Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Heliyon ; 10(2): e24368, 2024 Jan 30.
Artigo em Inglês | MEDLINE | ID: mdl-38298614

RESUMO

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.
Artigo em Inglês | MEDLINE | ID: mdl-37391383

RESUMO

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.


Assuntos
Fibras Musculares Esqueléticas , RNA , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Diferenciação Celular , RNA Mensageiro/genética , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Cobre/metabolismo
3.
Commun Biol ; 6(1): 403, 2023 04 18.
Artigo em Inglês | MEDLINE | ID: mdl-37072620

RESUMO

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.


Assuntos
Proteínas de Transporte de Cátions , Via Secretória , Animais , Camundongos , Humanos , Monofenol Mono-Oxigenase/genética , Monofenol Mono-Oxigenase/metabolismo , Zinco/metabolismo , Cobre/metabolismo , Pigmentação , Glicoproteínas de Membrana/metabolismo , Oxirredutases/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo
4.
Proc Natl Acad Sci U S A ; 120(10): e2216722120, 2023 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-36848556

RESUMO

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.


Assuntos
Cobre , Complexo IV da Cadeia de Transporte de Elétrons , Hidrazinas , Ionóforos , Ferredoxinas/metabolismo
5.
Nat Rev Cancer ; 22(2): 102-113, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34764459

RESUMO

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.


Assuntos
Cobre , Neoplasias , Autofagia , Proliferação de Células , Cobre/metabolismo , Humanos , Transdução de Sinais
6.
Biochim Biophys Acta Mol Cell Res ; 1868(2): 118893, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33091507

RESUMO

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.


Assuntos
Antineoplásicos/administração & dosagem , Carcinogênese/metabolismo , Cobre/metabolismo , Terapia de Alvo Molecular/métodos , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Carcinogênese/patologia , Progressão da Doença , Homeostase/efeitos dos fármacos , Humanos , Neoplasias/patologia
7.
Oral Oncol ; 109: 104808, 2020 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-32540611

RESUMO

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.

8.
Sci Rep ; 10(1): 7856, 2020 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-32398691

RESUMO

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.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Cobre/farmacologia , Metalotioneína/metabolismo , Animais , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , ATPases Transportadoras de Cobre/deficiência , ATPases Transportadoras de Cobre/genética , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Metalotioneína/deficiência , Metalotioneína/genética , Camundongos , Mutação , Transporte Proteico/efeitos dos fármacos
9.
Diabetologia ; 62(12): 2340-2353, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31396659

RESUMO

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).


Assuntos
Adipócitos/metabolismo , Envelhecimento/metabolismo , ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Metabolismo dos Lipídeos/genética , Lipodistrofia/metabolismo , Células 3T3-L1 , Tecido Adiposo Branco/metabolismo , Envelhecimento/genética , Animais , Peso Corporal/fisiologia , ATPases Transportadoras de Cobre/genética , Dieta Hiperlipídica , Metabolismo Energético/fisiologia , Resistência à Insulina/fisiologia , Lipodistrofia/genética , Lipólise/genética , Camundongos , Camundongos Knockout
10.
Proc Natl Acad Sci U S A ; 116(14): 6836-6841, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30890638

RESUMO

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.


Assuntos
Carcinoma Pulmonar de Lewis/enzimologia , ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Neoplasias Mamárias Animais/enzimologia , Proteínas de Neoplasias/metabolismo , Proteína-Lisina 6-Oxidase/metabolismo , Animais , Neoplasias da Mama/enzimologia , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , ATPases Transportadoras de Cobre/genética , Feminino , Humanos , Transporte de Íons , Masculino , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Metanálise como Assunto , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Proteínas de Neoplasias/genética , Proteína-Lisina 6-Oxidase/genética
11.
J Biol Chem ; 292(40): 16626-16637, 2017 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-28798231

RESUMO

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.


Assuntos
Células Epiteliais/metabolismo , Interleucina-1beta/metabolismo , Antagonistas do Receptor Purinérgico P2X/farmacologia , Piridinas/farmacologia , Receptores Purinérgicos P2X7/metabolismo , Síndrome de Sjogren/metabolismo , Glândula Submandibular/metabolismo , Tetrazóis/farmacologia , Animais , Antígenos CD28/genética , Antígenos CD28/metabolismo , Modelos Animais de Doenças , Células Epiteliais/patologia , Inflamassomos , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-18/genética , Interleucina-18/metabolismo , Transporte de Íons/efeitos dos fármacos , Transporte de Íons/genética , Camundongos , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Potássio/metabolismo , Receptores Purinérgicos P2X7/genética , Síndrome de Sjogren/genética , Síndrome de Sjogren/patologia , Sódio/metabolismo , Glândula Submandibular/patologia
12.
Infect Immun ; 85(9)2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28652309

RESUMO

Copper is an essential yet potentially toxic trace element that is required by all aerobic organisms. A key regulator of copper homeostasis in mammalian cells is the copper-transporting P-type ATPase ATP7A, which mediates copper transport from the cytoplasm into the secretory pathway, as well as copper export across the plasma membrane. Previous studies have shown that ATP7A-dependent copper transport is required for killing phagocytosed Escherichia coli in a cultured macrophage cell line. In this investigation, we expanded on these studies by generating Atp7aLysMcre mice, in which the Atp7a gene was specifically deleted in cells of the myeloid lineage, including macrophages. Primary macrophages isolated from Atp7aLysMcre mice exhibit decreased copper transport into phagosomal compartments and a reduced ability to kill Salmonella enterica serovar Typhimurium compared to that of macrophages isolated from wild-type mice. The Atp7aLysMcre mice were also more susceptible to systemic infection by S Typhimurium than wild-type mice. Deletion of the S Typhimurium copper exporters, CopA and GolT, was found to decrease infection in wild-type mice but not in the Atp7aLysMcre mice. These studies suggest that ATP7A-dependent copper transport into the phagosome mediates host defense against S Typhimurium, which is counteracted by copper export from the bacteria via CopA and GolT. These findings reveal unique and opposing functions for copper transporters of the host and pathogen during infection.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Interações Hospedeiro-Patógeno , Macrófagos/enzimologia , Salmonella typhimurium/enzimologia , Adenosina Trifosfatases/genética , Animais , Proteínas de Transporte de Cátions/genética , Cobre/toxicidade , Feminino , Macrófagos/imunologia , Masculino , Camundongos Knockout , Salmonelose Animal/microbiologia , Salmonelose Animal/patologia , Salmonella typhimurium/efeitos dos fármacos , Salmonella typhimurium/fisiologia , Virulência
13.
Metallomics ; 8(9): 993-1001, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27337370

RESUMO

The ATP7A protein is a ubiquitous copper-transporting P-type ATPase that is mutated in the lethal pediatric disorder of copper metabolism, Menkes disease. The steady-state location of ATP7A is within the trans-Golgi network (TGN), where it delivers copper to copper-dependent enzymes within the secretory pathway. However, ATP7A constantly cycles between the TGN and the plasma membrane, and in the presence of high copper concentrations, the exocytic arm of this cycling pathway is enhanced to promote a steady-state distribution of ATP7A to post-Golgi vesicles and the plasma membrane. A single di-leucine endocytic motif within the cytosolic carboxy tail of ATP7A (1487LL) was previously shown to be essential for TGN localization by functioning in retrieval from the plasma membrane, however, the requirement of other di-leucine signals in this region has not been fully investigated. While there has been some success in identifying sequence elements within ATP7A required for trafficking and catalysis, progress has been hampered by the instability of the ATP7A cDNA in high-copy plasmids during replication in Escherichia coli. In this study, we find that the use of DNA synthesis to generate silent mutations across the majority of both mouse and human ATP7A open reading frames was sufficient to stabilize these genes in high-copy plasmids, thus permitting the generation of full-length expression constructs. Using the stabilized mouse Atp7a construct, we identify a second di-leucine motif in the carboxy tail of ATP7A (1459LL) as essential for steady-state localization in the TGN by functioning in endosome-to-TGN trafficking. Taken together, these findings demonstrate that multiple di-leucine signals are required for recycling ATP7A from the plasma membrane to the TGN and illustrate the utility of large-scale codon reassignment as a simple and effective approach to circumvent cDNA instability in high-copy plasmids.


Assuntos
ATPases Transportadoras de Cobre/metabolismo , Cobre/metabolismo , Endocitose/fisiologia , Leucina/metabolismo , Rede trans-Golgi/metabolismo , Animais , Membrana Celular , ATPases Transportadoras de Cobre/genética , Células HEK293 , Humanos , Leucina/química , Leucina/genética , Camundongos , Mutação , Transporte Proteico
14.
Dis Model Mech ; 9(1): 25-38, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26747866

RESUMO

The deleterious effects of a disrupted copper metabolism are illustrated by hereditary diseases caused by mutations in the genes coding for the copper transporters ATP7A and ATP7B. Menkes disease, involving ATP7A, is a fatal neurodegenerative disorder of copper deficiency. Mutations in ATP7B lead to Wilson disease, which is characterized by a predominantly hepatic copper accumulation. The low incidence and the phenotypic variability of human copper toxicosis hamper identification of causal genes or modifier genes involved in the disease pathogenesis. The Labrador retriever was recently characterized as a new canine model for copper toxicosis. Purebred dogs have reduced genetic variability, which facilitates identification of genes involved in complex heritable traits that might influence phenotype in both humans and dogs. We performed a genome-wide association study in 235 Labrador retrievers and identified two chromosome regions containing ATP7A and ATP7B that were associated with variation in hepatic copper levels. DNA sequence analysis identified missense mutations in each gene. The amino acid substitution ATP7B:p.Arg1453Gln was associated with copper accumulation, whereas the amino acid substitution ATP7A:p.Thr327Ile partly protected against copper accumulation. Confocal microscopy indicated that aberrant copper metabolism upon expression of the ATP7B variant occurred because of mis-localization of the protein in the endoplasmic reticulum. Dermal fibroblasts derived from ATP7A:p.Thr327Ile dogs showed copper accumulation and delayed excretion. We identified the Labrador retriever as the first natural, non-rodent model for ATP7B-associated copper toxicosis. Attenuation of copper accumulation by the ATP7A mutation sheds an interesting light on the interplay of copper transporters in body copper homeostasis and warrants a thorough investigation of ATP7A as a modifier gene in copper-metabolism disorders. The identification of two new functional variants in ATP7A and ATP7B contributes to the biological understanding of protein function, with relevance for future development of therapy.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/genética , Cobre/toxicidade , Modelos Animais de Doenças , Degeneração Hepatolenticular/genética , Síndrome dos Cabelos Torcidos/genética , Sequência de Aminoácidos , Animais , ATPases Transportadoras de Cobre , Cães , Retículo Endoplasmático/metabolismo , Feminino , Variação Genética , Estudo de Associação Genômica Ampla , Genótipo , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Dados de Sequência Molecular , Mutação de Sentido Incorreto , Fenótipo , Estrutura Terciária de Proteína , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos
15.
Am J Physiol Cell Physiol ; 309(10): C660-8, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26269458

RESUMO

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


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte de Cátions/metabolismo , Cobre/metabolismo , Síndrome dos Cabelos Torcidos/metabolismo , Adenosina Trifosfatases/genética , Animais , Proteínas de Transporte de Cátions/genética , ATPases Transportadoras de Cobre , Feminino , Regulação da Expressão Gênica/fisiologia , Integrases , Masculino , Síndrome dos Cabelos Torcidos/genética , Síndrome dos Cabelos Torcidos/patologia , Camundongos , Camundongos Knockout , Mutação
16.
J Pathol ; 236(2): 241-50, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25639447

RESUMO

ATP7A is a copper-transporting P-type ATPase that is essential for cellular copper homeostasis. Loss-of-function mutations in the ATP7A gene result in Menkes disease, a fatal neurodegenerative disorder resulting in seizures, hypotonia and failure to thrive, due to systemic copper deficiency. Most recently, rare missense mutations in ATP7A that do not impact systemic copper homeostasis have been shown to cause X-linked spinal muscular atrophy type 3 (SMAX3), a distal hereditary motor neuropathy. An understanding of the mechanistic and pathophysiological basis of SMAX3 is currently lacking, in part because the disease-causing mutations have been shown to confer both loss- and gain-of-function properties to ATP7A, and because there is currently no animal model of the disease. In this study, the Atp7a gene was specifically deleted in the motor neurons of mice, resulting in a degenerative phenotype consistent with the clinical features in affected patients with SMAX3, including the progressive deterioration of gait, age-dependent muscle atrophy, denervation of neuromuscular junctions and a loss of motor neuron cell bodies. Taken together, these data reveal autonomous requirements for ATP7A that reveal essential roles for copper in the maintenance and function of the motor neuron, and suggest that SMAX3 is caused by a loss of ATP7A function that specifically impacts the spinal motor neuron.


Assuntos
Adenosina Trifosfatases/deficiência , Proteínas de Transporte de Cátions/deficiência , Doenças Genéticas Ligadas ao Cromossomo X/genética , Atrofia Muscular Espinal/genética , Adenosina Trifosfatases/genética , Animais , Proteínas de Transporte de Cátions/genética , Cobre/metabolismo , ATPases Transportadoras de Cobre , Deleção de Genes , Doenças Genéticas Ligadas ao Cromossomo X/patologia , Doenças Genéticas Ligadas ao Cromossomo X/fisiopatologia , Coxeadura Animal/genética , Coxeadura Animal/fisiopatologia , Camundongos Endogâmicos C57BL , Doença dos Neurônios Motores/genética , Doença dos Neurônios Motores/patologia , Doença dos Neurônios Motores/fisiopatologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Neurônios Motores/fisiologia , Músculo Esquelético/inervação , Atrofia Muscular Espinal/patologia , Atrofia Muscular Espinal/fisiopatologia , Mutação de Sentido Incorreto/genética , Medula Espinal/química
17.
Neurobiol Dis ; 81: 154-61, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25583185

RESUMO

ATP7A mutations impair copper metabolism resulting in three distinct genetic disorders in humans. These diseases are characterized by neurological phenotypes ranging from intellectual disability to neurodegeneration. Severe ATP7A loss-of-function alleles trigger Menkes disease, a copper deficiency condition where systemic and neurodegenerative phenotypes dominate clinical outcomes. The pathogenesis of these manifestations has been attributed to the hypoactivity of a limited number of copper-dependent enzymes, a hypothesis that we refer as the oligoenzymatic pathogenic hypothesis. This hypothesis, which has dominated the field for 25 years, only explains some systemic Menkes phenotypes. However, we argue that this hypothesis does not fully account for the Menkes neurodegeneration or neurodevelopmental phenotypes. Here, we propose revisions of the oligoenzymatic hypothesis that could illuminate the pathogenesis of Menkes neurodegeneration and neurodevelopmental defects through unsuspected overlap with other neurological conditions including Parkinson's, intellectual disability, and schizophrenia.


Assuntos
Adenosina Trifosfatases/genética , Proteínas de Transporte de Cátions/genética , Síndrome dos Cabelos Torcidos/genética , Mutação/genética , Doenças Neurodegenerativas/genética , Transtornos do Neurodesenvolvimento/genética , Animais , Biologia Computacional , ATPases Transportadoras de Cobre , Humanos , Síndrome dos Cabelos Torcidos/complicações , Camundongos , Doenças Neurodegenerativas/etiologia , Transtornos do Neurodesenvolvimento/etiologia
18.
J Invest Dermatol ; 135(3): 834-841, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25337692

RESUMO

Omeprazole is a proton pump inhibitor used in the treatment of peptic ulcer disease and gastrosophageal reflux disease and acts by irreversibly blocking ATP4A, a P-type H+/K+ ATPase in gastric parietal cells. We found that omeprazole and its closely related congeners inhibited melanogenesis at micromolar concentrations in B16 mouse melanoma cells, normal human epidermal melanocytes, and in a reconstructed human skin model. Omeprazole topically applied to the skin of UV-irradiated human subjects significantly reduced pigment levels after 3 weeks compared with untreated controls. Omeprazole had no significant inhibitory effect on the activities of purified human tyrosinase or on the mRNA levels of tyrosinase, dopachrome tautomerase, Pmel17, or MITF mRNA levels. Although melanocytes do not express ATP4A, they do express ATP7A, a copper transporting P-type ATPase in the trans-Golgi network that is required for copper acquisition by tyrosinase. ATP7A relocalization from the trans-Golgi network to the plasma membrane in response to elevated copper concentrations in melanocytes was inhibited by omeprazole. Omeprazole treatment increased the proportion of EndoH sensitive tyrosinase, indicating that tyrosinase maturation was impaired. In addition, omeprazole reduced tyrosinase protein abundance in the presence of cycloheximide, suggestive of increased degradation. Our findings are consistent with the hypothesis that omeprazole reduces melanogenesis by inhibiting ATP7A and by enhancing degradation of tyrosinase.


Assuntos
Adenosina Trifosfatases/antagonistas & inibidores , Proteínas de Transporte de Cátions/antagonistas & inibidores , Melaninas/antagonistas & inibidores , Melanócitos/metabolismo , Melanoma/metabolismo , Omeprazol/farmacologia , Inibidores da Bomba de Prótons/farmacologia , Neoplasias Cutâneas/metabolismo , Adenosina Trifosfatases/metabolismo , Animais , Proteínas de Transporte de Cátions/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , ATPases Transportadoras de Cobre , Cicloeximida/farmacologia , Modelos Animais de Doenças , Humanos , Melaninas/metabolismo , Melanócitos/efeitos dos fármacos , Melanócitos/patologia , Melanoma/patologia , Camundongos , Monofenol Mono-Oxigenase/metabolismo , Pigmentação/efeitos dos fármacos , Inibidores da Síntese de Proteínas/farmacologia , Pele/metabolismo , Pele/patologia , Neoplasias Cutâneas/patologia , Raios Ultravioleta
19.
FEBS Lett ; 588(17): 2928-35, 2014 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-24911202

RESUMO

Free zinc is required for proper lipopolysaccharide (LPS)-stimulated signaling, but potential sites of action in the pathway have not been defined. In this work, we provide in vitro and ex vivo evidence that zinc is not required for phosphorylation or ubiquitylation of IRAK1, a kinase functioning early in the TLR4 pathway. However, degradation of ubiquitylated IRAK1 occurred via a zinc-dependent, proteasome-independent pathway. These results provide evidence of a novel site of action for zinc during TLR4-mediated inflammatory responses.


Assuntos
Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Receptor 4 Toll-Like/metabolismo , Zinco/metabolismo , Animais , Linhagem Celular , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Imunidade Inata/efeitos dos fármacos , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Macrófagos/metabolismo , Camundongos , Fosforilação/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise/efeitos dos fármacos , Ubiquitinação/efeitos dos fármacos , Zinco/farmacologia
20.
Am J Physiol Cell Physiol ; 307(1): C83-96, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24760984

RESUMO

Hyposalivation resulting from salivary gland dysfunction leads to poor oral health and greatly reduces the quality of life of patients. Current treatments for hyposalivation are limited. However, regenerative medicine to replace dysfunctional salivary glands represents a revolutionary approach. The ability of dispersed salivary epithelial cells or salivary gland-derived progenitor cells to self-organize into acinar-like spheres or branching structures that mimic the native tissue holds promise for cell-based reconstitution of a functional salivary gland. However, the mechanisms involved in salivary epithelial cell aggregation and tissue reconstitution are not fully understood. This study investigated the role of the P2Y2 nucleotide receptor (P2Y2R), a G protein-coupled receptor that is upregulated following salivary gland damage and disease, in salivary gland reconstitution. In vitro results with the rat parotid acinar Par-C10 cell line indicate that P2Y2R activation with the selective agonist UTP enhances the self-organization of dispersed salivary epithelial cells into acinar-like spheres. Other results indicate that the P2Y2R-mediated response is dependent on epidermal growth factor receptor activation via the metalloproteases ADAM10/ADAM17 or the α5ß1 integrin/Cdc42 signaling pathway, which leads to activation of the MAPKs JNK and ERK1/2. Ex vivo data using primary submandibular gland cells from wild-type and P2Y2R(-/-) mice confirmed that UTP-induced migratory responses required for acinar cell self-organization are mediated by the P2Y2R. Overall, this study suggests that the P2Y2R is a promising target for salivary gland reconstitution and identifies the involvement of two novel components of the P2Y2R signaling cascade in salivary epithelial cells, the α5ß1 integrin and the Rho GTPase Cdc42.


Assuntos
Agregação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Glândula Parótida/efeitos dos fármacos , Agonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y2/efeitos dos fármacos , Glândula Submandibular/efeitos dos fármacos , Uridina Trifosfato/farmacologia , Proteínas ADAM/antagonistas & inibidores , Proteínas ADAM/metabolismo , Proteína ADAM10 , Proteína ADAM17 , Animais , Linhagem Celular , Células Epiteliais/metabolismo , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Integrina alfa5beta1/antagonistas & inibidores , Integrina alfa5beta1/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Glândula Parótida/citologia , Glândula Parótida/metabolismo , Fosforilação , Inibidores de Proteases/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Ratos , Receptores Purinérgicos P2Y2/deficiência , Receptores Purinérgicos P2Y2/genética , Glândula Submandibular/citologia , Glândula Submandibular/metabolismo , Transfecção , Proteína cdc42 de Ligação ao GTP/antagonistas & inibidores , Proteína cdc42 de Ligação ao GTP/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA