Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Cell Mol Life Sci ; 78(4): 1781-1798, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32797246

RESUMO

Zinc has been known to be essential for cell division for over 40 years but the molecular pathways involved remain elusive. Cellular zinc import across biological membranes necessitates the help of zinc transporters such as the SLC39A family of ZIP transporters. We have discovered a molecular process that explains why zinc is required for cell division, involving two highly regulated zinc transporters, as a heteromer of ZIP6 and ZIP10, providing the means of cellular zinc entry at a specific time of the cell cycle that initiates a pathway resulting in the onset of mitosis. Crucially, when the zinc influx across this heteromer is blocked by ZIP6 or ZIP10 specific antibodies, there is no evidence of mitosis, confirming the requirement for zinc influx as a trigger of mitosis. The zinc that influxes into cells to trigger mitosis additionally changes the phosphorylation state of STAT3 converting it from a transcription factor to a protein that complexes with this heteromer and pS38Stathmin, the form allowing microtubule rearrangement as required in mitosis. This discovery now explains the specific cellular role of ZIP6 and ZIP10 and how they have special importance in the mitosis process compared to other ZIP transporter family members. This finding offers new therapeutic opportunities for inhibition of cell division in the many proliferative diseases that exist, such as cancer.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte de Cátions/genética , Mitose/genética , Fator de Transcrição STAT3/genética , Regulação da Expressão Gênica , Humanos , Células MCF-7 , Fosforilação/genética , Multimerização Proteica/genética , Transdução de Sinais/genética , Zinco/química , Zinco/metabolismo
2.
PLoS Genet ; 8(6): e1002766, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22737083

RESUMO

Mutations in the human Zip4 gene cause acrodermatitis enteropathica, a rare, pseudo-dominant, lethal genetic disorder. We created a tamoxifen-inducible, enterocyte-specific knockout of this gene in mice which mimics this human disorder. We found that the enterocyte Zip4 gene in mice is essential throughout life, and loss-of-function of this gene rapidly leads to wasting and death unless mice are nursed or provided excess dietary zinc. An initial effect of the knockout was the reprogramming of Paneth cells, which contribute to the intestinal stem cell niche in the crypts. Labile zinc in Paneth cells was lost, followed by diminished Sox9 (sex determining region Y-box 9) and lysozyme expression, and accumulation of mucin, which is normally found in goblet cells. This was accompanied by dysplasia of the intestinal crypts and significantly diminished small intestine cell division, and attenuated mTOR1 activity in villus enterocytes, indicative of increased catabolic metabolism, and diminished protein synthesis. This was followed by disorganization of the absorptive epithelium. Elemental analyses of small intestine, liver, and pancreas from Zip4-intestine knockout mice revealed that total zinc was dramatically and rapidly decreased in these organs whereas iron, manganese, and copper slowly accumulated to high levels in the liver as the disease progressed. These studies strongly suggest that wasting and lethality in acrodermatitis enteropathica patients reflects the loss-of-function of the intestine zinc transporter ZIP4, which leads to abnormal Paneth cell gene expression, disruption of the intestinal stem cell niche, and diminished function of the intestinal mucosa. These changes, in turn, cause a switch from anabolic to catabolic metabolism and altered homeostasis of several essential metals, which, if untreated by excess dietary zinc, leads to dramatic weight loss and death.


Assuntos
Acrodermatite/genética , Proteínas de Transporte de Cátions/genética , Mucosa Intestinal , Intestinos , Nicho de Células-Tronco , Zinco , Acrodermatite/patologia , Animais , Proteínas de Transporte de Cátions/metabolismo , Modelos Animais de Doenças , Enterócitos/metabolismo , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patologia , Metais/metabolismo , Camundongos , Camundongos Knockout , Celulas de Paneth/metabolismo , Fatores de Transcrição SOX9/metabolismo , Nicho de Células-Tronco/genética , Serina-Treonina Quinases TOR/metabolismo , Zinco/deficiência , Zinco/metabolismo
3.
Nucleic Acids Res ; 40(11): 4850-60, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22362755

RESUMO

Dnmt1 is frequently overexpressed in cancers, which contributes significantly to cancer-associated epigenetic silencing of tumor suppressor genes. However, the mechanism of Dnmt1 overexpression remains elusive. Herein, we elucidate a pathway through which nuclear receptor SHP inhibits zinc-dependent induction of Dnmt1 by antagonizing metal-responsive transcription factor-1 (MTF-1). Zinc treatment induces Dnmt1 transcription by increasing the occupancy of MTF-1 on the Dnmt1 promoter while decreasing SHP expression. SHP in turn represses MTF-1 expression and abolishes zinc-mediated changes in the chromatin configuration of the Dnmt1 promoter. Dnmt1 expression is increased in SHP-knockout (sko) mice but decreased in SHP-transgenic (stg) mice. In human hepatocellular carcinoma (HCC), increased DNMT1 expression is negatively correlated with SHP levels. Our study provides a molecular explanation for increased Dnmt1 expression in HCC and highlights SHP as a potential therapeutic target.


Assuntos
Carcinoma Hepatocelular/genética , DNA (Citosina-5-)-Metiltransferases/genética , Proteínas de Ligação a DNA/metabolismo , Neoplasias Hepáticas/genética , Receptores Citoplasmáticos e Nucleares/metabolismo , Proteínas Repressoras/metabolismo , Fatores de Transcrição/metabolismo , Zinco/farmacologia , Animais , Carcinoma Hepatocelular/enzimologia , Linhagem Celular , Linhagem Celular Tumoral , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/biossíntese , DNA (Citosina-5-)-Metiltransferases/metabolismo , Proteínas de Ligação a DNA/antagonistas & inibidores , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Hepatócitos/enzimologia , Humanos , Fígado/enzimologia , Neoplasias Hepáticas/enzimologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Receptores Citoplasmáticos e Nucleares/genética , Fatores de Transcrição/antagonistas & inibidores , Transcrição Gênica/efeitos dos fármacos , Fator MTF-1 de Transcrição
4.
Biochim Biophys Acta ; 1809(1): 56-62, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21035574

RESUMO

Metallothionein (MT) is a small, cysteine-rich protein active in zinc homeostasis, cadmium detoxification, and protection against reactive oxygen species. Mouse MT-I gene transcription is regulated by metal response element-binding transcription factor-1 (MTF-1), which is recruited to the promoter by zinc. We examined alterations in the chromatin structure of the MT-I promoter associated with enhanced transcriptional activation. MTF-1 proved essential for zinc-induced epigenetic changes in the MT-I promoter. Chromatin immunoprecipitation assays demonstrated that zinc treatment rapidly decreased Lys4-trimethylated and Lys9-acetylated histone H3 in the promoter and decreased total histone H3 but not histone H3.3. Micrococcal nuclease sensitivity of the MT-I promoter was increased by zinc. Thus, the chromatin structure in the promoter may be locally disrupted by zinc-induced nucleosome removal. Without MTF-1 these changes were not observed, and an MTF-1 deletion mutant recruited to the MT-I promoter by zinc that did not recruit the coactivator p300 or activate MT-I transcription did not affect histone H3 in the MT-I promoter in response to zinc. Interleukin-6, which induces MT-I transcription independently of MTF-1, did not reduce histone H3 levels in the promoter. Rapid disruption of nucleosome structure at the MT-I promoter is mediated by zinc-responsive recruitment of an active MTF-1-coactivator complex.


Assuntos
Cromatina/metabolismo , Proteínas de Ligação a DNA/metabolismo , Metalotioneína/genética , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Acetilação/efeitos dos fármacos , Animais , Sítios de Ligação/genética , Western Blotting , Células Cultivadas , Cromatina/genética , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/genética , Embrião de Mamíferos/citologia , Epigênese Genética/efeitos dos fármacos , Epigenômica , Fibroblastos/citologia , Fibroblastos/metabolismo , Histonas/genética , Histonas/metabolismo , Interleucina-6/farmacologia , Metilação/efeitos dos fármacos , Camundongos , Camundongos Knockout , Mutação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Transcrição/genética , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/genética , Zinco/metabolismo , Zinco/farmacologia , Fator MTF-1 de Transcrição
5.
PLoS One ; 5(10)2010 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-20957146

RESUMO

BACKGROUND: The zinc transporter ZIP4 (Slc39a4) is important for proper mammalian development and is an essential gene in mice. Recent studies suggest that this gene may also play a role in pancreatic cancer. METHODS/PRINCIPAL FINDINGS: Herein, we present evidence that this essential zinc transporter is expressed in hepatocellular carcinomas. Zip4 mRNA and protein were dramatically elevated in hepatocytes in the majority of human hepatocellular carcinomas relative to noncancerous surrounding tissues, as well as in hepatocytes in hepatocellular carcinomas occurring in farnesoid X receptor-knockout mice. Interestingly, meta-analysis of microarray data in the Geo and Oncomine databases suggests that Zip4 mRNA may also be elevated in many types of cancer. Potential mechanisms of action of ZIP4 were examined in cultured cell lines. RNAi knockdown of Zip4 in mouse Hepa cells significantly increased apoptosis and modestly slowed progression from G(0)/G(1) to S phase when cells were released from hydroxyurea block into zinc-deficient medium. Cell migration assays revealed that RNAi knockdown of Zip4 in Hepa cells depressed in vitro migration whereas forced over-expression in Hepa cells and MCF-7 cells enhanced in vitro migration. CONCLUSIONS: ZIP4 may play a role in the acquisition of zinc by hepatocellular carcinomas, and potentially many different cancerous cell-types, leading to repressed apoptosis, enhanced growth rate and enhanced invasive behavior.


Assuntos
Apoptose/genética , Proteínas de Transporte de Cátions/genética , Ciclo Celular/genética , Movimento Celular/genética , Neoplasias Hepáticas Experimentais/patologia , Animais , Northern Blotting , Humanos , Neoplasias Hepáticas Experimentais/genética , Camundongos , Metástase Neoplásica , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Zinco/deficiência
6.
Am J Physiol Regul Integr Comp Physiol ; 297(1): R194-201, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19458277

RESUMO

The lactating mammary gland is composed of multiple cell types that tightly coordinate the accumulation, production, and secretion of milk components, including essential metals such as zinc (Zn). Our previous studies in animal and cell models implicated the Zn transporter Zip3 (Slc39a3) in mammary gland Zn acquisition. Herein, we investigated this hypothesis directly by utilizing Zip3-null mice. Our data verify that Zip3 is expressed in secretory mammary cells; however, Zip3 does not play a major role in Zn import from the maternal circulation. Importantly, the primary localization of Zip3 was associated with the luminal membrane of the secretory mammary cells. Consistent with this localization, Zn transfer studies using (65)Zn revealed that Zn retention in the secreted milk pool and milk Zn concentration was higher in Zip3-null compared with wild-type mice. Although total mammary gland Zn concentration was not altered, Zip3-null mice also had altered mammary tissue architecture, increased number of apoptotic cells, and reduced mammary gland weight implicating subtle changes in Zip3-mediated intracellular Zn pools in apoptosis regulation. Taken together, our data indicate that Zip3 does not participate in the acquisition of Zn from maternal circulation for secretion into milk but, in contrast, primarily plays a role in the reuptake and cellular retention of Zn in the mammary gland from the previously secreted milk pool, thus regulating cellular function.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Células Epiteliais/metabolismo , Lactação/metabolismo , Glândulas Mamárias Animais/metabolismo , Leite/metabolismo , Zinco/metabolismo , Animais , Animais Lactentes , Apoptose , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Membrana Celular/metabolismo , Células Epiteliais/patologia , Feminino , Genótipo , Transporte de Íons , Glândulas Mamárias Animais/patologia , Camundongos , Camundongos Knockout , Fenótipo , Zinco/sangue , Radioisótopos de Zinco
7.
Mol Cancer Res ; 6(3): 483-90, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18337454

RESUMO

Mammalian metallothionein (MT) genes are transcriptionally activated by the essential metal zinc as well as by environmental stresses, including toxic metal overload and redox fluctuations. In addition to playing a key role in zinc homeostasis, MT proteins can protect against metal- and oxidant-induced cellular damage, and may participate in other fundamental physiologic and pathologic processes such as cell survival, proliferation, and neoplasia. Previously, our group reported a requirement for metal-responsive transcription factor-1 (MTF-1) in hypoxia-induced transcription of mouse MT-I and human MT-IIA genes. Here, we provide evidence that the protumorigenic hypoxia-inducible transcription factor-1alpha (HIF-1alpha) is essential for induction of MT-1 by hypoxia, but not zinc. Chromatin immunoprecipitation assays revealed that MTF-1 and HIF-1alpha are both recruited to the mouse MT-I promoter in response to hypoxia, but not zinc. In the absence of HIF-1alpha, MTF-1 is recruited to the MT-I promoter but fails to activate MT-I gene expression in response to hypoxia. Thus, HIF-1alpha seems to function as a coactivator of MT-I gene transcription by interacting with MTF-1 during hypoxia. Coimmunoprecipitation studies suggest interaction between MTF-1 and HIF-1alpha, either directly or as mediated by other factors. It is proposed that association of these important transcription factors in a multiprotein complex represents a common strategy to control unique sets of hypoxia-inducible genes in both normal and diseased tissue.


Assuntos
Hipóxia Celular/fisiologia , Proteínas de Ligação a DNA/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Metalotioneína/genética , Fatores de Transcrição/fisiologia , Células 3T3 , Animais , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular Transformada , Deleção de Genes , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Metalotioneína/biossíntese , Camundongos , Interferência de RNA , Zinco/farmacologia , Fator MTF-1 de Transcrição
8.
Genesis ; 45(6): 339-52, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17506078

RESUMO

Fourteen members of the Slc39a superfamily of metal ion uptake transporters have been identified in mice and humans, but the physiological functions of most remain obscure. Herein, we created mice with Zip2 (Slc39a2) genes in which the open reading frame was replaced with that of the enhanced green fluorescent protein (EGFP), to study temporal and spatial patterns of Zip2 gene expression and examine the physiological roles of this transporter. Expression of this gene was remarkably cell-type specific and developmentally regulated in pericentral hepatocytes, developing keratinocytes, and a subset of immature dendritic cells in the immune system. In addition, the Zip2 gene was transiently expressed in giant trophoblast cells in the placenta. Although the Zip2 gene was not essential under conditions of normal dietary zinc, it played an important role in adapting to dietary zinc deficiency during pregnancy, and in the homeostasis of iron in the liver as well as iron and calcium in developing embryos. These studies suggest that active expression of the Zip2 gene in these few specific cell types, aforementioned, plays a particularly important role during zinc deficiency. These studies further reveal novel interactions between zinc transporter function and the homeostasis of other essential metals.


Assuntos
Proteínas de Transporte de Cátions/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Zinco/metabolismo , Animais , Cálcio/metabolismo , Proteínas de Transporte de Cátions/genética , Células Dendríticas/química , Células Dendríticas/metabolismo , Feminino , Expressão Gênica , Marcação de Genes , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Hepatócitos/química , Hepatócitos/metabolismo , Homeostase , Ferro/metabolismo , Queratinócitos/química , Queratinócitos/metabolismo , Camundongos , Camundongos Knockout , Gravidez , Trofoblastos/química , Trofoblastos/metabolismo
9.
Mol Cell Biol ; 26(15): 5580-7, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847313

RESUMO

Mouse metal response element-binding transcription factor-1 (MTF-1) regulates the transcription of genes in response to a variety of stimuli, including exposure to zinc or cadmium, hypoxia, and oxidative stress. Each of these stresses may increase labile cellular zinc, leading to nuclear translocation, DNA binding, and transcriptional activation of metallothionein genes (MT genes) by MTF-1. Several lines of evidence suggest that the highly conserved six-zinc finger DNA-binding domain of MTF-1 also functions as a zinc-sensing domain. In this study, we investigated the potential role of the peptide linkers connecting the four N-terminal zinc fingers of MTF-1 in their zinc-sensing function. Each of these three linkers is unique, completely conserved among all known vertebrate MTF-1 orthologs, and different from the canonical Cys2His2 zinc finger TGEKP linker sequence. Replacing the RGEYT linker between zinc fingers 1 and 2 with TGEKP abolished the zinc-sensing function of MTF-1, resulting in constitutive DNA binding, nuclear translocation, and transcriptional activation of the MT-I gene. In contrast, swapping the TKEKP linker between fingers 2 and 3 with TGEKP had little effect on the metal-sensing functions of MTF-1, whereas swapping the canonical linker for the shorter TGKT linker between fingers 3 and 4 rendered MTF-1 less sensitive to zinc-dependent activation both in vivo and in vitro. These observations suggest a mechanism by which physiological concentrations of accessible cellular zinc affect MTF-1 activity. Zinc may modulate highly specific, linker-mediated zinc finger interactions in MTF-1, thus affecting its zinc- and DNA-binding activities, resulting in translocation to the nucleus and binding to the MT-I gene promoter.


Assuntos
Proteínas de Ligação a DNA/química , Proteínas de Ligação a DNA/metabolismo , Peptídeos/metabolismo , Fatores de Transcrição/química , Fatores de Transcrição/metabolismo , Dedos de Zinco , Zinco/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Regulação da Expressão Gênica , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Peptídeos/química , Peptídeos/genética , Regiões Promotoras Genéticas , Fatores de Transcrição/genética , Fator MTF-1 de Transcrição
10.
J Biol Chem ; 280(31): 28529-40, 2005 Aug 05.
Artigo em Inglês | MEDLINE | ID: mdl-16055450

RESUMO

Six Cys(2)His(2) zinc fingers (F1-6) comprise the DNA binding domain of metal-responsive element binding transcription factor-1 (MTF-1). F1-6 is necessary for basal and zinc-induced expression of metallothionein genes. Analysis of NMR structural and dynamic data for an F1-6 protein construct demonstrates that each zinc finger adopts a stable betabetaalpha fold in the presence of stoichiometric Zn(II), provided that all cysteine ligands are in a reduced state. Parallel studies of protein constructs spanning the four N-terminal core DNA binding fingers (F1-4) and two C-terminal low DNA affinity fingers (F5-6) reveal similar stable zinc finger structures. In both the F1-6 and F5-6 proteins, the finger 5 cysteines were found to readily oxidize at neutral pH. Detailed spectral density and hydrodynamic analysis of (15)N relaxation data revealed quasi-ordered anisotropic rotational diffusion properties of the six F1-6 zinc fingers that could influence MTF-1 DNA binding function. A more general effect on the rotational diffusion properties of Cys(2)His(2) zinc fingers was also uncovered that is dependent upon the position of each finger within multifinger domains. Analysis of NMR (1)H-(15)N-heteronuclear single quantum coherence spectral peak intensities measured as a function of added Zn(II) in conjunction with Zn(II) binding modeling studies indicated that the Zn(II) affinities of all MTF-1 zinc fingers are within approximately 10-50-fold. These analyses further suggested that metal sensing by MTF-1 in eukaryotic cells involves multiple zinc fingers and occurs over a 100-fold or less range of accessible Zn(II) concentration.


Assuntos
Fatores de Transcrição/metabolismo , Dedos de Zinco , Zinco/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Proteínas de Ligação a DNA , Cinética , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Conformação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fator MTF-1 de Transcrição
11.
Biochem J ; 382(Pt 1): 33-41, 2004 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-15142038

RESUMO

Post-translational modification of MTF-1 (metal-response-element-binding transcription factor-1) was suggested to play a role in its metalloregulatory functions. In the present study, pulse labelling and two-dimensional electrophoresis-Western blotting were used to demonstrate that, although MTF-1 is highly modified in vivo, its phosphorylation level does not rapidly change in response to metals, nor does its overall modification pattern. Recombinant MTF-1 was found to serve as an in vitro substrate for casein kinase II, c-Jun N-terminal kinase and protein kinase C, but inhibition of these kinases in vivo did not significantly change the modification pattern of MTF-1. Northern blotting revealed that inhibitors of casein kinase II and c-Jun N-terminal kinase severely attenuate the metal-induced transcription of the native chromatin-packaged metallothionein-I and zinc transporter-1 genes, whereas protein kinase C inhibitors exerted gene- and cell-type-specific effects on the metal regulation and basal expression of these two genes. A chromatin immunoprecipitation assay was used to demonstrate that none of these inhibitors prevent the metal-dependent recruitment of MTF-1 to the MT-I promoter. In brief, results of the present study suggest that protein kinases may not alter the phosphorylation state of MTF-1 during the rapid-response phase to metals, nor do they regulate the metal-dependent formation of a stable MTF-1-chromatin complex. Instead, protein kinases may exert their interdependent effects on metal-induced gene expression by acting on cofactors that interact with MTF-1.


Assuntos
Cádmio/fisiologia , Transdução de Sinais/genética , Fatores de Transcrição/genética , Transcrição Gênica/genética , Zinco/fisiologia , Animais , Cádmio/metabolismo , Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Caseína Quinase II/antagonistas & inibidores , Caseína Quinase II/metabolismo , Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Inibidores Enzimáticos/farmacologia , Fibroblastos/química , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos , Fosfatos/metabolismo , Fosforilação , Regiões Promotoras Genéticas/genética , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C/metabolismo , Especificidade da Espécie , Especificidade por Substrato , Fatores de Transcrição/biossíntese , Fatores de Transcrição/metabolismo , Zinco/farmacologia , Fator MTF-1 de Transcrição
12.
Nucleic Acids Res ; 31(23): 6710-21, 2003 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-14627804

RESUMO

The in vivo association of transcription factors with the metallothionein-I promoter was examined using chromatin immunoprecipitation (ChIP) assays. The results demonstrated that c-fos is rapidly recruited along with the metal response element-binding transcription factor-1 (MTF-1) to this promoter in response to zinc or cadmium, and that this recruitment is reversed in the visceral yolk sac by a zinc-deficient diet in vivo, and in cultured cells after lowering the zinc concentration in the medium or during prolonged zinc exposure. In contrast, the interactions of c-jun, USF-1, USF-2 and Sp1 with this promoter are metal-independent. Studies of knockout cells revealed that the recruitment of c-fos to the MT-I promoter requires MTF-1, but that c-fos is not essential for recruitment of MTF-1 and metal-induction of MT-I gene expression. Studies of Hepa cells stably-transfected with reporter genes driven by the MT-I promoter suggested two in vivo binding sites for USF-1 and -2. In contrast, Sp1 was apparently associated with a single binding site (upstream of -153 bp). In addition, maximal recruitment of c-fos by metals required sequences and/or other proteins that interact upstream of -153 bp. In summary, these studies extend our understanding of the complexity and dynamics of the transcription factor complex that forms at the MT-I promoter in vivo in response to metals.


Assuntos
Metalotioneína/genética , Metais/farmacologia , Regiões Promotoras Genéticas/genética , Fatores de Transcrição/metabolismo , Animais , Cádmio/farmacologia , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/metabolismo , Fibroblastos , Substâncias Macromoleculares , Camundongos , Testes de Precipitina , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-fos/metabolismo , Proteínas Proto-Oncogênicas c-jun/metabolismo , Fator de Transcrição Sp1/metabolismo , Fatores Estimuladores Upstream , Zinco/farmacologia , Fator MTF-1 de Transcrição
13.
J Biol Chem ; 278(35): 33474-81, 2003 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-12801924

RESUMO

The human ZIP4 gene (SLC39A4) is a candidate for the genetic disorder of zinc metabolism acrodermatitis enteropathica. To understand its role in zinc homeostasis, we examined the function and expression of mouse ZIP4. This gene encodes a well conserved eight-transmembrane protein that can specifically increase the influx of zinc into transfected cells. Expression of this gene is robust in tissues involved in nutrient uptake, such as the intestines and embryonic visceral yolk sac, and is dynamically regulated by zinc. Dietary zinc deficiency causes a marked increase in the accumulation of ZIP4 mRNA in these tissues, whereas injection of zinc or increasing zinc content of the diet rapidly reduces its abundance. Zinc can also regulate the accumulation of ZIP4 protein at the apical surface of enterocytes and visceral endoderm cells. These results provide compelling evidence that ZIP4 is a zinc transporter that plays an important role in zinc homeostasis, a process that is defective in acrodermatitis enteropathica in humans.


Assuntos
Acrodermatite/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Proteínas de Transporte de Cátions/fisiologia , Sequência de Aminoácidos , Animais , Transporte Biológico , Northern Blotting , Proteínas de Transporte de Cátions/química , Cátions , Membrana Celular/metabolismo , Relação Dose-Resposta a Droga , Feminino , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Intestino Delgado/embriologia , Cinética , Masculino , Metais/química , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Peptídeos/química , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , RNA/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Distribuição Tecidual , Transfecção , Zinco/metabolismo
14.
Nucleic Acids Res ; 30(14): 3130-40, 2002 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-12136095

RESUMO

The zinc finger protein, metal response element-binding transcription factor-1 (MTF-1) regulates the expression of genes in response to metal ions and oxidative stress. The precise mechanisms by which this occurs are not understood. To further examine this problem, mouse MTF-1 was expressed in Saccharomyces cerevisiae and tested for the ability to activate metal response element-driven reporter gene expression. Zinc was an effective inducer of reporter gene expression. In general, the magnitude of zinc induction was dependent on the concentration of zinc in the culture medium, but independent of the amount of MTF-1 expression. Zinc induction also occurred with either integrated or episomal reporter plasmids containing the native mouse metallothionein-I proximal promoter. Deletion of fingers 5 and 6 of MTF-1, which function in a zinc-dependent manner to stabilize the DNA-binding activity of the protein in vitro, did not diminish the zinc induction of either episomal or integrated promoters. However, a Gal4 DNA-binding domain- MTF-1 fusion protein, which binds constitutively to the Gal4-responsive promoter, was not zinc inducible but caused constitutive activation of reporter gene expression. This suggests that zinc activation of the DNA-binding activity of MTF-1 is the rate limiting step in its metalloregulatory function in yeast. In contrast, MTF-1 was not responsive to either cadmium or hydrogen peroxide, suggesting that distinct co-activators or signal transduction cascades not found in yeast are required to mediate MTF-1 activation of gene expression by this toxic metal and by oxidative stress.


Assuntos
Cádmio/farmacologia , Estresse Oxidativo/fisiologia , Saccharomyces cerevisiae/efeitos dos fármacos , Fatores de Transcrição/fisiologia , Zinco/farmacologia , Animais , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Proteínas de Ligação a DNA , Relação Dose-Resposta a Droga , Expressão Gênica , Regulação Fúngica da Expressão Gênica/efeitos dos fármacos , Genes Reporter/genética , Peróxido de Hidrogênio/farmacologia , Metalotioneína/genética , Camundongos , Regiões Promotoras Genéticas/genética , Proteínas Recombinantes de Fusão/efeitos dos fármacos , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Elementos de Resposta/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/crescimento & desenvolvimento , Transdução de Sinais , Fatores de Transcrição/genética , beta-Galactosidase/efeitos dos fármacos , beta-Galactosidase/genética , beta-Galactosidase/metabolismo , Fator MTF-1 de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA