Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Mol Ther Nucleic Acids ; 28: 877-891, 2022 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-35694213

RESUMEN

Advances in gene therapy research have resulted in the successful development of new therapies for clinical use. Here, we explored a gene targeting approach to deplete ephrinB2 from colorectal cancer cells using an inducible lentiviral vector. EphrinB2, a transmembrane ephrin ligand, promotes colorectal cancer cell growth and viability and predicts poor patient survival when expressed at high levels in colorectal cancer tissues. We discovered that lentiviral vector integration and expression in the host DNA frequently drive divergent host gene transcription, generating antisense reads coupled with splicing events and generation of chimeric vector/host transcripts. Antisense transcription of host DNA was linked to development of an integrated stress response and cell death. Despite recent successes, off-target effects remain a concern in genetic medicine. Our results provide evidence that divergent gene transcription is a previously unrecognized off-target effect of lentiviral vector integration with built-in properties for regulation of gene expression.

2.
J Exp Clin Cancer Res ; 41(1): 208, 2022 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-35754026

RESUMEN

BACKGROUND: There is no universally accepted treatment for patients with advanced papillary renal cell carcinoma (PRCC). The presence of activating mutations in MET, as well as gain of chromosome 7, where the MET gene is located, are the most common genetic alterations associated with PRCC, leading to the clinical evaluation of MET tyrosine kinase inhibitors (TKIs) in this cancer. However, TKIs targeting MET selectively, as well as multitargeted TKIs with activity against MET demonstrate modest efficacy in PRCC and primary and secondary treatment failure is common; other approaches are urgently needed to improve outcomes in these patients. METHODS: High throughput screening with small molecule libraries identified HSP90 inhibitors as agents of interest based on antitumor activity against patient derived PRCC cell lines. We investigated the activity of the orally available HSP90 inhibitor, SNX2112 in vitro, using 2D/3D PRCC cell culture models and in vivo, in mice tumor xenograft models. The molecular pathways mediating antitumor activity of SNX2112 were assessed by Western blot analysis, Flow cytometry, RNA-seq analysis, Real Time qPCR and imaging approaches. RESULTS: SNX2112 significantly inhibited cellular proliferation, induced G2/M cell cycle arrest and apoptosis in PRCC lines overexpressing MET. In contrast to TKIs targeting MET, SNX2112 inhibited both MET and known downstream mediators of MET activity (AKT, pAKT1/2 and pERK1/2) in PRCC cell lines. RNAi silencing of AKT1/2 or ERK1/2 expression significantly inhibited growth in PRCC cells. Furthermore, SNX2112 inhibited a unique set of E2F and MYC targets and G2M-associated genes. Interestingly, interrogation of the TCGA papillary RCC cohort revealed that these genes were overexpressed in PRCC and portend a poor prognosis. Finally, SNX-2112 demonstrated strong antitumor activity in vivo and prolonged survival of mice bearing human PRCC xenograft. CONCLUSIONS: These results demonstrate that HSP90 inhibition is associated with potent activity in PRCC, and implicate the PI3K/AKT and MEK/ERK1/2 pathways as important mediators of tumorigenesis. These data also provide the impetus for further clinical evaluation of HSP90, AKT, MEK or E2F pathway inhibitors in PRCC.


Asunto(s)
Antineoplásicos , Carcinoma de Células Renales , Neoplasias Renales , Animales , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Carcinoma de Células Renales/patología , Proteínas HSP90 de Choque Térmico/genética , Humanos , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , Neoplasias Renales/patología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt
3.
Blood ; 138(24): 2469-2484, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34525183

RESUMEN

Chimeric antigen receptor (CAR) T-cell toxicities resembling hemophagocytic lymphohistiocytosis (HLH) occur in a subset of patients with cytokine release syndrome (CRS). As a variant of conventional CRS, a comprehensive characterization of CAR T-cell-associated HLH (carHLH) and investigations into associated risk factors are lacking. In the context of 59 patients infused with CD22 CAR T cells where a substantial proportion developed carHLH, we comprehensively describe the manifestations and timing of carHLH as a CRS variant and explore factors associated with this clinical profile. Among 52 subjects with CRS, 21 (40.4%) developed carHLH. Clinical features of carHLH included hyperferritinemia, hypertriglyceridemia, hypofibrinogenemia, coagulopathy, hepatic transaminitis, hyperbilirubinemia, severe neutropenia, elevated lactate dehydrogenase, and occasionally hemophagocytosis. Development of carHLH was associated with preinfusion natural killer(NK) cell lymphopenia and higher bone marrow T-cell:NK cell ratio, which was further amplified with CAR T-cell expansion. Following CRS, more robust CAR T-cell and CD8 T-cell expansion in concert with pronounced NK cell lymphopenia amplified preinfusion differences in those with carHLH without evidence for defects in NK cell mediated cytotoxicity. CarHLH was further characterized by persistent elevation of HLH-associated inflammatory cytokines, which contrasted with declining levels in those without carHLH. In the setting of CAR T-cell mediated expansion, clinical manifestations and immunophenotypic profiling in those with carHLH overlap with features of secondary HLH, prompting consideration of an alternative framework for identification and management of this toxicity profile to optimize outcomes following CAR T-cell infusion.


Asunto(s)
Síndrome de Liberación de Citoquinas/etiología , Inmunoterapia Adoptiva/efectos adversos , Linfohistiocitosis Hemofagocítica/etiología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Adulto , Linfocitos T CD8-positivos/inmunología , Síndrome de Liberación de Citoquinas/diagnóstico , Síndrome de Liberación de Citoquinas/inmunología , Femenino , Humanos , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/inmunología , Masculino , Estudios Retrospectivos
4.
Sci Signal ; 14(664)2021 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-33402335

RESUMEN

Understanding the mechanisms of the Warburg shift to aerobic glycolysis is critical to defining the metabolic basis of cancer. Hereditary leiomyomatosis and renal cell carcinoma (HLRCC) is an aggressive cancer characterized by biallelic inactivation of the gene encoding the Krebs cycle enzyme fumarate hydratase, an early shift to aerobic glycolysis, and rapid metastasis. We observed impairment of the mitochondrial respiratory chain in tumors from patients with HLRCC. Biochemical and transcriptomic analyses revealed that respiratory chain dysfunction in the tumors was due to loss of expression of mitochondrial DNA (mtDNA)-encoded subunits of respiratory chain complexes, caused by a marked decrease in mtDNA content and increased mtDNA mutations. We demonstrated that accumulation of fumarate in HLRCC tumors inactivated the core factors responsible for replication and proofreading of mtDNA, leading to loss of respiratory chain components, thereby promoting the shift to aerobic glycolysis and disease progression in this prototypic model of glucose-dependent human cancer.


Asunto(s)
Carcinoma de Células Renales/genética , Ciclo del Ácido Cítrico , Daño del ADN , ADN Mitocondrial/metabolismo , Fumarato Hidratasa/genética , Neoplasias Renales/genética , Leiomiomatosis/enzimología , Síndromes Neoplásicos Hereditarios/enzimología , Neoplasias Cutáneas/enzimología , Neoplasias Uterinas/enzimología , Adulto , Anciano , Carcinoma de Células Renales/etiología , Carcinoma de Células Renales/metabolismo , Reparación del ADN , Replicación del ADN , Femenino , Fumarato Hidratasa/deficiencia , Perfilación de la Expresión Génica , Humanos , Neoplasias Renales/etiología , Neoplasias Renales/metabolismo , Leiomiomatosis/complicaciones , Masculino , Persona de Mediana Edad , Mitocondrias/genética , Mitocondrias/metabolismo , Mutación , Síndromes Neoplásicos Hereditarios/complicaciones , Neoplasias Cutáneas/complicaciones , Neoplasias Uterinas/complicaciones , Adulto Joven
5.
Oncotarget ; 9(26): 18454-18479, 2018 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-29719618

RESUMEN

We report a novel mechanism of action of ONC201 as a mitochondria-targeting drug in cancer cells. ONC201 was originally identified as a small molecule that induces transcription of TNF-related apoptosis-inducing ligand (TRAIL) and subsequently kills cancer cells by activating TRAIL death receptors. In this study, we examined ONC201 toxicity on multiple human breast and endometrial cancer cell lines. ONC201 attenuated cell viability in all cancer cell lines tested. Unexpectedly, ONC201 toxicity was not dependent on either TRAIL receptors nor caspases. Time-lapse live cell imaging revealed that ONC201 induces cell membrane ballooning followed by rupture, distinct from the morphology of cells undergoing apoptosis. Further investigation found that ONC201 induces phosphorylation of AMP-dependent kinase and ATP loss. Cytotoxicity and ATP depletion were significantly enhanced in the absence of glucose, suggesting that ONC201 targets mitochondrial respiration. Further analysis indicated that ONC201 indirectly inhibits mitochondrial respiration. Confocal and electron microscopic analysis demonstrated that ONC201 triggers mitochondrial structural damage and functional impairment. Moreover, ONC201 decreased mitochondrial DNA (mtDNA). RNAseq analysis revealed that ONC201 suppresses expression of multiple mtDNA-encoded genes and nuclear-encoded mitochondrial genes involved in oxidative phosphorylation and other mitochondrial functions. Importantly, fumarate hydratase deficient cancer cells and multiple cancer cell lines with reduced amounts of mtDNA were resistant to ONC201. These results indicate that cells not dependent on mitochondrial respiration are ONC201-resistant. Our data demonstrate that ONC201 kills cancer cells by disrupting mitochondrial function and further suggests that cancer cells that are dependent on glycolysis will be resistant to ONC201.

6.
Nat Commun ; 9(1): 1526, 2018 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-29670103

RESUMEN

ATM drives DNA repair by phosphorylating the histone variant H2AX. While ATM mutations elicit prominent neurobehavioral phenotypes, neural roles for H2AX have been elusive. We report impaired motor learning and balance in H2AX-deficient mice. Mitigation of reactive oxygen species (ROS) with N-acetylcysteine (NAC) reverses the behavioral deficits. Mouse embryonic fibroblasts deficient for H2AX exhibit increased ROS production and failure to activate the antioxidant response pathway controlled by the transcription factor NRF2. The NRF2 targets GCLC and NQO1 are depleted in the striatum of H2AX knockouts, one of the regions most vulnerable to ROS-mediated damage. These findings establish a role for ROS in the behavioral deficits of H2AX knockout mice and reveal a physiologic function of H2AX in mediating influences of oxidative stress on NRF2-transcriptional targets and behavior.


Asunto(s)
Conducta Animal , Histonas/deficiencia , Factor 2 Relacionado con NF-E2/metabolismo , Estrés Oxidativo , Acetilcisteína/química , Animales , Antioxidantes/química , Cuerpo Estriado/metabolismo , Daño del ADN , Fibroblastos/metabolismo , Células HEK293 , Heterocigoto , Histonas/fisiología , Humanos , Ratones , Ratones Noqueados , Microscopía Confocal , Modelos Neurológicos , Destreza Motora , Oxidación-Reducción , Fenotipo , Fosforilación , Especies Reactivas de Oxígeno/metabolismo
7.
Proc Natl Acad Sci U S A ; 115(8): E1867-E1875, 2018 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-29432154

RESUMEN

Moxetumomab pasudotox (Moxe) is a chimeric protein composed of an anti-CD22 Fv fused to a portion of Pseudomonas exotoxin A and kills CD22-expressing leukemia cells. It is very active in hairy-cell leukemia, but many children with relapsed/refractory acute lymphoblastic leukemia (ALL) either respond transiently or are initially resistant. Resistance to Moxe in cultured cells is due to low expression of diphthamide genes (DPH), but only two of six ALL blast samples from resistant patients had low DPH expression. To develop a more clinically relevant model of resistance, we treated NSG mice bearing KOPN-8 or Reh cells with Moxe. More than 99.9% of the cancer cells were killed by Moxe, but relapse occurred from discrete bone marrow sites. The resistant cells would no longer grow in cell culture and showed major chromosomal changes and changes in phenotype with greatly decreased CD22. RNA deep sequencing of resistant KOPN-8 blasts revealed global changes in gene expression, indicating dedifferentiation toward less-mature B cell precursors, and showed an up-regulation of myeloid genes. When Moxe was combined with 5-azacytidine, resistance was prevented and survival increased to over 5 months in the KOPN-8 model and greatly improved in the Reh model. We conclude that Moxe resistance in mice is due to a new mechanism that could not be observed using cultured cells and is prevented by treatment with 5-azacytidine.


Asunto(s)
Azacitidina/uso terapéutico , Toxinas Bacterianas/uso terapéutico , Exotoxinas/uso terapéutico , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/uso terapéutico , Azacitidina/administración & dosificación , Toxinas Bacterianas/administración & dosificación , Médula Ósea , Línea Celular Tumoral , Regulación hacia Abajo , Resistencia a Antineoplásicos , Exotoxinas/administración & dosificación , Humanos , Leucemia , Ratones , Neoplasias Experimentales , Leucemia-Linfoma Linfoblástico de Células Precursoras , Recurrencia
8.
Nucleus ; 8(5): 573-582, 2017 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-28857661

RESUMEN

The nuclear lamina is a proteinaceous meshwork situated underneath the inner nuclear membrane and is composed of nuclear lamin proteins, which are type-V intermediate filaments. The LMNA gene gives rise to lamin A and lamin C through alternative splicing. Mutations in LMNA cause multiple diseases known as laminopathies, including Hutchinson-Gilford Progeria Syndrome (HGPS), a premature aging disorder caused by a point mutation that activates a cryptic 5' splice site in exon 11, resulting in a 150 bp deletion in the LMNA mRNA and the production of the dominant lamin A isoform progerin. During RNA sequencing analysis of wild type and HGPS patient skin fibroblasts, we discovered two novel LMNA isoforms. LMNAΔ447 and LMNAΔ297 use an alternative 3' splice acceptor site in the 3' untranslated region, and either the HGPS cryptic 5' splice site in exon 11 or the wild type 5' splice site. Both isoforms are present at low levels in HGPS patient and wild type cells in multiple cell types. We validate and quantify the expression levels of these novel isoforms in HGPS and wild type fibroblasts. Overexpression of either LMNAΔ447 or LMNAΔ297 is not sufficient to induce the typical HGPS cellular disease phenotypes and no significant difference in the two isoforms were found between young and old fibroblasts. These results identify and characterize two novel RNA isoforms of LMNA produced through alternative splicing.


Asunto(s)
Lamina Tipo A/genética , Isoformas de ARN/genética , Empalme Alternativo , Secuencia de Bases , Humanos , Mutación , Fenotipo , Progeria/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
9.
Cell Cycle ; 15(18): 2398-404, 2016 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-27315462

RESUMEN

The epithelial-mesenchymal transition (EMT) is thought to be essential for cancer metastasis. While chromatin remodeling is involved in EMT, which processes contribute to this remodeling remain poorly investigated. Recently, we showed that silencing or removal of the histone variant H2A.X induced mesenchymal-like characteristics, including activation of the EMT transcription factors, Slug and Zeb1 in human colon cancer cells. Here, we provide the evidence that H2A.X loss in human non-tumorigenic breast cell line MCF10A results in a robust EMT activation, as substantiated by a genome-wide expression analysis. Cells deficient for H2A.X exhibit enhanced migration and invasion, along with an activation of a set of mesenchymal genes and a concomitant repression of epithelial genes. In the breast model, the EMT-related transcription factor Twist1 cooperates with Slug to regulate EMT upon H2A.X Loss. Of interest, H2A.X expression level tightly correlates with Twist1, and to a lesser extent with Slug in the panel of human breast cancer cell lines of the NCI-60 datasets. These new findings indicate that H2A.X is involved in the EMT processes in cells of different origins but pairing with transcription factors for EMT may be tissue specific.


Asunto(s)
Mama/patología , Transición Epitelial-Mesenquimal , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción de la Familia Snail/metabolismo , Proteína 1 Relacionada con Twist/metabolismo , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Femenino , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Modelos Biológicos
10.
Proc Natl Acad Sci U S A ; 113(13): 3545-50, 2016 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-26969725

RESUMEN

The ancient, highly conserved, Wnt signaling pathway regulates cell fate in all metazoans. We have previously shown that combined null mutations of the specificity protein (Sp) 1/Klf-like zinc-finger transcription factors Sp5 and Sp8 (i.e., Sp5/8) result in an embryonic phenotype identical to that observed when core components of the Wnt/ß-catenin pathway are mutated; however, their role in Wnt signal transduction is unknown. Here, we show in mouse embryos and differentiating embryonic stem cells that Sp5/8 are gene-specific transcriptional coactivators in the Wnt/ß-catenin pathway. Sp5/8 bind directly to GC boxes in Wnt target gene enhancers and to adjacent, or distally positioned, chromatin-bound T-cell factor (Tcf) 1/lymphoid enhancer factor (Lef) 1 to facilitate recruitment of ß-catenin to target gene enhancers. Because Sp5 is itself directly activated by Wnt signals, we propose that Sp5 is a Wnt/ß-catenin pathway-specific transcript on factor that functions in a feed-forward loop to robustly activate select Wnt target genes.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor de Unión 1 al Potenciador Linfoide/metabolismo , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/genética , beta Catenina/metabolismo , Animales , Proteínas de Unión al ADN/genética , Desarrollo Embrionario/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Elementos de Facilitación Genéticos , Femenino , Factor Nuclear 1-alfa del Hepatocito/genética , Factor de Unión 1 al Potenciador Linfoide/genética , Ratones , Ratones Transgénicos , Embarazo , Factores de Transcripción/genética , Activación Transcripcional , beta Catenina/genética
11.
Clin Cancer Res ; 22(9): 2237-49, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26712692

RESUMEN

PURPOSE: Uveal melanoma is a rare melanoma variant with no effective therapies once metastases develop. Although durable cancer regression can be achieved in metastatic cutaneous melanoma with immunotherapies that augment naturally existing antitumor T-cell responses, the role of these treatments for metastatic uveal melanoma remains unclear. We sought to define the relative immunogenicity of these two melanoma variants and determine whether endogenous antitumor immune responses exist against uveal melanoma. EXPERIMENTAL DESIGN: We surgically procured liver metastases from uveal melanoma (n = 16) and cutaneous melanoma (n = 35) patients and compared the attributes of their respective tumor cell populations and their infiltrating T cells (TIL) using clinical radiology, histopathology, immune assays, and whole-exomic sequencing. RESULTS: Despite having common melanocytic lineage, uveal melanoma and cutaneous melanoma metastases differed in their melanin content, tumor differentiation antigen expression, and somatic mutational profile. Immunologic analysis of TIL cultures expanded from these divergent forms of melanoma revealed cutaneous melanoma TIL were predominantly composed of CD8(+) T cells, whereas uveal melanoma TIL were CD4(+) dominant. Reactivity against autologous tumor was significantly greater in cutaneous melanoma TIL compared with uveal melanoma TIL. However, we identified TIL from a subset of uveal melanoma patients which had robust antitumor reactivity comparable in magnitude with cutaneous melanoma TIL. Interestingly, the absence of melanin pigmentation in the parental tumor strongly correlated with the generation of highly reactive uveal melanoma TIL. CONCLUSIONS: The discovery of this immunogenic group of uveal melanoma metastases should prompt clinical efforts to determine whether patients who harbor these unique tumors can benefit from immunotherapies that exploit endogenous antitumor T-cell populations. Clin Cancer Res; 22(9); 2237-49. ©2015 AACR.


Asunto(s)
Activación de Linfocitos/inmunología , Melanoma/inmunología , Neoplasias de la Úvea/inmunología , Adolescente , Adulto , Anciano , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/inmunología , Citotoxicidad Inmunológica/inmunología , Femenino , Humanos , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Neoplasias Cutáneas/inmunología , Linfocitos T Citotóxicos/inmunología , Células Tumorales Cultivadas , Adulto Joven , Melanoma Cutáneo Maligno
12.
Mol Cell Biol ; 36(5): 693-713, 2015 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-26667036

RESUMEN

The integrated stress response (ISR) controls cellular adaptations to nutrient deprivation, redox imbalances, and endoplasmic reticulum (ER) stress. ISR genes are upregulated in stressed cells, primarily by the bZIP transcription factor ATF4 through its recruitment to cis-regulatory C/EBP:ATF response elements (CAREs) together with a dimeric partner of uncertain identity. Here, we show that C/EBPγ:ATF4 heterodimers, but not C/EBPß:ATF4 dimers, are the predominant CARE-binding species in stressed cells. C/EBPγ and ATF4 associate with genomic CAREs in a mutually dependent manner and coregulate many ISR genes. In contrast, the C/EBP family members C/EBPß and C/EBP homologous protein (CHOP) were largely dispensable for induction of stress genes. Cebpg(-/-) mouse embryonic fibroblasts (MEFs) proliferate poorly and exhibit oxidative stress due to reduced glutathione levels and impaired expression of several glutathione biosynthesis pathway genes. Cebpg(-/-) mice (C57BL/6 background) display reduced body size and microphthalmia, similar to ATF4-null animals. In addition, C/EBPγ-deficient newborns die from atelectasis and respiratory failure, which can be mitigated by in utero exposure to the antioxidant, N-acetyl-cysteine. Cebpg(-/-) mice on a mixed strain background showed improved viability but, upon aging, developed significantly fewer malignant solid tumors than WT animals. Our findings identify C/EBPγ as a novel antioxidant regulator and an obligatory ATF4 partner that controls redox homeostasis in normal and cancerous cells.


Asunto(s)
Factor de Transcripción Activador 4/metabolismo , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Estrés Oxidativo , Factor de Transcripción Activador 4/análisis , Factor de Transcripción Activador 4/genética , Animales , Proteínas Potenciadoras de Unión a CCAAT/análisis , Proteínas Potenciadoras de Unión a CCAAT/genética , Línea Celular , Femenino , Feto/anomalías , Feto/metabolismo , Eliminación de Gen , Regulación de la Expresión Génica , Glutatión/metabolismo , Humanos , Masculino , Ratones Endogámicos C57BL , Neoplasias/genética , Neoplasias/metabolismo , Multimerización de Proteína , Elementos de Respuesta , Factor de Transcripción CHOP/metabolismo
13.
Development ; 141(22): 4285-97, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25371364

RESUMEN

Neuromesodermal (NM) stem cells generate neural and paraxial presomitic mesoderm (PSM) cells, which are the respective progenitors of the spinal cord and musculoskeleton of the trunk and tail. The Wnt-regulated basic helix-loop-helix (bHLH) transcription factor mesogenin 1 (Msgn1) has been implicated as a cooperative regulator working in concert with T-box genes to control PSM formation in zebrafish, although the mechanism is unknown. We show here that, in mice, Msgn1 alone controls PSM differentiation by directly activating the transcriptional programs that define PSM identity, epithelial-mesenchymal transition, motility and segmentation. Forced expression of Msgn1 in NM stem cells in vivo reduced the contribution of their progeny to the neural tube, and dramatically expanded the unsegmented mesenchymal PSM while blocking somitogenesis and notochord differentiation. Expression of Msgn1 was sufficient to partially rescue PSM differentiation in Wnt3a(-/-) embryos, demonstrating that Msgn1 functions downstream of Wnt3a as the master regulator of PSM differentiation. Our data provide new insights into how cell fate decisions are imposed by the expression of a single transcriptional regulator.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Mesodermo/embriología , Músculo Esquelético/embriología , Sistema Nervioso/embriología , Animales , Inmunoprecipitación de Cromatina , Ensayo de Cambio de Movilidad Electroforética , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Inmunohistoquímica , Hibridación in Situ , Luciferasas , Mesodermo/citología , Ratones , Ratones Noqueados , Análisis por Micromatrices , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Wnt3A/genética
14.
Cell Rep ; 8(4): 1049-62, 2014 Aug 21.
Artículo en Inglés | MEDLINE | ID: mdl-25131201

RESUMEN

Appropriate DNA double-strand break (DSB) repair factor choice is essential for ensuring accurate repair outcome and genomic integrity. The factors that regulate this process remain poorly understood. Here, we identify two repressive chromatin components, the macrohistone variant macroH2A1 and the H3K9 methyltransferase and tumor suppressor PRDM2, which together direct the choice between the antagonistic DSB repair mediators BRCA1 and 53BP1. The macroH2A1/PRDM2 module mediates an unexpected shift from accessible to condensed chromatin that requires the ataxia telangiectasia mutated (ATM)-dependent accumulation of both proteins at DSBs in order to promote DSB-flanking H3K9 dimethylation. Remarkably, loss of macroH2A1 or PRDM2, as well as experimentally induced chromatin decondensation, impairs the retention of BRCA1, but not 53BP1, at DSBs. As a result, macroH2A1 and/or PRDM2 depletion causes epistatic defects in DSB end resection, homology-directed repair, and the resistance to poly(ADP-ribose) polymerase (PARP) inhibition-all hallmarks of BRCA1-deficient tumors. Together, these findings identify dynamic, DSB-associated chromatin reorganization as a critical modulator of BRCA1-dependent genome maintenance.


Asunto(s)
Proteína BRCA1/fisiología , Histonas/fisiología , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular Tumoral , Ensamble y Desensamble de Cromatina , Roturas del ADN de Doble Cadena , Proteínas de Unión al ADN/metabolismo , Inestabilidad Genómica , Células HEK293 , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Metilación , Proteínas Nucleares/metabolismo , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Reparación del ADN por Recombinación , Factores de Transcripción/metabolismo
15.
PLoS One ; 9(5): e96650, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24827921

RESUMEN

The pmel-1 T cell receptor transgenic mouse has been extensively employed as an ideal model system to study the mechanisms of tumor immunology, CD8+ T cell differentiation, autoimmunity and adoptive immunotherapy. The 'zygosity' of the transgene affects the transgene expression levels and may compromise optimal breeding scheme design. However, the integration sites for the pmel-1 mouse have remained uncharacterized. This is also true for many other commonly used transgenic mice created before the modern era of rapid and inexpensive next-generation sequencing. Here, we show that whole genome sequencing can be used to determine the exact pmel-1 genomic integration site, even with relatively 'shallow' (8X) coverage. The results were used to develop a validated polymerase chain reaction-based genotyping assay. For the first time, we provide a quick and convenient polymerase chain reaction method to determine the dosage of pmel-1 transgene for this freely and publically available mouse resource. We also demonstrate that next-generation sequencing provides a feasible approach for mapping foreign DNA integration sites, even when information of the original vector sequences is only partially known.


Asunto(s)
Cromosomas Humanos Par 2/química , Mutagénesis Insercional , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Transgenes , Antígeno gp100 del Melanoma/genética , Animales , Secuencia de Bases , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Mapeo Cromosómico , Dosificación de Gen , Expresión Génica , Vectores Genéticos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Ratones , Ratones Transgénicos , Datos de Secuencia Molecular , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Antígeno gp100 del Melanoma/inmunología
16.
PLoS One ; 7(4): e36040, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22563437

RESUMEN

We have previously reported increased apoptosis of regulatory T cells (Tregs) in recent-onset Type 1 Diabetes subjects (RO T1D) in the honeymoon phase and in multiple autoantibody-positive (Ab+) subjects, some of which are developing T1D. We have also reported that increased Treg apoptosis was associated with High HLA risk and that it subsided with cessation of honeymoon period. In this report, we present results generated using genetics, genomics, functional cell-based assays and flow cytometry to assess cellular changes at the T-cell level during T1D pathogenesis. We measured ex vivo Treg apoptosis and Treg function, surface markers expression, expression of HLA class II genes, the influence of HLA risk on Treg apoptosis and function, and evaluated contribution of genes reported to be involved in the apoptosis process. This integrated comprehensive approach uncovered important information that can serve as a basis for future studies aimed to modulate Treg cell responsiveness to apoptotic signals in autoimmunity. For example, T1D will progress in those subjects where increased Treg apoptosis is accompanied with decreased Treg function. Furthermore, Tregs from High HLA risk healthy controls had increased Treg apoptosis levels and overexpressed FADD but not Fas/FasL. Tregs from RO T1D subjects in the honeymoon phase were primarily dying through withdrawal of growth hormones with contribution of oxidative stress, mitochondrial apoptotic pathways, and employment of TNF-receptor family members. Ab+ subjects, however, expressed high inflammation level, which probably contributed to Treg apoptosis, although other apoptotic pathways were also activated: withdrawal of growth hormones, oxidative stress, mitochondrial apoptosis and Fas/FasL apoptotic pathways. The value of these results lie in potentially different preventive treatment subjects would receive depending on disease progression stage when treated.


Asunto(s)
Apoptosis/genética , Diabetes Mellitus Tipo 1/metabolismo , Antígenos de Histocompatibilidad Clase II/metabolismo , Linfocitos T Reguladores/metabolismo , Apoptosis/inmunología , Autoanticuerpos/metabolismo , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/patología , Proteína Ligando Fas/metabolismo , Proteína de Dominio de Muerte Asociada a Fas/metabolismo , Genotipo , Hormona del Crecimiento/metabolismo , Cadenas alfa de HLA-DQ/genética , Cadenas alfa de HLA-DQ/inmunología , Cadenas alfa de HLA-DQ/metabolismo , Cadenas beta de HLA-DQ/genética , Cadenas beta de HLA-DQ/inmunología , Cadenas beta de HLA-DQ/metabolismo , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Mitocondrias/metabolismo , Estrés Oxidativo , Receptores del Factor de Necrosis Tumoral/metabolismo , Riesgo , Linfocitos T Reguladores/inmunología , Receptor fas/metabolismo
17.
Physiol Genomics ; 43(11): 697-709, 2011 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-21406607

RESUMEN

Inflammation is common to many disorders and responsible for tissue and organ damage. In many disorders, the associated peripheral cytokine milieu is dilute and difficult to measure, necessitating development of more sensitive and informative biomarkers for mechanistic studies, earlier diagnosis, and monitoring therapeutic interventions. Previously, we have shown that plasma of recent-onset (RO) Type 1 diabetes patients induces a disease-specific proinflammatory transcriptional profile in fresh peripheral blood mononuclear cells (PBMC) compared with that of healthy controls (HC). To eliminate assay variance introduced through the use of multiple donors or multiple draws of the same person over time, we evaluated human leukemia cell lines as potential surrogates for fresh PBMC. We 1) tested seven different cell lines in their power to differentiate RO from HC plasma and 2) compared the similarity of the signatures generated across the seven cell lines to that obtained with fresh PBMC. While each cell line tested exhibited a distinct transcriptional response when cultured with RO or HC plasma, the expression profile induced in any single cell line shared little identity with that of the other cell lines or fresh PBMC. In terms of regulated biological pathways, the transcriptional response of each cell line shared varying degrees of functional identity with fresh PBMC. These results indicate that use of human leukemia cell lines as surrogates for fresh PBMC has potential in detecting perturbations to the peripheral cytokine milieu. However, the response of each is distinct, possessing varying degrees of functional relatedness to that observed with PBMC.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Perfilación de la Expresión Génica , Mediadores de Inflamación/metabolismo , Biomarcadores/metabolismo , Línea Celular Tumoral , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inmunología , Humanos , Leucemia , Leucocitos Mononucleares/metabolismo , Linfoma
18.
PLoS One ; 5(12): e15154, 2010 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-21151941

RESUMEN

BACKGROUND: In type 1 diabetes (T1D), a prototypic autoimmune disease, effector T cells destroy beta cells. Normally, CD4(+)CD25(+high), or natural regulatory T cells (Tregs), counter this assault. In autoimmunity, the failure to suppress CD4(+)CD25(low) T cells is important for disease development. However, both Treg dysfunction and hyperactive responder T-cell proliferation contribute to disease. METHODS/PRINCIPAL FINDINGS: We investigated human CD4(+)CD25(low) T cells and compared them to CD4(+)CD25(-) T cells in otherwise equivalent in vitro proliferative conditions. We then asked whether these differences in suppression are exacerbated in T1D. In both single and co-culture with Tregs, the CD4(+)CD25(low) T cells divided more rapidly than CD4(+)CD25(-) T cells, which manifests as increased proliferation/reduced suppression. Time-course experiments showed that this difference could be explained by higher IL-2 production from CD4+CD25(low) compared to CD4+CD25- T cells. There was also a significant increase in CD4+CD25(low) T-cell proliferation compared to CD4+CD25- T cells during suppression assays from RO T1D and at-risk subjects (n = 28, p = 0.015 and p = 0.024 respectively). CONCLUSIONS/SIGNIFICANCE: The in vitro dual suppression assays proposed here could highlight the impaired sensitivity of certain responder T cells to the suppressive effect of Tregs in human autoimmune diseases.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Linfocitos T/citología , Enfermedades Autoinmunes/inmunología , Linfocitos T CD4-Positivos/citología , Separación Celular , Técnicas de Cocultivo , Citometría de Flujo , Humanos , Células Secretoras de Insulina/inmunología , Interleucina-2/metabolismo , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Cinética , Leucocitos Mononucleares/citología , Riesgo , Linfocitos T Reguladores/citología
19.
Cell Tissue Res ; 339(3): 585-95, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20143240

RESUMEN

CD4+CD25+(high) regulatory T cells (Tregs) play a pivotal role in the control of the immune response. A growing body of evidence suggests the reduced function of these cells in autoimmune diseases, including type 1 diabetes (T1D). Restoration of their function can potentially delay further disease development. In the present study, we have converted conventional effector T cells into induced Tregs (iTregs) in recent-onset (RO) T1D (n=9) and compared them with the same cells generated in controls (n=12) and in long-standing (LS) T1D subjects (n=9). The functional potential of in-vitro-generated Tregs was measured by using an in vitro proliferation assay. We noted that the suppressive potential of iTregs exceeded that of natural regulatory T cells (nTregs) only in the RO T1D subjects. We showed that iTregs from RO T1D subjects had increased expression of Foxp3, E3 ubiquitin ligase (ITCH) and TGF-beta-inducible early gene 1 (TIEG1) compared with control and LS T1D subjects. We also expanded natural, thymically derived Tregs (nTregs) and compared the functional ability of these cells between subject groups. Expanded cells from all three subject groups were suppressive. RO T1D subjects were the only group in which both iTregs and expanded Tregs were functional, suggesting that the inflammatory milieu impacts in vitro Treg generation. Future longitudinal studies should delineate the actual contribution of the stage of disease to the quality of in-vitro-generated Tregs.


Asunto(s)
Diabetes Mellitus Tipo 1/enzimología , Diabetes Mellitus Tipo 1/inmunología , Tolerancia Inmunológica/inmunología , Proteínas Represoras/metabolismo , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Ubiquitina-Proteína Ligasas/metabolismo , Adulto , Edad de Inicio , Apoptosis , Estudios de Casos y Controles , Recuento de Células , Niño , Diabetes Mellitus Tipo 1/epidemiología , Factores de Transcripción de la Respuesta de Crecimiento Precoz/metabolismo , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Masculino
20.
Eur J Immunol ; 39(9): 2571-83, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19701891

RESUMEN

The transcription factor Foxp3 is essential for the development of functional, natural Treg (nTreg), which plays a prominent role in self-tolerance. Suppressive Foxp3(+) Treg cells can be generated from naïve T cells ex vivo, following TCR and TGF-beta1 stimulations. However, the molecular contributions from the different arms of these pathways leading to Foxp3 expression are not fully understood. TGF-beta1-activated Smad3 plays a major role in the expression of Foxp3, since TGF-beta1-induced-Treg generation from Smad3(-/-) mice is markedly reduced and abolished by inactivating Smad2. In the TCR pathway, deletion of Bcl10, which activates NF-kappaB, markedly reduces both IL-2 and Foxp3 production. However, partial rescue of Foxp3 expression occurs on addition of exogenous IL-2. TGF-beta1 significantly attenuates NF-kappaB binding to the Foxp3 promoter, while inducing Foxp3 expression. Furthermore, deletion of p50, a NF-kappaB subunit, results in increased Foxp3 expression despite a decline in the IL-2 production. We posit several TCR-NF-kappaB pathways, some increasing (Bcl10-IL-2-Foxp3) while others decreasing (p50-Foxp3) Foxp3 expression, with the former predominating. A better understanding of Foxp3 regulation could be useful in dissecting the cause of Treg dysfunction in several autoimmune diseases and for generating more potent TGF-beta1-induced-Treg cells for therapeutic purposes.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Factores de Transcripción Forkhead/biosíntesis , Subunidad p50 de NF-kappa B/metabolismo , Proteína smad3/metabolismo , Linfocitos T Reguladores/inmunología , Animales , Antineoplásicos/farmacología , Linfocitos T CD4-Positivos/efectos de los fármacos , Células Cultivadas , Factores de Transcripción Forkhead/genética , Interleucina-2/farmacología , Ratones , Ratones Endogámicos C57BL , Subunidad p50 de NF-kappa B/antagonistas & inhibidores , Péptidos/farmacología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Proteína smad3/genética , Linfocitos T Reguladores/efectos de los fármacos , Factor de Crecimiento Transformador beta1/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA