Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Heliyon ; 10(2): e24368, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38298614

ABSTRACT

Radiation therapy is a first-line treatment for head and neck cancer; however, it typically leads to hyposalivation stemming from fibrosis of the salivary gland. Current strategies to restore glandular function are dependent on the presence of residual functional salivary gland tissue, a condition commonly not met in patients with extensive fibrotic coverage of the salivary gland resulting from radiation therapy. Fibrosis is defined by the pathological accumulation of connective tissue (i.e., extracellular matrix) and excessive deposition of crosslinked (fibrillar) collagen that can impact a range of tissues and given that collagen crosslinking is necessary for fibrosis formation, inhibiting this process is a reasonable focus for developing anti-fibrotic therapies. Collagen crosslinking is catalyzed by the lysyl oxidase family of secreted copper-dependent metalloenzymes, and since that copper is an essential cofactor in all lysyl oxidase family members, we tested whether localized delivery of a copper chelator into the submandibular gland of irradiated mice could suppress collagen deposition and preserve the structure and function of this organ. Our results demonstrate that transdermal injection of tetrathiomolybdate into salivary glands significantly reduced the early deposition of fibrillar collagen in irradiated mice and preserved the integrity and function of submandibular gland epithelial tissue. Together, these studies identify copper metabolism as a novel therapeutic target to control radiation induced damage to the salivary gland and the current findings further indicate the therapeutic potential of repurposing clinically approved copper chelators as neoadjuvant treatments for radiation therapy.

2.
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
3.
Elife ; 122023 05 12.
Article in English | MEDLINE | ID: mdl-37171075

ABSTRACT

Mitochondria influence cellular function through both cell-autonomous and non-cell autonomous mechanisms, such as production of paracrine and endocrine factors. Here, we demonstrate that mitochondrial regulation of the secretome is more extensive than previously appreciated, as both genetic and pharmacological disruption of the electron transport chain caused upregulation of the Alzheimer's disease risk factor apolipoprotein E (APOE) and other secretome components. Indirect disruption of the electron transport chain by gene editing of SLC25A mitochondrial membrane transporters as well as direct genetic and pharmacological disruption of either complexes I, III, or the copper-containing complex IV of the electron transport chain elicited upregulation of APOE transcript, protein, and secretion, up to 49-fold. These APOE phenotypes were robustly expressed in diverse cell types and iPSC-derived human astrocytes as part of an inflammatory gene expression program. Moreover, age- and genotype-dependent decline in brain levels of respiratory complex I preceded an increase in APOE in the 5xFAD mouse model. We propose that mitochondria act as novel upstream regulators of APOE-dependent cellular processes in health and disease.


Subject(s)
Apolipoprotein E4 , Mitochondria , Animals , Humans , Mice , Apolipoprotein E4/genetics , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Astrocytes/metabolism , Genotype , Mitochondria/metabolism , Mitochondria/pathology
4.
Commun Biol ; 6(1): 403, 2023 04 18.
Article in English | MEDLINE | ID: mdl-37072620

ABSTRACT

Tyrosinase (TYR) and tyrosinase-related proteins 1 and 2 (TYRP1 and TYRP2) are essential for pigmentation. They are generally classified as type-3 copper proteins, with binuclear copper active sites. Although there is experimental evidence for a copper cofactor in TYR, delivered via the copper transporter, ATP7A, the presence of copper in TYRP1 and TYRP2 has not been demonstrated. Here, we report that the expression and function of TYRP1 requires zinc, mediated by ZNT5-ZNT6 heterodimers (ZNT5-6) or ZNT7-ZNT7 homodimers (ZNT7). Loss of ZNT5-6 and ZNT7 function results in hypopigmentation in medaka fish and human melanoma cells, and is accompanied by immature melanosomes and reduced melanin content, as observed in TYRP1 dysfunction. The requirement of ZNT5-6 and ZNT7 for TYRP1 expression is conserved in human, mouse, and chicken orthologs. Our results provide novel insights into the pigmentation process and address questions regarding metalation in tyrosinase protein family.


Subject(s)
Cation Transport Proteins , Secretory Pathway , Animals , Mice , Humans , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , Zinc/metabolism , Copper/metabolism , Pigmentation , Membrane Glycoproteins/metabolism , Oxidoreductases/metabolism , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism
5.
Proc Natl Acad Sci U S A ; 120(10): e2216722120, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36848556

ABSTRACT

Recent studies have uncovered the therapeutic potential of elesclomol (ES), a copper-ionophore, for copper deficiency disorders. However, we currently do not understand the mechanism by which copper brought into cells as ES-Cu(II) is released and delivered to cuproenzymes present in different subcellular compartments. Here, we have utilized a combination of genetic, biochemical, and cell-biological approaches to demonstrate that intracellular release of copper from ES occurs inside and outside of mitochondria. The mitochondrial matrix reductase, FDX1, catalyzes the reduction of ES-Cu(II) to Cu(I), releasing it into mitochondria where it is bioavailable for the metalation of mitochondrial cuproenzyme- cytochrome c oxidase. Consistently, ES fails to rescue cytochrome c oxidase abundance and activity in copper-deficient cells lacking FDX1. In the absence of FDX1, the ES-dependent increase in cellular copper is attenuated but not abolished. Thus, ES-mediated copper delivery to nonmitochondrial cuproproteins continues even in the absence of FDX1, suggesting alternate mechanism(s) of copper release. Importantly, we demonstrate that this mechanism of copper transport by ES is distinct from other clinically used copper-transporting drugs. Our study uncovers a unique mode of intracellular copper delivery by ES and may further aid in repurposing this anticancer drug for copper deficiency disorders.


Subject(s)
Copper , Electron Transport Complex IV , Hydrazines , Ionophores , Ferredoxins/metabolism
6.
Invest Ophthalmol Vis Sci ; 63(6): 18, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35727180

ABSTRACT

Purpose: To develop a mouse model of human dry eye disease (DED) for investigation of sex differences in autoimmune-associated dry eye pathology. Methods: Ocular surface disease was assessed by quantifying corneal epithelial damage with lissamine green stain in the NOD.H-2h4,IFNγ-/-,CD28-/- (NOD.H-2h4 DKO) mouse model of Sjögren's syndrome (SS). Lacrimal gland function was assessed by tear volume quantification with phenol red thread and lacrimal gland inflammation (i.e., dacryoadenitis) was assessed by quantification of immune cell foci, flow cytometric analysis of immune cell composition, and expression of proinflammatory markers. Results: The NOD.H-2h4 DKO mouse model of SS exhibits greater age-dependent increases in corneal damage than in NOD.H-2h4 parental mice and demonstrates an earlier disease onset in females compared to males. The severity of ocular surface disease correlates with loss of goblet cell density, increased conjunctivitis, and dacryoadenitis that is more pronounced in NOD.H-2h4 DKO than NOD.H-2h4 mice. B cells dominate lacrimal infiltrates in 16-week-old NOD.H-2h4 and NOD.H-2h4 DKO mice, but T helper cells and macrophages are also present. Lacrimal gland expression of proinflammatory genes, including the P2X7 and P2Y2 purinergic receptors, is greater in NOD.H-2h4 DKO than NOD.H-2h4 mice and correlates with dacryoadenitis. Conclusions: Our results demonstrate for the first time that autoimmune dry eye disease occurs in both sexes of NOD.H-2h4 DKO and NOD.H-2h4 mice, with earlier onset in female NOD.H-2h4 DKO mice when compared to males of the same strain. This study demonstrates that both NOD.H-2h4 and NOD.H-2h4 DKO mice are novel models that closely resemble SS-related and sex-dependent DED.


Subject(s)
Dacryocystitis , Dry Eye Syndromes , Lacrimal Apparatus , Sjogren's Syndrome , Animals , Dacryocystitis/pathology , Disease Models, Animal , Dry Eye Syndromes/metabolism , Female , Lacrimal Apparatus/metabolism , Male , Mice , Mice, Inbred NOD , Sjogren's Syndrome/genetics , Sjogren's Syndrome/metabolism
7.
Nat Rev Cancer ; 22(2): 102-113, 2022 02.
Article in English | MEDLINE | ID: mdl-34764459

ABSTRACT

Copper is an essential nutrient whose redox properties make it both beneficial and toxic to the cell. Recent progress in studying transition metal signalling has forged new links between researchers of different disciplines that can help translate basic research in the chemistry and biology of copper into clinical therapies and diagnostics to exploit copper-dependent disease vulnerabilities. This concept is particularly relevant in cancer, as tumour growth and metastasis have a heightened requirement for this metal nutrient. Indeed, the traditional view of copper as solely an active site metabolic cofactor has been challenged by emerging evidence that copper is also a dynamic signalling metal and metalloallosteric regulator, such as for copper-dependent phosphodiesterase 3B (PDE3B) in lipolysis, mitogen-activated protein kinase kinase 1 (MEK1) and MEK2 in cell growth and proliferation and the kinases ULK1 and ULK2 in autophagy. In this Perspective, we summarize our current understanding of the connection between copper and cancer and explore how challenges in the field could be addressed by using the framework of cuproplasia, which is defined as regulated copper-dependent cell proliferation and is a representative example of a broad range of metalloplasias. Cuproplasia is linked to a diverse array of cellular processes, including mitochondrial respiration, antioxidant defence, redox signalling, kinase signalling, autophagy and protein quality control. Identifying and characterizing new modes of copper-dependent signalling offers translational opportunities that leverage disease vulnerabilities to this metal nutrient.


Subject(s)
Copper , Neoplasms , Autophagy , Cell Proliferation , Copper/metabolism , Humans , Signal Transduction
8.
J Inorg Biochem ; 219: 111424, 2021 06.
Article in English | MEDLINE | ID: mdl-33765639

ABSTRACT

Copper is an essential metal for virtually all organisms, yet little is known about the extracellular sources of this micronutrient. In serum, the most abundant extracellular Cu-binding molecule is the multi­copper oxidase ceruloplasmin (Cp). Cp levels increase during infection and inflammation, and pathogens can be exposed to high Cp at sites of infection. It is not known whether Cp might serve as a Cu source for microbial pathogens and we tested this using the opportunistic fungal pathogen Candida albicans. We find that C. albicans can use whole serum as a Cu source and that this Cu is sensed by the transcription factor protein Mac1. Mac1 activates expression of Mn-SOD3 superoxide dismutase and represses Cu/Zn-SOD1 during Cu starvation and both responses are regulated by serum Cu. We also show that purified human Cp can act as a sole source of Cu for the fungus and likewise modulates the Mac1 Cu stress response. To investigate whether Cp is a Cu source in serum, we compared the ability of C. albicans to use serum from wild type versus Cp-/- mutant mice. We find that serum lacking Cp is deficient in its ability to trigger the Mac1 Cu response. C. albicans did accumulate Cu from Cp-/- serum, but this Cu was not efficiently sensed by Mac1. We conclude that Cp and non-Cp Cu sources are not equivalent and are handled differently by the fungal cell. Overall, these studies are the first to show that Cp is a preferred source of Cu for a pathogen.


Subject(s)
Candida albicans/metabolism , Candidiasis/metabolism , Ceruloplasmin/metabolism , Copper/metabolism , Animals , Copper/blood , Female , Fungal Proteins/metabolism , Humans , Male , Mice , Nuclear Proteins/metabolism , Superoxide Dismutase/metabolism , Superoxide Dismutase-1/metabolism , Transcription Factors/metabolism
9.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118893, 2021 02.
Article in English | MEDLINE | ID: mdl-33091507

ABSTRACT

The last 25 years have witnessed tremendous progress in identifying and characterizing proteins that regulate the uptake, intracellular trafficking and export of copper. Although dietary copper is required in trace amounts, sufficient quantities of this metal are needed to sustain growth and development in humans and other mammals. However, copper is also a rate-limiting nutrient for the growth and proliferation of cancer cells. Oral copper chelators taken with food have been shown to confer anti-neoplastic and anti-metastatic benefits in animals and humans. Recent studies have begun to identify specific roles for copper in pathways of oncogenic signaling and resistance to anti-neoplastic drugs. Here, we review the general mechanisms of cellular copper homeostasis and discuss roles of copper in cancer progression, highlighting metabolic vulnerabilities that may be targetable in the development of anticancer therapies.


Subject(s)
Antineoplastic Agents/administration & dosage , Carcinogenesis/metabolism , Copper/metabolism , Molecular Targeted Therapy/methods , Neoplasms/drug therapy , Neoplasms/metabolism , Signal Transduction/drug effects , Animals , Carcinogenesis/pathology , Disease Progression , Homeostasis/drug effects , Humans , Neoplasms/pathology
10.
Oral Oncol ; 109: 104808, 2020 Jun 12.
Article in English | MEDLINE | ID: mdl-32540611

ABSTRACT

OBJECTIVES: To assess functional expression of the P2Y2 nucleotide receptor (P2Y2R) in head and neck squamous cell carcinoma (HNSCC) cell lines and define its role in nucleotide-induced epidermal growth factor receptor (EGFR) transactivation. The use of anti-EGFR therapeutics to treat HNSCC is hindered by intrinsic and acquired drug resistance. Defining novel pathways that modulate EGFR signaling could identify additional targets to treat HNSCC. MATERIALS AND METHODS: In human HNSCC cell lines CAL27 and FaDu and the mouse oral cancer cell line MOC2, P2Y2R contributions to extracellular nucleotide-induced changes in intracellular free Ca2+ concentration and EGFR and extracellular signal-regulated kinase (ERK1/2) phosphorylation were determined using the ratiometric Ca2+ indicator fura-2 and immunoblot analysis, respectively. Genetic knockout of P2Y2Rs using CRISPR technology or pharmacological inhibition with P2Y2R-selective antagonist AR-C118925 defined P2Y2R contributions to in vivo tumor growth. RESULTS: P2Y2R agonists UTP and ATP increased intracellular Ca2+ levels and ERK1/2 and EGFR phosphorylation in CAL27 and FaDu cells, responses that were inhibited by AR-C118925 or P2Y2R knockout. P2Y2R-mediated EGFR phosphorylation was also attenuated by inhibition of the adamalysin family of metalloproteases or Src family kinases. P2Y2R knockout reduced UTP-induced CAL27 cell proliferation in vitro and significantly reduced CAL27 and FaDu tumor xenograft volume in vivo. In a syngeneic mouse model of oral cancer, AR-C118925 administration reduced MOC2 tumor volume. CONCLUSION: P2Y2Rs mediate HNSCC cell responses to extracellular nucleotides and genetic or pharmacological blockade of P2Y2R signaling attenuates tumor cell proliferation and tumorigenesis, suggesting that the P2Y2R represents a novel therapeutic target in HNSCC.

11.
Sci Rep ; 10(1): 7856, 2020 05 12.
Article in English | MEDLINE | ID: mdl-32398691

ABSTRACT

Copper (Cu) is an essential, yet potentially toxic nutrient, as illustrated by inherited diseases of copper deficiency and excess. Elevated expression of the ATP7A Cu exporter is known to confer copper tolerance, however, the contribution of metal-binding metallothioneins is less clear. In this study, we investigated the relative contributions of ATP7A and the metallothioneins MT-I and MT-II to cell viability under conditions of Cu excess or deficiency. Although the loss of ATP7A increased sensitivity to low Cu concentrations, the absence of MTs did not significantly affect Cu tolerance. However, the absence of all three proteins caused a synthetic lethal phenotype due to extreme Cu sensitivity, indicating that MTs are critical for Cu tolerance only in the absence of ATP7A. A lack of MTs resulted in the trafficking of ATP7A from the trans-Golgi complex in a Cu-dependent manner, suggesting that MTs regulate the delivery of Cu to ATP7A. Under Cu deficiency conditions, the absence of MTs and / or ATP7A enhanced cell proliferation compared to wild type cells, suggesting that these proteins compete with essential Cu-dependent pathways when Cu is scarce. These studies reveal new roles for ATP7A and metallothioneins under both Cu deficiency and excess.


Subject(s)
Copper-Transporting ATPases/metabolism , Copper/pharmacology , Metallothionein/metabolism , Animals , Cell Line , Cell Survival/drug effects , Copper-Transporting ATPases/deficiency , Copper-Transporting ATPases/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Golgi Apparatus/drug effects , Golgi Apparatus/metabolism , Metallothionein/deficiency , Metallothionein/genetics , Mice , Mutation , Protein Transport/drug effects
12.
Science ; 368(6491): 620-625, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32381719

ABSTRACT

Loss-of-function mutations in the copper (Cu) transporter ATP7A cause Menkes disease. Menkes is an infantile, fatal, hereditary copper-deficiency disorder that is characterized by progressive neurological injury culminating in death, typically by 3 years of age. Severe copper deficiency leads to multiple pathologies, including impaired energy generation caused by cytochrome c oxidase dysfunction in the mitochondria. Here we report that the small molecule elesclomol escorted copper to the mitochondria and increased cytochrome c oxidase levels in the brain. Through this mechanism, elesclomol prevented detrimental neurodegenerative changes and improved the survival of the mottled-brindled mouse-a murine model of severe Menkes disease. Thus, elesclomol holds promise for the treatment of Menkes and associated disorders of hereditary copper deficiency.


Subject(s)
Copper/metabolism , Hydrazines/therapeutic use , Menkes Kinky Hair Syndrome/drug therapy , Animals , Biological Transport/drug effects , Brain/metabolism , Brain/pathology , Cell Line , Copper Transporter 1/genetics , Disease Models, Animal , Electron Transport Complex IV/metabolism , Hydrazines/pharmacology , Male , Menkes Kinky Hair Syndrome/metabolism , Menkes Kinky Hair Syndrome/pathology , Mice , Mice, Knockout , Mitochondria/metabolism , Neurodegenerative Diseases/prevention & control , Rats
13.
Metallomics ; 12(3): 416-426, 2020 03 25.
Article in English | MEDLINE | ID: mdl-31976503

ABSTRACT

Animals carefully control homeostasis of Cu, a metal that is both potentially toxic and an essential nutrient. During infection, various shifts in Cu homeostasis can ensue. In mice infected with Candida albicans, serum Cu progressively rises and at late stages of infection, liver Cu rises, while kidney Cu declines. The basis for these changes in Cu homeostasis was poorly understood. We report here that the progressive rise in serum Cu is attributable to liver production of the multicopper oxidase ceruloplasmin (Cp). Through studies using Cp-/- mice, we find this elevated Cp helps recover serum Fe levels at late stages of infection, consistent with a role for Cp in loading transferrin with Fe. Cp also accounts for the elevation in liver Cu seen during infection, but not for the fluctuations in kidney Cu. The Cu exporting ATPase ATP7B is one candidate for kidney Cu control, but we find no change in the pattern of kidney Cu loss during infection of Atp7b-/- mice, implying alternative mechanisms. To test whether fungal infiltration of kidney tissue was required for kidney Cu loss, we explored other paradigms of infection. Infection with the intravascular malaria parasite Plasmodium berghei caused a rise in serum Cu and decrease in kidney Cu similar to that seen with C. albicans. Thus, dynamics in kidney Cu homeostasis appear to be a common feature among vastly different infection paradigms. The implications for such Cu homeostasis control in immunity are discussed.


Subject(s)
Candida albicans/physiology , Candidiasis/metabolism , Copper/metabolism , Animals , Candidiasis/blood , Ceruloplasmin/metabolism , Copper/blood , Female , Homeostasis , Kidney/metabolism , Liver/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
14.
Diabetologia ; 62(12): 2340-2353, 2019 12.
Article in English | MEDLINE | ID: mdl-31396659

ABSTRACT

AIMS/HYPOTHESIS: ATPase copper transporting α (ATP7A), also known as Menkes disease protein, is a P-type ATPase that transports copper across cell membranes. The critical role of ATP7A-mediated copper homeostasis has been well recognised in various organs, such as the intestine, macrophages and the nervous system. However, the importance of adipocyte ATP7A-mediated copper homeostasis on fat metabolism is not well understood. Here, we sought to reveal the contribution of adipose ATP7A to whole-body fat metabolism in mice. METHODS: We generated adipocyte-specific Atp7a-knockout (ASKO) mice using the Cre/loxP system, with Cre expression driven by the adiponectin promoter. ASKO mice and littermate control mice were aged on a chow diet or fed with a high-fat diet (HFD); body weight, fat mass, and glucose and insulin metabolism were analysed. Histological analysis, transmission electron microscopy and RNA-sequencing (RNA-Seq) analysis of white adipose tissue (WAT) were used to understand the physiological and molecular changes associated with loss of copper homeostasis in adipocytes. RESULTS: Significantly increased copper concentrations were observed in adipose tissues of ASKO mice compared with control mice. Aged or HFD-fed ASKO mice manifested a lipoatrophic phenotype characterised by a progressive generalised loss of WAT. Dysfunction of adipose tissues in these ASKO mice was confirmed by decreased levels of both serum leptin and adiponectin and increased levels of triacylglycerol and insulin. Systemic metabolism was also impaired in these mice, as evidenced by a pronounced glucose intolerance, insulin resistance and hepatic steatosis. Moreover, we demonstrate a significant induction of lipolysis and DNA-damage signalling pathways in gonadal WAT from aged and HFD-fed ASKO mice. In vitro studies suggest that copper overload is responsible for increased lipolysis and DNA damage. CONCLUSIONS/INTERPRETATION: Our results show a previously unappreciated role of adipocyte Atp7a in the regulation of ageing-related metabolic disease and identify new metallophysiologies in whole-body fat metabolism. DATA AVAILABILITY: The datasets generated during the current study are available in the Genome Sequence Archive in BIG Data Center, Beijing Institute of Genomics (BIG), Chinese Academy of Sciences, under accession number CRA001769 (http://bigd.big.ac.cn/gsa).


Subject(s)
Adipocytes/metabolism , Aging/metabolism , Copper-Transporting ATPases/metabolism , Copper/metabolism , Lipid Metabolism/genetics , Lipodystrophy/metabolism , 3T3-L1 Cells , Adipose Tissue, White/metabolism , Aging/genetics , Animals , Body Weight/physiology , Copper-Transporting ATPases/genetics , Diet, High-Fat , Energy Metabolism/physiology , Insulin Resistance/physiology , Lipodystrophy/genetics , Lipolysis/genetics , Mice , Mice, Knockout
15.
Proc Natl Acad Sci U S A ; 116(14): 6836-6841, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30890638

ABSTRACT

Lysyl oxidase (LOX) and LOX-like (LOXL) proteins are copper-dependent metalloenzymes with well-documented roles in tumor metastasis and fibrotic diseases. The mechanism by which copper is delivered to these enzymes is poorly understood. In this study, we demonstrate that the copper transporter ATP7A is necessary for the activity of LOX and LOXL enzymes. Silencing of ATP7A inhibited LOX activity in the 4T1 mammary carcinoma cell line, resulting in a loss of LOX-dependent mechanisms of metastasis, including the phosphorylation of focal adhesion kinase and myeloid cell recruitment to the lungs, in an orthotopic mouse model of breast cancer. ATP7A silencing was also found to attenuate LOX activity and metastasis of Lewis lung carcinoma cells in mice. Meta-analysis of breast cancer patients found that high ATP7A expression was significantly correlated with reduced survival. Taken together, these results identify ATP7A as a therapeutic target for blocking LOX- and LOXL-dependent malignancies.


Subject(s)
Carcinoma, Lewis Lung/enzymology , Copper-Transporting ATPases/metabolism , Copper/metabolism , Mammary Neoplasms, Animal/enzymology , Neoplasm Proteins/metabolism , Protein-Lysine 6-Oxidase/metabolism , Animals , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Copper-Transporting ATPases/genetics , Female , Humans , Ion Transport , Male , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Meta-Analysis as Topic , Mice , Mice, Inbred BALB C , Neoplasm Metastasis , Neoplasm Proteins/genetics , Protein-Lysine 6-Oxidase/genetics
16.
J Biol Chem ; 294(8): 2815-2826, 2019 02 22.
Article in English | MEDLINE | ID: mdl-30593504

ABSTRACT

Zinc is an essential trace element that serves as a cofactor for enzymes in critical biochemical processes and also plays a structural role in numerous proteins. Zinc transporter ZIP4 (ZIP4) is a zinc importer required for dietary zinc uptake in the intestine and other cell types. Studies in cultured cells have reported that zinc stimulates the endocytosis of plasma membrane-localized ZIP4 protein, resulting in reduced cellular zinc uptake. Thus, zinc-regulated trafficking of ZIP4 is a key means for regulating cellular zinc homeostasis, but the underlying mechanisms are not well understood. In this study, we used mutational analysis, immunoblotting, HEK293 cells, and immunofluorescence microscopy to identify a histidine-containing motif (398HTH) in the first extracellular loop that is required for high sensitivity to low zinc concentrations in a zinc-induced endocytic response of mouse ZIP4 (mZIP4). Moreover, using synthetic peptides with selective substitutions and truncated mZIP4 variants, we provide evidence that histidine residues in this motif coordinate a zinc ion in mZIP4 homodimers at the plasma membrane. These findings suggest that 398HTH is an important zinc-sensing motif for eliciting high-affinity zinc-stimulated endocytosis of mZIP4 and provide insight into cellular mechanisms for regulating cellular zinc homeostasis in mammalian cells.


Subject(s)
Cation Transport Proteins/metabolism , Endocytosis/physiology , Extracellular Matrix/metabolism , Histidine/chemistry , Mutant Proteins/metabolism , Mutation , Zinc/pharmacology , Amino Acid Motifs , Amino Acid Sequence , Cation Transport Proteins/chemistry , Cation Transport Proteins/genetics , Cell Membrane/metabolism , Endocytosis/drug effects , HEK293 Cells , Histidine/metabolism , Humans , Mutant Proteins/chemistry , Mutant Proteins/genetics , Protein Transport , Sequence Homology
17.
Cell Syst ; 6(3): 368-380.e6, 2018 Mar 28.
Article in English | MEDLINE | ID: mdl-29397366

ABSTRACT

Rare neurological diseases shed light onto universal neurobiological processes. However, molecular mechanisms connecting genetic defects to their disease phenotypes are elusive. Here, we obtain mechanistic information by comparing proteomes of cells from individuals with rare disorders with proteomes from their disease-free consanguineous relatives. We use triple-SILAC mass spectrometry to quantify proteomes from human pedigrees affected by mutations in ATP7A, which cause Menkes disease, a rare neurodegenerative and neurodevelopmental disorder stemming from systemic copper depletion. We identified 214 proteins whose expression was altered in ATP7A-/y fibroblasts. Bioinformatic analysis of ATP7A-mutant proteomes identified known phenotypes and processes affected in rare genetic diseases causing copper dyshomeostasis, including altered mitochondrial function. We found connections between copper dyshomeostasis and the UCHL1/PARK5 pathway of Parkinson disease, which we validated with mitochondrial respiration and Drosophila genetics assays. We propose that our genealogical "omics" strategy can be broadly applied to identify mechanisms linking a genomic locus to its phenotypes.


Subject(s)
Copper/metabolism , Ubiquitin Thiolesterase/genetics , Adenosine Triphosphatases/genetics , Animals , Cation Transport Proteins/genetics , Computational Biology/methods , Copper-Transporting ATPases/genetics , Copper-Transporting ATPases/metabolism , Disease Models, Animal , Drosophila , Female , Fibroblasts/metabolism , Homeostasis/genetics , Humans , Male , Menkes Kinky Hair Syndrome/genetics , Mice , Mice, Inbred C57BL , Mitochondria/genetics , Mutation , Pedigree , Phenotype , Proteomics/methods , Rare Diseases/metabolism , Ubiquitin Thiolesterase/metabolism
18.
Hum Mol Genet ; 26(23): 4617-4628, 2017 12 01.
Article in English | MEDLINE | ID: mdl-28973536

ABSTRACT

SCO1 is a ubiquitously expressed, mitochondrial protein with essential roles in cytochrome c oxidase (COX) assembly and the regulation of copper homeostasis. SCO1 patients present with severe forms of early onset disease, and ultimately succumb from liver, heart or brain failure. However, the inherent susceptibility of these tissues to SCO1 mutations and the clinical heterogeneity observed across SCO1 pedigrees remain poorly understood phenomena. To further address this issue, we generated Sco1hrt/hrt and Sco1stm/stm mice in which Sco1 was specifically deleted in heart and striated muscle, respectively. Lethality was observed in both models due to a combined COX and copper deficiency that resulted in a dilated cardiomyopathy. Left ventricular dilation and loss of heart function was preceded by a temporal decrease in COX activity and copper levels in the longer-lived Sco1stm/stm mice. Interestingly, the reduction in copper content of Sco1stm/stm cardiomyocytes was due to the mislocalisation of CTR1, the high affinity transporter that imports copper into the cell. CTR1 was similarly mislocalized to the cytosol in the heart of knockin mice carrying a homozygous G115S substitution in Sco1, which in humans causes a hypertrophic cardiomyopathy. Our current findings in the heart are in marked contrast to our prior observations in the liver, where Sco1 deletion results in a near complete absence of CTR1 protein. These data collectively argue that mutations perturbing SCO1 function have tissue-specific consequences for the machinery that ultimately governs copper homeostasis, and further establish the importance of aberrant mitochondrial signaling to the etiology of copper handling disorders.


Subject(s)
Cation Transport Proteins/metabolism , Copper/metabolism , Electron Transport Complex IV/metabolism , Mitochondria, Heart/metabolism , Myocardium/metabolism , Animals , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/metabolism , Cell Membrane/metabolism , Copper/deficiency , Copper Transporter 1 , Disease Models, Animal , Electron Transport Complex IV/genetics , Homeostasis , Ion Transport , Metallochaperones/genetics , Metallochaperones/metabolism , Mice , Mice, Transgenic , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Molecular Chaperones , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Signal Transduction
19.
J Biol Chem ; 292(40): 16626-16637, 2017 10 06.
Article in English | MEDLINE | ID: mdl-28798231

ABSTRACT

Salivary gland inflammation is a hallmark of Sjögren's syndrome (SS), a common autoimmune disease characterized by lymphocytic infiltration of the salivary gland and loss of saliva secretion, predominantly in women. The P2X7 receptor (P2X7R) is an ATP-gated nonselective cation channel that induces inflammatory responses in cells and tissues, including salivary gland epithelium. In immune cells, P2X7R activation induces the production of proinflammatory cytokines, including IL-1ß and IL-18, by inducing the oligomerization of the multiprotein complex NLRP3-type inflammasome. Here, our results show that in primary mouse submandibular gland (SMG) epithelial cells, P2X7R activation also induces the assembly of the NLRP3 inflammasome and the maturation and release of IL-1ß, a response that is absent in SMG cells isolated from mice deficient in P2X7Rs (P2X7R-/-). P2X7R-mediated IL-1ß release in SMG epithelial cells is dependent on transmembrane Na+ and/or K+ flux and the activation of heat shock protein 90 (HSP90), a protein required for the activation and stabilization of the NLRP3 inflammasome. Also, using the reactive oxygen species (ROS) scavengers N-acetyl cysteine and Mito-TEMPO, we determined that mitochondrial reactive oxygen species are required for P2X7R-mediated IL-1ß release. Lastly, in vivo administration of the P2X7R antagonist A438079 in the CD28-/-, IFNγ-/-, NOD.H-2h4 mouse model of salivary gland exocrinopathy ameliorated salivary gland inflammation and enhanced carbachol-induced saliva secretion. These findings demonstrate that P2X7R antagonism in vivo represents a promising therapeutic strategy to limit salivary gland inflammation and improve secretory function.


Subject(s)
Epithelial Cells/metabolism , Interleukin-1beta/metabolism , Purinergic P2X Receptor Antagonists/pharmacology , Pyridines/pharmacology , Receptors, Purinergic P2X7/metabolism , Sjogren's Syndrome/metabolism , Submandibular Gland/metabolism , Tetrazoles/pharmacology , Animals , CD28 Antigens/genetics , CD28 Antigens/metabolism , Disease Models, Animal , Epithelial Cells/pathology , Inflammasomes , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-18/genetics , Interleukin-18/metabolism , Ion Transport/drug effects , Ion Transport/genetics , Mice , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Potassium/metabolism , Receptors, Purinergic P2X7/genetics , Sjogren's Syndrome/genetics , Sjogren's Syndrome/pathology , Sodium/metabolism , Submandibular Gland/pathology
20.
J Cancer ; 8(11): 1952-1958, 2017.
Article in English | MEDLINE | ID: mdl-28819394

ABSTRACT

The ATP7A protein is a ubiquitously expressed copper-translocating P-type ATPase that controls cytoplasmic copper concentrations by mediating cellular copper egress. In vitro studies have previously demonstrated that ATP7A abundance in various tumor cell lines is correlated with increased resistance to cisplatin, a widely-used chemotherapy agent. However, to date no studies have examined a role for ATP7A in tumor growth or cisplatin sensitivity in vivo. In this study, we deleted ATP7A in H-RAS transformed tumorigenic mouse embryonic fibroblasts (MEFRAS7A-). Interestingly, loss of ATP7A was found to markedly suppress tumorigenesis in MEFRAS7A- cells relative to wild type parental cells. This was associated with hyperaccumulation of copper and sensitivity to reactive oxygen species and hypoxia. Tumor grafts lacking ATP7A were markedly more sensitive to cisplatin chemotherapy compared to ATP7A-expressing control tumors. These findings identify ATP7A at the nexus between tumorigenesis and cisplatin resistance pathways, underscoring its potential as a therapeutic target for regulating both tumor growth and the efficacy of cisplatin treatment.

SELECTION OF CITATIONS
SEARCH DETAIL
...