Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Metallomics ; 8(9): 816-23, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27399171

ABSTRACT

When we were asked to produce articles for this volume, it seemed appropriate to us to co-author an article on the history and impact of copper research in Melbourne. It is appropriate because over many years, decades in fact, we worked closely together and with Professor David Danks to identify the molecular defect in Menkes disease. This work was always carried out with the intention of understanding the nature of the copper homeostatic mechanisms and a "copper pathway" in the cell, that David had the prescience to predict must exist despite scepticism from granting agencies! He indeed inspired us to pursue research careers in this field. This article outlines some of this history.


Subject(s)
Copper/therapeutic use , Menkes Kinky Hair Syndrome/drug therapy , Animals , Cation Transport Proteins/genetics , Copper/deficiency , Homeostasis , Humans , Menkes Kinky Hair Syndrome/diagnosis , Menkes Kinky Hair Syndrome/genetics , Mutation/genetics
2.
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
3.
Clin Sci (Lond) ; 130(8): 565-74, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26957644

ABSTRACT

Copper is a biometal essential for normal brain development and function, thus copper deficiency or excess results in central nervous system disease. Well-characterized disorders of disrupted copper homoeostasis with neuronal degeneration include Menkes disease and Wilson's disease but a large body of evidence also implicates disrupted copper pathways in other neurodegenerative disorders, including Parkinson's disease, Alzheimer's disease, Amyotrophic lateral sclerosis, Huntington's disease and prion diseases. In this short review we critically evaluate the data regarding changes in systemic and brain copper levels in Parkinson's disease, where alterations in brain copper are associated with regional neuronal cell death and disease pathology. We review copper regulating mechanisms in the human brain and the effects of dysfunction within these systems. We then examine the evidence for a role for copper in pathogenic processes in Parkinson's disease and consider reports of diverse copper-modulating strategies in in vitro and in vivo models of this disorder. Copper-modulating therapies are currently advancing through clinical trials for Alzheimer's and Huntington's disease and may also hold promise as disease modifying agents in Parkinson's disease.


Subject(s)
Antiparkinson Agents/therapeutic use , Brain/drug effects , Copper/metabolism , Parkinson Disease/drug therapy , Animals , Brain/metabolism , Brain/physiopathology , Homeostasis , Humans , Oxidative Stress/drug effects , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , alpha-Synuclein/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.
Physiol Rep ; 2(1): e00195, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24744874

ABSTRACT

Placentae and mammary epithelial cells are unusual in robustly expressing two copper "pumps", ATP7A and B, raising the question of their individual roles in these tissues in pregnancy and lactation. Confocal microscopic evidence locates ATP7A to the fetal side of syncytiotrophoblasts, suggesting a role in pumping Cu towards the fetus; and to the basolateral (blood) side of lactating mammary epithelial cells, suggesting a role in recycling Cu to the blood. We tested these concepts in wild-type C57BL6 mice and their transgenic counterparts that expressed hATP7A at levels 10-20× those of endogenous mAtp7a. In lactation, overexpression of ATP7A reduced the Cu concentrations of the mammary gland and milk ~50%. Rates of transfer of tracer (64)Cu to the suckling pups were similarly reduced over 30-48 h, as was the total Cu in 10-day -old pups. During the early and middle periods of gestation, the transgenic litters had higher Cu concentrations than the wild-type, placental Cu showing the reverse trend; but this difference was lost by the first postnatal day. The transgenic mice expressed ATP7A in some hepatocytes, so we investigated the possibility that metalation of ceruloplasmin (Cp) might be enhanced. Rates of (64)Cu incorporation into Cp, oxidase activity, and ratios of holo to apoceruloplasmin were unchanged. We conclude that in the lactating mammary gland, the role of ATP7A is to return Cu to the blood, while in the placenta it mediates Cu delivery to the fetus and is the rate-limiting step for fetal Cu nutrition during most of gestation in mice.

6.
Prog Neurobiol ; 116: 33-57, 2014 May.
Article in English | MEDLINE | ID: mdl-24440710

ABSTRACT

Copper is an important trace element that is required for essential enzymes. However, due to its redox activity, copper can also lead to the generation of toxic reactive oxygen species. Therefore, cellular uptake, storage as well as export of copper have to be tightly regulated in order to guarantee sufficient copper supply for the synthesis of copper-containing enzymes but also to prevent copper-induced oxidative stress. In brain, copper is of importance for normal development. In addition, both copper deficiency as well as excess of copper can seriously affect brain functions. Therefore, this organ possesses ample mechanisms to regulate its copper metabolism. In brain, astrocytes are considered as important regulators of copper homeostasis. Impairments of homeostatic mechanisms in brain copper metabolism have been associated with neurodegeneration in human disorders such as Menkes disease, Wilson's disease and Alzheimer's disease. This review article will summarize the biological functions of copper in the brain and will describe the current knowledge on the mechanisms involved in copper transport, storage and export of brain cells. The role of copper in diseases that have been connected with disturbances in brain copper homeostasis will also be discussed.


Subject(s)
Brain/physiology , Copper/metabolism , Animals , Astrocytes/physiology , Brain/physiopathology , Homeostasis , Humans , Neurodegenerative Diseases/physiopathology , Neurons/physiology
7.
Neurobiol Aging ; 35(4): 858-66, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24176624

ABSTRACT

Synchrotron-based x-ray fluorescence microscopy, immunofluorescence, and Western blotting were used to investigate changes in copper (Cu) and Cu-associated pathways in the vulnerable substantia nigra (SN) and locus coeruleus (LC) and in nondegenerating brain regions in cases of Parkinson's disease (PD) and appropriate healthy and disease controls. In PD and incidental Lewy body disease, levels of Cu and Cu transporter protein 1, were significantly reduced in surviving neurons in the SN and LC. Specific activity of the cuproprotein superoxide dismutase 1 was unchanged in the SN in PD but was enhanced in the parkinsonian anterior cingulate cortex, a region with α-synuclein pathology, normal Cu, and limited cell loss. These data suggest that regions affected by α-synuclein pathology may display enhanced vulnerability and cell loss if Cu-dependent protective mechanisms are compromised. Additional investigation of copper pathology in PD may identify novel targets for the development of protective therapies for this disorder.


Subject(s)
Cation Transport Proteins/metabolism , Copper/metabolism , Locus Coeruleus/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Substantia Nigra/metabolism , Copper Transporter 1 , Gyrus Cinguli/enzymology , Humans , Locus Coeruleus/cytology , Molecular Targeted Therapy , Neurons/metabolism , Parkinson Disease/drug therapy , Substantia Nigra/cytology , Superoxide Dismutase/metabolism , Superoxide Dismutase-1 , alpha-Synuclein
8.
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
9.
J Cell Sci ; 126(Pt 12): 2551-60, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23729740

ABSTRACT

Nonsense-mediated mRNA decay (NMD) is an evolutionarily conserved mRNA surveillance system that degrades mRNA transcripts that harbour a premature translation-termination codon (PTC), thus reducing the synthesis of truncated proteins that would otherwise have deleterious effects. Although extensive research has identified a conserved repertoire of NMD factors, these studies have been performed with a restricted set of genes and gene constructs with relatively few exons. As a consequence, NMD mechanisms are poorly understood for genes with large 3' terminal exons, and the applicability of the current models to large multi-exon genes is not clear. In this Commentary, we present an overview of the current understanding of NMD and discuss how analysis of nonsense mutations in the collagen gene family has provided new mechanistic insights into this process. Although NMD of the collagen genes with numerous small exons is consistent with the widely accepted exon-junction complex (EJC)-dependent model, the degradation of Col10a1 transcripts with nonsense mutations cannot be explained by any of the current NMD models. Col10a1 NMD might represent a fail-safe mechanism for genes that have large 3' terminal exons. Defining the mechanistic complexity of NMD is important to allow us to understand the pathophysiology of the numerous genetic disorders caused by PTC mutations.


Subject(s)
Codon, Nonsense , Collagen/genetics , Nonsense Mediated mRNA Decay , RNA Stability/genetics , RNA/genetics , RNA/metabolism , Animals , Collagen/metabolism , Exons , Humans , Protein Biosynthesis
10.
Article in English | MEDLINE | ID: mdl-23503037

ABSTRACT

This short review will summarize the current knowledge on the uptake, storage, and export of copper ions by astrocytes and will address the potential roles of astrocytes in copper homeostasis in the normal and diseased brain. Astrocytes in culture efficiently accumulate copper by processes that include both the copper transporter Ctr1 and Ctr1-independent mechanisms. Exposure of astrocytes to copper induces an increase in cellular glutathione (GSH) content as well as synthesis of metallothioneins, suggesting that excess of copper is stored as complex with GSH and in metallothioneins. Furthermore, exposure of astrocytes to copper accelerates the release of GSH and glycolytically generated lactate. Astrocytes are able to export copper and express the Menkes protein ATP7A. This protein undergoes reversible, copper-dependent trafficking between the trans-Golgi network and vesicular structures. The ability of astrocytes to efficiently take up, store and export copper suggests that astrocytes play a key role in the supply of neurons with copper and that astrocytes should be considered as target for therapeutic interventions that aim to correct disturbances in brain copper homeostasis.

11.
Metallomics ; 5(1): 43-51, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23076575

ABSTRACT

Disturbances in brain copper result in rare and severe neurological disorders and may play a role in the pathogenesis and progression of multiple neurodegenerative diseases. Our current understanding of mammalian brain copper transport is based on model systems outside the central nervous system and no data are available regarding copper transport systems in the human brain. To address this deficit, we quantified regional copper concentrations and examined the distribution and cellular localization of the copper transport proteins Copper transporter 1, Atox1, ATP7A, and ATP7B in multiple regions of the human brain using inductively coupled plasma-mass spectrometry, Western blot and immunohistochemistry. We identified significant relationships between copper transporter levels and brain copper concentrations, supporting a role for these proteins in copper transport in the human brain. Interestingly, the substantia nigra contained twice as much copper than that in other brain regions, suggesting an important role for copper in this brain region. Furthermore, ATP7A levels were significantly greater in the cerebellum, compared with other brain regions, supporting an important role for ATP7A in cerebellar neuronal health. This study provides novel data regarding copper regulation in the human brain, critical to understand the mechanisms by which brain copper levels can be altered, leading to neurological disease.


Subject(s)
Adenosine Triphosphatases/analysis , Brain Chemistry , Cation Transport Proteins/analysis , Copper/analysis , Metallochaperones/analysis , Blotting, Western , Copper Transport Proteins , Copper Transporter 1 , Copper-Transporting ATPases , Humans , Immunohistochemistry , Mass Spectrometry , Molecular Chaperones
12.
Biochem Pharmacol ; 85(2): 207-15, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23123662

ABSTRACT

Dorsal root ganglion (DRG) neurons are affected by platinum-induced neurotoxicity and neurodegenerative processes associated with disturbed copper homeostasis and transport. This study aimed to understand the role of copper transporter 1 (Ctr1) in the uptake and toxicity of copper and platinum drugs in cultured rat DRG neurons, and the functional activities of rat Ctr1 (rCtr1) as a membrane transporter of copper and platinum drugs. Heterologous expression of rCtr1 in HEK293 cells (HEK/rCtr1 cells) increased the uptake and cytotoxicity of copper, oxaliplatin, cisplatin and carboplatin, in comparison to isogenic vector-transfected control cells. Cultured rat DRG neurons endogenously expressed rCtr1 protein on their neuronal cell body plasma membranes and cytoplasm, and displayed substantial capacity for taking up copper, but were resistant to copper toxicity. The uptake of copper by both cultured rat DRG neurons and HEK/rCtr1 cells was saturable and inhibited by cold temperature, silver and zinc, consistent with it being mediated by rCtr1. Cultured rat DRG neurons accumulated platinum during their exposure to oxaliplatin and were sensitive to oxaliplatin cytotoxicity. The accumulation of platinum by both cultured rat DRG neurons and HEK/rCtr1 cells, during oxaliplatin exposure, was saturable and temperature dependent, but was inhibited by copper only in HEK/rCtr1 cells. In conclusion, rCtr1 can transport copper and platinum drugs, and sensitizes cells to their cytotoxicities. DRG neurons display substantial capacity for accumulating copper via a transport process mediated by rCtr1, but appear able to resist copper toxicity and use alternative mechanisms to take up oxaliplatin.


Subject(s)
Antineoplastic Agents/metabolism , Cation Transport Proteins/metabolism , Copper/adverse effects , Ganglia, Spinal/drug effects , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Organoplatinum Compounds/metabolism , Animals , Antineoplastic Agents/adverse effects , Antineoplastic Agents/pharmacology , Biological Transport/drug effects , Carboplatin/adverse effects , Carboplatin/metabolism , Carboplatin/pharmacology , Cation Transport Proteins/antagonists & inhibitors , Cation Transport Proteins/genetics , Cells, Cultured , Cisplatin/adverse effects , Cisplatin/metabolism , Cisplatin/pharmacology , Copper/metabolism , Copper/pharmacology , Copper Sulfate/adverse effects , Copper Sulfate/metabolism , Copper Sulfate/pharmacology , Copper Transporter 1 , Ganglia, Spinal/cytology , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Membrane Transport Modulators/pharmacology , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , Neurons/cytology , Neurons/metabolism , Organoplatinum Compounds/adverse effects , Organoplatinum Compounds/pharmacology , Oxaliplatin , Rats , Rats, Wistar , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism
13.
Met Ions Life Sci ; 13: 359-87, 2013.
Article in English | MEDLINE | ID: mdl-24470097

ABSTRACT

Copper is an essential trace metal that is required for the catalysis of several important cellular enzymes. However, since an excess of copper can also harm cells due to its potential to catalyze the generation of toxic reactive oxygen species, transport of copper and the cellular copper content are tightly regulated. This chapter summarizes the current knowledge on the importance of copper for cellular processes and on the mechanisms involved in cellular copper uptake, storage and export. In addition, we will give an overview on disturbances of copper homeostasis that are characterized by copper overload or copper deficiency or have been connected with neurodegenerative disorders.


Subject(s)
Copper , Neurodegenerative Diseases , Reactive Oxygen Species/metabolism , Animals , Copper/adverse effects , Copper/deficiency , Copper/metabolism , Ion Transport , Male , Neurodegenerative Diseases/chemically induced , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/pathology
14.
Hum Mol Genet ; 21(8): 1794-807, 2012 Apr 15.
Article in English | MEDLINE | ID: mdl-22210628

ABSTRACT

ATP7A is a P-type ATPase that regulates cellular copper homeostasis by activity at the trans-Golgi network (TGN) and plasma membrane (PM), with the location normally governed by intracellular copper concentration. Defects in ATP7A lead to Menkes disease or its milder variant, occipital horn syndrome or to a newly discovered condition, ATP7A-related distal motor neuropathy (DMN), for which the precise pathophysiology has been obscure. We investigated two ATP7A motor neuropathy mutations (T994I, P1386S) previously associated with abnormal intracellular trafficking. In the patients' fibroblasts, total internal reflection fluorescence microscopy indicated a shift in steady-state equilibrium of ATP7A(T994I) and ATP7A(P1386S), with exaggerated PM localization. Transfection of Hek293T cells and NSC-34 motor neurons with the mutant alleles tagged with the Venus fluorescent protein also revealed excess PM localization. Endocytic retrieval of the mutant alleles from the PM to the TGN was impaired. Immunoprecipitation assays revealed an abnormal interaction between ATP7A(T994I) and p97/VCP, an ubiquitin-selective chaperone which is mutated in two autosomal dominant forms of motor neuron disease: amyotrophic lateral sclerosis and inclusion body myopathy with early-onset Paget disease and fronto-temporal dementia. Small-interfering RNA (SiRNA) knockdown of p97/VCP corrected ATP7A(T994I) mislocalization. Flow cytometry documented that non-permeabilized ATP7A(P1386S) fibroblasts bound a carboxyl-terminal ATP7A antibody, consistent with relocation of the ATP7A di-leucine endocytic retrieval signal to the extracellular surface and partially destabilized insertion of the eighth transmembrane helix. Our findings illuminate the mechanisms underlying ATP7A-related DMN and establish a link between p97/VCP and genetically distinct forms of motor neuron degeneration.


Subject(s)
Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Membrane/metabolism , Motor Neuron Disease/genetics , Motor Neuron Disease/metabolism , Adenosine Triphosphatases/chemistry , Alleles , Cation Transport Proteins/chemistry , Copper-Transporting ATPases , Endocytosis , Fibroblasts , HEK293 Cells , Humans , Motor Neurons/metabolism , Mutation , Protein Binding , Protein Structure, Secondary , Protein Transport , Valosin Containing Protein , trans-Golgi Network/metabolism
15.
Biometals ; 25(2): 373-82, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22080201

ABSTRACT

Copper transport and accumulation were studied in virgin and lactating C57BL/6 mice, with and without expression of ceruloplasmin (Cp), to assess the importance of Cp to these processes. One hour after i.p. injection of tracer (64)Cu, liver and kidney accounted for 80% of the radioactivity, and mammary gland 1%, while in lactating Cp+/+ mice 2-4 days post partum, uptake by mammary gland was 9-fold higher and that of liver and other organs was decreased, with (64)Cu rapidly appearing in milk. Parallel studies in Cp-/- mice (siblings from same colony) gave virtually identical results. However, their milk contained less (64)Cu, and actual copper contents determined by furnace atomic absorption were less than half those for milk from normal dams. Liver copper concentrations of pups born to Cp-/- dams also were half those of pups from wild type dams. Copper in pup brains was unaffected; but iron concentrations were reduced. We conclude that absence of Cp, while not affecting entry of exchangeable copper from the blood into the mammary gland, does have a significant effect on the availability of this metal to the newborn through the milk and in the form of stores accumulating in gestation.


Subject(s)
Ceruloplasmin/physiology , Copper/metabolism , Fetus/metabolism , Liver/metabolism , Mammary Glands, Animal/metabolism , Animals , Animals, Newborn , Biological Transport , Brain/metabolism , Lactation/metabolism , Mice
16.
J Biol Chem ; 287(4): 2485-99, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-22130675

ABSTRACT

ATP7A and ATP7B are copper-transporting P(1B)-type ATPases (Cu-ATPases) that are critical for regulating intracellular copper homeostasis. Mutations in the genes encoding ATP7A and ATP7B lead to copper deficiency and copper toxicity disorders, Menkes and Wilson diseases, respectively. Clusterin and COMMD1 were previously identified as interacting partners of these Cu-ATPases. In this study, we confirmed that clusterin and COMMD1 interact to down-regulate both ATP7A and ATP7B. Overexpression and knockdown of clusterin/COMMD1 decreased and increased, respectively, endogenous levels of ATP7A and ATP7B, consistent with a role in facilitating Cu-ATPase degradation. We demonstrate that whereas the clusterin/ATP7B interaction was enhanced by oxidative stress or mutation of ATP7B, the COMMD1/ATP7B interaction did not change under oxidative stress conditions, and only increased with ATP7B mutations that led to its misfolding. Clusterin and COMMD1 facilitated the degradation of ATP7B containing the same Wilson disease-causing C-terminal mutations via different degradation pathways, clusterin via the lysosomal pathway and COMMD1 via the proteasomal pathway. Furthermore, endogenous ATP7B existed in a complex with clusterin and COMMD1, but these interactions were neither competitive nor cooperative and occurred independently of each other. Together these data indicate that clusterin and COMMD1 represent alternative and independent systems regulating Cu-ATPase quality control, and consequently contributing to the maintenance of copper homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Clusterin/metabolism , Proteolysis , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphatases/genetics , Animals , CHO Cells , Cation Transport Proteins/genetics , Clusterin/genetics , Copper-Transporting ATPases , Cricetinae , Cricetulus , HEK293 Cells , Hepatolenticular Degeneration/genetics , Hepatolenticular Degeneration/metabolism , Humans , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Mice , Mutation , Rats
17.
J Biol Chem ; 286(12): 10073-83, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21242307

ABSTRACT

The copper-transporting P(1B)-type ATPases (Cu-ATPases) ATP7A and ATP7B are key regulators of physiological copper levels. They function to maintain intracellular copper homeostasis by delivering copper to secretory compartments and by trafficking toward the cell periphery to export excess copper. Mutations in the genes encoding ATP7A and ATP7B lead to copper deficiency and toxicity disorders, Menkes and Wilson diseases, respectively. This report describes the interaction between the Cu-ATPases and clusterin and demonstrates a chaperone-like role for clusterin in facilitating their degradation. Clusterin interacted with both ATP7A and ATP7B in mammalian cells. This interaction increased under conditions of oxidative stress and with mutations in ATP7B that led to its misfolding and mislocalization. A Wilson disease patient mutation (G85V) led to enhanced ATP7B turnover, which was further exacerbated when cells overexpressed clusterin. We demonstrated that clusterin-facilitated degradation of mutant ATP7B is likely to involve the lysosomal pathway. The knockdown and overexpression of clusterin increased and decreased, respectively, the Cu-ATPase-mediated copper export capacity of cells. These results highlight a new role for intracellular clusterin in mediating Cu-ATPase quality control and hence in the normal maintenance of copper homeostasis, and in promoting cell survival in the context of disease. Based on our findings, it is possible that variations in clusterin expression and function could contribute to the variable clinical expression of Menkes and Wilson diseases.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Clusterin/metabolism , Hepatolenticular Degeneration/metabolism , Menkes Kinky Hair Syndrome/metabolism , Protein Folding , Adenosine Triphosphatases/genetics , Animals , CHO Cells , Cation Transport Proteins/genetics , Clusterin/genetics , Copper/metabolism , Copper-Transporting ATPases , Cricetinae , Cricetulus , HEK293 Cells , Hep G2 Cells , Hepatolenticular Degeneration/genetics , Humans , Lysosomes/genetics , Lysosomes/metabolism , Menkes Kinky Hair Syndrome/genetics
18.
Mol Pain ; 6: 53, 2010 Sep 13.
Article in English | MEDLINE | ID: mdl-20836889

ABSTRACT

BACKGROUND: ATP7A, ATP7B and CTR1 are metal transporting proteins that control the cellular disposition of copper and platinum drugs, but their expression in dorsal root ganglion (DRG) tissue and their role in platinum-induced neurotoxicity are unknown. To investigate the DRG expression of ATP7A, ATP7B and CTR1, lumbar DRG and reference tissues were collected for real time quantitative PCR, RT-PCR, immunohistochemistry and Western blot analysis from healthy control adult rats or from animals treated with intraperitoneal oxaliplatin (1.85 mg/kg) or drug vehicle twice weekly for 8 weeks. RESULTS: In DRG tissue from healthy control animals, ATP7A mRNA was clearly detectable at levels similar to those found in the brain and spinal cord, and intense ATP7A immunoreactivity was localised to the cytoplasm of cell bodies of smaller DRG neurons without staining of satellite cells, nerve fibres or co-localisation with phosphorylated heavy neurofilament subunit (pNF-H). High levels of CTR1 mRNA were detected in all tissues from healthy control animals, and strong CTR1 immunoreactivity was associated with plasma membranes and vesicular cytoplasmic structures of the cell bodies of larger-sized DRG neurons without co-localization with ATP7A. DRG neurons with strong expression of ATP7A or CTR1 had distinct cell body size profiles with minimal overlap between them. Oxaliplatin treatment did not alter the size profile of strongly ATP7A-immunoreactive neurons but significantly reduced the size profile of strongly CTR1-immunoreactive neurons. ATP7B mRNA was barely detectable, and no specific immunoreactivity for ATP7B was found, in DRG tissue from healthy control animals. CONCLUSIONS: In conclusion, adult rat DRG tissue exhibits a specific pattern of expression of copper transporters with distinct subsets of peripheral sensory neurons intensely expressing either ATP7A or CTR1, but not both or ATP7B. The neuron subtype-specific and largely non-overlapping distribution of ATP7A and CTR1 within rat DRG tissue may be required to support the potentially differing cuproenzyme requirements of distinct subsets of sensory neurons, and could influence the transport and neurotoxicity of oxaliplatin.


Subject(s)
Adenosine Triphosphatases/genetics , Aging/metabolism , Cation Transport Proteins/genetics , Ganglia, Spinal/metabolism , Gene Expression Regulation , Adenosine Triphosphatases/metabolism , Aging/drug effects , Animals , Cation Transport Proteins/metabolism , Cell Size/drug effects , Copper Transporter 1 , Copper-Transporting ATPases , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Gene Expression Regulation/drug effects , Lumbar Vertebrae/cytology , Lumbar Vertebrae/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Organoplatinum Compounds/pharmacology , Oxaliplatin , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction
19.
J Biol Chem ; 285(35): 27111-27121, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20566629

ABSTRACT

The copper-transporting P-type ATPases (Cu-ATPases), ATP7A and ATP7B, are essential for the regulation of intracellular copper homeostasis. In this report we describe new roles for glutathione (GSH) and glutaredoxin1 (GRX1) in Cu homeostasis through their regulation of Cu-ATPase activity. GRX1 is a thiol oxidoreductase that catalyzes the reversible reduction of GSH-mixed disulfides to their respective sulfhydryls (deglutathionylation). Here, we demonstrated that glutathionylation of the Cu-ATPases and their interaction with GRX1 were affected by alterations in Cu levels. The data support our hypothesis that the Cu-ATPases serve as substrates for Cu-dependent GRX1-mediated deglutathionylation. This in turn liberates the Cu-ATPase cysteinyl thiol groups for Cu binding and transport. GSH depletion experiments led to reversible inhibition of the Cu-ATPases that correlated with effects on intracellular Cu levels and GRX1 activity. Finally, knockdown of GRX1 expression resulted in an increase in intracellular Cu accumulation. Together, these data directly implicate GSH and GRX1 with important new roles in redox regulation of the Cu-ATPases, through modulation of Cu binding by the Cu-ATPase cysteine motifs.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Copper/metabolism , Glutaredoxins/metabolism , Glutathione/metabolism , Protein Processing, Post-Translational/physiology , Adenosine Triphosphatases/genetics , Animals , Biological Transport/physiology , CHO Cells , Cation Transport Proteins/genetics , Copper-Transporting ATPases , Cricetinae , Cricetulus , Gene Knockdown Techniques , Glutaredoxins/genetics , Glutathione/genetics , Hep G2 Cells , Humans , Protein Binding/physiology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...