA alleles in neurodegenerative CNS, seen in MS, by comparing available clinical with molecular data through the prism of HLA-immunogenetics. Finally, we discuss available and experimental pharmacological approaches for MS targeting the trimolecular complex that are based on cell phenotype modulation and HLA genotype involvement and try to reveal fertile ground for the potential development of novel drugs.


Asunto(s)
Alelos , Macrófagos , Esclerosis Múltiple , Humanos , Esclerosis Múltiple/genética , Esclerosis Múltiple/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Antígenos de Histocompatibilidad Clase II/inmunología , Presentación de Antígeno/genética , Predisposición Genética a la Enfermedad , Animales , Polimorfismo Genético
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA