Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Br Dent J ; 234(8): 588, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37117362
2.
Nat Commun ; 13(1): 5818, 2022 10 07.
Artículo en Inglés | MEDLINE | ID: mdl-36207323

RESUMEN

Human papillomavirus (HPV)-associated cervical cancer is a leading cause of cancer deaths in women. Here we present an integrated multi-omic analysis of 643 cervical squamous cell carcinomas (CSCC, the most common histological variant of cervical cancer), representing patient populations from the USA, Europe and Sub-Saharan Africa and identify two CSCC subtypes (C1 and C2) with differing prognosis. C1 and C2 tumours can be driven by either of the two most common HPV types in cervical cancer (16 and 18) and while HPV16 and HPV18 are overrepresented among C1 and C2 tumours respectively, the prognostic difference between groups is not due to HPV type. C2 tumours, which comprise approximately 20% of CSCCs across these cohorts, display distinct genomic alterations, including loss or mutation of the STK11 tumour suppressor gene, increased expression of several immune checkpoint genes and differences in the tumour immune microenvironment that may explain the shorter survival associated with this group. In conclusion, we identify two therapy-relevant CSCC subtypes that share the same defining characteristics across three geographically diverse cohorts.


Asunto(s)
Carcinoma de Células Escamosas , Infecciones por Papillomavirus , Neoplasias del Cuello Uterino , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Femenino , Papillomavirus Humano 16/genética , Humanos , Papillomaviridae/genética , Infecciones por Papillomavirus/complicaciones , Infecciones por Papillomavirus/genética , Infecciones por Papillomavirus/patología , Pronóstico , Microambiente Tumoral , Neoplasias del Cuello Uterino/patología
3.
Cell Death Differ ; 29(12): 2459-2471, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36138226

RESUMEN

Oncohistones represent compelling evidence for a causative role of epigenetic perturbations in cancer. Giant cell tumours of bone (GCTs) are characterised by a mutated histone H3.3 as the sole genetic driver present in bone-forming osteoprogenitor cells but absent from abnormally large bone-resorbing osteoclasts which represent the hallmark of these neoplasms. While these striking features imply a pathogenic interaction between mesenchymal and myelomonocytic lineages during GCT development, the underlying mechanisms remain unknown. We show that the changes in the transcriptome and epigenome in the mesenchymal cells caused by the H3.3-G34W mutation contribute to increase osteoclast recruitment in part via reduced expression of the TGFß-like soluble factor, SCUBE3. Transcriptional changes in SCUBE3 are associated with altered histone marks and H3.3G34W enrichment at its enhancer regions. In turn, osteoclasts secrete unregulated amounts of SEMA4D which enhances proliferation of mutated osteoprogenitors arresting their maturation. These findings provide a mechanism by which GCTs undergo differentiation in response to denosumab, a drug that depletes the tumour of osteoclasts. In contrast, hTERT alterations, commonly found in malignant GCT, result in the histone-mutated neoplastic cells being independent of osteoclasts for their proliferation, predicting unresponsiveness to denosumab. We provide a mechanism for the initiation of GCT, the basis of which is dysfunctional cross-talk between bone-forming and bone-resorbing cells. The findings highlight the role of tumour/microenvironment bidirectional interactions in tumorigenesis and how this is exploited in the treatment of GCT.


Asunto(s)
Neoplasias Óseas , Tumor Óseo de Células Gigantes , Humanos , Tumor Óseo de Células Gigantes/genética , Tumor Óseo de Células Gigantes/tratamiento farmacológico , Tumor Óseo de Células Gigantes/patología , Histonas/genética , Histonas/metabolismo , Denosumab/metabolismo , Denosumab/uso terapéutico , Neoplasias Óseas/genética , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Osteoclastos/metabolismo , Remodelación Ósea/genética , Microambiente Tumoral , Proteínas de Unión al Calcio/metabolismo
4.
Cell Rep ; 39(7): 110819, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35584681

RESUMEN

T cell pathology in the skin leads to monocyte influx, but we have little understanding of the fate of recruited cells within the diseased niche, or the long-term impact on cutaneous immune homeostasis. By combining a murine model of acute graft-versus-host disease (aGVHD) with analysis of patient samples, we demonstrate that pathology initiates dermis-specific macrophage differentiation and show that aGVHD-primed macrophages continue to dominate the dermal compartment at the relative expense of quiescent MHCIIint cells. Exposure of the altered dermal niche to topical haptens after disease resolution results in hyper-activation of regulatory T cells (Treg), but local breakdown in tolerance. Disease-imprinted macrophages express increased IL-1ß and are predicted to elicit altered TNF superfamily interactions with cutaneous Treg, and we demonstrate the direct loss of T cell regulation within the resolved skin. Thus, T cell pathology leaves an immunological scar in the skin marked by failure to re-set immune homeostasis.


Asunto(s)
Enfermedad Injerto contra Huésped , Piel , Animales , Humanos , Tolerancia Inmunológica , Macrófagos/metabolismo , Ratones , Monocitos/metabolismo , Piel/metabolismo , Linfocitos T Reguladores
5.
Eur J Immunol ; 52(4): 566-581, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35092032

RESUMEN

T-bet is the lineage-specifying transcription factor for CD4+ TH 1 cells. T-bet has also been found in other CD4+ T cell subsets, including TH 17 cells and Treg, where it modulates their functional characteristics. However, we lack information on when and where T-bet is expressed during T cell differentiation and how this impacts T cell differentiation and function. To address this, we traced the ontogeny of T-bet-expressing cells using a fluorescent fate-mapping mouse line. We demonstrate that T-bet is expressed in a subset of CD4+ T cells that have naïve cell surface markers and transcriptional profile and that this novel cell population is phenotypically and functionally distinct from previously described populations of naïve and memory CD4+ T cells. Naïve-like T-bet-experienced cells are polarized to the TH 1 lineage, predisposed to produce IFN-γ upon cell activation, and resist repolarization to other lineages in vitro and in vivo. These results demonstrate that lineage-specifying factors can polarize T cells in the absence of canonical markers of T cell activation and that this has an impact on the subsequent T-helper response.


Asunto(s)
Proteínas de Dominio T Box , Células TH1 , Animales , Diferenciación Celular , Regulación de la Expresión Génica , Activación de Linfocitos , Ratones , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Linfocitos T Reguladores/metabolismo , Células Th17/metabolismo , Células Th2
6.
Life Sci Alliance ; 4(11)2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34531288

RESUMEN

Gene expression programs controlled by lineage-determining transcription factors are often conserved between species. However, infectious diseases have exerted profound evolutionary pressure, and therefore the genes regulated by immune-specific transcription factors might be expected to exhibit greater divergence. T-bet (Tbx21) is the immune-specific, lineage-specifying transcription factor for T helper type I (Th1) immunity, which is fundamental for the immune response to intracellular pathogens but also underlies inflammatory diseases. We compared T-bet genomic targets between mouse and human CD4+ T cells and correlated T-bet binding patterns with species-specific gene expression. Remarkably, we found that the majority of T-bet target genes are conserved between mouse and human, either via preservation of binding sites or via alternative binding sites associated with transposon-linked insertion. Species-specific T-bet binding was associated with differences in transcription factor-binding motifs and species-specific expression of associated genes. These results provide a genome-wide cross-species comparison of Th1 gene regulation that will enable more accurate translation of genetic targets and therapeutics from pre-clinical models of inflammatory and infectious diseases and cancer into human clinical trials.


Asunto(s)
Regulación de la Expresión Génica/genética , Proteínas de Dominio T Box/genética , Células TH1/fisiología , Animales , Sitios de Unión/genética , Bases de Datos Genéticas , Expresión Génica/genética , Genoma/genética , Humanos , Ratones , Unión Proteica/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/inmunología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Transcriptoma/genética
7.
PLoS One ; 16(8): e0254697, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34424918

RESUMEN

The PAF complex (PAFC) coordinates transcription elongation and mRNA processing and its CDC73/parafibromin subunit functions as a tumour suppressor. The NF2/Merlin tumour suppressor functions both at the cell cortex and nucleus and is a key mediator of contact inhibition but the molecular mechanisms remain unclear. In this study we have used affinity proteomics to identify novel Merlin interacting proteins and show that Merlin forms a complex with multiple proteins involved in RNA processing including the PAFC and the CHD1 chromatin remodeller. Tumour-derived inactivating mutations in both Merlin and the CDC73 PAFC subunit mutually disrupt their interaction and growth suppression by Merlin requires CDC73. Merlin interacts with the PAFC in a cell density-dependent manner and we identify a role for FAT cadherins in regulating the Merlin-PAFC interaction. Our results suggest that in addition to its function within the Hippo pathway, Merlin is part of a tumour suppressor network regulated by cell-cell adhesion which coordinates post-initiation steps of the transcription cycle of genes mediating contact inhibition.


Asunto(s)
Adhesión Celular/genética , ADN Helicasas/genética , Proteínas de Unión al ADN/genética , Neoplasias/genética , Neurofibromina 2/genética , Proteínas Supresoras de Tumor/genética , Proliferación Celular/genética , Cromatina/genética , Ensamble y Desensamble de Cromatina/genética , Inhibición de Contacto/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Neoplasias/patología , Unión Proteica/genética , Mapas de Interacción de Proteínas/genética , Transducción de Señal/genética
8.
Cancer Discov ; 10(7): 998-1017, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32349972

RESUMEN

Loss-of-function mutations of EZH2, the enzymatic component of PRC2, have been associated with poor outcome and chemotherapy resistance in T-cell acute lymphoblastic leukemia (T-ALL). Using isogenic T-ALL cells, with and without CRISPR/Cas9-induced EZH2-inactivating mutations, we performed a cell-based synthetic lethal drug screen. EZH2-deficient cells exhibited increased sensitivity to structurally diverse inhibitors of CHK1, an interaction that could be validated genetically. Furthermore, small-molecule inhibition of CHK1 had efficacy in delaying tumor progression in isogenic EZH2-deficient, but not EZH2 wild-type, T-ALL cells in vivo, as well as in a primary cell model of PRC2-mutant ALL. Mechanistically, EZH2 deficiency resulted in a gene-expression signature of immature T-ALL cells, marked transcriptional upregulation of MYCN, increased replication stress, and enhanced dependency on CHK1 for cell survival. Finally, we demonstrate this phenotype is mediated through derepression of a distal PRC2-regulated MYCN enhancer. In conclusion, we highlight a novel and clinically exploitable pathway in high-risk EZH2-mutated T-ALL. SIGNIFICANCE: Loss-of-function mutations of PRC2 genes are associated with chemotherapy resistance in T-ALL, yet no specific therapy for this aggressive subtype is currently clinically available. Our work demonstrates that loss of EZH2 activity leads to MYCN-driven replication stress, resulting in increased sensitivity to CHK1 inhibition, a finding with immediate clinical relevance.This article is highlighted in the In This Issue feature, p. 890.


Asunto(s)
Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Proliferación Celular , Humanos , Mutación , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética
9.
Br Dent J ; 227(12): 1035-1040, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31873258

RESUMEN

Complex oral surgery procedures are occurring more commonly in primary care, performed by both specialist oral surgeons and non-specialists/tier 2 providers. Practitioners in the UK are working in an increasingly litigious environment which is often at the forefront of a practitioner's mind and prevention of complaints and claims of negligence is vital in maintaining confidence in our practise. In this ever-increasing litigious environment practitioners need to be aware of the risks associated with oral surgery procedures and have to be able to assess and treatment plan patients effectively and safely. Consent is critical to all procedures, and the recent Montgomery case reinforces the importance of this process which must be individual to each patient. It is vital that as practitioners we are aware of our scope of practice and are able to fully inform patients of the risks and benefits of treatment. Practitioners must know their limits and when to refer on to secondary care. This paper discusses the essentials of risk management in oral surgery and describes key stages in the management of patients undergoing an oral surgery procedure. Initial assessment of the patient, which includes medical and dental history will be discussed followed by investigations to support the clinical diagnosis. Once diagnosis has been confirmed, the process of obtaining consent and the legal implications of this are discussed. Finally, a discussion on the appropriate evidence-based management of common complications which may arise when performing oral surgery will complete the paper.


Asunto(s)
Mala Praxis , Procedimientos Quirúrgicos Orales , Cirugía Bucal , Humanos , Atención Primaria de Salud , Gestión de Riesgos
10.
Prim Dent J ; 8(2): 49-52, 2019 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-31431206

RESUMEN

There are some dento-legal mantras that have, by default, come to be understood as the truth, for example: "if it ain't in the notes, it didn't happen" and "Good records, good defence; poor records, poor defence; no records, no defence." the opposite view was offered by the trial judge in a dental clinical negligence case in recent years. the judge's view can be summed up as: "if you don't make a note of something that has happened, you leave yourself at risk of proceedings like these."


Asunto(s)
Registros Odontológicos , Humanos
11.
Sci Immunol ; 4(38)2019 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-31444235

RESUMEN

A dense population of embryo-derived Langerhans cells (eLCs) is maintained within the sealed epidermis without contribution from circulating cells. When this network is perturbed by transient exposure to ultraviolet light, short-term LCs are temporarily reconstituted from an initial wave of monocytes but thought to be superseded by more permanent repopulation with undefined LC precursors. However, the extent to which this process is relevant to immunopathological processes that damage LC population integrity is not known. Using a model of allogeneic hematopoietic stem cell transplantation, where alloreactive T cells directly target eLCs, we have asked whether and how the original LC network is ultimately restored. We find that donor monocytes, but not dendritic cells, are the precursors of long-term LCs in this context. Destruction of eLCs leads to recruitment of a wave of monocytes that engraft in the epidermis and undergo a sequential pathway of differentiation via transcriptionally distinct EpCAM+ precursors. Monocyte-derived LCs acquire the capacity of self-renewal, and proliferation in the epidermis matched that of steady-state eLCs. However, we identified a bottleneck in the differentiation and survival of epidermal monocytes, which, together with the slow rate of renewal of mature LCs, limits repair of the network. Furthermore, replenishment of the LC network leads to constitutive entry of cells into the epidermal compartment. Thus, immune injury triggers functional adaptation of mechanisms used to maintain tissue-resident macrophages at other sites, but this process is highly inefficient in the skin.


Asunto(s)
Células de Langerhans/inmunología , Monocitos/inmunología , Animales , Células Cultivadas , Humanos , Células de Langerhans/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos
12.
Nat Cell Biol ; 20(12): 1389-1399, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30420664

RESUMEN

The linear-ubiquitin chain assembly complex (LUBAC) modulates signalling via various immune receptors. In tumour necrosis factor (TNF) signalling, linear (also known as M1) ubiquitin enables full gene activation and prevents cell death. However, the mechanisms underlying cell death prevention remain ill-defined. Here, we show that LUBAC activity enables TBK1 and IKKε recruitment to and activation at the TNF receptor 1 signalling complex (TNFR1-SC). While exerting only limited effects on TNF-induced gene activation, TBK1 and IKKε are essential to prevent TNF-induced cell death. Mechanistically, TBK1 and IKKε phosphorylate the kinase RIPK1 in the TNFR1-SC, thereby preventing RIPK1-dependent cell death. This activity is essential in vivo, as it prevents TNF-induced lethal shock. Strikingly, NEMO (also known as IKKγ), which mostly, but not exclusively, binds the TNFR1-SC via M1 ubiquitin, mediates the recruitment of the adaptors TANK and NAP1 (also known as AZI2). TANK is constitutively associated with both TBK1 and IKKε, while NAP1 is associated with TBK1. We discovered a previously unrecognized cell death checkpoint that is mediated by TBK1 and IKKε, and uncovered an essential survival function for NEMO, whereby it enables the recruitment and activation of these non-canonical IKKs to prevent TNF-induced cell death.


Asunto(s)
Quinasa I-kappa B/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Células A549 , Animales , Muerte Celular/efectos de los fármacos , Células Cultivadas , Células HeLa , Humanos , Ratones Noqueados , Fosforilación/efectos de los fármacos , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitinación/efectos de los fármacos
13.
JCI Insight ; 3(5)2018 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-29515032

RESUMEN

Graft-versus-host disease (GVHD) is a life-threatening complication of allogeneic stem cell transplantation induced by the influx of donor-derived effector T cells (TE) into peripheral tissues. Current treatment strategies rely on targeting systemic T cells; however, the precise location and nature of instructions that program TE to become pathogenic and trigger injury are unknown. We therefore used weighted gene coexpression network analysis to construct an unbiased spatial map of TE differentiation during the evolution of GVHD and identified wide variation in effector programs in mice and humans according to location. Idiosyncrasy of effector programming in affected organs did not result from variation in T cell receptor repertoire or the selection of optimally activated TE. Instead, TE were reprogrammed by tissue-autonomous mechanisms in target organs for site-specific proinflammatory functions that were highly divergent from those primed in lymph nodes. In the skin, we combined the correlation-based network with a module-based differential expression analysis and showed that Langerhans cells provided in situ instructions for a Notch-dependent T cell gene cluster critical for triggering local injury. Thus, the principal determinant of TE pathogenicity in GVHD is the final destination, highlighting the need for target organ-specific approaches to block immunopathology while avoiding global immune suppression.


Asunto(s)
Reprogramación Celular/inmunología , Enfermedad Injerto contra Huésped/inmunología , Células de Langerhans/inmunología , Piel/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Trasplante de Médula Ósea/efectos adversos , Células Cultivadas , Reprogramación Celular/genética , Modelos Animales de Enfermedad , Femenino , Regulación de la Expresión Génica/inmunología , Enfermedad Injerto contra Huésped/patología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Células de Langerhans/metabolismo , Lectinas Tipo C/genética , Lectinas Tipo C/metabolismo , Masculino , Lectinas de Unión a Manosa/genética , Lectinas de Unión a Manosa/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Familia de Multigenes/genética , Familia de Multigenes/inmunología , Cultivo Primario de Células , Receptores Notch/metabolismo , Piel/citología , Piel/patología , Linfocitos T Citotóxicos/metabolismo , Quimera por Trasplante , Trasplante Homólogo/efectos adversos
14.
Cancer Cell ; 32(5): 684-700.e9, 2017 11 13.
Artículo en Inglés | MEDLINE | ID: mdl-29107533

RESUMEN

Gain-of-function mutations in histone 3 (H3) variants are found in a substantial proportion of pediatric high-grade gliomas (pHGG), often in association with TP53 loss and platelet-derived growth factor receptor alpha (PDGFRA) amplification. Here, we describe a somatic mouse model wherein H3.3K27M and Trp53 loss alone are sufficient for neoplastic transformation if introduced in utero. H3.3K27M-driven lesions are clonal, H3K27me3 depleted, Olig2 positive, highly proliferative, and diffusely spreading, thus recapitulating hallmark molecular and histopathological features of pHGG. Addition of wild-type PDGFRA decreases latency and increases tumor invasion, while ATRX knockdown is associated with more circumscribed tumors. H3.3K27M-tumor cells serially engraft in recipient mice, and preliminary drug screening reveals mutation-specific vulnerabilities. Overall, we provide a faithful H3.3K27M-pHGG model which enables insights into oncohistone pathogenesis and investigation of future therapies.


Asunto(s)
Células Madre Embrionarias/metabolismo , Glioma/genética , Histonas/genética , Células-Madre Neurales/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/genética , Proteína p53 Supresora de Tumor/genética , Animales , Encéfalo/metabolismo , Encéfalo/patología , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Glioma/metabolismo , Glioma/patología , Humanos , Ratones , Mutación , Clasificación del Tumor , Invasividad Neoplásica , Interferencia de ARN , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteína Nuclear Ligada al Cromosoma X/genética , Proteína Nuclear Ligada al Cromosoma X/metabolismo
15.
PLoS Pathog ; 13(8): e1006524, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28837697

RESUMEN

Metabolic changes within the cell and its niche affect cell fate and are involved in many diseases and disorders including cancer and viral infections. Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS). KSHV latently infected cells express only a subset of viral genes, mainly located within the latency-associated region, among them 12 microRNAs. Notably, these miRNAs are responsible for inducing the Warburg effect in infected cells. Here we identify a novel mechanism enabling KSHV to manipulate the metabolic nature of the tumour microenvironment. We demonstrate that KSHV infected cells specifically transfer the virus-encoded microRNAs to surrounding cells via exosomes. This flow of genetic information results in a metabolic shift toward aerobic glycolysis in the surrounding non-infected cells. Importantly, this exosome-mediated metabolic reprogramming of neighbouring cells supports the growth of infected cells, thereby contributing to viral fitness. Finally, our data show that this miRNA transfer-based regulation of cell metabolism is a general mechanism used by other herpesviruses, such as EBV, as well as for the transfer of non-viral onco-miRs. This exosome-based crosstalk provides viruses with a mechanism for non-infectious transfer of genetic material without production of new viral particles, which might expose them to the immune system. We suggest that viruses and cancer cells use this mechanism to shape a specific metabolic niche that will contribute to their fitness.


Asunto(s)
Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/patogenicidad , Sarcoma de Kaposi/virología , Microambiente Tumoral/fisiología , Virulencia/fisiología , Western Blotting , Línea Celular , Exoma/fisiología , Infecciones por Herpesviridae/metabolismo , Herpesvirus Humano 8/fisiología , Humanos , MicroARNs/genética , Microscopía Electrónica de Transmisión , Reacción en Cadena de la Polimerasa , ARN Viral/genética , Sarcoma de Kaposi/metabolismo
16.
PLoS Genet ; 13(2): e1006587, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28187197

RESUMEN

The polarization of CD4+ T cells into distinct T helper cell lineages is essential for protective immunity against infection, but aberrant T cell polarization can cause autoimmunity. The transcription factor T-bet (TBX21) specifies the Th1 lineage and represses alternative T cell fates. Genome-wide association studies have identified single nucleotide polymorphisms (SNPs) that may be causative for autoimmune diseases. The majority of these polymorphisms are located within non-coding distal regulatory elements. It is considered that these genetic variants contribute to disease by altering the binding of regulatory proteins and thus gene expression, but whether these variants alter the binding of lineage-specifying transcription factors has not been determined. Here, we show that SNPs associated with the mucosal inflammatory diseases Crohn's disease, ulcerative colitis (UC) and celiac disease, but not rheumatoid arthritis or psoriasis, are enriched at T-bet binding sites. Furthermore, we identify disease-associated variants that alter T-bet binding in vitro and in vivo. ChIP-seq for T-bet in individuals heterozygous for the celiac disease-associated SNPs rs1465321 and rs2058622 and the IBD-associated SNPs rs1551398 and rs1551399, reveals decreased binding to the minor disease-associated alleles. Furthermore, we show that rs1465321 is an expression quantitative trait locus (eQTL) for the neighboring gene IL18RAP, with decreased T-bet binding associated with decreased expression of this gene. These results suggest that genetic polymorphisms may predispose individuals to mucosal autoimmune disease through alterations in T-bet binding. Other disease-associated variants may similarly act by modulating the binding of lineage-specifying transcription factors in a tissue-selective and disease-specific manner.


Asunto(s)
Enfermedad Celíaca/genética , Colitis Ulcerosa/genética , Enfermedad de Crohn/genética , Predisposición Genética a la Enfermedad/genética , Polimorfismo de Nucleótido Simple , Proteínas de Dominio T Box/genética , Animales , Sitios de Unión/genética , Western Blotting , Linfocitos T CD4-Positivos/metabolismo , Enfermedad Celíaca/metabolismo , Células Cultivadas , Colitis Ulcerosa/metabolismo , Enfermedad de Crohn/metabolismo , Expresión Génica , Estudio de Asociación del Genoma Completo/métodos , Humanos , Subunidad beta del Receptor de Interleucina-18/genética , Subunidad beta del Receptor de Interleucina-18/metabolismo , Ratones Noqueados , Unión Proteica/genética , Secuencias Reguladoras de Ácidos Nucleicos/genética , Proteínas de Dominio T Box/metabolismo , Células TH1/metabolismo
18.
J Clin Oncol ; 34(34): 4132-4141, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27863190

RESUMEN

Purpose In squamous cell carcinomas of the head and neck (HNSCC), the increasing incidence of oropharyngeal squamous cell carcinomas (OPSCCs) is attributable to human papillomavirus (HPV) infection. Despite commonly presenting at late stage, HPV-driven OPSCCs are associated with improved prognosis compared with HPV-negative disease. HPV DNA is also detectable in nonoropharyngeal (non-OPSCC), but its pathogenic role and clinical significance are unclear. The objectives of this study were to determine whether HPV plays a causal role in non-OPSCC and to investigate whether HPV confers a survival benefit in these tumors. Methods Meta-analysis was used to build a cross-tissue gene-expression signature for HPV-driven cancer. Classifiers trained by machine-learning approaches were used to predict the HPV status of 520 HNSCCs profiled by The Cancer Genome Atlas project. DNA methylation data were similarly used to classify 464 HNSCCs and these analyses were integrated with genomic, histopathology, and survival data to permit a comprehensive comparison of HPV transcript-positive OPSCC and non-OPSCC. Results HPV-driven tumors accounted for 4.1% of non-OPSCCs. Regardless of anatomic site, HPV+ HNSCCs shared highly similar gene expression and DNA methylation profiles; nonkeratinizing, basaloid histopathological features; and lack of TP53 or CDKN2A alterations. Improved overall survival, however, was largely restricted to HPV-driven OPSCCs, which were associated with increased levels of tumor-infiltrating lymphocytes compared with HPV-driven non-OPSCCs. Conclusion Our analysis identified a causal role for HPV in transcript-positive non-OPSCCs throughout the head and neck. Notably, however, HPV-driven non-OPSCCs display a distinct immune microenvironment and clinical behavior compared with HPV-driven OPSCCs.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/virología , Neoplasias de Cabeza y Cuello/virología , Papillomaviridae/genética , Infecciones por Papillomavirus/virología , Metilación de ADN , Pruebas de ADN del Papillomavirus Humano , Humanos , Neoplasias Orofaríngeas/virología , Pronóstico , Análisis de Supervivencia
19.
Eur J Immunol ; 46(1): 192-203, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26464217

RESUMEN

Dendritic cells (DCs) play a vital role in innate and adaptive immunities. Inducible depletion of CD11c(+) DCs engineered to express a high-affinity diphtheria toxin receptor has been a powerful tool to dissect DC function in vivo. However, despite reports showing that loss of DCs induces transient monocytosis, the monocyte population that emerges and the potential impact of monocytes on studies of DC function have not been investigated. We found that depletion of CD11c(+) cells from CD11c.DTR mice induced the expansion of a variant CD64(+) Ly6C(+) monocyte population in the spleen and blood that was distinct from conventional monocytes. Expansion of CD64(+) Ly6C(+) monocytes was independent of mobilization from the BM via CCR2 but required the cytokine, G-CSF. Indeed, this population was also expanded upon exposure to exogenous G-CSF in the absence of DC depletion. CD64(+) Ly6C(+) monocytes were characterized by upregulation of innate signaling apparatus despite the absence of inflammation, and an increased capacity to produce TNF-α following LPS stimulation. Thus, depletion of CD11c(+) cells induces expansion of a unique CD64(+) Ly6C(+) monocyte population poised to synthesize TNF-α. This finding will require consideration in experiments using depletion strategies to test the role of CD11c(+) DCs in immunity.


Asunto(s)
Células Dendríticas/inmunología , Monocitos/citología , Monocitos/inmunología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Antígenos Ly/inmunología , Antígeno CD11c/inmunología , Citometría de Flujo , Ratones , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Receptores de IgG/inmunología , Factor de Necrosis Tumoral alfa/inmunología
20.
Sci Transl Med ; 7(281): 281ra42, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25834108

RESUMEN

Granulocyte colony-stimulating factor (G-CSF) is routinely used to collect peripheral blood stem cells (PBSCs) from healthy donors for allogeneic hematopoietic stem cell transplantation (allo-HSCT). We show that, in both humans and mice, G-CSF mobilizes a subset of CD34(+) cells with mature monocyte features. These cells, which are phenotypically and functionally conserved in mice and humans, are transcriptionally distinct from myeloid and monocytic precursors but similar to mature monocytes and endowed with immunosuppressive properties. In response to interferon-γ released by activated T cells, these cells produce nitric oxide, which induces allogeneic T cell death both in vitro and in vivo. These apoptotic T cells are engulfed by macrophages that release transforming growth factor-ß and promote regulatory T cell expansion. Indeed, the fraction of CD34(+) monocytes in peripheral blood CD34(+) cells inversely correlates with the incidence of acute graft-versus-host disease (GVHD) in humans. Therefore, G-CSF-mobilized cells are an attractive candidate population to be expanded ex vivo for cellular therapy against GVHD.


Asunto(s)
Antígenos CD34/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética , Monocitos/metabolismo , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Tolerancia Inmunológica/efectos de los fármacos , Terapia de Inmunosupresión , Interferón gamma/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Ratones , Monocitos/efectos de los fármacos , Óxido Nítrico/biosíntesis , Óxido Nítrico Sintasa de Tipo II/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...