Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Biotechnol ; 37(8): 945-952, 2019 08.
Article in English | MEDLINE | ID: mdl-31359006

ABSTRACT

Engineered nucleases have gained broad appeal for their ability to mediate highly efficient genome editing. However the specificity of these reagents remains a concern, especially for therapeutic applications, given the potential mutagenic consequences of off-target cleavage. Here we have developed an approach for improving the specificity of zinc finger nucleases (ZFNs) that engineers the FokI catalytic domain with the aim of slowing cleavage, which should selectively reduce activity at low-affinity off-target sites. For three ZFN pairs, we engineered single-residue substitutions in the FokI domain that preserved full on-target activity but showed a reduction in off-target indels of up to 3,000-fold. By combining this approach with substitutions that reduced the affinity of zinc fingers, we developed ZFNs specific for the TRAC locus that mediated 98% knockout in T cells with no detectable off-target activity at an assay background of ~0.01%. We anticipate that this approach, and the FokI variants we report, will enable routine generation of nucleases for gene editing with no detectable off-target activity.


Subject(s)
DNA Cleavage , Gene Editing/methods , T-Lymphocytes , Base Sequence , DNA/genetics , DNA/metabolism , Flow Cytometry , Hematopoietic Stem Cells , Humans , K562 Cells , Protein Domains , RNA, Messenger
2.
J Exp Med ; 212(5): 809-24, 2015 May 04.
Article in English | MEDLINE | ID: mdl-25847946

ABSTRACT

Rag1 and Rag2 gene expression in CD4(+)CD8(+) double-positive (DP) thymocytes depends on the activity of a distant anti-silencer element (ASE) that counteracts the activity of an intergenic silencer. However, the mechanistic basis for ASE activity is unknown. Here, we show that the ASE physically interacts with the distant Rag1 and Rag2 gene promoters in DP thymocytes, bringing the two promoters together to form an active chromatin hub. Moreover, we show that the ASE functions as a classical enhancer that can potently activate these promoters in the absence of the silencer or other locus elements. In thymocytes lacking the chromatin organizer SATB1, we identified a partial defect in Tcra gene rearrangement that was associated with reduced expression of Rag1 and Rag2 at the DP stage. SATB1 binds to the ASE and Rag promoters, facilitating inclusion of Rag2 in the chromatin hub and the loading of RNA polymerase II to both the Rag1 and Rag2 promoters. Our results provide a novel framework for understanding ASE function and demonstrate a novel role for SATB1 as a regulator of Rag locus organization and gene expression in DP thymocytes.


Subject(s)
Cell Differentiation/immunology , Chromatin/immunology , DNA-Binding Proteins/immunology , Homeodomain Proteins/immunology , Matrix Attachment Region Binding Proteins/immunology , Response Elements/immunology , Thymocytes/immunology , Animals , Cell Differentiation/genetics , Chromatin/genetics , DNA-Binding Proteins/genetics , Gene Expression Regulation/genetics , Homeodomain Proteins/genetics , Matrix Attachment Region Binding Proteins/genetics , Mice , Mice, Knockout , Thymocytes/cytology
3.
J Cutan Pathol ; 41(11): 846-52, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25263506

ABSTRACT

BACKGROUND: High mitotic figure count (MFC) correlates with low survival rate in Merkel cell carcinoma (MCC). However, the prognostic impact of histone biomarkers as surrogates of MFC in MCC is unknown. We evaluated the prognostic significance of the immunodetection of mitotic figures and of G2+ tumor nuclei with histone-associated mitotic markers H3K79me3T80ph (H3KT) and phosphohistone H3 (PHH3) in MCC. METHODS: Immunohistochemical analyses of H3KT and PHH3 and proliferative marker Ki-67 were performed in a series of 21 cases of MCC. The significance of the pathologic data and immunoreactivity with these markers was evaluated with Pearson correlation and paired Student t-test. Univariate Cox proportional hazards regression models were performed to assess the relationships between these markers and survival. RESULTS: H3KT detected a higher number of mitotic figure (p<0.0001) and G2+ tumor nuclei (p<0.0052) than did PHH3. Furthermore, the MFC combined with G2+ tumor nuclei detected with H3KT compared to PHH3 and manual MFC was a significant predictor of impaired survival in patients with MCC (p=0.035; HR=1.0172), corresponding to a 1.72% increased risk of death for each unit increase in H3KT. CONCLUSIONS: Biomarker analysis of proliferative rates with histone markers may have relevance in stratifying risk in patients with MCC.


Subject(s)
Biomarkers, Tumor/analysis , Carcinoma, Merkel Cell/pathology , Cell Nucleus/pathology , Histones/analysis , Skin Neoplasms/pathology , Aged , Aged, 80 and over , Carcinoma, Merkel Cell/mortality , Cell Proliferation , Female , Humans , Immunohistochemistry , Male , Middle Aged , Mitosis , Prognosis , Proportional Hazards Models , Skin Neoplasms/mortality
4.
J Skin Cancer ; 2012: 823534, 2012.
Article in English | MEDLINE | ID: mdl-23227340

ABSTRACT

The current study characterizes the mitosis-associated histone dual modification on the core of histone H3: trimethylation of histone H3 lysine 79 and simultaneous phosphorylation of H3 threonine 80 (H3K79me3T80ph). Through the use of protein and microscopy-based techniques, we find that H3K79me3T80ph shares a similar spatial and temporal regulation as H3S10ph but additionally requires methyltransferase activity. In addition, we find that Aurora kinase activity is necessary for the catalysis of H3K79me3T80ph in vivo. Finally, our analysis of H3K79me3T80ph using a tissue microarray indicates that H3K79me3T80ph marks a subset of primary cutaneous melanomas with metastatic potential indicating that H3K79me3T80ph may identify a subset of invasive melanomas with a more aggressive clinical behaviour.

5.
Methods ; 58(3): 243-54, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22782115

ABSTRACT

Mammalian genomes are organized into multiple layers of higher-order chromatin structure, and in this organization chromatin looping is a striking and crucial feature that brings together distal genomic loci into close spatial proximity. Such three-dimensional organization of chromatin has been suggested to be functionally important in gene regulation. Many important questions need to be addressed, such as what types of nuclear proteins are responsible for folding chromatin into loops, whether there are any genomic marks that serve as the core sites of chromatin folding events, how distal genomic sites are brought together, and what are the biological consequences for interactions between distal genomic loci. In order to address these fundamental questions, it is essential to devise and employ methods that can capture higher-order structures formed by specific nuclear proteins at high resolution. In this article, in order to describe methods of analyzing protein-mediated chromatin interactions, we will use as an example a global genome-organizer protein, SATB1, which mediates chromatin looping.


Subject(s)
Chromatin/genetics , Matrix Attachment Region Binding Proteins/physiology , Animals , Base Sequence , Binding Sites , Chromatin Immunoprecipitation/methods , Chromosome Mapping/methods , Cross-Linking Reagents/chemistry , Formaldehyde/chemistry , Humans , Models, Molecular , Molecular Sequence Data , Nucleic Acid Conformation
6.
Exp Gerontol ; 45(4): 279-85, 2010 Apr.
Article in English | MEDLINE | ID: mdl-19818845

ABSTRACT

HDAC1 is a member of the class I of histone deacetylases that also includes HDAC2, -3 and -8. Although HDAC1 has been mostly studied in the context of cancer, recent evidence strongly suggests that it plays critical roles in cellular senescence, aging of the liver, myelination, and adult neurogenesis. Here we review such roles and discuss the entangled relationships between HDAC1 with histone acetyltransferases and other HDACs including SIRT1.


Subject(s)
Aging/physiology , Cellular Senescence/physiology , Histone Deacetylase 1/physiology , Neoplasms/physiopathology , Animals , Histones/physiology , Humans , Mice , Myelin Sheath/physiology , Neurogenesis/physiology
7.
Pigment Cell Melanoma Res ; 22(1): 14-29, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19040501

ABSTRACT

Melanoma is a highly heterogeneous cancer that comes in different flavors (lentigo maligna melanoma, superficial spreading melanoma, nodular melanoma, acral lentiginous/mucosal melanoma and other less common subtypes including malignant cellular blue nevus, desmoplastic melanoma, nevoid melanoma, and animal-type melanoma) and colors (black/bluish or unpigmented). Pathologists have known for many years that melanoma displays notable changes in the nuclear architecture including thick chromatic rims, presence of mitosis, nuclear grooves, and more. It is now evident from other cancers that such changes reflect not only genomic alterations but also non-genomic changes in both the structure of DNA and the structure of chromatin to which the DNA is bound (nucleosomes). Although aberrant gene expression resulting from DNA methylation has been known for many years, genome alterations resulting from histone modifications became evident in the current decade. In prostate and other cancers, some histone marks have clinical diagnostic and/or prognostic value. Here, we review the current data on epigenetic research in melanoma skin cancers, discuss ways to modify the epigenetic landscape of melanoma for inhibiting its growth, and propose strategies for identifying novel melanoma markers.


Subject(s)
Epigenesis, Genetic , Melanoma/genetics , Humans
8.
J Cutan Pathol ; 36(2): 197-205, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18647205

ABSTRACT

BACKGROUND: The p27(KIP1) and p57(KIP2) proteins belong to the CIP/KIP family of cyclin-dependent kinase inhibitors involved in the growth arrest and cellular senescence. High levels of p27(KIP1) unexpectedly have been detected in invasive malignant melanomas (MM), whereas the role of p57(KIP2) in melanocytic lesions is unknown. We therefore chose to study the expression of p27(KIP1) and p57(KIP2) in melanocytic neoplasms. DESIGN: The expression of p27(KIP1) and p57(KIP2) were examined by immunohistochemistry in 40 melanocytic neoplasms and by Western blot analysis in cultured human melanocytes. RESULTS: Expression of both nuclear p27(KIP1) and p57(KIP2) (> 10% of cells with nuclear labeling) was observed in most cases with non-proliferating melanocytes (8/10, benign nevi and 9/10 DN, dysplastic nevi), but in only a few cases containing proliferating melanocytes (3/11 RN, recurrent nevi and 2/9 MM, melanoma) (p < 0.002). In proliferating melanocytes, there was an inverse correlation of nuclear expression of p27(KIP1) and p57(KIP2) in both RN (p27(KIP1) = 3/11 RN and p57(KIP2) = 8/11 RN) and MM (p27(KIP1) = 7/9 MM and p57(KIP2) = 2/9 MM) (p < 0.05). Western blot analysis detected p57(KIP2) only in proliferating melanocytes. p27(KIP1) was detected in both proliferating and senescent melanocytes. CONCLUSION: The difference in expression patterns of p27(KIP1) and p57(KIP2) in proliferating and senescent melanocytes suggests the interplay between these proteins may play a functional role in melanocytic tumorigenesis.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p57/biosynthesis , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/metabolism , Melanocytes/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Cell Proliferation , Cells, Cultured , Cellular Senescence , Cyclin-Dependent Kinase Inhibitor p27 , Humans , Melanocytes/pathology , Melanoma/pathology , Skin Neoplasms/pathology
9.
Aging Cell ; 6(4): 577-91, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17578512

ABSTRACT

The retinoblastoma (RB)/p16(INK4a) pathway regulates senescence of human melanocytes in culture and oncogene-induced senescence of melanocytic nevi in vivo. This senescence response is likely due to chromatin modifications because RB complexes from senescent melanocytes contain increased levels of histone deacetylase (HDAC) activity and tethered HDAC1. Here we show that HDAC1 is prominently detected in p16(INK4a)-positive, senescent intradermal melanocytic nevi but not in proliferating, recurrent nevus cells that localize to the epidermal/dermal junction. To assess the role of HDAC1 in the senescence of melanocytes and nevi, we used tetracycline-based inducible expression systems in cultured melanocytic cells. We found that HDAC1 drives a sequential and cooperative activity of chromatin remodeling effectors, including transient recruitment of Brahma (Brm1) into RB/HDAC1 mega-complexes, formation of heterochromatin protein 1 beta (HP1 beta)/SUV39H1 foci, methylation of H3-K9, stable association of RB with chromatin and significant global heterochromatinization. These chromatin changes coincide with expression of typical markers of senescence, including the senescent-associated beta-galactosidase marker. Notably, formation of RB/HP1 beta foci and early tethering of RB to chromatin depends on intact Brm1 ATPase activity. As cells reached senescence, ejection of Brm1 from chromatin coincided with its dissociation from HP1 beta/RB and relocalization to protein complexes of lower molecular weight. These results provide new insights into the role of the RB pathway in regulating cellular senescence and implicate HDAC1 as a likely mediator of early chromatin remodeling events.


Subject(s)
Cellular Senescence/physiology , Chromatin/metabolism , Histone Deacetylases/metabolism , Melanocytes/physiology , Nevus, Pigmented/pathology , Cell Line , Chromatin/genetics , Chromatin Assembly and Disassembly , Chromobox Protein Homolog 5 , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Gene Silencing , Genes, p16 , Heterochromatin/genetics , Heterochromatin/metabolism , Histone Deacetylase 1 , Histone Deacetylases/genetics , Histones/genetics , Histones/metabolism , Humans , Melanocytes/cytology , Melanocytes/metabolism , Nevus, Pigmented/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...