Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
2.
Proc Natl Acad Sci U S A ; 111(46): 16407-12, 2014 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-25362053

RESUMO

Masculinization of external genitalia is an essential process in the formation of the male reproductive system. Prominent characteristics of this masculinization are the organ size and the sexual differentiation of the urethra. Although androgen is a pivotal inducer of the masculinization, the regulatory mechanism under the control of androgen is still unknown. Here, we address this longstanding question about how androgen induces masculinization of the embryonic external genitalia through the identification of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog B (Mafb) gene. Mafb is expressed prominently in the mesenchyme of male genital tubercle (GT), the anlage of external genitalia. MAFB expression is rarely detected in the mesenchyme of female GTs. However, exposure to exogenous androgen induces its mesenchymal expression in female GTs. Furthermore, MAFB expression is prominently down-regulated in male GTs of androgen receptor (Ar) KO mice, indicating that AR signaling is necessary for its expression. It is revealed that Mafb KO male GTs exhibit defective embryonic urethral formation, giving insight into the common human congenital anomaly hypospadias. However, the size of Mafb KO male GTs is similar with that of wild-type males. Moreover, androgen treatment fails to induce urethral masculinization of the GTs in Mafb KO mice. The current results provide evidence that Mafb is an androgen-inducible, sexually dimorphic regulator of embryonic urethral masculinization.


Assuntos
Genitália Masculina/embriologia , Fator de Transcrição MafB/fisiologia , Mesoderma/metabolismo , Caracteres Sexuais , Diferenciação Sexual/fisiologia , Uretra/embriologia , Androgênios/fisiologia , Animais , Modelos Animais de Doenças , Feminino , Genitália Feminina/embriologia , Genitália Feminina/metabolismo , Genitália Masculina/metabolismo , Hipospadia/embriologia , Hipospadia/genética , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/deficiência , Fator de Transcrição MafB/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Androgênicos/deficiência , Receptores Androgênicos/fisiologia , Uretra/anormalidades , Uretra/metabolismo
3.
Dev Biol ; 385(1): 2-12, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-24183936

RESUMO

Specification and maturation of insulin(+) cells accompanies a transition in expression of Maf family of transcription factors. In development, MafA is expressed after specification of insulin(+) cells that are expressing another Maf factor, MafB; after birth, these insulin(+) MafA(+) cells stop MafB expression and gain glucose responsiveness. Current differentiation protocols for deriving insulin-producing ß-cells from stem cells result in ß-cells lacking both MafA expression and glucose-stimulated insulin secretion. So driving expression of MafA, a ß-cell maturation factor in endocrine precursors could potentially generate glucose-responsive MafA(+) ß cells. Using inducible transgenic mice, we characterized the final stages of ß-cell differentiation and maturation with MafA pause/release experiments. We found that forcing MafA transgene expression, out of its normal developmental context, in Ngn3(+) endocrine progenitors blocked endocrine differentiation and prevented the formation of hormone(+) cells. However, this arrest was reversible such that with stopping the transgene expression, the cells resumed their differentiation to hormone(+) cells, including α-cells, indicating that the block likely occurred after progenitors had committed to a specific hormonal fate. Interestingly, this delayed resumption of endocrine differentiation resulted in a greater proportion of immature insulin(+)MafB(+) cells at P5, demonstrating that during maturation the inhibition of MafB in ß-cell transitioning from insulin(+)MafB(+) to insulin(+)MafB(-) stage is regulated by cell-autonomous mechanisms. These results demonstrate the importance of proper context of initiating MafA expression on the endocrine differentiation and suggest that generating mature Insulin(+)MafA(+) ß-cells will require the induction of MafA in a narrow temporal window to achieve normal endocrine differentiation.


Assuntos
Células Endócrinas/metabolismo , Fatores de Transcrição Maf Maior/metabolismo , Fator de Transcrição MafB/metabolismo , Células-Tronco/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Diferenciação Celular/genética , Células Endócrinas/citologia , Regulação da Expressão Gênica no Desenvolvimento , Insulina/biossíntese , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição Maf Maior/biossíntese , Fatores de Transcrição Maf Maior/genética , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Camundongos , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Pâncreas/citologia , Pâncreas/metabolismo
4.
BMC Cancer ; 15: 461, 2015 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-26055874

RESUMO

BACKGROUND: Mounting evidence suggests that miRNAs have major functions in tumor pathogenesis, and this study aimed to identify the candidate miRNA and investigate its role in nasopharyngeal carcinoma (NPC). METHODS: MiRNA and mRNA expressions were screened by microarray assays. The cell proliferation, colony formation and migration ability were measured by MTT, soft agar and wound healing assays, respectively. The tumor growth suppression was evaluated by xenografting in nude mice. The plasma miR-223 levels in NPC patients were detected by TaqMan analysis. Real-time quantitative PCR and Western blotting were used to confirm miR-223 and MAFB expression levels. The targeting relationship between miR-223 and MAFB was verified using dual luciferase reporter assay. RESULTS: The miR-223 expression was decreased in CNE-1, CNE-2 cells as compared with NP69 cells, an immortalized human nasopharyngeal epithelial cell line, and its level also reduced in NPC patients' plasma as compared with healthy controls. Exogenous expression of miR-223 in CNE-2 cells could inhibit cell proliferation both in vitro and in vivo. Extrogenous miR-223 in CNE-2 cells would decrease the ability of colony formation and migration. MAFB, a transcription factor of Maf family members, was identified as a target gene of miR-223. We found that migration and invasion abilities were inhibited by MAFB silencing. CONCLUSIONS: MiR-223 negatively regulates the growth and migration of NPC cells via reducing MAFB expression, and this finding provides a novel insight into understanding miR-223 regulation mechanism in nasopharyngeal carcinoma tumorigenesis.


Assuntos
Proliferação de Células/genética , Fator de Transcrição MafB/biossíntese , MicroRNAs/genética , Neoplasias Nasofaríngeas/genética , Animais , Carcinogênese , Carcinoma , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Fator de Transcrição MafB/antagonistas & inibidores , Fator de Transcrição MafB/genética , Camundongos , MicroRNAs/biossíntese , MicroRNAs/sangue , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/sangue , Neoplasias Nasofaríngeas/patologia , RNA Mensageiro/biossíntese , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Respirology ; 16(1): 124-30, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-20969674

RESUMO

BACKGROUND AND OBJECTIVE: Alveolar macrophages (AM) play an important role in the pathogenesis of COPD, and their numbers are significantly increased in patients with COPD. We previously demonstrated that expression of the transcription factor, MafB, was upregulated in AM of mice exposed to cigarette smoke. The aim of this study was to investigate whether the expression of MafB is associated with the degree of airflow limitation (AFL) in smokers. METHODS: Lung tissue specimens were obtained from male patients undergoing resection of small peripheral lung tumours. The patients were classified into three groups according to smoking index and FEV1 /FVC: (i) non-smokers or non-heavy ex-smokers without AFL (FEV1 /FVC ≥ 0.7, smoking index ≤ 400) (n=8); (ii) heavy current smokers without AFL (FEV1 /FVC ≥ 0.7, smoking index ≥ 800) (n=8); and (iii) heavy current smokers with AFL (FEV1 /FVC < 0.6, smoking index ≥ 800) (n=8). The intensity of immunostaining for MafB in AM was quantified by image analysis. RESULTS: Immunostaining for MafB was significantly enhanced in AM of smokers with AFL compared with AM of subjects without AFL. Smoking index, FEV1/FVC and FEF(25-75%) (% predicted) were significantly correlated with the intensity of MafB immunostaining. Multiple linear regression analysis revealed that FEV1 % was also an independent negative predictor of the intensity of MafB immunostaining. CONCLUSIONS: The degree of immunostaining for MafB in AM was correlated with the degree of AFL in smokers. MafB may be involved in the pathophysiology of COPD.


Assuntos
Pulmão/química , Macrófagos Alveolares/metabolismo , Fator de Transcrição MafB/biossíntese , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , Fumar/fisiopatologia , Idoso , Humanos , Imuno-Histoquímica , Pulmão/fisiopatologia , Macrófagos Alveolares/química , Fator de Transcrição MafB/análise , Masculino , Pessoa de Meia-Idade , Espirometria
6.
Eur J Haematol ; 84(4): 298-303, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20002157

RESUMO

The identification of aberrantly hypermethylated genes may lead to the development of new diagnostic markers and the identification of novel targets of epigenetic therapy in myelodysplastic syndromes (MDS). We therefore investigated the methylation status of transcription factor genes KLF5, KLF11, and MAFB, shown to be aberrantly methylated in myelogeneous leukaemia cells, in a series of 115 MDS patient as well as in 25 control subjects. Using quantitative high-resolution pyrosequencing methodology, KLF11, MAFB, and KLF5 were shown for the first time to be hypermethylated in 17 (15%), 8 (7%), and 2 (1.7%) cases, respectively, but not in any of the patients with an isolated 5q-deletion. Patient samples harbouring KLF11 methylation displayed reduced KLF11 mRNA expression and KLF11 hypermethylation correlated with a high International Prognostic Scoring System score (P < 0.05). In conclusion, epigenetic inactivation and subsequent transcriptional repression of the KLF11 gene is quite frequent in MDS. Patients with an isolated 5q-deletion seem to harbour a distinct epigenetic profile.


Assuntos
Proteínas de Ciclo Celular/biossíntese , Metilação de DNA , Epigênese Genética , Inativação Gênica , Genes Supressores de Tumor , Síndromes Mielodisplásicas/metabolismo , Proteínas Repressoras/biossíntese , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas Reguladoras de Apoptose , Proteínas de Ciclo Celular/genética , Feminino , Humanos , Fatores de Transcrição Kruppel-Like/biossíntese , Fatores de Transcrição Kruppel-Like/genética , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Masculino , Pessoa de Meia-Idade , Síndromes Mielodisplásicas/genética , Proteínas Repressoras/genética
7.
J Immunol ; 181(8): 5660-72, 2008 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-18832725

RESUMO

Although a considerable number of reports indicate an involvement of the Hox-A10 gene in the molecular control of hemopoiesis, the conclusions of such studies are quite controversial given that they support, in some cases, a role in the stimulation of stem cell self-renewal and myeloid progenitor expansion, whereas in others they implicate this transcription factor in the induction of monocyte-macrophage differentiation. To clarify this issue, we analyzed the biological effects and the transcriptome changes determined in human primary CD34(+) hemopoietic progenitors by retroviral transduction of a full-length Hox-A10 cDNA. The results obtained clearly indicated that this homeogene is an inducer of monocyte differentiation, at least partly acting through the up-regulation of the MafB gene, recently identified as the master regulator of such a maturation pathway. By using a combined approach based on computational analysis, EMSA experiments, and luciferase assays, we were able to demonstrate the presence of a Hox-A10-binding site in the promoter region of the MafB gene, which suggested the likely molecular mechanism underlying the observed effect. Stimulation of the same cells with the vitamin D(3) monocyte differentiation inducer resulted in a clear increase of Hox-A10 and MafB transcripts, indicating the existence of a precise transactivation cascade involving vitamin D(3) receptor, Hox-A10, and MafB transcription factors. Altogether, these data allow one to conclude that the vitamin D(3)/Hox-A10 pathway supports MafB function during the induction of monocyte differentiation.


Assuntos
Antígenos CD34 , Diferenciação Celular/imunologia , Colecalciferol/farmacologia , Proteínas de Homeodomínio/imunologia , Fator de Transcrição MafB/imunologia , Monócitos/imunologia , Células Progenitoras Mieloides/imunologia , Vitaminas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Células HL-60 , Hematopoese/efeitos dos fármacos , Hematopoese/genética , Hematopoese/imunologia , Proteínas Homeobox A10 , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Células K562 , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Monócitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Elementos de Resposta/genética , Elementos de Resposta/imunologia , Retroviridae , Transdução Genética , Células U937 , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/genética , Regulação para Cima/imunologia
8.
J Neurochem ; 104(5): 1364-71, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18036148

RESUMO

Non-visual opsins mediate various light-dependent physiological events. Our previous search for non-visual opsin genes in zebrafish led to the discovery of VAL-opsin (VAL-opsinA) in deep brain cells and retinal horizontal cells of the adult fish. In this study, we report the identification and characterization of its duplicated gene, VAL-opsinB, in zebrafish. A molecular phylogenetic analysis indicates that VAL-opsinB is orthologous to a previously reported salmon gene and that the duplication of the VAL-opsin gene occurred in the teleost lineage. The recombinant protein of zebrafish VAL-opsinB forms a green-sensitive photopigment when reconstituted with 11-cis-retinal. VAL-opsinB expression was detected in a limited number of cells of the brain and the eye, and the expression pattern is distinct from that of the VAL-opsinA gene. Such a differential expression pattern suggests that VAL-opsinA and VAL-opsinB are involved in different physiological events in zebrafish.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Genes Duplicados/fisiologia , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Proteínas do Tecido Nervoso/biossíntese , Proteínas do Tecido Nervoso/genética , Opsinas de Bastonetes/biossíntese , Opsinas de Bastonetes/genética , Proteínas de Peixe-Zebra/biossíntese , Proteínas de Peixe-Zebra/genética , Animais , Filogenia , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Células Horizontais da Retina/embriologia , Células Horizontais da Retina/fisiologia , Peixe-Zebra
9.
Artigo em Inglês | MEDLINE | ID: mdl-17671361

RESUMO

The MafB transcription factor (residues 211-305) has been overexpressed in and purified from Escherichia coli. A protein-DNA complex between the MafB homodimer and the 21 bp Maf-recognition sequence known as Cmare has been successfully reconstituted in vitro and subsequently crystallized. The diffraction properties of the protein-DNA complex crystals were improved using a combination of protein-construct boundary optimization and targeted mutagenesis to promote crystal lattice stability. Both native and mercury-derivatized crystals have been prepared using these optimized conditions. The crystals belong to space group P4(1)2(1)2 or P4(3)2(1)2, with unit-cell parameters a = b = 94.8, c = 197.9 A. An anomalous difference Patterson map computed using data collected from crystals grown in the presence of HgCl(2) reveals four peaks. This corresponds to two copies of the protein-DNA complex in the asymmetric unit, with a solvent content of 62% and a Matthews coefficient of 3.22 A(3) Da(-1).


Assuntos
DNA/metabolismo , Fator de Transcrição MafB/genética , Fator de Transcrição MafB/isolamento & purificação , Animais , Cristalização , Cristalografia por Raios X , DNA/isolamento & purificação , Regulação da Expressão Gênica/fisiologia , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/metabolismo , Camundongos , Proteínas Recombinantes/biossíntese , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo
10.
PLoS One ; 12(1): e0170568, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28118374

RESUMO

Primary auditory neurons (PANs) connect cochlear sensory hair cells in the mammalian inner ear to cochlear nucleus neurons in the brainstem. PANs develop from neuroblasts delaminated from the proneurosensory domain of the otocyst and keep maturing until the onset of hearing after birth. There are two types of PANs: type I, which innervate the inner hair cells (IHCs), and type II, which innervate the outer hair cells (OHCs). Glial cells surrounding these neurons originate from neural crest cells and migrate to the spiral ganglion. Several transcription factors are known to regulate the development and differentiation of PANs. Here we systematically examined the spatiotemporal expression of five transcription factors: Sox2, Sox10, Gata3, Mafb, and Prox1 from early delamination at embryonic day (E) 10.5 to adult. We found that Sox2 and Sox10 were initially expressed in the proneurosensory cells in the otocyst (E10.5). By E12.75 both Sox2 and Sox10 were downregulated in the developing PANs; however, Sox2 expression transiently increased in the neurons around birth. Furthermore, both Sox2 and Sox10 continued to be expressed in spiral ganglion glial cells. We also show that Gata3 and Prox1 were first expressed in all developing neurons, followed by a decrease in expression of Gata3 and Mafb in type I PANs and Prox1 in type II PANs as they matured. Moreover, we describe two subtypes of type II neurons based on Peripherin expression. These results suggest that Sox2, Gata3 and Prox1 play a role during neurogenesis as well as maturation of the PANs.


Assuntos
Cóclea/embriologia , Fator de Transcrição GATA3/biossíntese , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/biossíntese , Proteínas do Tecido Nervoso/biossíntese , Neurogênese , Fatores de Transcrição SOXB1/biossíntese , Células Receptoras Sensoriais/metabolismo , Gânglio Espiral da Cóclea/embriologia , Proteínas Supressoras de Tumor/biossíntese , Animais , Núcleo Celular/metabolismo , Cóclea/crescimento & desenvolvimento , Cóclea/metabolismo , Fator de Transcrição GATA3/genética , Técnicas de Introdução de Genes , Genes Reporter , Idade Gestacional , Proteínas de Homeodomínio/genética , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Camundongos , Proteínas do Tecido Nervoso/genética , Crista Neural/metabolismo , Células-Tronco Neurais/metabolismo , Neurogênese/genética , Neuroglia/metabolismo , Periferinas/biossíntese , Periferinas/genética , Proteínas Recombinantes de Fusão/biossíntese , Fatores de Transcrição SOXB1/genética , Fatores de Transcrição SOXE/biossíntese , Fatores de Transcrição SOXE/genética , Células Receptoras Sensoriais/classificação , Gânglio Espiral da Cóclea/metabolismo , Proteínas Supressoras de Tumor/genética
11.
Mech Dev ; 122(12): 1300-9, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16274963

RESUMO

The early transcriptional hierarchy that subdivides the vertebrate hindbrain into seven to eight segments, the rhombomeres (r1-r8), is largely unknown. The Kreisler (MafB, Krml1, Val) gene is earliest gene expressed in an r5/r6-restricted manner and is essential for r5 and r6 development. We have identified the S5 regulatory element that directs early Kreisler expression in the future r5/r6 domain in 0-10 somite stage embryos. variant Hepatocyte Nuclear Factor 1 (vHNF1/HNF1beta/LF-3B) is transiently expressed in the r5/r6 domain of 0-10 somite stage embryos and a vHNF1binding site within this element is essential but not sufficient for r5/r6-specific expression. Thus, early inductive events that initiate Kreisler expression are clearly distinct from later-acting ones that modulate its expression levels. This site and some of the surrounding sequences are evolutionarily conserved in the genomic DNA upstream of the Kreisler gene among species as divergent as mouse, humans, and chickens. This provides the first evidence of a direct requirement for vHNF1 in initiation of Kreisler expression, suggests that the role of vHNF1 is evolutionarily conserved, and indicates that vHNF1 collaborates with other transcription factors, which independently bind to the S5 regulatory region, to establish the r5/r6 domain.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Fator 1 Nuclear de Hepatócito/fisiologia , Proteínas de Homeodomínio/fisiologia , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Rombencéfalo/embriologia , Animais , Sequência de Bases , Sítios de Ligação/genética , Diferenciação Celular/genética , Sequência Conservada , Elementos Facilitadores Genéticos , Variação Genética , Fator 1 Nuclear de Hepatócito/biossíntese , Fator 1 Nuclear de Hepatócito/genética , Proteínas de Homeodomínio/biossíntese , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Rombencéfalo/citologia , Rombencéfalo/metabolismo
12.
J Clin Invest ; 126(5): 1857-70, 2016 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-27064285

RESUMO

Type 2 diabetes is characterized by insulin resistance, hyperglycemia, and progressive ß cell dysfunction. Excess glucose and lipid impair ß cell function in islet cell lines, cultured rodent and human islets, and in vivo rodent models. Here, we examined the mechanistic consequences of glucotoxic and lipotoxic conditions on human islets in vivo and developed and/or used 3 complementary models that allowed comparison of the effects of hyperglycemic and/or insulin-resistant metabolic stress conditions on human and mouse islets, which responded quite differently to these challenges. Hyperglycemia and/or insulin resistance impaired insulin secretion only from human islets in vivo. In human grafts, chronic insulin resistance decreased antioxidant enzyme expression and increased superoxide and amyloid formation. In human islet grafts, expression of transcription factors NKX6.1 and MAFB was decreased by chronic insulin resistance, but only MAFB decreased under chronic hyperglycemia. Knockdown of NKX6.1 or MAFB expression in a human ß cell line recapitulated the insulin secretion defect seen in vivo. Contrary to rodent islet studies, neither insulin resistance nor hyperglycemia led to human ß cell proliferation or apoptosis. These results demonstrate profound differences in how excess glucose or lipid influence mouse and human insulin secretion and ß cell activity and show that reduced expression of key islet-enriched transcription factors is an important mediator of glucotoxicity and lipotoxicity.


Assuntos
Regulação da Expressão Gênica , Proteínas de Homeodomínio/biossíntese , Células Secretoras de Insulina/metabolismo , Transplante das Ilhotas Pancreáticas , Fator de Transcrição MafB/biossíntese , Animais , Xenoenxertos , Proteínas de Homeodomínio/genética , Humanos , Células Secretoras de Insulina/patologia , Células Secretoras de Insulina/transplante , Fator de Transcrição MafB/genética , Camundongos , Camundongos Knockout
13.
J Clin Invest ; 125(2): 809-23, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25574839

RESUMO

Osteoclasts are bone-resorbing cells that are important for maintenance of bone remodeling and mineral homeostasis. Regulation of osteoclast differentiation and activity is important for the pathogenesis and treatment of diseases associated with bone loss. Here, we demonstrate that retinoid X receptors (RXRs) are key elements of the transcriptional program of differentiating osteoclasts. Loss of RXR function in hematopoietic cells resulted in formation of giant, nonresorbing osteoclasts and increased bone mass in male mice and protected female mice from bone loss following ovariectomy, which induces osteoporosis in WT females. The increase in bone mass associated with RXR deficiency was due to lack of expression of the RXR-dependent transcription factor v-maf musculoaponeurotic fibrosarcoma oncogene family, protein B (MAFB) in osteoclast progenitors. Evaluation of osteoclast progenitor cells revealed that RXR homodimers directly target and bind to the Mafb promoter, and this interaction is required for proper osteoclast proliferation, differentiation, and activity. Pharmacological activation of RXRs inhibited osteoclast differentiation due to the formation of RXR/liver X receptor (LXR) heterodimers, which induced expression of sterol regulatory element binding protein-1c (SREBP-1c), resulting in indirect MAFB upregulation. Our study reveals that RXR signaling mediates bone homeostasis and suggests that RXRs have potential as targets for the treatment of bone pathologies such as osteoporosis.


Assuntos
Remodelação Óssea/fisiologia , Diferenciação Celular/fisiologia , Receptores Nucleares Órfãos/metabolismo , Osteoclastos/metabolismo , Multimerização Proteica/fisiologia , Receptores X de Retinoides/metabolismo , Animais , Feminino , Receptores X do Fígado , Fator de Transcrição MafB/biossíntese , Fator de Transcrição MafB/genética , Masculino , Camundongos , Camundongos Knockout , Receptores Nucleares Órfãos/genética , Osteoclastos/citologia , Osteoporose/genética , Osteoporose/metabolismo , Receptores X de Retinoides/genética , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Transcrição Gênica/fisiologia , Regulação para Cima/fisiologia
14.
Oncogene ; 31(31): 3647-54, 2012 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-22120711

RESUMO

MafB, a member of the large Maf transcription factor family, is essential for the embryonic and terminal differentiation of pancreatic α- and ß-cells. However, the role of MafB in the control of adult islet-cell proliferation remains unknown. Considering its oncogenic potential in several other tissues, we investigated the possible alteration of its expression in adult mouse ß-cells under different conditions of proliferation. We found that MafB, in general silenced in these cells, was reexpressed in ∼30% of adaptive ß-cells both in gestational female mice and in mice fed with a high-fat diet. Importantly, reactivated MafB expression was also observed in the early ß-cell lesions and insulinomas that developed in ß-cell specific Men1 mutant mice, appearing in >80% of ß-cells in hyperplasic or dysplastic islets from the mutant mice >4 months of age. Moreover, MafB expression could be induced by glucose stimulation in INS-1 rat insulinoma cells. The induction was further reinforced following Men1 knockdown by siRNA. Furthermore, MafB overexpression in cultured ßTC3 cells enhanced cell foci formation both in culture medium and on soft agar, accompanied with the increased expression of Cyclin B1 and D2. Conversely, MafB downregulation by siRNA transfection reduced BrdU incorporation in INS-1E cells. Taken together, our data reveal that Men1 inactivation leads to MafB reexpression in mouse ß-cells in vivo, and provides evidence that deregulated ectopic MafB expression may have a hitherto unknown role in adult ß-cell proliferation and Men1-related tumorigenesis.


Assuntos
Proliferação de Células , Células Secretoras de Insulina/metabolismo , Insulinoma/metabolismo , Fator de Transcrição MafB/biossíntese , Neoplasias Pancreáticas/metabolismo , Proteínas Proto-Oncogênicas/biossíntese , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Ciclina B1/biossíntese , Ciclina D2/biossíntese , Dieta Hiperlipídica , Feminino , Glucose/farmacologia , Insulinoma/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neoplasias Pancreáticas/patologia , Gravidez , Proteínas Proto-Oncogênicas/genética , RNA Interferente Pequeno/metabolismo , Ratos
15.
J Burn Care Res ; 33(6): 747-58, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22868453

RESUMO

Studies have shown that monocytes are hyporesponsive and that dendritic cells (DCs) are depleted in burn patients. We have recently shown in a mouse model that burn injury alters the transcriptional regulation in bone marrow progenitors and inhibits myeloid-derived DC (mDC) production. In the present study, using human burn patient peripheral blood mononuclear cells, we have shown an overexpression of MafB with a corresponding reduction in peripheral blood mononuclear cell-derived mDCs. We isolated mononuclear cells from burn patient (23­68% TBSA) and control volunteer peripheral blood samples by Ficoll gradient centrifugation and cultured mDCs by using a standard ex vivo culture system. Fluorescence-activated cell sorter analysis was used to select myeloid cells based on the cell surface expression of CD45+. The mDC fraction was identified by the expression of human leukocyte antigen (HLA)-DR+CD11c+, and we found a significant reduction in HLA-DR+ leukocytes for up to 4 weeks postburn. MafB expression was then examined in HLA-DR+CD14+ monocytes. Burn injury alters the phenotype of CD14+ monocytes augmenting MafB expression and reducing their differentiation into mDCs. MafB was then silenced in ex vivo culture prior to DC differentiation by using small interfering RNA technique. MafB gene silencing improved the differentiation potential of CD14+ cells into mDCs, increasing the percentage of mDCs by >75%. Furthermore, GATA-1+ and HLA-DR+ mDCs were increased following MafB silencing. Although burn injury augments the number of peripheral blood monocytes, the frequency of mDC is reduced. This impairment is likely secondary to the down-regulation of mDC differentiation by high MafB-expressing monocytes following burn injury.


Assuntos
Queimaduras/imunologia , Células Dendríticas/imunologia , Fator de Transcrição MafB/biossíntese , Monócitos/imunologia , Adulto , Idoso , Análise de Variância , Queimaduras/metabolismo , Antígeno CD11c/metabolismo , Células Cultivadas , Feminino , Citometria de Fluxo , Fator de Transcrição GATA1/metabolismo , Antígenos HLA-DR/metabolismo , Humanos , Antígenos Comuns de Leucócito/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Masculino , Microscopia Confocal , Pessoa de Meia-Idade , Monócitos/metabolismo , Fenótipo , RNA Interferente Pequeno
16.
J Leukoc Biol ; 91(1): 69-81, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21984745

RESUMO

We have previously shown that perturbed bone marrow progenitor development promotes hyporesponsive monocytes following experimental burn sepsis. Clinical and experimental sepsis is associated with monocyte deactivation and depletion of mDCs. Decrease in circulating DCs is reported in burn patients who develop sepsis. In our 15% TBSA scald burn model, we demonstrate a significant reduction in the circulating MHC-II(+) population and mDCs (Gr1(neg)CD11b(+)CD11c(+)) with a corresponding decrease in bone marrow MHC-II(+) cells and mDCs for up to 14 days following burn. We explored the underlying mechanism(s) that regulate bone marrow development of monocytes and DCs following burn injury. We found a robust bone marrow response with a significant increase in multipotential HSCs (LSK) and bipotential GMPs following burn injury. GMPs from burn mice exhibit a significant reduction in GATA-1, which is essential for DC development, but express high levels of MafB and M-CSFRs, both associated with monocyte production. GMPs obtained from burn mice differentiated 1.7 times more into Mϕ and 1.6-fold less into DCs compared with sham. Monocytes and DCs expressed 50% less MHC-II in burn versus sham. Increased monocyte commitment in burn GMPs was a result of high MafB and M-CSFR expressions. Transient silencing of MafB (siRNA) in GMP-derived monocytes from burn mice partially restored DC differentiation deficits and increased GATA-1 expression. We provide evidence that high MafB following burn plays an inhibitory role in monocyte-derived DC differentiation by regulating M-CSFR and GATA-1 expressions.


Assuntos
Queimaduras/imunologia , Queimaduras/metabolismo , Células Dendríticas/imunologia , Inibidores do Crescimento/biossíntese , Células-Tronco Hematopoéticas/imunologia , Fator de Transcrição MafB/biossíntese , Monócitos/imunologia , Sepse/imunologia , Animais , Queimaduras/patologia , Diferenciação Celular/imunologia , Células Dendríticas/citologia , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Fator de Transcrição GATA1/genética , Fator de Transcrição GATA1/fisiologia , Inibidores do Crescimento/genética , Inibidores do Crescimento/fisiologia , Células-Tronco Hematopoéticas/citologia , Fator de Transcrição MafB/genética , Fator de Transcrição MafB/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos , Monócitos/citologia , Monócitos/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/fisiologia , Sepse/metabolismo , Sepse/patologia , Regulação para Cima/imunologia
17.
Blood ; 109(8): 3253-9, 2007 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-17158225

RESUMO

Receptor activator of nuclear factor kappaB ligand (RANKL) induces osteoclast formation from hematopoietic cells via regulation of various transcription factors. Here, we show that MafB negatively regulates RANKL-induced osteoclast differentiation. Expression levels of MafB are significantly reduced by RANKL during osteoclastogenesis. Overexpression of MafB in bone marrow-derived monocyte/macrophage lineage cells (BMMs) inhibits the formation of TRAP(+) multinuclear osteoclasts, but phagocytic activity of BMMs is retained. Furthermore, overexpression of MafB in BMMs attenuates the gene induction of NFATc1 and osteoclast-associated receptor (OSCAR) during RANKL-mediated osteoclastogenesis. In addition, MafB proteins interfere with the DNA-binding ability of c-Fos, Mitf, and NFATc1, inhibiting their transactivation of NFATc1 and OSCAR. Furthermore, reduced expression of MafB by RNAi enhances osteoclastogenesis and increases expression of NFATc1 and OSCAR. Taken together, our results suggest that MafB can act as an important modulator of RANKL-mediated osteoclastogenesis.


Assuntos
Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Macrófagos/metabolismo , Fator de Transcrição MafB/biossíntese , Osteoclastos/metabolismo , Ligante RANK/biossíntese , Animais , Células da Medula Óssea/citologia , Células Cultivadas , Regulação da Expressão Gênica/genética , Células Gigantes/citologia , Células Gigantes/metabolismo , Células-Tronco Hematopoéticas , Macrófagos/citologia , Fator de Transcrição MafB/genética , Camundongos , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/citologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ligante RANK/genética , RNA Interferente Pequeno/genética , Receptores de Superfície Celular/metabolismo , Ativação Transcricional
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA