Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Phys Chem B ; 128(22): 5336-5343, 2024 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-38780400

RESUMO

Copper, an essential metal for various cellular processes, requires tight regulation to prevent cytotoxicity. Intracellular pathways crucial for maintaining optimal copper levels involve soluble and membrane transporters, namely, metallochaperones and P-type ATPases, respectively. In this study, we used a simulation workflow based on free-energy perturbation (FEP) theory and parallel bias metadynamics (PBMetaD) to predict the Cu(I) exchange mechanism between the human Cu(I) chaperone, Atox1, and one of its two physiological partners, ATP7A. ATP7A, also known as the Menkes disease protein, is a transmembrane protein and one of the main copper-transporting ATPases. It pumps copper into the trans-Golgi network for the maturation of cuproenzymes and is also essential for the efflux of excess copper across the plasma membrane. In this analysis, we utilized the nuclear magnetic resonance (NMR) structure of the Cu(I)-mediated complex between Atox1 and the first soluble domain of the Menkes protein (Mnk1) as a starting point. Independent free-energy simulations were conducted to investigate the dissociation of both Atox1 and Mnk1. The calculations revealed that the two dissociations require free energy values of 6.3 and 6.2 kcal/mol, respectively, following a stepwise dissociation mechanism.


Assuntos
Proteínas de Transporte de Cobre , ATPases Transportadoras de Cobre , Cobre , Metalochaperonas , Chaperonas Moleculares , Simulação de Dinâmica Molecular , Cobre/química , Cobre/metabolismo , Proteínas de Transporte de Cobre/química , Proteínas de Transporte de Cobre/metabolismo , Humanos , Metalochaperonas/química , Metalochaperonas/metabolismo , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Chaperonas Moleculares/química , Chaperonas Moleculares/metabolismo , Termodinâmica , Multimerização Proteica
2.
PLoS Comput Biol ; 18(9): e1010074, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36070320

RESUMO

ATP7B is a human copper-transporting P1B-type ATPase that is involved in copper homeostasis and resistance to platinum drugs in cancer cells. ATP7B consists of a copper-transporting core and a regulatory N-terminal tail that contains six metal-binding domains (MBD1-6) connected by linker regions. The MBDs can bind copper, which changes the dynamics of the regulatory domain and activates the protein, but the underlying mechanism remains unknown. To identify possible copper-specific structural dynamics involved in transport regulation, we constructed a model of ATP7B spanning the N-terminal tail and core catalytic domains and performed molecular dynamics (MD) simulations with (holo) and without (apo) copper ions bound to the MBDs. In the holo protein, MBD2, MBD3 and MBD5 showed enhanced mobilities, which resulted in a more extended N-terminal regulatory region. The observed separation of MBD2 and MBD3 from the core protein supports a mechanism where copper binding activates the ATP7B protein by reducing interactions among MBD1-3 and between MBD1-3 and the core protein. We also observed an increased interaction between MBD5 and the core protein that brought the copper-binding site of MBD5 closer to the high-affinity internal copper-binding site in the core protein. The simulation results assign specific, mechanistic roles to the metal-binding domains involved in ATP7B regulation that are testable in experimental settings.


Assuntos
ATPases Transportadoras de Cobre , Cobre , Sítios de Ligação , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Proteínas de Ligação a DNA/metabolismo , Humanos , Domínios Proteicos
3.
Nat Commun ; 13(1): 5121, 2022 08 31.
Artigo em Inglês | MEDLINE | ID: mdl-36045128

RESUMO

Copper is essential for living cells, yet toxic at elevated concentrations. Class 1B P-type (P1B-) ATPases are present in all kingdoms of life, facilitating cellular export of transition metals including copper. P-type ATPases follow an alternating access mechanism, with inward-facing E1 and outward-facing E2 conformations. Nevertheless, no structural information on E1 states is available for P1B-ATPases, hampering mechanistic understanding. Here, we present structures that reach 2.7 Å resolution of a copper-specific P1B-ATPase in an E1 conformation, with complementing data and analyses. Our efforts reveal a domain arrangement that generates space for interaction with ion donating chaperones, and suggest a direct Cu+ transfer to the transmembrane core. A methionine serves a key role by assisting the release of the chaperone-bound ion and forming a cargo entry site together with the cysteines of the CPC signature motif. Collectively, the findings provide insights into P1B-mediated transport, likely applicable also to human P1B-members.


Assuntos
ATPases Transportadoras de Cobre , Cobre , Adenosina Trifosfatases/química , Adenosina Trifosfatases/metabolismo , Transporte Biológico , Cobre/química , Cobre/metabolismo , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Humanos , Chaperonas Moleculares/metabolismo
4.
Cells ; 10(11)2021 11 10.
Artigo em Inglês | MEDLINE | ID: mdl-34831341

RESUMO

Macroautophagy/autophagy plays an important role in cellular copper clearance. The means by which the copper metabolism and autophagy pathways interact mechanistically is vastly unexplored. Dysfunctional ATP7B, a copper-transporting ATPase, is involved in the development of monogenic Wilson disease, a disorder characterized by disturbed copper transport. Using in silico prediction, we found that ATP7B contains a number of potential binding sites for LC3, a central protein in the autophagy pathway, the so-called LC3 interaction regions (LIRs). The conserved LIR3, located at the C-terminal end of ATP7B, was found to directly interact with LC3B in vitro. Replacing the two conserved hydrophobic residues W1452 and L1455 of LIR3 significantly reduced interaction. Furthermore, autophagy was induced in normal human hepatocellular carcinoma cells (HepG2) leading to enhanced colocalization of ATP7B and LC3B on the autophagosome membranes. By contrast, HepG2 cells deficient of ATP7B (HepG2 ATP7B-/-) showed autophagy deficiency at elevated copper condition. This phenotype was complemented by heterologous ATP7B expression. These findings suggest a cooperative role of ATP7B and LC3B in autophagy-mediated copper clearance.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Sequência de Aminoácidos , Transporte Biológico/efeitos dos fármacos , Cobre/farmacologia , ATPases Transportadoras de Cobre/química , Células Hep G2 , Humanos , Ligação Proteica/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos
5.
Int J Pharm ; 609: 121193, 2021 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-34673167

RESUMO

Copper homeostasis is finely regulated in human to avoid any detrimental impact of free intracellular copper ions. Upon copper accumulation, biliary excretion is triggered in liver thanks to trafficking of the ATP7B copper transporter to bile canaliculi. However, in Wilson's disease this protein is mutated leading to copper accumulation. Current therapy uses Cu chelators acting extracellularly and requiring a life-long treatment with side effects. Herein, a new Cu(I) pro-chelator was encapsulated in long-term stable nanostructured lipid carriers. Cellular assays revealed that the pro-chelator protects hepatocytes against Cu-induced cell death. Besides, the cellular stresses induced by moderate copper concentrations, including protein unfolding, are counteracted by the pro-chelator. These data showed the pro-chelator efficiency to deliver intracellularly an active chelator that copes with copper stress and surpasses current and under development chelators. Although its biological activity is more mitigated, the pro-chelator nanolipid formulation led to promising results. This innovative approach is of outmost importance in the quest of better treatments for Wilson's disease.


Assuntos
Degeneração Hepatolenticular , Quelantes , Cobre , ATPases Transportadoras de Cobre/química , Hepatócitos , Degeneração Hepatolenticular/tratamento farmacológico , Humanos
6.
Int J Mol Sci ; 22(15)2021 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-34361043

RESUMO

Intravesicular pH plays a crucial role in melanosome maturation and function. Melanosomal pH changes during maturation from very acidic in the early stages to neutral in late stages. Neutral pH is critical for providing optimal conditions for the rate-limiting, pH-sensitive melanin-synthesizing enzyme tyrosinase (TYR). This dramatic change in pH is thought to result from the activity of several proteins that control melanosomal pH. Here, we computationally investigated the pH-dependent stability of several melanosomal membrane proteins and compared them to the pH dependence of the stability of TYR. We confirmed that the pH optimum of TYR is neutral, and we also found that proteins that are negative regulators of melanosomal pH are predicted to function optimally at neutral pH. In contrast, positive pH regulators were predicted to have an acidic pH optimum. We propose a competitive mechanism among positive and negative regulators that results in pH equilibrium. Our findings are consistent with previous work that demonstrated a correlation between the pH optima of stability and activity, and they are consistent with the expected activity of positive and negative regulators of melanosomal pH. Furthermore, our data suggest that disease-causing variants impact the pH dependence of melanosomal proteins; this is particularly prominent for the OCA2 protein. In conclusion, melanosomal pH appears to affect the activity of multiple melanosomal proteins.


Assuntos
Antígenos de Neoplasias/química , ATPases Transportadoras de Cobre/química , Melanossomas/metabolismo , Proteínas de Membrana Transportadoras/química , Simulação de Dinâmica Molecular , Monofenol Mono-Oxigenase/química , Prótons , Antígenos de Neoplasias/metabolismo , ATPases Transportadoras de Cobre/metabolismo , Humanos , Concentração de Íons de Hidrogênio , Melanossomas/química , Proteínas de Membrana Transportadoras/metabolismo , Monofenol Mono-Oxigenase/metabolismo , Estabilidade Proteica
7.
Metallomics ; 12(12): 1941-1950, 2020 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-33094790

RESUMO

Nanobodies are genetically engineered single domain antibodies derived from the unusual heavy-chain only antibodies found in llamas and camels. The small size of the nanobodies and flexible selection schemes make them uniquely versatile tools for protein biochemistry and cell biology. We have developed a panel of nanobodies against the metal binding domains of the human copper transporter ATP7B, a multidomain membrane protein with a complex regulation of enzymatic activity and intracellular localization. To enable the use of the nanobodies as tools to investigate copper transport in the cell, we characterized their binding sites and affinity by isothermal titration calorimetry and NMR. We have identified nanobodies against each of the first four metal binding domains of ATP7B, with a wide affinity range, as evidenced by dissociation constants from below 10-9 to 10-6 M. We found both the inhibitory and activating nanobodies among those tested. The diverse properties of the nanobodies make the panel useful for the structural studies of ATP7B, immunoaffinity purification of the protein, modulation of its activity in the cell, protein dynamics studies, and as mimics of copper chaperone ATOX1, the natural interaction partner of ATP7B.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Anticorpos de Domínio Único/farmacologia , Sítios de Ligação/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , ATPases Transportadoras de Cobre/química , Humanos , Simulação de Acoplamento Molecular , Domínios Proteicos/efeitos dos fármacos
8.
Int J Mol Sci ; 21(15)2020 Aug 02.
Artigo em Inglês | MEDLINE | ID: mdl-32748830

RESUMO

Copper's essentiality and toxicity mean it requires a sophisticated regulation system for its acquisition, cellular distribution and excretion, which until now has remained elusive. Herein, we applied continuous wave (CW) and pulsed electron paramagnetic resonance (EPR) spectroscopy in solution to resolve the copper trafficking mechanism in humans, by considering the route travelled by Cu(I) from the metallochaperone Atox1 to the metal binding domains of ATP7B. Our study revealed that Cu(I) is most likely mediated by the binding of the Atox1 monomer to metal binding domain 1 (MBD1) and MBD4 of ATP7B in the final part of its extraction pathway, while the other MBDs mediate this interaction and participate in copper transfer between the various MBDs to the ATP7B membrane domain. This research also proposes that MBD1-3 and MBD4-6 act as two independent units.


Assuntos
Proteínas de Transporte de Cobre/metabolismo , ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica/métodos , Chaperonas Moleculares/metabolismo , Sítios de Ligação , Cobre/química , Proteínas de Transporte de Cobre/química , ATPases Transportadoras de Cobre/química , Humanos , Modelos Moleculares , Chaperonas Moleculares/química , Ligação Proteica , Domínios Proteicos
9.
Mol Biochem Parasitol ; 235: 111245, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31751595

RESUMO

Copper is an essential component of cuproproteins but can be toxic to cells, therefore copper metabolism is very carefully regulated within cells. To gain insight into trypanosome copper metabolism, Trypanosoma spp. genomic databases were screened for the presence of copper-containing and -transporting proteins. Among other genes encoding copper-binding proteins, a copper-transporting P-type ATPase (CuATPase) gene was identified. Sequence and phylogenetic analyses suggest that the gene codes for a Cu+ transporter belonging to the P1B-1 ATPase subfamily that has an N-terminal domain with copper binding motifs. The N-terminal cytosolic domains of the proteins from Trypanosoma congolense and Trypanosoma brucei brucei were recombinantly expressed in Escherichia coli as maltose binding protein (MBP) fusion proteins. These N-terminal domains bound copper in vitro and within E. coli cells, more than the control MBP fusion partner alone. The copper binding properties of the recombinant proteins were further confirmed when they inhibited copper catalysed ascorbate oxidation. Native CuATPases were detected in a western blot of lysates of T. congolense IL3000 and T. b. brucei ILTat1.1 bloodstream form parasites using affinity purified IgY antibodies against N-terminal domain peptides. The CuATPase was also detected by immunofluorescence in T. b. brucei bloodstream form parasites where it was associated with subcellular vesicles. In conclusion, Trypanosoma species express a copper-transporting P1B-1-type ATPase and together with other copper-binding proteins identified in the genomes of kinetoplastid parasites may constitute potential targets for anti-trypanosomal drug discovery.


Assuntos
ATPases Transportadoras de Cobre , Cobre/metabolismo , Trypanosoma , Animais , Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/imunologia , Proteínas de Transporte/genética , Proteínas de Transporte/imunologia , Proteínas de Transporte/metabolismo , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/imunologia , ATPases Transportadoras de Cobre/metabolismo , Vesículas Citoplasmáticas , Escherichia coli/genética , Transporte Proteico , Proteínas Recombinantes/genética , Trypanosoma/genética , Trypanosoma/metabolismo , Trypanosoma brucei brucei/genética , Trypanosoma brucei brucei/metabolismo , Trypanosoma congolense/genética , Trypanosoma congolense/metabolismo
10.
Annu Rev Biochem ; 89: 583-603, 2020 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-31874046

RESUMO

P-type ATPases are found in all kingdoms of life and constitute a wide range of cation transporters, primarily for H+, Na+, K+, Ca2+, and transition metal ions such as Cu(I), Zn(II), and Cd(II). They have been studied through a wide range of techniques, and research has gained very significant insight on their transport mechanism and regulation. Here, we review the structure, function, and dynamics of P2-ATPases including Ca2+-ATPases and Na,K-ATPase. We highlight mechanisms of functional transitions that are associated with ion exchange on either side of the membrane and how the functional cycle is regulated by interaction partners, autoregulatory domains, and off-cycle states. Finally, we discuss future perspectives based on emerging techniques and insights.


Assuntos
Trifosfato de Adenosina/química , ATPases Transportadoras de Cobre/química , ATPase Trocadora de Hidrogênio-Potássio/química , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/química , ATPase Trocadora de Sódio-Potássio/química , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Cátions Bivalentes , Cátions Monovalentes , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , ATPase Trocadora de Hidrogênio-Potássio/genética , ATPase Trocadora de Hidrogênio-Potássio/metabolismo , Humanos , Transporte de Íons , Modelos Moleculares , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Secundária de Proteína , Prótons , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/genética , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Imagem Individual de Molécula , ATPase Trocadora de Sódio-Potássio/genética , ATPase Trocadora de Sódio-Potássio/metabolismo , Especificidade por Substrato
11.
J Chem Inf Model ; 59(12): 5230-5243, 2019 12 23.
Artigo em Inglês | MEDLINE | ID: mdl-31751128

RESUMO

Genetic variations in the gene encoding the copper-transport protein ATP7B are the primary cause of Wilson's disease. Controversially, clinical prevalence seems much smaller than the prevalence estimated by genetic screening tools, causing fear that many people are undiagnosed, although early diagnosis and treatment is essential. To address this issue, we benchmarked 16 state-of-the-art computational disease-prediction methods against established data of missense ATP7B mutations. Our results show that the quality of the methods varies widely. We show the importance of optimizing the threshold of the methods used to distinguish pathogenic from nonpathogenic mutations against data of clinically confirmed pathogenic and nonpathogenic mutations. We find that most methods use thresholds that predict too many ATP7B mutations to be pathogenic. Thus, our findings explain the current controversy on Wilson's disease prevalence because meta-analysis and text search methods include many computational estimates that lead to higher disease prevalence than clinically observed. As proteins and diseases differ widely, a one-size-fits-all threshold cannot distinguish pathogenic and nonpathogenic mutations efficiently, as shown here. We also show that amino acid changes with small evolutionary substitution probability, mainly due to amino acid volume, are more associated with the disease, implying a pathological effect on the conformational state of the protein, which could affect copper transport or adenosine triphosphate recognition and hydrolysis. These findings may be a first step toward a more quantitative genotype-phenotype relationship of Wilson's disease.


Assuntos
ATPases Transportadoras de Cobre/genética , Degeneração Hepatolenticular/enzimologia , Degeneração Hepatolenticular/genética , Mutação , Sequência de Aminoácidos , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Frequência do Gene , Degeneração Hepatolenticular/patologia , Humanos , Prevalência
12.
Int J Mol Sci ; 20(14)2019 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-31337158

RESUMO

Appropriate maintenance of Cu(I) homeostasis is an essential requirement for proper cell function because its misregulation induces the onset of major human diseases and mortality. For this reason, several research efforts have been devoted to dissecting the inner working mechanism of Cu(I)-binding proteins and transporters. A commonly adopted strategy relies on mutations of cysteine residues, for which Cu(I) has an exquisite complementarity, to serines. Nevertheless, in spite of the similarity between these two amino acids, the structural and functional impact of serine mutations on Cu(I)-binding biomolecules remains unclear. Here, we applied various biochemical and biophysical methods, together with all-atom simulations, to investigate the effect of these mutations on the stability, structure, and aggregation propensity of Cu(I)-binding proteins, as well as their interaction with specific partner proteins. Among Cu(I)-binding biomolecules, we focused on the eukaryotic Atox1-ATP7B system, and the prokaryotic CueR metalloregulator. Our results reveal that proteins containing cysteine-to-serine mutations can still bind Cu(I) ions; however, this alters their stability and aggregation propensity. These results contribute to deciphering the critical biological principles underlying the regulatory mechanism of the in-cell Cu(I) concentration, and provide a basis for interpreting future studies that will take advantage of cysteine-to-serine mutations in Cu(I)-binding systems.


Assuntos
Substituição de Aminoácidos , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Cisteína/genética , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutação , Serina/genética , ATPases Transportadoras de Cobre/genética , Humanos , Metalochaperonas/química , Metalochaperonas/genética , Metalochaperonas/metabolismo , Modelos Moleculares , Conformação Proteica , Análise Espectral , Relação Estrutura-Atividade
13.
Metallomics ; 11(9): 1472-1480, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31321400

RESUMO

In human cells, copper (Cu) ions are transported by the cytoplasmic Cu chaperone Atox1 to the Wilson disease protein (ATP7B) in the Golgi for loading of Cu-dependent enzymes. ATP7B is a membrane-spanning protein which, in contrast to non-mammalian homologs, has six cytoplasmic metal-binding domains (MBDs). To address the reason for multiple MBDs, we introduced strategic mutations in which one, two or three MBDs had been blocked for Cu binding via cysteine-to-serine mutations (but all six MBDs are present in all) in a yeast system that probes Cu flow through Atox1 and ATP7B. The results, combined with earlier work, support a mechanistic model in which MBD1-3 forms a regulatory unit of ATP7B Cu transport. Cu delivery via Atox1 to this unit, followed by loading of Cu in MBD3, promotes release of inhibitory interactions. Whereas the Cu site in MBD4 can be mutated without a large effect, an intact Cu site in either MBD5 or MBD6 is required for Cu transport. All MBDs, expressed as single-domain proteins, can replace Atox1 and deliver Cu to full-length ATP7B. However, only MBD6 can deliver Cu to truncated ATP7B where all six MBDs are removed, suggesting a docking role for this structural unit.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Degeneração Hepatolenticular/metabolismo , Sítios de Ligação , Transporte Biológico Ativo , Proteínas de Transporte de Cobre/metabolismo , ATPases Transportadoras de Cobre/química , Humanos , Chaperonas Moleculares/metabolismo , Domínios Proteicos
14.
Metallomics ; 11(7): 1288-1297, 2019 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-31187846

RESUMO

Copper's essentiality and toxicity require a meticulous mechanism for its acquisition, cellular distribution and excretion, which remains hitherto elusive. Herein, we jointly employed electron paramagnetic resonance spectroscopy and all-atom simulations to resolve the copper trafficking mechanism in humans considering the route travelled by Cu(i) from the metallochaperone Atox1 to the metal binding domains 3 and 4 of ATP7B. Our study shows that Cu(i) in the final part of its extraction pathway is most likely mediated by binding of Atox1 monomer to MBD4 of ATP7B. This interaction takes place through weak metal-stabilized protein-protein interactions.


Assuntos
Proteínas de Transporte de Cobre/metabolismo , ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Chaperonas Moleculares/metabolismo , Sítios de Ligação , Transporte Biológico , ATPases Transportadoras de Cobre/química , Espectroscopia de Ressonância de Spin Eletrônica , Humanos , Modelos Moleculares , Domínios Proteicos , Mapas de Interação de Proteínas
15.
Metallomics ; 11(6): 1128-1139, 2019 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-31070637

RESUMO

Single nucleotide polymorphisms (SNPs) are the largest source of sequence variation in the human genome. However, their functional significance is not well understood. We show that SNPs in the Wilson disease gene, ATP7B, that produce amino-acid substitutions K832R and R952K, modulate ATP7B properties in vitro and influence serum copper (Cu) status in vivo. The presence of R832 is associated with a lower ATP7B abundance and a diminished trafficking in response to elevated Cu. The K832R substitution alters surface exposure of amino acid residues in the actuator domain and increases its conformational flexibility. All SNP-related ATP7B variants (R832/R952, R832/K952, K832/K952, and K832/R952) have Cu-transport activity. However, the activity of ATP7B-K832/K952 is lower compared to other variants. In humans, the presence of K952 is associated with a higher fraction of exchangeable Cu in serum. Thus, SNPs may modulate the properties of ATP7B and the organism Cu status.


Assuntos
ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Polimorfismo de Nucleotídeo Único , Sequência de Aminoácidos , Substituição de Aminoácidos , Cobre/sangue , ATPases Transportadoras de Cobre/química , Células HEK293 , Degeneração Hepatolenticular/sangue , Degeneração Hepatolenticular/genética , Degeneração Hepatolenticular/metabolismo , Humanos , Simulação de Dinâmica Molecular , Conformação Proteica , Transporte Proteico
16.
PLoS One ; 14(5): e0215779, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31059521

RESUMO

Wilson disease (WD) is an autosomal recessive disorder, characterized by excessive deposition of copper in various parts of the body, mainly in the liver and brain. It is caused by mutations in ATP7B. We report here the genetic analysis of 102 WD families from a south Indian population. Thirty-six different ATP7B mutations, including 13 novel ones [p.Ala58fs*19, p.Lys74fs*9, p.Gln281*, p.Pro350fs*12, p.Ser481*, p.Leu735Arg, p.Val752Gly, p.Asn812fs*2, p.Val845Ala, p.His889Pro, p.Ile1184fs*1, p.Val1307Glu and p.Ala1339Pro], were identified in 76/102 families. Interestingly, the mutation analysis of affected individuals in two families identified two different homozygous mutations in each family, and thus each affected individual from these families harbored two mutations in each ATP7B allele. Of 36 mutations, 28 were missense, thus making them the most prevalent mutations identified in the present study. Nonsense, insertion and deletion represented 3/36, 2/36 and 3/36 mutations, respectively. The haplotype analysis suggested founder effects for all the 14 recurrent mutations. Our study thus expands the mutational landscape of ATP7B with a total number of 758 mutations. The mutations identified during the present study will facilitate carrier and pre-symptomatic detection, and prenatal genetic diagnosis in affected families.


Assuntos
ATPases Transportadoras de Cobre/genética , Análise Mutacional de DNA , Degeneração Hepatolenticular/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Haplótipos , Humanos , Índia , Fenótipo
17.
Nat Commun ; 10(1): 186, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30643139

RESUMO

Tetrathiomolybdate (TM) is used in the clinic for the treatment of Wilson's disease by targeting the cellular copper efflux protein ATP7B (WLN). Interestingly, both TM and WLN are associated with the efficacy of cisplatin, a widely used anticancer drug. Herein, we show that TM induces dimerization of the metal-binding domain of ATP7B (WLN4) through a unique sulfur-bridged Mo2S6O2 cluster. TM expels copper ions from Cu-WLN4 and forms a copper-free dimer. The binding of Mo to cysteine residues of WLN4 inhibits platination of the protein. Reaction with multi-domain proteins indicates that TM can also connect two domains in the same molecule, forming Mo-bridged intramolecular crosslinks. These results provide structural and chemical insight into the mechanism of action of TM against ATPase, and reveal the molecular mechanism by which TM attenuates the cisplatin resistance mediated by copper efflux proteins.


Assuntos
Antineoplásicos/farmacologia , Quelantes/farmacologia , Cisplatino/farmacologia , ATPases Transportadoras de Cobre/metabolismo , Molibdênio/farmacologia , Antineoplásicos/uso terapêutico , Quelantes/uso terapêutico , Cisplatino/uso terapêutico , Cobre/metabolismo , ATPases Transportadoras de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/química , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , Reagentes de Ligações Cruzadas/uso terapêutico , Cristalografia por Raios X , Cisteína/química , Cisteína/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Molibdênio/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Platina/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína
18.
Biophys J ; 115(6): 1007-1018, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30173886

RESUMO

The copper-transporting ATPase ATP7B is essential for loading of copper ions to copper-dependent enzymes in the secretory pathway; its inactivation results in Wilson disease. In contrast to copper-ion uptake by the cytoplasmic domains, ATP7B-mediated copper-ion release in the Golgi has not been explored yet. We demonstrate here that a luminal loop in ATP7B, rich in histidine/methionine residues, binds reduced copper (Cu(I)) ions, and identified copper-binding residues play an essential role in ATP7B-mediated metal ion release. NMR experiments on short-peptide models demonstrate that three methionine and two histidine residues are specifically involved in Cu(I) ion binding; with these residues replaced by alanines, no Cu(I) ion interaction is detected. Although more than one Cu(I) ion can interact with the wild-type peptide, removing either all histidine or all methionine residues reduces the stoichiometry to one Cu(I) ion binding per peptide. Using a yeast complementation assay, we show that for efficient copper transport by full-length ATP7B, the complete set of histidine and methionine residues in the lumen loop are required. The replacement of histidine/methionine residues by alanines does not perturb overall ATP7B structure, as the localization of ATP7B variants in yeast cells matches that of the wild-type protein. Thus, in similarity to ATP7A, ATP7B also appears to have a luminal "exit" copper ion site.


Assuntos
ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Sequência de Aminoácidos , Sítios de Ligação , Humanos , Modelos Moleculares , Ligação Proteica , Conformação Proteica em alfa-Hélice , Especificidade por Substrato
19.
Hum Mutat ; 39(12): 1926-1941, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30120852

RESUMO

Wilson disease (WD), a copper metabolism disorder, occurs due to the presence of mutations in the gene encoding ATP7B, a protein that primarily facilitates hepatic copper excretion. A better understanding of spectrum and functional significance of ATP7B variants is critical to formulating targeted and personalized therapies. Henceforth, we screened and sequenced 21 exons of ATP7B gene from 50 WD patients and 60 healthy subjects. We identified 28 variants comprising, seven novels in 20% alleles, while eight variations affecting 23% alleles were first time reported in Indian cohort. The c.813C>A, p.(Cys271*) (10%) was the most frequent mutation. Bioinformatics analysis revealed five of seven novel variants viz. c.1600C>A, p.(Pro534Thr); c.1616C>A, p.(Pro539His); c.1924G>T, p.(Asp642Tyr); c.2168G>C, p.(Arg723Thr); c.2174G>C, p.(Arg725Thr) resulted in protein misfolding. Sequence conservation analysis of ATP7B regions containing novel variants documented an evolutionarily conserved nature. Functional analysis of these novel variants in five different cell lines lacking inherent ATP7B expression demonstrated sensitivity to CuCl2 -treatment, experiencing augmented cellular copper retention and decreased copper excretion as well as ceruloplasmin secretion to that of wildtype-ATP7B expressing cells. Interestingly, pharmacological chaperone 4-phenylbutyrate, a clinically approved compound, partially restored protein function of ATP7B mutants. These findings might enable novel treatment strategies in WD by clinically enhancing the protein expression of mutant ATP7B with residual copper export activity.


Assuntos
ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/genética , Degeneração Hepatolenticular/genética , Mutação , Polimorfismo de Nucleotídeo Único , Adolescente , Adulto , Estudos de Casos e Controles , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Criança , Pré-Escolar , Estudos de Coortes , ATPases Transportadoras de Cobre/metabolismo , Feminino , Células HeLa , Degeneração Hepatolenticular/metabolismo , Humanos , Índia , Masculino , Mutação/efeitos dos fármacos , Fenilbutiratos/farmacologia , Dobramento de Proteína , Adulto Jovem
20.
Eur J Hum Genet ; 26(12): 1810-1818, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30087448

RESUMO

Approximately 2% of the human genome accounts for protein-coding genes, yet most known Mendelian disease-causing variants lie in exons or splice sites. Individuals who symptomatically present with monogenic disorders but do not possess function-altering variants in the protein-coding regions of causative genes may harbor variants in the surrounding gene regulatory domains. We present such a case: a male of Afghani descent was clinically diagnosed with Wilson Disease-a disorder of systemic copper buildup-but was found to have no function-altering coding variants in ATP7B (ENST00000242839.4), the typically causative gene. Our analysis revealed the homozygous variant chr13:g.52,586,149T>C (NC_000013.10, hg19) 676 bp into the ATP7B promoter, which disrupts a metal regulatory transcription factor 1 (MTF1) binding site and diminishes expression of ATP7B in response to copper intake, likely resulting in Wilson Disease. Our approach to identify the causative variant can be generalized to systematically discover function-altering non-coding variants underlying disease and motivates evaluation of gene regulatory variants.


Assuntos
ATPases Transportadoras de Cobre/genética , Degeneração Hepatolenticular/genética , Sítios de Ligação , Pré-Escolar , ATPases Transportadoras de Cobre/química , ATPases Transportadoras de Cobre/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Hep G2 , Degeneração Hepatolenticular/patologia , Homozigoto , Humanos , Masculino , Mutação , Regiões Promotoras Genéticas , Ligação Proteica , Fatores de Transcrição/metabolismo , Fator MTF-1 de Transcrição
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA