Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Cell ; 185(7): 1223-1239.e20, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35290801

RESUMEN

While CRISPR screens are helping uncover genes regulating many cell-intrinsic processes, existing approaches are suboptimal for identifying extracellular gene functions, particularly in the tissue context. Here, we developed an approach for spatial functional genomics called Perturb-map. We applied Perturb-map to knock out dozens of genes in parallel in a mouse model of lung cancer and simultaneously assessed how each knockout influenced tumor growth, histopathology, and immune composition. Moreover, we paired Perturb-map and spatial transcriptomics for unbiased analysis of CRISPR-edited tumors. We found that in Tgfbr2 knockout tumors, the tumor microenvironment (TME) was converted to a fibro-mucinous state, and T cells excluded, concomitant with upregulated TGFß and TGFß-mediated fibroblast activation, indicating that TGFß-receptor loss on cancer cells increased TGFß bioavailability and its immunosuppressive effects on the TME. These studies establish Perturb-map for functional genomics within the tissue at single-cell resolution with spatial architecture preserved and provide insight into how TGFß responsiveness of cancer cells can affect the TME.


Asunto(s)
Neoplasias , Microambiente Tumoral , Animales , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Genómica , Ratones , Neoplasias/genética , Factor de Crecimiento Transformador beta/genética
2.
Nat Immunol ; 22(7): 914-927, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34099919

RESUMEN

To better define the control of immune system regulation, we generated an atlas of microRNA (miRNA) expression from 63 mouse immune cell populations and connected these signatures with assay for transposase-accessible chromatin using sequencing (ATAC-seq), chromatin immunoprecipitation followed by sequencing (ChIP-seq) and nascent RNA profiles to establish a map of miRNA promoter and enhancer usage in immune cells. miRNA complexity was relatively low, with >90% of the miRNA compartment of each population comprising <75 miRNAs; however, each cell type had a unique miRNA signature. Integration of miRNA expression with chromatin accessibility revealed putative regulatory elements for differentially expressed miRNAs, including miR-21a, miR-146a and miR-223. The integrated maps suggest that many miRNAs utilize multiple promoters to reach high abundance and identified dominant and divergent miRNA regulatory elements between lineages and during development that may be used by clustered miRNAs, such as miR-99a/let-7c/miR-125b, to achieve distinct expression. These studies, with web-accessible data, help delineate the cis-regulatory elements controlling miRNA signatures of the immune system.


Asunto(s)
Perfilación de la Expresión Génica , Sistema Inmunológico/metabolismo , MicroARNs/genética , Regiones Promotoras Genéticas , Transcriptoma , Animales , Células Cultivadas , Inmunoprecipitación de Cromatina , Biología Computacional , Regulación del Desarrollo de la Expresión Génica , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , RNA-Seq
3.
Cell ; 175(4): 1141-1155.e16, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30343902

RESUMEN

CRISPR pools are being widely employed to identify gene functions. However, current technology, which utilizes DNA as barcodes, permits limited phenotyping and bulk-cell resolution. To enable novel screening capabilities, we developed a barcoding system operating at the protein level. We synthesized modules encoding triplet combinations of linear epitopes to generate >100 unique protein barcodes (Pro-Codes). Pro-Code-expressing vectors were introduced into cells and analyzed by CyTOF mass cytometry. Using just 14 antibodies, we detected 364 Pro-Code populations; establishing the largest set of protein-based reporters. By pairing each Pro-Code with a different CRISPR, we simultaneously analyzed multiple phenotypic markers, including phospho-signaling, on dozens of knockouts. Pro-Code/CRISPR screens found two interferon-stimulated genes, the immunoproteasome component Psmb8 and a chaperone Rtp4, are important for antigen-dependent immune editing of cancer cells and identified Socs1 as a negative regulator of Pd-l1. The Pro-Code technology enables simultaneous high-dimensional protein-level phenotyping of 100s of genes with single-cell resolution.


Asunto(s)
Sistemas CRISPR-Cas , Citometría de Flujo/métodos , Genómica/métodos , Espectrometría de Masas/métodos , Análisis de la Célula Individual/métodos , Animales , Epítopos/química , Epítopos/clasificación , Epítopos/genética , Células HEK293 , Humanos , Inmunofenotipificación/métodos , Células Jurkat , Ratones Endogámicos BALB C , Proteoma/química , Proteoma/clasificación , Proteoma/genética , Células THP-1
4.
Immunity ; 48(2): 271-285.e5, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29466757

RESUMEN

Stem cells are critical for the maintenance of many tissues, but whether their integrity is maintained in the face of immunity is unclear. Here we found that cycling epithelial stem cells, including Lgr5+ intestinal stem cells, as well as ovary and mammary stem cells, were eliminated by activated T cells, but quiescent stem cells in the hair follicle and muscle were resistant to T cell killing. Immune evasion was an intrinsic property of the quiescent stem cells resulting from systemic downregulation of the antigen presentation machinery, including MHC class I and TAP proteins, and is mediated by the transactivator NLRC5. This process was reversed upon stem cell entry into the cell cycle. These studies identify a link between stem cell quiescence, antigen presentation, and immune evasion. As cancer-initiating cells can derive from stem cells, these findings may help explain how the earliest cancer cells evade immune surveillance.


Asunto(s)
Folículo Piloso/citología , Evasión Inmune , Vigilancia Inmunológica , Células Madre/inmunología , Animales , Presentación de Antígeno , Péptidos y Proteínas de Señalización Intracelular/fisiología , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos C57BL , Músculos/citología , Receptores Acoplados a Proteínas G/fisiología , Escape del Tumor
5.
Nature ; 595(7868): 578-584, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34135508

RESUMEN

Macrophages have a key role in shaping the tumour microenvironment (TME), tumour immunity and response to immunotherapy, which makes them an important target for cancer treatment1,2. However, modulating macrophages has proved extremely difficult, as we still lack a complete understanding of the molecular and functional diversity of the tumour macrophage compartment. Macrophages arise from two distinct lineages. Tissue-resident macrophages self-renew locally, independent of adult haematopoiesis3-5, whereas short-lived monocyte-derived macrophages arise from adult haematopoietic stem cells, and accumulate mostly in inflamed lesions1. How these macrophage lineages contribute to the TME and cancer progression remains unclear. To explore the diversity of the macrophage compartment in human non-small cell lung carcinoma (NSCLC) lesions, here we performed single-cell RNA sequencing of tumour-associated leukocytes. We identified distinct populations of macrophages that were enriched in human and mouse lung tumours. Using lineage tracing, we discovered that these macrophage populations differ in origin and have a distinct temporal and spatial distribution in the TME. Tissue-resident macrophages accumulate close to tumour cells early during tumour formation to promote epithelial-mesenchymal transition and invasiveness in tumour cells, and they also induce a potent regulatory T cell response that protects tumour cells from adaptive immunity. Depletion of tissue-resident macrophages reduced the numbers and altered the phenotype of regulatory T cells, promoted the accumulation of CD8+ T cells and reduced tumour invasiveness and growth. During tumour growth, tissue-resident macrophages became redistributed at the periphery of the TME, which becomes dominated by monocyte-derived macrophages in both mouse and human NSCLC. This study identifies the contribution of tissue-resident macrophages to early lung cancer and establishes them as a target for the prevention and treatment of early lung cancer lesions.


Asunto(s)
Carcinogénesis , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Macrófagos/inmunología , Microambiente Tumoral , Animales , Linfocitos T CD8-positivos/inmunología , Transición Epitelial-Mesenquimal , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Invasividad Neoplásica , Linfocitos T Reguladores/inmunología
6.
EMBO J ; 34(10): 1336-48, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25787857

RESUMEN

The severity and intensity of autoimmune disease in immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) patients and in scurfy mice emphasize the critical role played by thymus-derived regulatory T cells (tTregs) in maintaining peripheral immune tolerance. However, although tTregs are critical to prevent lethal autoimmunity and excessive inflammatory responses, their suppressive mechanism remains elusive. Here, we demonstrate that tTregs selectively inhibit CD27/CD70-dependent Th1 priming, while leaving the IL-12-dependent pathway unaffected. Immunized mice depleted of tTregs showed an increased response of IFN-γ-secreting CD4(+) T cells that was strictly reliant on a functional CD27/CD70 pathway. In vitro studies revealed that tTregs downregulate CD70 from the plasma membrane of dendritic cells (DCs) in a CD27-dependent manner. CD70 downregulation required contact between Tregs and DCs and resulted in endocytosis of CD27 and CD70 into the DC. These findings reveal a novel mechanism by which tTregs can maintain tolerance or prevent excessive, proinflammatory Th1 responses.


Asunto(s)
Ligando CD27/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Timo/inmunología , Timo/metabolismo , Animales , Ligando CD27/genética , Células Dendríticas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo
7.
Stem Cells ; 34(7): 1971-84, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-26992046

RESUMEN

Transplantation of mesenchymal stem cells (MSCs) into injured or diseased tissue-for the in situ delivery of a wide variety of MSC-secreted therapeutic proteins-is an emerging approach for the modulation of the clinical course of several diseases and traumata. From an emergency point-of-view, allogeneic MSCs have numerous advantages over patient-specific autologous MSCs since "off-the-shelf" cell preparations could be readily available for instant therapeutic intervention following acute injury. Although we confirmed the in vitro immunomodulatory capacity of allogeneic MSCs on antigen-presenting cells with standard coculture experiments, allogeneic MSC grafts were irrevocably rejected by the host's immune system upon either intramuscular or intracerebral transplantation. In an attempt to modulate MSC allograft rejection in vivo, we transduced MSCs with an interleukin-13 (IL13)-expressing lentiviral vector. Our data clearly indicate that prolonged survival of IL13-expressing allogeneic MSC grafts in muscle tissue coincided with the induction of an alternatively activated macrophage phenotype in vivo and a reduced number of alloantigen-reactive IFNγ- and/or IL2-producing CD8(+) T cells compared to nonmodified allografts. Similarly, intracerebral IL13-expressing MSC allografts also exhibited prolonged survival and induction of an alternatively activated macrophage phenotype, although a peripheral T cell component was absent. In summary, this study demonstrates that both innate and adaptive immune responses are effectively modulated in vivo by locally secreted IL13, ultimately resulting in prolonged MSC allograft survival in both muscle and brain tissue. Stem Cells 2016;34:1971-1984.


Asunto(s)
Supervivencia de Injerto/inmunología , Interleucina-13/farmacología , Isoantígenos/inmunología , Activación de Linfocitos/efectos de los fármacos , Macrófagos/metabolismo , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , Linfocitos T/inmunología , Aloinjertos/efectos de los fármacos , Aloinjertos/inmunología , Animales , Formación de Anticuerpos/efectos de los fármacos , Células Presentadoras de Antígenos/efectos de los fármacos , Células Dendríticas/citología , Células Dendríticas/efectos de los fármacos , Ingeniería Genética , Inmunomodulación/efectos de los fármacos , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Ratones , Microglía/efectos de los fármacos , Microglía/patología , Linfocitos T/efectos de los fármacos
8.
Dev Biol ; 363(2): 362-72, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22261149

RESUMEN

The maternal-zygotic transition (MZT) is an embryonic event that overlaps with and plays key roles in primary germ layer specification in vertebrates. During MZT, maternally supplied mRNAs are degraded while zygotic transcripts are synthesized to either reinforce the already specified cell fate or to trigger new cell identity. Here, we show that forced expression of the RNA-binding protein, XSeb4R, in animal pole blastomeres of Xenopus embryos, inappropriately stabilizes transcripts there, including maternal Sox3. This leads to the impaired ability of the ectodermal progenitors to respond to factors regulating brain patterning and their eventual loss by apoptosis. XSeb4R protein binds specifically to the 3'UTR of Sox3 mRNA. XSeb4R gain-of-function in ectodermal explants reveals increased stability of the maternal Sox3 transcripts, associated with a robust Sox3 protein production. Conversely, whereas XSeb4R depletion abolishes VegT expression, the amount of the maternal Sox3 mRNA is rather increased but without augmentation in the amount of Sox3 protein. Moreover, XSeb4R protein knockdown leads to the modification of the ectoderm-mesoderm boundary, marked by expanded/shifted expression of the mesodermal marker genes such as Xbra and Apod, followed by an expression inhibition of Epi. K., an ectodermal marker. Overall, our data suggest XSeb4R as a novel player in gene expression regulation, acting at the posttranscriptional level during ectoderm specification in Xenopus.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Procesamiento Postranscripcional del ARN , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción SOXB1/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis/embriología , Xenopus laevis/genética , Cigoto/crecimiento & desarrollo , Animales , Apoptosis , Blastómeros/metabolismo , Ectodermo/metabolismo , Femenino , Mesodermo/metabolismo , Unión Proteica , Xenopus laevis/metabolismo
9.
Biotechniques ; 66(6): 295-302, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31039627

RESUMEN

CRISPR/Cas9 technology has evolved as the most powerful approach to generate genetic models both for fundamental and preclinical research. Despite its apparent simplicity, the outcome of a genome-editing experiment can be substantially impacted by technical parameters and biological considerations. Here, we present guidelines and tools to optimize CRISPR/Cas9 genome-targeting efficiency and specificity. The nature of the target locus, the design of the single guide RNA and the choice of the delivery method should all be carefully considered prior to a genome-editing experiment. Different methods can also be used to detect off-target cleavages and decrease the risk of unwanted mutations. Together, these optimized tools and proper controls are essential to the assessment of CRISPR/Cas9 genome-editing experiments.


Asunto(s)
Sistemas CRISPR-Cas , Edición Génica/métodos , Animales , Técnicas de Inactivación de Genes/métodos , Sitios Genéticos , Células HEK293 , Humanos , ARN Guía de Kinetoplastida/genética , Pez Cebra/genética
10.
Nat Med ; 25(5): 814-824, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30962585

RESUMEN

Indolent non-Hodgkin's lymphomas (iNHLs) are incurable with standard therapy and are poorly responsive to checkpoint blockade. Although lymphoma cells are efficiently killed by primed T cells, in vivo priming of anti-lymphoma T cells has been elusive. Here, we demonstrate that lymphoma cells can directly prime T cells, but in vivo immunity still requires cross-presentation. To address this, we developed an in situ vaccine (ISV), combining Flt3L, radiotherapy, and a TLR3 agonist, which recruited, antigen-loaded and activated intratumoral, cross-presenting dendritic cells (DCs). ISV induced anti-tumor CD8+ T cell responses and systemic (abscopal) cancer remission in patients with advanced stage iNHL in an ongoing trial ( NCT01976585 ). Non-responding patients developed a population of PD1+CD8+ T cells after ISV, and murine tumors became newly responsive to PD1 blockade, prompting a follow-up trial of the combined therapy. Our data substantiate that recruiting and activating intratumoral, cross-priming DCs is achievable and critical to anti-tumor T cell responses and PD1-blockade efficacy.


Asunto(s)
Vacunas contra el Cáncer , Linfoma de Células B/terapia , Adulto , Anciano , Animales , Presentación de Antígeno , Linfocitos T CD8-positivos/inmunología , Carboximetilcelulosa de Sodio/análogos & derivados , Carboximetilcelulosa de Sodio/uso terapéutico , Línea Celular Tumoral , Terapia Combinada , Células Dendríticas/inmunología , Femenino , Humanos , Inmunoterapia Adoptiva , Linfoma de Células B/inmunología , Linfoma de Células B/patología , Masculino , Proteínas de la Membrana/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Poli I-C/uso terapéutico , Polilisina/análogos & derivados , Polilisina/uso terapéutico , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Receptor Toll-Like 3/agonistas , Vacunación
11.
Cancer Discov ; 9(8): 1124-1141, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31186238

RESUMEN

PD-1 immune checkpoint inhibitors have produced encouraging results in patients with hepatocellular carcinoma (HCC). However, what determines resistance to anti-PD-1 therapies is unclear. We created a novel genetically engineered mouse model of HCC that enables interrogation of how different genetic alterations affect immune surveillance and response to immunotherapies. Expression of exogenous antigens in MYC;Trp53 -/- HCCs led to T cell-mediated immune surveillance, which was accompanied by decreased tumor formation and increased survival. Some antigen-expressing MYC;Trp53 -/- HCCs escaped the immune system by upregulating the ß-catenin (CTNNB1) pathway. Accordingly, expression of exogenous antigens in MYC;CTNNB1 HCCs had no effect, demonstrating that ß-catenin promoted immune escape, which involved defective recruitment of dendritic cells and consequently impaired T-cell activity. Expression of chemokine CCL5 in antigen-expressing MYC;CTNNB1 HCCs restored immune surveillance. Finally, ß-catenin-driven tumors were resistant to anti-PD-1. In summary, ß-catenin activation promotes immune escape and resistance to anti-PD-1 and could represent a novel biomarker for HCC patient exclusion. SIGNIFICANCE: Determinants of response to anti-PD-1 immunotherapies in HCC are poorly understood. Using a novel mouse model of HCC, we show that ß-catenin activation promotes immune evasion and resistance to anti-PD-1 therapy and could potentially represent a novel biomarker for HCC patient exclusion.See related commentary by Berraondo et al., p. 1003.This article is highlighted in the In This Issue feature, p. 983.


Asunto(s)
Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/metabolismo , Resistencia a Antineoplásicos , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/metabolismo , Escape del Tumor , beta Catenina/metabolismo , Animales , Antineoplásicos Inmunológicos/farmacología , Antineoplásicos Inmunológicos/uso terapéutico , Biomarcadores de Tumor , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/tratamiento farmacológico , Línea Celular Tumoral , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Expresión Génica , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Neoplasias Hepáticas/patología , Ratones , Oncogenes , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Transducción de Señal , Resultado del Tratamiento , Escape del Tumor/genética , Ensayos Antitumor por Modelo de Xenoinjerto , beta Catenina/genética
12.
Prog Mol Biol Transl Sci ; 136: 131-54, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26615095

RESUMEN

Regulatory T cells (Tregs) are critical to maintain the homeostasis of the organism, i.e., to prevent (excessive) inflammatory reactions against self and nonself. Data in the literature suggest that a variety of suppressive mechanisms exist that may act selectively depending on the tissue environment. An interesting question is whether dendritic cells (DCs), which are involved in immunity and tolerance, are targets of Tregs. We review herein some experimental evidence indicating that some suppressive mechanisms involve DCs, altering their function and inducing an immune control at the very first step of the immune response.


Asunto(s)
Células Dendríticas/inmunología , Vigilancia Inmunológica , Linfocitos T Reguladores/inmunología , Animales , Humanos , Terapia de Inmunosupresión , Modelos Inmunológicos
13.
Front Immunol ; 5: 112, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24734030

RESUMEN

It was recently proposed that T lymphocytes, which closely interact with APCs, can extract surface molecules from the presenting cells when they dissociate. These observations question the classical view of discrete interactions between phenotypically defined cell populations. In this review, we summarize some reports suggesting that membrane exchange at the immune synapse can be a vector for intercellular communication and envisage some consequences on the biology of T cells.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA