Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 289
Filter
1.
PLoS Genet ; 19(1): e1010558, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36626371

ABSTRACT

Copper (Cu) has a multifaceted role in brain development, function, and metabolism. Two homologous Cu transporters, Atp7a (Menkes disease protein) and Atp7b (Wilson disease protein), maintain Cu homeostasis in the tissue. Atp7a mediates Cu entry into the brain and activates Cu-dependent enzymes, whereas the role of Atp7b is less clear. We show that during postnatal development Atp7b is necessary for normal morphology and function of choroid plexus (ChPl). Inactivation of Atp7b causes reorganization of ChPl' cytoskeleton and cell-cell contacts, loss of Slc31a1 from the apical membrane, and a decrease in the length and number of microvilli and cilia. In ChPl lacking Atp7b, Atp7a is upregulated but remains intracellular, which limits Cu transport into the brain and results in significant Cu deficit, which is reversed only in older animals. Cu deficiency is associated with down-regulation of Atp7a in locus coeruleus and catecholamine imbalance, despite normal expression of dopamine-ß-hydroxylase. In addition, there are notable changes in the brain lipidome, which can be attributed to inhibition of diacylglyceride-to-phosphatidylethanolamine conversion. These results identify the new role for Atp7b in developing brain and identify metabolic changes that could be exacerbated by Cu chelation therapy.


Subject(s)
Copper , Menkes Kinky Hair Syndrome , Mice , Animals , Copper-Transporting ATPases , Copper/metabolism , Choroid Plexus/metabolism , Menkes Kinky Hair Syndrome/metabolism , Brain/metabolism
2.
Proc Natl Acad Sci U S A ; 116(25): 12167-12172, 2019 06 18.
Article in English | MEDLINE | ID: mdl-31160463

ABSTRACT

Copper is controlled by a sophisticated network of transport and storage proteins within mammalian cells, yet its uptake and efflux occur with rapid kinetics. Present as Cu(I) within the reducing intracellular environment, the nature of this labile copper pool remains elusive. While glutathione is involved in copper homeostasis and has been assumed to buffer intracellular copper, we demonstrate with a ratiometric fluorescent indicator, crisp-17, that cytosolic Cu(I) levels are buffered to the vicinity of 1 aM, where negligible complexation by glutathione is expected. Enabled by our phosphine sulfide-stabilized phosphine (PSP) ligand design strategy, crisp-17 offers a Cu(I) dissociation constant of 8 aM, thus exceeding the binding affinities of previous synthetic Cu(I) probes by four to six orders of magnitude. Two-photon excitation microscopy with crisp-17 revealed rapid, reversible increases in intracellular Cu(I) availability upon addition of the ionophoric complex CuGTSM or the thiol-selective oxidant 2,2'-dithiodipyridine (DTDP). While the latter effect was dramatically enhanced in 3T3 cells grown in the presence of supplemental copper and in cultured Menkes mutant fibroblasts exhibiting impaired copper efflux, basal Cu(I) availability in these cells showed little difference from controls, despite large increases in total copper content. Intracellular copper is thus tightly buffered by endogenous thiol ligands with significantly higher affinity than glutathione. The dual utility of crisp-17 to detect normal intracellular buffered Cu(I) levels as well as to probe the depth of the labile copper pool in conjunction with DTDP provides a promising strategy to characterize perturbations of cellular copper homeostasis.


Subject(s)
Copper/metabolism , Menkes Kinky Hair Syndrome/metabolism , Buffers , Fibroblasts/metabolism , Fluorescent Dyes , Glutathione/metabolism , Ligands , Microscopy, Fluorescence, Multiphoton/methods , Mutation , Phosphines/metabolism
3.
Int J Mol Sci ; 23(19)2022 Sep 28.
Article in English | MEDLINE | ID: mdl-36232742

ABSTRACT

Kidneys play an especial role in copper redistribution in the organism. The epithelial cells of proximal tubules perform the functions of both copper uptake from the primary urine and release to the blood. These cells are equipped on their apical and basal membrane with copper transporters CTR1 and ATP7A. Mosaic mutant mice displaying a functional dysfunction of ATP7A are an established model of Menkes disease. These mice exhibit systemic copper deficiency despite renal copper overload, enhanced by copper therapy, which is indispensable for their life span extension. The aim of this study was to analyze the expression of Slc31a1 and Slc31a2 genes (encoding CTR1/CTR2 proteins) and the cellular localization of the CTR1 protein in suckling, young and adult mosaic mutants. Our results indicate that in the kidney of both intact and copper-injected 14-day-old mutants showing high renal copper content, CTR1 mRNA level is not up-regulated compared to wild-type mice given a copper injection. The expression of the Slc31a1 gene in 45-day-old mice is even reduced compared with intact wild-type animals. In suckling and young copper-injected mutants, the CTR1 protein is relocalized from the apical membrane to the cytoplasm of epithelial cells of proximal tubules, the process which prevents copper transport from the primary urine and, thus, protects cells against copper toxicity.


Subject(s)
Copper Transporter 1 , Copper , Epithelial Cells , Kidney Tubules, Proximal , Menkes Kinky Hair Syndrome , Animals , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Membrane/genetics , Cell Membrane/metabolism , Copper/metabolism , Copper/toxicity , Copper Transporter 1/genetics , Copper Transporter 1/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Cytoplasm/genetics , Cytoplasm/metabolism , Disease Models, Animal , Epithelial Cells/metabolism , Gene Expression , Kidney Tubules, Proximal/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Menkes Kinky Hair Syndrome/etiology , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Mice , Protein Transport/genetics , Protein Transport/physiology , RNA, Messenger/metabolism , SLC31 Proteins/genetics , SLC31 Proteins/metabolism
4.
Pflugers Arch ; 472(10): 1415-1429, 2020 10.
Article in English | MEDLINE | ID: mdl-32506322

ABSTRACT

Copper is an essential element in cells; it can act as either a recipient or a donor of electrons, participating in various reactions. However, an excess of copper ions in cells is detrimental as these copper ions can generate free radicals and increase oxidative stress. In multicellular organisms, copper metabolism involves uptake, distribution, sequestration, and excretion, at both the cellular and systemic levels. Mammalian enterocytes take in bioavailable copper ions from the diet in a Ctr1-dependent manner. After incorporation, cuprous ions are delivered to ATP7A, which pumps Cu+ from enterocytes into the blood. Copper ions arrive at the liver through the portal vein and are incorporated into hepatocytes by Ctr1. Then, Cu+ can be secreted into the bile or the blood via the Atox1/ATP7B/ceruloplasmin route. In the bloodstream, this micronutrient can reach peripheral tissues and is again incorporated by Ctr1. In peripheral tissue cells, cuprous ions are either sequestrated by molecules such as metallothioneins or targeted to utilization pathways by chaperons such as Atox1, Cox17, and CCS. Copper metabolism must be tightly controlled in order to achieve homeostasis and avoid disorders. A hereditary or acquired copper unbalance, including deficiency, overload, or misdistribution, may cause or aggravate certain diseases such as Menkes disease, Wilson disease, neurodegenerative diseases, anemia, metabolic syndrome, cardiovascular diseases, and cancer. A full understanding of copper metabolism and its roles in diseases underlies the identification of novel effective therapies for such diseases.


Subject(s)
Copper/metabolism , Hepatolenticular Degeneration/metabolism , Menkes Kinky Hair Syndrome/metabolism , Animals , Copper/deficiency , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Hepatolenticular Degeneration/genetics , Humans , Menkes Kinky Hair Syndrome/genetics , Molecular Chaperones/genetics , Molecular Chaperones/metabolism
5.
J Inherit Metab Dis ; 41(6): 1285-1291, 2018 11.
Article in English | MEDLINE | ID: mdl-30132231

ABSTRACT

INTRODUCTION: Menkes disease (MD) is an X-linked recessive disorder caused by dysfunction of a copper-transporting protein, leading to severe neurodegeneration in early childhood. We investigated whether a lipophilic copper chelator, disulfiram, could enhance copper absorption from the intestine and transport copper across the blood-brain barrier in MD model mice. METHODS: Wild type and MD model mice were pretreated with disulfiram for 30 min before oral administration of 64CuCl2. Each organ was sequentially analyzed for radioactivity with γ counting. Copper uptake into the brain parenchyma was assessed by ex vivo autoradiography. RESULTS: In wild type mice, orally administered copper was initially detected in the intestine within 2 h, reaching a maximum level in the liver (19.6 ± 3.8 percentage injected dose per gram [%ID/g]) at 6 h. In MD model mice, the copper reached the maximum level in the liver (5.3 ± 1.5 %ID/g) at 4 h, which was lower than that of wild type mice (19.0 ± 7.4 %ID/g) (P < 0.05). Pretreatment of disulfiram in MD model mice increased the copper level in the brain (0.59 ± 0.28 %ID/g) at 24 h compared with MD model mice without disulfiram (0.07 ± 0.05 %ID/g) (P < 0.05). Ex vivo autoradiography revealed that high levels of copper uptake was observed in the cerebral cortex upon disulfiram pretreatment. CONCLUSION: Our data demonstrated that disulfiram enhanced the delivery of orally administered copper into the central nervous system in MD model mice. The administration of disulfiram will enable patients to avoid unpleasant subcutaneous copper injection in the future.


Subject(s)
Copper/pharmacology , Disulfiram/therapeutic use , Drug Carriers , Menkes Kinky Hair Syndrome/drug therapy , Animals , Autoradiography , Blood-Brain Barrier/metabolism , Copper/metabolism , Disease Models, Animal , Male , Menkes Kinky Hair Syndrome/blood , Menkes Kinky Hair Syndrome/metabolism , Mice , Mice, Inbred C3H
6.
IUBMB Life ; 69(4): 263-270, 2017 04.
Article in English | MEDLINE | ID: mdl-28271632

ABSTRACT

The essential transition metal copper is important in lipid metabolism, redox balance, iron mobilization, and many other critical processes in eukaryotic organisms. Genetic diseases where copper homeostasis is disrupted, including Menkes disease and Wilson disease, indicate the importance of copper balance to human health. The severe consequences of insufficient copper supply are illustrated by Menkes disease, caused by mutation in the X-linked ATP7A gene encoding a protein that transports copper from intestinal epithelia into the bloodstream and across the blood-brain barrier. Inadequate copper supply to the body due to poor diet quality or malabsorption can disrupt several molecular level pathways and processes. Though much of the copper distribution machinery has been described and consequences of disrupted copper handling have been characterized in human disease as well as animal models, physiological consequences of sub-optimal copper due to poor nutrition or malabsorption have not been extensively studied. Recent work indicates that insufficient copper may be important in a number of common diseases including obesity, ischemic heart disease, and metabolic syndrome. Specifically, marginal copper deficiency (CuD) has been reported as a potential etiologic factor in diseases characterized by disrupted lipid metabolism such as non-alcoholic fatty-liver disease (NAFLD). In this review, we discuss the available data suggesting that a significant portion of the North American population may consume insufficient copper, the potential mechanisms by which CuD may promote lipid biosynthesis, and the interaction between CuD and dietary fructose in the etiology of NAFLD. © 2016 IUBMB Life, 69(4):263-270, 2017.


Subject(s)
Copper/metabolism , Lipid Metabolism/genetics , Lipids/biosynthesis , Non-alcoholic Fatty Liver Disease/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Blood-Brain Barrier/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper-Transporting ATPases , Diet , Humans , Iron/metabolism , Lipids/genetics , Liver/metabolism , Liver/pathology , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Non-alcoholic Fatty Liver Disease/diet therapy , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/pathology
7.
Folia Biol (Praha) ; 63(5-6): 165-173, 2017.
Article in English | MEDLINE | ID: mdl-29687769

ABSTRACT

Menkes disease is a severe X-linked recessive disorder caused by a defect in the ATP7A gene, which encodes a membrane copper-transporting ATPase. Deficient activity of the ATP7A protein results in decreased intestinal absorption of copper, low copper level in serum and defective distribution of copper in tissues. The clinical symptoms are caused by decreased activities of copper-dependent enzymes and include neurodegeneration, connective tissue disorders, arterial changes and hair abnormalities. Without therapy, the disease is fatal in early infancy. Rapid diagnosis of Menkes disease and early start of copper therapy is critical for the effectiveness of treatment. We report a molecular biology-based strategy that allows early diagnosis of copper transport defects and implementation of individual therapies before the full development of pathological symptoms. Low serum copper and decreased activity of copperdependent mitochondrial cytochrome c oxidase in isolated platelets found in three patients indicated a possibility of functional defects in copper-transporting proteins, especially in the ATPA7 protein, a copper- transporting P-type ATPase. Rapid mutational screening of the ATP7A gene using high-resolution melting analysis of DNA indicated presence of mutations in the patients. Molecular investigation for mutations in the ATP7A gene revealed three nonsense mutations: c.2170C>T (p.Gln724Ter); c.3745G>T (p.Glu1249Ter); and c.3862C>T (p.Gln1288Ter). The mutation c.3745G>T (p.Glu1249Ter) has not been identified previously. Molecular analysis of the ATOX1 gene as a possible modulating factor of Menkes disease did not reveal presence of pathogenic mutations. Molecular diagnostics allowed early onset of individual therapies, adequate genetic counselling and prenatal diagnosis in the affected families.


Subject(s)
Carrier Proteins/metabolism , Copper-Transporting ATPases/metabolism , Copper/blood , Menkes Kinky Hair Syndrome/metabolism , Mutation/genetics , Carrier Proteins/genetics , Child , Copper/metabolism , Copper Transport Proteins , Copper-Transporting ATPases/genetics , Humans , Infant , Male , Menkes Kinky Hair Syndrome/genetics , Metallochaperones/genetics , Metallochaperones/metabolism , Models, Biological , Molecular Chaperones
8.
Arch Dis Child Educ Pract Ed ; 102(6): 319-327, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28751535

ABSTRACT

In paediatrics, one of our main aims in the diagnostic process is to identify any treatable conditions. The copper metabolism disorder Wilson's disease (WD) is one such condition that is caused by mutations in the ATP7B gene. Delay in treatment could result in irreversible disability or even death. Although liver disease is the most common presenting feature in children, some children may initially present with a subtle neurological presentation only. In patients presenting with dystonia, tremor, dysarthria or with a deterioration in school performance, there should be a high index of suspicion for WD. However, the differential of these clinical presentations is wide and exclusion of WD is difficult. No single diagnostic test can exclude WD and each of the biochemical tests has limitations. In this article, we discuss copper metabolism disorders including WD and Menke's disease. We then discuss the available diagnostic tests and how to investigate children for these rare disorders.


Subject(s)
Hepatolenticular Degeneration/diagnosis , Menkes Kinky Hair Syndrome/diagnosis , Adolescent , Algorithms , C-Reactive Protein/metabolism , Ceruloplasmin/metabolism , Child , Child, Preschool , Copper/metabolism , Female , Genetic Testing , Hepatolenticular Degeneration/complications , Hepatolenticular Degeneration/metabolism , Humans , Infant , Liver Function Tests , Male , Menkes Kinky Hair Syndrome/complications , Menkes Kinky Hair Syndrome/metabolism , Young Adult
9.
Nihon Rinsho ; 74(7): 1151-5, 2016 Jul.
Article in Japanese | MEDLINE | ID: mdl-27455805

ABSTRACT

Copper is one of essential trace elements. Copper deficiency lead to growth and developmental failure and/or neurological dysfunction. However, excess copper is also problems for human life. There are two disorders of inborn error of copper metabolism, Menkes disease and Wilson disease. Menkes disease is an X linked recessive disorder with copper deficiency and Wilson disease is an autosomal recessive disorder with copper accumulation. These both disorders result from the defective functioning of copper transport P-type ATPase, ATP7A of Menkes disease and ATP7B of Wilson disease. In this paper, the author describes about copper metabolism of human, and clinical feature, diagnosis and treatment of Menkes disease and Wilson disease.


Subject(s)
Copper/metabolism , Genetic Predisposition to Disease , Hepatolenticular Degeneration/metabolism , Menkes Kinky Hair Syndrome/metabolism , Hepatolenticular Degeneration/diagnosis , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/genetics , Humans , Menkes Kinky Hair Syndrome/diagnosis , Menkes Kinky Hair Syndrome/drug therapy , Menkes Kinky Hair Syndrome/genetics , Prognosis
10.
Am J Physiol Cell Physiol ; 309(10): C660-8, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26269458

ABSTRACT

Menkes disease is a fatal neurodegenerative disorder arising from a systemic copper deficiency caused by loss-of-function mutations in a ubiquitously expressed copper transporter, ATP7A. Although this disorder reveals an essential role for copper in the developing human nervous system, the role of ATP7A in the pathogenesis of signs and symptoms in affected patients, including severe mental retardation, ataxia, and excitotoxic seizures, remains unknown. To directly examine the role of ATP7A within the central nervous system, we generated Atp7a(Nes) mice, in which the Atp7a gene was specifically deleted within neural and glial cell precursors without impairing systemic copper homeostasis, and compared these mice with the mottled brindle (mo-br) mutant, a murine model of Menkes disease in which Atp7a is defective in all cells. Whereas mo-br mice displayed neurodegeneration, demyelination, and 100% mortality prior to weaning, the Atp7a(Nes) mice showed none of these phenotypes, exhibiting only mild sensorimotor deficits, increased anxiety, and susceptibility to NMDA-induced seizure. Our results indicate that the pathophysiology of severe neurological signs and symptoms in Menkes disease is the result of copper deficiency within the central nervous system secondary to impaired systemic copper homeostasis and does not arise from an intrinsic lack of ATP7A within the developing brain. Furthermore, the sensorimotor deficits, hypophagia, anxiety, and sensitivity to NMDA-induced seizure in the Atp7a(Nes) mice reveal unique autonomous requirements for ATP7A in the nervous system. Taken together, these data reveal essential roles for copper acquisition in the central nervous system in early development and suggest novel therapeutic approaches in affected patients.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Copper/metabolism , Menkes Kinky Hair Syndrome/metabolism , Adenosine Triphosphatases/genetics , Animals , Cation Transport Proteins/genetics , Copper-Transporting ATPases , Female , Gene Expression Regulation/physiology , Integrases , Male , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/pathology , Mice , Mice, Knockout , Mutation
11.
Mol Genet Metab ; 113(4): 294-300, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25456742

ABSTRACT

Mottled-dappled (Mo-dp) is a mouse model of Menkes disease caused by a large, previously uncharacterized deletion in the 5' region of Atp7a, the mouse ortholog of ATP7A. Affected mutants die in utero at embryonic day 17, and show bending and thickening of the ribs and distortion of the pectoral and pelvic girdles and limbs. To characterize this allele, we designed a custom 4x180K microarray on the mouse X chromosome and performed comparative genomic hybridization using extracted DNA from normal and carrier Mo-dp females, and identified an approximately 9 kb deletion. We used PCR to fine-map the breakpoints and amplify a junction fragment of 630 bp. Sequencing of the junction fragment disclosed the exact breakpoint locations and that the Mo-dp deletion is precisely 8990 bp, including approximately 2 kb in the promoter region of Atp7a. Western blot analysis of Mo-dp heterozygous brains showed diminished amounts of Atp7a protein, consistent with reduced expression due to the promoter region deletion on one allele. In heterozygous females, brain copper levels tended to be lower compared to wild type whereas neurochemical analyses revealed higher dihydroxyphenylacetic acid:dihydroxyphenylglycol (DOPAC:DHPG) and dopamine:norepinephrine (DA:NE) ratios compared to normal (P=0.002 and 0.029, respectively), consistent with partial deficiency of dopamine-beta-hydroxylase, a copper-dependent enzyme. Heterozygous females showed no significant differences in body weight compared to wild type females. Our results delineate the molecular details of the Mo-dp mutation for the first time and define novel biochemical findings in heterozygous female carriers of this allele.


Subject(s)
Adenosine Triphosphatases/genetics , Cation Transport Proteins/genetics , Disease Models, Animal , Menkes Kinky Hair Syndrome/genetics , Alleles , Animals , Blotting, Western , Body Weight , Brain/metabolism , Comparative Genomic Hybridization , Copper/metabolism , Copper-Transporting ATPases , Dopamine beta-Hydroxylase/metabolism , Female , Heterozygote , Humans , Menkes Kinky Hair Syndrome/metabolism , Mice , Oligonucleotide Array Sequence Analysis , Phenotype , Polymerase Chain Reaction , Sequence Analysis, DNA , Sequence Deletion , X Chromosome/genetics
12.
Ann Neurol ; 73(2): 259-65, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23224983

ABSTRACT

OBJECTIVE: Menkes disease is a lethal neurodegenerative disorder of infancy caused by mutations in a copper-transporting adenosine triphosphatase gene, ATP7A. Among its multiple cellular tasks, ATP7A transfers copper to dopamine beta hydroxylase (DBH) within the lumen of the Golgi network or secretory granules, catalyzing the conversion of dopamine to norepinephrine. In a well-established mouse model of Menkes disease, mottled-brindled (mo-br), we tested whether systemic administration of L-threo-dihydroxyphenylserine (L-DOPS), a drug used successfully to treat autosomal recessive norepinephrine deficiency, would improve brain neurochemical abnormalities and neuropathology. METHODS: At 8, 10, and 12 days of age, wild-type and mo-br mice received intraperitoneal injections of 200µg/g body weight of L-DOPS, or mock solution. Five hours after the final injection, the mice were euthanized, and brains were removed. We measured catecholamine metabolites affected by DBH via high-performance liquid chromatography with electrochemical detection, and assessed brain histopathology. RESULTS: Compared to mock-treated controls, mo-br mice that received intraperitoneal L-DOPS showed significant increases in brain norepinephrine (p < 0.001) and its deaminated metabolite, dihydroxyphenylglycol (p < 0.05). The ratio of a non-beta-hydroxylated metabolite in the catecholamine biosynthetic pathway, dihydroxyphenylacetic acid, to the beta-hydroxylated metabolite, dihydroxyphenylglycol, improved equivalently to results obtained previously with brain-directed ATP7A gene therapy (p < 0.01). However, L-DOPS treatment did not arrest global brain pathology or improve somatic growth, as gene therapy had. INTERPRETATION: We conclude that (1) L-DOPS crosses the blood-brain barrier in mo-br mice and corrects brain neurochemical abnormalities, (2) norepinephrine deficiency is not the cause of neurodegeneration in mo-br mice, and (3) L-DOPS treatment may ameliorate noradrenergic hypofunction in Menkes disease.


Subject(s)
Brain Chemistry/drug effects , Brain/drug effects , Droxidopa/pharmacology , Menkes Kinky Hair Syndrome/drug therapy , Nerve Degeneration/drug therapy , 3,4-Dihydroxyphenylacetic Acid/metabolism , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Antiparkinson Agents/pharmacokinetics , Antiparkinson Agents/pharmacology , Blood-Brain Barrier/metabolism , Brain/metabolism , Brain/pathology , Brain Chemistry/physiology , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper/metabolism , Copper-Transporting ATPases , Disease Models, Animal , Dopamine/biosynthesis , Dopamine/metabolism , Dopamine beta-Hydroxylase/metabolism , Droxidopa/pharmacokinetics , Female , Male , Menkes Kinky Hair Syndrome/metabolism , Menkes Kinky Hair Syndrome/pathology , Mice , Mice, Inbred C57BL , Mice, Neurologic Mutants , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Norepinephrine/biosynthesis , Norepinephrine/deficiency , Norepinephrine/metabolism
13.
Biometals ; 27(4): 611-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24748564

ABSTRACT

Copper (Cu) is an essential microelement found in all living organisms with the unique ability to adopt two different redox states-in the oxidized (Cu(2+)) and reduced (Cu(+)). It is required for survival and serves as an important catalytic cofactor in redox chemistry for proteins that carry out fundamental biological functions, important in growth and development. The deficit of copper can result in impaired energy production, abnormal glucose and cholesterol metabolism, increased oxidative damage, increased tissue iron (Fe) accrual, altered structure and function of circulating blood and immune cells, abnormal neuropeptides synthesis and processing, aberrant cardiac electrophysiology, impaired myocardial contractility, and persistent effects on the neurobehavioral and the immune system. Increased copper level has been found in several disorders like e.g.: Wilson's disease or Menke's disease. New findings with the great potential for impact in medicine include the use of copper-lowering therapy for antiangiogenesis, antifibrotic and anti-inflammatory purposes. The role of copper in formation of amyloid plaques in Alzheimer's disease, and successful treatment of this disorder in rodent model by copper chelating are also of interest. In this work we will try to describe essential aspects of copper in chosen diseases. We will represent the evidence available on adverse effect derived from copper deficiency and copper excess. We will try to review also the copper biomarkers (chosen enzymes) that help reflect the level of copper in the body.


Subject(s)
Copper/physiology , Alzheimer Disease/metabolism , Animals , Copper/toxicity , Environmental Pollutants/toxicity , Hepatolenticular Degeneration/metabolism , Humans , Menkes Kinky Hair Syndrome/metabolism , Metalloproteins/metabolism , Oxidation-Reduction , Oxidative Stress
14.
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
15.
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
16.
Anal Bioanal Chem ; 405(6): 1809-20, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23079951

ABSTRACT

Copper plays an important role in numerous biological processes across all living systems predominantly because of its versatile redox behavior. Cellular copper homeostasis is tightly regulated and disturbances lead to severe disorders such as Wilson disease and Menkes disease. Age-related changes of copper metabolism have been implicated in other neurodegenerative disorders such as Alzheimer disease. The role of copper in these diseases has been a topic of mostly bioinorganic research efforts for more than a decade, metal-protein interactions have been characterized, and cellular copper pathways have been described. Despite these efforts, crucial aspects of how copper is associated with Alzheimer disease, for example, are still only poorly understood. To take metal-related disease research to the next level, emerging multidimensional imaging techniques are now revealing the copper metallome as the basis to better understand disease mechanisms. This review describes how recent advances in X-ray fluorescence microscopy and fluorescent copper probes have started to contribute to this field, specifically in Wilson disease and Alzheimer disease. It furthermore provides an overview of current developments and future applications in X-ray microscopic methods.


Subject(s)
Copper/chemistry , Imaging, Three-Dimensional/methods , Molecular Imaging/methods , Spectrometry, X-Ray Emission/instrumentation , Alzheimer Disease/diagnosis , Alzheimer Disease/metabolism , Cations, Divalent , Copper/metabolism , Fluorescent Dyes , Hepatolenticular Degeneration/diagnosis , Hepatolenticular Degeneration/metabolism , Homeostasis , Humans , Imaging, Three-Dimensional/instrumentation , Menkes Kinky Hair Syndrome/diagnosis , Menkes Kinky Hair Syndrome/metabolism , Molecular Imaging/instrumentation , Plaque, Amyloid/chemistry , Spectroscopy, Fourier Transform Infrared
17.
Cell Mol Life Sci ; 69(1): 149-63, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21667063

ABSTRACT

Menkes disease (MD) is an X-linked recessive disorder characterized by copper deficiency resulting in a diminished function of copper-dependent enzymes. Most MD patients die in early childhood, although mild forms of MD have also been described. A diversity of mutations in the gene encoding of the Golgi-resident copper-transporting P(1B)-type ATPase ATP7A underlies MD. To elucidate the molecular consequences of the ATP7A mutations, various mutations in ATP7A associated with distinct phenotypes of MD (L873R, C1000R, N1304S, and A1362D) were analyzed in detail. All mutants studied displayed changes in protein expression and intracellular localization parallel to a dramatic decline in their copper-transporting capacity compared to ATP7A the wild-type. We restored these observed defects in ATP7A mutant proteins by culturing the cells at 30°C, which improves the quality of protein folding, similar to that which as has recently has been demonstrated for misfolded ATP7B, a copper transporter homologous to ATP7A. Further, the effect of the canine copper toxicosis protein COMMD1 on ATP7A function was examined as COMMD1 has been shown to regulate the proteolysis of ATP7B proteins. Interestingly, in addition to adjusted growth temperature, binding of COMMD1 partially restored the expression, subcellular localization, and copper-exporting activities of the ATP7A mutants. However, no effect of pharmacological chaperones was observed. Together, the presented data might provide a new direction for developing therapies to improve the residual exporting activity of unstable ATP7A mutant proteins, and suggests a potential role for COMMD1 in this process.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Adenosine Triphosphatases , Cation Transport Proteins , Menkes Kinky Hair Syndrome , Adenosine Triphosphatases/deficiency , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Cation Transport Proteins/deficiency , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Cell Culture Techniques/methods , Cell Line, Tumor , Copper/deficiency , Copper/metabolism , Copper-Transporting ATPases , Dogs , Golgi Apparatus/metabolism , HEK293 Cells , Humans , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Mice , Mutation/genetics , Protein Folding , Protein Transport/genetics , Proteolysis , Temperature
18.
Nat Genet ; 6(4): 374-8, 1994 Apr.
Article in English | MEDLINE | ID: mdl-8054977

ABSTRACT

The murine homologue of the Menkes disease gene (MNK) was isolated from cDNA libraries, using human cDNA clones as probes, and by PCR. The predicted amino acid sequence shows a high level of identity (89.9%) with the human protein, and the predicted functional domains in the human protein are present. Using probes to the mouse Mnk gene, we found that the mottled dappled mutation was caused by alteration in the Mnk locus and lack of expression of Mnk RNA. Tissues of the blotchy mouse contained two larger sizes of MNK mRNA demonstrating a likely defect in RNA splicing. Thus, the mottled locus is homologous to the human MNK locus and dappled and blotchy are allelic mutations in this gene.


Subject(s)
Adenosine Triphosphatases/genetics , Carrier Proteins/genetics , Cation Transport Proteins , Disease Models, Animal , Hair Color/genetics , Menkes Kinky Hair Syndrome/genetics , Mice, Mutant Strains/genetics , Recombinant Fusion Proteins , Adenosine Triphosphatases/biosynthesis , Adenosine Triphosphatases/chemistry , Alleles , Amino Acid Sequence , Animals , Base Sequence , Blotting, Southern , Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Copper-Transporting ATPases , Female , Fetal Diseases/genetics , Fetal Diseases/metabolism , Gene Expression Regulation , Humans , Male , Menkes Kinky Hair Syndrome/embryology , Menkes Kinky Hair Syndrome/metabolism , Mice , Mice, Mutant Strains/embryology , Molecular Sequence Data , Protein Structure, Tertiary , RNA, Messenger/genetics , Sequence Homology, Amino Acid
19.
Biomolecules ; 13(12)2023 12 05.
Article in English | MEDLINE | ID: mdl-38136617

ABSTRACT

Menkes' disease (MD) and Wilson's disease (WD) are two major copper (Cu) metabolism-related disorders caused by mutations of the ATP7A and ATP7B ATPase gene, respectively. While Cu is involved in DNA strand breaks signaling and repair, the response of cells from both diseases to ionizing radiation, a common DNA strand breaks inducer, has not been investigated yet. To this aim, three MD and two WD skin fibroblasts lines were irradiated at two Gy X-rays and clonogenic cell survival, micronuclei, anti-γH2AX, -pATM, and -MRE11 immunofluorescence assays were applied to evaluate the DNA double-strand breaks (DSB) recognition and repair. MD and WD cells appeared moderately radiosensitive with a delay in the radiation-induced ATM nucleo-shuttling (RIANS) associated with impairments in the DSB recognition. Such delayed RIANS was notably caused in both MD and WD cells by a highly expressed ATP7B protein that forms complexes with ATM monomers in cytoplasm. Interestingly, a Cu pre-treatment of cells may influence the activity of the MRE11 nuclease and modulate the radiobiological phenotype. Lastly, some high-passage MD cells cultured in routine may transform spontaneously becoming immortalized. Altogether, our findings suggest that exposure to ionizing radiation may impact on clinical features of MD and WD, which requires cautiousness when affected patients are submitted to radiodiagnosis and, eventually, radiotherapy.


Subject(s)
Hepatolenticular Degeneration , Menkes Kinky Hair Syndrome , Humans , Copper/metabolism , Protein Kinases/metabolism , Radiation, Ionizing , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Hepatolenticular Degeneration/genetics , Fibroblasts/metabolism , DNA/metabolism , Ataxia Telangiectasia Mutated Proteins/metabolism
20.
J Biol Chem ; 286(30): 26585-94, 2011 Jul 29.
Article in English | MEDLINE | ID: mdl-21646353

ABSTRACT

The copper-transporting ATPase ATP7A has an essential role in human physiology. ATP7A transfers the copper cofactor to metalloenzymes within the secretory pathway; inactivation of ATP7A results in an untreatable neurodegenerative disorder, Menkes disease. Presently, the mechanism of ATP7A-mediated copper release into the secretory pathway is not understood. We demonstrate that the characteristic His/Met-rich segment Met(672)-Pro(707) (HM-loop) that connects the first two transmembrane segments of ATP7A is important for copper release. Mutations within this loop do not prevent the ability of ATP7A to form a phosphorylated intermediate during ATP hydrolysis but inhibit subsequent dephosphorylation, a step associated with copper release. The HM-loop inserted into a scaffold protein forms two structurally distinct binding sites and coordinates copper in a mixed His-Met environment with an ∼2:1 stoichiometry. Binding of either copper or silver, a Cu(I) analog, induces structural changes in the loop. Mutations of 4 Met residues to Ile or two His-His pairs to Ala-Gly decrease affinity for copper. Altogether, the data suggest a two-step process, where copper released from the transport sites binds to the first His(Met)(2) site, triggering a structural change and binding to a second 2-coordinate His-His or His-Met site. We also show that copper binding within the HM-loop stabilizes Cu(I) and protects it from oxidation, which may further aid the transfer of copper from ATP7A to acceptor proteins. The mechanism of copper entry into the secretory pathway is discussed.


Subject(s)
Adenosine Triphosphatases/metabolism , Cation Transport Proteins/metabolism , Copper/metabolism , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/genetics , Adenosine Triphosphate/chemistry , Adenosine Triphosphate/genetics , Adenosine Triphosphate/metabolism , Binding Sites , Biological Transport/genetics , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cell Line , Copper/chemistry , Copper-Transporting ATPases , Humans , Menkes Kinky Hair Syndrome/genetics , Menkes Kinky Hair Syndrome/metabolism , Mutation , Protein Binding , Protein Structure, Secondary , Silver/chemistry , Silver/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL