Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Mol Endocrinol ; 67(3): 71-82, 2021 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-34223824

RESUMO

Glucose transporter type 2 (GLUT2), encoded by the SLC2A2 gene, is an essential component of glucose-stimulated insulin secretion in pancreatic islet ß-cells. Like that of the gene encoding insulin, expression of the SLC2A2 gene expression is closely linked to ß-cell functionality in rodents, but the mechanism by which ß-cell-specific expression of SLC2A2 is controlled remains unclear. In this report, to identify putative enhancer elements of the mouse Slc2a2 gene, we examined evolutional conservation of the nucleotide sequence of its genomic locus, together with ChIP-seq data of histone modifications and various transcription factors published in previous studies. Using luciferase reporter assays, we found that an evolutionarily conserved region (ECR) located approximately 40 kbp downstream of the transcription start site of Slc2a2 functions as an active enhancer in the MIN6 ß-cell line. We also found that three ß-cell-enriched transcription factors, MafA, NeuroD1, and HNF1ß, synergistically activate transcription through this 3' downstream distal enhancer (ECR3') and the proximal promoter region of the gene. Our data also indicate that the simultaneous binding of HNF1ß to its target sites within the promoter and ECR3' of Slc2a2 is indispensable for transcriptional activation, and that binding of MafA and NeuroD1 to their respective target sites within the ECR3' enhances transcription. Co-immunoprecipitation experiments suggested that MafA, NeuroD1, and HNF1ß interact with each other. Overall, these results suggest that promoter-enhancer communication through MafA, NeuroD1, and HNF1ß is critical for Slc2a2 gene expression. These findings provide clues to help elucidate the mechanism of regulation of Slc2a2 gene expression in ß-cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Regulação da Expressão Gênica , Transportador de Glucose Tipo 2/genética , Fator 1-beta Nuclear de Hepatócito/metabolismo , Células Secretoras de Insulina/metabolismo , Fatores de Transcrição Maf Maior/metabolismo , Animais , Sequência Conservada , Elementos Facilitadores Genéticos , Transportador de Glucose Tipo 2/metabolismo , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica , Elementos de Resposta , Ativação Transcricional
2.
J Mol Endocrinol ; 63(4): 297-308, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31614335

RESUMO

Insulin plays a central role in glucose homeostasis and is produced exclusively by pancreatic islet ß-cells. Insulin gene transcription is regulated by a set of ß-cell-enriched transcription factors that bind to cis-regulatory elements within the promoter region, and regulation of the insulin gene promoter is closely linked to ß-cell functionality. PIASy, a member of the PIAS family of SUMO E3 ligases, is thought to affect insulin gene transcription, but its mechanism of action is not fully understood. Here, we demonstrate that PIASy interacts with MafA and represses insulin gene promoter activity. MafA is a ß-cell-restricted basic leucine-zipper transcriptional activator that binds to the C1 element of the insulin gene promoter. In line with previous studies showing the transactivator domain of MafA is SUMOylated, PIASy enhanced the SUMOylation of MafA. However, a SUMOylation-deficient mutant of MafA was still repressed by PIASy, indicating that this modification is dispensable for repression. Using a series of MafA and PIASy mutants, we found that the basic domain of MafA and the amino-terminal region of PIASy containing the SAP domain are necessary for their interaction. In addition, SUMO-interacting motif 1 (SIM1) at the carboxyl-terminal region of PIASy was required to repress the synergistic transactivation of MafA, Pdx1, and Beta2, transcription factors playing central roles in ß-cell differentiation and function. The PINIT and SP-RING domains in the middle region of PIASy were dispensable. These findings suggest that PIASy binds to MafA through the SAP domain and negatively regulates the insulin gene promoter through a novel SIM1-dependent mechanism.


Assuntos
Insulina/genética , Fatores de Transcrição Maf Maior/metabolismo , Proteínas de Ligação a Poli-ADP-Ribose/metabolismo , Proteínas Inibidoras de STAT Ativados/metabolismo , Animais , Regulação da Expressão Gênica , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Camundongos , Proteínas de Ligação a Poli-ADP-Ribose/química , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Inibidoras de STAT Ativados/química , Domínios e Motivos de Interação entre Proteínas , Sumoilação , Transativadores/metabolismo , Ativação Transcricional , Dedos de Zinco
4.
J Biol Chem ; 293(10): 3524-3534, 2018 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-29348175

RESUMO

Insulin mRNA expression in pancreatic islet ß-cells is up-regulated by extracellular glucose concentration, but the underlying mechanism remains incompletely understood. MafA is a transcriptional activator specifically enriched in ß-cells that binds to the insulin gene promoter. Its expression is transcriptionally and posttranscriptionally regulated by glucose. Moreover, AMP-activated protein kinase (AMPK), a regulator of cellular energy homeostasis, is inhibited by high glucose, and this inhibition is essential for the up-regulation of insulin gene expression and glucose-stimulated insulin secretion (GSIS). Here we mutagenized the insulin promoter and found that the MafA-binding element C1/RIPE3b is required for glucose- or AMPK-induced alterations in insulin gene promoter activity. Under high-glucose conditions, pharmacological activation of AMPK in isolated mouse islets or MIN6 cells by metformin or 5-aminoimidazole-4-carboxamide riboside decreased MafA protein levels and mRNA expression of insulin and GSIS-related genes (i.e. glut2 and sur1). Overexpression of constitutively active AMPK also reduced MafA and insulin expression. Conversely, pharmacological AMPK inhibition by dorsomorphin (compound C) or expression of a dominant-negative form of AMPK increased MafA and insulin expression under low-glucose conditions. However, AMPK activation or inhibition did not change the expression levels of the ß-cell-enriched transcription factors Pdx1 and Beta2/NeuroD1. AMPK activation accelerated MafA protein degradation, which is not dependent on the proteasome. We also noted that MafA overexpression prevents metformin-induced decreases in insulin and GSIS-related gene expression. These findings indicate that high glucose concentrations inhibit AMPK, thereby increasing MafA protein levels and activating the insulin promoter.

5.
Exp Dermatol ; 26(11): 1039-1045, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28418611

RESUMO

Mammalian epidermis is composed of four morphologically and functionally distinct layers of keratinocytes. The innermost basal layer consists of proliferating self-renewing keratinocytes, which also undergo asymmetric cell division to differentiate into postmitotic suprabasal cells throughout life. Control of the balance between growth and differentiation of basal cells is important for epidermal homeostasis to prevent skin disorders including malignancies; however, the underlying mechanism remains to be elucidated. Recently, MafB was identified as one of the transcription factors that regulate epidermal keratinocyte differentiation. MafB is expressed in postmitotic differentiating keratinocytes, and epidermal differentiation is partially impaired in MafB-deficient mice. To further establish the roles of MafB in the epidermis in vivo, we generated mice transgenic for MafB under the control of the basal cell-specific keratin (Krt) 14 promoter. In the epidermis of transgenic mice at embryonic day 18.5, the number of proliferating Krt14-positive basal-like cells was increased, and the granular and cornified layers were thickened. Furthermore, these MafB transgenic mice developed papillomas spontaneously with age. Therefore, MafB promotes differentiation in postmitotic keratinocytes and simultaneously has potential to promote growth when ectopically expressed in undifferentiated basal keratinocytes.


Assuntos
Diferenciação Celular/genética , Epiderme/metabolismo , Queratinócitos/metabolismo , Fator de Transcrição MafB/genética , Papiloma/genética , Neoplasias Cutâneas/genética , Animais , Proliferação de Células/genética , Epiderme/patologia , Epiderme/fisiopatologia , Feminino , Homeostase/genética , Queratina-14/genética , Queratina-14/metabolismo , Queratina-15/metabolismo , Queratinócitos/fisiologia , Masculino , Camundongos , Camundongos Transgênicos , Papiloma/patologia , Regiões Promotoras Genéticas , Neoplasias Cutâneas/patologia
6.
J Invest Dermatol ; 136(9): 1848-1857, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27208706

RESUMO

Mammalian epidermis is a stratified epithelium composed of distinct layers of keratinocytes. The outermost cornified layer is a primary barrier that consists of a cornified envelope, an insoluble structure assembled by cross-linked scaffold proteins, and a surrounding mixture of lipids. Skin keratinocytes undergo a multistep differentiation process, but the mechanism underlying this process is not fully understood. We demonstrate that the transcription factor MafB is expressed in differentiating keratinocytes in mice and is transcriptionally upregulated upon human keratinocyte differentiation in vitro. In MafB-deficient mice, epidermal differentiation was partially impaired and the cornified layer was thinner than in wild-type mice. On the basis of transcriptional profiling, we detected reduced expression levels of a subset of cornified envelope genes, for example, filaggrin and repetin, in the MafB(-/-) epidermis. By contrast, the expression levels of lipid metabolism-related genes, such as Alox12e and Smpd3, increased. The upregulated genes in the MafB(-/-) epidermis were enriched for putative target genes of the transcription factors Gata3, Grhl3, and Klf4. Immunohistochemical analysis of skin biopsy samples revealed that the expression levels of filaggrin and MafB were significantly reduced in patients with human atopic dermatitis and psoriasis vulgaris. Our results indicate that MafB is a component of the gene expression program that regulates epidermal keratinocyte differentiation.


Assuntos
Diferenciação Celular/genética , Células Epidérmicas , Fator de Transcrição MafB/genética , Fatores de Transcrição/genética , Animais , Dermatite Atópica/genética , Dermatite Atópica/fisiopatologia , Regulação para Baixo , Proteínas Filagrinas , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Immunoblotting , Queratinócitos/citologia , Queratinócitos/fisiologia , Fator 4 Semelhante a Kruppel , Camundongos , Análise em Microsséries , Organogênese/genética , Psoríase/genética , Psoríase/fisiopatologia , Reação em Cadeia da Polimerase em Tempo Real , Sensibilidade e Especificidade
7.
Acta Diabetol ; 53(4): 651-60, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27017486

RESUMO

AIMS: MafA is a critical regulator of insulin expression and mature ß-cell function. MafA binds to the insulin promoter through its carboxyl-terminal basic domain-leucine zipper (bZip) region and activates transcription synergistically with the ß-cell-enriched transactivators Beta2 (NeuroD1) and Pdx1. MafA protein is highly phosphorylated in ß-cells, and phosphorylation at multiple sites within its amino-terminal region is critical for its DNA-binding and transactivating abilities, as well as for regulation of its degradation. Here, we investigated whether phosphorylation of MafA affects its interaction with Beta2. METHODS: By mutational analysis, we identified interaction domains of MafA and Beta2. Using in situ proximity ligation assay (PLA), we explored mechanism of phosphorylation-dependent binding of MafA with Beta2. We also searched for a pathophysiological condition that would induce lower levels of MafA phosphorylation. RESULTS: Mutational analysis revealed that the phosphorylation sites within the amino-terminal region of MafA were not necessary for interaction with Beta2. In situ PLA suggested that phosphorylation induces conformational or configurational changes in MafA, thereby regulating the interaction with Beta2. We also found that long-term culture of the MIN6 insulinoma cell line under high-glucose conditions resulted in a decrease in ß-cell-specific transcripts including insulin, along with a decrease in MafA phosphorylation and DNA binding. CONCLUSION: Phosphorylation of MafA plays a critical role in ß-cell function by regulating multiple functionalities, including binding to DNA, interaction with Beta2, and transactivation.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Células Secretoras de Insulina/metabolismo , Fator de Transcrição MafB/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Animais , Sítios de Ligação , Linhagem Celular , Regulação da Expressão Gênica , Insulina/metabolismo , Camundongos , Fosforilação , Regiões Promotoras Genéticas
8.
Mol Cell Endocrinol ; 411: 113-20, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-25917456

RESUMO

Haploinsufficiency of the Gata3 gene, which encodes a zinc-finger transcription factor, is associated with the disorder hypoparathyroidism, deafness, and renal dysplasia (HDR) syndrome in humans. However, the roles of Gata3 in transcriptional regulation in the parathyroid glands are not well-understood. In this study, we show that Gata3 activates transcription of parathyroid hormone (PTH), which is secreted from parathyroid glands and is critical for regulating serum calcium and phosphate homeostasis. Gata3 interacted with Gcm2 and MafB, two known transcriptional regulators of parathyroid development, and synergistically stimulated the PTH promoter. An SP1-binding element (GC box) located within the PTH-promoter proximal region was critical for activating transcription by Gata3. In addition, the ubiquitous transcription factor SP1 also interacted with Gata3 as well as MafB and Gcm2, and HDR syndrome-associated Gata3 mutants were defective in activating the PTH promoter. These results suggest that Gata3 is a critical regulator of PTH gene expression.


Assuntos
Fator de Transcrição GATA3/metabolismo , Regulação da Expressão Gênica , Fator de Transcrição MafB/metabolismo , Proteínas Nucleares/metabolismo , Hormônio Paratireóideo/genética , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Surdez/genética , Surdez/metabolismo , Células HeLa , Humanos , Hipoparatireoidismo/genética , Hipoparatireoidismo/metabolismo , Nefropatias/genética , Nefropatias/metabolismo , Mesocricetus , Hormônio Paratireóideo/metabolismo , Regiões Promotoras Genéticas , Síndrome
9.
Development ; 141(14): 2885-94, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25005477

RESUMO

Apoptosis is an important mechanism for sculpting morphology. However, the molecular cascades that control apoptosis in developing limb buds remain largely unclear. Here, we show that MafB was specifically expressed in apoptotic regions of chick limb buds, and MafB/cFos heterodimers repressed apoptosis, whereas MafB/cJun heterodimers promoted apoptosis for sculpting the shape of the limbs. Chromatin immunoprecipitation sequencing in chick limb buds identified potential target genes and regulatory elements controlled by Maf and Jun. Functional analyses revealed that expression of p63 and p73, key components known to arrest the cell cycle, was directly activated by MafB and cJun. Our data suggest that dimeric combinations of MafB, cFos and cJun in developing chick limb buds control the number of apoptotic cells, and that MafB/cJun heterodimers lead to apoptosis via activation of p63 and p73.


Assuntos
Apoptose , Extremidades/embriologia , Fator de Transcrição MafB/metabolismo , Morfogênese , Multimerização Proteica , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Animais , Sítios de Ligação , Proteínas Morfogenéticas Ósseas/metabolismo , Sobrevivência Celular , Embrião de Galinha , Galinhas , Regulação para Baixo/genética , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Botões de Extremidades/citologia , Botões de Extremidades/embriologia , Botões de Extremidades/metabolismo , Macrófagos/metabolismo , Fator de Transcrição MafB/genética , Modelos Biológicos , Morfogênese/genética , Ligação Proteica , Proteínas Proto-Oncogênicas c-fos/genética , Proteínas Proto-Oncogênicas c-jun/genética , Transdução de Sinais/genética , Fator de Transcrição AP-1/metabolismo , Tretinoína/metabolismo
10.
J Mol Endocrinol ; 47(1): 119-127, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21830322

RESUMO

MAFA is a member of the MAF family of basic leucine zipper transcription factors and is a critical regulator of insulin gene expression and islet ß-cell function. To be degraded by the proteasome, MAFA must be phosphorylated by GSK3 and MAP kinases at multiple serine and threonine residues (Ser49, Thr53, Thr57, Ser61, and Ser65) within its amino-terminal domain. In this study, we report that MAFA degradation is stimulated by PA28γ (REGγ and PSME3), a member of a family of proteasome activators that bind and activate the 20S proteasome. To date, only a few PA28γ-proteasome pathway substrates have been identified, including steroid receptor coactivator 3 (SRC3) and the cell cycle inhibitor p21(CIP1). PA28γ binds to MAFA, induces its proteasomal degradation, and thereby attenuates MAFA-driven transcriptional activation of the insulin promoter. Co-expression of GSK3 enhanced the PA28γ-mediated degradation of MAFA, but mutants that contained alanine substitutions at the MAFA phosphorylation sites did not bind PA28γ and were resistant to degradation. We also found that a PA28γ mutant (N151Y) that did not stimulate p21 degradation enhanced MAFA degradation, and another mutant (K188D) that promoted greater p21 degradation did not enhance MAFA degradation.These results suggest that PA28γ stimulates MAFA degradation through a novel molecular mechanism that is distinct from that for the degradation of p21.


Assuntos
Autoantígenos/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Insulina/genética , Fatores de Transcrição Maf Maior/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Transcrição Gênica , Animais , Autoantígenos/genética , Genes Reporter , Células HEK293 , Humanos , Luciferases de Renilla/biossíntese , Luciferases de Renilla/genética , Camundongos , Mutação de Sentido Incorreto , Células NIH 3T3 , Fosforilação , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/genética , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Proteínas Recombinantes de Fusão/genética
11.
J Mol Endocrinol ; 47(1): 119-27, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21646385

RESUMO

MAFA is a member of the MAF family of basic leucine zipper transcription factors and is a critical regulator of insulin gene expression and islet ß-cell function. To be degraded by the proteasome, MAFA must be phosphorylated by GSK3 and MAP kinases at multiple serine and threonine residues (Ser49, Thr53, Thr57, Ser61, and Ser65) within its amino-terminal domain. In this study, we report that MAFA degradation is stimulated by PA28γ (REGγ and PSME3), a member of a family of proteasome activators that bind and activate the 20S proteasome. To date, only a few PA28γ-proteasome pathway substrates have been identified, including steroid receptor coactivator 3 (SRC3) and the cell cycle inhibitor p21 (CIP1). PA28γ binds to MAFA, induces its proteasomal degradation, and thereby attenuates MAFA-driven transcriptional activation of the insulin promoter. Co-expression of GSK3 enhanced the PA28γ-mediated degradation of MAFA, but mutants that contained alanine substitutions at the MAFA phosphorylation sites did not bind PA28γ and were resistant to degradation. We also found that a PA28γ mutant (N151Y) that did not stimulate p21 degradation enhanced MAFA degradation, and another mutant (K188D) that promoted greater p21 degradation did not enhance MAFA degradation. These results suggest that PA28γ stimulates MAFA degradation through a novel molecular mechanism that is distinct from that for the degradation of p21.

12.
J Bone Miner Res ; 26(10): 2463-72, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21713993

RESUMO

Serum calcium and phosphate homeostasis is critically regulated by parathyroid hormone (PTH) secreted by the parathyroid glands. Parathyroid glands develop from the bilateral parathyroid-thymus common primordia. In mice, the expression of transcription factor Glial cell missing 2 (Gcm2) begins in the dorsal/anterior part of the primordium on embryonic day 9.5 (E9.5), specifying the parathyroid domain. The parathyroid primordium then separates from the thymus primordium and migrates to its adult location beside the thyroid gland by E15.5. Genetic ablation of gcm2 results in parathyroid agenesis in mice, indicating that Gcm2 is essential for early parathyroid organogenesis. However, the regulation of parathyroid development at later stages is not well understood. Here we show that transcriptional activator v-maf musculoaponeurotic fibrosarcoma oncogene homologue B (MafB) is developmentally expressed in parathyroid cells after E11.5. MafB expression was lost in the parathyroid primordium of gcm2 null mice. The parathyroid glands of mafB(+/-) mice were mislocalized between the thymus and thyroid. In mafB(-/-) mice, the parathyroid did not separate from the thymus. Furthermore, in mafB(-/-) mice, PTH expression and secretion were impaired; expression levels of renal cyp27b1, one of the target genes of PTH, was decreased; and bone mineralization was reduced. We also demonstrate that although Gcm2 alone does not stimulate the PTH gene promoter, it associates with MafB to synergistically activate PTH expression. Taken together, our results suggest that MafB regulates later steps of parathyroid development, that is, separation from the thymus and migration toward the thyroid. MafB also regulates the expression of PTH in cooperation with Gcm2.


Assuntos
Fator de Transcrição MafB/metabolismo , Proteínas Nucleares/metabolismo , Glândulas Paratireoides/embriologia , Hormônio Paratireóideo/metabolismo , Fatores de Transcrição/metabolismo , Animais , Sequência de Bases , Primers do DNA , Ensaio de Desvio de Mobilidade Eletroforética , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Hormônio Paratireóideo/sangue , Reação em Cadeia da Polimerase em Tempo Real
13.
J Biol Chem ; 286(12): 10449-56, 2011 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-21278380

RESUMO

Pancreatic ß-cell-restricted expression of insulin is established through several critical cis-regulatory elements located in the insulin gene promoter region. The principal cis elements are A-boxes, E1, and C1/RIPE3b. The ß-cell-enriched transcription factors Pdx1 and Beta2 bind to the A-boxes and E1 element, respectively. A ß-cell-specific trans-acting factor binding to C1/RIPE3b (termed RIPE3b1 activator) was detected by electrophoretic mobility shift assay and has been identified as MafA, a member of the Maf family of basic leucine zipper (bZip) proteins. Here, ATF2, a member of the ATF/CREB family of basic leucine zipper proteins, was identified as a component of the RIPE3b1 activator. ATF2 alone was unable to bind to the C1/RIPE3b element but acquired binding capacity upon complex formation with MafA. ATF2 also interacted with Pdx1 and Beta2, and co-expression of ATF2, MafA, Pdx1, and Beta2 resulted in a synergistic activation of the insulin promoter. Immunohistochemical analysis of mouse pancreas tissue sections showed that ATF2 is enriched in islet endocrine cells, including ß-cells. RNAi-mediated knockdown of MafA or ATF2 in the MIN6 ß-cell line resulted in a significant decrease in endogenous levels of insulin mRNA. These data indicate that ATF2 is an essential component of the positive regulators of the insulin gene expression.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Proteínas de Homeodomínio/metabolismo , Células Secretoras de Insulina/metabolismo , Insulina/biossíntese , Fatores de Transcrição Maf Maior/metabolismo , Transativadores/metabolismo , Transcrição Gênica/fisiologia , Fator 2 Ativador da Transcrição/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Regulação da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Células HeLa , Proteínas de Homeodomínio/genética , Humanos , Insulina/genética , Células Secretoras de Insulina/citologia , Fatores de Transcrição Maf Maior/genética , Camundongos , Células NIH 3T3 , Ligação Proteica , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Elementos de Resposta/fisiologia , Transativadores/genética
14.
Diabetes ; 59(10): 2579-87, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20682694

RESUMO

OBJECTIVE: Tissue-specific self-antigens are ectopically expressed within the thymus and play an important role in the induction of central tolerance. Insulin is expressed in both pancreatic islets and the thymus and is considered to be the primary antigen for type 1 diabetes. Here, we report the role of the insulin transactivator MafA in the expression of insulin in the thymus and susceptibility to type 1 diabetes. RESEARCH DESIGN AND METHODS: The expression profiles of transcriptional factors (Pdx1, NeuroD, Mafa, and Aire) in pancreatic islets and the thymus were examined in nonobese diabetic (NOD) and control mice. Thymic Ins2 expression and serum autoantibodies were examined in Mafa knockout mice. Luciferase reporter assay was performed for newly identified polymorphisms of mouse Mafa and human MAFA. A case-control study was applied for human MAFA polymorphisms. RESULTS: Mafa, Ins2, and Aire expression was detected in the thymus. Mafa expression was lower in NOD thymus than in the control and was correlated with Ins2 expression. Targeted disruption of MafA reduced thymic Ins2 expression and induced autoantibodies against pancreatic islets. Functional polymorphisms of MafA were newly identified in NOD mice and humans, and polymorphisms of human MAFA were associated with susceptibility to type 1 diabetes but not to autoimmune thyroid disease. CONCLUSIONS: These data indicate that functional polymorphisms of MafA are associated with reduced expression of insulin in the thymus and susceptibility to type 1 diabetes in the NOD mouse as well as human type 1 diabetes.


Assuntos
Diabetes Mellitus Tipo 1/genética , Insulina/genética , Fatores de Transcrição Maf Maior/fisiologia , Animais , Proteínas de Ligação a DNA/genética , Feminino , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Predisposição Genética para Doença , Humanos , Imuno-Histoquímica , Ilhotas Pancreáticas/citologia , Ilhotas Pancreáticas/fisiologia , Fatores de Transcrição Maf Maior/deficiência , Fatores de Transcrição Maf Maior/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Regiões Promotoras Genéticas , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timo/fisiologia , Fatores de Transcrição/genética , Transcrição Gênica , Proteína AIRE
15.
Genes Cells ; 15(9): 971-82, 2010 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-20718938

RESUMO

Dysregulated expression of Maf proteins (namely c-Maf, MafA and MafB) leads to multiple myeloma in humans and oncogenic transformation of chicken embryonic fibroblasts. Maf proteins are transcriptional activators of tissue-specific gene expression and regulators of cell differentiation. For example, MafA is a critical regulator of crystallin genes and the lens differentiation program in chickens. In mammals, MafA is essential for the development of mature insulin-producing beta-cells of pancreas. It has been shown that MafA protein stability is regulated by phosphorylations at multiple serine and threonine residues. Here, we report that Maf proteins are also post-translationally modified by small ubiquitin-like modifier (SUMO) proteins at a conserved lysine residue in the amino-terminal transactivator domain. A SUMOylation-deficient mutant of MafA (K32R) was more potent than wild-type MafA in transactivating luciferase reporter construct driven by alphaA-crystallin or insulin gene promoter. In ovo electroporation into developing chicken embryo showed that the K32R mutant induced ectopic delta-crystallin gene expression more efficiently than the wild-type MafA. We also demonstrated that the K32R mutant had enhanced ability to induce colony formation of a chicken fibroblast cell line DF-1. Therefore, SUMOylation is a functional post-translational modification of MafA that negatively regulates its transcriptional and transforming activities.


Assuntos
Transformação Celular Neoplásica/genética , Fatores de Transcrição Maf Maior/genética , Sumoilação , Transcrição Gênica/genética , Animais , Linhagem Celular , Linhagem Celular Tumoral , Embrião de Galinha , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Células HeLa , Humanos , Lisina/genética , Lisina/metabolismo , Fatores de Transcrição Maf Maior/metabolismo , Fator de Transcrição MafB/genética , Fator de Transcrição MafB/metabolismo , Camundongos , Mutação , Células NIH 3T3 , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/genética , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Transfecção , delta-Cristalinas/genética
16.
J Dermatol Sci ; 57(3): 178-82, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20060689

RESUMO

BACKGROUND: The hair follicle of mammalian skin consists of a group of concentric epithelial cell layers. The inner root sheath (IRS), which surrounds the hardening hair shaft beneath the skin surface, is subdivided into three layers, termed the cuticle of the IRS, Huxley's layer, and Henle's layer. The IRS forms a follicular wall in the hair canal and helps guide the developing hair shaft. c-Maf and MafB, members of the Maf family of transcription factors, play important roles in the developmental processes of various tissues and in cell type-specific gene expression. OBJECTIVE: The aim of this study is to reveal the pattern of expression and functional roles of c-Maf and MafB in the hair follicle. METHODS: We determined the precise location of c-Maf and MafB expression using immunofluorescent staining of mouse skin sections with layer-specific markers. We also analyzed whiskers of c-maf- and mafB-null mice (c-maf(-/-) and mafB(-/-), respectively) using scanning electron microscopy. RESULTS: c-Maf and MafB were differentially expressed in the Huxley's and Henle's layers of the IRS. Scanning electron microscopic analysis showed irregular cuticle patterning of whiskers of c-maf(-/-) and mafB(-/-) mice. The cuticles of mafB(-/-) mice were also thinner than those of wild-type mice. CONCLUSION: c-Maf and MafB are expressed in the IRS layers in a lineage-restricted manner and are involved in hair morphogenesis.


Assuntos
Folículo Piloso/crescimento & desenvolvimento , Folículo Piloso/metabolismo , Fator de Transcrição MafB/metabolismo , Proteínas Proto-Oncogênicas c-maf/metabolismo , Vibrissas/metabolismo , Animais , Camundongos , Camundongos Endogâmicos C57BL , Morfogênese , RNA Mensageiro/metabolismo , Vibrissas/anormalidades , Vibrissas/ultraestrutura
17.
Toxicol Sci ; 109(1): 143-51, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19240039

RESUMO

Phthalate esters are commonly used plasticizers; however, some are suspected to cause reproductive toxicity. Administration of high doses of di-(2-ethylhexyl) phthalate (DEHP) induces germ cell death in male rodents. Mono-(2-ethylhexyl) phthalate (MEHP), a hydrolyzed metabolite of DEHP, appears to be responsible for this testicular toxicity; however, the underlying mechanism of this chemical's action remains unknown. Here, using a one-step affinity purification procedure, we identified glycogen debranching enzyme (GDE) as a phthalate-binding protein. GDE has oligo-1,4-1,4-glucanotransferase and amylo-1,6-glucosidase activities, which are responsible for the complete degradation of glycogen to glucose. Our findings demonstrate that MEHP inhibits the activity of oligo-1,4-1,4-glucanotransferase, but not of amylo-1,6-glucosidase. Among various phthalate esters tested, MEHP specifically binds to and inhibits GDE. We also show that DEHP administration affects glycogen metabolism in rat testis. Thus, inhibition of GDE by MEHP may play a role in germ cell apoptosis in the testis.


Assuntos
Dietilexilftalato/análogos & derivados , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Glicogenólise/efeitos dos fármacos , Testículo/efeitos dos fármacos , Análise de Variância , Animais , Células COS , Chlorocebus aethiops , Dietilexilftalato/metabolismo , Dietilexilftalato/toxicidade , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Cinética , Fígado/metabolismo , Masculino , Metabolômica , Ligação Proteica , Ratos , Ratos Wistar , Espermatozoides/metabolismo , Testículo/metabolismo
18.
J Biotechnol ; 135(4): 385-92, 2008 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-18588926

RESUMO

Viral capsids of simian virus 40 (SV40) are highly efficient gene delivery vehicles that infect a broad range of cells and tissues. To develop a controlled, cell type-specific delivery system, we sought to display foreign peptides on the capsid surface by genetically manipulating the major capsid protein Vp1. Here we report the identification of two sites within the surface loops of Vp1 that can accommodate foreign peptides in such a way that the foreign peptides are displayed on the surface of the virus-like particles (VLPs) without interfering with VLP assembly or the packaging of viral DNA. Insertion of Flag-tags but not RGD integrin-binding motifs at these sites strongly inhibited cell attachment of VLPs, which normally associate with host cells through cell surface molecules such as major histocompatibility complex (MHC) class I and ganglioside GM1. Instead, VLPs carrying the RGD motifs bound to integrin in vitro and to the cell surface in an RGD-dependent manner. Thus, insertion of foreign sequences into the surface loops of Vp1 can reduce natural virus-cell interactions and even confer an ability to bind to a new target receptor. This study demonstrates the potential usefulness of this strategy for the development of novel delivery vehicles with different cell tropisms.


Assuntos
Peptídeos/metabolismo , Vírus 40 dos Símios/metabolismo , Vírion/metabolismo , Motivos de Aminoácidos , Sequência de Aminoácidos , Animais , Células COS , Proteínas do Capsídeo/metabolismo , Adesão Celular , Chlorocebus aethiops , Empacotamento do DNA , DNA Viral/metabolismo , Glicina , Integrina alfaVbeta3/metabolismo , Dados de Sequência Molecular , Mutagênese Insercional , Proteínas Mutantes/metabolismo , Oligopeptídeos , Estrutura Secundária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Virais de Fusão/química , Proteínas Virais de Fusão/metabolismo
19.
J Biotechnol ; 134(1-2): 181-92, 2008 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-18243389

RESUMO

The capsid of SV40 is regarded as a potential nano-capsule for delivery of biologically active materials. The SV40 capsid is composed of 72 pentamers of the VP1 major capsid protein and 72 copies of the minor coat proteins VP2/3. We have previously demonstrated that, when expressed in insect Sf9 cells by the baculovirus system, VP1 self-assembles into virus-like particles (VP1-VLPs), which are morphologically indistinguishable from the SV40 virion and can be easily purified. Here, we show that heterologous proteins fused to VP2/3 can be efficiently incorporated into the VP1-VLPs. Using EGFP as a model protein, we have optimized this encapsulation system and found that fusion to the C-terminus of VP2/3 is preferable and that the C-terminal VP1-interaction domain of VP2/3 is sufficient for incorporation into VLPs. The VLPs encapsulating EGFP retain the ability to attach to the cell surface and enter the cells. Using this system, we have encapsulated yeast cytosine deaminase (yCD), a prodrug-modifying enzyme that converts 5-fluorocytosine to 5-fluorouracil, into VLPs. When CV-1 cells are challenged by the yCD-encapsulating VLPs, they become sensitive to 5-fluorocytosine-induced cell death. Therefore, proteins of interest can be encapsulated in VP1-VLPs by fusion to VP2/3 and successfully delivered to cells.


Assuntos
Proteínas do Capsídeo/genética , Nanotecnologia/métodos , Vírus 40 dos Símios/genética , Baculoviridae/genética , Baculoviridae/ultraestrutura , Proteínas do Capsídeo/metabolismo , Citosina Desaminase/genética , Citosina Desaminase/metabolismo , Engenharia Genética/métodos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Microscopia Confocal , Microscopia Eletrônica , Modelos Biológicos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Vírus 40 dos Símios/ultraestrutura
20.
Genes Cells ; 12(11): 1267-79, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17986010

RESUMO

The simian virus 40 (SV40) particle is mainly composed of the major capsid protein termed VP1. VP1 self-assembles into virus-like particles (VLPs) of approximately 40 nm in diameter when over-expressed in bacteria or in insect cells, but purified VP1 does not form such a structure under physiological conditions, and thus, the mechanism of VP1 assembly is not well understood. Using a highly purified VP1 assembly/disassembly system in vitro, here we provide evidence that DNA is a factor that contributes to VP1 assembly into 40-nm spherical particles. At pH 5, for example, VP1 preferentially assembles into 40-nm particles in the presence of DNA, whereas VP1 assembles into tubular structures in the absence of DNA. Electron microscopic observations revealed that the concentration of DNA and its length are important for the formation of 40-nm particles. In addition, sucrose gradient sedimentation analysis and DNase I-sensitivity assays indicated that DNA of up to 2,000 bp is packaged into the 40-nm particles under the conditions examined. We propose that DNA may facilitate the formation of 40-nm spherical particles by acting as a scaffold that increases the local concentration of VP1 and/or by acting as an allosteric effector that alters the structure of VP1.


Assuntos
Proteínas do Capsídeo/metabolismo , DNA Circular/metabolismo , Vírus 40 dos Símios/fisiologia , Vírion/fisiologia , Montagem de Vírus/fisiologia , Cálcio/metabolismo , Proteínas do Capsídeo/ultraestrutura , DNA Circular/ultraestrutura , Concentração de Íons de Hidrogênio , Microscopia Eletrônica , Sódio/metabolismo , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...