Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 11(1): 6523, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33753776

RESUMO

Insecticidal double-stranded RNAs (dsRNAs) silence expression of vital genes by activating the RNA interference (RNAi) mechanism in insect cells. Despite high commercial interest in insecticidal dsRNA, information on resistance to dsRNA is scarce, particularly for dsRNA products with non-transgenic delivery (ex. foliar/topical application) nearing regulatory review. We report the development of the CEAS 300 population of Colorado potato beetle (Leptinotarsa decemlineata Say) (Coleoptera: Chrysomelidae) with > 11,100-fold resistance to a dsRNA targeting the V-ATPase subunit A gene after nine episodes of selection using non-transgenic delivery by foliar coating. Resistance was associated with lack of target gene down-regulation in CEAS 300 larvae and cross-resistance to another dsRNA target (COPI ß; Coatomer subunit beta). In contrast, CEAS 300 larvae showed very low (~ 4-fold) reduced susceptibility to the Cry3Aa insecticidal protein from Bacillus thuringiensis. Resistance to dsRNA in CEAS 300 is transmitted as an autosomal recessive trait and is polygenic. These data represent the first documented case of resistance in an insect pest with high pesticide resistance potential using dsRNA delivered through non-transgenic techniques. Information on the genetics of resistance and availability of dsRNA-resistant L. decemlineata guide the design of resistance management tools and allow research to identify resistance alleles and estimate resistance risks.


Assuntos
Besouros/efeitos dos fármacos , Resistência a Medicamentos/genética , Inseticidas/farmacologia , RNA de Cadeia Dupla/farmacologia , Animais , Bacillus thuringiensis/genética , Toxinas de Bacillus thuringiensis/genética , Toxinas de Bacillus thuringiensis/farmacologia , Besouros/genética , Besouros/patogenicidade , Colorado , Endotoxinas/genética , Endotoxinas/farmacologia , Proteínas Hemolisinas/genética , Proteínas Hemolisinas/farmacologia , Proteínas de Insetos/genética , Larva/genética , Larva/crescimento & desenvolvimento , Interferência de RNA , RNA de Cadeia Dupla/genética , Solanum tuberosum/crescimento & desenvolvimento , Solanum tuberosum/parasitologia
2.
Antimicrob Agents Chemother ; 58(6): 3399-410, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24709254

RESUMO

A small-molecule inhibitor of hepatitis C virus (HCV) designated AP89652 was identified by screening a compound library with an HCV genotype 1b subgenomic replicon assay. AP89652 contains two chiral centers, and testing of two syn enantiomers revealed that activity in the replicon assay resided with only one, AP80978, whose 50% effective concentration (EC50) (the concentration at which a 50% reduction in Renilla luciferase levels was observed relative to an untreated control) was 630 nM. AP80978 was inhibitory against HCV genotypes 1a and 1b but not genotype 2a. In a replicon clearance assay, the potency and clearance rate of AP80978 were similar to those of telaprevir (VX950) and cyclosporine (CsA). AP80978 was nontoxic when tested against a panel of human cell lines, and inhibitory activity was HCV specific in that there was limited activity against negative-strand viruses, an alphavirus, and flaviviruses. By selection of resistant replicons and assessment of activity in genotype 1b/2a intergenotypic replicons, the viral protein target of this compound was identified as NS4B. NS4B F98V/L substitutions were confirmed by site-directed mutagenesis as AP80978 resistance-associated mutations. When tested against HCV produced in cell culture, the compound was significantly more potent than other HCV inhibitors, including VX950, CsA, and 2'-C-methyladenosine (2'C-meA). In addition, AP80977, the enantiomer that was inactive in the replicon assay, had activity against the virus, although it was lower than the activity of AP80978. These results suggest that AP80978 has the potential to be optimized into an effective antiviral drug and is a useful tool to further study the role of NS4B in HCV replication.


Assuntos
Antivirais/farmacologia , Furanos/farmacologia , Hepacivirus/efeitos dos fármacos , Hepatite C/tratamento farmacológico , Piridinas/farmacologia , Tiofenos/farmacologia , Proteínas não Estruturais Virais/antagonistas & inibidores , Adenosina/análogos & derivados , Adenosina/farmacologia , Bioensaio , Técnicas de Cultura de Células , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Ciclosporina/farmacologia , Genótipo , Hepacivirus/genética , Hepatite C/virologia , Ensaios de Triagem em Larga Escala , Humanos , Luciferases de Renilla , Mutagênese Sítio-Dirigida , Oligopeptídeos/farmacologia , Replicon/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
3.
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
4.
Biol Chem ; 388(12): 1301-12, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18020946

RESUMO

Dietary zinc deficiency in mice is accompanied by enhanced expression of the zinc uptake transporter Slc39a4 (Zip4) and repressed expression of Slc39a5 (Zip5) in tissues which regulate zinc homeostasis (intestine, pancreas and visceral yolk sac). Herein, mechanisms controlling this differential expression were investigated. The induction of Zip4 mRNA during zinc deficiency, and its repression in response to zinc repletion were found to reflect changes in Zip4 mRNA stability and not changes in the relative rate of transcription of this gene. During zinc deficiency, ZIP4 protein levels are increased and this protein is localized on the apical membranes. Administration of an oral gavage of zinc caused ZIP4 internalization and degradation in enterocytes and visceral endoderm cells. Similarly, ZIP4 is induced by zinc deficiency in cultured mouse Hepa cells and is rapidly degraded in response to added zinc. Zip5 mRNA abundance does not change in response to zinc, but the translation of this mRNA was found to be zinc-responsive. During zinc deficiency, Zip5 mRNA remains associated with polysomes, while the protein is internalized and degraded in enterocytes, acinar cells and endoderm cells. After zinc-gavage, ZIP5 is rapidly resynthesized and targeted to the basolateral membranes of these cell types.


Assuntos
Proteínas de Transporte de Cátions/biossíntese , Proteínas de Transporte de Cátions/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Zinco/farmacologia , Animais , Northern Blotting , Western Blotting , Linhagem Celular , Centrifugação com Gradiente de Concentração , Dieta , Feminino , Imuno-Histoquímica , Imunoprecipitação , Cinética , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Técnicas de Cultura de Órgãos , Polirribossomos/genética , Gravidez , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , RNA Mensageiro/isolamento & purificação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Saco Vitelino/transplante
5.
Hum Mol Genet ; 16(12): 1391-9, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17483098

RESUMO

The human Zip4 gene (Slc39a4) is mutated in the rare recessive genetic disorder of zinc metabolism acrodermatitis enteropathica, but the physiological functions of Zip4 are not well understood. Herein we demonstrate that homozygous Zip4-knockout mouse embryos die during early morphogenesis and heterozygous offspring are significantly underrepresented. At mid-gestation, an array of developmental defects including exencephalia, anophthalmia and severe growth retardation were noted in heterozygous embryos, and at weaning, many (63/280) heterozygous offspring were hydrocephalic, growth retarded and missing one or both eyes. Maternal dietary zinc deficiency during pregnancy exacerbated these effects, whereas zinc excess ameliorated these effects and protected embryonic development of heterozygotes but failed to rescue homozygous embryos. Heterozygous Zip4 embryos were not underrepresented in litters from wild-type mothers, but were approximately 10 times more likely to develop abnormally than were their wild-type littermates during zinc deficiency. Thus, both embryonic and maternal Zip4 gene expressions are critical for proper zinc homeostasis. These studies suggest that heterozygous mutations in the acrodermatitis gene Zip4 may be associated with a wider range of developmental defects than was previously appreciated, particularly when dietary zinc is limiting.


Assuntos
Proteínas de Transporte de Cátions/genética , Desenvolvimento Embrionário , Heterozigoto , Zinco/deficiência , Acrodermatite/genética , Alelos , Animais , Proteínas de Transporte de Cátions/metabolismo , Células Cultivadas , Embrião de Mamíferos/metabolismo , Endoderma/metabolismo , Feminino , Homozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Camundongos Knockout , Microscopia de Fluorescência , Modelos Genéticos , Zinco/metabolismo
6.
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
7.
Dev Biol ; 295(2): 571-9, 2006 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16682017

RESUMO

Several ZIP genes (SLC39A family of metal transporters) play roles in zinc homeostasis. Herein, the temporal and spatial patterns of expression of the mouse ZIP1, 3, 4, and 5 genes in the developing intestine and the effects of maternal dietary zinc deficiency on these patterns of expression were examined. ZIP1 and ZIP3 genes, conserved members of the ZIP subfamily II, were found to be coexpressed during development. Expression of these genes was detected on day 14 of gestation in smooth muscle and the pseudostratified endoderm. By 5 days post-partum, prominent expression became restricted to muscle and connective stroma. In contrast, expression of ZIP4 and ZIP5 genes, members of the ZIP subfamily called LIV-1, coincided with epithelial morphogenesis. ZIP5 expression was detected on d16 of gestation and localized to the basolateral membranes of the single-layered epithelium. ZIP4 expression was detected on d18 of gestation and localized to the apical membrane of villus epithelial cells. When dams were fed a zinc-deficient diet beginning at parturition, ZIP4 expression in the nursing neonate was greatly induced. In contrast, neonatal ZIP5 expression remained unchanged, but this protein was removed from the basolateral membrane of the enterocyte. These responses to dietary zinc deficiency mimic those found in the adult intestine. These studies reveal cell-type-specific expression of ZIP genes during development of the intestine, and suggest that the mouse intestine can elicit an adaptive response to dietary zinc availability at birth.


Assuntos
Proteínas de Transporte de Cátions/genética , Regulação da Expressão Gênica , Intestinos/crescimento & desenvolvimento , Zinco/metabolismo , Adaptação Fisiológica/genética , Animais , Animais Recém-Nascidos , Embrião de Mamíferos , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Homeostase , Camundongos , Distribuição Tecidual , Zinco/fisiologia
8.
Genesis ; 44(5): 239-51, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16652366

RESUMO

Subfamily II of the solute-linked carrier 39A superfamily contains three well-conserved zinc transporters (ZIPs1, 2, 3) whose physiological functions are unknown. We generated mice homozygous for knockout alleles of ZIP1 and both ZIP1 and ZIP 3 (double-knockout). These mice were apparently normal when dietary zinc was replete, but when dietary zinc was limited during pregnancy embryos from ZIP1 or ZIP3 knockout mice were two to three times more likely to develop abnormally than those in wildtype mice, and 91% (71/78) of embryos developed abnormally in ZIP1, ZIP3 double-knockout mice. Analysis of the patterns of expression of these genes in mice revealed predominate expression in intestinal stromal cells, nephric-tubular epithelial cells, pancreatic ductal epithelial cells, and hepatocytes surrounding the central vein. This suggests that these zinc transporters function, at least in part, in the redistribution and/or retention of zinc rather than its acquisition from the diet. In conclusion, mutations in the ZIP1 and ZIP3 zinc transporter genes are silent when dietary intake of zinc is normal, but can dramatically compromise the success of pregnancy when dietary intake of zinc is limiting.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Desenvolvimento Embrionário , Complicações na Gravidez/metabolismo , Zinco/deficiência , Adaptação Fisiológica , Animais , Proteínas de Transporte de Cátions/genética , Anormalidades Congênitas/etiologia , Dieta , Perda do Embrião/etiologia , Embrião de Mamíferos/metabolismo , Feminino , Expressão Gênica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Gravidez , Zinco/metabolismo
9.
Mol Cell Biol ; 25(13): 5607-15, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15964816

RESUMO

The mouse ZIP3 (SLC39A3) gene encodes an eight-transmembrane-domain protein that has been conserved in mammals and can function to transport zinc. To analyze the expression of ZIP3 in the early embryo and neonate and to determine its in vivo function, we generated ZIP3 null mice in which the ZIP3 open reading frame was replaced with that of the enhanced green fluorescent protein (EGFP) reporter. EGFP fluorescence revealed that ZIP3 was expressed in the inner cell mass of the blastocyst and later during embryonic development in many tissues. Elevated expression was apparent in the embryonic brain and neurotube and neonatal gonads. Homozygous knockout mice were viable and fertile and under normal growth conditions exhibited no obvious phenotypic abnormalities. Deletion of ZIP3 did not alter zinc homeostasis at the molecular level as assessed by essential metal levels and the expression of zinc-responsive genes. In knockout mice stressed with a zinc-deficient diet during pregnancy or at weaning, a subtle increase in the sensitivity to abnormal morphogenesis of the embryo and to depletion of thymic pre-T cells, respectively, was noted. These results suggest that this protein plays an ancillary role in zinc homeostasis in mice.


Assuntos
Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Zinco/metabolismo , Animais , Animais Recém-Nascidos , Blastocisto/metabolismo , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Cruzamentos Genéticos , Eletroporação , Desenvolvimento Embrionário , Feminino , Genes Reporter , Vetores Genéticos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Gravidez , Células-Tronco/citologia , Distribuição Tecidual , Zinco/análise , Zinco/deficiência
10.
J Biol Chem ; 279(47): 49082-90, 2004 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-15358787

RESUMO

The ZIP5 gene encodes a protein closely related to ZIP4, a zinc transporter mutated in the human genetic disorder acrodermatitis enteropathica. Herein, we demonstrate that mouse ZIP5 and ZIP4 genes are co-expressed in several tissues involved in zinc homeostasis (intestine, pancreas, embryonic yolk sac). However, unlike expression of the ZIP4 gene, which is induced during periods of zinc deficiency, ZIP5 gene expression is unaltered by dietary zinc. Immunohistochemistry localizes ZIP5 to the basolateral surfaces of enterocytes, acinar cells, and visceral endoderm cells in mice fed a zinc-adequate diet. However, this protein is removed from these cell surfaces and internalized during dietary zinc deficiency. In contrast, ZIP4 is induced and recruited to the apical surface of enterocytes and endoderm cells during zinc deficiency. In the pancreas, ZIP4 is expressed in beta-cells, whereas ZIP5 is expressed in acinar cells. These results suggest that the function of ZIP5 is antagonistic to that of ZIP4 in the control of zinc homeostasis; rather than functioning in the acquisition of dietary zinc, as does ZIP4, ZIP5 may function in the removal of zinc from the body. Thus, during periods when dietary zinc is replete, ZIP5 may function to remove zinc from the blood via the pancreas and intestine, the major sites of zinc excretion in mammals, whereas the acquisition of dietary zinc by intestinal ZIP4 would be minimal. In contrast, during periods of dietary zinc deficiency when secretion of zinc by the pancreas and intestine is minimized, ZIP5 is removed from the cell surface, and the intestinal uptake of zinc is augmented by induction of ZIP4.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Endoderma/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Zinco/farmacologia , Sequência de Aminoácidos , Animais , Transporte Biológico , Northern Blotting , Western Blotting , Proteínas de Transporte de Cátions/química , Membrana Celular/metabolismo , Biologia Computacional , Sequência Conservada , DNA Complementar/metabolismo , Suplementos Nutricionais , Evolução Molecular , Éxons , Feminino , Imuno-Histoquímica , Insulina/metabolismo , Camundongos , Microscopia de Fluorescência , Modelos Genéticos , Dados de Sequência Molecular , Pâncreas/citologia , Pâncreas/embriologia , Pâncreas/metabolismo , RNA/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Distribuição Tecidual , Saco Vitelino/metabolismo , Zinco/química , Zinco/metabolismo
11.
J Biol Chem ; 279(23): 24631-9, 2004 Jun 04.
Artigo em Inglês | MEDLINE | ID: mdl-15054103

RESUMO

The mouse mZip1 and mZip3 zinc transporters have been implicated in zinc acquisition by the cells of many tissues. This hypothesis raised the question of whether activity of these proteins is regulated to maintain zinc homeostasis. Neither mZIP1 nor mZIP3 mRNA levels are highly regulated by zinc status. Therefore, we investigated whether zinc controls the activity of these proteins post-translationally by altering their subcellular distribution. When expressed in transfected cells grown in zinc-replete medium, both mZip1 and mZip3 were largely present in intracellular organelles. However, these proteins were found to rapidly transit between the plasma membrane and intracellular compartments in zinc-replete cells. Zinc deficiency increased plasma membrane levels of mZip1 and mZip3 by decreasing their rates of endocytosis. Greater zinc deficiency was required to alter mZip3 distribution than was needed to affect mZip1. Increased surface levels correlated with increased zinc uptake activity. Taken together, these results suggest that post-translational control of mZip1 and mZip3 localization plays a role in zinc homeostasis. Moreover, our results indicate that zinc-responsive endocytosis is a conserved mechanism controlling activity of many mammalian zinc uptake transporters.


Assuntos
Proteínas de Transporte/metabolismo , Endocitose , Zinco/metabolismo , Animais , Proteínas de Transporte de Cátions , Linhagem Celular , Membrana Celular/metabolismo , Humanos , Immunoblotting , Camundongos , Microscopia de Fluorescência , Plasmídeos/metabolismo , Processamento de Proteína Pós-Traducional , RNA Mensageiro/metabolismo , Frações Subcelulares/metabolismo , Fatores de Tempo , Transfecção , Zinco/química
12.
Anal Biochem ; 326(1): 33-41, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14769333

RESUMO

No clinical assays for the direct detection of heparin in blood exist. To create a heparin sensor, the hyaluronan (HA)-binding domain (HABD) of a protein that binds heparin and HA was engineered. GST fusion proteins containing one to three HABD modules were cloned, expressed, and purified. The affinities of each construct for heparin and for HA were determined by a competitive enzyme-linked immunosorbent assay using immobilized HA or heparin. Each of the constructs showed modest affinity for immobilized HA. However, heparin was 100-fold more potent than HA as a competing ligand. With immobilized heparin, affinity increased as the HABD copy number increased. The three-copy construct, GST-HB3, detected unfractionated free heparin (UFH) as low as 39ng/ml (equivalent to approximately 0.1U/ml) with a signal-to-noise ratio of 5.6. GST-HB3 also showed 100-fold selectivity for heparin in preference to other glycosaminoglycans. The plot of logKd vs log [Na+] showed 2.5 ionic interactions per heparin-HB3 interaction. GST-HB3 showed a linear detection of both UFH (15kDa) and low-molecular-weight heparin (LMWH; 6kDa) added to human plasma. For UFH, the range examined was 78 to over 2000ng/ml (equivalent to 0.2 to 5.0U/ml). For LMWH, the useful range was 312 to over 2000ng/ml. The coefficient of variance for the assay was < 9% for six serial heparin dilutions and <12% for three plasma samples. In clinical use, GST-HB3 could accurately measure therapeutic heparin levels in plasma (0.2 to 2U/ml).


Assuntos
Heparina/análise , Sequência de Aminoácidos , Sítios de Ligação , Ensaio de Imunoadsorção Enzimática , Heparina/sangue , Heparina/química , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Ácido Hialurônico/metabolismo , Ligantes , Dados de Sequência Molecular , Estrutura Molecular , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Cloreto de Sódio/farmacologia , Soluções/química , Titulometria
13.
Hum Mol Genet ; 13(5): 563-71, 2004 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-14709598

RESUMO

The Zip4 protein is involved in dietary zinc uptake from the intestinal lumen. The human ZIP4 gene (SLC39A4) was identified because of its association with acrodermatitis enteropathica (AE), a genetic disorder of zinc absorption. To date, several SLC39A4 mutations have been identified in AE patients. To investigate the effects of these mutations on function of the Zip4 transporter, we introduced six AE-associated missense mutations into the orthologous mouse ZIP4 gene for functional expression in cultured cells. All mutations decreased 65Zn uptake activity of mZip4, thereby providing a causal link to AE. The mutants fell into two groups based on their phenotypic effects. Several alleles (G340D, L382P, G384R, G643R) failed to localize on the cell surface at high levels. These defects were attributable to misfolding and/or mislocalization in the secretory pathway. Two other alleles (P200L and G539R) accumulated to high levels in the plasma membrane and had wild-type apparent Km values for 65Zn uptake. However, these mutations decreased the Vmax of uptake to approximately 30% of wild-type. We showed previously that wild-type mZip4 is regulated post-translationally in response to zinc status. In zinc-replete cells, mZip4 is found largely in intracellular compartments. In zinc-limited cells, surface levels increase markedly because the rate of endocytosis decreases. Surprisingly, endocytosis of both P200L and G539R is no longer zinc responsive; these proteins are endocytosed at a slow rate regardless of zinc status. These effects suggest a zinc sensing mechanism for regulating Zip4 trafficking in response to zinc.


Assuntos
Acrodermatite/genética , Alelos , Proteínas de Transporte de Cátions/metabolismo , Endocitose/genética , Zinco/metabolismo , Animais , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Modelos Animais de Doenças , Glicosilação , Immunoblotting , Camundongos , Microscopia de Fluorescência , Mutação de Sentido Incorreto/genética , Plasmídeos/genética , Estrutura Terciária de Proteína , Transfecção
14.
J Biol Chem ; 279(6): 4523-30, 2004 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-14612438

RESUMO

Zinc is an essential nutrient for all organisms. Its requirement in humans is illustrated dramatically by the genetic disorder acrodermatitis enteropathica (AE). AE is caused by the reduced uptake of dietary zinc by enterocytes, and the ensuing systemic zinc deficiency leads to dermatological lesions and immune and reproductive dysfunction. The gene responsible for AE, SLC39A4, encodes a member of the ZIP family of metal transporters, hZIP4. The mouse ZIP4 protein, mZIP4, stimulates zinc uptake in cultured cells, and studies in mice have demonstrated that zinc treatment decreases mZIP4 mRNA levels in the gut. In this study, we demonstrated using transfected cultured cells that the mZIP4 protein is also regulated at a post-translational level in response to zinc availability. Zinc deficiency increased mZIP4 protein levels at the plasma membrane, and this was associated with increased zinc uptake. Significantly, treating cells with low micromolar zinc concentrations stimulated the rapid endocytosis of the transporter. Zinc-regulated localization of the human ZIP4 protein was also demonstrated in cultured cells. These findings suggest that zinc-regulated trafficking of human and mouse ZIP4 is a key mechanism controlling dietary zinc absorption and cellular zinc homeostasis.


Assuntos
Proteínas de Transporte de Cátions/metabolismo , Zinco/metabolismo , Animais , Proteínas de Transporte de Cátions/química , Proteínas de Transporte de Cátions/genética , Linhagem Celular , Membrana Celular/metabolismo , Endocitose , Humanos , Camundongos , Modelos Moleculares , Processamento de Proteína Pós-Traducional , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Zinco/deficiência , Zinco/farmacologia
15.
J Biol Chem ; 278(50): 50142-50, 2003 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-14525987

RESUMO

Zinc is an essential metal for all eukaryotes, and cells have evolved a complex system of proteins to maintain the precise balance of zinc uptake, intracellular storage, and efflux. In mammals, zinc uptake appears to be mediated by members of the Zrt/Irt-like protein (ZIP) superfamily of metal ion transporters. Herein, we have studied a subfamily of zip genes (zip1, zip2, and zip3) that is conserved in mice and humans. These eight-transmembrane domain proteins contain a conserved 12-amino acid signature sequence within the fourth transmembrane domain. All three of these mouse ZIP proteins function to specifically increase the uptake of zinc in transfected cultured cells, similar to the previously demonstrated functions of human ZIP1 and ZIP2 (Gaither, L. A., and Eide, D. J. (2000) J. Biol. Chem. 275, 5560-5564; Gaither, L. A., and Eide, D. J. (2001) J. Biol. Chem. 276, 22258-22264). No ZIP3 orthologs have been previously studied. Furthermore, this first systematic comparative study of the in vivo expression and dietary zinc regulation of this subfamily of zip genes revealed that 1) zip1 mRNA is abundant in many mouse tissues, whereas zip2 and zip3 mRNAs are very rare or moderately rare, respectively, and tissue-restricted in their accumulation; and 2) unlike mouse metallothionein I and zip4 mRNAs (Dufner-Beattie, J., Wang, F., Kuo, Y.-M., Gitschier, J., Eide, D., and Andrews, G. K. (2003) J. Biol. Chem. 278, 33474-33481), the abundance of zip1, zip2, and zip3 mRNAs is not regulated by dietary zinc in the intestine and visceral endoderm, tissues involved in nutrient absorption. These studies suggest that all three of these ZIP proteins may play cell-specific roles in zinc homeostasis rather than primary roles in the acquisition of dietary zinc.


Assuntos
Proteínas de Transporte/química , Proteínas de Transporte/genética , Proteínas de Transporte/fisiologia , Sequência de Aminoácidos , Animais , Transporte Biológico , Northern Blotting , Proteínas de Transporte de Cátions , Cátions , Linhagem Celular , Células Cultivadas , DNA Complementar/metabolismo , Relação Dose-Resposta a Droga , Feminino , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/metabolismo , Masculino , Camundongos , Modelos Genéticos , Dados de Sequência Molecular , Plasmídeos/metabolismo , Estrutura Terciária de Proteína , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade , Fatores de Tempo , Distribuição Tecidual , Transfecção , Zinco/química , Zinco/deficiência , Zinco/metabolismo
16.
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
17.
J Nutr ; 133(1): 45-50, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12514265

RESUMO

Herein, the function of pancreatic metallothionein (MT)-I during zinc deficiency in pregnancy was examined using transgenic mice, which constitutively express the mouse MT-I gene driven by the rat elastase I promoter. Pancreatic MT protein levels and zinc levels were elevated significantly in the transgenic mice compared with those in control mice. Pregnant transgenic and control mice were fed zinc-deficient (1 micro g/g beginning at d 8) or zinc-adequate (50 micro g/g) diets during pregnancy, and the effects on the morphology of embryos were determined at d 14 of pregnancy (d 1 = vaginal plug). As other indicators of zinc deficiency, maternal pancreatic MT levels, as well as the expression of zinc-regulated genes in the embryonic visceral yolk sac were examined. Under these experimental conditions of moderate dietary zinc deficiency, 21.3% of the embryos in control mice exhibited morphological defects, whereas only 5.8% of the embryos in the elastase-MT-I transgenic females had developed abnormally by d 14. Surprisingly, dietary zinc deficiency caused a >95% decrease in pancreatic MT protein concentration in these transgenic mice. This suggests the post-transcriptional control of MT protein levels during zinc deficiency because the rat elastase I promoter is not metal-regulated. The decrease in pancreatic MT protein levels was paralleled by a dramatic decrease in the relative abundance of MT-I mRNA and a dramatic increase in the relative abundance of the zinc/iron regulated transporter-related zinc transporter-4 (ZIP4) mRNA in the embryonic visceral yolk sac. Thus, the constitutive overexpression of pancreatic MT-I in these mice attenuated, but did not prevent the effects of maternal or embryonic zinc deficiency under these conditions. Overall, these findings are consistent with the hypothesis that mouse pancreatic MT-I may participate in providing a labile pool of maternal zinc for the developing embryo during periods of zinc deficiency.


Assuntos
Metalotioneína/fisiologia , Pancrelipase/fisiologia , Zinco/deficiência , Animais , Dieta , Feminino , Genótipo , Homeostase , Metalotioneína/genética , Camundongos , Camundongos Transgênicos , Pancrelipase/metabolismo , Gravidez , Zinco/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA