Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Traffic ; 25(1): e12920, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37886910

RESUMEN

Wilson disease (WD) is caused by mutations in the ATP7B gene that encodes a copper (Cu) transporting ATPase whose trafficking from the Golgi to endo-lysosomal compartments drives sequestration of excess Cu and its further excretion from hepatocytes into the bile. Loss of ATP7B function leads to toxic Cu overload in the liver and subsequently in the brain, causing fatal hepatic and neurological abnormalities. The limitations of existing WD therapies call for the development of new therapeutic approaches, which require an amenable animal model system for screening and validation of drugs and molecular targets. To achieve this objective, we generated a mutant Caenorhabditis elegans strain with a substitution of a conserved histidine (H828Q) in the ATP7B ortholog cua-1 corresponding to the most common ATP7B variant (H1069Q) that causes WD. cua-1 mutant animals exhibited very poor resistance to Cu compared to the wild-type strain. This manifested in a strong delay in larval development, a shorter lifespan, impaired motility, oxidative stress pathway activation, and mitochondrial damage. In addition, morphological analysis revealed several neuronal abnormalities in cua-1 mutant animals exposed to Cu. Further investigation suggested that mutant CUA-1 is retained and degraded in the endoplasmic reticulum, similarly to human ATP7B-H1069Q. As a consequence, the mutant protein does not allow animals to counteract Cu toxicity. Notably, pharmacological correctors of ATP7B-H1069Q reduced Cu toxicity in cua-1 mutants indicating that similar pathogenic molecular pathways might be activated by the H/Q substitution and, therefore, targeted for rescue of ATP7B/CUA-1 function. Taken together, our findings suggest that the newly generated cua-1 mutant strain represents an excellent model for Cu toxicity studies in WD.


Asunto(s)
Degeneración Hepatolenticular , Animales , Humanos , Degeneración Hepatolenticular/genética , Degeneración Hepatolenticular/tratamiento farmacológico , Degeneración Hepatolenticular/metabolismo , Cobre/toxicidad , Cobre/metabolismo , Caenorhabditis elegans/genética , Caenorhabditis elegans/metabolismo , ATPasas Transportadoras de Cobre/genética , ATPasas Transportadoras de Cobre/metabolismo , Hepatocitos/metabolismo
2.
J Clin Invest ; 133(1)2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36301669

RESUMEN

Signaling circuits crucial to systemic physiology are widespread, yet uncovering their molecular underpinnings remains a barrier to understanding the etiology of many metabolic disorders. Here, we identified a copper-linked signaling circuit activated by disruption of mitochondrial function in the murine liver or heart that resulted in atrophy of the spleen and thymus and caused a peripheral white blood cell deficiency. We demonstrated that the leukopenia was caused by α-fetoprotein, which required copper and the cell surface receptor CCR5 to promote white blood cell death. We further showed that α-fetoprotein expression was upregulated in several cell types upon inhibition of oxidative phosphorylation. Collectively, our data argue that α-fetoprotein may be secreted by bioenergetically stressed tissue to suppress the immune system, an effect that may explain the recurrent or chronic infections that are observed in a subset of mitochondrial diseases or in other disorders with secondary mitochondrial dysfunction.


Asunto(s)
Cobre , Enfermedades Mitocondriales , Ratones , Animales , Cobre/metabolismo , alfa-Fetoproteínas/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Enfermedades Mitocondriales/metabolismo , Terapia de Inmunosupresión
3.
Front Cell Dev Biol ; 10: 856300, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35433682

RESUMEN

Copper (Cu) is an essential trace element for key biochemical reactions. Dietary or genetic copper deficiencies are associated with anemia, cardiomyopathy, and neurodegeneration. The essential requirement for copper in humans is illustrated by Menkes disease, a fatal neurodegenerative disorder of early childhood caused by mutations in the ATP7A copper transporter. Recent groundbreaking studies have demonstrated that a copper delivery small molecule compound, elesclomol (ES), is able to substantially ameliorate pathology and lethality in a mouse model of Menkes disease when injected as an ES-Cu2+ complex. It is well appreciated that drugs administered through oral means are more convenient with better efficacy than injection methods. Here we show, using genetic models of copper-deficient C. elegans and mice, that dietary ES supplementation fully rescues copper deficiency phenotypes. Worms lacking either the homolog of the CTR1 copper importer or the ATP7 copper exporter showed normal development when fed ES. Oral gavage with ES rescued intestine-specific Ctr1 knockout mice from early postnatal lethality without additional copper supplementation. Our findings reveal that ES facilitates copper delivery from dietary sources independent of the intestinal copper transporter CTR1 and provide insight into oral administration of ES as an optimal therapeutic for Menkes disease and possibly other disorders of copper insufficiency.

4.
Eur J Med Chem ; 212: 113103, 2021 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-33387904

RESUMEN

The Kelch-like ECH-associated protein 1 (Keap1)-Nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway plays a pivotal role in the cellular defense system against oxidative stress by inducing antioxidant and anti-inflammatory effects. We previously developed Nrf2 activators that potentially protect the death of dopaminergic (DAergic) neuronal cells against oxidative stress in Parkinson's disease (PD). In this study, we designed and synthesized a class of halogenated vinyl sulfones by inserting halogens and pyridine to maximize Nrf2 activation efficacy. Among the synthesized compounds, (E)-3-chloro-2-(2-((2-chlorophenyl)sulfonyl)vinyl)pyridine (9d) significantly exhibited potent Nrf2 activating efficacy (9d: EC50 = 26 nM) at least 10-fold compared with the previous developed compounds (1 and 2). Furthermore, treating with 9d remarkably increased Nrf2 nuclear translocation and Nrf2 protein levels in microglial BV-2 cells. 9d was shown to induce the expression of antioxidant response genes HO-1, GCLC, GCLM, and SOD-1 at both the mRNA and protein levels and suppress proinflammatory cytokines and enzymes. Also, 9d remarkably protected DAergic neurons and restored the PD-associated motor dysfunction in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced mouse model.


Asunto(s)
Desarrollo de Medicamentos , Factor 2 Relacionado con NF-E2/metabolismo , Enfermedad de Parkinson/tratamiento farmacológico , Sulfonas/farmacología , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Halogenación , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Estructura Molecular , Enfermedad de Parkinson/metabolismo , Relación Estructura-Actividad , Sulfonas/síntesis química , Sulfonas/química
5.
Science ; 368(6491): 620-625, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32381719

RESUMEN

Loss-of-function mutations in the copper (Cu) transporter ATP7A cause Menkes disease. Menkes is an infantile, fatal, hereditary copper-deficiency disorder that is characterized by progressive neurological injury culminating in death, typically by 3 years of age. Severe copper deficiency leads to multiple pathologies, including impaired energy generation caused by cytochrome c oxidase dysfunction in the mitochondria. Here we report that the small molecule elesclomol escorted copper to the mitochondria and increased cytochrome c oxidase levels in the brain. Through this mechanism, elesclomol prevented detrimental neurodegenerative changes and improved the survival of the mottled-brindled mouse-a murine model of severe Menkes disease. Thus, elesclomol holds promise for the treatment of Menkes and associated disorders of hereditary copper deficiency.


Asunto(s)
Cobre/metabolismo , Hidrazinas/uso terapéutico , Síndrome del Pelo Ensortijado/tratamiento farmacológico , Animales , Transporte Biológico/efectos de los fármacos , Encéfalo/metabolismo , Encéfalo/patología , Línea Celular , Transportador de Cobre 1/genética , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/metabolismo , Hidrazinas/farmacología , Masculino , Síndrome del Pelo Ensortijado/metabolismo , Síndrome del Pelo Ensortijado/patología , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/prevención & control , Ratas
6.
J Biol Chem ; 294(8): 2815-2826, 2019 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-30593504

RESUMEN

Zinc is an essential trace element that serves as a cofactor for enzymes in critical biochemical processes and also plays a structural role in numerous proteins. Zinc transporter ZIP4 (ZIP4) is a zinc importer required for dietary zinc uptake in the intestine and other cell types. Studies in cultured cells have reported that zinc stimulates the endocytosis of plasma membrane-localized ZIP4 protein, resulting in reduced cellular zinc uptake. Thus, zinc-regulated trafficking of ZIP4 is a key means for regulating cellular zinc homeostasis, but the underlying mechanisms are not well understood. In this study, we used mutational analysis, immunoblotting, HEK293 cells, and immunofluorescence microscopy to identify a histidine-containing motif (398HTH) in the first extracellular loop that is required for high sensitivity to low zinc concentrations in a zinc-induced endocytic response of mouse ZIP4 (mZIP4). Moreover, using synthetic peptides with selective substitutions and truncated mZIP4 variants, we provide evidence that histidine residues in this motif coordinate a zinc ion in mZIP4 homodimers at the plasma membrane. These findings suggest that 398HTH is an important zinc-sensing motif for eliciting high-affinity zinc-stimulated endocytosis of mZIP4 and provide insight into cellular mechanisms for regulating cellular zinc homeostasis in mammalian cells.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Endocitosis/fisiología , Matriz Extracelular/metabolismo , Histidina/química , Proteínas Mutantes/metabolismo , Mutación , Zinc/farmacología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Proteínas de Transporte de Catión/química , Proteínas de Transporte de Catión/genética , Membrana Celular/metabolismo , Endocitosis/efectos de los fármacos , Células HEK293 , Histidina/metabolismo , Humanos , Proteínas Mutantes/química , Proteínas Mutantes/genética , Transporte de Proteínas , Homología de Secuencia
7.
Proc Natl Acad Sci U S A ; 115(32): 8161-8166, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30038027

RESUMEN

Copper is an essential cofactor of cytochrome c oxidase (CcO), the terminal enzyme of the mitochondrial respiratory chain. Inherited loss-of-function mutations in several genes encoding proteins required for copper delivery to CcO result in diminished CcO activity and severe pathologic conditions in affected infants. Copper supplementation restores CcO function in patient cells with mutations in two of these genes, COA6 and SCO2, suggesting a potential therapeutic approach. However, direct copper supplementation has not been therapeutically effective in human patients, underscoring the need to identify highly efficient copper transporting pharmacological agents. By using a candidate-based approach, we identified an investigational anticancer drug, elesclomol (ES), that rescues respiratory defects of COA6-deficient yeast cells by increasing mitochondrial copper content and restoring CcO activity. ES also rescues respiratory defects in other yeast mutants of copper metabolism, suggesting a broader applicability. Low nanomolar concentrations of ES reinstate copper-containing subunits of CcO in a zebrafish model of copper deficiency and in a series of copper-deficient mammalian cells, including those derived from a patient with SCO2 mutations. These findings reveal that ES can restore intracellular copper homeostasis by mimicking the function of missing transporters and chaperones of copper, and may have potential in treating human disorders of copper metabolism.


Asunto(s)
Antineoplásicos/farmacología , Cobre/deficiencia , Drogas en Investigación/farmacología , Complejo IV de Transporte de Electrones/metabolismo , Hidrazinas/farmacología , Mitocondrias/efectos de los fármacos , Animales , Antineoplásicos/uso terapéutico , Transporte Biológico/genética , Proteínas Portadoras/genética , Línea Celular , Coenzimas/deficiencia , Cobre/uso terapéutico , Transportador de Cobre 1 , Suplementos Dietéticos , Modelos Animales de Enfermedad , Reposicionamiento de Medicamentos , Drogas en Investigación/uso terapéutico , Fibroblastos , Humanos , Hidrazinas/uso terapéutico , Proteínas de Transporte de Membrana/genética , Errores Innatos del Metabolismo/tratamiento farmacológico , Errores Innatos del Metabolismo/genética , Errores Innatos del Metabolismo/metabolismo , Mitocondrias/metabolismo , Proteínas Mitocondriales/genética , Chaperonas Moleculares , Mutagénesis Sitio-Dirigida , Mutación , Ratas , Saccharomyces cerevisiae , Pez Cebra , Proteínas de Pez Cebra/genética
8.
J Biol Chem ; 293(28): 10911-10925, 2018 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-29784876

RESUMEN

Copper plays key roles in catalytic and regulatory biochemical reactions essential for normal growth, development, and health. Dietary copper deficiencies or mutations in copper homeostasis genes can lead to abnormal musculoskeletal development, cognitive disorders, and poor growth. In yeast and mammals, copper is acquired through the activities of the CTR1 family of high-affinity copper transporters. However, the mechanisms of systemic responses to dietary or tissue-specific copper deficiency remain unclear. Here, taking advantage of the animal model Caenorhabditis elegans for studying whole-body copper homeostasis, we investigated the role of a C. elegans CTR1 homolog, CHCA-1, in copper acquisition and in worm growth, development, and behavior. Using sequence homology searches, we identified 10 potential orthologs to mammalian CTR1 Among these genes, we found that chca-1, which is transcriptionally up-regulated in the intestine and hypodermis of C. elegans during copper deficiency, is required for normal growth, reproduction, and maintenance of systemic copper balance under copper deprivation. The intestinal copper transporter CUA-1 normally traffics to endosomes to sequester excess copper, and we found here that loss of chca-1 caused CUA-1 to mislocalize to the basolateral membrane under copper overload conditions. Moreover, animals lacking chca-1 exhibited significantly reduced copper avoidance behavior in response to toxic copper conditions compared with WT worms. These results establish that CHCA-1-mediated copper acquisition in C. elegans is crucial for normal growth, development, and copper-sensing behavior.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/crecimiento & desarrollo , Caenorhabditis elegans/metabolismo , Proteínas de Transporte de Catión/metabolismo , Membrana Celular/metabolismo , Cobre/metabolismo , Animales , Proteínas de Caenorhabditis elegans/genética , Proteínas de Transporte de Catión/genética , Transportador de Cobre 1 , Homeostasis , Intestinos , Transporte Iónico , Reproducción , Activación Transcripcional
9.
Gastroenterology ; 154(1): 168-180.e5, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-28958857

RESUMEN

BACKGROUND & AIMS: Wilson disease is a disorder of copper (Cu) misbalance caused by mutations in ATP7B. ATP7B is highly expressed in the liver-the major site of Cu accumulation in patients with Wilson disease. The intestine also expresses ATP7B, but little is known about the contribution of intestinal ATP7B to normal intestinal copper homeostasis or to Wilson disease manifestations. We characterized the role of ATP7B in mouse intestinal organoids and tissues. METHODS: We collected intestinal tissues from ATP7B-knockout (Atp7b-/-) and control mice, and established 3-dimensional enteroids. Immunohistochemistry and x-ray fluorescence were used to characterize the distribution of ATP7B and Cu in tissues. Electron microscopy, histologic analyses, and immunoblotting were used to determine the effects of ATP7B loss. Enteroids derived from control and ATP7B-knockout mice were incubated with excess Cu or with Cu-chelating reagents; effects on cell fat content and ATP7B levels and localization were determined by fluorescent confocal microscopy. RESULTS: ATP7B maintains a Cu gradient along the duodenal crypt-villus axis and buffers Cu levels in the cytosol of enterocytes. These functions are mediated by rapid Cu-dependent enlargement of ATP7B-containing vesicles and increased levels of ATP7B. Intestines of Atp7b-/- mice had reduced Cu storage pools in intestine, Cu depletion, accumulation of triglyceride-filled vesicles in enterocytes, mislocalization of apolipoprotein B, and loss of chylomicrons. In primary 3-dimensional enteroids, administration of excess Cu or Cu chelators impaired assembly of chylomicrons. CONCLUSIONS: ATP7B regulates vesicular storage of Cu in mouse intestine. ATP7B buffers Cu levels in enterocytes to maintain a range necessary for formation of chylomicrons. Misbalance of Cu and lipid in the intestine could account for gastrointestinal manifestations of Wilson disease.


Asunto(s)
ATPasas Transportadoras de Cobre/metabolismo , Degeneración Hepatolenticular/etiología , Degeneración Hepatolenticular/metabolismo , Intestinos/enzimología , Animales , Modelos Animales de Enfermedad , Femenino , Degeneración Hepatolenticular/patología , Intestinos/patología , Masculino , Ratones , Ratones Noqueados
10.
Hum Mol Genet ; 26(23): 4617-4628, 2017 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-28973536

RESUMEN

SCO1 is a ubiquitously expressed, mitochondrial protein with essential roles in cytochrome c oxidase (COX) assembly and the regulation of copper homeostasis. SCO1 patients present with severe forms of early onset disease, and ultimately succumb from liver, heart or brain failure. However, the inherent susceptibility of these tissues to SCO1 mutations and the clinical heterogeneity observed across SCO1 pedigrees remain poorly understood phenomena. To further address this issue, we generated Sco1hrt/hrt and Sco1stm/stm mice in which Sco1 was specifically deleted in heart and striated muscle, respectively. Lethality was observed in both models due to a combined COX and copper deficiency that resulted in a dilated cardiomyopathy. Left ventricular dilation and loss of heart function was preceded by a temporal decrease in COX activity and copper levels in the longer-lived Sco1stm/stm mice. Interestingly, the reduction in copper content of Sco1stm/stm cardiomyocytes was due to the mislocalisation of CTR1, the high affinity transporter that imports copper into the cell. CTR1 was similarly mislocalized to the cytosol in the heart of knockin mice carrying a homozygous G115S substitution in Sco1, which in humans causes a hypertrophic cardiomyopathy. Our current findings in the heart are in marked contrast to our prior observations in the liver, where Sco1 deletion results in a near complete absence of CTR1 protein. These data collectively argue that mutations perturbing SCO1 function have tissue-specific consequences for the machinery that ultimately governs copper homeostasis, and further establish the importance of aberrant mitochondrial signaling to the etiology of copper handling disorders.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Animales , Cardiomiopatía Hipertrófica/genética , Cardiomiopatía Hipertrófica/metabolismo , Membrana Celular/metabolismo , Cobre/deficiencia , Transportador de Cobre 1 , Modelos Animales de Enfermedad , Complejo IV de Transporte de Electrones/genética , Homeostasis , Transporte Iónico , Metalochaperonas/genética , Metalochaperonas/metabolismo , Ratones , Ratones Transgénicos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Chaperonas Moleculares , Miocitos Cardíacos/metabolismo , Oxidación-Reducción , Transducción de Señal
11.
Sci Rep ; 7(1): 12001, 2017 09 20.
Artículo en Inglés | MEDLINE | ID: mdl-28931909

RESUMEN

Copper (Cu) is an essential cofactor for various enzymatic activities including mitochondrial electron transport, iron mobilization, and peptide hormone maturation. Consequently, Cu dysregulation is associated with fatal neonatal disease, liver and cardiac dysfunction, and anemia. While the Cu transporter ATP7A plays a major role in both intestinal Cu mobilization to the periphery and prevention of Cu over-accumulation, it is unclear how regulation of ATP7A contributes to Cu homeostasis in response to systemic Cu fluctuation. Here we show, using Cu-deficient mouse models, that steady-state levels of ATP7A are lower in peripheral tissues (including the heart, spleen, and liver) under Cu deficiency and that subcutaneous administration of Cu to these animals restore normal ATP7A levels in these tissues. Strikingly, ATP7A in the intestine is regulated in the opposite manner - low systemic Cu increases ATP7A while subcutaneous Cu administration decreases ATP7A suggesting that intestine-specific non-autonomous regulation of ATP7A abundance may serve as a key homeostatic control for Cu export into the circulation. Our results support a systemic model for how a single transporter can be inversely regulated in a tissue-specific manner to maintain organismal Cu homeostasis.


Asunto(s)
ATPasas Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Homeostasis , Especificidad de Órganos , Animales , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Células Cultivadas , Cobre/deficiencia , Cobre/farmacología , Transportador de Cobre 1 , ATPasas Transportadoras de Cobre/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Hígado/citología , Hígado/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Fenantrolinas/farmacología , Ratas
12.
J Biol Chem ; 292(1): 1-14, 2017 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-27881675

RESUMEN

Copper plays key catalytic and regulatory roles in biochemical processes essential for normal growth, development, and health. Defects in copper metabolism cause Menkes and Wilson's disease, myeloneuropathy, and cardiovascular disease and are associated with other pathophysiological states. Consequently, it is critical to understand the mechanisms by which organisms control the acquisition, distribution, and utilization of copper. The intestinal enterocyte is a key regulatory point for copper absorption into the body; however, the mechanisms by which intestinal cells transport copper to maintain organismal copper homeostasis are poorly understood. Here, we identify a mechanism by which organismal copper homeostasis is maintained by intestinal copper exporter trafficking that is coordinated with extraintestinal copper levels in Caenorhabditis elegans Specifically, we show that CUA-1, the C. elegans homolog of ATP7A/B, localizes to lysosome-like organelles (gut granules) in the intestine under copper overload conditions for copper detoxification, whereas copper deficiency results in a redistribution of CUA-1 to basolateral membranes for copper efflux to peripheral tissues. Worms defective in gut granule biogenesis exhibit defects in copper sequestration and increased susceptibility to toxic copper levels. Interestingly, however, a splice isoform CUA-1.2 that lacks a portion of the N-terminal domain is targeted constitutively to the basolateral membrane irrespective of dietary copper concentration. Our studies establish that CUA-1 is a key intestinal copper exporter and that its trafficking is regulated to maintain systemic copper homeostasis. C. elegans could therefore be exploited as a whole-animal model system to study regulation of intra- and intercellular copper trafficking pathways.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Transporte de Catión/metabolismo , Membrana Celular/metabolismo , Cobre/metabolismo , Homeostasis , Mucosa Intestinal/metabolismo , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Intestinos/crecimiento & desarrollo , Transporte de Proteínas
13.
Proc Natl Acad Sci U S A ; 110(46): E4279-88, 2013 Nov 12.
Artículo en Inglés | MEDLINE | ID: mdl-24167251

RESUMEN

Copper is an essential catalytic cofactor for enzymatic activities that drive a range of metabolic biochemistry including mitochondrial electron transport, iron mobilization, and peptide hormone maturation. Copper dysregulation is associated with fatal infantile disease, liver, and cardiac dysfunction, neuropathy, and anemia. Here we report that mammals regulate systemic copper acquisition and intracellular mobilization via cleavage of the copper-binding ecto-domain of the copper transporter 1 (Ctr1). Although full-length Ctr1 is critical to drive efficient copper import across the plasma membrane, cleavage of the ecto-domain is required for Ctr1 to mobilize endosomal copper stores. The biogenesis of the truncated form of Ctr1 requires the structurally related, previously enigmatic copper transporter 2 (Ctr2). Ctr2(-/-) mice are defective in accumulation of truncated Ctr1 and exhibit increased tissue copper levels, and X-ray fluorescence microscopy demonstrates that copper accumulates as intracellular foci. These studies identify a key regulatory mechanism for mammalian copper transport through Ctr2-dependent accumulation of a Ctr1 variant lacking the copper- and cisplatin-binding ecto-domain.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Animales , Transporte Biológico/fisiología , Southern Blotting , Proteínas de Transporte de Catión/biosíntesis , Proteínas de Transporte de Catión/genética , Cisplatino/metabolismo , Cobre/metabolismo , Transportador de Cobre 1 , Espectrometría de Masas , Ratones , Ratones Noqueados , Microscopía Fluorescente , Estructura Terciaria de Proteína/genética , Interferencia de ARN , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas SLC31
14.
Mol Cell Biol ; 32(7): 1284-95, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22290441

RESUMEN

Copper (Cu) is essential for development and proliferation, yet the cellular requirements for Cu in these processes are not well defined. We report that Cu plays an unanticipated role in the mitogen-activated protein (MAP) kinase pathway. Ablation of the Ctr1 high-affinity Cu transporter in flies and mouse cells, mutation of Ctr1, and Cu chelators all reduce the ability of the MAP kinase kinase Mek1 to phosphorylate the MAP kinase Erk. Moreover, mice bearing a cardiac-tissue-specific knockout of Ctr1 are deficient in Erk phosphorylation in cardiac tissue. in vitro investigations reveal that recombinant Mek1 binds two Cu atoms with high affinity and that Cu enhances Mek1 phosphorylation of Erk in a dose-dependent fashion. Coimmunoprecipitation experiments suggest that Cu is important for promoting the Mek1-Erk physical interaction that precedes the phosphorylation of Erk by Mek1. These results demonstrate a role for Ctr1 and Cu in activating a pathway well known to play a key role in normal physiology and in cancer.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Sistema de Señalización de MAP Quinasas , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Proteínas ras/metabolismo , Animales , Proteínas de Transporte de Catión/genética , Línea Celular , Transportador de Cobre 1 , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Activación Enzimática , Eliminación de Gen , MAP Quinasa Quinasa 1/metabolismo , Ratones , Fosforilación , Unión Proteica
15.
J Biol Chem ; 285(42): 32385-92, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20699218

RESUMEN

Copper is an essential trace element that functions in a diverse array of biochemical processes that include mitochondrial respiration, neurotransmitter biogenesis, connective tissue maturation, and reactive oxygen chemistry. The Ctr1 protein is a high-affinity Cu(+) importer that is structurally and functionally conserved in yeast, plants, fruit flies, and humans and that, in all of these organisms, is localized to the plasma membrane and intracellular vesicles. Although intestinal epithelial cell-specific deletion of Ctr1 in mice demonstrated a critical role for Ctr1 in dietary copper absorption, some controversy exists over the localization of Ctr1 in intestinal epithelial cells in vivo. In this work, we assess the localization of Ctr1 in intestinal epithelial cells through two independent mechanisms. Using immunohistochemistry, we demonstrate that Ctr1 localizes to the apical membrane in intestinal epithelial cells of the mouse, rat, and pig. Moreover, biotinylation of intestinal luminal proteins from mice fed a control or a copper-deficient diet showed elevated levels of both total and apical membrane Ctr1 protein in response to transient dietary copper limitation. Experiments in cultured HEK293T cells demonstrated that alterations in the levels of the glycosylated form of Ctr1 in response to copper availability were a time-dependent, copper-specific posttranslational response. Taken together, these results demonstrate apical localization of Ctr1 in intestinal epithelia across three mammalian species and suggest that increased Ctr1 apical localization in response to dietary copper limitation may represent an adaptive response to homeostatically modulate Ctr1 availability at the site of intestinal copper absorption.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/metabolismo , Células Epiteliales/metabolismo , Mucosa Intestinal/citología , Estabilidad Proteica , Secuencia de Aminoácidos , Animales , Proteínas de Transporte de Catión/genética , Polaridad Celular , Transportador de Cobre 1 , Dieta , Células Epiteliales/citología , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Ratas , Porcinos
16.
Cell Metab ; 11(5): 353-63, 2010 May 05.
Artículo en Inglés | MEDLINE | ID: mdl-20444417

RESUMEN

Copper (Cu) is an essential cofactor for a variety of metabolic functions, and the regulation of systemic Cu metabolism is critical to human health. Dietary Cu is absorbed through the intestine, stored in the liver, and mobilized into the circulation; however, systemic Cu homeostasis is poorly understood. We generated mice with a cardiac-specific knockout of the Ctr1 Cu transporter (Ctr1(hrt/hrt)), resulting in cardiac Cu deficiency and severe cardiomyopathy. Unexpectedly, Ctr1(hrt/hrt) mice exhibited increased serum Cu levels and a concomitant decrease in hepatic Cu stores. Expression of the ATP7A Cu exporter, thought to function predominantly in intestinal Cu acquisition, was strongly increased in liver and intestine of Ctr1(hrt/hrt) mice. These studies identify ATP7A as a candidate for hepatic Cu mobilization in response to peripheral tissue demand, and illuminate a systemic regulation in which the Cu status of the heart is signaled to organs that take up and store Cu.


Asunto(s)
Cobre/metabolismo , Miocardio/metabolismo , Transducción de Señal , Adenosina Trifosfatasas/metabolismo , Animales , Cardiomiopatía Dilatada/etiología , Proteínas de Transporte de Catión/deficiencia , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/metabolismo , Cobre/deficiencia , Transportador de Cobre 1 , ATPasas Transportadoras de Cobre , Drosophila/metabolismo , Mucosa Intestinal/metabolismo , Hígado/metabolismo , Ratones , Ratones Noqueados
17.
Nat Chem Biol ; 4(3): 176-85, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18277979

RESUMEN

Copper (Cu) is a redox-active metal ion essential for most aerobic organisms. Cu serves as a catalytic and structural cofactor for enzymes that function in energy generation, iron acquisition, oxygen transport, cellular metabolism, peptide hormone maturation, blood clotting, signal transduction and a host of other processes. The inability to control Cu balance is associated with genetic diseases of overload and deficiency and has recently been tied to neurodegenerative disorders and fungal virulence. The essential nature of Cu, the existence of human genetic disorders of Cu metabolism and the potential impact of Cu deposition in the environment have been driving forces for detailed investigations in microbial and eukaryotic model systems. Here we review recent advances in the identification and function of cellular and systemic molecules that drive Cu accumulation, distribution and sensing.


Asunto(s)
Cobre/metabolismo , Metaloproteínas/metabolismo , Animales , Transporte Biológico , Compartimento Celular , Cobre/química , Cryptococcus neoformans/metabolismo , Cryptococcus neoformans/patogenicidad , Células Eucariotas/química , Células Eucariotas/metabolismo , Humanos , Metaloproteínas/química , Mitocondrias/química , Mitocondrias/metabolismo , Chaperonas Moleculares/metabolismo , Virulencia
18.
J Med Genet ; 44(10): 641-6, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17483305

RESUMEN

Owing to mutations in the copper-transporting P-type ATPase, ATP7A (or MNK), patients with Menkes disease (MD) have an inadequate supply of copper to various copper-dependent enzymes. The ATP7A protein is located in the trans-Golgi network, where it transports copper via secretory compartments to copper-dependent enzymes. Raised copper concentrations result in the trafficking of ATP7A to the plasma membrane, where it functions in copper export. An important model of MD is the Mottled mouse, which possesses mutations in Atp7A. The Mottled mouse displays three distinct phenotypic severities: embryonic lethal, perinatal lethal and a longer-lived viable phenotype. However, the effects of mutations from these phenotypic classes on the ATP7A protein are unknown. In this study, we found that these classes of mutation differentially affect the copper transport and trafficking functions of the ATP7A protein. The embryonic lethal mutation, Atp7a(mo11H) (11H), caused mislocalisation of the protein to the endoplasmic reticulum, impaired glycosylation, and abolished copper delivery to the secretory pathway. In contrast, the perinatal lethal and viable mutations, Atp7a(moMac) (Macular) and Atp7a(moVbr) (Viable brindle) both resulted in a reduction in copper delivery to the secretory pathway and constitutive trafficking of the ATP7A protein to the plasma membrane in the absence of additional copper. In the case of Viable brindle, this hypertrafficking response was dependent on the catalytic phosphorylation site of ATP7A, whereas no such requirement was found for the Macular mutation. These findings provide evidence that the degree of MD severity in mice is associated with both copper transport and trafficking defects in the ATP7A protein.


Asunto(s)
Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/fisiología , Proteínas de Transporte de Catión/genética , Proteínas de Transporte de Catión/fisiología , Síndrome del Pelo Ensortijado/genética , Animales , ATPasas Transportadoras de Cobre , Retículo Endoplásmico/metabolismo , Aparato de Golgi/metabolismo , Ratones , Microscopía Fluorescente , Modelos Biológicos , Modelos Genéticos , Mutación , Fenotipo , Fosforilación , Plásmidos/metabolismo , Proteína Disulfuro Isomerasas/metabolismo , Transporte de Proteínas
19.
J Biol Chem ; 282(10): 6992-7000, 2007 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-17202136

RESUMEN

Zinc is an essential nutrient. Genetic evidence for this nutritional requirement in humans is the zinc deficiency disease, acrodermatitis enteropathica. This disorder is caused by mutations in hZIP4 (SLC39A4), a zinc importer required for zinc uptake in enterocytes and other cell types. Studies in mice have demonstrated that levels of the mZIP4 mRNA are reduced by elevated dietary zinc, resulting in a decreased abundance of the ZIP4 protein at the plasma membrane. Moreover, studies in cultured cells have demonstrated that low micromolar concentrations of zinc stimulate the endocytosis of the mZIP4 protein resulting in a reduction in cellular zinc uptake. In this study, we demonstrate an additional level of hZIP4 regulation involving ubiquitination and degradation of this transporter in elevated zinc concentrations. Mutational analysis identified a cytoplasmic histidine-rich domain that was essential for ubiquitin-dependent degradation of ZIP4 and protection against zinc toxicity. However, this motif was dispensable for zinc-induced endocytosis. These findings indicate that ubiquitin-mediated degradation of the ZIP4 protein is critical for regulating zinc homeostasis in response to the upper tier of physiological zinc concentrations, via a process that is distinct from zinc-stimulated endocytosis.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Citoprotección , Proteínas/fisiología , Ubiquitina/metabolismo , Zinc/toxicidad , Proteínas de Transporte de Catión/genética , Línea Celular , Endocitosis , Homeostasis , Humanos , ARN Mensajero/análisis , Zinc/metabolismo
20.
Cell Metab ; 4(3): 235-44, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16950140

RESUMEN

The trace element copper (Cu) is a cofactor for biochemical functions ranging from energy generation to iron (Fe) acquisition, angiogenesis, and free radical detoxification. While Cu is essential for life, the molecules that mediate dietary Cu uptake have not been identified. Ctr1 is a homotrimeric protein, conserved from yeast to humans, that transports Cu across the plasma membrane with high affinity and specificity. Here we describe the generation of intestinal epithelial cell-specific Ctr1 knockout mice. These mice exhibit striking neonatal defects in Cu accumulation in peripheral tissues, hepatic Fe overload, cardiac hypertrophy, and severe growth and viability defects. Consistent with an intestinal Cu absorption block, the growth and viability defects can be partially rescued by a single postnatal Cu administration, indicative of a critical neonatal metabolic requirement for Cu that is provided by intestinal Ctr1. These studies identify Ctr1 as the major factor driving intestinal Cu absorption in mammals.


Asunto(s)
Proteínas de Transporte de Catión/metabolismo , Cobre/deficiencia , Absorción Intestinal/genética , Mucosa Intestinal/metabolismo , Hierro/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Animales , Animales Recién Nacidos , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/fisiopatología , Proteínas de Transporte de Catión/genética , Membrana Celular/metabolismo , Cobre/farmacología , Transportador de Cobre 1 , Modelos Animales de Enfermedad , Enanismo/genética , Enanismo/metabolismo , Enanismo/fisiopatología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/metabolismo , Cardiopatías Congénitas/fisiopatología , Mucosa Intestinal/fisiopatología , Intestinos/fisiopatología , Síndromes de Malabsorción/genética , Síndromes de Malabsorción/metabolismo , Síndromes de Malabsorción/fisiopatología , Proteínas de Transporte de Membrana/genética , Ratones , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...