Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 190
Filter
1.
Nat Commun ; 15(1): 3834, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38714741

ABSTRACT

Sleep disorders increase the risk and mortality of heart disease, but the brain-heart interaction has not yet been fully elucidated. Cuproptosis is a copper-dependent type of cell death activated by the excessive accumulation of intracellular copper. Here, we showed that 16 weeks of sleep fragmentation (SF) resulted in elevated copper levels in the male mouse heart and exacerbated myocardial ischemia-reperfusion injury with increased myocardial cuproptosis and apoptosis. Mechanistically, we found that SF promotes sympathetic overactivity, increases the germination of myocardial sympathetic nerve terminals, and increases the level of norepinephrine in cardiac tissue, thereby inhibits VPS35 expression and leads to impaired ATP7A related copper transport and copper overload in cardiomyocytes. Copper overload further leads to exacerbated cuproptosis and apoptosis, and these effects can be rescued by excision of the sympathetic nerve or administration of copper chelating agent. Our study elucidates one of the molecular mechanisms by which sleep disorders aggravate myocardial injury and suggests possible targets for intervention.


Subject(s)
Apoptosis , Copper , Mice, Inbred C57BL , Myocardial Reperfusion Injury , Myocytes, Cardiac , Sleep Deprivation , Animals , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Male , Copper/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Mice , Sleep Deprivation/physiopathology , Sleep Deprivation/metabolism , Sleep Deprivation/complications , Copper-Transporting ATPases/metabolism , Copper-Transporting ATPases/genetics , Norepinephrine/metabolism , Norepinephrine/pharmacology , Myocardium/metabolism , Myocardium/pathology , Sympathetic Nervous System/metabolism , Disease Models, Animal
2.
Nat Commun ; 15(1): 2690, 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38538615

ABSTRACT

Copper transporting P-type (P1B-1-) ATPases are essential for cellular homeostasis. Nonetheless, the E1-E1P-E2P-E2 states mechanism of P1B-1-ATPases remains poorly understood. In particular, the role of the intrinsic metal binding domains (MBDs) is enigmatic. Here, four cryo-EM structures and molecular dynamics simulations of a P1B-1-ATPase are combined to reveal that in many eukaryotes the MBD immediately prior to the ATPase core, MBD-1, serves a structural role, remodeling the ion-uptake region. In contrast, the MBD prior to MBD-1, MBD-2, likely assists in copper delivery to the ATPase core. Invariant Tyr, Asn and Ser residues in the transmembrane domain assist in positioning sulfur-providing copper-binding amino acids, allowing for copper uptake, binding and release. As such, our findings unify previously conflicting data on the transport and regulation of P1B-1-ATPases. The results are critical for a fundamental understanding of cellular copper homeostasis and for comprehension of the molecular bases of P1B-1-disorders and ongoing clinical trials.


Subject(s)
Cation Transport Proteins , Copper , Copper/chemistry , Copper-Transporting ATPases/metabolism , Amino Acid Sequence , Cation Transport Proteins/metabolism , Protein Domains , Binding Sites
3.
Biochim Biophys Acta Biomembr ; 1866(4): 184306, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38408697

ABSTRACT

Human copper transporters ATP7B and ATP7A deliver copper to biosynthetic pathways and maintain copper homeostasis in the cell. These enzymes combine several challenges for structural biology because they are large low abundance membrane proteins with many highly mobile domains and long disordered loops. No method has yet succeeded in solving the structure of the complete fully functional protein. Still, X-ray crystallography, Cryo-EM and NMR helped to piece together a structure based model of the enzyme activity and regulation by copper. We review the structures of ATP7B and ATP7A with an emphasis on the mechanistic insights into the unique aspects of the transport function and regulation of the human copper ATPases that have emerged from more than twenty years of research.


Subject(s)
Cation Transport Proteins , Copper , Humans , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Copper/chemistry , Cation Transport Proteins/metabolism , Homeostasis
4.
J Biol Chem ; 300(3): 105700, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38307383

ABSTRACT

Selective retrograde transport from endosomes back to the trans-Golgi network (TGN) is important for maintaining protein homeostasis, recycling receptors, and returning molecules that were transported to the wrong compartments. Two important transmembrane proteins directed to this pathway are the Cation-Independent Mannose-6-phosphate receptor (CI-MPR) and the ATP7B copper transporter. Among CI-MPR functions is the delivery of acid hydrolases to lysosomes, while ATP7B facilitates the transport of cytosolic copper ions into organelles or the extracellular space. Precise subcellular localization of CI-MPR and ATP7B is essential for the proper functioning of these proteins. This study shows that both CI-MPR and ATP7B interact with a variant of the clathrin adaptor 1 (AP-1) complex that contains a specific isoform of the γ-adaptin subunit called γ2. Through synchronized anterograde trafficking and cell-surface uptake assays, we demonstrated that AP-1γ2 is dispensable for ATP7B and CI-MPR exit from the TGN while being critically required for ATP7B and CI-MPR retrieval from endosomes to the TGN. Moreover, AP-1γ2 depletion leads to the retention of endocytosed CI-MPR in endosomes enriched in retromer complex subunits. These data underscore the importance of AP-1γ2 as a key component in the sorting and trafficking machinery of CI-MPR and ATP7B, highlighting its essential role in the transport of proteins from endosomes.


Subject(s)
Adaptor Protein Complex 1 , Copper-Transporting ATPases , Endosomes , Protein Transport , Receptor, IGF Type 2 , trans-Golgi Network , Humans , Endosomes/metabolism , HeLa Cells , Protein Transport/genetics , Receptor, IGF Type 2/genetics , Receptor, IGF Type 2/metabolism , trans-Golgi Network/genetics , trans-Golgi Network/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Adaptor Protein Complex 1/genetics , Adaptor Protein Complex 1/metabolism , Adaptor Protein Complex gamma Subunits/metabolism
5.
Mol Metab ; 80: 101872, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38185452

ABSTRACT

OBJECTIVES: Adipocyte fate determination is tightly regulated by extrinsic signaling pathways and intrinsic metabolic and morphologic changes that maintain adipose tissue function. Copper (Cu) homeostasis is required for the normal metabolism of mature adipocytes, whereas the role of Cu in adipogenesis is unclear. METHODS: To determine the role of Cu is adipocytes differentiation, we used 3T3-L1 adipocytes, immunocytochemistry, X-ray fluorescence, mass-spectrometry, pharmacological treatments, and manipulations of copper levels. RESULTS: In differentiating 3T3-L1 cells, adipogenic stimuli trigger the upregulation and trafficking of the Cu transporter Atp7a, thus causing Cu redistribution from the cytosol to vesicles. Disrupting Cu homeostasis by the deletion of Atp7a results in Cu elevation and inhibition of adipogenesis. The upregulation of C/EBPß, an initial step of adipogenesis, is not affected in Atp7a-/- cells, whereas the subsequent upregulation of PPARγ is inhibited. Comparison of changes in the Atp7a-/- and wild type cells proteomes during early adipogenesis revealed stabilization of ß-catenin, a negative regulator of adipogenesis. Cu chelation, or overexpression of the Cu transporter ATP7B in Atp7a-/- cells, restored ß-catenin down-regulation and intracellular targeting. CONCLUSIONS: Cu buffering during early adipogenesis contributes to termination of ß-catenin signaling. Abnormal upregulation of ß-catenin was also observed in vivo in the livers of Atp7b-/- mice, which accumulate Cu, suggesting a tissue-independent crosstalk between Cu homeostasis and the Wnt/ß-catenin pathway. These results point to a new regulatory role of Cu in adipocytes and contribute to better understanding of human disorders of Cu misbalance.


Subject(s)
Adipogenesis , Wnt Signaling Pathway , beta Catenin , Animals , Humans , Mice , Adipose Tissue/metabolism , beta Catenin/metabolism , Copper/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Peptide Fragments/metabolism
6.
Traffic ; 25(1): e12920, 2024 01.
Article in English | MEDLINE | ID: mdl-37886910

ABSTRACT

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


Subject(s)
Hepatolenticular Degeneration , Animals , Humans , Hepatolenticular Degeneration/genetics , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Copper/toxicity , Copper/metabolism , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Hepatocytes/metabolism
7.
J Cell Sci ; 137(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38032054

ABSTRACT

The homologous P-type copper-ATPases (Cu-ATPases) ATP7A and ATP7B are the key regulators of copper homeostasis in mammalian cells. In polarized epithelia, upon copper treatment, ATP7A and ATP7B traffic from the trans-Golgi network (TGN) to basolateral and apical membranes, respectively. We characterized the sorting pathways of Cu-ATPases between TGN and the plasma membrane and identified the machinery involved. ATP7A and ATP7B reside on distinct domains of TGN in limiting copper conditions, and in high copper, ATP7A traffics to basolateral membrane, whereas ATP7B traverses common recycling, apical sorting and apical recycling endosomes en route to apical membrane. Mass spectrometry identified regulatory partners of ATP7A and ATP7B that include the adaptor protein-1 complex. Upon knocking out pan-AP-1, sorting of both Cu-ATPases is disrupted. ATP7A loses its trafficking polarity and localizes on both apical and basolateral surfaces in high copper. By contrast, ATP7B loses TGN retention but retained its trafficking polarity to the apical domain, which became copper independent. Using isoform-specific knockouts, we found that the AP-1A complex provides directionality and TGN retention for both Cu-ATPases, whereas the AP-1B complex governs copper-independent trafficking of ATP7B solely. Trafficking phenotypes of Wilson disease-causing ATP7B mutants that disrupts putative ATP7B-AP1 interaction further substantiates the role of AP-1 in apical sorting of ATP7B.


Subject(s)
Copper , Hepatolenticular Degeneration , Animals , Humans , Adenosine Triphosphatases/metabolism , Cell Membrane/metabolism , Copper/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Hepatolenticular Degeneration/genetics , Mammals/metabolism , Peptide Fragments/metabolism , Transcription Factor AP-1/metabolism
8.
Front Cell Infect Microbiol ; 13: 1267931, 2023.
Article in English | MEDLINE | ID: mdl-38106478

ABSTRACT

Copper, a vital element in various physiological processes, is transported from the gastrointestinal tract to tissues and cells through diverse copper transporters. Among these transporters, ATP7A and ATP7B play significant roles in regulating systemic copper metabolism and exhibit precise regulation in their intracellular trafficking. These transporters undergo dynamic shuttling between the trans-Golgi network (TGN) and the plasma membrane via the endocytic recycling mechanism, which involves the retromer and other associated factors. Interestingly, the antimicrobial attribute of copper implies a potential connection between microbial infection and copper metabolism. Several microbes, including Salmonella enterica, Cryptococcus, Influenza A virus (IAV) and Zika virus (ZIKV) have been observed to impact the regulatory mechanisms of ATP7A/B, either directly or indirectly, as a means of survival. This review summarizes the key features and trafficking mechanisms of the copper transporters ATP7A/B, and examines the intricate interplay between microbes and copper metabolism. Ultimately, it highlights how microbes can perturb copper homeostasis through interactions with host factors, offering valuable insights into the mechanistic aspects of host-microbe interactions.


Subject(s)
Cation Transport Proteins , Zika Virus Infection , Zika Virus , Humans , Copper/metabolism , Adenosine Triphosphatases , Cation Transport Proteins/metabolism , Copper Transport Proteins , Copper-Transporting ATPases/metabolism , Peptide Fragments/metabolism
9.
Traffic ; 24(12): 587-609, 2023 12.
Article in English | MEDLINE | ID: mdl-37846526

ABSTRACT

In hepatocytes, the Wilson disease protein ATP7B resides on the trans-Golgi network (TGN) and traffics to peripheral lysosomes to export excess intracellular copper through lysosomal exocytosis. We found that in basal copper or even upon copper chelation, a significant amount of ATP7B persists in the endolysosomal compartment of hepatocytes but not in non-hepatic cells. These ATP7B-harbouring lysosomes lie in close proximity of ~10 nm to the TGN. ATP7B constitutively distributes itself between the sub-domain of the TGN with a lower pH and the TGN-proximal lysosomal compartments. The presence of ATP7B on TGN-lysosome colocalising sites upon Golgi disruption suggested a possible exchange of ATP7B directly between the TGN and its proximal lysosomes. Manipulating lysosomal positioning significantly alters the localisation of ATP7B in the cell. Contrary to previous understanding, we found that upon copper chelation in a copper-replete hepatocyte, ATP7B is not retrieved back to TGN from peripheral lysosomes; rather, ATP7B recycles to these TGN-proximal lysosomes to initiate the next cycle of copper transport. We report a hitherto unknown copper-independent lysosomal localisation of ATP7B and the importance of TGN-proximal lysosomes but not TGN as the terminal acceptor organelle of ATP7B in its retrograde pathway.


Subject(s)
Copper , Lysosomes , Copper/metabolism , Copper-Transporting ATPases/metabolism , Protein Transport , Lysosomes/metabolism , Exocytosis
10.
J Inorg Biochem ; 247: 112334, 2023 10.
Article in English | MEDLINE | ID: mdl-37499466

ABSTRACT

The deregulation of copper homoeostasis can promote various diseases such as Menkes disease or hypertrophic cardioencephalomyopathy. We have recently synthesized solid copper(II) complexes ([Cu(His)2Cl2] and [Cu(Ser)2]), stable in physiological media and with potential as therapeutic agents. This report describes: i) the biocompatibility of these complexes at concentrations up to 100 µM using a differentiated Caco-2 cells model; ii) their transport across the intestinal epithelium using a transepithelial resistance assay and monitoring the amount of copper complexes at the apical and basolateral sides of the cells. The results suggest that the flow occurs through paracellular routes. The intracellular copper retention was <2.7% with no significant differences in intracellular copper content between 6 h and 48 h, suggesting an early copper retention process. Furthermore, this is the first evidence that demonstrates [Cu(His)2Cl2] and [Cu(Ser)2] induce transcriptional downregulation of the four major copper transporters (CTR1, DMT1, ATP7A, ATP7B), and the upregulation of the metallothionein gene expression. A remarkable finding was the increase in cytochrome c oxidase activity observed after the treatment of differentiated Caco-2 cells with copper(II) complexes at concentrations of 50-100 µM. The understanding of the transport mechanisms of these copper(II) complexes across the intestinal epithelium and of their subsequent biological activities could contribute to the development of optimal pharmaceutical formulations for the therapy of copper deficiency-related diseases.


Subject(s)
Cation Transport Proteins , Copper , Humans , Copper/pharmacology , Caco-2 Cells , Rare Diseases/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Intestinal Mucosa/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism
11.
Metallomics ; 15(7)2023 07 10.
Article in English | MEDLINE | ID: mdl-37391383

ABSTRACT

Cu (Cu) is essential for several biochemical pathways due to its role as a catalytic cofactor or allosteric regulator of enzymes. Its import and distribution are tightly controlled by transporters and metallochaperones and Cu homeostasis is maintained by balancing Cu uptake and export. Genetic diseases are caused by impaired Cu transporters CTR1, ATP7A, or ATP7B but little is known about the regulatory mechanisms by which these proteins meet the fluctuating demands of Cu in specific tissues. Cu is required for differentiation of skeletal myoblasts to myotubes. Here, we demonstrate that ATP7A is needed for myotube formation and that its increased abundance during differentiation is mediated by stabilization of Atp7a mRNA via the 3' untranslated region. Increased ATP7A levels during differentiation resulted in increased Cu delivery to lysyl oxidase, a secreted cuproenzyme that needed for myotube formation. These studies identify a previously unknown role for Cu in regulating muscle differentiation and have broad implications for understanding Cu-dependent differentiation in other tissues.


Subject(s)
Muscle Fibers, Skeletal , RNA , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Cell Differentiation , RNA, Messenger/genetics , Muscle Fibers, Skeletal/metabolism , Muscle, Skeletal/metabolism , Copper/metabolism
12.
Metallomics ; 15(5)2023 05 02.
Article in English | MEDLINE | ID: mdl-37070960

ABSTRACT

As a copper (Cu) transport ATPase, ATP7B plays an important role in maintaining Cu homeostasis in the body and its dysfunction is associated with retinal disease. How ATP7B dysfunction and the subsequent Cu overload induce retinal damage, however, are unknown. Here, we show that atp7b-/- homozygous zebrafish larvae are insensitive to light stimulation, with a reduction in retinal cells but normal like morphological phenotypes. Additionally, a series of differentially expressed genes are unveiled in atp7b-/- mutated larvae, which enrich in photo-transduction, structural constituent of eye lens, sensory perception of light stimulus, oxidative phosphorylation, and ATPase activity. Moreover, we show the Cu accumulation in retinal cells in atp7b-/- mutated larvae, which results in endoplasmic reticulum (ER) stress and retinal cell apoptosis and subsequent retinal defects. The integral data in this study demonstrate that atp7b mutation leads to Cu accumulation in zebrafish retinal cells and the consequence ER stress and retinal cell death. These data may give some possible hints to explain retinal disease occurred in the Cu dysregulation syndromes Wilson's disease with ATP7B mutation.


Subject(s)
Cation Transport Proteins , Hepatolenticular Degeneration , Animals , Zebrafish/genetics , Zebrafish/metabolism , Cation Transport Proteins/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Hepatolenticular Degeneration/metabolism , Copper/metabolism , Mutation
13.
Cell Rep ; 42(5): 112417, 2023 05 30.
Article in English | MEDLINE | ID: mdl-37074913

ABSTRACT

The P-type ATPase ATP7B exports cytosolic copper and plays an essential role in the regulation of cellular copper homeostasis. Mutants of ATP7B cause Wilson disease (WD), an autosomal recessive disorder of copper metabolism. Here, we present cryoelectron microscopy (cryo-EM) structures of human ATP7B in the E1 state in the apo, the putative copper-bound, and the putative cisplatin-bound forms. In ATP7B, the N-terminal sixth metal-binding domain (MBD6) binds at the cytosolic copper entry site of the transmembrane domain (TMD), facilitating the delivery of copper from the MBD6 to the TMD. The sulfur-containing residues in the TMD of ATP7B mark the copper transport pathway. By comparing structures of the E1 state human ATP7B and E2-Pi state frog ATP7B, we propose the ATP-driving copper transport model of ATP7B. These structures not only advance our understanding of the mechanisms of ATP7B-mediated copper export but can also guide the development of therapeutics for the treatment of WD.


Subject(s)
Cation Transport Proteins , Hepatolenticular Degeneration , Humans , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper/metabolism , Copper Transport Proteins , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Cryoelectron Microscopy , Hepatolenticular Degeneration/metabolism
14.
Int Immunopharmacol ; 114: 109518, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36502594

ABSTRACT

BACKGROUND: ATP7A is an important copper transporter that regulates numerous cellular biological processes. However, the role of ATP7A in immunotherapy and targeted therapy, especially for hepatocellular carcinoma (HCC), remains unknown. METHODS: We analyzed ATP7A expression and its effect on digestive system tumor prognoses, assessed its expression in tissue microarrays from 319 HCC patients, and investigated the relationship between ATP7A expression and tumor immunity. Specifically, we evaluated the possible association between ATP7A and programmed death ligand 1 (PD-L1) expression in human HCC tissues. Finally, we analyzed the effect of ATP7A on sorafenib efficacy in HCC. RESULTS: ATP7A is generally highly expressed in digestive system tumors but related to poor prognosis only in HCC. ATP7A levels are positively associated with immune cell infiltration and immune checkpoint expression (especially PD-L1). HCC patients coexpressing APT7A and PD-L1 demonstrate poor prognoses. Moreover, HCC patients with high ATP7A levels were more sensitive to sorafenib and demonstrated higher survival rates after sorafenib treatment. CONCLUSIONS: This study provides insights into the correlation between ATP7A levels and tumor immune infiltration and immune checkpoint function in HCC, sheds light on the significance of ATP7A in cancer progression, and provides guidance for more effective and general therapeutic strategies.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Sorafenib/therapeutic use , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Copper Transport Proteins , Immunotherapy , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Peptide Fragments/metabolism
15.
Cells ; 11(21)2022 10 27.
Article in English | MEDLINE | ID: mdl-36359796

ABSTRACT

Copper is a transition metal essential for human life. Its homeostasis is regulated in the liver, which delivers copper to the whole body and excretes its excess outside the organism in the feces through the bile. These functions are regulated within hepatocytes, and the ATP7B copper transporter is central to making the switch between copper use and excretion. In Wilson disease, the gene coding for ATP7B is mutated, leading to copper overload, firstly, in the liver and the brain. To better understand the role of ATP7B in hepatocytes and to provide a smart tool for the development of novel therapies against Wilson disease, we used the CrispR/Cas9 tool to generate hepatocyte cell lines with the abolished expression of ATP7B. These cell lines revealed that ATP7B plays a major role at low copper concentrations starting in the micromolar range. Moreover, metal stress markers are induced at lower copper concentrations compared to parental cells, while redox stress remains not activated. As shown recently, the main drawback induced by copper exposure is protein unfolding that is drastically exacerbated in ATP7B-deficient cells. Our data enabled us to propose that the zinc finger domain of DNAJ-A1 would serve as a sensor of Cu stress. Therefore, these Wilson-like hepatocytes are of high interest to explore in more detail the role of ATP7B.


Subject(s)
Copper-Transporting ATPases , Copper , Hepatolenticular Degeneration , Humans , Cell Line , Copper/pharmacology , Copper/metabolism , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Hepatocytes/metabolism , Hepatolenticular Degeneration/genetics , Proteostasis Deficiencies/genetics , Proteostasis Deficiencies/metabolism
16.
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
17.
PLoS Comput Biol ; 18(9): e1010074, 2022 09.
Article in English | MEDLINE | ID: mdl-36070320

ABSTRACT

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.


Subject(s)
Copper-Transporting ATPases , Copper , Binding Sites , Copper-Transporting ATPases/chemistry , Copper-Transporting ATPases/metabolism , DNA-Binding Proteins/metabolism , Humans , Protein Domains
18.
Mol Cell ; 82(19): 3745-3749.e2, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36115342

ABSTRACT

The research article describing the discovery of ribosomal frameshifting in the bacterial CopA gene also reported the occurrence of frameshifting in the expression of the human ortholog ATP7B based on assays using dual luciferase reporters. An examination of the publicly available ribosome profiling data and the phylogenetic analysis of the proposed frameshifting site cast doubt on the validity of this claim and prompted us to reexamine the evidence. We observed similar apparent frameshifting efficiencies as the original authors using the same type of vector that synthesizes both luciferases as a single polyprotein. However, we noticed anomalously low absolute luciferase activities from the N-terminal reporter that suggests interference of reporter activity or levels by the ATP7B test cassette. When we tested the same proposed ATP7B frameshifting cassette in a more recently developed reporter system in which the reporters are released without being included in a polyprotein, no frameshifting was detected above background levels.


Subject(s)
Copper-Transporting ATPases/metabolism , Frameshifting, Ribosomal , Polyproteins , Frameshifting, Ribosomal/genetics , Humans , Luciferases/genetics , Nucleic Acid Conformation , Phylogeny , Polyproteins/genetics , Polyproteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Nat Commun ; 13(1): 5121, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36045128

ABSTRACT

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.


Subject(s)
Copper-Transporting ATPases , Copper , Adenosine Triphosphatases/chemistry , Adenosine Triphosphatases/metabolism , Biological Transport , Copper/chemistry , Copper/metabolism , Copper-Transporting ATPases/chemistry , Copper-Transporting ATPases/metabolism , Humans , Molecular Chaperones/metabolism
20.
Hum Cell ; 35(5): 1560-1576, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35907138

ABSTRACT

Circular RNAs (circRNAs) are a class of non-coding RNAs with a unique covalently closed loop structure. Recent studies indicate that dysregulation of circRNAs acts a role in cancer progression and chemotherapy resistance via interacting with RNA-binding proteins (RBPs). Herein, we identified circPBX3 to be involved in cisplatin resistance of ovarian cancer. In our study, two cisplatin-resistant ovarian cancer cell lines were established, and transcriptome RNA-sequencing was performed and circPBX3 was identified as significantly upregulated circRNA in these cells. The characteristics of circPBX3 and potential function of circPBX3 were evaluated. We found that circPBX3 was upregulated in ovarian tumor tissues and cisplatin-resistant ovarian cancer cells. CircPBX3 overexpression increased the half maximal inhibitory rate (IC50) of cisplatin, promoted colony formation and tumor xenografts growth, and reduced cell apoptosis of ovarian cancer cells under cisplatin treatment, while silencing circPBX3 showed opposite effects. Furthermore, circPBX3 could interact with the RNA-binding protein IGF2BP2, thus increased the stability of ATP7A mRNA and elevated ATP7A protein level. In addition, silencing ATP7A in ovarian cancer cells abrogated the effect of circPBX3 overexpression on cisplatin tolerance. Our findings provided a novel role of circPBX3 in cisplatin resistance of ovarian cancer.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Peptide Fragments/genetics , Peptide Fragments/metabolism , Peptide Fragments/pharmacology , RNA, Circular/genetics , RNA, Messenger , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...