Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Blood ; 138(10): 858-870, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34036317

RESUMEN

Invariant natural killer T (iNKT) cells are a T-cell subset with potent immunomodulatory properties. Experimental evidence in mice and observational studies in humans indicate that iNKT cells have antitumor potential as well as the ability to suppress acute and chronic graft-versus-host-disease (GVHD). Murine iNKT cells differentiate during thymic development into iNKT1, iNKT2, and iNKT17 sublineages, which differ transcriptomically and epigenomically and have subset-specific developmental requirements. Whether distinct iNKT sublineages also differ in their antitumor effect and their ability to suppress GVHD is currently unknown. In this work, we generated highly purified murine iNKT sublineages, characterized their transcriptomic and epigenomic landscape, and assessed specific functions. We show that iNKT2 and iNKT17, but not iNKT1, cells efficiently suppress T-cell activation in vitro and mitigate murine acute GVHD in vivo. Conversely, we show that iNKT1 cells display the highest antitumor activity against murine B-cell lymphoma cells both in vitro and in vivo. Thus, we report for the first time that iNKT sublineages have distinct and different functions, with iNKT1 cells having the highest antitumor activity and iNKT2 and iNKT17 cells having immune-regulatory properties. These results have important implications for the translation of iNKT cell therapies to the clinic for cancer immunotherapy as well as for the prevention and treatment of GVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Efecto Injerto vs Tumor/inmunología , Activación de Linfocitos , Linfoma de Células B , Células T Asesinas Naturales/inmunología , Neoplasias Experimentales , Animales , Epigenómica , Femenino , Perfilación de la Expresión Génica , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/prevención & control , Linfoma de Células B/inmunología , Linfoma de Células B/terapia , Masculino , Ratones , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/terapia
2.
Nature ; 523(7561): 486-90, 2015 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-26083756

RESUMEN

Cell-to-cell variation is a universal feature of life that affects a wide range of biological phenomena, from developmental plasticity to tumour heterogeneity. Although recent advances have improved our ability to document cellular phenotypic variation, the fundamental mechanisms that generate variability from identical DNA sequences remain elusive. Here we reveal the landscape and principles of mammalian DNA regulatory variation by developing a robust method for mapping the accessible genome of individual cells by assay for transposase-accessible chromatin using sequencing (ATAC-seq) integrated into a programmable microfluidics platform. Single-cell ATAC-seq (scATAC-seq) maps from hundreds of single cells in aggregate closely resemble accessibility profiles from tens of millions of cells and provide insights into cell-to-cell variation. Accessibility variance is systematically associated with specific trans-factors and cis-elements, and we discover combinations of trans-factors associated with either induction or suppression of cell-to-cell variability. We further identify sets of trans-factors associated with cell-type-specific accessibility variance across eight cell types. Targeted perturbations of cell cycle or transcription factor signalling evoke stimulus-specific changes in this observed variability. The pattern of accessibility variation in cis across the genome recapitulates chromosome compartments de novo, linking single-cell accessibility variation to three-dimensional genome organization. Single-cell analysis of DNA accessibility provides new insight into cellular variation of the 'regulome'.


Asunto(s)
Células/metabolismo , Cromatina/genética , Cromatina/metabolismo , Epigenómica , Análisis de la Célula Individual/métodos , Animales , Compartimento Celular , Ciclo Celular/genética , Línea Celular , Células/clasificación , ADN/genética , ADN/metabolismo , Epigénesis Genética , Genoma Humano/genética , Humanos , Microfluídica , Transducción de Señal , Factores de Transcripción/metabolismo , Transposasas/metabolismo
3.
Nature ; 478(7368): 197-203, 2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-21976023

RESUMEN

Activation of the aryl hydrocarbon receptor (AHR) by environmental xenobiotic toxic chemicals, for instance 2,3,7,8-tetrachlorodibenzo-p-dioxin (dioxin), has been implicated in a variety of cellular processes such as embryogenesis, transformation, tumorigenesis and inflammation. But the identity of an endogenous ligand activating the AHR under physiological conditions in the absence of environmental toxic chemicals is still unknown. Here we identify the tryptophan (Trp) catabolite kynurenine (Kyn) as an endogenous ligand of the human AHR that is constitutively generated by human tumour cells via tryptophan-2,3-dioxygenase (TDO), a liver- and neuron-derived Trp-degrading enzyme not yet implicated in cancer biology. TDO-derived Kyn suppresses antitumour immune responses and promotes tumour-cell survival and motility through the AHR in an autocrine/paracrine fashion. The TDO-AHR pathway is active in human brain tumours and is associated with malignant progression and poor survival. Because Kyn is produced during cancer progression and inflammation in the local microenvironment in amounts sufficient for activating the human AHR, these results provide evidence for a previously unidentified pathophysiological function of the AHR with profound implications for cancer and immune biology.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Glioma/metabolismo , Glioma/patología , Quinurenina/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Comunicación Autocrina , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/inmunología , Línea Celular Tumoral , Supervivencia Celular , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Glioma/genética , Glioma/inmunología , Humanos , Quinurenina/inmunología , Quinurenina/farmacología , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante de Neoplasias , Comunicación Paracrina , Receptores de Hidrocarburo de Aril/inmunología , Triptófano/metabolismo , Triptófano Oxigenasa/deficiencia , Triptófano Oxigenasa/genética , Triptófano Oxigenasa/metabolismo
4.
Proc Natl Acad Sci U S A ; 110(36): 14735-40, 2013 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-23959874

RESUMEN

Disruption of the blood-brain barrier (BBB) is a hallmark of acute inflammatory lesions in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis. This disruption may precede and facilitate the infiltration of encephalitogenic T cells. The signaling events that lead to this BBB disruption are incompletely understood but appear to involve dysregulation of tight-junction proteins such as claudins. Pharmacological interventions aiming at stabilizing the BBB in MS might have therapeutic potential. Here, we show that the orally available small molecule LY-317615, a synthetic bisindolylmaleimide and inhibitor of protein kinase Cß, which is clinically under investigation for the treatment of cancer, suppresses the transmigration of activated T cells through an inflamed endothelial cell barrier, where it leads to the induction of the tight-junction molecules zona occludens-1, claudin 3, and claudin 5 and other pathways critically involved in transendothelial leukocyte migration. Treatment of mice with ongoing experimental autoimmune encephalomyelitis with LY-317615 ameliorates inflammation, demyelination, axonal damage, and clinical symptoms. Although LY-317615 dose-dependently suppresses T-cell proliferation and cytokine production independent of antigen specificity, its therapeutic effect is abrogated in a mouse model requiring pertussis toxin. This abrogation indicates that the anti-inflammatory and clinical efficacy is mainly mediated by stabilization of the BBB, thus suppressing the transmigration of encephalitogenic T cells. Collectively, our data suggest the involvement of endothelial protein kinase Cß in stabilizing the BBB in autoimmune neuroinflammation and imply a therapeutic potential of BBB-targeting agents such as LY-317615 as therapeutic approaches for MS.


Asunto(s)
Barrera Hematoencefálica/efectos de los fármacos , Encefalomielitis Autoinmune Experimental/prevención & control , Indoles/farmacología , Proteína Quinasa C beta/antagonistas & inhibidores , Animales , Barrera Hematoencefálica/inmunología , Proliferación Celular/efectos de los fármacos , Claudina-3/inmunología , Claudina-3/metabolismo , Claudina-5/inmunología , Claudina-5/metabolismo , Citocinas/inmunología , Citocinas/metabolismo , Enfermedades Desmielinizantes/inmunología , Enfermedades Desmielinizantes/prevención & control , Relación Dosis-Respuesta a Droga , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/inmunología , Células Endoteliales/metabolismo , Femenino , Perfilación de la Expresión Génica , Inmunohistoquímica , Indoles/inmunología , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Microscopía Confocal , Proteína Quinasa C beta/inmunología , Proteína Quinasa C beta/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/inmunología , Uniones Estrechas/metabolismo , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/inmunología , Proteína de la Zonula Occludens-1/inmunología , Proteína de la Zonula Occludens-1/metabolismo
5.
Glia ; 63(1): 78-90, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25132599

RESUMEN

Tryptophan catabolism is increasingly recognized as a key and druggable molecular mechanism active in cancer, immune, and glioneural cells and involved in the modulation of antitumor immunity, autoimmunity and glioneural function. In addition to the pivotal rate limiting enzyme indoleamine-2,3-dioxygenase, expression of tryptophan-2,3-dioxygenase (TDO) has recently been described as an alternative pathway responsible for constitutive tryptophan degradation in malignant gliomas and other types of cancer. In addition, TDO has been implicated as a key regulator of neurotoxicity involved in neurodegenerative diseases and ageing. The pathways regulating TDO expression, however, are largely unknown. Here, a siRNA-based transcription factor profiling in human glioblastoma cells revealed that the expression of human TDO is suppressed by endogenous glucocorticoid signaling. Similarly, treatment of glioblastoma cells with the synthetic glucocorticoid dexamethasone led to a reduction of TDO expression and activity in vitro and in vivo. TDO inhibition was dependent on the immunophilin FKBP52, whose FK1 domain physically interacted with the glucocorticoid receptor as demonstrated by bimolecular fluorescence complementation and in situ proximity ligation assays. Accordingly, gene expression profile analyses revealed negative correlation of FKBP52 and TDO in glial and neural tumors and in normal brain. Knockdown of FKBP52 and treatment with the FK-binding immunosuppressant FK506 enhanced TDO expression and activity in glioblastoma cells. In summary, we identify a novel steroid-responsive FKBP52-dependent pathway suppressing the expression and activity of TDO, a central and rate-limiting enzyme in tryptophan metabolism, in human gliomas.


Asunto(s)
Glioblastoma/metabolismo , Transducción de Señal/efectos de los fármacos , Proteínas de Unión a Tacrolimus/metabolismo , Triptófano Oxigenasa/metabolismo , Triptófano/metabolismo , Envejecimiento/efectos de los fármacos , Animales , Línea Celular Tumoral , Dexametasona/farmacología , Glioblastoma/tratamiento farmacológico , Humanos , Ratones , Tacrolimus/farmacología , Triptófano Oxigenasa/antagonistas & inhibidores
6.
Elife ; 112022 12 29.
Artículo en Inglés | MEDLINE | ID: mdl-36579891

RESUMEN

HOTAIR is a 2.2-kb long noncoding RNA (lncRNA) whose dysregulation has been linked to oncogenesis, defects in pattern formation during early development, and irregularities during the process of epithelial-to-mesenchymal transition (EMT). However, the oncogenic transformation determined by HOTAIR in vivo and its impact on chromatin dynamics are incompletely understood. Here, we generate a transgenic mouse model with doxycycline-inducible expression of human HOTAIR in the context of the MMTV-PyMT breast cancer-prone background to systematically interrogate the cellular mechanisms by which human HOTAIR lncRNA acts to promote breast cancer progression. We show that sustained high levels of HOTAIR over time increased breast metastatic capacity and invasiveness in breast cancer cells, promoting migration and subsequent metastasis to the lung. Subsequent withdrawal of HOTAIR overexpression reverted the metastatic phenotype, indicating oncogenic lncRNA addiction. Furthermore, HOTAIR overexpression altered both the cellular transcriptome and chromatin accessibility landscape of multiple metastasis-associated genes and promoted EMT. These alterations are abrogated within several cell cycles after HOTAIR expression is reverted to basal levels, indicating an erasable lncRNA-associated epigenetic memory. These results suggest that a continual role for HOTAIR in programming a metastatic gene regulatory program. Targeting HOTAIR lncRNA may potentially serve as a therapeutic strategy to ameliorate breast cancer progression.


Asunto(s)
Neoplasias de la Mama , ARN Largo no Codificante , Animales , Femenino , Humanos , Ratones , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Proliferación Celular , Cromatina , Regulación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Ratones Transgénicos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Pulmonares/secundario
7.
Life Sci Alliance ; 4(8)2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34145026

RESUMEN

Sepsis, sequela of bloodstream infections and dysregulated host responses, is a leading cause of death globally. Neutrophils tightly regulate responses to pathogens to prevent organ damage. Profiling early host epigenetic responses in neutrophils may aid in disease recognition. We performed assay for transposase-accessible chromatin (ATAC)-seq of human neutrophils challenged with six toll-like receptor ligands and two organisms; and RNA-seq after Escherichia coli exposure for 1 and 4 h along with ATAC-seq. ATAC-seq of neutrophils facilitates detection of pathogen DNA. In addition, despite similarities in genomic distribution of differential chromatin changes across challenges, only a fraction overlaps between the challenges. Ligands depict shared signatures, but majority are unique in position, function, and challenge. Epigenomic changes are plastic, only ∼120 are shared by E coli challenges over time, resulting in varied differential genes and associated processes. We identify three classes of gene regulation, chromatin access changes in the promoter; changes in the promoter and distal enhancers; and controlling expression through changes solely in distal enhancers. These and transcription factor footprinting reveal timely and challenge specific mechanisms of transcriptional regulation in neutrophils.


Asunto(s)
Secuenciación de Inmunoprecipitación de Cromatina/métodos , Infecciones por Escherichia coli/genética , Escherichia coli/patogenicidad , Neutrófilos/microbiología , Sepsis/genética , Adulto , Epigenómica , Femenino , Regulación de la Expresión Génica , Humanos , Modelos Biológicos , Neutrófilos/química , Regiones Promotoras Genéticas , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN , Factores de Tiempo
8.
Stem Cells ; 27(4): 909-19, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19353519

RESUMEN

Mesenchymal stem cells (MSC) display unique suppressive properties on T-cell immunity, thus representing an attractive vehicle for the treatment of conditions associated with harmful T-cell responses such as organ-specific autoimmunity and graft-versus-host disease. Toll-like receptors (TLR) are primarily expressed on antigen-presenting cells and recognize conserved pathogen-derived components. Ligation of TLR activates multiple innate and adaptive immune response pathways to eliminate and protect against invading pathogens. In this work, we show that TLR expressed on human bone marrow-derived MSC enhanced the immunosuppressive phenotype of MSC. Immunosuppression mediated by TLR was dependent on the production of immunosuppressive kynurenines by the tryptophan-degrading enzyme indoleamine-2,3-dioxygenase-1 (IDO1). Induction of IDO1 by TLR involved an autocrine interferon (IFN)-beta signaling loop, which was dependent on protein kinase R (PKR), but independent of IFN-gamma. These data define a new role for TLR in MSC immunobiology, which is to augment the immunosuppressive properties of MSC in the absence of IFN-gamma rather than inducing proinflammatory immune response pathways. PKR and IFN-beta play a central, previously unidentified role in orchestrating the production of immunosuppressive kynurenines by MSC.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/inmunología , Interferón beta/inmunología , Células Madre Mesenquimatosas/inmunología , Transducción de Señal/inmunología , Receptores Toll-Like/inmunología , eIF-2 Quinasa/inmunología , Western Blotting , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Diferenciación Celular/inmunología , Cromatografía Líquida de Alta Presión , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Interferón beta/metabolismo , Quinurenina/biosíntesis , Quinurenina/inmunología , Prueba de Cultivo Mixto de Linfocitos , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Receptores Toll-Like/metabolismo , eIF-2 Quinasa/metabolismo
9.
Cell Death Dis ; 11(2): 102, 2020 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-32029721

RESUMEN

Despite the fact that Otto H. Warburg discovered the Warburg effect almost one hundred years ago, why cancer cells waste most of the glucose carbon as lactate remains an enigma. Warburg proposed a connection between the Warburg effect and cell dedifferentiation. Hypoxia is a common tumor microenvironmental stress that induces the Warburg effect and blocks tumor cell differentiation. The underlying mechanism by which this occurs is poorly understood, and no effective therapeutic strategy has been developed to overcome this resistance to differentiation. Using a neuroblastoma differentiation model, we discovered that hypoxia repressed cell differentiation through reducing cellular acetyl-CoA levels, leading to reduction of global histone acetylation and chromatin accessibility. The metabolic switch triggering this global histone hypoacetylation was the induction of pyruvate dehydrogenase kinases (PDK1 and PDK3). Inhibition of PDKs using dichloroacetate (DCA) restored acetyl-CoA generation and histone acetylation under hypoxia. Knocking down PDK1 induced neuroblastoma cell differentiation, highlighting the critical role of PDK1 in cell fate control. Importantly, acetate or glycerol triacetate (GTA) supplementation restored differentiation markers expression and neuron differentiation under hypoxia. Moreover, ATAC-Seq analysis demonstrated that hypoxia treatment significantly reduced chromatin accessibility at RAR/RXR binding sites, which can be restored by acetate supplementation. In addition, hypoxia-induced histone hypermethylation by increasing 2-hydroxyglutarate (2HG) and reducing α-ketoglutarate (αKG). αKG supplementation reduced histone hypermethylation upon hypoxia, but did not restore histone acetylation or differentiation markers expression. Together, these findings suggest that diverting pyruvate flux away from acetyl-CoA generation to lactate production is the key mechanism that Warburg effect drives dedifferentiation and tumorigenesis. We propose that combining differentiation therapy with acetate/GTA supplementation might represent an effective therapy against neuroblastoma.


Asunto(s)
Acetatos/farmacología , Antineoplásicos/farmacología , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Neuroblastoma/tratamiento farmacológico , Neurogénesis/efectos de los fármacos , Efecto Warburg en Oncología/efectos de los fármacos , Acetilación , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Histonas/metabolismo , Humanos , Masculino , Ratones , Neuroblastoma/genética , Neuroblastoma/metabolismo , Neuroblastoma/patología , Proyección Neuronal/efectos de los fármacos , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/metabolismo , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Hipoxia Tumoral , Microambiente Tumoral , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cell Rep ; 33(6): 108356, 2020 11 10.
Artículo en Inglés | MEDLINE | ID: mdl-33176144

RESUMEN

Fibroblast heterogeneity has been shown within the unwounded mouse dorsal dermis, with fibroblast subpopulations being identified according to anatomical location and embryonic lineage. Using lineage tracing, we demonstrate that paired related homeobox 1 (Prrx1)-expressing fibroblasts are responsible for acute and chronic fibroses in the ventral dermis. Single-cell transcriptomics further corroborated the inherent fibrotic characteristics of Prrx1 fibroblasts during wound repair. In summary, we identify and characterize a fibroblast subpopulation in the mouse ventral dermis with intrinsic scar-forming potential.


Asunto(s)
Dermis/metabolismo , Fibroblastos/metabolismo , Proteínas de Homeodominio/metabolismo , Animales , Humanos , Ratones
11.
Elife ; 82019 04 09.
Artículo en Inglés | MEDLINE | ID: mdl-30958261

RESUMEN

Simultaneous measurement of cell lineage and cell fates is a longstanding goal in biomedicine. Here we describe EMBLEM, a strategy to track cell lineage using endogenous mitochondrial DNA variants in ATAC-seq data. We show that somatic mutations in mitochondrial DNA can reconstruct cell lineage relationships at single cell resolution with high sensitivity and specificity. Using EMBLEM, we define the genetic and epigenomic clonal evolution of hematopoietic stem cells and their progenies in patients with acute myeloid leukemia. EMBLEM extends lineage tracing to any eukaryotic organism without genetic engineering.


Asunto(s)
Linaje de la Célula , Técnicas Citológicas/métodos , ADN Mitocondrial/genética , Células Madre Hematopoyéticas/patología , Leucemia Mieloide Aguda/patología , Mutación , Evolución Clonal , Humanos , Sensibilidad y Especificidad
12.
Nat Commun ; 9(1): 4590, 2018 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-30389926

RESUMEN

Here we introduce Protein-indexed Assay of Transposase Accessible Chromatin with sequencing (Pi-ATAC) that combines single-cell chromatin and proteomic profiling. In conjunction with DNA transposition, the levels of multiple cell surface or intracellular protein epitopes are recorded by index flow cytometry and positions in arrayed microwells, and then subject to molecular barcoding for subsequent pooled analysis. Pi-ATAC simultaneously identifies the epigenomic and proteomic heterogeneity in individual cells. Pi-ATAC reveals a casual link between transcription factor abundance and DNA motif access, and deconvolute cell types and states in the tumor microenvironment in vivo. We identify a dominant role for hypoxia, marked by HIF1α protein, in the tumor microvenvironment for shaping the regulome in a subset of epithelial tumor cells.


Asunto(s)
ADN/genética , Ambiente , Epigenómica , Epítopos/metabolismo , Proteínas/metabolismo , Análisis de la Célula Individual , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Hipoxia de la Célula/genética , Línea Celular Tumoral , Cromatina/metabolismo , Epigénesis Genética , Molécula de Adhesión Celular Epitelial/metabolismo , Linfocitos/metabolismo , Ratones , Motivos de Nucleótidos/genética , Reproducibilidad de los Resultados , Análisis de Secuencia de ADN , Factores de Transcripción/metabolismo , Transposasas/metabolismo
13.
Genome Biol ; 18(1): 15, 2017 01 24.
Artículo en Inglés | MEDLINE | ID: mdl-28118844

RESUMEN

BACKGROUND: Cell-to-cell heterogeneity is a major driver of cancer evolution, progression, and emergence of drug resistance. Epigenomic variation at the single-cell level can rapidly create cancer heterogeneity but is difficult to detect and assess functionally. RESULTS: We develop a strategy to bridge the gap between measurement and function in single-cell epigenomics. Using single-cell chromatin accessibility and RNA-seq data in K562 leukemic cells, we identify the cell surface marker CD24 as co-varying with chromatin accessibility changes linked to GATA transcription factors in single cells. Fluorescence-activated cell sorting of CD24 high versus low cells prospectively isolated GATA1 and GATA2 high versus low cells. GATA high versus low cells express differential gene regulatory networks, differential sensitivity to the drug imatinib mesylate, and differential self-renewal capacity. Lineage tracing experiments show that GATA/CD24hi cells have the capability to rapidly reconstitute the heterogeneity within the entire starting population, suggesting that GATA expression levels drive a phenotypically relevant source of epigenomic plasticity. CONCLUSION: Single-cell chromatin accessibility can guide prospective characterization of cancer heterogeneity. Epigenomic subpopulations in cancer impact drug sensitivity and the clonal dynamics of cancer evolution.


Asunto(s)
Epigénesis Genética , Epigenómica , Heterogeneidad Genética , Variación Genética , Neoplasias/genética , Análisis de la Célula Individual , Antígenos de Superficie/química , Antígenos de Superficie/genética , Antígenos de Superficie/metabolismo , Biomarcadores , Línea Celular Tumoral , Epigenómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Inmunofenotipificación , Células K562 , Neoplasias/metabolismo , Motivos de Nucleótidos , Reproducibilidad de los Resultados , Análisis de la Célula Individual/métodos
14.
Oncotarget ; 5(4): 1038-51, 2014 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-24657910

RESUMEN

Indoleamine-2,3-dioxygenase (IDO) inhibitors have entered clinical trials based on their ability to restore anti-tumor immunity in preclinical studies. However, the mechanisms leading to constitutive expression of IDO in human tumors are largely unknown. Here we analyzed the pathways mediating constitutive IDO expression in human cancer. IDO-positive tumor cells and tissues showed basal phosphorylation and acetylation of STAT3 as evidenced by western blotting and immunoprecipitation. Inhibition of IL-6 or STAT3 using siRNA and/or pharmacological inhibitors reduced IDO mRNA and protein expression as well as kynurenine formation. In turn, IDO enzymatic activity activated the AHR as shown by the induction of AHR target genes. IDO-mediated AHR activation induced IL-6 expression, while inhibition or knockdown of the AHR reduced IL-6 expression. IDO activity thus sustains its own expression via an autocrine AHR-IL-6-STAT3 signaling loop. Inhibition of the AHR-IL-6-STAT3 signaling loop restored T-cell proliferation in mixed leukocyte reactions performed in the presence of IDO-expressing human cancer cells. Identification of the IDO-AHR-IL-6-STAT3 signaling loop maintaining IDO expression in human cancers reveals novel therapeutic targets for the inhibition of this core pathway promoting immunosuppression of human cancers. The relevance of the IDO-AHR-IL-6-STAT3 transcriptional circuit is underscored by the finding that high expression of its members IDO, STAT3 and the AHR target gene CYP1B1 is associated with reduced relapse-free survival in lung cancer patients.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenasa/biosíntesis , Interleucina-6/metabolismo , Neoplasias/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo , Factor de Transcripción STAT3/metabolismo , Acetilación , Apoptosis/fisiología , Comunicación Autocrina , Carcinoma de Pulmón de Células no Pequeñas/enzimología , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Carcinoma de Pulmón de Células no Pequeñas/patología , Proliferación Celular/fisiología , Femenino , Humanos , Inmunohistoquímica , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias Pulmonares/enzimología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Neoplasias/enzimología , Neoplasias/patología , Neoplasias Ováricas/enzimología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Fosforilación , Transducción de Señal
15.
PLoS One ; 7(10): e47663, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23077658

RESUMEN

Enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of the Polycomb-repressive complex 2 (PRC2) that epigenetically silences gene transcription through histone H3 lysine trimethylation (H3K27me3). EZH2 has been implicated in stem cell maintenance and is overexpressed in hematological and solid malignancie`s including malignant glioma. EZH2 is thought to promote tumor progression by silencing tumor suppressor genes. Hence pharmacological disruption of the PRC2 is an attractive therapeutic strategy for cancer treatment. Here we show that EZH2 is expressed in human glioma and correlates with malignancy. Silencing of EZH2 reduced glioma cell proliferation and invasiveness. While we did not observe induction of cell cycle-associated tumor suppressor genes by silencing or pharmacological inhibition of EZH2, microarray analyses demonstrated a strong transcriptional reduction of the AXL receptor kinase. Neither histone nor DNA methylation appeared to be involved in the positive regulation of AXL by EZH2. Silencing AXL mimicked the antiinvasive effects of EZH2 knockdown. Finally, AXL expression is found in human gliomas with high EZH2 expression. Collectively these data suggest that EZH2 drives glioma invasiveness via transcriptional control of AXL independent of histone or DNA methylation.


Asunto(s)
Movimiento Celular/genética , Glioblastoma/metabolismo , Complejo Represivo Polycomb 2 , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Línea Celular Tumoral , Transformación Celular Neoplásica , Metilación de ADN , Proteína Potenciadora del Homólogo Zeste 2 , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Glioblastoma/genética , Glioma/genética , Humanos , Invasividad Neoplásica , Complejo Represivo Polycomb 2/antagonistas & inhibidores , Complejo Represivo Polycomb 2/genética , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Tirosina Quinasa del Receptor Axl
16.
PLoS One ; 6(5): e19823, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21625531

RESUMEN

1-methyl-D-tryptophan (1-D-MT) is currently being used in clinical trials in patients with relapsed or refractory solid tumors with the aim of inhibiting indoleamine-2,3-dioxygenase (IDO)-mediated tumor immune escape. IDO is expressed in tumors and tumor-draining lymph nodes and degrades tryptophan (trp) to create an immunsuppressive micromilieu both by depleting trp and by accumulating immunosuppressive metabolites of the kynurenine (kyn) pathway. Here we show that proliferation of alloreactive T-cells cocultured with IDO1-positive human cancer cells paradoxically was inhibited by 1-D-MT. Surprisingly incubation with 1-D-MT increased kyn production of human cancer cells. Cell-free assays revealed that 1-D-MT did not alter IDO1 enzymatic activity. Instead, 1-D-MT induced IDO1 mRNA and protein expression through pathways involving p38 MAPK and JNK signalling. Treatment of cancer patients with 1-D-MT has transcriptional effects that may promote rather than suppress anti-tumor immune escape by increasing IDO1 in the cancer cells. These off-target effects should be carefully analyzed in the ongoing clinical trials with 1-D-MT.


Asunto(s)
Indolamina-Pirrol 2,3,-Dioxigenasa/antagonistas & inhibidores , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/enzimología , Triptófano/análogos & derivados , Western Blotting , Ciclo Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Técnicas para Inmunoenzimas , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , ARN Mensajero/genética , ARN Interferente Pequeño/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Linfocitos T/metabolismo , Triptófano/farmacología , Células Tumorales Cultivadas , Regulación hacia Arriba , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA