Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Orthop Surg Res ; 16(1): 313, 2021 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-33990210

RESUMO

BACKGROUND: Growing evidence has implicated core-binding factor beta (Cbfb) as a contributor to osteoblast differentiation, which plays a key role in fracture healing. Herein, we aimed to assess whether Cbfb affects osteoblast differentiation after fibula fracture. METHODS: Initially, we established a Cbfb conditional knockout mouse model for subsequent studies. Immunohistochemical staining was conducted to detect the expression of proliferating cell nuclear antigen (PCNA) and collagen II in the fracture end. Next, we isolated and cultured osteoblasts from specific Cbfb conditional knockout mice for BrdU analysis, alkaline phosphatase (ALP) staining, and von Kossa staining to detect osteoblast viability, differentiation, and mineralization, respectively. Western blot analysis and reverse transcription-quantitative polymerase chain reaction (RT-qPCR) were used to detect the expression of osteoblast differentiation-related genes. RESULTS: The Cbfb conditional knockout mice exhibited downregulated expression of PCNA and collagen II, reduced ALP activity, and mineralization, as well as diminished expression of osteoblast differentiation-related genes. Further, Cbfb knockout exerted no obvious effects on osteoblast proliferation. CONCLUSIONS: Overall, these results substantiated that Cbfb could promote fibula fracture healing and osteoblast differentiation and thus provided a promising therapeutic target for clinical treatment of fibula fracture.


Assuntos
Diferenciação Celular/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Fíbula/lesões , Consolidação da Fratura/fisiologia , Osteoblastos/metabolismo , Animais , Proliferação de Células/fisiologia , Modelos Animais de Doenças , Camundongos
2.
Blood ; 137(19): 2579-2584, 2021 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-33821975

RESUMO

The core binding factor composed of CBFß and RUNX subunits plays a critical role in most hematopoietic lineages and is deregulated in acute myeloid leukemia (AML). The fusion oncogene CBFß-SMMHC expressed in AML with the chromosome inversion inv(16)(p13q22) acts as a driver oncogene in hematopoietic stem cells and induces AML. This review focuses on novel insights regarding the molecular mechanisms involved in CBFß-SMMHC-driven leukemogenesis and recent advances in therapeutic approaches to target CBFß-SMMHC in inv(16) AML.


Assuntos
Transformação Celular Neoplásica/genética , Inversão Cromossômica , Cromossomos Humanos Par 16/genética , Subunidade beta de Fator de Ligação ao Core/genética , Imunoterapia/métodos , Leucemia Mieloide Aguda/tratamento farmacológico , Terapia de Alvo Molecular , Cadeias Pesadas de Miosina/genética , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Animais , Antineoplásicos Imunológicos/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cromossomos Humanos Par 16/ultraestrutura , Terapia Combinada , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/fisiologia , Previsões , Gemtuzumab/uso terapêutico , Regulação Leucêmica da Expressão Gênica , Técnicas de Introdução de Genes , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Humanos , Leucemia Mieloide Aguda/genética , Camundongos , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Linfócitos T/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Leuk Res ; 96: 106409, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32652328

RESUMO

Acute myeloid leukemia (AML) is often characterized by the presence of specific and recurrent chromosomal abnormalities. Current treatments have greatly increased remission rate, but relapse still occurs. Therefore, novel therapeutic approaches are required. Previously, using a conditional Cbfb-MYH11 knockin mouse model, we showed that Cbfb-MYH11 induces the expression of a cytokine receptor, IL1RL1. Treatment with IL-33, the only known ligand of IL1RL1, promotes leukemia cell survival in vitro. We further found that IL1RL1+ cells survive better with chemotherapy than IL1RL1- population. However, the mechanism is not clear. Here, we show that IL-33 treatment decreased drug sensitivity in the human inv(16) AML cell line ME-1. By RT-PCR, we found that IL-33 increased the expression of IL-4 and IL-6 and led to the activation of both p38 MAPK and NF-κB. We also showed that IL-33 decreased apoptosis with increased phosphorylation of p38 MAPK. Moreover, pre-treatment with MAPK inhibitor attenuated the phosphorylation of p38 enhanced by IL-33 and reversed the anti-apoptotic effect by IL-33. Taken together, our findings give news insights into the potential mechanism of the anti-apoptotic effect by IL-33/IL1RL1 axis in AML which will help in future drug development.


Assuntos
Apoptose , Subunidade beta de Fator de Ligação ao Core/fisiologia , Interleucina-33/farmacologia , Leucemia Mieloide Aguda/patologia , Cadeias Pesadas de Miosina/fisiologia , NF-kappa B/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Regulação Leucêmica da Expressão Gênica , Proteína 1 Semelhante a Receptor de Interleucina-1/genética , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/genética , Fosforilação , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno/genética
4.
Sci Rep ; 10(1): 9921, 2020 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-32555437

RESUMO

Core Binding Factors (CBFs) are a small group of heterodimeric transcription factor complexes composed of DNA binding proteins, RUNXs, and a non-DNA binding protein, CBFB. The LH surge increases the expression of Runx1 and Runx2 in ovulatory follicles, while Cbfb is constitutively expressed. To investigate the physiological significance of CBFs, we generated a conditional mutant mouse model in which granulosa cell expression of Runx2 and Cbfb was deleted by the Esr2Cre. Female Cbfbflox/flox;Esr2cre/+;Runx2flox/flox mice were infertile; follicles developed to the preovulatory follicle stage but failed to ovulate. RNA-seq analysis of mutant mouse ovaries collected at 11 h post-hCG unveiled numerous CBFs-downstream genes that are associated with inflammation, matrix remodeling, wnt signaling, and steroid metabolism. Mutant mice also failed to develop corpora lutea, as evident by the lack of luteal marker gene expression, marked reduction of vascularization, and excessive apoptotic staining in unruptured poorly luteinized follicles, consistent with dramatic reduction of progesterone by 24 h after hCG administration. The present study provides in vivo evidence that CBFs act as essential transcriptional regulators of both ovulation and luteinization by regulating the expression of key genes that are involved in inflammation, matrix remodeling, cell differentiation, vascularization, and steroid metabolisms in mice.


Assuntos
Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Fertilidade , Células da Granulosa/metabolismo , Infertilidade Feminina/fisiopatologia , Luteinização , Ovulação , Animais , Feminino , Células da Granulosa/citologia , Camundongos , Camundongos Knockout , Reprodução
5.
Oncogene ; 39(12): 2624-2637, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32005976

RESUMO

Epithelial to mesenchymal transition (EMT) is a dynamic process that drives cancer cell plasticity and is thought to play a major role in metastasis. Here we show, using MDA-MB-231 cells as a model, that the plasticity of at least some metastatic breast cancer cells is dependent on the transcriptional co-regulator CBFß. We demonstrate that CBFß is essential to maintain the mesenchymal phenotype of triple-negative breast cancer cells and that CBFß-depleted cells undergo a mesenchymal to epithelial transition (MET) and re-organise into acini-like structures, reminiscent of those formed by epithelial breast cells. We subsequently show, using an inducible CBFß system, that the MET can be reversed, thus demonstrating the plasticity of CBFß-mediated EMT. Moreover, the MET can be reversed by expression of the EMT transcription factor Slug whose expression is dependent on CBFß. Finally, we demonstrate that loss of CBFß inhibits the ability of metastatic breast cancer cells to invade bone cell cultures and suppresses their ability to form bone metastases in vivo. Together our findings demonstrate that CBFß can determine the plasticity of the metastatic cancer cell phenotype, suggesting that its regulation in different micro-environments may play a key role in the establishment of metastatic tumours.


Assuntos
Neoplasias da Mama/patologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Metástase Neoplásica , Animais , Fator de Ligação a CCAAT , Linhagem Celular Tumoral , Feminino , Humanos , Camundongos , Fenótipo
6.
Exp Hematol ; 68: 2-9, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30391350

RESUMO

The transcription factor RUNX1 is required in the embryo for formation of the adult hematopoietic system. Here, we describe the seminal findings that led to the discovery of RUNX1 and of its critical role in blood cell formation in the embryo from hemogenic endothelium (HE). We also present RNA-sequencing data demonstrating that HE cells in different anatomic sites, which produce hematopoietic progenitors with dissimilar differentiation potentials, are molecularly distinct. Hemogenic and non-HE cells in the yolk sac are more closely related to each other than either is to hemogenic or non-HE cells in the major arteries. Therefore, a major driver of the different lineage potentials of the committed erythro-myeloid progenitors that emerge in the yolk sac versus hematopoietic stem cells that originate in the major arteries is likely to be the distinct molecular properties of the HE cells from which they are derived. We used bioinformatics analyses to predict signaling pathways active in arterial HE, which include the functionally validated pathways Notch, Wnt, and Hedgehog. We also used a novel bioinformatics approach to assemble transcriptional regulatory networks and predict transcription factors that may be specifically involved in hematopoietic cell formation from arterial HE, which is the origin of the adult hematopoietic system.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Hemangioblastos/fisiologia , Hematopoese/fisiologia , Animais , Artérias/citologia , Artérias/embriologia , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/fisiologia , Proteínas de Drosophila/genética , Sangue Fetal/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Leucemia Experimental/genética , Leucemia Experimental/virologia , Leucemia Mieloide Aguda/genética , Camundongos , Camundongos Knockout , Proteínas de Fusão Oncogênica/genética , Proteínas de Fusão Oncogênica/fisiologia , Transcrição Gênica , Saco Vitelino/citologia
7.
J Exp Med ; 214(10): 2933-2946, 2017 Oct 02.
Artigo em Inglês | MEDLINE | ID: mdl-28814567

RESUMO

The mouse Langerhans cell (LC) network is established through the differentiation of embryonic LC precursors. BMP7 and TGFß1 initiate cellular signaling that is essential for inducing LC differentiation and preserving LCs in a quiescent state, respectively. Here we show that loss of Cbfß2, one of two RNA splice variants of the Cbfb gene, results in long-term persistence of embryonic LC precursors after their developmental arrest at the transition into the EpCAM+ stage. This phenotype is caused by selective loss of BMP7-mediated signaling essential for LC differentiation, whereas TGFßR signaling is intact, maintaining cells in a quiescent state. Transgenic Cbfß2 expression at the neonatal stage, but not at the adult stage, restored differentiation from Cbfß2-deficient LC precursors. Loss of developmental potential in skin-residential precursor cells was accompanied by diminished BMP7-BMPR1A signaling. Collectively, our results reveal an essential requirement for the Cbfß2 variant in LC differentiation and provide novel insight into how the establishment and homeostasis of the LC network is regulated.


Assuntos
Subunidade beta de Fator de Ligação ao Core/deficiência , Células de Langerhans/fisiologia , Receptores de Fatores de Crescimento Transformadores beta/fisiologia , Animais , Proteína Morfogenética Óssea 7/fisiologia , Receptores de Proteínas Morfogenéticas Ósseas Tipo I/fisiologia , Diferenciação Celular/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Feminino , Citometria de Fluxo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isoformas de Proteínas , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia
9.
Leukemia ; 29(8): 1771-8, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25742748

RESUMO

CBFß-SMMHC (core-binding factor ß-smooth muscle myosin heavy chain), the fusion protein generated by the chromosome 16 inversion fusion gene, CBFB-MYH11, is known to initiate leukemogenesis. However, the mechanism through which CBFß-SMMHC contributes to leukemia development is not well understood. Previously, it was proposed that CBFß-SMMHC acts by dominantly repressing the transcription factor RUNX1 (Runt-related protein 1), but we recently showed that CBFß-SMMHC has activities that are independent of RUNX1 repression. In addition, we showed that a modified CBFß-SMMHC with decreased RUNX1-binding activity accelerates leukemogenesis. These results raise questions about the importance of RUNX1 in leukemogenesis by CBFß-SMMHC. To test this, we generated mice expressing Cbfb-MYH11 in a Runx1-deficient background, resulting from either homozygous Runx1-null alleles (Runx1(-/-)) or a single dominant-negative Runx1 allele (Runx1(+/lz)). We found that loss of Runx1 activity rescued the differentiation defects induced by Cbfb-MYH11 during primitive hematopoiesis. During definitive hematopoiesis, RUNX1 loss also significantly reduced the proliferation and differentiation defects induced by Cbfb-MYH11. Importantly, Cbfb-MYH11-induced leukemia had much longer latency in Runx1(+/lz) mice than in Runx1-sufficient mice. These data indicate that Runx1 activity is critical for Cbfb-MYH11-induced hematopoietic defects and leukemogenesis.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Hematopoese/fisiologia , Leucemia/metabolismo , Leucemia/patologia , Cadeias Pesadas de Miosina/fisiologia , Animais , Apoptose , Western Blotting , Diferenciação Celular , Proliferação de Células , Células Cultivadas , Citometria de Fluxo , Técnicas de Introdução de Genes , Humanos , Imunoprecipitação , Leucemia/mortalidade , Camundongos
10.
J Bone Miner Res ; 30(4): 706-14, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25262822

RESUMO

Runx family proteins, Runx1, Runx2, and Runx3, play important roles in skeletal development. Runx2 is required for osteoblast differentiation and chondrocyte maturation, and haplodeficiency of RUNX2 causes cleidocranial dysplasia, which is characterized by open fontanelles and sutures and hypoplastic clavicles. Cbfb forms a heterodimer with Runx family proteins and enhances their DNA-binding capacity. Cbfb-deficient (Cbfb(-/-) ) mice die at midgestation because of the lack of fetal liver hematopoiesis. We previously reported that the partial rescue of hematopoiesis in Cbfb(-/-) mice revealed the requirement of Cbfb in skeletal development. However, the precise functions of Cbfb in skeletal development still remain to be clarified. We deleted Cbfb in mesenchymal cells giving rise to both chondrocyte and osteoblast lineages by mating Cbfb(fl/fl) mice with Dermo1 Cre knock-in mice. Cbfb(fl/fl/Cre) mice showed dwarfism, both intramembranous and endochondral ossifications were retarded, and chondrocyte maturation and proliferation and osteoblast differentiation were inhibited. The differentiation of chondrocytes and osteoblasts were severely inhibited in vitro, and the reporter activities of Ihh, Col10a1, and Bglap2 promoter constructs were reduced in Cbfb(fl/fl/Cre) chondrocytes or osteoblasts. The proteins of Runx1, Runx2, and Runx3 were reduced in the cartilaginous limb skeletons and calvariae of Cbfb(fl/fl/Cre) embryos compared with the respective protein in the respective tissue of Cbfb(fl/fl) embryos at E15.5, although the reduction of Runx2 protein in calvariae was much milder than that in cartilaginous limb skeletons. All of the Runx family proteins were severely reduced in Cbfb(fl/fl/Cre) primary osteoblasts, and Runx2 protein was less stable in Cbfb(fl/fl/Cre) osteoblasts than Cbfb(fl/fl) osteoblasts. These findings indicate that Cbfb is required for skeletal development by regulating chondrocyte differentiation and proliferation and osteoblast differentiation; that Cbfb plays an important role in the stabilization of Runx family proteins; and that Runx2 protein stability is less dependent on Cbfb in calvariae than in cartilaginous limb skeletons.


Assuntos
Desenvolvimento Ósseo/fisiologia , Subunidade alfa 1 de Fator de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Animais , Diferenciação Celular , Condrócitos/citologia , Subunidade beta de Fator de Ligação ao Core/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoblastos/citologia
11.
J Bone Miner Res ; 30(4): 715-22, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25358268

RESUMO

Core binding factor beta (Cbfß), the partner protein of Runx family transcription factors, enhances Runx function by increasing the binding of Runx to DNA. Null mutations of Cbfb result in embryonic death, which can be rescued by restoring fetal hematopoiesis but only until birth, where bone formation is still nearly absent. Here, we address a direct role of Cbfß in skeletal homeostasis by generating osteoblast-specific Cbfß-deficient mice (Cbfb(Δob/Δob) ) from Cbfb-floxed mice crossed with mice expressing Cre from the Col1a1 promoter. Cbfb(Δob/Δob) mice showed normal growth and development but exhibited reduced bone mass, particularly of cortical bone. The reduction of bone mass in Cbfb(Δob/Δob) mice is similar to the phenotype of mice with haploinsufficiency of Runx2. Although the number of osteoblasts remained unchanged, the number of active osteoblasts decreased in Cbfb(Δob/Δob) mice and resulted in lower mineral apposition rate. Immunohistochemical and quantitative real-time PCR analyses showed that the expression of osteogenic markers, including Runx2, osterix, osteocalcin, and osteopontin, was significantly repressed in Cbfb(Δob/Δob) mice compared with wild-type mice. Cbfß deficiency also reduced Runx2 protein levels in osteoblasts. The mechanism was revealed by forced expression of Cbfß, which increased Runx2 protein levels in vitro by inhibiting polyubiquitination-mediated proteosomal degradation. Collectively, these findings indicate that Cbfß stabilizes Runx2 in osteoblasts by forming a complex and thus facilitates the proper maintenance of bone mass, particularly cortical bone.


Assuntos
Osso e Ossos/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Osteoblastos/metabolismo , Animais , Desenvolvimento Ósseo/fisiologia , Núcleo Celular/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core , Subunidade beta de Fator de Ligação ao Core/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Tamanho do Órgão
12.
J Cell Biochem ; 115(12): 2155-64, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25079347

RESUMO

Mitotic bookmarking is an epigenetic control mechanism that sustains gene expression in progeny cells; it is often found in genes related to the maintenance of cellular phenotype and growth control. RUNX transcription factors regulate a broad spectrum of RNA Polymerase (Pol II) transcribed genes important for lineage commitment but also regulate RNA Polymerase I (Pol I) driven ribosomal gene expression, thus coordinating control of cellular identity and proliferation. In this study, using fluorescence microscopy and biochemical approaches we show that the principal RUNX co-factor, CBFß, associates with nucleolar organizing regions (NORs) during mitosis to negatively regulate RUNX-dependent ribosomal gene expression. Of clinical relevance, we establish for the first time that the leukemogenic fusion protein CBFß-SMMHC (smooth muscle myosin heavy chain) also associates with ribosomal genes in interphase chromatin and mitotic chromosomes to promote and epigenetically sustain regulation of ribosomal genes through RUNX factor interactions. Our results demonstrate that CBFß contributes to the transcriptional regulation of ribosomal gene expression and provide further understanding of the epigenetic role of CBFß-SMMHC in proliferation and maintenance of the leukemic phenotype.


Assuntos
Subunidade beta de Fator de Ligação ao Core/fisiologia , Epigênese Genética , Cadeias Pesadas de Miosina/fisiologia , Proteínas de Fusão Oncogênica/fisiologia , Ribossomos/genética , Linhagem Celular Tumoral , Cromossomos/metabolismo , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , DNA Polimerase I/metabolismo , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia , Mitose , Ribossomos/metabolismo
13.
J Virol ; 88(20): 12112-22, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25122780

RESUMO

Viral infectivity factor (Vif) is required for lentivirus fitness and pathogenicity, except in equine infectious anemia virus (EIAV). Vif enhances viral infectivity by a Cullin5-Elongin B/C E3 complex to inactivate the host restriction factor APOBEC3. Core-binding factor subunit beta (CBF-ß) is a cell factor that was recently shown to be important for the primate lentiviral Vif function. Non-primate lentiviral Vif also degrades APOBEC3 through the proteasome pathway. However, it is unclear whether CBF-ß is required for the non-primate lentiviral Vif function. In this study, we demonstrated that the Vifs of non-primate lentiviruses, including feline immunodeficiency virus (FIV), bovine immunodeficiency virus (BIV), caprine arthritis encephalitis virus (CAEV), and maedi-visna virus (MVV), do not interact with CBF-ß. In addition, CBF-ß did not promote the stability of FIV, BIV, CAEV, and MVV Vifs. Furthermore, CBF-ß silencing or overexpression did not affect non-primate lentiviral Vif-mediated APOBEC3 degradation. Our results suggest that non-primate lentiviral Vif induces APOBEC3 degradation through a different mechanism than primate lentiviral Vif. Importance: The APOBEC3 protein family members are host restriction factors that block retrovirus replication. Vif, an accessory protein of lentivirus, degrades APOBEC3 to rescue viral infectivity by forming Cullin5-Elongin B/C-based E3 complex. CBF-ß was proved to be a novel regulator of primate lentiviral Vif function. In this study, we found that CBF-ß knockdown or overexpression did not affect FIV Vif's function, which induced polyubiquitination and degradation of APOBEC3 by recruiting the E3 complex in a manner similar to that of HIV-1 Vif. We also showed that other non-primate lentiviral Vifs did not require CBF-ß to degrade APOBEC3. CBF-ß did not interact with non-primate lentiviral Vifs or promote their stability. These results suggest that a different mechanism exists for the Vif-APOBEC interaction and that non-primates are not suitable animal models for exploring pharmacological interventions that disrupt Vif-CBF-ß interaction.


Assuntos
Subunidade beta de Fator de Ligação ao Core/fisiologia , Citosina Desaminase/metabolismo , Produtos do Gene vif/fisiologia , Lentivirus/fisiologia , Desaminases APOBEC , Sequência de Bases , Citidina Desaminase , Primers do DNA , Células HEK293 , Humanos , Lentivirus/classificação , Proteólise , Reação em Cadeia da Polimerase Via Transcriptase Reversa
14.
Proc Natl Acad Sci U S A ; 111(23): 8482-7, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24850862

RESUMO

The pathogenesis of cleidocranial dysplasia (CCD) as well as the specific role of core binding factor ß (Cbfß) and the Runt-related transcription factor (RUNX)/Cbfß complex in postnatal skeletogenesis remain unclear. We demonstrate that Cbfß ablation in osteoblast precursors, differentiating chondrocytes, osteoblasts, and odontoblasts via Osterix-Cre, results in severe craniofacial dysplasia, skeletal dysplasia, abnormal teeth, and a phenotype recapitulating the clinical features of CCD. Cbfß(f/f)Osterix-Cre mice have fewer proliferative and hypertrophic chondrocytes, fewer osteoblasts, and almost absent trabecular bone, indicating that Cbfß may maintain trabecular bone formation through its function in hypertrophic chondrocytes and osteoblasts. Cbfß(f/f)Collagen, type 1, alpha 1 (Col1α1)-Cre mice show decreased bone mineralization and skeletal deformities, but no radical deformities in teeth, mandibles, or cartilage, indicating that osteoblast lineage-specific ablation of Cbfß results in milder bone defects and less resemblance to CCD. Activating transcription factor 4 (Atf4) and Osterix protein levels in both mutant mice are dramatically reduced. ChIP assays show that Cbfß directly associates with the promoter regions of Atf4 and Osterix. Our data further demonstrate that Cbfß highly up-regulates the expression of Atf4 at the transcriptional regulation level. Overall, our genetic dissection approach revealed that Cbfß plays an indispensable role in postnatal skeletal development and homeostasis in various skeletal cell types, at least partially by up-regulating the expression of Atf4 and Osterix. It also revealed that CCD may result from functional defects of the Runx2/Cbfß heterodimeric complex in various skeletal cells. These insights into the role of Cbfß in postnatal skeletogenesis and CCD pathogenesis may assist in the development of new therapies for CCD and osteoporosis.


Assuntos
Condrócitos/fisiologia , Displasia Cleidocraniana/fisiopatologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Osteoblastos/fisiologia , Osteogênese/fisiologia , Fator 4 Ativador da Transcrição/genética , Fator 4 Ativador da Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Western Blotting , Células Cultivadas , Condrócitos/metabolismo , Displasia Cleidocraniana/genética , Displasia Cleidocraniana/metabolismo , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia de Fluorescência , Osteoblastos/metabolismo , Osteogênese/genética , Regiões Promotoras Genéticas/genética , Ligação Proteica , Multimerização Proteica , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Crânio/citologia , Crânio/metabolismo , Fator de Transcrição Sp7 , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
15.
Leukemia ; 28(4): 770-8, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24002588

RESUMO

Different mechanisms for CBFß-MYH11 function in acute myeloid leukemia with inv(16) have been proposed such as tethering of RUNX1 outside the nucleus, interference with transcription factor complex assembly and recruitment of histone deacetylases, all resulting in transcriptional repression of RUNX1 target genes. Here, through genome-wide CBFß-MYH11-binding site analysis and quantitative interaction proteomics, we found that CBFß-MYH11 localizes to RUNX1 occupied promoters, where it interacts with TAL1, FLI1 and TBP-associated factors (TAFs) in the context of the hematopoietic transcription factors ERG, GATA2 and PU.1/SPI1 and the coregulators EP300 and HDAC1. Transcriptional analysis revealed that upon fusion protein knockdown, a small subset of the CBFß-MYH11 target genes show increased expression, confirming a role in transcriptional repression. However, the majority of CBFß-MYH11 target genes, including genes implicated in hematopoietic stem cell self-renewal such as ID1, LMO1 and JAG1, are actively transcribed and repressed upon fusion protein knockdown. Together these results suggest an essential role for CBFß-MYH11 in regulating the expression of genes involved in maintaining a stem cell phenotype.


Assuntos
Inversão Cromossômica , Cromossomos Humanos Par 16 , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Leucemia Mieloide Aguda/genética , Cadeias Pesadas de Miosina/fisiologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Sítios de Ligação , Fator de Transcrição GATA2/fisiologia , Histona Desacetilases/fisiologia , Humanos , Regiões Promotoras Genéticas , Proteína Proto-Oncogênica c-fli-1/fisiologia , Proteínas Proto-Oncogênicas/fisiologia , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Ativação Transcricional
16.
Biochemistry ; 51(44): 8702-4, 2012 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-23098073

RESUMO

HIV-1 Vif masquerades as a receptor for a cellular E3 ligase harboring Elongin B, Elongin C, and Cullin 5 (EloB/C/Cul5) proteins that facilitate degradation of the antiretroviral factor APOBEC3G (A3G). This Vif-mediated activity requires human core-binding factor ß (CBFß) in contrast to cellular substrate receptors. We observed calorimetrically that Cul5 binds tighter to full-length Vif((1-192))/EloB/C/CBFß (K(d) = 5 ± 2 nM) than to Vif((95-192))/EloB/C (K(d) = 327 ± 40 nM), which cannot bind CBFß. A comparison of heat capacity changes supports a model in which CBFß prestabilizes Vif((1-192)) relative to Vif((95-192)), consistent with a stronger interaction of Cul5 with Vif's C-terminal Zn(2+)-binding motif. An additional interface between Cul5 and an N-terminal region of Vif appears to be plausible, which has therapeutic design implications.


Assuntos
Subunidade beta de Fator de Ligação ao Core/fisiologia , Proteínas Culina/metabolismo , Produtos do Gene vif do Vírus da Imunodeficiência Humana/metabolismo , Humanos , Termodinâmica , Ubiquitina-Proteína Ligases/metabolismo
17.
J Immunol ; 186(3): 1450-7, 2011 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-21178013

RESUMO

Hematopoietic lymphoid tissue inducer (LTi) cells are essential for the development of secondary lymphoid tissues including lymph nodes and Peyer's patches. Two transcription factors, the helix-loop-helix inhibitor Id2 and the retinoic acid-related orphan receptor γt (Rorγt), have been shown to be crucial for LTi cell development. However, it remains unclear how the specification of multipotent hematopoietic progenitor cells toward the LTi lineage is programmed. In this study, we report impaired lymphoid tissue organogenesis in mice in which the function of Runx1/Cbfß transcription factor complexes was attenuated by the loss of either the distal promoter-derived Runx1 or Cbfß2 variant protein. We found that LTi progenitors in fetal liver, defined previously as a lineage marker-negative α4ß7 integrin (α4ß7)(+) IL-7R α-chain (IL-7Rα)(+) population, can be subdivided into Rorγt-expressing IL-7Rα(high) cells and nonexpressing IL-7Rα(mid) cells. Whereas Id2 and Rorγt are required to direct α4ß7(+)IL-7Rα(mid) cells to become α4ß7(+)IL-7Rα(high) cells, Runx1/Cbfß2 complexes are necessary for the emergence of α4ß7(+)IL-7Rα(mid) cells. In addition, the loss of Cbfß2, but not P1-Runx1, resulted in an inefficient upregulation of Rorγt in residual α4ß7(+)IL-7Rα(+) LTi cells at anlagen. Our results thus revealed that Runx1/Cbfß2 complexes regulate the differentiation of LTi cells at two stages: an early specification of hematopoietic progenitors toward the LTi lineage and a subsequent activation of Rorγt expression at anlagen.


Assuntos
Diferenciação Celular/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Tecido Linfoide/imunologia , Animais , Linhagem da Célula/genética , Linhagem da Célula/imunologia , Subunidade alfa 2 de Fator de Ligação ao Core/deficiência , Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/deficiência , Subunidade beta de Fator de Ligação ao Core/genética , Variação Genética/imunologia , Proteína 2 Inibidora de Diferenciação/biossíntese , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/fisiologia , Fígado/embriologia , Fígado/imunologia , Fígado/patologia , Tecido Linfoide/citologia , Tecido Linfoide/embriologia , Camundongos , Camundongos Mutantes , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/imunologia , Células-Tronco Multipotentes/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/biossíntese , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/fisiologia
18.
Mol Cancer ; 9: 171, 2010 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-20591170

RESUMO

BACKGROUND: The transcription factor Runx2 has an established role in cancers that metastasize to bone. In metastatic breast cancer cells Runx2 is overexpressed and contributes to the invasive capacity of the cells by regulating the expression of several invasion genes. CBFbeta is a transcriptional co-activator that is recruited to promoters by Runx transcription factors and there is considerable evidence that CBFbeta is essential for the function of Runx factors. However, overexpression of Runx1 can partially rescue the lethal phenotype in CBFbeta-deficient mice, indicating that increased levels of Runx factors can, in some situations, overcome the requirement for CBFbeta. Since Runx2 is overexpressed in metastatic breast cancer cells, and there are no reports of CBFbeta expression in breast cells, we sought to determine whether Runx2 function in these cells was dependent on CBFbeta. Such an interaction might represent a viable target for therapeutic intervention to inhibit bone metastasis. RESULTS: We show that CBFbeta is expressed in the metastatic breast cancer cells, MDA-MB-231, and that it associates with Runx2. Matrigel invasion assays and RNA interference were used to demonstrate that CBFbeta contributes to the invasive capacity of these cells. Subsequent analysis of Runx2 target genes in MDA-MB-231 cells revealed that CBFbeta is essential for the expression of Osteopontin, Matrixmetalloproteinase-13, Matrixmetalloproteinase-9, and Osteocalcin but not for Galectin-3. Chromatin immunoprecipitation analysis showed that CBFbeta is recruited to both the Osteopontin and the Galectin-3 promoters. CONCLUSIONS: CBFbeta is expressed in metastatic breast cancer cells and is essential for cell invasion. CBFbeta is required for expression of several Runx2-target genes known to be involved in cell invasion. However, whilst CBFbeta is essential for invasion, not all Runx2-target genes require CBFbeta. We conclude that CBFbeta is required for a subset of Runx2-target genes that are sufficient to maintain the invasive phenotype of the cells. These findings suggest that the interaction between Runx2 and CBFbeta might represent a viable target for therapeutic intervention to inhibit bone metastasis.


Assuntos
Neoplasias da Mama/patologia , Subunidade beta de Fator de Ligação ao Core/fisiologia , Invasividade Neoplásica , Animais , Linhagem Celular Tumoral , Subunidade beta de Fator de Ligação ao Core/genética , Feminino , Humanos , Camundongos , Metástase Neoplásica , Interferência de RNA
19.
Cancer Cell ; 17(5): 455-68, 2010 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-20478528

RESUMO

Dominant RUNX1 inhibition has been proposed as a common pathway for CBF leukemia. CBF beta-SMMHC, a fusion protein in human acute myeloid leukemia (AML), dominantly inhibits RUNX1 largely through its RUNX1 high-affinity binding domain (HABD). However, the type I CBF beta-SMMHC fusion in AML patients lacks HABD. Here, we report that the type I CBF beta-SMMHC protein binds RUNX1 inefficiently. Knockin mice expressing CBF beta-SMMHC with a HABD deletion developed leukemia quickly, even though hematopoietic defects associated with Runx1-inhibition were partially rescued. A larger pool of leukemia-initiating cells, increased MN1 expression, and retention of RUNX1 phosphorylation are potential mechanisms for accelerated leukemia development in these mice. Our data suggest that RUNX1 dominant inhibition may not be a critical step for leukemogenesis by CBF beta-SMMHC.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , Subunidade beta de Fator de Ligação ao Core/fisiologia , Leucemia Experimental/fisiopatologia , Proteínas de Fusão Oncogênica/fisiologia , Animais , Subunidade beta de Fator de Ligação ao Core/metabolismo , Humanos , Camundongos , Camundongos Transgênicos , Proteínas de Fusão Oncogênica/metabolismo , Fosforilação , Ligação Proteica
20.
Cancer Treat Res ; 145: 127-47, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20306249

RESUMO

The existence of non-random mutations in critical regulators of cell growth and differentiation is a recurring theme in cancer pathogenesis and provides the basis for our modern, molecular approach to the study and treatment of malignant diseases. Nowhere is this more true than in the study of leukemogenesis, where research has converged upon a critical group of genes involved in hematopoietic stem and progenitor cell self-renewal and fate specification. Prominent among these is the heterodimeric transcriptional regulator, RUNX1/CBFbeta. RUNX1 is a site-specific DNA-binding protein whose consensus response element is found in the promoters of many hematopoietically relevant genes. CBFbeta interacts with RUNX1, stabilizing its interaction with DNA to promote the actions of RUNX1/CBFbeta in transcriptional control. Both the RUNX1 and the CBFbeta genes participate in proleukemic chromosomal alterations. Together they contribute to approximately one-third of acute myelogenous leukemia (AML) and one-quarter of acute lymphoblastic leukemia (ALL) cases, making RUNX1 and CBFbeta the most frequently affected genes known in the pathogenesis of acute leukemia. Investigating the mechanisms by which RUNX1, CBFbeta, and their proleukemic fusion proteins influence leukemogenesis has contributed greatly to our understanding of both normal and malignant hematopoiesis. Here we present an overview of the structural features of RUNX1/CBFbeta and their derivatives, their roles in transcriptional control, and their contributions to normal and malignant hematopoiesis.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/genética , Subunidade beta de Fator de Ligação ao Core/genética , Regulação Leucêmica da Expressão Gênica/genética , Leucemia Mieloide Aguda/genética , Mutação , Proteínas de Neoplasias/genética , Proteínas de Fusão Oncogênica/fisiologia , Transformação Celular Neoplásica/genética , Subunidade alfa 2 de Fator de Ligação ao Core/química , Subunidade alfa 2 de Fator de Ligação ao Core/fisiologia , Subunidade beta de Fator de Ligação ao Core/química , Subunidade beta de Fator de Ligação ao Core/fisiologia , Hematopoese , Humanos , Leucemia Mieloide Aguda/patologia , Proteínas de Neoplasias/química , Proteínas de Neoplasias/fisiologia , Proteínas de Fusão Oncogênica/genética , Transcrição Gênica , Translocação Genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA