Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
1.
Cell ; 186(21): 4546-4566.e27, 2023 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-37769657

RESUMEN

Neutrophils are abundant immune cells in the circulation and frequently infiltrate tumors in substantial numbers. However, their precise functions in different cancer types remain incompletely understood, including in the brain microenvironment. We therefore investigated neutrophils in tumor tissue of glioma and brain metastasis patients, with matched peripheral blood, and herein describe the first in-depth analysis of neutrophil phenotypes and functions in these tissues. Orthogonal profiling strategies in humans and mice revealed that brain tumor-associated neutrophils (TANs) differ significantly from blood neutrophils and have a prolonged lifespan and immune-suppressive and pro-angiogenic capacity. TANs exhibit a distinct inflammatory signature, driven by a combination of soluble inflammatory mediators including tumor necrosis factor alpha (TNF-ɑ) and Ceruloplasmin, which is more pronounced in TANs from brain metastasis versus glioma. Myeloid cells, including tumor-associated macrophages, emerge at the core of this network of pro-inflammatory mediators, supporting the concept of a critical myeloid niche regulating overall immune suppression in human brain tumors.

2.
Cell Rep Med ; 4(1): 100900, 2023 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-36652909

RESUMEN

Brain metastases (BrMs) are the most common form of brain tumors in adults and frequently originate from lung and breast primary cancers. BrMs are associated with high mortality, emphasizing the need for more effective therapies. Genetic profiling of primary tumors is increasingly used as part of the effort to guide targeted therapies against BrMs, and immune-based strategies for the treatment of metastatic cancer are gaining momentum. However, the tumor immune microenvironment (TIME) of BrM is extremely heterogeneous, and whether specific genetic profiles are associated with distinct immune states remains unknown. Here, we perform an extensive characterization of the immunogenomic landscape of human BrMs by combining whole-exome/whole-genome sequencing, RNA sequencing of immune cell populations, flow cytometry, immunofluorescence staining, and tissue imaging analyses. This revealed unique TIME phenotypes in genetically distinct lung- and breast-BrMs, thereby enabling the development of personalized immunotherapies tailored by the genetic makeup of the tumors.


Asunto(s)
Neoplasias Encefálicas , Neoplasias de la Mama , Melanoma , Neoplasias Cutáneas , Adulto , Humanos , Femenino , Neoplasias Encefálicas/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Inmunoterapia , Microambiente Tumoral/genética
3.
Nat Protoc ; 16(10): 4692-4721, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34462595

RESUMEN

Human tissue samples represent an invaluable source of information for the analysis of disease-specific cellular alterations and their variation between different pathologies. In cancer research, advancing a comprehensive understanding of the unique characteristics of individual tumor types and their microenvironment is of considerable importance for clinical translation. However, investigating human brain tumor tissue is challenging due to the often-limited availability of surgical specimens. Here we describe a multimodule integrated pipeline for the processing of freshly resected human brain tumor tissue and matched blood that enables analysis of the tumor microenvironment, with a particular focus on the tumor immune microenvironment (TIME). The protocol maximizes the information yield from limited tissue and includes both the preservation of bulk tissue, which can be performed within 1 h following surgical resection, as well as tissue dissociation for an in-depth characterization of individual TIME cell populations, which typically takes several hours depending on tissue quantity and further downstream processing. We also describe integrated modules for immunofluorescent staining of sectioned tissue, bulk tissue genomic analysis and fluorescence- or magnetic-activated cell sorting of digested tissue for subsequent culture or transcriptomic analysis by RNA sequencing. Applying this pipeline, we have previously described the overall TIME landscape across different human brain malignancies, and were able to delineate disease-specific alterations of tissue-resident versus recruited macrophage populations. This protocol will enable researchers to use this pipeline to address further research questions regarding the tumor microenvironment.


Asunto(s)
Neoplasias Encefálicas , Perfilación de la Expresión Génica , Humanos , Macrófagos , Análisis de Secuencia de ARN , Microambiente Tumoral
4.
Nucleic Acids Res ; 49(9): 5057-5073, 2021 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-33950194

RESUMEN

Activation-induced deaminase (AID) initiates antibody diversification in germinal center B cells by deaminating cytosines, leading to somatic hypermutation and class-switch recombination. Loss-of-function mutations in AID lead to hyper-IgM syndrome type 2 (HIGM2), a rare human primary antibody deficiency. AID-mediated deamination has been proposed as leading to active demethylation of 5-methycytosines in the DNA, although evidence both supports and casts doubt on such a role. In this study, using whole-genome bisulfite sequencing of HIGM2 B cells, we investigated direct AID involvement in active DNA demethylation. HIGM2 naïve and memory B cells both display widespread DNA methylation alterations, of which ∼25% are attributable to active DNA demethylation. For genes that undergo active demethylation that is impaired in HIGM2 individuals, our analysis indicates that AID is not directly involved. We demonstrate that the widespread alterations in the DNA methylation and expression profiles of HIGM2 naïve B cells result from premature overstimulation of the B-cell receptor prior to the germinal center reaction. Our data support a role for AID in B cell central tolerance in preventing the expansion of autoreactive cell clones, affecting the correct establishment of DNA methylation patterns.


Asunto(s)
Linfocitos B/inmunología , Citidina Desaminasa/fisiología , Metilación de ADN , Síndrome de Inmunodeficiencia con Hiper-IgM/genética , Síndrome de Inmunodeficiencia con Hiper-IgM/inmunología , Autoinmunidad , Linfocitos B/metabolismo , Citidina Desaminasa/deficiencia , Citidina Desaminasa/genética , Centro Germinal/inmunología , Humanos , Síndrome de Inmunodeficiencia con Hiper-IgM/metabolismo , Tolerancia Inmunológica , Memoria Inmunológica , Receptores de Antígenos de Linfocitos B/genética , Transcriptoma , Secuenciación Completa del Genoma
5.
PLoS Genet ; 16(12): e1008960, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33362210

RESUMEN

Most B cell lymphomas originate from B cells that have germinal center (GC) experience and bear chromosome translocations and numerous point mutations. GC B cells remodel their immunoglobulin (Ig) genes by somatic hypermutation (SHM) and class switch recombination (CSR) in their Ig genes. Activation Induced Deaminase (AID) initiates CSR and SHM by generating U:G mismatches on Ig DNA that can then be processed by Uracyl-N-glycosylase (UNG). AID promotes collateral damage in the form of chromosome translocations and off-target SHM, however, the exact contribution of AID activity to lymphoma generation and progression is not completely understood. Here we show using a conditional knock-in strategy that AID supra-activity alone is not sufficient to generate B cell transformation. In contrast, in the absence of UNG, AID supra-expression increases SHM and promotes lymphoma. Whole exome sequencing revealed that AID heavily contributes to lymphoma SHM, promoting subclonal variability and a wider range of oncogenic variants. Thus, our data provide direct evidence that UNG is a brake to AID-induced intratumoral heterogeneity and evolution of B cell lymphoma.


Asunto(s)
Citidina Desaminasa/genética , Heterogeneidad Genética , Linfoma de Células B/genética , Uracil-ADN Glicosidasa/genética , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Evolución Clonal , Citidina Desaminasa/metabolismo , Femenino , Linfoma de Células B/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación , Uracil-ADN Glicosidasa/metabolismo
6.
Leukemia ; 34(10): 2722-2735, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32576963

RESUMEN

Mutations in genes encoding subunits of the SWI/SNF chromatin remodeling complex are frequently found in different human cancers. While the tumor suppressor function of this complex is widely established in solid tumors, its role in hematologic malignancies is largely unknown. Recurrent point mutations in BCL7A gene, encoding a subunit of the SWI/SNF complex, have been reported in diffuse large B-cell lymphoma (DLBCL), but their functional impact remains to be elucidated. Here we show that BCL7A often undergoes biallelic inactivation, including a previously unnoticed mutational hotspot in the splice donor site of intron one. The splice site mutations render a truncated BCL7A protein, lacking a portion of the amino-terminal domain. Moreover, restoration of wild-type BCL7A expression elicits a tumor suppressor-like phenotype in vitro and in vivo. In contrast, splice site mutations block the tumor suppressor function of BCL7A by preventing its binding to the SWI/SNF complex. We also show that BCL7A restoration induces transcriptomic changes in genes involved in B-cell activation. In addition, we report that SWI/SNF complex subunits harbor mutations in more than half of patients with germinal center B-cell (GCB)-DLBCL. Overall, this work demonstrates the tumor suppressor function of BCL7A in DLBCL, and highlights that the SWI/SNF complex plays a relevant role in DLBCL pathogenesis.


Asunto(s)
Genes Supresores de Tumor , Linfoma de Células B Grandes Difuso/genética , Proteínas de Microfilamentos/genética , Mutación , Proteínas Oncogénicas/genética , Dominios y Motivos de Interacción de Proteínas/genética , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/patología , Cromatografía Liquida , Proteínas Cromosómicas no Histona/metabolismo , Análisis Mutacional de ADN , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Humanos , Activación de Linfocitos/inmunología , Linfoma de Células B Grandes Difuso/diagnóstico , Linfoma de Células B Grandes Difuso/terapia , Ratones , Proteínas de Microfilamentos/química , Imagen Molecular , Complejos Multiproteicos , Proteínas Oncogénicas/química , Unión Proteica , Espectrometría de Masas en Tándem , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Nat Commun ; 10(1): 5563, 2019 12 05.
Artículo en Inglés | MEDLINE | ID: mdl-31804490

RESUMEN

The prerequisite to prevent childhood B-cell acute lymphoblastic leukemia (B-ALL) is to decipher its etiology. The current model suggests that infection triggers B-ALL development through induction of activation-induced cytidine deaminase (AID; also known as AICDA) in precursor B-cells. This evidence has been largely acquired through the use of ex vivo functional studies. However, whether this mechanism governs native non-transplant B-ALL development is unknown. Here we show that, surprisingly, AID genetic deletion does not affect B-ALL development in Pax5-haploinsufficient mice prone to B-ALL upon natural infection exposure. We next test the effect of premature AID expression from earliest pro-B-cell stages in B-cell transformation. The generation of AID off-target mutagenic activity in precursor B-cells does not promote B-ALL. Likewise, known drivers of human B-ALL are not preferentially targeted by AID. Overall these results suggest that infections promote B-ALL through AID-independent mechanisms, providing evidence for a new model of childhood B-ALL development.


Asunto(s)
Linfocitos B/metabolismo , Transformación Celular Neoplásica/metabolismo , Citidina Desaminasa/metabolismo , Infecciones/fisiopatología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Animales , Linfocitos B/patología , Transformación Celular Neoplásica/genética , Niño , Citidina Desaminasa/genética , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Infecciones/genética , Estimación de Kaplan-Meier , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética
8.
J Exp Med ; 215(3): 761-771, 2018 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-29374026

RESUMEN

Activation-induced deaminase (AID) initiates antibody diversification in germinal center (GC) B cells through the deamination of cytosines on immunoglobulin genes. AID can also target other regions in the genome, triggering mutations or chromosome translocations, with major implications for oncogenic transformation. However, understanding the specificity of AID has proved extremely challenging. We have sequenced at very high depth >1,500 genomic regions from GC B cells and identified 275 genes targeted by AID, including 30 of the previously known 35 AID targets. We have also identified the most highly mutated hotspot for AID activity described to date. Furthermore, integrative analysis of the molecular features of mutated genes coupled to machine learning has produced a powerful predictive tool for AID targets. We also have found that base excision repair and mismatch repair back up each other to faithfully repair AID-induced lesions. Finally, our data establish a novel link between AID mutagenic activity and lymphomagenesis.


Asunto(s)
Citidina Desaminasa/genética , Hipermutación Somática de Inmunoglobulina/genética , Animales , Secuencia de Bases , Daño del ADN/genética , Reparación de la Incompatibilidad de ADN/genética , Secuenciación de Nucleótidos de Alto Rendimiento , Linfoma/genética , Linfoma/patología , Ratones , Mutación/genética
9.
Nat Commun ; 8: 16067, 2017 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-28677680

RESUMEN

In germinal centres (GC) mature B cells undergo intense proliferation and immunoglobulin gene modification before they differentiate into memory B cells or long-lived plasma cells (PC). GC B-cell-to-PC transition involves a major transcriptional switch that promotes a halt in cell proliferation and the production of secreted immunoglobulins. Here we show that the CCCTC-binding factor (CTCF) is required for the GC reaction in vivo, whereas in vitro the requirement for CTCF is not universal and instead depends on the pathways used for B-cell activation. CTCF maintains the GC transcriptional programme, allows a high proliferation rate, and represses the expression of Blimp-1, the master regulator of PC differentiation. Restoration of Blimp-1 levels partially rescues the proliferation defect of CTCF-deficient B cells. Thus, our data reveal an essential function of CTCF in maintaining the GC transcriptional programme and preventing premature PC differentiation.


Asunto(s)
Linfocitos B/fisiología , Factor de Unión a CCCTC/fisiología , Diferenciación Celular , Centro Germinal/metabolismo , Animales , Femenino , Masculino , Ratones , Células Plasmáticas , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Cultivo Primario de Células , Transcripción Genética
10.
Blood ; 129(17): 2408-2419, 2017 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-28188132

RESUMEN

Non-Hodgkin lymphoma comprises a variety of neoplasms, many of which arise from germinal center (GC)-experienced B cells. microRNA-28 (miR-28) is a GC-specific miRNA whose expression is lost in numerous mature B-cell neoplasms. Here we show that miR-28 regulates the GC reaction in primary B cells by impairing class switch recombination and memory B and plasma cell differentiation. Deep quantitative proteomics combined with transcriptome analysis identified miR-28 targets involved in cell-cycle and B-cell receptor signaling. Accordingly, we found that miR-28 expression diminished proliferation in primary and lymphoma cells in vitro. Importantly, miR-28 reexpression in human Burkitt (BL) and diffuse large B-cell lymphoma (DLBCL) xenografts blocked tumor growth, both when delivered in viral vectors or as synthetic, clinically amenable, molecules. Further, the antitumoral effect of miR-28 is conserved in a primary murine in vivo model of BL. Thus, miR-28 replacement is uncovered as a novel therapeutic strategy for DLBCL and BL treatment.


Asunto(s)
Linfocitos B/inmunología , Linfoma de Burkitt/terapia , Regulación Neoplásica de la Expresión Génica , Centro Germinal/inmunología , Linfoma de Células B Grandes Difuso/terapia , MicroARNs/genética , Animales , Linfocitos B/patología , Linfoma de Burkitt/genética , Linfoma de Burkitt/inmunología , Linfoma de Burkitt/patología , Diferenciación Celular , Proliferación Celular , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Centro Germinal/patología , Humanos , Cambio de Clase de Inmunoglobulina , Memoria Inmunológica , Lentivirus/genética , Lentivirus/metabolismo , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/inmunología , Linfoma de Células B Grandes Difuso/patología , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/patología , Proteómica , Transcriptoma , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...