Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Metallomics ; 8(9): 981-92, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27293072

ABSTRACT

ATP7A is a P-type ATPase essential for cellular copper (Cu) transport and homeostasis. Loss-of-function ATP7A mutations causing systemic Cu deficiency are associated with severe Menkes disease or its milder allelic variant, occipital horn syndrome. We previously identified two rare ATP7A missense mutations (P1386S and T994I) leading to a non-fatal form of motor neuron disorder, X-linked distal hereditary motor neuropathy (dHMNX), without overt signs of systemic Cu deficiency. Recent investigations using a tissue specific Atp7a knock out model have demonstrated that Cu plays an essential role in motor neuron maintenance and function, however the underlying pathogenic mechanisms of ATP7A mutations causing axonal degeneration remain unknown. We have generated an Atp7a conditional knock in mouse model of dHMNX expressing Atp7a(T985I), the orthologue of the human ATP7A(T994I) identified in dHMNX patients. Although a degenerative motor phenotype is not observed, the knock in Atp7a(T985I/Y) mice show altered Cu levels within the peripheral and central nervous systems, an increased diameter of the muscle fibres and altered myogenin and myostatin gene expression. Atp7a(T985I/Y) mice have reduced Atp7a protein levels and recapitulate the defective trafficking and altered post-translational regulatory mechanisms observed in the human ATP7A(T994I) patient fibroblasts. Our model provides a unique opportunity to characterise the molecular phenotype of dHMNX and the time course of cellular events leading to the process of axonal degeneration in this disease.


Subject(s)
Copper-Transporting ATPases/genetics , Copper/metabolism , Genetic Diseases, X-Linked/pathology , Motor Neuron Disease/pathology , Mutation , Animals , Behavior, Animal , Cells, Cultured , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Motor Neuron Disease/genetics , Motor Neuron Disease/metabolism , Myogenin/metabolism , Myostatin/metabolism
2.
Oncotarget ; 7(24): 37064-37080, 2016 Jun 14.
Article in English | MEDLINE | ID: mdl-27175597

ABSTRACT

Copper-ionophores that elevate intracellular bioavailable copper display significant therapeutic utility against prostate cancer cells in vitro and in TRAMP (Transgenic Adenocarcinoma of Mouse Prostate) mice. However, the pharmacological basis for their anticancer activity remains unclear, despite impending clinical trails. Herein we show that intracellular copper levels in prostate cancer, evaluated in vitro and across disease progression in TRAMP mice, were not correlative with copper-ionophore activity and mirrored the normal levels observed in patient prostatectomy tissues (Gleason Score 7 & 9). TRAMP adenocarcinoma cells harbored markedly elevated oxidative stress and diminished glutathione (GSH)-mediated antioxidant capacity, which together conferred selective sensitivity to prooxidant ionophoric copper. Copper-ionophore treatments [CuII(gtsm), disulfiram & clioquinol] generated toxic levels of reactive oxygen species (ROS) in TRAMP adenocarcinoma cells, but not in normal mouse prostate epithelial cells (PrECs). Our results provide a basis for the pharmacological activity of copper-ionophores and suggest they are amendable for treatment of patients with prostate cancer. Additionally, recent in vitro and mouse xenograft studies have suggested an increased copper requirement by prostate cancer cells. We demonstrated that prostate adenocarcinoma development in TRAMP mice requires a functional supply of copper and is significantly impeded by altered systemic copper distribution. The presence of a mutant copper-transporting Atp7b protein (tx mutation: A4066G/Met1356Val) in TRAMP mice changed copper-integration into serum and caused a remarkable reduction in prostate cancer burden (64% reduction) and disease severity (grade), abrogating adenocarcinoma development. Implications for current clinical trials are discussed.


Subject(s)
Adenocarcinoma/metabolism , Antineoplastic Agents/pharmacology , Copper/metabolism , Ionophores/pharmacology , Prostatic Neoplasms/metabolism , Animals , Copper-Transporting ATPases/genetics , Male , Mice , Mice, Transgenic
3.
Neurochem Res ; 41(3): 554-67, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26318862

ABSTRACT

Mitochondrial dysfunction, ubiquitin-proteasomal system impairment and excitotoxicity occur during the injury and death of neurons in neurodegenerative conditions. The aim of this work was to elucidate the cellular mechanisms that are universally altered by these conditions. Through overlapping expression profiles of rotenone-, lactacystin- and N-methyl-D-aspartate-treated cortical neurons, we have identified three affected biological processes that are commonly affected; oxidative stress, dysfunction of calcium signalling and inhibition of the autophagic-lysosomal pathway. These data provides many opportunities for therapeutic intervention in neurodegenerative conditions, where mitochondrial dysfunction, proteasomal inhibition and excitotoxicity are evident.


Subject(s)
Autophagy , Calcium Signaling , Lysosomes/metabolism , Neurons/metabolism , Oxidative Stress , Acetylcysteine/analogs & derivatives , Acetylcysteine/toxicity , Animals , Humans , Microarray Analysis , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology , Neurons/drug effects , Pesticides/toxicity , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/toxicity , Receptors, N-Methyl-D-Aspartate/metabolism , Rotenone/toxicity , Ubiquitin/metabolism
4.
J Neurosci ; 34(23): 8021-31, 2014 Jun 04.
Article in English | MEDLINE | ID: mdl-24899723

ABSTRACT

Mutations in the metallo-protein Cu/Zn-superoxide dismutase (SOD1) cause amyotrophic lateral sclerosis (ALS) in humans and an expression level-dependent phenotype in transgenic rodents. We show that oral treatment with the therapeutic agent diacetyl-bis(4-methylthiosemicarbazonato)copper(II) [Cu(II)(atsm)] increased the concentration of mutant SOD1 (SOD1G37R) in ALS model mice, but paradoxically improved locomotor function and survival of the mice. To determine why the mice with increased levels of mutant SOD1 had an improved phenotype, we analyzed tissues by mass spectrometry. These analyses revealed most SOD1 in the spinal cord tissue of the SOD1G37R mice was Cu deficient. Treating with Cu(II)(atsm) decreased the pool of Cu-deficient SOD1 and increased the pool of fully metallated (holo) SOD1. Tracking isotopically enriched (65)Cu(II)(atsm) confirmed the increase in holo-SOD1 involved transfer of Cu from Cu(II)(atsm) to SOD1, suggesting the improved locomotor function and survival of the Cu(II)(atsm)-treated SOD1G37R mice involved, at least in part, the ability of the compound to improve the Cu content of the mutant SOD1. This was supported by improved survival of SOD1G37R mice that expressed the human gene for the Cu uptake protein CTR1. Improving the metal content of mutant SOD1 in vivo with Cu(II)(atsm) did not decrease levels of misfolded SOD1. These outcomes indicate the metal content of SOD1 may be a greater determinant of the toxicity of the protein in mutant SOD1-associated forms of ALS than the mutations themselves. Improving the metal content of SOD1 therefore represents a valid therapeutic strategy for treating ALS caused by SOD1.


Subject(s)
Amyotrophic Lateral Sclerosis , Motor Neurons/drug effects , Mutation/genetics , Organometallic Compounds/administration & dosage , Superoxide Dismutase/genetics , Thiosemicarbazones/administration & dosage , Administration, Oral , Age Factors , Amyotrophic Lateral Sclerosis/drug therapy , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/mortality , Amyotrophic Lateral Sclerosis/pathology , Animals , Cation Transport Proteins/genetics , Chromatography, Gel , Coordination Complexes , Copper Transporter 1 , Disease Models, Animal , Humans , Locomotion/drug effects , Locomotion/genetics , Mice , Mice, Transgenic , Phenotype , Spinal Cord/drug effects , Spinal Cord/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1
5.
Diabetes ; 62(11): 3839-50, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23884884

ABSTRACT

Oxidative stress and endothelial dysfunction contribute to vascular complication in diabetes. Extracellular superoxide dismutase (SOD3) is one of the key antioxidant enzymes that obtains copper via copper transporter ATP7A. SOD3 is secreted from vascular smooth muscles cells (VSMCs) and anchors at the endothelial surface. The role of SOD3 and ATP7A in endothelial dysfunction in type 1 diabetes mellitus (T1DM) is entirely unknown. Here we show that the specific activity of SOD3, but not SOD1, is decreased, which is associated with increased O2(•-) production in aortas of streptozotocin-induced and genetically induced Ins2(Akita) T1DM mice. Exogenous copper partially rescued SOD3 activity in isolated T1DM vessels. Functionally, acetylcholine-induced, endothelium-dependent relaxation is impaired in T1DM mesenteric arteries, which is rescued by SOD mimetic tempol or gene transfer of SOD3. Mechanistically, ATP7A expression in T1DM vessels is dramatically decreased whereas other copper transport proteins are not altered. T1DM-induced endothelial dysfunction and decrease of SOD3 activity are rescued in transgenic mice overexpressing ATP7A. Furthermore, SOD3-deficient T1DM mice or ATP7A mutant T1DM mice augment endothelial dysfunction and vascular O2(•-) production versus T1DM mice. These effects are in part due to hypoinsulinemia in T1DM mice, since insulin treatment, but not high glucose, increases ATP7A expression in VSMCs and restores SOD3 activity in the organoid culture of T1DM vessels. In summary, a decrease in ATP7A protein expression contributes to impaired SOD3 activity, resulting in O2(•-) overproduction and endothelial dysfunction in blood vessels of T1DM. Thus, restoring copper transporter function is an essential therapeutic approach for oxidant stress-dependent vascular and metabolic diseases.


Subject(s)
Adenosine Triphosphatases/physiology , Cation Transport Proteins/physiology , Copper/metabolism , Superoxide Dismutase/metabolism , Adenosine Triphosphatases/genetics , Animals , Aorta/metabolism , Cation Transport Proteins/genetics , Cells, Cultured , Copper/pharmacology , Copper-Transporting ATPases , Cyclic N-Oxides/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Insulin/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , Rats , Spin Labels , Superoxide Dismutase/genetics , Superoxides/metabolism , Transfection
6.
Neurochem Int ; 62(5): 719-30, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23291249

ABSTRACT

Excitotoxicity, induced by the aberrant rise in cytosolic Ca(2+) level, is a major neuropathological process in numerous neurodegenerative disorders. It is triggered when extracellular glutamate (Glu) concentration reaches neuropathological levels resulting in dysregulation and hyper-activation of ionotropic glutamate receptor subtype (iGluRs). Even though all three members of the iGluRs, namely N-methyl-d-aspartate (NMDAR), α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPAR) and kainate (KAR) receptors are implicated in excitotoxicity, their individual contributions to downstream signaling transduction have not been explored. In this study, we report a comprehensive description of the recruitment of cellular processes in neurons upon iGluR activation during excitotoxicity through temporal (5h, 15h, and 24h) global gene profiling of AMPA, KA, NMDA, and Glu excitotoxic models. DNA microarray analyses of mouse primary cortical neurons treated with these four pharmacological agonists are further validated via real-time PCR. Bi-model analyses against Glu model demonstrate that NMDARs and KARs play a more pivotal role in Glu-mediated excitotoxicity, with a higher degree of global gene profiling overlaps, as compared to that of AMPARs. Comparison of global transcriptomic profiles reveals aberrant calcium ion binding and homeostasis, organellar (lysosomal and endoplasmic reticulum) stress, oxidative stress, cell cycle re-entry and activation of cell death processes as the main pathways that are significantly modulated across all excitotoxicity models. Singular profile analyses demonstrate substantial transcriptional regulation of numerous cell cycle proteins. For the first time, we show that iGluR activation forms the basis of cell cycle re-activation, and together with oxidative stress fulfill the "two-hit" hypothesis that accelerates neurodegeneration.


Subject(s)
Cell Cycle , Gene Expression Profiling , Neurons/metabolism , Oxidative Stress , Receptors, Ionotropic Glutamate/metabolism , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Animals , Cells, Cultured , Mice , Real-Time Polymerase Chain Reaction
7.
Neurochem Int ; 62(5): 653-63, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23186747

ABSTRACT

Mitochondrial dysfunction and oxidative stress are currently considered two key mechanisms contributing to pathobiology in neurodegenerative conditions. The current study investigated the temporal molecular events contributing to programmed cell death after treatment with the mitochondrial complex I inhibitor rotenone. Microarray analysis was performed using cultured neocortical neurons treated with 10nM rotenone for 8, 15, and 24h. Genes showing at least ±1.2-fold change in expression at one time point were considered significant. Transcriptomic analysis of the 4178 genes probes revealed major changes to nine biological processes, including those eliciting mitochondrial dysfunction, activation of calcium signaling, increased expression of apoptotic genes, and downplay of chaperones/co-chaperones, ubiquitin-proteasome system and autophagy. These data define targets for intervention where mitochondrial complex I dysfunction plays a substantial role, most notably Parkinson's disease.


Subject(s)
Autophagy/drug effects , Calcium Signaling/drug effects , Cell Death/drug effects , Cerebral Cortex/drug effects , Gene Expression Profiling , Lysosomes/drug effects , Mitochondria/drug effects , Proteasome Endopeptidase Complex/metabolism , Rotenone/pharmacology , Ubiquitin/metabolism , Animals , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Lysosomes/metabolism , Mice , Mitochondria/metabolism , Neurons/drug effects , Neurons/metabolism , Real-Time Polymerase Chain Reaction
8.
Am J Hum Genet ; 86(3): 343-52, 2010 Mar 12.
Article in English | MEDLINE | ID: mdl-20170900

ABSTRACT

Distal hereditary motor neuropathies comprise a clinically and genetically heterogeneous group of disorders. We recently mapped an X-linked form of this condition to chromosome Xq13.1-q21 in two large unrelated families. The region of genetic linkage included ATP7A, which encodes a copper-transporting P-type ATPase mutated in patients with Menkes disease, a severe infantile-onset neurodegenerative condition. We identified two unique ATP7A missense mutations (p.P1386S and p.T994I) in males with distal motor neuropathy in two families. These molecular alterations impact highly conserved amino acids in the carboxyl half of ATP7A and do not directly involve the copper transporter's known critical functional domains. Studies of p.P1386S revealed normal ATP7A mRNA and protein levels, a defect in ATP7A trafficking, and partial rescue of a S. cerevisiae copper transport knockout. Although ATP7A mutations are typically associated with severe Menkes disease or its milder allelic variant, occipital horn syndrome, we demonstrate here that certain missense mutations at this locus can cause a syndrome restricted to progressive distal motor neuropathy without overt signs of systemic copper deficiency. This previously unrecognized genotype-phenotype correlation suggests an important role of the ATP7A copper transporter in motor-neuron maintenance and function.


Subject(s)
Adenosine Triphosphatases/genetics , Cation Transport Proteins/genetics , Genetic Diseases, X-Linked/genetics , Motor Neuron Disease/genetics , Mutation, Missense , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Cation Transport Proteins/chemistry , Cation Transport Proteins/metabolism , Cells, Cultured , Child, Preschool , Copper/metabolism , Copper-Transporting ATPases , DNA Primers/genetics , Female , Genetic Association Studies , Genetic Complementation Test , Genetic Diseases, X-Linked/metabolism , Humans , Immunohistochemistry , Male , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Middle Aged , Models, Molecular , Molecular Sequence Data , Motor Neuron Disease/metabolism , Pedigree , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sequence Homology, Amino Acid , Syndrome , Young Adult
9.
Biochem Biophys Res Commun ; 372(4): 613-7, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18515074

ABSTRACT

Both copper transporting ATPases, ATP7A and ATP7B, are expressed in mammary epithelial cells but their role in copper delivery to milk has not been clarified. We investigated the role of ATP7A in delivery of copper to milk using transgenic mice that over-express human ATP7A. In mammary gland of transgenic mice, human ATP7A protein was 10- to 20-fold higher than in control mice, and was localized to the basolateral membrane of mammary epithelial cells in lactating mice. The copper concentration in the mammary gland of transgenic dams and stomach contents of transgenic pups was significantly reduced compared to non-transgenic mice. The mRNA levels of endogenous Atp7a, Atp7b, and Ctr1 copper transporters in the mammary gland were not altered by the expression of the ATP7A transgene, and the protein levels of Atp7b and ceruloplasmin were similar in transgenic and non-transgenic mice. These data suggest that ATP7A plays a role in removing excess copper from the mammary epithelial cells rather than supplying copper to milk.


Subject(s)
Adenosine Triphosphatases/physiology , Cation Transport Proteins/physiology , Copper/metabolism , Lactation/metabolism , Mammary Glands, Animal/enzymology , Adenosine Triphosphatases/genetics , Animals , Cation Transport Proteins/genetics , Copper/analysis , Copper-Transporting ATPases , Epithelial Cells/chemistry , Epithelial Cells/enzymology , Female , Gene Expression , Homozygote , Humans , Ion Transport , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/cytology , Mice , Mice, Transgenic , Milk/chemistry , Milk/enzymology
10.
J Nutr ; 138(4): 693-7, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18356322

ABSTRACT

The protein affected in Menkes disease, ATP7A, is a copper (Cu)-transporting P-type ATPase that plays an important role in Cu homeostasis, but the full extent of this role has not been defined at a systemic level. Transgenic mice that overexpress the human ATP7A from the chicken beta-actin composite promoter (CAG) were used to further investigate the physiological function of ATP7A. Overexpression of ATP7A in the mice caused disturbances in Cu homeostasis, with depletion of Cu in some tissues, especially the heart. To investigate the effect of overexpression of ATP7A when dietary Cu intake was markedly increased, normal and transgenic mice were exposed to drinking water containing 300 mg/L of Cu as Cu acetate for 3 mo. Cu exposure resulted in partial restoration of heart Cu concentrations in male transgenic mice. Despite the extended period of Cu exposure, Cu concentrations in the liver remained relatively unaffected, with a significant increase in male nontransgenic mice. Liver pathology was unremarkable except for small areas of fibrosis that were detected only in livers of the Cu-exposed transgenic mice. Intracellular localization of ATP7A in various tissues was not affected by Cu exposure. Plasma Cu concentration and ceruloplasmin oxidase activity were reduced in both Cu-exposed transgenic and nontransgenic mice. The expression levels of other candidate Cu homeostatic proteins, endogenous Atp7b, ceruloplasmin, Ctr1, and transgenic ATP7A were not altered significantly by Cu exposure. Overall, mice are remarkably resistant to high Cu loads and the overexpression of ATP7A has only moderate effects on the response to Cu exposure.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper/administration & dosage , Copper/pharmacology , Animals , Biological Transport , Brain/metabolism , Brain Chemistry , Ceruloplasmin/metabolism , Chickens , Copper/adverse effects , Copper/blood , Copper-Transporting ATPases , Down-Regulation , Female , Gene Expression Regulation , Humans , Intestine, Small/chemistry , Intestine, Small/metabolism , Kidney/chemistry , Kidney/metabolism , Liver/chemistry , Liver/metabolism , Male , Mice , Mice, Transgenic , Myocardium/chemistry , Myocardium/metabolism , Spleen/chemistry , Spleen/metabolism , Water
11.
Biochim Biophys Acta ; 1762(4): 485-93, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16488577

ABSTRACT

The brindled mouse is an accurate model of the fatal human X-linked copper deficiency disorder, Menkes disease. Males carrying the mutant allele of the Menkes gene orthologue Atp7a die in the second week of life. To determine whether the genetic defect in the brindled mice could be corrected by expression of the human Menkes gene, male transgenic mice expressing ATP7A from the chicken beta-actin composite promoter (CAG) were mated with female carriers of the brindled mutation (Atp7a(Mo-br)). Mutant males carrying the transgene survived and were fertile but the copper defect was not completely corrected. Unexpectedly males corrected with one transgenic line (T25#5) were mottled and resembled carrier females, this effect appeared to be caused by mosaic expression of the transgene. In contrast, males corrected with another line (T22#2) had agouti coats. Copper concentrations in tissues of the rescued mutants also resembled those of the heterozygous females, with high levels in kidney (84.6+/-4.9 microg/g in corrected males vs. 137.0+/-44.3 microg/g in heterozygotes) and small intestine (15.6+/-2.5 microg/g in corrected males vs. 15.7+/-2.8 microg/g in heterozygotes). The results show that the Menkes defect in mice is corrected by the human Menkes gene and that adequate correction is obtained even when the transgene expression does not match that of the endogenous gene.


Subject(s)
Adenosine Triphosphatases/genetics , Cation Transport Proteins/genetics , Copper/metabolism , Menkes Kinky Hair Syndrome/genetics , Adenosine Triphosphatases/metabolism , Animals , Cation Transport Proteins/metabolism , Chickens , Copper-Transporting ATPases , Disease Models, Animal , Female , Fertility , Humans , Intestine, Small/metabolism , Kidney/metabolism , Male , Menkes Kinky Hair Syndrome/metabolism , Mice , Mice, Transgenic , Mutation , Promoter Regions, Genetic
12.
Am J Physiol Regul Integr Comp Physiol ; 290(5): R1460-7, 2006 May.
Article in English | MEDLINE | ID: mdl-16397091

ABSTRACT

The Menkes protein (ATP7A) is defective in the Cu deficiency disorder Menkes disease and is an important contributor to the maintenance of physiological Cu homeostasis. To investigate more fully the role of ATP7A, transgenic mice expressing the human Menkes gene ATP7A from chicken beta-actin composite promoter (CAG) were produced. The transgenic mice expressed ATP7A in lung, heart, liver, kidney, small intestine, and brain but displayed no overt phenotype resulting from expression of the human protein. Immunohistochemical analysis revealed that ATP7A was found primarily in the cardiac muscle, smooth muscle of the lung, distal tubules of the kidney, intestinal enterocytes, and patches of hepatocytes, as well as in the hippocampus, cerebellum, and choroid plexus of the brain. In 60-day- and 300-day-old mice, Cu concentrations were reduced in most tissues, consistent with ATP7A playing a role in Cu efflux. The reduction in Cu was most pronounced in the hearts of older T22#2 females (24%), T22#2 males (18%), and T25#5 females (23%), as well as in the brains of 60-day-old T22#2 females and males (23% and 30%, respectively).


Subject(s)
Adenosine Triphosphatases/biosynthesis , Cation Transport Proteins/biosynthesis , Copper/physiology , Actins/metabolism , Animals , Blotting, Western , Chickens , Copper/metabolism , Copper-Transporting ATPases , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, Transgenic , Promoter Regions, Genetic/genetics , Spectrophotometry, Atomic , Tissue Distribution
13.
J Nutr ; 135(12): 2762-6, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16317117

ABSTRACT

The final steps in the absorption and excretion of copper at the molecular level are accomplished by 2 closely related proteins that catalyze the ATP-dependent transport of copper across the plasma membrane. These proteins, ATP7A and ATP7B, are encoded by the genes affected in human genetic copper-transport disorders, namely, Menkes and Wilson diseases. We studied the effect of copper perfusion of an isolated segment of the jejunum of ATP7A transgenic mice on the intracellular distribution of ATP7A by immunofluorescence of frozen sections. Our results indicate that ATP7A is retained in the trans-Golgi network under copper-limiting conditions, but relocalized to a vesicular compartment adjacent to the basolateral membrane in intestines perfused with copper. The findings support the hypothesis that the basolateral transport of copper from the enterocyte into the portal blood may involve ATP7A pumping copper into a vesicular compartment followed by exocytosis to release the copper, rather than direct pumping of copper across the basolateral membrane.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper/pharmacology , Intestinal Mucosa/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Adenosine Triphosphatases/drug effects , Animals , Base Sequence , Cation Transport Proteins/drug effects , Copper-Transporting ATPases , DNA Primers , Humans , In Vitro Techniques , Intestinal Mucosa/drug effects , Intestinal Mucosa/physiology , Mice , Mice, Transgenic , Oligonucleotides, Antisense , Plasmids , Recombinant Fusion Proteins/drug effects , Silver/pharmacology
14.
J Nutr ; 133(5 Suppl 1): 1481S-4S, 2003 05.
Article in English | MEDLINE | ID: mdl-12730448

ABSTRACT

Copper is an essential trace element that requires tightly regulated homeostatic mechanisms to ensure adequate supplies without any toxic effects because of the ability of the metal ion to catalyze the formation of free radicals. The Cu-ATPases, ATP7A and ATP7B, play an important role in the physiological regulation of copper. Adequate supplies of copper are particularly important in developing animals, and in humans this is illustrated by mutations of ATP7A that cause the copper deficiency condition Menkes disease, which is fatal in early childhood. In contrast, mutations in ATP7B result in the genetic toxicosis, Wilson disease. We propose that the physiological regulation of copper is accomplished mainly by the intracellular copper-regulated trafficking of the Cu-ATPases. This process allows the overall copper status in the body to be maintained when levels of copper in the diet alter. A study of the defects in mouse models of Menkes and Wilson diseases has demonstrated that both ATPases play an important role in supplying copper to the developing fetus and neonate.


Subject(s)
Copper/deficiency , Copper/metabolism , Mammals/growth & development , Animals , Biological Transport , Hepatolenticular Degeneration/genetics , Homeostasis , Humans , Menkes Kinky Hair Syndrome/genetics , Models, Biological
15.
Biometals ; 16(1): 175-84, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12572677

ABSTRACT

The Menkes protein (MNK) and Wilson protein (WND) are transmembrane, CPX-type Cu-ATPases with six metal binding sites (MBSs) in the N-terminal region containing the motif GMXCXXC. In cells cultured in low copper concentration MNK and WND localize to the transGolgi network but in high copper relocalize either to the plasma membrane (MNK) or a vesicular compartment (WND). In this paper we investigate the role of the MBSs in Cu-transport and trafficking. The copper transport activity of MBS mutants of MNK was determined by their ability to complement a strain of Saccharomyces cerevisiae deficient in CCC2 (delta ccc2), the yeast MNK/WND homologue. Mutants (CXXC to SXXS) of MBS1, MBS6, and MBSs1-3 were able to complement delta ccc2 while mutants of MBS4-6, MBS5-6 and all six MBS inactivated the protein. Each of the inactive mutants also failed to display Cu-induced trafficking suggesting a correlation between trafficking and transport activity. A similar correlation was found with mutants of MNK in which various MBSs were deleted, but two constructs with deletion of MBS5-6 were unable to traffic despite retaining 25% of copper transport activity. Chimeras in which the N-terminal MBSs of MNK were replaced with the corresponding MBSs of WND were used to investigate the region of the molecules that is responsible for the difference in Cu-trafficking of MNK and WND. The chimera which included the complete WND N-terminus localized to a vesicular compartment, similar to WND in elevated copper. Deletions of various MBSs of the WND N-terminus in the chimera indicate that a targeting signal in the region of MBS6 directs either WND/MNK or WND to a vesicular compartment of the cell.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Copper/metabolism , Copper/pharmacology , Recombinant Fusion Proteins , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Amino Acid Sequence , Animals , Binding Sites , CHO Cells , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Copper-Transporting ATPases , Cricetinae , Genetic Complementation Test , Humans , Models, Molecular , Molecular Sequence Data , Peptide Fragments/chemistry , Protein Structure, Secondary , Sequence Deletion , Transfection
16.
J Biol Chem ; 277(48): 46736-42, 2002 Nov 29.
Article in English | MEDLINE | ID: mdl-12228238

ABSTRACT

The Menkes protein (MNK; ATP7A) is a copper-transporting P-type ATPase that is defective in the copper deficiency disorder, Menkes disease. MNK is localized in the trans-Golgi network and transports copper to enzymes synthesized within secretory compartments. However, in cells exposed to excessive copper, MNK traffics to the plasma membrane where it functions in copper efflux. A conserved feature of all P-type ATPases is the formation of an acyl-phosphate intermediate, which occurs as part of the catalytic cycle during cation transport. In this study we investigated the effect of mutations within conserved catalytic regions of MNK on intracellular localization and trafficking from the trans-Golgi network (TGN). Our findings suggest that mutations that block formation of the phosphorylated catalytic intermediate also prevent copper-induced relocalization of MNK from the TGN. Furthermore, mutations in the phosphatase domain, which resulted in hyperphosphorylation of MNK, caused constitutive trafficking from the TGN to the plasma membrane. A similar effect on trafficking was observed with a phosphatase mutation in the closely related copper ATPase, ATP7B, affected in Wilson disease. These findings suggest that the copper-induced trafficking of the Menkes and Wilson disease copper ATPases is associated with the phosphorylated intermediate that is formed during the catalysis of these pumps. Our findings describe a novel mechanism for regulating the subcellular location of a transport protein involving the recognition of intermediate conformations during catalysis.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Copper/physiology , Recombinant Fusion Proteins , Adenosine Triphosphatases/genetics , Animals , Blotting, Western , CHO Cells , Catalysis , Cation Transport Proteins/genetics , Copper-Transporting ATPases , Cricetinae , Fluorescent Antibody Technique , Genetic Complementation Test , Mutagenesis , Phosphorylation , Protein Transport , Subcellular Fractions/enzymology
17.
DNA Cell Biol ; 21(4): 259-70, 2002 Apr.
Article in English | MEDLINE | ID: mdl-12042066

ABSTRACT

Copper is an essential trace element that can be extremely toxic in excess due to the pro-oxidant activity of copper ions. Inherited disorders of copper transport, Menkes disease (copper deficiency), and Wilson disease (copper toxicosis) are caused by mutations of two closely related Cu transporting-ATPases, and demonstrate the essentiality and potential toxicity of copper. Other copper toxicosis conditions in humans and animals have been described, but are not well understood at a molecular level. Copper homeostatic mechanisms are being discovered. One such mechanism is copper-induced trafficking of the Cu-ATPases, which allows cells to provide copper to secreted cupro-proteins but also to efflux excess copper. Oxidative damage induced by copper may be involved in the pathogenesis of neurodegenerative conditions such as Alzheimer's disease, familial amyotrophic lateral sclerosis, and prion diseases.


Subject(s)
Copper/metabolism , Hepatolenticular Degeneration , Menkes Kinky Hair Syndrome , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/metabolism , Animals , Biological Transport , Hepatolenticular Degeneration/genetics , Hepatolenticular Degeneration/metabolism , Homeostasis , Humans , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Oxidative Stress , Prion Diseases/genetics , Prion Diseases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...