Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Nat Immunol ; 23(10): 1495-1506, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36151395

RESUMEN

The immune system can eliminate tumors, but checkpoints enable immune escape. Here, we identify immune evasion mechanisms using genome-scale in vivo CRISPR screens across cancer models treated with immune checkpoint blockade (ICB). We identify immune evasion genes and important immune inhibitory checkpoints conserved across cancers, including the non-classical major histocompatibility complex class I (MHC class I) molecule Qa-1b/HLA-E. Surprisingly, loss of tumor interferon-γ (IFNγ) signaling sensitizes many models to immunity. The immune inhibitory effects of tumor IFN sensing are mediated through two mechanisms. First, tumor upregulation of classical MHC class I inhibits natural killer cells. Second, IFN-induced expression of Qa-1b inhibits CD8+ T cells via the NKG2A/CD94 receptor, which is induced by ICB. Finally, we show that strong IFN signatures are associated with poor response to ICB in individuals with renal cell carcinoma or melanoma. This study reveals that IFN-mediated upregulation of classical and non-classical MHC class I inhibitory checkpoints can facilitate immune escape.


Asunto(s)
Linfocitos T CD8-positivos , Neoplasias , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico , Evasión Inmune , Interferón gamma/genética , Interferón gamma/metabolismo , Subfamília C de Receptores Similares a Lectina de Células NK
2.
Nat Immunol ; 22(8): 1020-1029, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34312547

RESUMEN

T cell exhaustion is an induced state of dysfunction that arises in response to chronic infection and cancer. Exhausted CD8+ T cells acquire a distinct epigenetic state, but it is not known whether that chromatin landscape is fixed or plastic following the resolution of a chronic infection. Here we show that the epigenetic state of exhaustion is largely irreversible, even after curative therapy. Analysis of chromatin accessibility in HCV- and HIV-specific responses identifies a core epigenetic program of exhaustion in CD8+ T cells, which undergoes only limited remodeling before and after resolution of infection. Moreover, canonical features of exhaustion, including super-enhancers near the genes TOX and HIF1A, remain 'epigenetically scarred.' T cell exhaustion is therefore a conserved epigenetic state that becomes fixed and persists independent of chronic antigen stimulation and inflammation. Therapeutic efforts to reverse T cell exhaustion may require new approaches that increase the epigenetic plasticity of exhausted T cells.


Asunto(s)
Antígenos Virales/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Hepacivirus/inmunología , Hepatitis C Crónica/inmunología , Memoria Inmunológica/inmunología , 2-Naftilamina/uso terapéutico , Anilidas/uso terapéutico , Antivirales/uso terapéutico , Cromatina/metabolismo , Ciclopropanos/uso terapéutico , Epigénesis Genética/genética , Hepacivirus/efectos de los fármacos , Hepatitis C Crónica/tratamiento farmacológico , Proteínas del Grupo de Alta Movilidad/genética , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Lactamas Macrocíclicas/uso terapéutico , Prolina/análogos & derivados , Prolina/uso terapéutico , Ribavirina/uso terapéutico , Ritonavir/uso terapéutico , Sulfonamidas/uso terapéutico , Uracilo/análogos & derivados , Uracilo/uso terapéutico , Valina/uso terapéutico
3.
Nat Immunol ; 20(10): 1335-1347, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31527834

RESUMEN

CD8+ T cell exhaustion is a state of dysfunction acquired in chronic viral infection and cancer, characterized by the formation of Slamf6+ progenitor exhausted and Tim-3+ terminally exhausted subpopulations through unknown mechanisms. Here we establish the phosphatase PTPN2 as a new regulator of the differentiation of the terminally exhausted subpopulation that functions by attenuating type 1 interferon signaling. Deletion of Ptpn2 in CD8+ T cells increased the generation, proliferative capacity and cytotoxicity of Tim-3+ cells without altering Slamf6+ numbers during lymphocytic choriomeningitis virus clone 13 infection. Likewise, Ptpn2 deletion in CD8+ T cells enhanced Tim-3+ anti-tumor responses and improved tumor control. Deletion of Ptpn2 throughout the immune system resulted in MC38 tumor clearance and improved programmed cell death-1 checkpoint blockade responses to B16 tumors. Our results indicate that increasing the number of cytotoxic Tim-3+CD8+ T cells can promote effective anti-tumor immunity and implicate PTPN2 in immune cells as an attractive cancer immunotherapy target.


Asunto(s)
Adenocarcinoma/inmunología , Linfocitos T CD8-positivos/fisiología , Neoplasias del Colon/inmunología , Inmunoterapia/métodos , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Células Progenitoras Linfoides/fisiología , Melanoma/inmunología , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Neoplasias Cutáneas/inmunología , Animales , Senescencia Celular , Citotoxicidad Inmunológica , Femenino , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Tolerancia Inmunológica , Interferón Tipo I/metabolismo , Masculino , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Transducción de Señal , Familia de Moléculas Señalizadoras de la Activación Linfocitaria/metabolismo
4.
Nat Immunol ; 20(12): 1668-1680, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31636464

RESUMEN

Lymph node fibroblastic reticular cells (FRCs) respond to signals from activated T cells by releasing nitric oxide, which inhibits T cell proliferation and restricts the size of the expanding T cell pool. Whether interactions with FRCs also support the function or differentiation of activated CD8+ T cells is not known. Here we report that encounters with FRCs enhanced cytokine production and remodeled chromatin accessibility in newly activated CD8+ T cells via interleukin-6. These epigenetic changes facilitated metabolic reprogramming and amplified the activity of pro-survival pathways through differential transcription factor activity. Accordingly, FRC conditioning significantly enhanced the persistence of virus-specific CD8+ T cells in vivo and augmented their differentiation into tissue-resident memory T cells. Our study demonstrates that FRCs play a role beyond restricting T cell expansion-they can also shape the fate and function of CD8+ T cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Fibroblastos/fisiología , Ganglios Linfáticos/inmunología , Animales , Diferenciación Celular , Proliferación Celular , Supervivencia Celular , Células Cultivadas , Reprogramación Celular , Ensamble y Desensamble de Cromatina , Citotoxicidad Inmunológica , Epigénesis Genética , Regulación de la Expresión Génica , Memoria Inmunológica , Interleucina-6/genética , Interleucina-6/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Óxido Nítrico/metabolismo
6.
Nat Immunol ; 20(3): 326-336, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30778252

RESUMEN

T cell dysfunction is a hallmark of many cancers, but the basis for T cell dysfunction and the mechanisms by which antibody blockade of the inhibitory receptor PD-1 (anti-PD-1) reinvigorates T cells are not fully understood. Here we show that such therapy acts on a specific subpopulation of exhausted CD8+ tumor-infiltrating lymphocytes (TILs). Dysfunctional CD8+ TILs possess canonical epigenetic and transcriptional features of exhaustion that mirror those seen in chronic viral infection. Exhausted CD8+ TILs include a subpopulation of 'progenitor exhausted' cells that retain polyfunctionality, persist long term and differentiate into 'terminally exhausted' TILs. Consequently, progenitor exhausted CD8+ TILs are better able to control tumor growth than are terminally exhausted T cells. Progenitor exhausted TILs can respond to anti-PD-1 therapy, but terminally exhausted TILs cannot. Patients with melanoma who have a higher percentage of progenitor exhausted cells experience a longer duration of response to checkpoint-blockade therapy. Thus, approaches to expand the population of progenitor exhausted CD8+ T cells might be an important component of improving the response to checkpoint blockade.


Asunto(s)
Anticuerpos Bloqueadores/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Melanoma Experimental/prevención & control , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Animales , Anticuerpos Bloqueadores/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Línea Celular Tumoral , Femenino , Humanos , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Subgrupos Linfocitarios/virología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/virología , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/prevención & control , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/efectos de los fármacos , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/fisiología , Melanoma Experimental/inmunología , Melanoma Experimental/virología , Ratones Congénicos , Ratones Endogámicos C57BL , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo
7.
Nat Immunol ; 19(8): 838-848, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29988091

RESUMEN

Foxo transcription factors play an essential role in regulating specialized lymphocyte functions and in maintaining T cell quiescence. Here, we used a system in which Foxo1 transcription-factor activity, which is normally terminated upon cell activation, cannot be silenced, and we show that enforcing Foxo1 activity disrupts homeostasis of CD4 conventional and regulatory T cells. Despite limiting cell metabolism, continued Foxo1 activity is associated with increased activation of the kinase Akt and a cell-intrinsic proliferative advantage; however, survival and cell division are decreased in a competitive setting or growth-factor-limiting conditions. Via control of expression of the transcription factor Myc and the IL-2 receptor ß-chain, termination of Foxo1 signaling couples the increase in cellular cholesterol to biomass accumulation after activation, thereby facilitating immunological synapse formation and mTORC1 activity. These data reveal that Foxo1 regulates the integration of metabolic and mitogenic signals essential for T cell competitive fitness and the coordination of cell growth with cell division.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Proteína Forkhead Box O1/metabolismo , Linfocitos T Reguladores/fisiología , Animales , Proliferación Celular , Células Cultivadas , Colesterol/metabolismo , Proteína Forkhead Box O1/genética , Perfilación de la Expresión Génica , Homeostasis , Sinapsis Inmunológicas/metabolismo , Subunidad beta del Receptor de Interleucina-2/genética , Subunidad beta del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Transducción de Señal
8.
Immunity ; 54(3): 571-585.e6, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33497609

RESUMEN

CRISPR-Cas9 genome engineering has increased the pace of discovery for immunology and cancer biology, revealing potential therapeutic targets and providing insight into mechanisms underlying resistance to immunotherapy. However, endogenous immune recognition of Cas9 has limited the applicability of CRISPR technologies in vivo. Here, we characterized immune responses against Cas9 and other expressed CRISPR vector components that cause antigen-specific tumor rejection in several mouse cancer models. To avoid unwanted immune recognition, we designed a lentiviral vector system that allowed selective CRISPR antigen removal (SCAR) from tumor cells. The SCAR system reversed immune-mediated rejection of CRISPR-modified tumor cells in vivo and enabled high-throughput genetic screens in previously intractable models. A pooled in vivo screen using SCAR in a CRISPR-antigen-sensitive renal cell carcinoma revealed resistance pathways associated with autophagy and major histocompatibility complex class I (MHC class I) expression. Thus, SCAR presents a resource that enables CRISPR-based studies of tumor-immune interactions and prevents unwanted immune recognition of genetically engineered cells, with implications for clinical applications.


Asunto(s)
Carcinoma de Células Renales/inmunología , Pruebas Genéticas/métodos , Vectores Genéticos/genética , Inmunoterapia/métodos , Neoplasias Renales/inmunología , Células Asesinas Naturales/inmunología , Lentivirus/genética , Animales , Presentación de Antígeno , Autofagia , Carcinoma de Células Renales/terapia , Células Cultivadas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Ingeniería Genética , Antígenos de Histocompatibilidad Clase I/metabolismo , Neoplasias Renales/terapia , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Terapia Molecular Dirigida
9.
Nature ; 615(7950): 158-167, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36634707

RESUMEN

Despite the success of PD-1 blockade in melanoma and other cancers, effective treatment strategies to overcome resistance to cancer immunotherapy are lacking1,2. Here we identify the innate immune kinase TANK-binding kinase 1 (TBK1)3 as a candidate immune-evasion gene in a pooled genetic screen4. Using a suite of genetic and pharmacological tools across multiple experimental model systems, we confirm a role for TBK1 as an immune-evasion gene. Targeting TBK1 enhances responses to PD-1 blockade by decreasing the cytotoxicity threshold to effector cytokines (TNF and IFNγ). TBK1 inhibition in combination with PD-1 blockade also demonstrated efficacy using patient-derived tumour models, with concordant findings in matched patient-derived organotypic tumour spheroids and matched patient-derived organoids. Tumour cells lacking TBK1 are primed to undergo RIPK- and caspase-dependent cell death in response to TNF and IFNγ in a JAK-STAT-dependent manner. Taken together, our results demonstrate that targeting TBK1 is an effective strategy to overcome resistance to cancer immunotherapy.


Asunto(s)
Resistencia a Antineoplásicos , Evasión Inmune , Inmunoterapia , Proteínas Serina-Treonina Quinasas , Humanos , Evasión Inmune/genética , Evasión Inmune/inmunología , Inmunoterapia/métodos , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Organoides , Factores de Necrosis Tumoral/inmunología , Interferón gamma/inmunología , Esferoides Celulares , Caspasas , Quinasas Janus , Factores de Transcripción STAT
10.
Nature ; 595(7866): 309-314, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33953401

RESUMEN

Epigenetic dysregulation is a defining feature of tumorigenesis that is implicated in immune escape1,2. Here, to identify factors that modulate the immune sensitivity of cancer cells, we performed in vivo CRISPR-Cas9 screens targeting 936 chromatin regulators in mouse tumour models treated with immune checkpoint blockade. We identified the H3K9 methyltransferase SETDB1 and other members of the HUSH and KAP1 complexes as mediators of immune escape3-5. We also found that amplification of SETDB1 (1q21.3) in human tumours is associated with immune exclusion and resistance to immune checkpoint blockade. SETDB1 represses broad domains, primarily within the open genome compartment. These domains are enriched for transposable elements (TEs) and immune clusters associated with segmental duplication events, a central mechanism of genome evolution6. SETDB1 loss derepresses latent TE-derived regulatory elements, immunostimulatory genes, and TE-encoded retroviral antigens in these regions, and triggers TE-specific cytotoxic T cell responses in vivo. Our study establishes SETDB1 as an epigenetic checkpoint that suppresses tumour-intrinsic immunogenicity, and thus represents a candidate target for immunotherapy.


Asunto(s)
Silenciador del Gen , N-Metiltransferasa de Histona-Lisina/metabolismo , Neoplasias/genética , Neoplasias/inmunología , Animales , Antígenos Virales/inmunología , Sistemas CRISPR-Cas/genética , Cromatina/genética , Cromatina/metabolismo , Elementos Transponibles de ADN/genética , Modelos Animales de Enfermedad , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Ratones , Neoplasias/tratamiento farmacológico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Linfocitos T Citotóxicos/citología , Linfocitos T Citotóxicos/inmunología
11.
Nature ; 565(7737): 43-48, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30559380

RESUMEN

Most patients with cancer either do not respond to immune checkpoint blockade or develop resistance to it, often because of acquired mutations that impair antigen presentation. Here we show that loss of function of the RNA-editing enzyme ADAR1 in tumour cells profoundly sensitizes tumours to immunotherapy and overcomes resistance to checkpoint blockade. In the absence of ADAR1, A-to-I editing of interferon-inducible RNA species is reduced, leading to double-stranded RNA ligand sensing by PKR and MDA5; this results in growth inhibition and tumour inflammation, respectively. Loss of ADAR1 overcomes resistance to PD-1 checkpoint blockade caused by inactivation of antigen presentation by tumour cells. Thus, effective anti-tumour immunity is constrained by inhibitory checkpoints such as ADAR1 that limit the sensing of innate ligands. The induction of sufficient inflammation in tumours that are sensitized to interferon can bypass the therapeutic requirement for CD8+ T cell recognition of cancer cells and may provide a general strategy to overcome immunotherapy resistance.


Asunto(s)
Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/metabolismo , Puntos de Control del Ciclo Celular/efectos de los fármacos , Resistencia a Antineoplásicos/efectos de los fármacos , Melanoma Experimental/tratamiento farmacológico , Melanoma Experimental/genética , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Proteínas de Unión al ARN/metabolismo , Adenosina Desaminasa/genética , Animales , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Antígenos de Histocompatibilidad Clase I/inmunología , Inmunoterapia , Inflamación/genética , Inflamación/inmunología , Helicasa Inducida por Interferón IFIH1/metabolismo , Interferones/inmunología , Melanoma Experimental/inmunología , Melanoma Experimental/radioterapia , Ratones , Ratones Endogámicos C57BL , Fenotipo , Edición de ARN , ARN Bicatenario/genética , Proteínas de Unión al ARN/genética , Receptores Acoplados a Proteínas G/metabolismo
12.
Nucleic Acids Res ; 50(D1): D1391-D1397, 2022 01 07.
Artículo en Inglés | MEDLINE | ID: mdl-34534350

RESUMEN

Syngeneic mouse models are tumors derived from murine cancer cells engrafted on genetically identical mouse strains. They are widely used tools for studying tumor immunity and immunotherapy response in the context of a fully functional murine immune system. Large volumes of syngeneic mouse tumor expression profiles under different immunotherapy treatments have been generated, although a lack of systematic collection and analysis makes data reuse challenging. We present Tumor Immune Syngeneic MOuse (TISMO), a database with an extensive collection of syngeneic mouse model profiles with interactive visualization features. TISMO contains 605 in vitro RNA-seq samples from 49 syngeneic cancer cell lines across 23 cancer types, of which 195 underwent cytokine treatment. TISMO also includes 1518 in vivo RNA-seq samples from 68 syngeneic mouse tumor models across 19 cancer types, of which 832 were from immune checkpoint blockade (ICB) studies. We manually annotated the sample metadata, such as cell line, mouse strain, transplantation site, treatment, and response status, and uniformly processed and quality-controlled the RNA-seq data. Besides data download, TISMO provides interactive web interfaces to investigate whether specific gene expression, pathway enrichment, or immune infiltration level is associated with differential immunotherapy response. TISMO is available at http://tismo.cistrome.org.


Asunto(s)
Biomarcadores Farmacológicos , Neoplasias/genética , Programas Informáticos , Microambiente Tumoral/inmunología , Animales , Bases de Datos Genéticas , Modelos Animales de Enfermedad , Humanos , Inmunoterapia/tendencias , Ratones , Neoplasias/inmunología , Neoplasias/terapia , Microambiente Tumoral/genética
13.
Nature ; 547(7664): 413-418, 2017 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-28723893

RESUMEN

Immunotherapy with PD-1 checkpoint blockade is effective in only a minority of patients with cancer, suggesting that additional treatment strategies are needed. Here we use a pooled in vivo genetic screening approach using CRISPR-Cas9 genome editing in transplantable tumours in mice treated with immunotherapy to discover previously undescribed immunotherapy targets. We tested 2,368 genes expressed by melanoma cells to identify those that synergize with or cause resistance to checkpoint blockade. We recovered the known immune evasion molecules PD-L1 and CD47, and confirmed that defects in interferon-γ signalling caused resistance to immunotherapy. Tumours were sensitized to immunotherapy by deletion of genes involved in several diverse pathways, including NF-κB signalling, antigen presentation and the unfolded protein response. In addition, deletion of the protein tyrosine phosphatase PTPN2 in tumour cells increased the efficacy of immunotherapy by enhancing interferon-γ-mediated effects on antigen presentation and growth suppression. In vivo genetic screens in tumour models can identify new immunotherapy targets in unanticipated pathways.


Asunto(s)
Sistemas CRISPR-Cas/genética , Edición Génica , Inmunoterapia/métodos , Melanoma Experimental/inmunología , Melanoma Experimental/terapia , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Escape del Tumor/efectos de los fármacos , Escape del Tumor/inmunología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Genómica , Humanos , Interferones/inmunología , Mutación con Pérdida de Función , Melanoma Experimental/genética , Melanoma Experimental/patología , Ratones , FN-kappa B/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 2/deficiencia , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Escape del Tumor/genética , Respuesta de Proteína Desplegada , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Nucleic Acids Res ; 45(16): e148, 2017 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-28934479

RESUMEN

The T cell compartment must contain diversity in both T cell receptor (TCR) repertoire and cell state to provide effective immunity against pathogens. However, it remains unclear how differences in the TCR contribute to heterogeneity in T cell state. Single cell RNA-sequencing (scRNA-seq) can allow simultaneous measurement of TCR sequence and global transcriptional profile from single cells. However, current methods for TCR inference from scRNA-seq are limited in their sensitivity and require long sequencing reads, thus increasing the cost and decreasing the number of cells that can be feasibly analyzed. Here we present TRAPeS, a publicly available tool that can efficiently extract TCR sequence information from short-read scRNA-seq libraries. We apply it to investigate heterogeneity in the CD8+ T cell response in humans and mice, and show that it is accurate and more sensitive than existing approaches. Coupling TRAPeS with transcriptome analysis of CD8+ T cells specific for a single epitope from Yellow Fever Virus (YFV), we show that the recently described 'naive-like' memory population have significantly longer CDR3 regions and greater divergence from germline sequence than do effector-memory phenotype cells. This suggests that TCR usage is associated with the differentiation state of the CD8+ T cell response to YFV.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Regiones Determinantes de Complementariedad/química , Receptores de Antígenos de Linfocitos T/química , Análisis de Secuencia de ARN/métodos , Programas Informáticos , Algoritmos , Animales , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de la Célula Individual , Virus de la Fiebre Amarilla/inmunología
15.
J Immunol ; 197(5): 1809-22, 2016 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-27448585

RESUMEN

CD4(+) T cell help is critical for optimal CD8(+) T cell memory differentiation and maintenance in many experimental systems. In addition, many reports have identified reduced primary CD8(+) T cell responses in the absence of CD4(+) T cell help, which often coincides with reduced Ag or pathogen clearance. In this study, we demonstrate that absence of CD4(+) T cells at the time of adenovirus vector immunization of mice led to immediate impairments in early CD8(+) T cell functionality and differentiation. Unhelped CD8(+) T cells exhibited a reduced effector phenotype, decreased ex vivo cytotoxicity, and decreased capacity to produce cytokines. This dysfunctional state was imprinted within 3 d of immunization. Unhelped CD8(+) T cells expressed elevated levels of inhibitory receptors and exhibited transcriptomic exhaustion and anergy profiles by gene set enrichment analysis. Dysfunctional, impaired effector differentiation also occurred following immunization of CD4(+) T cell-deficient mice with a poxvirus vector. This study demonstrates that following priming with viral vectors, CD4(+) T cell help is required to promote both the expansion and acquisition of effector functions by CD8(+) T cells, which is accomplished by preventing immediate dysfunction.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Vacunas/inmunología , Adenoviridae/genética , Animales , Linfocitos T CD8-positivos/fisiología , Citocinas/inmunología , Citotoxicidad Inmunológica , Femenino , Regulación de la Expresión Génica , Vectores Genéticos , Inmunización , Memoria Inmunológica , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL
16.
JCI Insight ; 9(11)2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38687615

RESUMEN

A systems analysis was conducted to determine the potential molecular mechanisms underlying differential immunogenicity and protective efficacy results of a clinical trial of the radiation-attenuated whole-sporozoite PfSPZ vaccine in African infants. Innate immune activation and myeloid signatures at prevaccination baseline correlated with protection from P. falciparum parasitemia in placebo controls. These same signatures were associated with susceptibility to parasitemia among infants who received the highest and most protective PfSPZ vaccine dose. Machine learning identified spliceosome, proteosome, and resting DC signatures as prevaccination features predictive of protection after highest-dose PfSPZ vaccination, whereas baseline circumsporozoite protein-specific (CSP-specific) IgG predicted nonprotection. Prevaccination innate inflammatory and myeloid signatures were associated with higher sporozoite-specific IgG Ab response but undetectable PfSPZ-specific CD8+ T cell responses after vaccination. Consistent with these human data, innate stimulation in vivo conferred protection against infection by sporozoite injection in malaria-naive mice while diminishing the CD8+ T cell response to radiation-attenuated sporozoites. These data suggest a dichotomous role of innate stimulation for malaria protection and induction of protective immunity by whole-sporozoite malaria vaccines. The uncoupling of vaccine-induced protective immunity achieved by Abs from more protective CD8+ T cell responses suggests that PfSPZ vaccine efficacy in malaria-endemic settings may be constrained by opposing antigen presentation pathways.


Asunto(s)
Inmunidad Innata , Vacunas contra la Malaria , Malaria Falciparum , Plasmodium falciparum , Esporozoítos , Vacunas Atenuadas , Vacunas contra la Malaria/inmunología , Vacunas contra la Malaria/administración & dosificación , Inmunidad Innata/inmunología , Humanos , Animales , Malaria Falciparum/prevención & control , Malaria Falciparum/inmunología , Plasmodium falciparum/inmunología , Ratones , Vacunas Atenuadas/inmunología , Vacunas Atenuadas/administración & dosificación , Esporozoítos/inmunología , Esporozoítos/efectos de la radiación , Linfocitos T CD8-positivos/inmunología , Lactante , Proteínas Protozoarias/inmunología , Anticuerpos Antiprotozoarios/inmunología , Femenino , Parasitemia/inmunología , Parasitemia/prevención & control , Inmunoglobulina G/inmunología , Inmunoglobulina G/sangre , Eficacia de las Vacunas
17.
STAR Protoc ; 4(1): 102082, 2023 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-36861834

RESUMEN

Recognition of Cas9 and other proteins encoded in delivery vectors has limited CRISPR technology in vivo. Here, we present a protocol for genome engineering using selective CRISPR antigen removal (SCAR) lentiviral vectors in Renca mouse model. This protocol describes how to conduct an in vivo genetic screen with a sgRNA library and SCAR vectors that can be applied to different cell lines and contexts. For complete details on the use and execution of this protocol, please refer to Dubrot et al. (2021).1.


Asunto(s)
Sistemas CRISPR-Cas , ARN Guía de Sistemas CRISPR-Cas , Ratones , Animales , Sistemas CRISPR-Cas/genética , Biblioteca de Genes , Genoma , Línea Celular
18.
Nat Commun ; 14(1): 8048, 2023 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-38052854

RESUMEN

CAR-T therapy is a promising, novel treatment modality for B-cell malignancies and yet many patients relapse through a variety of means, including loss of CAR-T cells and antigen escape. To investigate leukemia-intrinsic CAR-T resistance mechanisms, we performed genome-wide CRISPR-Cas9 loss-of-function screens in an immunocompetent murine model of B-cell acute lymphoblastic leukemia (B-ALL) utilizing a modular guide RNA library. We identified IFNγR/JAK/STAT signaling and components of antigen processing and presentation pathway as key mediators of resistance to CAR-T therapy in vivo; intriguingly, loss of this pathway yielded the opposite effect in vitro (sensitized leukemia to CAR-T cells). Transcriptional characterization of this model demonstrated upregulation of these pathways in tumors relapsed after CAR-T treatment, and functional studies showed a surprising role for natural killer (NK) cells in engaging this resistance program. Finally, examination of data from B-ALL patients treated with CAR-T revealed an association between poor outcomes and increased expression of JAK/STAT and MHC-I in leukemia cells. Overall, our data identify an unexpected mechanism of resistance to CAR-T therapy in which tumor cell interaction with the in vivo tumor microenvironment, including NK cells, induces expression of an adaptive, therapy-induced, T-cell resistance program in tumor cells.


Asunto(s)
Linfoma de Burkitt , Leucemia , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Receptores Quiméricos de Antígenos , Humanos , Animales , Ratones , ARN Guía de Sistemas CRISPR-Cas , Inmunoterapia Adoptiva , Linfocitos T , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/terapia , Microambiente Tumoral
19.
Front Immunol ; 14: 1297932, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38213329

RESUMEN

Background: The GL261 and CT2A syngeneic tumor lines are frequently used as immunocompetent orthotopic mouse models of human glioblastoma (huGBM) but demonstrate distinct differences in their responses to immunotherapy. Methods: To decipher the cell-intrinsic mechanisms that drive immunotherapy resistance in CT2A-luc and to define the aspects of human cancer biology that these lines can best model, we systematically compared their characteristics using whole exome and transcriptome sequencing, and protein analysis through immunohistochemistry, Western blot, flow cytometry, immunopeptidomics, and phosphopeptidomics. Results: The transcriptional profiles of GL261-luc2 and CT2A-luc tumors resembled those of some huGBMs, despite neither line sharing the essential genetic or histologic features of huGBM. Both models exhibited striking hypermutation, with clonal hotspot mutations in RAS genes (Kras p.G12C in GL261-luc2 and Nras p.Q61L in CT2A-luc). CT2A-luc distinctly displayed mesenchymal differentiation, upregulated angiogenesis, and multiple defects in antigen presentation machinery (e.g. Tap1 p.Y488C and Psmb8 p.A275P mutations) and interferon response pathways (e.g. copy number losses of loci including IFN genes and reduced phosphorylation of JAK/STAT pathway members). The defect in MHC class I expression could be overcome in CT2A-luc by interferon-γ treatment, which may underlie the modest efficacy of some immunotherapy combinations. Additionally, CT2A-luc demonstrated substantial baseline secretion of the CCL-2, CCL-5, and CCL-22 chemokines, which play important roles as myeloid chemoattractants. Conclusion: Although the clinical contexts that can be modeled by GL261 and CT2A for huGBM are limited, CT2A may be an informative model of immunotherapy resistance due to its deficits in antigen presentation machinery and interferon response pathways.


Asunto(s)
Presentación de Antígeno , Glioblastoma , Humanos , Animales , Ratones , Quinasas Janus , Transducción de Señal , Factores de Transcripción STAT , Interferón gamma , Inmunoterapia
20.
J Immunother Cancer ; 10(3)2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35264433

RESUMEN

BACKGROUND: Oncogenes act in a cell-intrinsic way to promote tumorigenesis. Whether oncogenes also have a cell-extrinsic effect on suppressing the immune response to cancer is less well understood. METHODS: We use an in vivo expression screen of known cancer-associated somatic mutations in mouse syngeneic tumor models treated with checkpoint blockade to identify oncogenes that promote immune evasion. We then validated candidates from this screen in vivo and analyzed the tumor immune microenvironment of tumors expressing mutant protein to identify mechanisms of immune evasion. RESULTS: We found that expression of a catalytically active mutation in phospho-inositol 3 kinase (PI3K), PIK3CA c.3140A>G (H1047R) confers a selective growth advantage to tumors treated with immunotherapy that is reversed by pharmacological PI3K inhibition. PIK3CA H1047R-expression in tumors decreased the number of CD8+ T cells but increased the number of inhibitory myeloid cells following immunotherapy. Inhibition of myeloid infiltration by pharmacological or genetic modulation of Ccl2 in PIK3CA H1047R tumors restored sensitivity to programmed cell death protein 1 (PD-1) checkpoint blockade. CONCLUSIONS: PI3K activation enables tumor immune evasion by promoting an inhibitory myeloid microenvironment. Activating mutations in PI3K may be useful as a biomarker of poor response to immunotherapy. Our data suggest that some oncogenes promote tumorigenesis by enabling tumor cells to avoid clearance by the immune system. Identification of those mechanisms can advance rational combination strategies to increase the efficacy of immunotherapy.


Asunto(s)
Neoplasias , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos/metabolismo , Carcinogénesis , Fosfatidilinositol 3-Quinasa Clase I/genética , Modelos Animales de Enfermedad , Humanos , Evasión Inmune , Inositol , Ratones , Neoplasias/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA