Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 134(7)2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38271119

RESUMEN

Loss of BRCA2 (breast cancer 2) is lethal for normal cells. Yet it remains poorly understood how, in BRCA2 mutation carriers, cells undergoing loss of heterozygosity overcome the lethality and undergo tissue-specific neoplastic transformation. Here, we identified mismatch repair gene mutL homolog 1 (MLH1) as a genetic interactor of BRCA2 whose overexpression supports the viability of Brca2-null cells. Mechanistically, we showed that MLH1 interacts with Flap endonuclease 1 (FEN1) and competes to process the RNA flaps of Okazaki fragments. Together, they restrained the DNA2 nuclease activity on the reversed forks of lagging strands, leading to replication fork (RF) stability in BRCA2-deficient cells. In these cells, MLH1 also attenuated R-loops, allowing the progression of stable RFs, which suppressed genomic instability and supported cell viability. We demonstrated the significance of their genetic interaction by the lethality of Brca2-mutant mice and inhibition of Brca2-deficient tumor growth in mice by Mlh1 loss. Furthermore, we described estrogen as inducing MLH1 expression through estrogen receptor α (ERα), which might explain why the majority of BRCA2 mutation carriers develop ER-positive breast cancer. Taken together, our findings reveal a role of MLH1 in relieving replicative stress and show how it may contribute to the establishment of BRCA2-deficient breast tumors.


Asunto(s)
Proteína BRCA2 , Neoplasias Mamarias Animales , Animales , Ratones , Proteína BRCA2/genética , Proteína BRCA2/metabolismo , Homólogo 1 de la Proteína MutL/genética , Homólogo 1 de la Proteína MutL/metabolismo , Reparación de la Incompatibilidad de ADN , Replicación del ADN
2.
Front Immunol ; 13: 1067417, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36685559

RESUMEN

Introduction: Ex vivo gene therapy for treatment of Inborn errors of Immunity (IEIs) have demonstrated significant clinical benefit in multiple Phase I/II clinical trials. Current approaches rely on engineered retroviral vectors to randomly integrate copy(s) of gene-of-interest in autologous hematopoietic stem/progenitor cells (HSPCs) genome permanently to provide gene function in transduced HSPCs and their progenies. To circumvent concerns related to potential genotoxicities due to the random vector integrations in HSPCs, targeted correction with CRISPR-Cas9-based genome editing offers improved precision for functional correction of multiple IEIs. Methods: We compare the two approaches for integration of IL2RG transgene for functional correction of HSPCs from patients with X-linked Severe Combined Immunodeficiency (SCID-X1 or XSCID); delivery via current clinical lentivector (LV)-IL2RG versus targeted insertion (TI) of IL2RG via homology-directed repair (HDR) when using an adeno-associated virus (AAV)-IL2RG donor following double-strand DNA break at the endogenous IL2RG locus. Results and discussion: In vitro differentiation of LV- or TI-treated XSCID HSPCs similarly overcome differentiation block into Pre-T-I and Pre-T-II lymphocytes but we observed significantly superior development of NK cells when corrected by TI (40.7% versus 4.1%, p = 0.0099). Transplants into immunodeficient mice demonstrated robust engraftment (8.1% and 23.3% in bone marrow) for LV- and TI-IL2RG HSPCs with efficient T cell development following TI-IL2RG in all four patients' HSPCs. Extensive specificity analysis of CRISPR-Cas9 editing with rhAmpSeq covering 82 predicted off-target sites found no evidence of indels in edited cells before (in vitro) or following transplant, in stark contrast to LV's non-targeted vector integration sites. Together, the improved efficiency and safety of IL2RG correction via CRISPR-Cas9-based TI approach provides a strong rationale for a clinical trial for treatment of XSCID patients.


Asunto(s)
Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X , Animales , Ratones , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/genética , Enfermedades por Inmunodeficiencia Combinada Ligada al Cromosoma X/terapia , Dependovirus , Sistemas CRISPR-Cas , Ratones SCID , Células Madre Hematopoyéticas
3.
BMC Cancer ; 21(1): 310, 2021 Mar 24.
Artículo en Inglés | MEDLINE | ID: mdl-33761896

RESUMEN

BACKGROUND: Chromosomal inversions involving anaplastic lymphoma kinase (ALK) and echinoderm microtubule associated protein like 4 (EML4) generate a fusion protein EML4-ALK in non-small cell lung cancer (NSCLC). The understanding of EML4-ALK function can be improved by a functional study using normal human cells. METHODS: Here we for the first time conduct such study to examine the effects of EML4-ALK on cell proliferation, cellular senescence, DNA damage, gene expression profiles and transformed phenotypes. RESULTS: The lentiviral expression of EML4-ALK in mortal, normal human fibroblasts caused, through its constitutive ALK kinase activity, an early induction of cellular senescence with accumulated DNA damage, upregulation of p16INK4A and p21WAF1, and senescence-associated ß-galactosidase (SA-ß-gal) activity. In contrast, when EML4-ALK was expressed in normal human fibroblasts transduced with telomerase reverse transcriptase (hTERT), which is activated in the vast majority of NSCLC, the cells showed accelerated proliferation and acquired anchorage-independent growth ability in soft-agar medium, without accumulated DNA damage, chromosome aberration, nor p53 mutation. EML4-ALK induced the phosphorylation of STAT3 in both mortal and hTERT-transduced cells, but RNA sequencing analysis suggested that the different signaling pathways contributed to the different phenotypic outcomes in these cells. While EML4-ALK also induced anchorage-independent growth in hTERT-immortalized human bronchial epithelial cells in vitro, the expression of EML4-ALK alone did not cause detectable in vivo tumorigenicity in immunodeficient mice. CONCLUSIONS: Our data indicate that the expression of hTERT is critical for EML4-ALK to manifest its in vitro transforming activity in human cells. This study provides the isogenic pairs of human cells with and without EML4-ALK expression.


Asunto(s)
Carcinogénesis/genética , Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Proteínas de Fusión Oncogénica/metabolismo , Telomerasa/metabolismo , Animales , Carcinogénesis/patología , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular , Proliferación Celular/genética , Senescencia Celular/genética , Daño del ADN , Modelos Animales de Enfermedad , Células Epiteliales , Femenino , Fibroblastos , Regulación Neoplásica de la Expresión Génica , Vectores Genéticos/genética , Humanos , Lentivirus/genética , Neoplasias Pulmonares/patología , Ratones , Proteínas de Fusión Oncogénica/genética , RNA-Seq , Telomerasa/genética , Homeostasis del Telómero/genética , Transfección
4.
Blood ; 137(19): 2598-2608, 2021 05 13.
Artículo en Inglés | MEDLINE | ID: mdl-33623984

RESUMEN

Lentivector gene therapy for X-linked chronic granulomatous disease (X-CGD) has proven to be a viable approach, but random vector integration and subnormal protein production from exogenous promoters in transduced cells remain concerning for long-term safety and efficacy. A previous genome editing-based approach using Streptococcus pyogenes Cas9 mRNA and an oligodeoxynucleotide donor to repair genetic mutations showed the capability to restore physiological protein expression but lacked sufficient efficiency in quiescent CD34+ hematopoietic cells for clinical translation. Here, we report that transient inhibition of p53-binding protein 1 (53BP1) significantly increased (2.3-fold) long-term homology-directed repair to achieve highly efficient (80% gp91phox+ cells compared with healthy donor control subjects) long-term correction of X-CGD CD34+ cells.


Asunto(s)
Reparación del ADN , Edición Génica/métodos , Terapia Genética/métodos , Enfermedad Granulomatosa Crónica/terapia , Trasplante de Células Madre Hematopoyéticas , NADPH Oxidasa 2/genética , Proteína 1 de Unión al Supresor Tumoral P53/antagonistas & inhibidores , Animales , Proteínas Bacterianas , Caspasa 9 , Células Cultivadas , Reparación del ADN/genética , Dependovirus/genética , Exones/genética , Vectores Genéticos/genética , Vectores Genéticos/uso terapéutico , Enfermedad Granulomatosa Crónica/genética , Células Madre Hematopoyéticas/enzimología , Xenoinjertos , Humanos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , NADPH Oxidasa 2/deficiencia , Fagocitos/metabolismo , ARN Guía de Kinetoplastida/genética , ARN Mensajero/genética , Especies Reactivas de Oxígeno , Ribonucleoproteínas/genética , Eliminación de Secuencia , Streptococcus pyogenes/enzimología
5.
BMC Cancer ; 15: 886, 2015 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-26555296

RESUMEN

BACKGROUND: USP18 (ubiquitin-specific protease 18) removes ubiquitin-like modifier interferon stimulated gene 15 (ISG15) from conjugated proteins. USP18 null mice in a FVB/N background develop tumors as early as 2 months of age. These tumors are leiomyosarcomas and thus represent a new murine model for this disease. METHODS: Heterozygous USP18 +/- FVB/N mice were bred to generate wild-type, heterozygous and homozygous cohorts. Tumors were characterized immunohistochemically and two cell lines were derived from independent tumors. Cell lines were karyotyped and their responses to restoration of USP18 activity assessed. Drug testing and tumorigenic assays were also performed. USP18 immunohistochemical staining in a large series of human leiomyosacomas was examined. RESULTS: USP18 -/- FVB/N mice spontaneously develop tumors predominantly on the back of the neck with most tumors evident between 6-12 months (80 % penetrance). Immunohistochemical characterization of the tumors confirmed they were leiomyosarcomas, which originate from smooth muscle. Restoration of USP18 activity in sarcoma-derived cell lines did not reduce anchorage dependent or independent growth or xenograft tumor formation demonstrating that these cells no longer require USP18 suppression for tumorigenesis. Karyotyping revealed that both tumor-derived cell lines were aneuploid with extra copies of chromosomes 3 and 15. Chromosome 15 contains the Myc locus and MYC is also amplified in human leiomyosarcomas. MYC protein levels were elevated in both murine leiomyosarcoma cell lines. Stabilized P53 protein was detected in a subset of these murine tumors, another feature of human leiomyosarcomas. Immunohistochemical analyses of USP18 in human leiomyosarcomas revealed a range of staining intensities with the highest USP18 expression in normal vascular smooth muscle. USP18 tissue array analysis of primary leiomyosarcomas from 89 patients with a clinical database revealed cases with reduced USP18 levels had a significantly decreased time to metastasis (P = 0.0441). CONCLUSIONS: USP18 null mice develop leiomyosarcoma recapitulating key features of clinical leiomyosarcomas and patients with reduced-USP18 tumor levels have an unfavorable outcome. USP18 null mice and the derived cell lines represent clinically-relevant models of leiomyosarcoma and can provide insights into both leiomyosarcoma biology and therapy.


Asunto(s)
Carcinogénesis/genética , Leiomiosarcoma/genética , Ubiquitina Tiolesterasa/genética , Neoplasias Uterinas/genética , Animales , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Leiomiosarcoma/patología , Ratones , Ratones Noqueados , Metástasis de la Neoplasia , Proteína p53 Supresora de Tumor/genética , Ubiquitina Tiolesterasa/biosíntesis , Neoplasias Uterinas/patología
6.
Oncotarget ; 6(30): 29469-81, 2015 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-26320182

RESUMEN

Osteosarcoma (OS) is the most common bone tumor in pediatric patients. Metastasis is a major cause of mortality and morbidity. The rarity of this disease coupled with the challenges of drug development for metastatic cancers have slowed the delivery of improvements in long-term outcomes for these patients. In this study, we collected 18 OS cell lines, confirmed their expression of bone markers and complex karyotypes, and characterized their in vivo tumorgenicity and metastatic potential. Since prior reports included conflicting descriptions of the metastatic and in vivo phenotypes of these models, there was a need for a comparative assessment of metastatic phenotypes using identical procedures in the hands of a single investigative group. We expect that this single characterization will accelerate the study of this metastatic cancer. Using these models we evaluated the expression of six previously reported metastasis-related OS genes. Ezrin was the only gene consistently differentially expressed in all the pairs of high/low metastatic OS cells. We then used a subtractive gene expression approach of the high and low human metastatic cells to identify novel genes that may be involved in OS metastasis. PHLDA1 (pleckstrin homology-like domain, family A) was identified as one of the genes more highly expressed in the high metastatic compared to low metastatic cells. Knocking down PHLDA1 with siRNA or shRNA resulted in down regulation of the activities of MAPKs (ERK1/2), c-Jun N-terminal kinases (JNK), and p38 mitogen-activated protein kinases (MAPKs). Reducing the expression of PHLDA1 also delayed OS metastasis progression in mouse xenograft models.


Asunto(s)
Neoplasias Óseas/patología , Movimiento Celular , Neoplasias Pulmonares/secundario , Osteosarcoma/secundario , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Invasividad Neoplásica , Osteosarcoma/genética , Osteosarcoma/metabolismo , Fenotipo , Interferencia de ARN , Transducción de Señal , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transfección , Ensayos Antitumor por Modelo de Xenoinjerto , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
7.
J Exp Med ; 210(5): 987-1001, 2013 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-23630229

RESUMEN

The (histone) deacetylase Sirt1 is a mediator of genomic and epigenetic maintenance, both of which are critical aspects of stem cell homeostasis and tightly linked to their functional decline in aging and disease. We show that Sirt1 ablation in adult hematopoietic stem and progenitor cells (HSPCs) promotes aberrant HSPC expansion specifically under conditions of hematopoietic stress, which is associated with genomic instability as well as the accumulation of DNA damage and eventually results in a loss of long-term progenitors. We further demonstrate that progenitor cell expansion is mechanistically linked to the selective up-regulation of the HSPC maintenance factor and polycomb target gene Hoxa9. We show that Sirt1 binds to the Hoxa9 gene, counteracts acetylation of its histone target H4 lysine 16, and in turn promotes polycomb-specific repressive histone modification. Together, these findings demonstrate a dual role for Sirt1 in HSPC homeostasis, both via epigenetic regulation of a key developmental gene and by promoting genome stability in adult stem cells.


Asunto(s)
Epigénesis Genética , Eliminación de Gen , Genoma/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Sirtuina 1/genética , Estrés Fisiológico/genética , Células Madre Adultas/citología , Células Madre Adultas/metabolismo , Animales , Trasplante de Médula Ósea , Proliferación Celular , Cromatina/metabolismo , Citoprotección/genética , Daño del ADN/genética , Sitios Genéticos/genética , Inestabilidad Genómica/genética , Histonas/metabolismo , Proteínas de Homeodominio/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Unión Proteica/genética , Sirtuina 1/deficiencia , Sirtuina 1/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
8.
Chromosome Res ; 21(3): 311-28, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23681662

RESUMEN

Sister chromatids contain identical DNA sequence but are chiral with respect to both their helical handedness and their replication history. Emerging evidence from various model organisms suggests that certain stem cells segregate sister chromatids nonrandomly to either maintain genome integrity or to bias cellular differentiation in asymmetric cell divisions. Conventional methods for tracing of old vs. newly synthesized DNA strands generally lack resolution for individual chromosomes and employ halogenated thymidine analogs with profound cytotoxic effects on rapidly dividing cells. Here, we present a modified chromosome orientation fluorescence in situ hybridization (CO-FISH) assay, where identification of individual chromosomes and their replication history is achieved in subsequent hybridization steps with chromosome-specific DNA probes and PNA telomere probes. Importantly, we tackle the issue of BrdU cytotoxicity and show that our method is compatible with normal mouse ES cell biology, unlike a recently published related protocol. Results from our CO-FISH assay show that mitotic segregation of mouse chromosome 7 is random in ES cells, which contrasts previously published results from our laboratory and settles a controversy. Our straightforward protocol represents a useful resource for future studies on chromatid segregation patterns of in vitro-cultured cells from distinct model organisms.


Asunto(s)
Cromátides/metabolismo , Segregación Cromosómica , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Hibridación Fluorescente in Situ/métodos , Mitosis , Animales , Bromodesoxiuridina/metabolismo , Bromodesoxiuridina/toxicidad , Supervivencia Celular/efectos de los fármacos , Segregación Cromosómica/efectos de los fármacos , Cromosomas de los Mamíferos/metabolismo , Células Madre Embrionarias/efectos de los fármacos , Ratones , Mitosis/efectos de los fármacos
9.
Hum Mol Genet ; 21(18): 3993-4006, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22678057

RESUMEN

Single-nucleotide substitutions and small in-frame insertions or deletions identified in human breast cancer susceptibility genes BRCA1 and BRCA2 are frequently classified as variants of unknown clinical significance (VUS) due to the availability of very limited information about their functional consequences. Such variants can most reliably be classified as pathogenic or non-pathogenic based on the data of their co-segregation with breast cancer in affected families and/or their co-occurrence with a pathogenic mutation. Biological assays that examine the effect of variants on protein function can provide important information that can be used in conjunction with available familial data to determine the pathogenicity of VUS. In this report, we have used a previously described mouse embryonic stem (mES) cell-based functional assay to characterize eight BRCA2 VUS that affect highly conserved amino acid residues and map to the N-terminal PALB2-binding or the C-terminal DNA-binding domains. For several of these variants, very limited co-segregation information is available, making it difficult to determine their pathogenicity. Based on their ability to rescue the lethality of Brca2-deficient mES cells and their effect on sensitivity to DNA-damaging agents, homologous recombination and genomic integrity, we have classified these variants as pathogenic or non-pathogenic. In addition, we have used homology-based modeling as a predictive tool to assess the effect of some of these variants on the structural integrity of the C-terminal DNA-binding domain and also generated a knock-in mouse model to analyze the physiological significance of a residue reported to be essential for the interaction of BRCA2 with meiosis-specific recombinase, DMC1.


Asunto(s)
Proteína BRCA2/genética , Neoplasias de la Mama/genética , Células Madre Embrionarias/metabolismo , Mutación , Proteínas Nucleares/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Secuencia de Aminoácidos , Animales , Proteína BRCA2/química , Proteínas de Ciclo Celular , Supervivencia Celular , Células Cultivadas , Mapeo Cromosómico , Secuencia Conservada , Roturas del ADN de Doble Cadena , Reparación del ADN , Proteínas de Unión al ADN , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/fisiología , Proteína del Grupo de Complementación N de la Anemia de Fanconi , Femenino , Estudios de Asociación Genética , Humanos , Funciones de Verosimilitud , Masculino , Ratones , Ratones Transgénicos , Mitomicina/farmacología , Modelos Moleculares , Mutágenos/farmacología , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Estructura Cuaternaria de Proteína , Homología Estructural de Proteína
10.
Nat Med ; 18(2): 227-34, 2012 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-22231558

RESUMEN

In addition to allelic mutations, cancers are known to harbor alterations in their chromatin landscape. Here we show that genomic ablation of Smad ubiquitin regulatory factor 2 (Smurf2), a HECT-domain E3 ubiquitin ligase, results in dysregulation of both the DNA damage response and genomic stability, culminating in increased susceptibility to various types of cancers in aged mice. We show that Smurf2 regulates the monoubiquitination of histone H2B as well as the trimethylation of histone H3 at Lys4 and Lys79 by targeting ring finger protein 20 (RNF20) for proteasomal degradation in both mouse and human cells. We also show that Smurf2 and RNF20 are colocalized at the γ-H2AX foci of double-stranded DNA breaks in the nucleus. Thus, Smurf2 has a tumor suppression function that normally maintains genomic stability by controlling the epigenetic landscape of histone modifications through RNF20.


Asunto(s)
Cromatina/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Cromatina/metabolismo , Roturas del ADN de Doble Cadena , Daño del ADN/fisiología , Metilación de ADN/fisiología , Reparación del ADN/fisiología , Genes Supresores de Tumor/fisiología , Histonas/metabolismo , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias/fisiopatología , Ubiquitinación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA