Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Genet ; 55(9): 1542-1554, 2023 09.
Article in English | MEDLINE | ID: mdl-37580596

ABSTRACT

Cellular differentiation requires extensive alterations in chromatin structure and function, which is elicited by the coordinated action of chromatin and transcription factors. By contrast with transcription factors, the roles of chromatin factors in differentiation have not been systematically characterized. Here, we combine bulk ex vivo and single-cell in vivo CRISPR screens to characterize the role of chromatin factor families in hematopoiesis. We uncover marked lineage specificities for 142 chromatin factors, revealing functional diversity among related chromatin factors (i.e. barrier-to-autointegration factor subcomplexes) as well as shared roles for unrelated repressive complexes that restrain excessive myeloid differentiation. Using epigenetic profiling, we identify functional interactions between lineage-determining transcription factors and several chromatin factors that explain their lineage dependencies. Studying chromatin factor functions in leukemia, we show that leukemia cells engage homeostatic chromatin factor functions to block differentiation, generating specific chromatin factor-transcription factor interactions that might be therapeutically targeted. Together, our work elucidates the lineage-determining properties of chromatin factors across normal and malignant hematopoiesis.


Subject(s)
Chromatin , Leukemia , Humans , Chromatin/genetics , Cell Lineage/genetics , Hematopoiesis/genetics , Cell Differentiation/genetics , Transcription Factors/genetics
2.
Nucleic Acids Res ; 44(16): 7592-604, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27179028

ABSTRACT

Methylation of DNA at carbon 5 of cytosine is essential for mammalian development and implicated in transcriptional repression of genes and transposons. New patterns of DNA methylation characteristic of lineage-committed cells are established at the exit from pluripotency by de novo DNA methyltransferases enzymes, DNMT3A and DNMT3B, which are regulated by developmental signaling and require access to chromatin-organized DNA. Whether or not the capacity for de novo DNA methylation of developmentally regulated loci is preserved in differentiated somatic cells and can occur in the absence of exogenous signals is currently unknown. Here, we demonstrate that fibroblasts derived from chromatin remodeling ATPase LSH (HELLS)-null mouse embryos, which lack DNA methylation from centromeric repeats, transposons and a number of gene promoters, are capable of reestablishing DNA methylation and silencing of misregulated genes upon re-expression of LSH. We also show that the ability of LSH to bind ATP and the cellular concentration of DNMT3B are critical for cell-autonomous de novo DNA methylation in somatic cells. These data suggest the existence of cellular memory that persists in differentiated cells through many cell generations and changes in transcriptional state.


Subject(s)
Adenosine Triphosphatases/metabolism , DNA Helicases/metabolism , DNA Methylation , Fibroblasts/metabolism , 5-Methylcytosine/metabolism , Animals , Cell Differentiation/genetics , Cell Line , DNA (Cytosine-5-)-Methyltransferases/metabolism , Embryo, Mammalian/cytology , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Gene Silencing , Mice , Mutation/genetics , NIH 3T3 Cells , Promoter Regions, Genetic/genetics , Repetitive Sequences, Nucleic Acid/genetics , Retroelements/genetics , DNA Methyltransferase 3B
3.
Oncotarget ; 6(27): 23944-58, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26284587

ABSTRACT

Breast cancer is a heterogeneous disease that can be subdivided into clinical, histopathological and molecular subtypes (luminal A-like, luminal B-like/HER2-negative, luminal B-like/HER2-positive, HER2-positive, and triple-negative). The study of new molecular factors is essential to obtain further insights into the mechanisms involved in the tumorigenesis of each tumor subtype. RASSF2 is a gene that is hypermethylated in breast cancer and whose clinical value has not been previously studied. The hypermethylation of RASSF1 and RASSF2 genes was analyzed in 198 breast tumors of different subtypes. The effect of the demethylating agent 5-aza-2'-deoxycytidine in the re-expression of these genes was examined in triple-negative (BT-549), HER2 (SK-BR-3), and luminal cells (T-47D). Different patterns of RASSF2 expression for distinct tumor subtypes were detected by immunohistochemistry. RASSF2 hypermethylation was much more frequent in luminal subtypes than in non-luminal tumors (p = 0.001). The re-expression of this gene by lentiviral transduction contributed to the differential cell proliferation and response to antineoplastic drugs observed in luminal compared with triple-negative cell lines. RASSF2 hypermethylation is associated with better prognosis in multivariate statistical analysis (P = 0.039). In conclusion, RASSF2 gene is differently methylated in luminal and non-luminal tumors and is a promising suppressor gene with clinical involvement in breast cancer.


Subject(s)
Breast Neoplasms/diagnosis , Breast Neoplasms/genetics , DNA Methylation , Gene Expression Regulation, Neoplastic , Tumor Suppressor Proteins/physiology , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/chemistry , Azacitidine/chemistry , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Proliferation , Female , Gene Expression Profiling , HEK293 Cells , Humans , Hydroxamic Acids/chemistry , Immunohistochemistry , Kaplan-Meier Estimate , Middle Aged , Multivariate Analysis , Prognosis , Proportional Hazards Models , Treatment Outcome , Tumor Suppressor Proteins/genetics
4.
Mod Pathol ; 26(8): 1111-22, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23542458

ABSTRACT

Ras association (RalGDS/AF-6) domain family member 2 (RASSF2) is a gene involved in the progression of several human cancers, including breast, colorectal and lung cancer. The aims of this study were to determine the hypermethylation of the gene in squamous cervical cancer and precursor lesions, along with that of RASSF1 and the recently described EPB41L3, and to analyze the potential prognostic role of these genes. Methylation-specific PCR and bisulfite sequencing were used to analyze the methylation status of RASSF2 and EPB41L3 gene in 60 squamous cervical cancer, 76 cervical intraepithelial neoplasias grade III, 16 grade II, 14 grade I and 13 cases of normal tissue adjacent to cervical intraepithelial neoplasia. RASSF2 expression was evaluated by immunohistochemistry and the re-expression of RASSF2 and EPB41L3 was analyzed by quantitative reverse-transcription PCR in HeLa, SiHa, C33A and A431 cell lines treated with 5-aza-2'-deoxycytidine and/or trichostatin. RASSF1 hypermethylation and human papillomavirus type were also analyzed in all the cases by methylation-specific PCR and reverse line blot, respectively. RASSF2 hypermethylation was predominant in squamous cervical cancer (60.9%) compared with cervical intraepithelial neoplasias (4.2%) and was associated with a lower level of RASSF2 expression and vascular invasion in squamous cervical cancer. EPB41L3 and RASSF1 hypermethylations were also more frequent in cancer than in precursor lesions. Patients with RASSF2 hypermethylation had shorter survival time, independent of tumor stage (hazard ratio: 6.0; 95% confidence interval: 1.5-24.5). Finally, the expressions of RASSF2 and EPB41L3 were restored in several cell lines treated with 5-aza-2'-deoxycytidine. Taken together, our results suggest that RASSF2 potentially functions as a new tumor-suppressor gene that is inactivated through hypermethylation in cervical cancer and is related to the bad prognosis of these patients.


Subject(s)
Carcinoma, Squamous Cell/genetics , DNA Methylation/genetics , Genes, Tumor Suppressor , Tumor Suppressor Proteins/genetics , Uterine Cervical Neoplasms/genetics , Adult , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Microfilament Proteins/genetics , Middle Aged , Prognosis , Proportional Hazards Models , Reverse Transcriptase Polymerase Chain Reaction , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology , Uterine Cervical Dysplasia
5.
Pathol Int ; 62(7): 491-5, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22726069

ABSTRACT

Pleomorphic ductal carcinoma of the breast is a rare variant included in the morphological group of infiltrating ductal carcinoma. The pleomorphic carcinoma is composed predominantly of epithelial and multinucleated tumor giant cells. We report here two cases presenting a lesion composed microscopically of a proliferation of large pleomorphic cells with a predominance of multinucleated giant cells. These lesions were negative for estrogen receptor, progesterone receptor and Her2-neu (triple-negative phenotype). Basal markers (cytokeratin 5/6, cytokeratin 17 and epidermal growth factor receptor [EGFR]) were present, accompanied by the presence of histiocyte marker CD163 in most neoplastic giant cells. High-grade pleomorphic breast carcinomas with the triple-negative phenotype and expression of basal markers might be included in the basal subtype. This is the first report about the co-expression of macrophage marker CD163, with tumor (P53) or epithelial markers (CAM5.2), as indicated by double immunohistochemistry in pleomorphic ductal carcinoma of the breast.


Subject(s)
Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Giant Cells/pathology , Macrophages/pathology , Aged , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Biomarkers/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/surgery , ErbB Receptors/metabolism , Female , Giant Cells/metabolism , Humans , Keratins/metabolism , Macrophages/metabolism , Middle Aged , Receptors, Cell Surface/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...