Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Biosci Rep ; 44(6)2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38813981

ABSTRACT

The search for relevant molecular targets is one of the main tasks of modern tumor chemotherapy. To successfully achieve this, it is necessary to have the most complete understanding of the functioning of a transcriptional apparatus of the cell, particularly related to proliferation. The p53 protein plays an important role in regulating processes such as apoptosis, repair, and cell division, and the loss of its functionality often accompanies various types of tumors and contributes to the development of chemoresistance. Additionally, the proliferative activity of tumor cells is closely related to the metabolism of transition metals. For example, the metallochaperone Atox1 - a copper transporter protein - acts as a transcription activator for cyclin D1, promoting progression through the G1/S phase of the cell cycle. On the other hand, p53 suppresses cyclin D1 at the transcriptional level, thereby these proteins have divergent effects on cell cycle progression. However, the contribution of the interaction between these proteins to cell survival is poorly understood. This work demonstrates that not only exists a positive feedback loop between Atox1 and cyclin D1 but also that the activity of this loop depends on the status of the TP53 gene. Upon inactivation of TP53 in A549 and HepG2 cell lines, the expression of ATOX1 and CCND1 genes is enhanced, and their suppression in these cells leads to pronounced apoptosis. This fundamental observation may be useful in selecting more precise interventions for combined therapy of p53-negative tumors.


Subject(s)
Cell Survival , Copper Transport Proteins , Cyclin D1 , Tumor Suppressor Protein p53 , Humans , Cyclin D1/metabolism , Cyclin D1/genetics , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics , Cell Survival/drug effects , Hep G2 Cells , Copper Transport Proteins/metabolism , Copper Transport Proteins/genetics , A549 Cells , Gene Expression Regulation, Neoplastic , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Cell Proliferation , Apoptosis , Metallochaperones/metabolism , Metallochaperones/genetics , Cation Transport Proteins/metabolism , Cation Transport Proteins/genetics
2.
J Phys Chem B ; 128(22): 5336-5343, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38780400

ABSTRACT

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.


Subject(s)
Copper Transport Proteins , Copper-Transporting ATPases , Copper , Metallochaperones , Molecular Chaperones , Molecular Dynamics Simulation , Copper/chemistry , Copper/metabolism , Copper Transport Proteins/chemistry , Copper Transport Proteins/metabolism , Humans , Metallochaperones/chemistry , Metallochaperones/metabolism , Copper-Transporting ATPases/chemistry , Copper-Transporting ATPases/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Thermodynamics , Protein Multimerization
3.
Redox Biol ; 72: 103156, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38640584

ABSTRACT

Regulation of the oxidative stress response is crucial for the management and prognosis of traumatic brain injury (TBI). The copper chaperone Antioxidant 1 (Atox1) plays a crucial role in regulating intracellular copper ion balance and impacting the antioxidant capacity of mitochondria, as well as the oxidative stress state of cells. However, it remains unknown whether Atox1 is involved in modulating oxidative stress following TBI. Here, we investigated the regulatory role of Atox1 in oxidative stress on neurons both in vivo and in vitro, and elucidated the underlying mechanism through culturing hippocampal HT-22 cells with Atox1 mutation. The expression of Atox1 was significantly diminished following TBI, while mice with overexpressed Atox1 exhibited a more preserved hippocampal structure and reduced levels of oxidative stress post-TBI. Furthermore, the mice displayed notable impairments in learning and memory functions after TBI, which were ameliorated by the overexpression of Atox1. In the stretch injury model of HT-22 cells, overexpression of Atox1 mitigated oxidative stress by preserving the normal morphology and network connectivity of mitochondria, as well as facilitating the elimination of damaged mitochondria. Mechanistically, co-immunoprecipitation and mass spectrometry revealed the binding of Atox1 to DJ-1. Knockdown of DJ-1 in HT-22 cells significantly impaired the antioxidant capacity of Atox1. Mutations in the copper-binding motif or sequestration of free copper led to a substantial decrease in the interaction between Atox1 and DJ-1, with overexpression of DJ-1 failing to restore the antioxidant capacity of Atox1 mutants. The findings suggest that DJ-1 mediates the ability of Atox1 to withstand oxidative stress. And targeting Atox1 could be a potential therapeutic approach for addressing post-traumatic neurological dysfunction.


Subject(s)
Brain Injuries, Traumatic , Copper Transport Proteins , Hippocampus , Mitophagy , Neurons , Oxidative Stress , Protein Deglycase DJ-1 , Animals , Brain Injuries, Traumatic/metabolism , Brain Injuries, Traumatic/pathology , Brain Injuries, Traumatic/genetics , Mice , Hippocampus/metabolism , Hippocampus/pathology , Neurons/metabolism , Protein Deglycase DJ-1/metabolism , Protein Deglycase DJ-1/genetics , Copper Transport Proteins/metabolism , Copper Transport Proteins/genetics , Mitochondria/metabolism , Disease Models, Animal , Molecular Chaperones/metabolism , Molecular Chaperones/genetics , Male , Antioxidants/metabolism , Cell Line , Humans
4.
Int Arch Allergy Immunol ; 185(3): 201-211, 2024.
Article in English | MEDLINE | ID: mdl-38071964

ABSTRACT

INTRODUCTION: Long noncoding RNAs (lncRNAs) have been implicated in the pathogenesis of allergic rhinitis (AR). The current investigation is focused on elucidating the functional impact of a specific lncRNA, FGD5 antisense RNA 1 (FGD5-AS1), on the development and progression of AR through its interaction with miR-223-3p. METHODS: An experimental framework for AR was constructed in both cellular and animal models. Quantitative assessment of FGD5-AS1, miR-223-3p, and COX11 mRNA expression was conducted using real-time quantitative reverse transcription PCR. The expression of inflammatory factors, immunoglobulin E, LTC4, and ECP, was examined using ELISA. Apoptosis in human nasal epithelial cells was assessed by the flow cytometry method. The protein expression of COX11 was examined using Western blotting. Nasal mucosal function was further evaluated by hematoxylin and eosin staining. Furthermore, bioinformatics evaluations, dual-luciferase reporter assays, and a series of experimental procedures unveiled a putative competitive endogenous RNA regulatory mechanism. RESULTS: We found the expression of lncRNA FGD5-AS1 was decreased in AR. In vitro lncRNA FGD5-AS1 attenuated the production of inflammatory cytokines in nasal epithelial cells. Furthermore, elevated FGD5-AS1 expression significantly alleviated AR symptoms by reducing nasal epithelial apoptosis and inflammation. MiR-223-3p was identified as a direct target of FGD5-AS1. Moreover, miRNA-223-3p directly downregulated the expression of COX11 mRNA. Subsequent experiments confirmed that FGD5-AS1 regulated AR through the miR-223-3p/COX11 axis, thereby inhibiting inflammation. CONCLUSION: The FGD5-AS1/miR-223-3p/COX11 axis plays a pivotal role in the pathogenesis of AR, suggesting that FGD5-AS1 could serve as a potential diagnostic biomarker and therapeutic target for AR.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Rhinitis, Allergic , Animals , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Inflammation/genetics , Rhinitis, Allergic/genetics , RNA, Messenger , Cell Proliferation , Copper Transport Proteins/genetics , Copper Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Electron Transport Chain Complex Proteins/genetics , Electron Transport Chain Complex Proteins/metabolism , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism
5.
Int J Toxicol ; 43(2): 134-145, 2024.
Article in English | MEDLINE | ID: mdl-37859596

ABSTRACT

Antioxidant 1 copper chaperone (Atox1) may contribute to preventing DDP cochlear damage by regulating copper transport family and cell cycle proteins. A rat model of cochlear damage was developed by placing gelatin sponges treated with DDP in the cochlea. HEI-OC1 cells were treated with 133 µM DDP as a cell model. DDP-induced ototoxicity in rats was confirmed by immunofluorescence (IF) imaging. The damage of DDP to HEI-OC1 cells was assessed by using CCK-8, TUNEL, and flow cytometry. The relationship between Atox1, a member of the copper transport protein family, and the damage to in vivo/vitro models was explored by qRT-PCR, western blot, CCK-8, TUNEL, and flow cytometry. DDP had toxic and other side effects causing cochlear damage and promoted HEI-OC1 cell apoptosis and cell cycle arrest. The over-expression of Atox1 (oe-Atox1) was accomplished by transfecting lentiviral vectors into in vitro/vivo models. We found that oe-Atox1 increased the levels of Atox1, copper transporter 1 (CTR1), and SOD3 in HEI-OC1 cells and decreased the expression levels of ATPase copper transporting α (ATP7A) and ATPase copper transporting ß (ATP7B). In addition, the transfection of oe-Atox1 decreased cell apoptosis rate and the number of G2/M stage cells. Similarly, the expression of myosin VI and phalloidin of cochlea cells in vivo decreased. Atox1 ameliorated DDP-induced damage to HEI-OC1 cells or rats' cochlea by regulating the levels of members of the copper transport family.


Subject(s)
Cisplatin , Copper Transport Proteins , Molecular Chaperones , Animals , Rats , Cell Cycle , Cisplatin/toxicity , Cochlea , Copper/metabolism , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Sincalide/pharmacology , Copper Transport Proteins/metabolism
6.
Int J Mol Sci ; 24(23)2023 Nov 23.
Article in English | MEDLINE | ID: mdl-38068960

ABSTRACT

Genetic defects in the nuclear encoded subunits and assembly factors of cytochrome c oxidase (mitochondrial complex IV) are very rare and are associated with a wide variety of phenotypes. Biallelic pathogenic variants in the COX11 protein were previously identified in two unrelated children with infantile-onset mitochondrial encephalopathies. Through comprehensive clinical, genetic and functional analyses, here we report on a new patient harboring novel heterozygous variants in COX11, presenting with Leigh-like features, and provide additional experimental evidence for a direct correlation between COX11 protein expression and sensitivity to oxidative stress. To sort out the contribution of the single mutations to the phenotype, we employed a multi-faceted approach using Saccharomyces cerevisiae as a genetically manipulable system, and in silico structure-based analysis of human COX11. Our results reveal differential effects of the two novel COX11 mutations on yeast growth, respiration, and cellular redox status, as well as their potential impact on human protein stability and function. Strikingly, the functional deficits observed in patient fibroblasts are recapitulated in yeast models, validating the conservation of COX11's role in mitochondrial integrity across evolutionarily distant organisms. This study not only expands the mutational landscape of COX11-associated mitochondrial disorders but also underscores the continued translational relevance of yeast models in dissecting complex molecular pathways.


Subject(s)
Mitochondrial Diseases , Saccharomyces cerevisiae Proteins , Child , Humans , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/metabolism , Amino Acid Sequence , Membrane Proteins/metabolism , Electron Transport Complex IV/genetics , Electron Transport Complex IV/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Mutation , Mitochondrial Diseases/genetics , Fibroblasts/metabolism , Copper Transport Proteins/metabolism , Electron Transport Chain Complex Proteins/metabolism
7.
Int J Mol Sci ; 24(21)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37958561

ABSTRACT

The copper transporter (COPT/Ctr) gene family plays a critical part in maintaining the balance of the metal, and many diverse species depend on COPT to move copper (Cu) across the cell membrane. In Arabidopsis thaliana, Oryza sativa, Medicago sativa, Zea mays, Populus trichocarpa, Vitis vinifera, and Solanum lycopersicum, a genome-wide study of the COPT protein family was performed. To understand the major roles of the COPT gene family in Kandelia obovata (Ko), a genome-wide study identified four COPT genes in the Kandelia obovata genome for the first time. The domain and 3D structural variation, phylogenetic tree, chromosomal distributions, gene structure, motif analysis, subcellular localization, cis-regulatory elements, synteny and duplication analysis, and expression profiles in leaves and Cu were all investigated in this research. Structural and sequence investigations show that most KoCOPTs have three transmembrane domains (TMDs). According to phylogenetic research, these KoCOPTs might be divided into two subgroups, just like Populus trichocarpa. KoCOPT gene segmental duplications and positive selection pressure were discovered by universal analysis. According to gene structure and motif analysis, most KoCOPT genes showed consistent exon-intron and motif organization within the same group. In addition, we found five hormones and four stress- and seven light-responsive cis-elements in the KoCOPTs promoters. The expression studies revealed that all four genes changed their expression levels in response to copper (CuCl2) treatments. In summary, our study offers a thorough overview of the Kandelia obovata COPT gene family's expression pattern and functional diversity, making it easier to characterize each KoCOPT gene's function in the future.


Subject(s)
Genes, Plant , Rhizophoraceae , Copper/metabolism , Copper Transport Proteins/metabolism , Gene Expression Profiling , Gene Expression Regulation, Plant , Genome-Wide Association Study , Multigene Family , Phylogeny , Plant Proteins/metabolism , Rhizophoraceae/genetics
8.
Nat Metab ; 5(11): 1931-1952, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37813994

ABSTRACT

Reversible acetylation of mitochondrial proteins is a regulatory mechanism central to adaptive metabolic responses. Yet, how such functionally relevant protein acetylation is achieved remains unexplored. Here we reveal an unprecedented role of the MYST family lysine acetyltransferase MOF in energy metabolism via mitochondrial protein acetylation. Loss of MOF-KANSL complex members leads to mitochondrial defects including fragmentation, reduced cristae density and impaired mitochondrial electron transport chain complex IV integrity in primary mouse embryonic fibroblasts. We demonstrate COX17, a complex IV assembly factor, as a bona fide acetylation target of MOF. Loss of COX17 or expression of its non-acetylatable mutant phenocopies the mitochondrial defects observed upon MOF depletion. The acetylation-mimetic COX17 rescues these defects and maintains complex IV activity even in the absence of MOF, suggesting an activatory role of mitochondrial electron transport chain protein acetylation. Fibroblasts from patients with MOF syndrome who have intellectual disability also revealed respiratory defects that could be restored by alternative oxidase, acetylation-mimetic COX17 or mitochondrially targeted MOF. Overall, our findings highlight the critical role of MOF-KANSL complex in mitochondrial physiology and provide new insights into MOF syndrome.


Subject(s)
Fibroblasts , Mitochondria , Humans , Animals , Mice , Acetylation , Fibroblasts/metabolism , Mitochondria/metabolism , Energy Metabolism , Electron Transport Complex IV/metabolism , Copper Transport Proteins/metabolism
9.
Mol Pharm ; 20(8): 4138-4152, 2023 08 07.
Article in English | MEDLINE | ID: mdl-37358225

ABSTRACT

Lipid nanoparticle (LNP) delivery systems are widely used in the delivery of small-molecule drugs and nucleic acids. In this study, we prepared LNP-miR-155 by lipid nanomaterial technology and investigated the effects of LNP-miR-155 on ß-catenin/transcription factor 4 (TCF4)/solute carrier family 31 member 1/copper transporter 1 (SLC31A1/CTR1) signaling and copper transport in colorectal cancer. For this, we used an LNP-miR-155 cy5 inhibitor and LNP-miR-155 cy5 mimics for the transfection of HT-29/SW480 cells. The transfection efficiency and uptake efficiency were detected by immunofluorescence. Relevant cell assays confirmed that the LNP-miR-155 cy5 inhibitor mediates the regulation of copper transport through the ß-catenin/TCF4/SLC31A1 axis. The LNP-miR-155 cy5 inhibitor reduced cell proliferation, migration, and colony formation and promoted cell apoptosis. We also confirmed that miR-155 downregulates HMG box-containing protein 1 (HBP1) and adenomatous polyposis coli (APC) in cells and activates the function of ß-catenin/TCF4 signaling. In addition, we found that the copper transporter, SLC31A1, is highly expressed in colorectal cancer cells. Furthermore, we also found that the complex ß-catenin/TCF4 promotes the transcription of SLC31A1 by binding to its promoter region, which sustains the transport of copper from the extracellular region to the intracellular region and increases the activities of Cu2+-ATPase and superoxide dismutase (SOD). In summary, the LNP-miR-155 cy5 inhibitor regulates ß-catenin/TCF4 by downregulating SLC31A1-mediated copper transport and intracellular copper homeostasis.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , beta Catenin/metabolism , Transcription Factor 4/metabolism , Copper Transport Proteins/metabolism , Copper/pharmacology , Copper/metabolism , Colorectal Neoplasms/genetics , MicroRNAs/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Cell Line, Tumor , Copper Transporter 1/metabolism , High Mobility Group Proteins/metabolism , Repressor Proteins/metabolism
10.
Int J Biol Macromol ; 241: 124404, 2023 Jun 30.
Article in English | MEDLINE | ID: mdl-37054854

ABSTRACT

Copper acquisition and subsequent delivery to target proteins are essential for many biological processes. However, the cellular levels of this trace element must be controlled because of its potential toxicity. The COPT1 protein rich in potential metal-binding amino acids functions in high affinity copper uptake at the plasma membrane of Arabidopsis cells. The functional role of these putative metal-binding residues is largely unknown. Through truncations and site-directed mutagenesis, we identified His43, a single residue within the extracellular N-terminal domain as absolutely critical for copper uptake of COPT1. Substitution of this residue with leucine, methionine or cysteine almost inactivated transport function of COPT1, implying that His43 fails to serves as a copper ligand in the regulation of COPT1 activity. Deletion of all extracellular N-terminal metal-binding residues completely blocked copper-stimulated degradation but did not alter the subcellular distribution and multimerization of COPT1. Although mutation of His43 to alanine and serine retained the transporter activity in yeast cells, the mutant protein was unstable and degraded in the proteasome in Arabidopsis cells. Our results demonstrate a pivotal role for the extracellular residue His43 in high affinity copper transport activity, and suggest common molecular mechanisms for regulating both metal transport and protein stability of COPT1.


Subject(s)
Arabidopsis Proteins , Arabidopsis , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Histidine/genetics , Histidine/metabolism , Membrane Transport Proteins/genetics , Copper Transport Proteins/metabolism , Copper/chemistry , Copper Transporter 1/metabolism , Biological Transport , Protein Stability
11.
Proc Natl Acad Sci U S A ; 119(37): e2206905119, 2022 09 13.
Article in English | MEDLINE | ID: mdl-36067318

ABSTRACT

The protein mediator of ERBB2-driven cell motility 1 (Memo1) is connected to many signaling pathways that play key roles in cancer. Memo1 was recently postulated to bind copper (Cu) ions and thereby promote the generation of reactive oxygen species (ROS) in cancer cells. Since the concentration of Cu as well as ROS are increased in cancer cells, both can be toxic if not well regulated. Here, we investigated the Cu-binding capacity of Memo1 using an array of biophysical methods at reducing as well as oxidizing conditions in vitro. We find that Memo1 coordinates two reduced Cu (Cu(I)) ions per protein, and, by doing so, the metal ions are shielded from ROS generation. In support of biological relevance, we show that the cytoplasmic Cu chaperone Atox1, which delivers Cu(I) in the secretory pathway, can interact with and exchange Cu(I) with Memo1 in vitro and that the two proteins exhibit spatial proximity in breast cancer cells. Thus, Memo1 appears to act as a Cu(I) chelator (perhaps shuttling the metal ion to Atox1 and the secretory path) that protects cells from Cu-mediated toxicity, such as uncontrolled formation of ROS. This Memo1 functionality may be a safety mechanism to cope with the increased demand of Cu ions in cancer cells.


Subject(s)
Copper Transport Proteins , Copper , Intracellular Signaling Peptides and Proteins , Metallochaperones , Molecular Chaperones , Cell Line, Tumor , Copper/metabolism , Copper Transport Proteins/genetics , Copper Transport Proteins/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ions/metabolism , Metallochaperones/genetics , Metallochaperones/metabolism , Models, Molecular , Molecular Chaperones/genetics , Molecular Chaperones/metabolism , Oxidation-Reduction , Protein Binding , Reactive Oxygen Species/metabolism
12.
Hum Mutat ; 43(12): 1970-1978, 2022 12.
Article in English | MEDLINE | ID: mdl-36030551

ABSTRACT

Primary mitochondrial diseases are a group of genetically and clinically heterogeneous disorders resulting from oxidative phosphorylation (OXPHOS) defects. COX11 encodes a copper chaperone that participates in the assembly of complex IV and has not been previously linked to human disease. In a previous study, we identified that COX11 knockdown decreased cellular adenosine triphosphate (ATP) derived from respiration, and that ATP levels could be restored with coenzyme Q10 (CoQ10 ) supplementation. This finding is surprising since COX11 has no known role in CoQ10 biosynthesis. Here, we report a novel gene-disease association by identifying biallelic pathogenic variants in COX11 associated with infantile-onset mitochondrial encephalopathies in two unrelated families using trio genome and exome sequencing. Functional studies showed that mutant COX11 fibroblasts had decreased ATP levels which could be rescued by CoQ10 . These results not only suggest that COX11 variants cause defects in energy production but reveal a potential metabolic therapeutic strategy for patients with COX11 variants.


Subject(s)
Mitochondrial Diseases , Mitochondrial Encephalomyopathies , Humans , Mitochondrial Encephalomyopathies/genetics , Mitochondrial Encephalomyopathies/metabolism , Mitochondrial Diseases/genetics , Mitochondrial Diseases/metabolism , Mitochondria/metabolism , Adenosine Triphosphate/metabolism , Copper Transport Proteins/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Electron Transport Chain Complex Proteins/metabolism
13.
Essays Biochem ; 66(7): 977-986, 2022 12 16.
Article in English | MEDLINE | ID: mdl-35757906

ABSTRACT

Copper (Cu) ion dys-homeostasis and α-synclein amyloid deposits are two hallmarks of Parkinson's disease (PD). Here, I will discuss the connections between these features, with a major focus on the role of Cu in the α-synuclein (aS) amyloid formation process. The structurally disordered aS monomer can bind to both redox states of Cu (i.e., oxidized Cu(II) and reduced Cu(I)) with high affinity in vitro. Notably, the presence of Cu(II) (in absence of aS N-terminal acetylation) and Cu(I) (when in complex with the copper chaperone Atox1) modulate aS assembly into ß-structured amyloids in opposite directions in vitro. Albeit the link to biological relevance is not fully unraveled, existing observations clearly emphasize the need for more knowledge on this interplay and its consequences to eventually combat destructive reactions that promote PD.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/metabolism , Copper/metabolism , Binding Sites , alpha-Synuclein/metabolism , Amyloid/metabolism , Ions/metabolism , Copper Transport Proteins/metabolism , Molecular Chaperones/metabolism
14.
Biophys J ; 121(7): 1194-1204, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35202609

ABSTRACT

Abnormal cellular copper levels have been clearly implicated in genetic diseases, cancer, and neurodegeneration. Ctr1, a high-affinity copper transporter, is a homotrimeric integral membrane protein that provides the main route for cellular copper uptake. Together with a sophisticated copper transport system, Ctr1 regulates Cu(I) metabolism in eukaryotes. Despite its pivotal role in normal cell function, the molecular mechanism of copper uptake and transport via Ctr1 remains elusive. In this study, electron paramagnetic resonance (EPR), UV-visible spectroscopy, and all-atom simulations were employed to explore Cu(I) binding to full-length human Ctr1 (hCtr1), thereby elucidating how metal binding at multiple distinct sites affects the hCtr1 conformational dynamics. We demonstrate that each hCtr1 monomer binds up to five Cu(I) ions and that progressive Cu(I) binding triggers a marked structural rearrangement in the hCtr1 C-terminal region. The observed Cu(I)-induced conformational remodeling suggests that the C-terminal region may play a dual role, serving both as a channel gate and as a shuttle mediating the delivery of copper ions from the extracellular hCtr1 selectivity filter to intracellular metallochaperones. Our findings thus contribute to a more complete understanding of the mechanism of hCtr1-mediated Cu(I) uptake and provide a conceptual basis for developing mechanism-based therapeutics for treating pathological conditions linked to de-regulated copper metabolism.


Subject(s)
Cation Transport Proteins , Copper Transport Proteins , Copper Transporter 1 , Copper , Copper/chemistry , Copper/metabolism , Copper Transport Proteins/chemistry , Copper Transport Proteins/metabolism , Copper Transporter 1/chemistry , Copper Transporter 1/metabolism , Humans , Ions/chemistry , Ions/metabolism
15.
Nat Commun ; 12(1): 7311, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34911956

ABSTRACT

Copper serves as a co-factor for a host of metalloenzymes that contribute to malignant progression. The orally bioavailable copper chelating agent tetrathiomolybdate (TM) has been associated with a significant survival benefit in high-risk triple negative breast cancer (TNBC) patients. Despite these promising data, the mechanisms by which copper depletion impacts metastasis are poorly understood and this remains a major barrier to advancing TM to a randomized phase II trial. Here, using two independent TNBC models, we report a discrete subpopulation of highly metastatic SOX2/OCT4+ cells within primary tumors that exhibit elevated intracellular copper levels and a marked sensitivity to TM. Global proteomic and metabolomic profiling identifies TM-mediated inactivation of Complex IV as the primary metabolic defect in the SOX2/OCT4+ cell population. We also identify AMPK/mTORC1 energy sensor as an important downstream pathway and show that AMPK inhibition rescues TM-mediated loss of invasion. Furthermore, loss of the mitochondria-specific copper chaperone, COX17, restricts copper deficiency to mitochondria and phenocopies TM-mediated alterations. These findings identify a copper-metabolism-metastasis axis with potential to enrich patient populations in next-generation therapeutic trials.


Subject(s)
Copper/metabolism , Mitochondria/metabolism , Triple Negative Breast Neoplasms/metabolism , Animals , Cell Line, Tumor , Copper Transport Proteins/genetics , Copper Transport Proteins/metabolism , Female , Humans , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Neoplasm Metastasis , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Oxidative Phosphorylation , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
16.
Open Biol ; 11(12): 210128, 2021 12.
Article in English | MEDLINE | ID: mdl-34847776

ABSTRACT

Imbalanced copper homeostasis and perturbation of membrane trafficking are two common symptoms that have been associated with the pathogenesis of neurodegenerative and neurodevelopmental diseases. Accumulating evidence from biophysical, cellular and in vivo studies suggest that membrane trafficking orchestrates both copper homeostasis and neural functions-however, a systematic review of how copper homeostasis and membrane trafficking interplays in neurons remains lacking. Here, we summarize current knowledge of the general trafficking itineraries for copper transporters and highlight several critical membrane trafficking regulators in maintaining copper homeostasis. We discuss how membrane trafficking regulators may alter copper transporter distribution in different membrane compartments to regulate intracellular copper homeostasis. Using Parkinson's disease and MEDNIK as examples, we further elaborate how misregulated trafficking regulators may interplay parallelly or synergistically with copper dyshomeostasis in devastating pathogenesis in neurodegenerative diseases. Finally, we explore multiple unsolved questions and highlight the existing challenges to understand how copper homeostasis is modulated through membrane trafficking.


Subject(s)
Copper Transport Proteins/metabolism , Copper/metabolism , Nervous System Diseases/metabolism , Nervous System/metabolism , Animals , Gene Expression Regulation , Homeostasis , Humans , Parkinson Disease/metabolism , Signal Transduction
17.
Int J Biol Macromol ; 192: 600-610, 2021 Dec 01.
Article in English | MEDLINE | ID: mdl-34655579

ABSTRACT

Copper (Cu) plays a key role as cofactor in the plant proteins participating in essential cellular processes, such as electron transport and free radical scavenging. Despite high-affinity Cu transporters (COPTs) being key participants in Cu homeostasis maintenance, very little is known about COPTs in tomato (Solanum lycopersicum) even though it is the most consumed fruit worldwide and this crop is susceptible to suboptimal Cu conditions. In this study, a six-member family of COPT (SlCOPT1-6) was identified and characterized. SlCOPTs have a conserved architecture consisting of three transmembrane domains and ß-strains. However, the presence of essential methionine residues, a methionine-enriched amino-terminal region, an Mx3Mx12Gx3G Cu-binding motif and a cysteine rich carboxy-terminal region, all required for their functionality, is more variable among members. Accordingly, functional complementation assays in yeast indicate that SlCOPT1 and SlCOPT2 are able to transport Cu inside the cell, while SlCOPT3 and SlCOPT5 are only partially functional. In addition, protein interaction network analyses reveal the connection between SlCOPTs and Cu PIB-type ATPases, other metal transporters, and proteins related to the peroxisome. Gene expression analyses uncover organ-dependency, fruit vasculature tissue specialization and ripening-dependent gene expression profiles, as well as different response to Cu deficiency or toxicity in an organ-dependent manner.


Subject(s)
Copper Transport Proteins/chemistry , Copper Transport Proteins/metabolism , Solanum lycopersicum/metabolism , Amino Acid Sequence , Conserved Sequence , Copper/chemistry , Copper/metabolism , Copper Transport Proteins/genetics , Gene Expression , Solanum lycopersicum/chemistry , Solanum lycopersicum/genetics , Molecular Conformation , Multigene Family , Phylogeny , Plant Proteins/chemistry , Plant Proteins/metabolism , Structure-Activity Relationship
18.
FEMS Microbiol Lett ; 368(19)2021 10 26.
Article in English | MEDLINE | ID: mdl-34665227

ABSTRACT

Nitrogen limitation was previously shown to be an important regulator of several genes associated with virulence in Cryptococcus neoformans. Among the most highly expressed genes under low-nitrogen conditions were CTR4 and CGP1, encoding a copper transporter and a microtubule-associated protein, respectively. However, the functional association of these genes with nitrogen limitation-a nutritional stress experienced in both environment and host-remains to be determined. Moreover, whether increased CTR4 and CGP1 expression is linked to the enhanced cryptococcal drug tolerance previously observed in low-nitrogen conditions is yet to be elucidated. Therefore, the present study explored the role of Cgp1 and Ctr4 in C. neoformans nitrogen stress adaptation and antifungal susceptibility. Our results showed that these genes play a role in the growth of C. neoformans in nitrogen-limited media, nitrogen source assimilation and growth on nitrogen-poor woody debris. Furthermore, we demonstrate that both Ctr4 and Cgp1 contribute to oxidative stress and antifungal susceptibility, with a ctr4∆ mutant being more susceptible to fluconazole and a cgp1∆ mutant being more susceptible to fluconazole and amphotericin B. Overall, our findings improve our understanding of the role of Ctr4 and Cgp1 in cryptococcal drug tolerance and adaptation to nitrogen availability.


Subject(s)
Copper Transport Proteins , Cryptococcus neoformans , Fungal Proteins , Microtubule-Associated Proteins , Nitrogen , Antifungal Agents/pharmacology , Copper Transport Proteins/metabolism , Cryptococcus neoformans/drug effects , Cryptococcus neoformans/genetics , Cryptococcus neoformans/metabolism , Fluconazole/pharmacology , Fungal Proteins/metabolism , Humans , Microbial Sensitivity Tests , Microtubule-Associated Proteins/metabolism , Nitrogen/metabolism
19.
Angew Chem Int Ed Engl ; 60(48): 25346-25355, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34550632

ABSTRACT

Mammalian cell nuclei contain copper, and cancer cells are known to accumulate aberrantly high copper levels, yet the mechanisms underlying nuclear accumulation and copper's broader functional significance remain poorly understood. Here, by combining APEX2-based proximity labeling focused on the copper chaperone Atox1 with mass spectrometry we identified a previously unrecognized nuclear copper binding protein, Cysteine-rich protein 2 (CRIP2), that interacts with Atox1 in the nucleus. We show that Atox1 transfers copper to CRIP2, which induces a change in CRIP2's secondary structure that ultimately promotes its ubiquitin-mediated proteasomal degradation. Finally, we demonstrate that depletion of CRIP2-as well as copper-induced CRIP2 degradation-elevates ROS levels and activates autophagy in H1299 cells. Thus, our study establishes that CRIP2 as an autophagic suppressor protein and implicates CRIP2-mediated copper metabolism in the activation of autophagy in cancer cells.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Autophagy , Copper Transport Proteins/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Endonucleases/metabolism , LIM Domain Proteins/metabolism , Molecular Chaperones/metabolism , Multifunctional Enzymes/metabolism , Cell Line, Tumor , Copper/metabolism , Humans
20.
Neurosci Lett ; 763: 136173, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34400285

ABSTRACT

PURPOSE: Autosomal dominant acute necrotizing encephalopathy (ADANE) is caused by missense mutations in the gene encoding Ran-binding protein 2 (RANBP2), a nuclear pore protein regulating mitochondrial localization and function. Previous studies have found that RANBP2 binds to COX11 and suppresses its inhibitory activity over hexokinase1. To further elucidate mitochondrial dysfunction in ADANE, we analyzed the interaction between mutated RANBP2 and COX11. METHODS: We extracted cDNA from a patient and constructed pGEX wild-type or mutant-type vectors including RANBP2 c.1754C>T, the commonest variant in ADANE. We transformed E. coli competent cells with the vectors and had them express GST-RANBP2 recombinant protein, and conducted a pull-down assay of RANBP2 and COX11. RESULTS: The amount of COX11 bound to mutated RANBP2 was significantly smaller than that bound to the wild-type RANBP2. CONCLUSION: Mutated RANBP2 had an attenuated binding ability to COX11. Whether this change indeed decreases ATP production remains to be further explored.


Subject(s)
Copper Transport Proteins/metabolism , Electron Transport Chain Complex Proteins/metabolism , Leukoencephalitis, Acute Hemorrhagic/genetics , Mitochondrial Proteins/metabolism , Molecular Chaperones/metabolism , Nuclear Pore Complex Proteins/metabolism , Protein Binding/genetics , Case-Control Studies , Cells, Cultured , Child, Preschool , Copper Transport Proteins/isolation & purification , Electron Transport Chain Complex Proteins/isolation & purification , Energy Metabolism/genetics , Healthy Volunteers , Humans , Leukoencephalitis, Acute Hemorrhagic/blood , Leukoencephalitis, Acute Hemorrhagic/pathology , Lymphocytes , Male , Mitochondria/pathology , Mitochondrial Proteins/isolation & purification , Molecular Chaperones/genetics , Molecular Chaperones/isolation & purification , Mutation, Missense , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/isolation & purification , Pedigree , Primary Cell Culture , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Recombinant Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...