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/metabolismoRESUMO
Extreme differences in allele frequency between West Africans and Eurasians were observed for a leucine-to-valine substitution (Leu372Val) in the human intestinal zinc uptake transporter, ZIP4, yet no further evidence was found for a selective sweep around the ZIP4 gene (SLC39A4). By interrogating allele frequencies in more than 100 diverse human populations and resequencing Neanderthal DNA, we confirmed the ancestral state of this locus and found a strong geographical gradient for the derived allele (Val372), with near fixation in West Africa. In extensive coalescent simulations, we show that the extreme differences in allele frequency, yet absence of a classical sweep signature, can be explained by the effect of a local recombination hotspot, together with directional selection favoring the Val372 allele in Sub-Saharan Africans. The possible functional effect of the Leu372Val substitution, together with two pathological mutations at the same codon (Leu372Pro and Leu372Arg) that cause acrodermatitis enteropathica (a disease phenotype characterized by extreme zinc deficiency), was investigated by transient overexpression of human ZIP4 protein in HeLa cells. Both acrodermatitis mutations cause absence of the ZIP4 transporter cell surface expression and nearly absent zinc uptake, while the Val372 variant displayed significantly reduced surface protein expression, reduced basal levels of intracellular zinc, and reduced zinc uptake in comparison with the Leu372 variant. We speculate that reduced zinc uptake by the ZIP4-derived Val372 isoform may act by starving certain pathogens of zinc, and hence may have been advantageous in Sub-Saharan Africa. Moreover, these functional results may indicate differences in zinc homeostasis among modern human populations with possible relevance for disease risk.
Assuntos
Acrodermatite/genética , Proteínas de Transporte de Cátions/genética , Genética Populacional , Seleção Genética/genética , Zinco/deficiência , Acrodermatite/patologia , África Subsaariana , Regulação da Expressão Gênica/genética , Frequência do Gene , Células HeLa , Humanos , MutaçãoRESUMO
Dietary zinc deficiency puts human health at risk, so we explored strategies for enhancing zinc absorption. In the small intestine, the zinc transporter ZIP4 functions as an essential component of zinc absorption. Overexpression of ZIP4 protein increases zinc uptake and thereby cellular zinc levels, suggesting that food components with the ability to increase ZIP4 could potentially enhance zinc absorption via the intestine. In the present study, we used mouse Hepa cells, which regulate mouse Zip4 (mZip4) in a manner indistinguishable from that in intestinal enterocytes, to screen for suitable food components that can increase the abundance of ZIP4. Using this ZIP4-targeting strategy, two such soybean extracts were identified that were specifically able to decrease mZip4 endocytosis in response to zinc. These soybean extracts also effectively increased the abundance of apically localized mZip4 in transfected polarized Caco2 and Madin-Darby canine kidney cells and, moreover, two apically localized mZip4 acrodermatitis enteropathica mutants. Soybean components were purified from one extract and soyasaponin Bb was identified as an active component that increased both mZip4 protein abundance and zinc levels in Hepa cells. Finally, we confirmed that soyasaponin Bb is capable of enhancing cell surface endogenous human ZIP4 in human cells. Our results suggest that ZIP4 targeting may represent a new strategy to improve zinc absorption in humans.
Assuntos
Proteínas de Transporte de Cátions/agonistas , Enterócitos/metabolismo , Fármacos Gastrointestinais/metabolismo , Glycine max/química , Absorção Intestinal , Extratos Vegetais/metabolismo , Zinco/metabolismo , Animais , Células CACO-2 , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular , Membrana Celular/metabolismo , Deficiências Nutricionais/metabolismo , Deficiências Nutricionais/prevenção & controle , Suplementos Nutricionais , Cães , Endocitose , Enterócitos/citologia , Fármacos Gastrointestinais/análise , Fármacos Gastrointestinais/química , Fármacos Gastrointestinais/uso terapêutico , Regulação da Expressão Gênica , Humanos , Camundongos , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , Estabilidade Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Saponinas/análise , Saponinas/metabolismo , Sementes/química , Zinco/deficiênciaRESUMO
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/metabolismoRESUMO
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 TranscriçãoRESUMO
BACKGROUND: Population pharmacokinetic data suggest axitinib plasma exposure correlates with efficacy in metastatic renal-cell carcinoma. Axitinib dose titration might optimise exposure and improve outcomes. We prospectively assessed the efficacy and safety of axitinib dose titration in previously untreated patients with metastatic renal-cell carcinoma. METHODS: In this randomised, double-blind, multicentre, phase 2 study, patients were enrolled from 49 hospitals and outpatient clinics in the Czech Republic, Germany, Japan, Russia, Spain, and USA. Patients with treatment-naive metastatic renal-cell carcinoma received axitinib 5 mg twice daily during a 4 week lead-in period. Those patients with blood pressure 150/90 mm Hg or lower, no grade 3 or 4 treatment-related toxic effects, no dose reductions, and no more than two antihypertensive drugs for 2 consecutive weeks were stratified by Eastern Cooperative Oncology Group performance status (0 vs 1), and then randomly assigned (1:1) to either masked titration with axitinib to total twice daily doses of 7 mg, and then 10 mg, if tolerated, or placebo titration. Patients who did not meet these criteria continued without titration. The primary objective was comparison of the proportion of patients achieving an objective response between randomised groups. Safety analyses were based on all patients who received at least one dose of axitinib. FINDINGS: Between Sept 2, 2009, and Feb 28, 2011, we enrolled 213 patients, of whom 112 were randomly assigned to either the axitinib titration group (56 patients) or the placebo titration group (56 patients). 91 were not eligible for titration, and ten withdrew during the lead-in period. 30 patients (54%, 95% CI 40-67) in the axitinib titration group had an objective response, as did 19 patients (34%, 22-48]) in the placebo titration group (one-sided p=0·019). 54 (59%, 95% CI 49-70) of non-randomised patients achieved an objective response. Common grade 3 or worse, all-causality adverse events in treated patients were hypertension (ten [18%] of 56 in the axitinib titration group vs five [9%] of 56 in the placebo titration group vs 45 [49%] of 91 in the non-randomised group), diarrhoea (seven [13%] vs two [4%] vs eight [9%]), and decreased weight (four [7%] vs three [5%] vs six [7%]). One or more all-causality serious adverse events were reported in 15 (27%) patients in the axitinib titration group, 13 (23%) patients in the placebo titration group, and 35 (38%) non-randomised patients. The most common serious adverse events in all 213 patients were disease progression and dehydration (eight each [4%]), and diarrhoea, vomiting, pneumonia, and decreased appetite (four each [2%]). INTERPRETATION: The greater proportion of patients in the axitinib titration group achieving an objective response supports the concept of individual axitinib dose titration in selected patients with metastatic renal-cell carcinoma. Axitinib shows clinical activity with a manageable safety profile in treatment-naive patients with this disease.
Assuntos
Antineoplásicos/administração & dosagem , Carcinoma de Células Renais/tratamento farmacológico , Carcinoma de Células Renais/secundário , Imidazóis/administração & dosagem , Indazóis/administração & dosagem , Neoplasias Renais/tratamento farmacológico , Neoplasias Renais/patologia , Inibidores de Proteínas Quinases/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos/efeitos adversos , Axitinibe , Carcinoma de Células Renais/mortalidade , Progressão da Doença , Intervalo Livre de Doença , Método Duplo-Cego , Europa (Continente) , Feminino , Humanos , Imidazóis/efeitos adversos , Indazóis/efeitos adversos , Japão , Estimativa de Kaplan-Meier , Neoplasias Renais/mortalidade , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Inibidores de Proteínas Quinases/efeitos adversos , Fatores de Tempo , Resultado do Tratamento , Estados UnidosRESUMO
CA1 pyramidal neurons are the final integrators of information flow leaving the hippocampus, yet are singularly vulnerable to activity-dependent cell death. Zinc (Zn) entry into cells may add to this vulnerability. Here, we find that Slc39a1 and Slc39a3, members of the Zip (Zrt/Irt-like protein) plasmalemmal Zn transporter family, are predominantly expressed in the hippocampus. We examined Zip-1,3-deficient mice to investigate their role in neurodegeneration following intense synaptic activation. When isolated by blockade of NMDA receptors and voltage-gated calcium channels, the absence of both transporters slowed passive Zn uptake into CA1 neurons measured with intracellular fluorescent Zn dyes. In vivo CA1 cell damage following kainic acid exposure was greatly attenuated. Consistent with the hypothesis that Zn entry contributes to neurodegeneration, Znt-3-deficient mice lacking synaptic Zn also show less hippocampal cell damage following kainic acid injection. Zip transporters may provide selective therapeutic targets to protect these neurons from early Zn-induced neurodegeneration following injury.
Assuntos
Região CA1 Hipocampal/fisiopatologia , Proteínas de Transporte de Cátions/deficiência , Degeneração Neural/etiologia , Convulsões/patologia , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Modelos Animais de Doenças , Eletroencefalografia/métodos , Agonistas de Aminoácidos Excitatórios/efeitos adversos , Fluoresceínas , Regulação Viral da Expressão Gênica/genética , Proteínas de Fluorescência Verde/genética , Ácido Caínico/efeitos adversos , Camundongos , Camundongos Transgênicos , Degeneração Neural/genética , Neurônios/efeitos dos fármacos , Neurônios/fisiologia , Compostos Orgânicos , Técnicas de Patch-Clamp/métodos , Receptores de N-Metil-D-Aspartato/metabolismo , Convulsões/induzido quimicamente , Gravação em Vídeo/métodos , Zinco/metabolismoRESUMO
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 TranscriçãoRESUMO
Translation of the basolateral zinc transporter ZIP5 is repressed during zinc deficiency but Zip5 mRNA remains associated with polysomes and can be rapidly translated when zinc is repleted. Herein, we examined the mechanisms regulating translation of Zip5. The 3'-untranslated region (UTR) of Zip5 mRNA is well conserved among mammals and is predicted by mFOLD to form a very stable stem-loop structure. Three algorithms predict this structure to be flanked by repeated seed sites for miR-328 and miR-193a. RNAse footprinting supports the notion that a stable stem-loop structure exists in this 3'-UTR and electrophoretic mobility shift assays detect polysomal protein(s) binding specifically to the stem-loop structure in the Zip5 3'-UTR. miR-328 and miR-193a are expressed in tissues known to regulate Zip5 mRNA translation in response to zinc availability and both are polysome-associated consistent with Zip5 mRNA localization. Transient transfection assays using native and mutant Zip5 3'-UTRs cloned 3' to luciferase cDNA revealed that the miRNA seed sites and the stem-loop function together to augment translation of Zip5 mRNA when zinc is replete.
Assuntos
Regiões 3' não Traduzidas/fisiologia , Proteínas de Transporte de Cátions/genética , Biossíntese de Proteínas , Animais , Sequência de Bases , Células Cultivadas , Camundongos , MicroRNAs/fisiologia , Dados de Sequência Molecular , Polirribossomos/metabolismo , Ratos , Ribonucleoproteínas/químicaRESUMO
We report the design of novel, potent cPLA(2)α inhibitors that possess an α-methyl-2-ketothiazole that acts as a serine-reactive moiety. We describe the optimization of the series for potency and metabolic stability towards ketone reduction. This was achieved by attenuating the reactivity of the ketone using a combination of electronic and steric effects.
Assuntos
Desenho de Fármacos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Fosfolipases A2 do Grupo IV/antagonistas & inibidores , Cetonas/química , Tiazóis/síntese química , Tiazóis/farmacologia , Animais , Estabilidade de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/química , Células HL-60 , Humanos , Concentração Inibidora 50 , Cetonas/síntese química , Cetonas/farmacologia , Espectroscopia de Ressonância Magnética , Estrutura Molecular , Oxirredução , Ratos , Serina/química , Tiazóis/químicaRESUMO
By careful analysis of experimental X-ray ligand crystallographic protein data across several inhibitor series we have discovered a novel, potent and selective series of iNOS inhibitors exemplified by compound 8.
Assuntos
Inibidores Enzimáticos/química , Isoxazóis/química , Óxido Nítrico Sintase Tipo II/antagonistas & inibidores , Piridinas/química , Animais , Sítios de Ligação , Simulação por Computador , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/farmacologia , Humanos , Isoxazóis/síntese química , Isoxazóis/farmacologia , Camundongos , Microssomos Hepáticos/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Estrutura Terciária de Proteína , Piridinas/farmacologia , RatosRESUMO
Nitric oxide synthase (NOS) enzymes synthesize nitric oxide, a signal for vasodilatation and neurotransmission at low concentrations and a defensive cytotoxin at higher concentrations. The high active site conservation among all three NOS isozymes hinders the design of selective NOS inhibitors to treat inflammation, arthritis, stroke, septic shock and cancer. Our crystal structures and mutagenesis results identified an isozyme-specific induced-fit binding mode linking a cascade of conformational changes to a new specificity pocket. Plasticity of an isozyme-specific triad of distant second- and third-shell residues modulates conformational changes of invariant first-shell residues to determine inhibitor selectivity. To design potent and selective NOS inhibitors, we developed the anchored plasticity approach: anchor an inhibitor core in a conserved binding pocket, then extend rigid bulky substituents toward remote specificity pockets, which become accessible upon conformational changes of flexible residues. This approach exemplifies general principles for the design of selective enzyme inhibitors that overcome strong active site conservation.
Assuntos
Desenho de Fármacos , Inibidores Enzimáticos , Inflamação/tratamento farmacológico , Inflamação/enzimologia , Óxido Nítrico Sintase/antagonistas & inibidores , Sequência de Aminoácidos , Aminopiridinas/química , Aminopiridinas/farmacologia , Animais , Bovinos , Cristalografia por Raios X , Modelos Animais de Doenças , Inibidores Enzimáticos/síntese química , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Expressão Gênica , Humanos , Isoenzimas/antagonistas & inibidores , Masculino , Camundongos , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Mutação , Óxido Nítrico Sintase/genética , Óxido Nítrico Sintase/metabolismo , Quinazolinas/química , Quinazolinas/farmacologia , RatosRESUMO
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 TranscriçãoRESUMO
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 ZincoRESUMO
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 TranscriçãoRESUMO
The SLC39A family of zinc transporters can be divided into four subfamilies (I, II, LIV-1, and gufA) in vertebrates, but studies of their functions have been restricted exclusively to members of subfamilies II and LIV-1. In this study, we characterized SLC39A9 (ZIP9), the only member of subfamily I in vertebrates. Confocal microscopy demonstrated that transiently expressed, HA-tagged human ZIP9 (hZIP9-HA) was localized to the trans-Golgi network regardless of zinc status. Disruption of the ZIP9 gene in DT40 cells did not change the growth rate, sensitivity to high zinc and manganese concentrations during long-term culture, or cellular zinc status after short-term incubation with zinc. The alkaline phosphatase activity of ZIP9(-/-) cells did not change in cells cultured in medium containing normal zinc levels. In contrast, the activity of this enzyme decreased in wild-type cells cultured in zinc deficient medium but less so in ZIP9(-/-) cells under these conditions. Stable over-expression of hZIP9-HA moderately decreased alkaline phophatase activity. These results suggest that ZIP9 functions to regulate zinc homeostasis in the secretory pathway without significantly altering cytosolic zinc homeostasis.
Assuntos
Proteínas de Transporte de Cátions/metabolismo , Homeostase , Via Secretória , Zinco/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Proteínas de Transporte de Cátions/deficiência , Proteínas de Transporte de Cátions/genética , Células HeLa , Humanos , Dados de Sequência Molecular , Rede trans-Golgi/metabolismoRESUMO
Mouse MT-I (metallothionein-I) transcription is regulated by MTF-1 (metal-response-element-binding transcription factor-1) which is recruited to the promoter in response to zinc. Cr(VI) [chromium(VI)] pretreatment blocks zinc-activation of the endogenous MT-I gene and attenuates zinc-activation of MT-I-promoter-driven luciferase reporter genes in transient transfection assays. Chromatin immunoprecipitation assays revealed that Cr(VI) only modestly reduces recruitment of MTF-1 to the MT-I promoter in response to zinc, but drastically reduces the recruitment of RNA polymerase II. These results suggest that Cr(VI) inhibits the ability of MTF-1 to transactivate this gene in response to zinc. Zinc has recently been shown to induce the formation of a co-activator complex containing MTF-1 and the histone acetyltransferase p300 which plays an essential role in the activation of MT-I transcription. In the present study, co-immunoprecipitation assays demonstrated that Cr(VI) pretreatment blocks the zinc-induced formation of this co-activator complex. Thus Cr(VI) inhibits mouse MT-I gene expression in response to zinc by interfering with the ability of MTF-1 to form a co-activator complex containing p300 and recruiting RNA polymerase II to the promoter.
Assuntos
Cromo/farmacologia , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteína p300 Associada a E1A/antagonistas & inibidores , Metalotioneína/genética , Fatores de Transcrição/antagonistas & inibidores , Transcrição Gênica , Zinco/metabolismo , Animais , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína p300 Associada a E1A/genética , Proteína p300 Associada a E1A/metabolismo , Metalotioneína/metabolismo , Camundongos , Regiões Promotoras Genéticas , RNA Polimerase II/genética , RNA Polimerase II/metabolismo , Elementos de Resposta , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Transfecção , Fator MTF-1 de TranscriçãoRESUMO
The essential metals copper, zinc, and iron play key roles in embryonic, fetal, and postnatal development in higher eukaryotes. Recent advances in our understanding of the molecules involved in the intricate control of the homeostasis of these metals and the availability of natural mutations and targeted mutations in many of the genes involved have allowed for elucidation of the diverse roles of these metals during development. Evidence suggests that the ability of the embryo to control the homeostasis of these metals becomes essential at the blastocyst stage and during early morphogenesis. However, these metals play unique roles throughout development and exert pleiotropic, metal-specific, and often cell-specific effects on morphogenesis, growth, and differentiation. Herein, we briefly review the major players known to be involved in the homeostasis of each of these essential metals and their known roles in development.
Assuntos
Cobre/metabolismo , Desenvolvimento Embrionário/genética , Desenvolvimento Fetal/genética , Homeostase/genética , Ferro/metabolismo , Zinco/metabolismo , Animais , Cobre/fisiologia , Desenvolvimento Embrionário/fisiologia , Desenvolvimento Fetal/fisiologia , Homeostase/fisiologia , Humanos , Ferro/fisiologia , Zinco/fisiologiaRESUMO
A novel mechanism for antagonism of the human chemokine receptors CCR4 and CCR5 has been discovered with a series of small-molecule compounds that seems to interact with an allosteric, intracellular site on the receptor. The existence of this site is supported by a series of observations: 1) intracellular access of these antagonists is required for their activity; 2) specific, saturable binding of a radiolabeled antagonist requires the presence of CCR4; and 3) through engineering receptor chimeras by reciprocal transfer of C-terminal domains between CCR4 and CCR5, compound binding and the selective structure-activity relationships for antagonism of these receptors seem to be associated with the integrity of that intracellular region. Published antagonists from other chemical series do not seem to bind to the novel site, and their interaction with either CCR4 or CCR5 is not affected by alteration of the C-terminal domain. The precise location of the proposed binding site remains to be determined, but the known close association of the C-terminal domain, including helix 8, as a proposed intracellular region that interacts with transduction proteins (e.g., G proteins and beta-arrestin) suggests that this could be a generic allosteric site for chemokine receptors and perhaps more broadly for class A G protein-coupled receptors. The existence of such a site that can be targeted for drug discovery has implications for screening assays for receptor antagonists, which would need, therefore, to consider compound properties for access to this intracellular site.
Assuntos
Sítio Alostérico , Antagonistas dos Receptores CCR5 , Quimiocinas CC/antagonistas & inibidores , Receptores CCR4/antagonistas & inibidores , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Sequência de Aminoácidos , Animais , Células CHO , Sinalização do Cálcio , Linhagem Celular , Quimiocina CCL22/metabolismo , Quimiocina CCL5/metabolismo , Quimiocinas CC/química , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Cricetinae , Cricetulus , Interpretação Estatística de Dados , Humanos , Concentração Inibidora 50 , Rim/citologia , Dados de Sequência Molecular , Estrutura Molecular , Receptores CCR4/química , Receptores CCR4/genética , Receptores CCR4/metabolismo , Receptores CCR5/química , Receptores CCR5/genética , Receptores CCR5/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Homologia de Sequência de Aminoácidos , TransfecçãoRESUMO
The SLC39A (solute carrier 39A) [ZIP (Zrt-Irt-like protein)] family consists of 14 members which are thought to control zinc uptake into the cytoplasm. Among these, ZIP4 is known to be particularly important for zinc homoeostasis. Mutations in this gene cause acrodermatitis enteropathica, a rare recessive-lethal human genetic disorder. In the present paper, our studies of the regulation and function of the mouse Zip4 gene are briefly reviewed. Mouse Zip4 is expressed at highest levels in tissues involved in absorption of dietary or maternal zinc, and the gene and protein are dynamically regulated by multiple post-transcriptional mechanisms in response to zinc availability. ZIP4 accumulates at the apical surface of enterocytes and endoderm cells when zinc is deficient, because of increased stability of the mRNA and stabilization of the protein. In contrast, when zinc is replenished, the mRNA is destabilized and the protein is internalized and degraded rapidly. The critical importance of ZIP4 in zinc homoeostasis is revealed in mice with targeted deletions of this gene. Homozygous Zip4-knockout embryos die during early morphogenesis and heterozygous offspring are significantly underrepresented and display an array of developmental defects, including exencephalia, anophthalmia and severe growth retardation. Mice heterozygous for Zip4-knockout are hypersensitive to zinc deficiency, which suggests that humans heterozygous for this gene may also be very sensitive to zinc deficiency.