Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Nat Commun ; 13(1): 1859, 2022 04 06.
Artigo em Inglês | MEDLINE | ID: mdl-35388001

RESUMO

The cohesin complex participates in the organization of 3D genome through generating and maintaining DNA loops. Stromal antigen 2 (STAG2), a core subunit of the cohesin complex, is frequently mutated in various cancers. However, the impact of STAG2 inactivation on 3D genome organization, especially the long-range enhancer-promoter contacts and subsequent gene expression control in cancer, remains poorly understood. Here we show that depletion of STAG2 in melanoma cells leads to expansion of topologically associating domains (TADs) and enhances the formation of acetylated histone H3 lysine 27 (H3K27ac)-associated DNA loops at sites where binding of STAG2 is switched to its paralog STAG1. We further identify Interferon Regulatory Factor 9 (IRF9) as a major direct target of STAG2 in melanoma cells via integrated RNA-seq, STAG2 ChIP-seq and H3K27ac HiChIP analyses. We demonstrate that loss of STAG2 activates IRF9 through modulating the 3D genome organization, which in turn enhances type I interferon signaling and increases the expression of PD-L1. Our findings not only establish a previously unknown role of the STAG2 to STAG1 switch in 3D genome organization, but also reveal a functional link between STAG2 and interferon signaling in cancer cells, which may enhance the immune evasion potential in STAG2-mutant cancer.


Assuntos
Proteínas Cromossômicas não Histona , Melanoma , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas Cromossômicas não Histona/metabolismo , Genoma , Humanos , Interferons/genética , Melanoma/genética
2.
Mol Cancer Res ; 19(3): 507-515, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33234577

RESUMO

Hematopoietic-derived cells are integral components of the tumor microenvironment and serve as critical mediators of tumor-host interactions. Host cells derived from myeloid and lymphoid lineages perform well-established functions linked to cancer development, progression, and response to therapy. It is unclear whether host erythroid cells also contribute to shaping the path that cancer can take, but emerging evidence points to this possibility. Here, we show that tumor-promoting environmental stress and tumor-induced hemodynamic changes trigger renal erythropoietin production and erythropoietin-dependent expansion of splenic erythroid cell populations in mice. These erythroid cells display molecular features indicative of an immature erythroid phenotype, such as the expression of both CD71 and TER119 and the retention of intact nuclei, and express genes encoding immune checkpoint molecules. Nucleated erythroid cells with similar properties are present in mouse and human tumor tissues. Antibody-mediated erythropoietin blockade reduces tumor-responsive erythroid cell induction and tumor growth. These findings reveal the potential of tumor-induced erythropoietin and erythroid cells as targets for cancer treatment. IMPLICATIONS: : Our study identifies erythropoietin and erythroid cells as novel players in tumor-host interactions and highlights the involvement of multiorgan signaling events in their induction in response to environmental stress and tumor growth.


Assuntos
Células Eritroides/metabolismo , Proteínas de Checkpoint Imunológico/metabolismo , Animais , Diferenciação Celular , Humanos , Camundongos , Transdução de Sinais
3.
Curr Opin Hematol ; 24(4): 345-352, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28463873

RESUMO

PURPOSE OF REVIEW: Loss of IKAROS in committed B cell precursors causes a block in differentiation while at the same time augments aberrant cellular properties, such as bone marrow stromal adhesion, self-renewal and resistance to glucocorticoid-mediated cell death. B cell acute lymphoblastic leukaemias originating from these early stages of B cell differentiation and associated with IKAROS mutations share a high-risk cellular phenotype suggesting that deregulation of IKAROS-based mechanisms cause a highly malignant disease process. RECENT STUDIES: Recent studies show that IKAROS is critical for the activity of super-enhancers at genes required for pre-B cell receptor (BCR) signalling and differentiation, working either downstream of or in parallel with B cell master regulators such as EBF1 and PAX5. IKAROS also directly represses a cryptic regulatory network of transcription factors prevalent in mesenchymal and epithelial precursors that includes YAP1, TEAD1/2, LHX2 and LMO2, and their targets, which are not normally expressed in lymphocytes. IKAROS prevents not only expression of these 'extra-lineage' transcription factors but also their cooperation with endogenous B cell master regulators, such as EBF1 and PAX5, leading to the formation of a de novo for lymphocytes super-enhancer network. IKAROS coordinates with the Polycomb repression complex (PRC2) to provide stable repression of associated genes during B cell development. However, induction of regulatory factors normally repressed by IKAROS starts a feed-forward loop that activates de-novo enhancers and elevates them to super-enhancer status, thereby diminishing PRC2 repression and awakening aberrant epithelial-like cell properties in B cell precursors. SUMMARY: Insight into IKAROS-based transcriptional circuits not only sets new paradigms for cell differentiation but also provides new approaches for classifying and treating high-risk human B-ALL that originates from these early stages of B cell differentiation.


Assuntos
Linfócitos B/metabolismo , Transformação Celular Neoplásica/genética , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Transcrição Gênica , Animais , Linfócitos B/patologia , Diferenciação Celular/genética , Proliferação de Células/genética , Autorrenovação Celular/genética , Elementos Facilitadores Genéticos , Humanos , Fator de Transcrição Ikaros/metabolismo , Proteínas do Grupo Polycomb/metabolismo , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos B/metabolismo , Ligação Proteica
4.
Genes Dev ; 30(17): 1971-90, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27664237

RESUMO

IKAROS is required for the differentiation of highly proliferative pre-B-cell precursors, and loss of IKAROS function indicates poor prognosis in precursor B-cell acute lymphoblastic leukemia (B-ALL). Here we show that IKAROS regulates this developmental stage by positive and negative regulation of superenhancers with distinct lineage affiliations. IKAROS defines superenhancers at pre-B-cell differentiation genes together with B-cell master regulators such as PAX5, EBF1, and IRF4 but is required for a highly permissive chromatin environment, a function that cannot be compensated for by the other transcription factors. IKAROS is also highly enriched at inactive enhancers of genes normally expressed in stem-epithelial cells. Upon IKAROS loss, expression of pre-B-cell differentiation genes is attenuated, while a group of extralineage transcription factors that are directly repressed by IKAROS and depend on EBF1 relocalization at their enhancers for expression is induced. LHX2, LMO2, and TEAD-YAP1, normally kept separate from native B-cell transcription regulators by IKAROS, now cooperate directly with them in a de novo superenhancer network with its own feed-forward transcriptional reinforcement. Induction of de novo superenhancers antagonizes Polycomb repression and superimposes aberrant stem-epithelial cell properties in a B-cell precursor. This dual mechanism of IKAROS regulation promotes differentiation while safeguarding against a hybrid stem-epithelial-B-cell phenotype that underlies high-risk B-ALL.


Assuntos
Diferenciação Celular/genética , Elementos Facilitadores Genéticos/fisiologia , Células Epiteliais/citologia , Regulação Leucêmica da Expressão Gênica , Fator de Transcrição Ikaros/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/fisiopatologia , Células Precursoras de Linfócitos B/citologia , Animais , Epigênese Genética , Células Epiteliais/patologia , Fator de Transcrição Ikaros/genética , Camundongos , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Células Precursoras de Linfócitos B/patologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
5.
Int J Hematol ; 100(3): 220-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25085254

RESUMO

The Ikaros family of DNA-binding proteins are critical regulators of lymphocyte differentiation. In multipotent, hematopoietic progenitors, Ikaros supports transcriptional priming of genes promoting lymphocyte differentiation. Ikaros targets the Nucleosome Remodeling Deacetylase (NuRD) complex to lymphoid lineage genes, thereby increasing chromatin accessibility and transcriptional priming. After lymphoid lineage specification, Ikaros expression is raised to levels characteristic of intermediate B cell and T cell precursors, which is necessary to support maturation and prevent leukemogenesis. Loss of Ikaros in T cell precursors allows the NuRD complex to repress lymphocyte genes and extends its targeting to genes that support growth and proliferation, causing their activation and triggering a cascade of events that leads to leukemogenesis. Loss of Ikaros in B cell precursors blocks differentiation and perpetuates stromal adhesion by enhancing integrin signaling. The combination of integrin and cytokine signaling in Ikaros-deficient pre-B cells promotes their survival and self-renewal. The stages of lymphocyte differentiation that are highly dependent on Ikaros are underscored by changes in Ikaros transcription, supported by a complex network of stage-specific regulatory networks that converge upon the Ikzf1 locus. It is increasingly apparent that understanding the regulatory networks that operate upstream and downstream of Ikaros is critical not only for our understanding of normal lymphopoiesis, but also in placing the right finger on the mechanisms that support hematopoietic malignancies in mouse and human.


Assuntos
Epigênese Genética/imunologia , Fator de Transcrição Ikaros/genética , Linfopoese/genética , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos T/imunologia , Animais , Autoantígenos/genética , Autoantígenos/imunologia , Diferenciação Celular , Linhagem da Célula/imunologia , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Fator de Transcrição Ikaros/imunologia , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/imunologia , Camundongos , Células Precursoras de Linfócitos B/citologia , Células Precursoras de Linfócitos T/citologia , Isoformas de Proteínas/genética , Isoformas de Proteínas/imunologia , Transdução de Sinais
6.
Neuron ; 83(1): 122-34, 2014 Jul 02.
Artigo em Inglês | MEDLINE | ID: mdl-24991957

RESUMO

Precise control of gene expression plays fundamental roles in brain development, but the roles of chromatin regulators in neuronal connectivity have remained poorly understood. We report that depletion of the NuRD complex by in vivo RNAi and conditional knockout of the core NuRD subunit Chd4 profoundly impairs the establishment of granule neuron parallel fiber/Purkinje cell synapses in the rodent cerebellar cortex in vivo. By interfacing genome-wide sequencing of transcripts and ChIP-seq analyses, we uncover a network of repressed genes and distinct histone modifications at target gene promoters that are developmentally regulated by the NuRD complex in the cerebellum in vivo. Finally, in a targeted in vivo RNAi screen of NuRD target genes, we identify a program of NuRD-repressed genes that operate as critical regulators of presynaptic differentiation in the cerebellar cortex. Our findings define NuRD-dependent promoter decommissioning as a developmentally regulated programming mechanism that drives synaptic connectivity in the mammalian brain.


Assuntos
Química Encefálica/genética , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/genética , Regiões Promotoras Genéticas/genética , Sinapses/genética , Animais , Animais Recém-Nascidos , Células Cultivadas , Montagem e Desmontagem da Cromatina/genética , Proteínas de Ligação a DNA/genética , Humanos , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Células de Purkinje/fisiologia , Células de Purkinje/ultraestrutura , Ratos , Ratos Long-Evans , Ratos Sprague-Dawley , Proteína 4 de Ligação ao Retinoblastoma/genética , Sinapses/ultraestrutura
7.
Anticancer Res ; 34(3): 1251-3, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24596368

RESUMO

BACKGROUND/AIM: Survivin is expressed in the nucleus and/or cytoplasm of various types of malignant tumor cells. Nuclear survivin is indispensable for complete mitosis, while cytoplasmic survivin functions as an apoptosis inhibitor. We examined the difference in the survivin expression among stromal cells of fibroadenoma, and benign and malignant phyllodes tumors. MATERIALS AND METHODS: Tumor sections were immunohistochemically stained with an anti-human survivin antibody and the labeling index of survivin was calculated. RESULTS: In stromal cells of all tumors, survivin was expressed in the nuclei but not in the cytoplasm. The labeling indices of the stromal cells in five malignant phyllodes tumors (20.5±3.0) were significantly greater than those observed in eight fibroadenomas (1.9±0.6) or nine benign phyllodes tumors (3.0±0.9). CONCLUSION: In the present study it was shown that stromal cells in malignant phyllodes tumors express nuclear survivin more extensively than stromal cells in benign phyllodes tumors or fibroadenomas.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Núcleo Celular/metabolismo , Fibroadenoma/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Tumor Filoide/metabolismo , Células Estromais/metabolismo , Neoplasias da Mama/patologia , Citoplasma/metabolismo , Feminino , Fibroadenoma/patologia , Seguimentos , Humanos , Técnicas Imunoenzimáticas , Estadiamento de Neoplasias , Tumor Filoide/patologia , Prognóstico , Células Estromais/patologia , Survivina
8.
Nat Immunol ; 15(3): 294-304, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24509510

RESUMO

Deletion of the DNA-binding domain of the transcription factor Ikaros generates dominant-negative isoforms that interfere with its activity and correlate with poor prognosis in human precursor B cell acute lymphoblastic leukemia (B-ALL). Here we found that conditional inactivation of the Ikaros DNA-binding domain in early pre-B cells arrested their differentiation at a stage at which integrin-dependent adhesion to niches augmented signaling via mitogen-activated protein kinases, proliferation and self-renewal and attenuated signaling via the pre-B cell signaling complex (pre-BCR) and the differentiation of pre-B cells. Transplantation of polyclonal Ikaros-mutant pre-B cells resulted in long-latency oligoclonal pre-B-ALL, which demonstrates that loss of Ikaros contributes to multistep B cell leukemogenesis. Our results explain how normal pre-B cells transit from a highly proliferative and stroma-dependent phase to a stroma-independent phase during which differentiation is enabled, and suggest potential therapeutic strategies for Ikaros-mutant B-ALL.


Assuntos
Diferenciação Celular/imunologia , Proliferação de Células , Fator de Transcrição Ikaros/imunologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia , Células Precursoras de Linfócitos B/patologia , Transferência Adotiva , Animais , Apoptose , Separação Celular , Sobrevivência Celular , Proteínas de Ligação a DNA/imunologia , Proteínas de Ligação a DNA/metabolismo , Progressão da Doença , Citometria de Fluxo , Imunofluorescência , Fator de Transcrição Ikaros/metabolismo , Immunoblotting , Camundongos , Camundongos Transgênicos , Leucemia-Linfoma Linfoblástico de Células Precursoras/imunologia , Células Precursoras de Linfócitos B/imunologia , Células Precursoras de Linfócitos B/metabolismo
9.
Blood ; 122(18): 3149-59, 2013 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-24002445

RESUMO

Ikaros is a critical regulator of lymphocyte development and homeostasis; thus, understanding its transcriptional regulation is important from both developmental and clinical perspectives. Using a mouse transgenic reporter approach, we functionally characterized a network of highly conserved cis-acting elements at the Ikzf1 locus. We attribute B-cell and myeloid but not T-cell specificity to the main Ikzf1 promoter. Although this promoter was unable to counter local chromatin silencing effects, each of the 6 highly conserved Ikzf1 intronic enhancers alleviated silencing. Working together, the Ikzf1 enhancers provided locus control region activity, allowing reporter expression in a position and copy-independent manner. Only 1 of the Ikzf1 enhancers was responsible for the progressive upregulation of Ikaros expression from hematopoietic stem cells to lymphoid-primed multipotent progenitors to T-cell precursors, which are stages of differentiation dependent on Ikaros for normal outcome. Thus, Ikzf1 is regulated by both epigenetic and transcriptional factors that target its enhancers in both redundant and specific fashions to provide an expression profile supportive of normal lymphoid lineage progression and homeostasis. Mutations in the Ikzf1 regulatory elements and their interacting factors are likely to have adverse effects on lymphopoiesis and contribute to leukemogenesis.


Assuntos
Elementos Facilitadores Genéticos/genética , Fator de Transcrição Ikaros/genética , Sequências Reguladoras de Ácido Nucleico/genética , Ativação Transcricional , Animais , Linfócitos B/metabolismo , Sequência de Bases , Sítios de Ligação/genética , Encéfalo/metabolismo , Epigênese Genética , Citometria de Fluxo , Redes Reguladoras de Genes , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Fator de Transcrição Ikaros/metabolismo , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Modelos Genéticos , Dados de Sequência Molecular , Células Mieloides/metabolismo , Homologia de Sequência de Aminoácidos , Linfócitos T/metabolismo , Fatores de Transcrição/metabolismo
11.
Gan To Kagaku Ryoho ; 40(12): 2470-2, 2013 Nov.
Artigo em Japonês | MEDLINE | ID: mdl-24394147

RESUMO

A 67-year-old man with bladder cancer who was treated with transurethral resection of bladder tumour(TUR-Bt)and chemotherapy at the age of 59 years was diagnosed as having urothelial cancer by biopsy 8 years later. Detailed examination revealed the presence of synchronous triple cancer, with hepatocellular cancer and gastric cancer. Subsequently, semi-total gastrectomy, partial hepatectomy(S6), radio frequency ablation(S5, S7), and cholecystectomy were performed. Histologically, the gastric tumor was a moderately differentiated tubular adenocarcinoma, the hepatic tumor was a moderately differentiated hepatocellular carcinoma, the bladder tumor was a transitional cell carcinoma, and the ureteral tumor was an urothelial carcinoma.


Assuntos
Neoplasias Hepáticas/patologia , Neoplasias Primárias Múltiplas/patologia , Neoplasias Gástricas/patologia , Neoplasias da Bexiga Urinária/patologia , Neoplasias Urológicas/patologia , Idoso , Antimetabólitos Antineoplásicos/uso terapêutico , Quimioterapia Adjuvante , Combinação de Medicamentos , Humanos , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/cirurgia , Masculino , Neoplasias Primárias Múltiplas/tratamento farmacológico , Neoplasias Primárias Múltiplas/cirurgia , Ácido Oxônico/uso terapêutico , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/cirurgia , Tegafur/uso terapêutico , Resultado do Tratamento , Neoplasias da Bexiga Urinária/tratamento farmacológico , Neoplasias da Bexiga Urinária/cirurgia , Neoplasias Urológicas/tratamento farmacológico , Neoplasias Urológicas/cirurgia
12.
Med Mol Morphol ; 45(3): 173-7, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23001300

RESUMO

Extra-gastrointestinal stromal tumors (E-GISTs) not associated with the alimentary tract in the pelvic cavity are extremely rare. We treated a 49-year-old Japanese man with such an E-GIST in the pelvic cavity who underwent an intrapelvic tumorectomy with a total prostatectomy and partial rectum resection. Gross examination of the specimen revealed an 8.1 × 5 × 4 cm white-grayish mass. Histological findings showed uniform spindle cells with scant atypia that formed interlacing bundles or whorl patterns. These neoplastic cells did not invade adjacent organs, including the gut. Immunohistochemical findings revealed that the neoplastic cells were positive for c-kit, CD34, and vimentin. Molecular analysis showed a c-kit mutation at exon 9 with duplication of Ala and Tyr. Our diagnosis was E-GIST, which belongs to the intermediate group of GIST. Following the operation, we administered imatinib mesylate for 6 months. After stopping for 5 months, it was administered again for local recurrence. We are planning our future strategy for this case including surgical resection as necessary.


Assuntos
Pelve , Antígenos CD34/genética , Antígenos CD34/metabolismo , Antineoplásicos/uso terapêutico , Benzamidas , Éxons/genética , Tumores do Estroma Gastrointestinal/diagnóstico , Tumores do Estroma Gastrointestinal/tratamento farmacológico , Tumores do Estroma Gastrointestinal/patologia , Tumores do Estroma Gastrointestinal/cirurgia , Humanos , Mesilato de Imatinib , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Mutação , Pelve/patologia , Pelve/cirurgia , Piperazinas/uso terapêutico , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Pirimidinas/uso terapêutico , Vimentina/genética , Vimentina/metabolismo
13.
Nat Immunol ; 13(1): 86-94, 2011 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-22080921

RESUMO

Cell fate depends on the interplay between chromatin regulators and transcription factors. Here we show that activity of the Mi-2ß nucleosome-remodeling and histone-deacetylase (NuRD) complex was controlled by the Ikaros family of lymphoid lineage-determining proteins. Ikaros, an integral component of the NuRD complex in lymphocytes, tethered this complex to active genes encoding molecules involved in lymphoid differentiation. Loss of Ikaros DNA-binding activity caused a local increase in chromatin remodeling and histone deacetylation and suppression of lymphoid cell-specific gene expression. Without Ikaros, the NuRD complex also redistributed to transcriptionally poised genes that were not targets of Ikaros (encoding molecules involved in proliferation and metabolism), which induced their reactivation. Thus, release of NuRD from Ikaros regulation blocks lymphocyte maturation and mediates progression to a leukemic state by engaging functionally opposing epigenetic and genetic networks.


Assuntos
Linfócitos/enzimologia , Complexo Mi-2 de Remodelação de Nucleossomo e Desacetilase/metabolismo , Animais , Sequência de Bases , Diferenciação Celular/genética , Montagem e Desmontagem da Cromatina , Perfilação da Expressão Gênica , Redes Reguladoras de Genes , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Leucemia/genética , Linfócitos/imunologia , Camundongos , Motivos de Nucleotídeos , Ligação Proteica , Timócitos/metabolismo
14.
Curr Opin Immunol ; 22(2): 154-60, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20299195

RESUMO

Bioinformatic studies on a revised hierarchy of hematopoietic progenitors have provided a genome-wide view of lineage-affiliated transcriptional programs directing early hematopoiesis. Unexpectedly, lymphoid, myeloid, and erythroid gene expression programs were primed with similar frequency at the multipotent progenitor stage indicating a stochastic nature to this process. Multilineage transcriptional priming is quickly resolved upon erythroid lineage restriction with both lymphoid and myeloid transcriptional programs rapidly extinguished. However, expression of lymphoid and myeloid factors remains active past nominal lymphoid and myeloid lineage restrictions, revealing a common genetic network utilized by both pathways. Priming and resolution of multilineage potential is dependent on the activity of the DNA binding factor Ikaros. Ikaros primes the lymphoid transcriptional program in the HSC and represses the stem cell and other disparate transcriptional programs downstream of the HSC. Loss of Ikaros removes the lymphoid leg of the immune system and may confer aberrant self-renewing properties to myeloid progenitors.


Assuntos
Linhagem da Célula/genética , Fator de Transcrição Ikaros/imunologia , Linfopoese , Animais , Linhagem da Célula/imunologia , Biologia Computacional , Genoma , Humanos , Ativação Transcricional/imunologia
15.
Immunity ; 30(4): 493-507, 2009 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-19345118

RESUMO

The mechanisms regulating lineage potential during early hematopoiesis were investigated. First, a cascade of lineage-affiliated gene expression signatures, primed in hematopoietic stem cells (HSCs) and differentially propagated in lineage-restricted progenitors, was identified. Lymphoid transcripts were primed as early as the HSC, together with myeloid and erythroid transcripts. Although this multilineage priming was resolved upon subsequent lineage restrictions, an unexpected cosegregation of lymphoid and myeloid gene expression and potential past a nominal myeloid restriction point was identified. Finally, we demonstrated that whereas the zinc finger DNA-binding factor Ikaros was required for induction of lymphoid lineage priming in the HSC, it was also necessary for repression of genetic programs compatible with self-renewal and multipotency downstream of the HSC. Taken together, our studies provide new insight into the priming and restriction of lineage potentials during early hematopoiesis and identify Ikaros as a key bivalent regulator of this process.


Assuntos
Linhagem da Célula , Redes Reguladoras de Genes , Genoma , Células-Tronco Hematopoéticas/imunologia , Fator de Transcrição Ikaros/metabolismo , Linfócitos/imunologia , Animais , Regulação da Expressão Gênica , Hematopoese/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
16.
Genes Dev ; 22(9): 1174-89, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18451107

RESUMO

The ability of somatic stem cells to self-renew and differentiate into downstream lineages is dependent on specialized chromatin environments that keep stem cell-specific genes active and key differentiation factors repressed but poised for activation. The epigenetic factors that provide this type of regulation remain ill-defined. Here we provide the first evidence that the SNF2-like ATPase Mi-2beta of the Nucleosome Remodeling Deacetylase (NuRD) complex is required for maintenance of and multilineage differentiation in the early hematopoietic hierarchy. Shortly after conditional inactivation of Mi-2beta, there is an increase in cycling and a decrease in quiescence in an HSC (hematopoietic stem cell)-enriched bone marrow population. These cycling mutant cells readily differentiate into the erythroid lineage but not into the myeloid and lymphoid lineages. Together, these effects result in an initial expansion of mutant HSC and erythroid progenitors that are later depleted as more differentiated proerythroblasts accumulate at hematopoietic sites exhibiting features of erythroid leukemia. Examination of gene expression in the mutant HSC reveals changes in the expression of genes associated with self-renewal and lineage priming and a pivotal role of Mi-2beta in their regulation. Thus, Mi-2beta provides the hematopoietic system with immune cell capabilities as well as with an extensive regenerative capacity.


Assuntos
Adenosina Trifosfatases/metabolismo , Diferenciação Celular/fisiologia , Cromatina/metabolismo , Células-Tronco Hematopoéticas/metabolismo , Adenosina Trifosfatases/genética , Animais , Antígenos CD/análise , Antígenos CD34/análise , Apoptose , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Ciclo Celular , Diferenciação Celular/genética , Linhagem da Célula , Proliferação de Células , Células Cultivadas , DNA Helicases , Eritrócitos/citologia , Eritrócitos/metabolismo , Feminino , Citometria de Fluxo/métodos , Perfilação da Expressão Gênica , Células-Tronco Hematopoéticas/citologia , Linfócitos/citologia , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Células Mieloides/citologia , Células Mieloides/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos/métodos , Receptores da Transferrina/análise
17.
Surg Today ; 37(10): 901-4, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17879044

RESUMO

Hyperplastic polyps (HPs) of the stomach have been reported to be mostly benign. However, in rare cases, carcinomas have been found in HPs. We treated a 59-year-old Japanese male who underwent a total gastrectomy, and a gross examination of the resected stomach revealed a 4.8 x 3.8-cm polyp on the greater curvature of the antrum and multiple small polyps in the whole gastric mucosa. Histologically, the large polyp consisted mainly of hyperplastic foveolar epithelium, while the presence of variously colored lobules demonstrated a poorly differentiated adenocarcinoma mixed with signet-ring cell carcinoma. Hyperplastic polyps should therefore be carefully examined microscopically as a polypectomy specimen and in resected stomach specimens.


Assuntos
Adenocarcinoma/diagnóstico , Neoplasias Gástricas/diagnóstico , Adenocarcinoma/fisiopatologia , Adenocarcinoma/cirurgia , Carcinoma de Células em Anel de Sinete/patologia , Carcinoma de Células em Anel de Sinete/cirurgia , Gastrectomia , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias Gástricas/fisiopatologia , Neoplasias Gástricas/cirurgia
18.
Pathol Int ; 57(10): 698-702, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17803660

RESUMO

An 80-year-old woman with a pleomorphic carcinoma (PC) producing hCG was admitted to Nippon Steel Hirohata Hospital because of an abnormal shadow on CT seen during a follow-up examination after surgery for breast cancer. A right upper lobectomy was performed due to rapid growth of the shadow 3 months later. Macroscopically the tumor was a 4.8 x 4.0 cm well-circumscribed grayish-white mass. On histology the tumor consisted mostly of intermingled spindle and polygonal cells, while evidence of poorly differentiated adenocarcinoma was seen in a few areas. A diagnosis of PC was made due to hCG expression in approximately 20% of the spindle and polygonal cells on immunohistology. Six months after the operation metastasis to the liver and adrenal gland was seen on CT. The patient died due to metastases 1 year after the operation, even though the patient had been at stage 1B at the time of the operation and appropriate chemotherapy had been given. PC patients with immunohistochemical hCG expression have elevated risk of local recurrence and metastasis.


Assuntos
Adenocarcinoma/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinossarcoma/metabolismo , Gonadotropina Coriônica/metabolismo , Neoplasias Pulmonares/metabolismo , Adenocarcinoma/secundário , Adenocarcinoma/terapia , Idoso de 80 Anos ou mais , Carcinoma Pulmonar de Células não Pequenas/patologia , Carcinoma Pulmonar de Células não Pequenas/secundário , Carcinossarcoma/secundário , Carcinossarcoma/terapia , Terapia Combinada , Evolução Fatal , Feminino , Humanos , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , Radiografia Torácica , Tomografia Computadorizada por Raios X
19.
Curr Opin Immunol ; 19(2): 116-22, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17307348

RESUMO

Hematopoiesis is the developmental process by which all blood and immune cells are generated. A decade-old scheme has supported an early and complete separation of the erythro-myeloid from the lymphoid lineages. Recent advances have re-drawn this map, separating lymphoid and myeloid from erythroid programs early in development. Subsequently, the fate restriction of both the lympho-myeloid and the erythro-megakaryocyte progenitors is dependent on Ikaros and its associated chromatin regulators. Genetic studies of this family of nuclear factors are now providing unique insight into the functional molecular signatures that bestow plasticity to the hematopoietic stem cell and its early progeny.


Assuntos
Cromatina/genética , Células Precursoras Eritroides/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Hematopoese/genética , Fator de Transcrição Ikaros/fisiologia , Células-Tronco Multipotentes/metabolismo , Animais , Fator de Transcrição Ikaros/genética , Camundongos
20.
Nat Immunol ; 7(4): 382-91, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16518393

RESUMO

Ikaros is expressed in early hematopoietic progenitors and is required for lymphoid differentiation. In the absence of Ikaros, there is a lack of markers defining fate restriction along lympho-myeloid pathways, but it is unclear whether formation of specific progenitors or expression of their markers is affected. Here we use a reporter based on Ikaros regulatory elements to separate early progenitors in wild-type and Ikaros-null mice. We found previously undetected Ikaros-null lympho-myeloid progenitors lacking the receptor tyrosine kinase Flt3 that were capable of myeloid but not lymphoid differentiation. In contrast, lack of Ikaros in the common myeloid progenitor resulted in increased formation of erythro-megakaryocytes at the expense of myeloid progenitors. Using this approach, we identify previously unknown pivotal functions for Ikaros in distinct fate 'decisions' in the early hematopoietic hierarchy.


Assuntos
Diferenciação Celular/imunologia , Hematopoese/imunologia , Células-Tronco Hematopoéticas/imunologia , Fator de Transcrição Ikaros/imunologia , Animais , Cruzamentos Genéticos , Proteínas de Fluorescência Verde , Células-Tronco Hematopoéticas/citologia , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Células Mieloides/citologia , Células Mieloides/imunologia , Organismos Livres de Patógenos Específicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA