Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
Mais filtros

Bases de dados
Tipo de documento
Intervalo de ano de publicação
2.
Nat Immunol ; 18(2): 173-183, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27992401

RESUMO

Most Foxp3+ regulatory T (Treg) cells develop in the thymus as a functionally mature T cell subpopulation specialized for immune suppression. Their cell fate appears to be determined before Foxp3 expression; yet molecular events that prime Foxp3- Treg precursor cells are largely obscure. We found that Treg cell-specific super-enhancers (Treg-SEs), which were associated with Foxp3 and other Treg cell signature genes, began to be activated in Treg precursor cells. T cell-specific deficiency of the genome organizer Satb1 impaired Treg-SE activation and the subsequent expression of Treg signature genes, causing severe autoimmunity due to Treg cell deficiency. These results suggest that Satb1-dependent Treg-SE activation is crucial for Treg cell lineage specification in the thymus and that its perturbation is causative of autoimmune and other immunological diseases.


Assuntos
Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Linfócitos T Reguladores/fisiologia , Ativação Transcricional/imunologia , Animais , Autoimunidade , Linhagem da Célula , Células Cultivadas , Elementos Facilitadores Genéticos/genética , Epigênese Genética , Fatores de Transcrição Forkhead/genética , Tolerância Imunológica , Masculino , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Especificidade de Órgãos , Células Precursoras de Linfócitos T/fisiologia
3.
J Immunol ; 211(2): 209-218, 2023 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-37256264

RESUMO

Double-positive thymocytes that have passed positive selection migrate from the cortex to the medulla, where negative selection and the development of thymic regulatory T cells (tTregs) take place. Medullary thymic epithelial cells (mTECs) play important roles in these selections, and their differentiation and maintenance depend on interaction with positively selected CD4+ single-positive cells. Therefore, migration and differentiation after positive selection must be coordinated to establish immune tolerance. However, the regulatory mechanisms of these processes are not fully understood. SATB1 is a genome organizer highly expressed in double-positive thymocytes, and SATB1 deletion causes various defects in T-cell development, including impaired positive and negative selection and tTreg differentiation. Here, we show that SATB1 is critical for temporally coordinated thymocyte trafficking after positive selection in mice. Satb1 knockout (ΔSatb1) led to precocious thymic egress caused by augmented S1pr1 upregulation in positively selected thymocytes, accompanied by lower induction of Ccr7, Tnfsf11, and Cd40lg. Altered thymocyte trafficking and functionality affected the differentiation of mTECs and, in turn, tTreg differentiation. Thus, SATB1 is required to establish immune tolerance, at least in part, by ensuring timely thymic egress and mTEC differentiation.


Assuntos
Proteínas de Ligação à Região de Interação com a Matriz , Timócitos , Animais , Camundongos , Diferenciação Celular , Células Epiteliais , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos Knockout , Timo , Fatores de Transcrição
4.
Mol Cancer ; 23(1): 40, 2024 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-38383439

RESUMO

Finding effective therapeutic targets to treat NRAS-mutated melanoma remains a challenge. Long non-coding RNAs (lncRNAs) recently emerged as essential regulators of tumorigenesis. Using a discovery approach combining experimental models and unbiased computational analysis complemented by validation in patient biospecimens, we identified a nuclear-enriched lncRNA (AC004540.4) that is upregulated in NRAS/MAPK-dependent melanoma, and that we named T-RECS. Considering potential innovative treatment strategies, we designed antisense oligonucleotides (ASOs) to target T-RECS. T-RECS ASOs reduced the growth of melanoma cells and induced apoptotic cell death, while having minimal impact on normal primary melanocytes. Mechanistically, treatment with T-RECS ASOs downregulated the activity of pro-survival kinases and reduced the protein stability of hnRNPA2/B1, a pro-oncogenic regulator of MAPK signaling. Using patient- and cell line- derived tumor xenograft mouse models, we demonstrated that systemic treatment with T-RECS ASOs significantly suppressed the growth of melanoma tumors, with no noticeable toxicity. ASO-mediated T-RECS inhibition represents a promising RNA-targeting approach to improve the outcome of MAPK pathway-activated melanoma.


Assuntos
Melanoma , RNA Longo não Codificante , Humanos , Camundongos , Animais , Melanoma/patologia , RNA Longo não Codificante/genética , Apoptose/genética , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/uso terapêutico , Linhagem Celular Tumoral , Proteínas de Membrana/genética , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo
6.
Immunity ; 38(6): 1105-15, 2013 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-23791645

RESUMO

How hematopoietic stem cells (HSCs) produce particular lineages is insufficiently understood. We searched for key factors that direct HSC to lymphopoiesis. Comparing gene expression profiles for HSCs and early lymphoid progenitors revealed that Satb1, a global chromatin regulator, was markedly induced with lymphoid lineage specification. HSCs from Satb1-deficient mice were defective in lymphopoietic activity in culture and failed to reconstitute T lymphopoiesis in wild-type recipients. Furthermore, Satb1 transduction of HSCs and embryonic stem cells robustly promoted their differentiation toward lymphocytes. Whereas genes that encode Ikaros, E2A, and Notch1 were unaffected, many genes involved in lineage decisions were regulated by Satb1. Satb1 expression was reduced in aged HSCs with compromised lymphopoietic potential, but forced Satb1 expression partly restored that potential. Thus, Satb1 governs the initiating process central to the replenishing of lymphoid lineages. Such activity in lymphoid cell generation may be of clinical importance and useful to overcome immunosenescence.


Assuntos
Células-Tronco Hematopoéticas/fisiologia , Linfopoese , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Linfócitos T/fisiologia , Animais , Diferenciação Celular/genética , Linhagem da Célula/genética , Sobrevivência Celular/genética , Células Cultivadas , Senescência Celular/genética , Montagem e Desmontagem da Cromatina/genética , Regulação da Expressão Gênica , Humanos , Linfopoese/genética , Proteínas de Ligação à Região de Interação com a Matriz/genética , Proteínas de Ligação à Região de Interação com a Matriz/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transgenes/genética
7.
Int J Mol Sci ; 22(18)2021 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-34576286

RESUMO

Sjögren's syndrome (SS) is a chronic autoimmune disease characterized by dysfunction of salivary and lacrimal glands, resulting in xerostomia (dry mouth) and keratoconjunctivitis sicca (dry eyes). Autoantibodies, such as anti-SSA and anti-SSB antibodies, are hallmarks and important diagnostic factors for SS. In our previous study, we demonstrated that SS-like xerostomia was observed in SATB1 conditional knockout (SATB1cKO) mice, in which the floxed SATB1 gene was specifically deleted in hematopoietic cells as early as 4 weeks of age. In these mice, autoantibodies were not detected until 8 weeks of age in SATB1cKO mice, although exocrine gland function reached its lowest at this age. Therefore, other markers may be necessary for the diagnosis of SS in the early phase. Here, we found that mRNA expression of the interferonγ (IFN-γ) gene and the IFN-responsive indoleamine 2,3-dioxygenase (IDO) gene is upregulated in the salivary glands of SATB1cKO mice after 3 and 4 weeks of age, respectively. We detected l-kynurenine (l-KYN), an intermediate of l-tryptophan (l-Trp) metabolism mediated by IDO, in the serum of SATB1cKO mice after 4 weeks of age. In addition, the upregulation of IDO expression was significantly suppressed by the administration of IFN-γ neutralizing antibodies in SATB1cKO mice. These results suggest that the induction of IFN-dependent IDO expression is an initial event that occurs immediately after the onset of SS in SATB1cKO mice. These results also imply that serum l-KYN could be used as a marker for SS diagnosis in the early phases of the disease before autoantibodies are detectable.


Assuntos
Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/deficiência , Síndrome de Sjogren/enzimologia , Animais , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Interferon gama/metabolismo , Cinurenina/sangue , Cinurenina/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Saliva/metabolismo , Glândulas Salivares/metabolismo , Síndrome de Sjogren/sangue , Triptofano/metabolismo , Regulação para Cima
8.
Nature ; 514(7521): 257-61, 2014 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-25119036

RESUMO

Homeodomain proteins, described 30 years ago, exert essential roles in development as regulators of target gene expression; however, the molecular mechanisms underlying transcriptional activity of homeodomain factors remain poorly understood. Here investigation of a developmentally required POU-homeodomain transcription factor, Pit1 (also known as Pou1f1), has revealed that, unexpectedly, binding of Pit1-occupied enhancers to a nuclear matrin-3-rich network/architecture is a key event in effective activation of the Pit1-regulated enhancer/coding gene transcriptional program. Pit1 association with Satb1 (ref. 8) and ß-catenin is required for this tethering event. A naturally occurring, dominant negative, point mutation in human PIT1(R271W), causing combined pituitary hormone deficiency, results in loss of Pit1 association with ß-catenin and Satb1 and therefore the matrin-3-rich network, blocking Pit1-dependent enhancer/coding target gene activation. This defective activation can be rescued by artificial tethering of the mutant R271W Pit1 protein to the matrin-3 network, bypassing the pre-requisite association with ß-catenin and Satb1 otherwise required. The matrin-3 network-tethered R271W Pit1 mutant, but not the untethered protein, restores Pit1-dependent activation of the enhancers and recruitment of co-activators, exemplified by p300, causing both enhancer RNA transcription and target gene activation. These studies have thus revealed an unanticipated homeodomain factor/ß-catenin/Satb1-dependent localization of target gene regulatory enhancer regions to a subnuclear architectural structure that serves as an underlying mechanism by which an enhancer-bound homeodomain factor effectively activates developmental gene transcriptional programs.


Assuntos
Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas de Ligação a RNA/metabolismo , Transcrição Gênica , Animais , Células Cultivadas , Proteínas de Homeodomínio/genética , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos , Hipófise/embriologia , Hipófise/metabolismo , Ligação Proteica , Fator de Transcrição Pit-1/genética , Fator de Transcrição Pit-1/metabolismo , Transcrição Gênica/genética , beta Catenina/metabolismo
9.
PLoS Genet ; 13(9): e1006966, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28863138

RESUMO

Mammalian genomes contain several dozens of large (>0.5 Mbp) lineage-specific gene loci harbouring functionally related genes. However, spatial chromatin folding, organization of the enhancer-promoter networks and their relevance to Topologically Associating Domains (TADs) in these loci remain poorly understood. TADs are principle units of the genome folding and represents the DNA regions within which DNA interacts more frequently and less frequently across the TAD boundary. Here, we used Chromatin Conformation Capture Carbon Copy (5C) technology to characterize spatial chromatin interaction network in the 3.1 Mb Epidermal Differentiation Complex (EDC) locus harbouring 61 functionally related genes that show lineage-specific activation during terminal keratinocyte differentiation in the epidermis. 5C data validated by 3D-FISH demonstrate that the EDC locus is organized into several TADs showing distinct lineage-specific chromatin interaction networks based on their transcription activity and the gene-rich or gene-poor status. Correlation of the 5C results with genome-wide studies for enhancer-specific histone modifications (H3K4me1 and H3K27ac) revealed that the majority of spatial chromatin interactions that involves the gene-rich TADs at the EDC locus in keratinocytes include both intra- and inter-TAD interaction networks, connecting gene promoters and enhancers. Compared to thymocytes in which the EDC locus is mostly transcriptionally inactive, these interactions were found to be keratinocyte-specific. In keratinocytes, the promoter-enhancer anchoring regions in the gene-rich transcriptionally active TADs are enriched for the binding of chromatin architectural proteins CTCF, Rad21 and chromatin remodeler Brg1. In contrast to gene-rich TADs, gene-poor TADs show preferential spatial contacts with each other, do not contain active enhancers and show decreased binding of CTCF, Rad21 and Brg1 in keratinocytes. Thus, spatial interactions between gene promoters and enhancers at the multi-TAD EDC locus in skin epithelial cells are cell type-specific and involve extensive contacts within TADs as well as between different gene-rich TADs, forming the framework for lineage-specific transcription.


Assuntos
Diferenciação Celular/genética , Cromatina/genética , DNA Helicases/genética , Proteínas Nucleares/genética , Fosfoproteínas/genética , Proteínas Repressoras/genética , Fatores de Transcrição/genética , Animais , Fator de Ligação a CCCTC , Proteínas de Ciclo Celular , Montagem e Desmontagem da Cromatina/genética , Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Epiderme/metabolismo , Epigênese Genética , Genoma , Queratinócitos , Camundongos , Regiões Promotoras Genéticas , Pele/metabolismo
10.
BMC Biol ; 17(1): 104, 2019 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-31830989

RESUMO

BACKGROUND: Polarity is necessary for epithelial cells to perform distinct functions at their apical and basal surfaces. Oral epithelial cell-derived ameloblasts at secretory stage (SABs) synthesize large amounts of enamel matrix proteins (EMPs), largely amelogenins. EMPs are unidirectionally secreted into the enamel space through their apical cytoplasmic protrusions, or Tomes' processes (TPs), to guide the enamel formation. Little is known about the transcriptional regulation underlying the establishment of cell polarity and unidirectional secretion of SABs. RESULTS: The higher-order chromatin architecture of eukaryotic genome plays important roles in cell- and stage-specific transcriptional programming. A genome organizer, special AT-rich sequence-binding protein 1 (SATB1), was discovered to be significantly upregulated in ameloblasts compared to oral epithelial cells using a whole-transcript microarray analysis. The Satb1-/- mice possessed deformed ameloblasts and a thin layer of hypomineralized and non-prismatic enamel. Remarkably, Satb1-/- ameloblasts at the secretory stage lost many morphological characteristics found at the apical surface of wild-type (wt) SABs, including the loss of Tomes' processes, defective inter-ameloblastic adhesion, and filamentous actin architecture. As expected, the secretory function of Satb1-/- SABs was compromised as amelogenins were largely retained in cells. We found the expression of epidermal growth factor receptor pathway substrate 8 (Eps8), a known regulator for actin filament assembly and small intestinal epithelial cytoplasmic protrusion formation, to be SATB1 dependent. In contrast to wt SABs, EPS8 could not be detected at the apical surface of Satb1-/- SABs. Eps8 expression was greatly reduced in small intestinal epithelial cells in Satb1-/- mice as well, displaying defective intestinal microvilli. CONCLUSIONS: Our data show that SATB1 is essential for establishing secretory ameloblast cell polarity and for EMP secretion. In line with the deformed apical architecture, amelogenin transport to the apical secretory front and secretion into enamel space were impeded in Satb1-/- SABs resulting in a massive cytoplasmic accumulation of amelogenins and a thin layer of hypomineralized enamel. Our studies strongly suggest that SATB1-dependent Eps8 expression plays a critical role in cytoplasmic protrusion formation in both SABs and in small intestines. This study demonstrates the role of SATB1 in the regulation of amelogenesis and the potential application of SATB1 in ameloblast/enamel regeneration.


Assuntos
Ameloblastos/fisiologia , Amelogênese , Polaridade Celular , Esmalte Dentário/crescimento & desenvolvimento , Proteínas de Ligação à Região de Interação com a Matriz/genética , Animais , Diferenciação Celular , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Camundongos
11.
J Immunol ; 196(2): 563-72, 2016 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-26667169

RESUMO

Special AT-rich sequence binding protein 1 (SATB1) is a genome organizer that is expressed by T cells. T cell development is severely impaired in SATB1 null mice; however, because SATB1 null mice die by 3 wk of age, the roles of SATB1 in T cell development have not been well clarified. In this study, we generated and analyzed SATB1 conditional knockout (cKO) mice, in which the SATB1 gene was deleted from all hematopoietic cells. T cell numbers were reduced in these mice, mainly because of a deficiency in positive selection at the CD4(+)CD8(+) double-positive stage during T cell development in the thymus. We also found that SATB1 cKO mice developed autoimmune diseases within 16 wk after birth. In SATB1 cKO mice, the numbers of Foxp3(+) regulatory T (Treg) cells were significantly reduced at 2 wk of age compared with wild-type littermates. Although the numbers gradually increased upon aging, Treg cells in SATB1 cKO mice were still less than those in wild-type littermates at adulthood. Suppressive functions of Treg cells, which play a major role in establishment of peripheral tolerance, were also affected in the absence of SATB1. In addition, negative selection during T cell development in the thymus was severely impaired in SATB1 deficient mice. These results suggest that SATB1 plays an essential role in establishment of immune tolerance.


Assuntos
Tolerância Imunológica/imunologia , Proteínas de Ligação à Região de Interação com a Matriz/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia , Animais , Diferenciação Celular/imunologia , Citometria de Fluxo , Imuno-Histoquímica , Proteínas de Ligação à Região de Interação com a Matriz/deficiência , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , Timo/crescimento & desenvolvimento , Timo/imunologia
12.
Genes Dev ; 23(22): 2625-38, 2009 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-19933152

RESUMO

Satb1 and the closely related Satb2 proteins regulate gene expression and higher-order chromatin structure of multigene clusters in vivo. In examining the role of Satb proteins in murine embryonic stem (ES) cells, we find that Satb1(-/-) cells display an impaired differentiation potential and augmented expression of the pluripotency determinants Nanog, Klf4, and Tbx3. Metastable states of self-renewal and differentiation competence have been attributed to heterogeneity of ES cells in the expression of Nanog. Satb1(-/-) cultures have a higher proportion of Nanog(high) cells, and an increased potential to reprogram human B lymphocytes in cell fusion experiments. Moreover, Satb1-deficient ES cells show an increased expression of Satb2, and we find that forced Satb2 expression in wild-type ES cells antagonizes differentiation-associated silencing of Nanog and enhances the induction of NANOG in cell fusions with human B lymphocytes. An antagonistic function of Satb1 and Satb2 is also supported by the almost normal differentiation potential of Satb1(-/-)Satb2(-/-) ES cells. Taken together with the finding that both Satb1 and Satb2 bind the Nanog locus in vivo, our data suggest that the balance of Satb1 and Satb2 contributes to the plasticity of Nanog expression and ES cell pluripotency.


Assuntos
Diferenciação Celular , Células-Tronco Embrionárias/citologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linfócitos B/metabolismo , Linhagem Celular , Humanos , Fator 4 Semelhante a Kruppel , Camundongos , Proteína Homeobox Nanog
14.
Proc Natl Acad Sci U S A ; 109(47): 19071-8, 2012 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-23144223

RESUMO

Neurons within each layer in the mammalian cortex have stereotypic projections. Four genes-Fezf2, Ctip2, Tbr1, and Satb2-regulate these projection identities. These genes also interact with each other, and it is unclear how these interactions shape the final projection identity. Here we show, by generating double mutants of Fezf2, Ctip2, and Satb2, that cortical neurons deploy a complex genetic switch that uses mutual repression to produce subcortical or callosal projections. We discovered that Tbr1, EphA4, and Unc5H3 are critical downstream targets of Satb2 in callosal fate specification. This represents a unique role for Tbr1, implicated previously in specifying corticothalamic projections. We further show that Tbr1 expression is dually regulated by Satb2 and Ctip2 in layers 2-5. Finally, we show that Satb2 and Fezf2 regulate two disease-related genes, Auts2 (Autistic Susceptibility Gene2) and Bhlhb5 (mutated in Hereditary Spastic Paraplegia), providing a molecular handle to investigate circuit disorders in neurodevelopmental diseases.


Assuntos
Redes Reguladoras de Genes , Neocórtex/crescimento & desenvolvimento , Neocórtex/metabolismo , Neurônios/metabolismo , Proteínas Repressoras/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Axônios/enzimologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Moléculas de Adesão Celular Neuronais/metabolismo , Córtex Cerebral/metabolismo , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteínas Ligadas por GPI/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Loci Gênicos/genética , Isoenzimas/metabolismo , Camundongos , Mutação/genética , Proteínas do Tecido Nervoso/metabolismo , Receptores de Netrina , Proteínas Nucleares/metabolismo , Ligação Proteica , Receptor EphA4/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Repressoras/genética , Proteínas com Domínio T , Tálamo/metabolismo , Fatores de Transcrição , Proteínas Supressoras de Tumor/metabolismo
15.
Nat Genet ; 38(11): 1278-88, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17057718

RESUMO

SATB1 (special AT-rich sequence binding protein 1) organizes cell type-specific nuclear architecture by anchoring specialized DNA sequences and recruiting chromatin remodeling factors to control gene transcription. We studied the role of SATB1 in regulating the coordinated expression of Il5, Il4 and Il13, located in the 200-kb T-helper 2 (T(H)2) cytokine locus on mouse chromosome 11. We show that on T(H)2 cell activation, SATB1 expression is rapidly induced to form a unique transcriptionally active chromatin structure at the cytokine locus. In this structure, chromatin is folded into numerous small loops, all anchored to SATB1 at their base. In addition, histone H3 is acetylated at Lys9 and Lys14, and the T(H)2-specific factors GATA3, STAT6 and c-Maf, the chromatin-remodeling enzyme Brg1 and RNA polymerase II are all bound across the 200-kb region. Before activation, the T(H)2 cytokine locus is already associated with GATA3 and STAT6, showing some looping, but these are insufficient to induce cytokine gene expression. Using RNA interference, we show that on cell activation, SATB1 is required not only for compacting chromatin into dense loops at the 200-kb cytokine locus but also for inducing Il4, Il5, Il13 and c-Maf expression. Thus, SATB1 is a necessary determinant for the hitherto unidentified higher-order, transcriptionally active chromatin structure that forms on T(H)2 cell activation.


Assuntos
Cromatina/química , Cromatina/metabolismo , Citocinas/metabolismo , Proteínas de Ligação à Região de Interação com a Matriz/fisiologia , Transcrição Gênica/fisiologia , Acetilação , Hidrolases Anidrido Ácido , Animais , Sítios de Ligação , Imunoprecipitação da Cromatina/métodos , Cromossomos/química , DNA Helicases/metabolismo , Enzimas Reparadoras do DNA/genética , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Fator de Transcrição GATA3/metabolismo , Regulação da Expressão Gênica , Histona Acetiltransferases/metabolismo , Histonas/metabolismo , Região de Controle de Locus Gênico , Ativação Linfocitária/fisiologia , Camundongos , Camundongos Endogâmicos AKR , Modelos Biológicos , Proteínas Nucleares/metabolismo , Conformação de Ácido Nucleico , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-maf/metabolismo , RNA Polimerase II/metabolismo , Fator de Transcrição STAT6/metabolismo , Células Th2/metabolismo , Fatores de Transcrição/metabolismo
16.
Semin Cancer Biol ; 23(2): 72-9, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22771615

RESUMO

When cells change functions or activities (such as during differentiation, response to extracellular stimuli, or migration), gene expression undergoes large-scale reprogramming, in cell type- and function-specific manners. Large changes in gene regulation require changes in chromatin architecture, which involve recruitment of chromatin remodeling enzymes and epigenomic modification enzymes to specific genomic loci. Transcription factors must also be accurately assembled at these loci. SATB1 is a genome organizer protein that facilitates these processes, providing a nuclear architectural platform that anchors hundreds of genes, through its interaction with specific genomic sequences; this activity allows expression of all these genes to be regulated in parallel, and enables cells to thereby alter their function. We review and describe future perspectives on SATB1 function in higher-order chromatin structure and gene regulation, and its role in metastasis of breast cancer and other tumor types.


Assuntos
Montagem e Desmontagem da Cromatina/genética , Proteínas de Ligação à Região de Interação com a Matriz/fisiologia , Neoplasias/genética , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Cromatina/química , Cromatina/metabolismo , Progressão da Doença , Epigênese Genética/genética , Feminino , Genoma Humano/fisiologia , Humanos , Proteínas de Ligação à Região de Interação com a Matriz/genética , Modelos Biológicos
17.
Nature ; 452(7184): 187-93, 2008 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-18337816

RESUMO

Mechanisms underlying global changes in gene expression during tumour progression are poorly understood. SATB1 is a genome organizer that tethers multiple genomic loci and recruits chromatin-remodelling enzymes to regulate chromatin structure and gene expression. Here we show that SATB1 is expressed by aggressive breast cancer cells and its expression level has high prognostic significance (P < 0.0001), independent of lymph-node status. RNA-interference-mediated knockdown of SATB1 in highly aggressive (MDA-MB-231) cancer cells altered the expression of >1,000 genes, reversing tumorigenesis by restoring breast-like acinar polarity and inhibiting tumour growth and metastasis in vivo. Conversely, ectopic SATB1 expression in non-aggressive (SKBR3) cells led to gene expression patterns consistent with aggressive-tumour phenotypes, acquiring metastatic activity in vivo. SATB1 delineates specific epigenetic modifications at target gene loci, directly upregulating metastasis-associated genes while downregulating tumour-suppressor genes. SATB1 reprogrammes chromatin organization and the transcription profiles of breast tumours to promote growth and metastasis; this is a new mechanism of tumour progression.


Assuntos
Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica/genética , Proteínas de Ligação à Região de Interação com a Matriz/metabolismo , Metástase Neoplásica/genética , Animais , Biomarcadores Tumorais/análise , Neoplasias da Mama/diagnóstico , Linhagem Celular , Linhagem Celular Tumoral , Polaridade Celular , Progressão da Doença , Epigênese Genética/genética , Perfilação da Expressão Gênica , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metástase Linfática/diagnóstico , Metástase Linfática/genética , Metástase Linfática/patologia , Proteínas de Ligação à Região de Interação com a Matriz/deficiência , Proteínas de Ligação à Região de Interação com a Matriz/genética , Camundongos , Camundongos Nus , Metástase Neoplásica/diagnóstico , Metástase Neoplásica/patologia , Transplante de Neoplasias , Fenótipo , Prognóstico , Interferência de RNA
18.
Nat Genet ; 37(1): 31-40, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15608638

RESUMO

Mutations in MECP2 are associated with Rett syndrome, an X-linked neurodevelopmental disorder. To identify genes targeted by Mecp2, we sequenced 100 in vivo Mecp2-binding sites in mouse brain. Several sequences mapped to an imprinted gene cluster on chromosome 6, including Dlx5 and Dlx6, whose transcription was roughly two times greater in brains of Mecp2-null mice compared with those of wild-type mice. The maternally expressed gene DLX5 showed a loss of imprinting in lymphoblastoid cells from individuals with Rett syndrome. Because Dlx5 regulates production of enzymes that synthesize gamma-aminobutyric acid (GABA), loss of imprinting of Dlx5 may alter GABAergic neuron activity in individuals with Rett syndrome. In mouse brain, Dlx5 imprinting was relaxed, yet Mecp2-mediated silent-chromatin structure existed at the Dlx5-Dlx6 locus in brains of wild-type, but not Mecp2-null, mice. Mecp2 targeted histone deacetylase 1 to a sharply defined, approximately 1-kb region at the Dlx5-Dlx6 locus and promoted repressive histone methylation at Lys9 at this site. Chromatin immunoprecipitation-combined loop assays showed that Mecp2 mediated the silent chromatin-derived 11-kb chromatin loop at the Dlx5-Dlx6 locus. This loop was absent in chromatin of brains of Mecp2-null mice, and Dlx5-Dlx6 interacted with far distant sequences, forming distinct active chromatin-associated loops. These results show that formation of a silent-chromatin loop is a new mechanism underlying gene regulation by Mecp2.


Assuntos
Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/genética , Impressão Genômica , Proteínas de Homeodomínio/genética , Proteínas Repressoras/genética , Síndrome de Rett/genética , Animais , Cromatina/genética , Ilhas de CpG/fisiologia , Metilação de DNA , Perfilação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Humanos , Proteína 2 de Ligação a Metil-CpG , Camundongos , Família Multigênica , Neurônios/metabolismo , Testes de Precipitina , Fatores de Transcrição , Ácido gama-Aminobutírico/metabolismo
20.
Methods ; 58(3): 243-54, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22782115

RESUMO

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.


Assuntos
Cromatina/genética , Proteínas de Ligação à Região de Interação com a Matriz/fisiologia , Animais , Sequência de Bases , Sítios de Ligação , Imunoprecipitação da Cromatina/métodos , Mapeamento Cromossômico/métodos , Reagentes de Ligações Cruzadas/química , Formaldeído/química , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Conformação de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA