Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Adv Exp Med Biol ; 1433: 51-68, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37751135

RESUMEN

The histone lysine demethylase 2 (KDM2) family of α-Ketoglutarate-Fe++-dependent dioxygenases were the first Jumonji-domain-containing proteins reported to harbor demethylase activity. This landmark discovery paved the way for the characterization of more than 25 enzymes capable of demethylating lysine residues on histones-an epigenetic modification previously thought to be irreversible. The KDM2 family is comprised of KDM2A and KDM2B which share significant structural similarities and demethylate lysine 36 on histone H3. However, they exert distinct cellular functions and are frequently deregulated in a broad spectrum of human cancers. With the advent of next generation sequencing and development of genetically engineered mouse models, it was shown that KDM2A and KDM2B play critical roles in stem cell biology, somatic cell reprograming, and organismal development by regulating cell fate and lineage commitment decisions. Thus, understanding the biochemistry and elucidating the context-dependent function of these enzymes is an emerging new frontier for the development of small molecule inhibitors to treat cancer and other diseases.


Asunto(s)
Proteínas F-Box , Histona Demetilasas , Humanos , Animales , Ratones , Lisina , Diferenciación Celular , Epigénesis Genética , Secuenciación de Nucleótidos de Alto Rendimiento , Histonas , Histona Demetilasas con Dominio de Jumonji/genética
2.
Hepatology ; 65(2): 678-693, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28114741

RESUMEN

Exposure to genotoxins such as ethanol-derived acetaldehyde leads to DNA damage and liver injury and promotes the development of cancer. We report here a major role for the transforming growth factor ß/mothers against decapentaplegic homolog 3 adaptor ß2-Spectrin (ß2SP, gene Sptbn1) in maintaining genomic stability following alcohol-induced DNA damage. ß2SP supports DNA repair through ß2SP-dependent activation of Fanconi anemia complementation group D2 (Fancd2), a core component of the Fanconi anemia complex. Loss of ß2SP leads to decreased Fancd2 levels and sensitizes ß2SP mutants to DNA damage by ethanol treatment, leading to phenotypes that closely resemble those observed in animals lacking both aldehyde dehydrogenase 2 and Fancd2 and resemble human fetal alcohol syndrome. Sptbn1-deficient cells are hypersensitive to DNA crosslinking agents and have defective DNA double-strand break repair that is rescued by ectopic Fancd2 expression. Moreover, Fancd2 transcription in response to DNA damage/transforming growth factor ß stimulation is regulated by the ß2SP/mothers against decapentaplegic homolog 3 complex. CONCLUSION: Dysfunctional transforming growth factor ß/ß2SP signaling impacts the processing of genotoxic metabolites by altering the Fanconi anemia DNA repair pathway. (Hepatology 2017;65:678-693).


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Inestabilidad Genómica/genética , Preñez , Espectrina/genética , Factor de Crecimiento Transformador beta2/genética , Análisis de Varianza , Animales , Animales Recién Nacidos , Daño del ADN/genética , Reparación del ADN/genética , Etanol/farmacología , Femenino , Trastornos del Espectro Alcohólico Fetal/genética , Trastornos del Espectro Alcohólico Fetal/patología , Humanos , Inmunohistoquímica , Peroxidación de Lípido/genética , Ratones , Ratones Transgénicos , Embarazo , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Transducción de Señal
3.
Nano Res ; 15(3): 2300-2314, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36089987

RESUMEN

Despite the promise of immunotherapy such as the immune checkpoint inhibitors (ICIs) anti-PD-1 and anti-CTLA-4 for advanced melanoma, only 26%-52% of patients respond, and many experience grade III/IV immune-related adverse events. Motivated by the need for an effective therapy for patients non-responsive to clinically approved ICIs, we have developed a novel nanoimmunotherapy that combines locally administered Prussian blue nanoparticle-based photothermal therapy (PBNP-PTT) with systemically administered agonistic anti-CD137 monoclonal antibody therapy (aCD137). PBNP-PTT was administered at various thermal doses to melanoma cells in vitro, and was combined with aCD137 in vivo to test treatment effects on melanoma tumor progression, animal survival, immunological protection against tumor rechallenge, and hepatotoxicity. When administered at a melanoma-specific thermal dose, PBNP-PTT elicits immunogenic cell death (ICD) in melanoma cells and upregulates markers associated with antigen presentation and immune cell co-stimulation in vitro. Consequently, PBNP-PTT eliminates primary melanoma tumors in vivo, yielding long-term tumor-free survival. However, the antitumor immune effects generated by PBNP-PTT cannot eliminate secondary tumors, despite significantly slowing their growth. The addition of aCD137 enables significant abscopal efficacy and improvement of survival, functioning through activated dendritic cells and tumor-infiltrating CD8+ T cells, and generates CD4+ and CD8+ T cell memory that manifests in the rejection of tumor rechallenge, with no long-term hepatotoxicity. This study describes for the first time a novel and effective nanoimmunotherapy combination of PBNP-PTT with aCD137 mAb therapy for melanoma.

4.
Adv Nanobiomed Res ; 1(8)2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34435194

RESUMEN

A combination therapy using Prussian blue nanoparticles (PBNP) as photothermal therapy (PTT) agents coated with CpG oligodeoxynucleotides, an immunologic adjuvant, as a nanoimmunotherapy (CpG-PBNP-PTT) for neuroblastoma (NB) is described. NB driven by MYCN amplification confers high risk and correlates with a dismal prognosis, accounting for the majority of NB-related mortality. The efficacy of the CpG-PBNP-PTT nanoimmunotherapy in a clinically relevant, TH-MYCN murine NB model (9464D) overexpressing MYCN is tested. When administered to 9464D NB cells in vitro, CpG-PBNP-PTT triggers thermal dose-dependent immunogenic cell death and tumor cell priming for immune recognition in vitro, measured by the expression of specific costimulatory and antigen-presenting molecules. In vivo, intratumorally administered CpG-PBNP-PTT generates complete tumor regression and significantly higher long-term survival compared to controls. Furthermore, CpG-PBNP-PTT-treated mice reject tumor rechallenge. Ex vivo studies confirm these therapeutic responses result from the generation of robust T cell-mediated immunological memory. Consequently, in a synchronous 9464D tumor model, CpG-PBNP-PTT induces complete tumor regression on the treated flank and significantly slows tumor progression on the untreated flank, improving animal survival. These findings demonstrate that localized administration of the CpG-PBNP-PTT nanoimmunotherapy drives potent systemic T cell responses in solid tumors such as NB and therefore has therapeutic implications for NB.

5.
Cell Rep ; 37(8): 110036, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34818550

RESUMEN

Balance between the hematopoietic stem cell (HSC) duality to either possess self-renewal capacity or differentiate into multipotency progenitors (MPPs) is crucial for maintaining homeostasis of the hematopoietic stem/progenitor cell (HSPC) compartment. To retain the HSC self-renewal activity, KIT, a receptor tyrosine kinase, in HSCs is activated by its cognate ligand KITLG originating from niche cells. Here, we show that AT-rich interaction domain 4B (ARID4B) interferes with KITLG/KIT signaling, consequently allowing HSC differentiation. Conditional Arid4b knockout in mouse hematopoietic cells blocks fetal HSC differentiation, preventing hematopoiesis. Mechanistically, ARID4B-deficient HSCs self-express KITLG and overexpress KIT. As to downstream pathways of KITLG/KIT signaling, inhibition of Src family kinases rescues the HSC differentiation defect elicited by ARID4B loss. In summary, the intrinsic ARID4B-KITLG/KIT-Src axis is an HSPC regulatory program that enables the differentiation state, while KIT stimulation by KITLG from niche cells preserves the HSPC undifferentiated pool.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Comunicación Autocrina , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Proliferación Celular/fisiología , Autorrenovación de las Células/fisiología , Proteínas de Unión al ADN/fisiología , Femenino , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas c-kit/genética , Transducción de Señal/fisiología , Factor de Células Madre/metabolismo , Nicho de Células Madre/fisiología , Factores de Transcripción/metabolismo , Familia-src Quinasas/metabolismo
6.
Nat Commun ; 11(1): 3018, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32541668

RESUMEN

Chronic pancreatitis represents a risk factor for the development of pancreatic cancer. We find that heterozygous loss of histone H2A lysine 119 deubiquitinase BAP1 (BRCA1 Associated Protein-1) associates with a history of chronic pancreatitis and occurs in 25% of pancreatic ductal adenocarcinomas and 40% of acinar cell carcinomas. Deletion or heterozygous loss of Bap1 in murine pancreata causes genomic instability, tissue damage, and pancreatitis with full penetrance. Concomitant expression of KrasG12D leads to predominantly intraductal papillary mucinous neoplasms and mucinous cystic neoplasms, while pancreatic intraepithelial neoplasias are rarely detected. These lesions progress to metastatic pancreatic cancer with high frequency. Lesions with histological features mimicking Acinar Cell Carcinomas are also observed in some tumors. Heterozygous mice also develop pancreatic cancer suggesting a haploinsufficient tumor suppressor role for BAP1. Mechanistically, BAP1 regulates genomic stability, in a catalytic independent manner, and its loss confers sensitivity to irradiation and platinum-based chemotherapy in pancreatic cancer.


Asunto(s)
Neoplasias Pancreáticas/metabolismo , Pancreatitis Crónica/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Estudios de Cohortes , Regulación Neoplásica de la Expresión Génica , Haploinsuficiencia , Humanos , Ratones , Neoplasias Pancreáticas/genética , Pancreatitis Crónica/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética
7.
Nat Commun ; 9(1): 4221, 2018 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-30310060

RESUMEN

The CCCTC-binding zinc-finger protein (CTCF)-mediated network of long-range chromatin interactions is important for genome organization and function. Although this network has been considered largely invariant, we find that it exhibits extensive cell-type-specific interactions that contribute to cell identity. Here, we present Lollipop, a machine-learning framework, which predicts CTCF-mediated long-range interactions using genomic and epigenomic features. Using ChIA-PET data as benchmark, we demonstrate that Lollipop accurately predicts CTCF-mediated chromatin interactions both within and across cell types, and outperforms other methods based only on CTCF motif orientation. Predictions are confirmed computationally and experimentally by Chromatin Conformation Capture (3C). Moreover, our approach identifies other determinants of CTCF-mediated chromatin wiring, such as gene expression within the loops. Our study contributes to a better understanding about the underlying principles of CTCF-mediated chromatin interactions and their impact on gene expression.


Asunto(s)
Factor de Unión a CCCTC/metabolismo , Cromatina/metabolismo , Epigénesis Genética , Genoma , Línea Celular , Redes Reguladoras de Genes , Humanos , Curva ROC
8.
Cancer Cell ; 33(3): 512-526.e8, 2018 03 12.
Artículo en Inglés | MEDLINE | ID: mdl-29533787

RESUMEN

KDM6A, an X chromosome-encoded histone demethylase and member of the COMPASS-like complex, is frequently mutated in a broad spectrum of malignancies and contributes to oncogenesis with poorly characterized mechanisms. We found that KDM6A loss induced squamous-like, metastatic pancreatic cancer selectively in females through deregulation of the COMPASS-like complex and aberrant activation of super-enhancers regulating ΔNp63, MYC, and RUNX3 oncogenes. This subtype of tumor developed in males had concomitant loss of UTY and KDM6A, suggesting overlapping roles, and points to largely demethylase independent tumor suppressor functions. We also demonstrate that KDM6A-deficient pancreatic cancer is selectively sensitive to BET inhibitors, which reversed squamous differentiation and restrained tumor growth in vivo, highlighting a therapeutic niche for patient tailored therapies.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Histona Demetilasas/deficiencia , Mutación/genética , Proteínas Nucleares/deficiencia , Neoplasias Pancreáticas/genética , Animales , Carcinoma de Células Escamosas , Línea Celular Tumoral , Proliferación Celular/genética , Histona Demetilasas/genética , Humanos , Ratones , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
9.
Exp Hematol ; 44(9): 778-782, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27208808

RESUMEN

The epigenetic control of gene expression is central to the development of the hematopoietic system and the execution of lineage-specific transcriptional programs. During the last 10 years, mounting evidence has implicated the family of lysine-specific histone demethylases as critical regulators of normal hematopoiesis, whereas their deregulation is found in a broad spectrum of hematopoietic malignancies. Here, we review recent findings on the role of these enzymes in normal and malignant hematopoiesis and highlight how aberrant epigenetic regulation facilitates hematopoietic cell transformation through subversion of cell fate and lineage commitment programs.


Asunto(s)
Neoplasias Hematológicas/genética , Neoplasias Hematológicas/metabolismo , Hematopoyesis/genética , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Regulación de la Expresión Génica , Histonas/metabolismo , Humanos , Metilación , Transducción de Señal
10.
J Clin Invest ; 126(3): 905-20, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26808549

RESUMEN

The development of the hematopoietic system is a dynamic process that is controlled by the interplay between transcriptional and epigenetic networks to determine cellular identity. These networks are critical for lineage specification and are frequently dysregulated in leukemias. Here, we identified histone demethylase KDM2B as a critical regulator of definitive hematopoiesis and lineage commitment of murine hematopoietic stem and progenitor cells (HSPCs). RNA sequencing of Kdm2b-null HSPCs and genome-wide ChIP studies in human leukemias revealed that KDM2B cooperates with polycomb and trithorax complexes to regulate differentiation, lineage choice, cytokine signaling, and cell cycle. Furthermore, we demonstrated that KDM2B exhibits a dichotomous role in hematopoietic malignancies. Specifically, we determined that KDM2B maintains lymphoid leukemias, but restrains RAS-driven myeloid transformation. Our study reveals that KDM2B is an important mediator of hematopoietic cell development and has opposing roles in tumor progression that are dependent on cellular context.


Asunto(s)
Proteínas F-Box/fisiología , Hematopoyesis , Histona Demetilasas con Dominio de Jumonji/fisiología , Linfoma/enzimología , Animales , Diferenciación Celular , Línea Celular Tumoral , Linaje de la Célula , Transformación Celular Neoplásica , Regulación Leucémica de la Expresión Génica , Humanos , Linfoma/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas p21(ras)/genética
11.
Sci Rep ; 6: 30217, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27456065

RESUMEN

Disruption of the TGF-ß pathway is associated with liver fibrosis and suppression of liver tumorigenesis, conditions associated with low Vitamin D (VD) levels. However, potential contributions of VD to liver tumor progression in the context of TGF-ß signaling remain unexplored. Our analyses of VD deprivation (VDD) in in vivo models of liver tumor formation revealed striking three-fold increases in tumor burden in Smad3(+/-) mice, with a three-fold increase in TLR7 expression compared to controls. ChIP and transcriptional assays confirm Smad3 binding at two TLR7 promoter SBE sites. Molecular interactions between TGF-ß pathway and VDD were validated clinically, where an absence of VD supplementation was associated with low TGF-ß pathway member expression levels and ß-catenin activation in fibrotic/cirrhotic human liver tissues. Subsequent supplementing VD led to restoration of TGF-ß member expression with lower ß-catenin levels. Bioinformatics analysis provides positive supportive correlation between somatic mutations for VD-related genes and the TGF-ß pathway. We conclude that VDD promotes tumor growth in the context of Smad3 disruption, potentially through regulation of TLR7 expression and ß-catenin activation. VD could therefore be a strong candidate for liver cancer prevention in the context of aberrant Smad3 signaling.


Asunto(s)
Neoplasias Hepáticas Experimentales/patología , Glicoproteínas de Membrana/metabolismo , Proteína smad3/genética , Receptor Toll-Like 7/metabolismo , Factor de Crecimiento Transformador beta/genética , Deficiencia de Vitamina D/complicaciones , Proteínas Wnt/metabolismo , Animales , Humanos , Neoplasias Hepáticas Experimentales/complicaciones , Masculino , Ratones , Ratones Transgénicos , Transducción de Señal , Vitamina D/administración & dosificación
12.
Cell Rep ; 8(1): 163-76, 2014 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-24953653

RESUMEN

Timely and selective recruitment of transcription factors to their appropriate DNA-binding sites represents a critical step in regulating gene activation; however, the regulatory strategies underlying each factor's effective recruitment to specific promoter and/or enhancer regions are not fully understood. Here, we identify an unexpected regulatory mechanism by which promoter-specific binding, and therefore function, of peroxisome proliferator-activator receptor γ (PPARγ) in adipocytes requires G protein suppressor 2 (GPS2) to prime the local chromatin environment via inhibition of the ubiquitin ligase RNF8 and stabilization of the H3K9 histone demethylase KDM4A/JMJD2. Integration of genome-wide profiling data indicates that the pioneering activity of GPS2/KDM4A is required for PPARγ-mediated regulation of a specific transcriptional program, including the lipolytic enzymes adipose triglyceride lipase (ATGL) and hormone-sensitive lipase (HSL). Hence, our findings reveal that GPS2 exerts a biologically important function in adipose tissue lipid mobilization by directly regulating ubiquitin signaling and indirectly modulating chromatin remodeling to prime selected genes for activation.


Asunto(s)
Histona Demetilasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , PPAR gamma/metabolismo , Regiones Promotoras Genéticas , Adipocitos/metabolismo , Animales , Ensamble y Desensamble de Cromatina , Lipasa/genética , Lipasa/metabolismo , Lipólisis/genética , Ratones , Esterol Esterasa/genética , Esterol Esterasa/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA