Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
Dis Model Mech ; 14(1)2021 01 01.
Article in English | MEDLINE | ID: mdl-33262129

ABSTRACT

Copper is an essential transition metal for all eukaryotes. In mammals, intestinal copper absorption is mediated by the ATP7A copper transporter, whereas copper excretion occurs predominantly through the biliary route and is mediated by the paralog ATP7B. Both transporters have been shown to be recycled actively between the endosomal network and the plasma membrane by a molecular machinery known as the COMMD/CCDC22/CCDC93 or CCC complex. In fact, mutations in COMMD1 can lead to impaired biliary copper excretion and liver pathology in dogs and in mice with liver-specific Commd1 deficiency, recapitulating aspects of this phenotype. Nonetheless, the role of the CCC complex in intestinal copper absorption in vivo has not been studied, and the potential redundancy of various COMMD family members has not been tested. In this study, we examined copper homeostasis in enterocyte-specific and hepatocyte-specific COMMD gene-deficient mice. We found that, in contrast to effects in cell lines in culture, COMMD protein deficiency induced minimal changes in ATP7A in enterocytes and did not lead to altered copper levels under low- or high-copper diets, suggesting that regulation of ATP7A in enterocytes is not of physiological consequence. By contrast, deficiency of any of three COMMD genes (Commd1, Commd6 or Commd9) resulted in hepatic copper accumulation under high-copper diets. We found that each of these deficiencies caused destabilization of the entire CCC complex and suggest that this might explain their shared phenotype. Overall, we conclude that the CCC complex plays an important role in ATP7B endosomal recycling and function.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Copper-Transporting ATPases/metabolism , Copper/metabolism , Gene Expression Regulation , Mutation , Animals , Cell Line, Tumor , Ceruloplasmin/biosynthesis , Copper/blood , Endosomes/metabolism , Enterocytes/metabolism , Hepatocytes/metabolism , Homeostasis , Humans , Mice , Mice, Knockout , Phenotype
2.
Nat Commun ; 10(1): 4271, 2019 09 19.
Article in English | MEDLINE | ID: mdl-31537807

ABSTRACT

Protein recycling through the endolysosomal system relies on molecular assemblies that interact with cargo proteins, membranes, and effector molecules. Among them, the COMMD/CCDC22/CCDC93 (CCC) complex plays a critical role in recycling events. While CCC is closely associated with retriever, a cargo recognition complex, its mechanism of action remains unexplained. Herein we show that CCC and retriever are closely linked through sharing a common subunit (VPS35L), yet the integrity of CCC, but not retriever, is required to maintain normal endosomal levels of phosphatidylinositol-3-phosphate (PI(3)P). CCC complex depletion leads to elevated PI(3)P levels, enhanced recruitment and activation of WASH (an actin nucleation promoting factor), excess endosomal F-actin and trapping of internalized receptors. Mechanistically, we find that CCC regulates the phosphorylation and endosomal recruitment of the PI(3)P phosphatase MTMR2. Taken together, we show that the regulation of PI(3)P levels by the CCC complex is critical to protein recycling in the endosomal compartment.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Endosomes/metabolism , Microfilament Proteins/metabolism , Phosphatidylinositol Phosphates/metabolism , Proteins/metabolism , Vesicular Transport Proteins/metabolism , Actins/metabolism , Animals , Cell Line, Tumor , HEK293 Cells , HeLa Cells , Humans , Lysosomes/metabolism , Membrane Proteins/metabolism , Mice , Phosphorylation , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , RNA Interference , RNA, Small Interfering/genetics
3.
Nat Commun ; 10(1): 2193, 2019 05 16.
Article in English | MEDLINE | ID: mdl-31097705

ABSTRACT

Filamentous actin (F-actin) networks facilitate key processes like cell shape control, division, polarization and motility. The dynamic coordination of F-actin networks and its impact on cellular activities are poorly understood. We report an antagonistic relationship between endosomal F-actin assembly and cortical actin bundle integrity during Drosophila airway maturation. Double mutants lacking receptor tyrosine phosphatases (PTP) Ptp10D and Ptp4E, clear luminal proteins and disassemble apical actin bundles prematurely. These defects are counterbalanced by reduction of endosomal trafficking and by mutations affecting the tyrosine kinase Btk29A, and the actin nucleation factor WASH. Btk29A forms protein complexes with Ptp10D and WASH, and Btk29A phosphorylates WASH. This phosphorylation activates endosomal WASH function in flies and mice. In contrast, a phospho-mimetic WASH variant induces endosomal actin accumulation, premature luminal endocytosis and cortical F-actin disassembly. We conclude that PTPs and Btk29A regulate WASH activity to balance the endosomal and cortical F-actin networks during epithelial tube maturation.


Subject(s)
Drosophila Proteins/metabolism , Endosomes/metabolism , Morphogenesis/physiology , Protein-Tyrosine Kinases/metabolism , Vesicular Transport Proteins/metabolism , Actins/metabolism , Animals , Animals, Genetically Modified , Cell Line , Drosophila Proteins/genetics , Drosophila melanogaster , Embryo, Nonmammalian/diagnostic imaging , Epithelium/diagnostic imaging , Epithelium/growth & development , Fibroblasts , Intravital Microscopy , Mice , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microscopy, Confocal , Phosphorylation/physiology , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Protein-Tyrosine Kinases/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Respiratory System/diagnostic imaging , Respiratory System/growth & development , Vesicular Transport Proteins/genetics
4.
Traffic ; 19(8): 578-590, 2018 08.
Article in English | MEDLINE | ID: mdl-29667289

ABSTRACT

The tubular endolysosomal network is a quality control system that ensures the proper delivery of internalized receptors to specific subcellular destinations in order to maintain cellular homeostasis. Although retromer was originally described in yeast as a regulator of endosome-to-Golgi receptor recycling, mammalian retromer has emerged as a central player in endosome-to-plasma membrane recycling of a variety of receptors. Over the past decade, information regarding the mechanism by which retromer facilitates receptor trafficking has emerged, as has the identification of numerous retromer-associated molecules including the WASH complex, sorting nexins (SNXs) and TBC1d5. Moreover, the recent demonstration that several SNXs can directly interact with retromer cargo to facilitate endosome-to-Golgi retrieval has provided new insight into how these receptors are trafficked in cells. The mechanism by which SNX17 cargoes are recycled out of the endosomal system was demonstrated to involve a retromer-like complex termed the retriever, which is recruited to WASH positive endosomes through an interaction with the COMMD/CCDC22/CCDC93 (CCC) complex. Lastly, the mechanisms by which bacterial and viral pathogens highjack this complex sorting machinery in order to escape the endolysosomal system or remain hidden within the cells are beginning to emerge. In this review, we will highlight recent studies that have begun to unravel the intricacies by which the retromer and associated molecules contribute to receptor trafficking and how deregulation at this sorting domain can contribute to disease or facilitate pathogen infection.


Subject(s)
Endosomes/metabolism , Endosomes/physiology , Protein Transport/physiology , Animals , Cell Membrane/metabolism , Cell Membrane/physiology , Golgi Apparatus/metabolism , Golgi Apparatus/physiology , Humans , trans-Golgi Network/metabolism , trans-Golgi Network/physiology
5.
Circ Res ; 122(12): 1648-1660, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29545368

ABSTRACT

RATIONALE: COMMD (copper metabolism MURR1 domain)-containing proteins are a part of the CCC (COMMD-CCDC22 [coiled-coil domain containing 22]-CCDC93 [coiled-coil domain containing 93]) complex facilitating endosomal trafficking of cell surface receptors. Hepatic COMMD1 inactivation decreases CCDC22 and CCDC93 protein levels, impairs the recycling of the LDLR (low-density lipoprotein receptor), and increases plasma low-density lipoprotein cholesterol levels in mice. However, whether any of the other COMMD members function similarly as COMMD1 and whether perturbation in the CCC complex promotes atherogenesis remain unclear. OBJECTIVE: The main aim of this study is to unravel the contribution of evolutionarily conserved COMMD proteins to plasma lipoprotein levels and atherogenesis. METHODS AND RESULTS: Using liver-specific Commd1, Commd6, or Commd9 knockout mice, we investigated the relation between the COMMD proteins in the regulation of plasma cholesterol levels. Combining biochemical and quantitative targeted proteomic approaches, we found that hepatic COMMD1, COMMD6, or COMMD9 deficiency resulted in massive reduction in the protein levels of all 10 COMMDs. This decrease in COMMD protein levels coincided with destabilizing of the core (CCDC22, CCDC93, and chromosome 16 open reading frame 62 [C16orf62]) of the CCC complex, reduced cell surface levels of LDLR and LRP1 (LDLR-related protein 1), followed by increased plasma low-density lipoprotein cholesterol levels. To assess the direct contribution of the CCC core in the regulation of plasma cholesterol levels, Ccdc22 was deleted in mouse livers via CRISPR/Cas9-mediated somatic gene editing. CCDC22 deficiency also destabilized the complete CCC complex and resulted in elevated plasma low-density lipoprotein cholesterol levels. Finally, we found that hepatic disruption of the CCC complex exacerbates dyslipidemia and atherosclerosis in ApoE3*Leiden mice. CONCLUSIONS: Collectively, these findings demonstrate a strong interrelationship between COMMD proteins and the core of the CCC complex in endosomal LDLR trafficking. Hepatic disruption of either of these CCC components causes hypercholesterolemia and exacerbates atherosclerosis. Our results indicate that not only COMMD1 but all other COMMDs and CCC components may be potential targets for modulating plasma lipid levels in humans.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Atherosclerosis/prevention & control , Cholesterol, LDL/blood , Cytoskeletal Proteins/metabolism , Endosomes/metabolism , Receptors, LDL/metabolism , Adaptor Proteins, Signal Transducing/genetics , Animals , Atherosclerosis/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Line , Cholesterol/analysis , Chromatography, High Pressure Liquid , Cytoskeletal Proteins/genetics , Gene Deletion , Gene Expression , HEK293 Cells , Hep G2 Cells , Humans , Liver/chemistry , Liver/metabolism , Low Density Lipoprotein Receptor-Related Protein-1 , Male , Mice , Mice, Knockout , Protein Transport , Triglycerides/analysis , Tumor Suppressor Proteins/metabolism
6.
PLoS One ; 11(10): e0165385, 2016.
Article in English | MEDLINE | ID: mdl-27788210

ABSTRACT

Copper metabolism MURR1 domain 1 (COMMD1) protein is a multifunctional protein, and its expression has been correlated with patients' survival in different types of cancer. In vitro studies revealed that COMMD1 plays a role in sensitizing cancer cell lines to cisplatin, however, the mechanism and its role in platinum sensitivity in cancer has yet to be established. We evaluated the role of COMMD1 in cisplatin sensitivity in A2780 ovarian cancer cells and the relation between COMMD1 expression and response to platinum-based therapy in advanced stage high-grade serous ovarian cancer (HGSOC) patients. We found that elevation of nuclear COMMD1 expression sensitized A2780 ovarian cancer cells to cisplatin-mediated cytotoxicity. This was accompanied by a more effective G2/M checkpoint, and decreased protein expression of the DNA repair gene BRCA1, and the apoptosis inhibitor BCL2. Furthermore, COMMD1 expression was immunohistochemically analyzed in two tissue micro-arrays (TMAs), representing a historical cohort and a randomized clinical trial-based cohort of advanced stage HGSOC tumor specimens. Expression of COMMD1 was observed in all ovarian cancer samples, however, specifically nuclear expression of COMMD1 was only observed in a subset of ovarian cancers. In our historical cohort, nuclear COMMD1 expression was associated with an improved response to chemotherapy (OR = 0.167; P = 0.038), although this association could not be confirmed in the second cohort, likely due to sample size. Taken together, these results suggest that nuclear expression of COMMD1 sensitize ovarian cancer to cisplatin, possibly by modulating the G2/M checkpoint and through controlling expression of genes involved in DNA repair and apoptosis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cisplatin/pharmacology , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Adaptor Proteins, Signal Transducing/genetics , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/therapeutic use , Cohort Studies , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , M Phase Cell Cycle Checkpoints/drug effects , Neoplasm Grading , Neoplasm Staging , Ovarian Neoplasms/genetics
7.
Nat Commun ; 7: 10961, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26965651

ABSTRACT

The low-density lipoprotein receptor (LDLR) plays a pivotal role in clearing atherogenic circulating low-density lipoprotein (LDL) cholesterol. Here we show that the COMMD/CCDC22/CCDC93 (CCC) and the Wiskott-Aldrich syndrome protein and SCAR homologue (WASH) complexes are both crucial for endosomal sorting of LDLR and for its function. We find that patients with X-linked intellectual disability caused by mutations in CCDC22 are hypercholesterolaemic, and that COMMD1-deficient dogs and liver-specific Commd1 knockout mice have elevated plasma LDL cholesterol levels. Furthermore, Commd1 depletion results in mislocalization of LDLR, accompanied by decreased LDL uptake. Increased total plasma cholesterol levels are also seen in hepatic COMMD9-deficient mice. Inactivation of the CCC-associated WASH complex causes LDLR mislocalization, increased lysosomal degradation of LDLR and impaired LDL uptake. Furthermore, a mutation in the WASH component KIAA0196 (strumpellin) is associated with hypercholesterolaemia in humans. Altogether, this study provides valuable insights into the mechanisms regulating cholesterol homeostasis and LDLR trafficking.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cholesterol, LDL/metabolism , Endosomes/metabolism , Hypercholesterolemia/genetics , Liver/metabolism , Microfilament Proteins/genetics , Proteins/genetics , Receptors, LDL/metabolism , Triglycerides/metabolism , Adolescent , Adult , Animals , Animals, Genetically Modified , Child , Child, Preschool , Chromatography, Liquid , Dogs , Female , Fluorescent Antibody Technique , HEK293 Cells , Humans , Immunoprecipitation , Male , Mice , Mice, Knockout , Middle Aged , Mutation , Protein Transport/genetics , Transcriptome , Young Adult
8.
Biochim Biophys Acta ; 1842(11): 2257-65, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25072958

ABSTRACT

The transcription factor NF-κB plays a critical role in the inflammatory response and it has been implicated in various diseases, including non-alcoholic fatty liver disease (NAFLD). Although transient NF-κB activation may protect tissues from stress, a prolonged NF-κB activation can have a detrimental effect on tissue homeostasis and therefore accurate termination is crucial. Copper Metabolism MURR1 Domain-containing 1 (COMMD1), a protein with functions in multiple pathways, has been shown to suppress NF-κB activity. However, its action in controlling liver inflammation has not yet been investigated. To determine the cell-type-specific contribution of Commd1 to liver inflammation, we used hepatocyte and myeloid-specific Commd1-deficient mice. We also used a mouse model of NAFLD to study low-grade chronic liver inflammation: we fed the mice a high fat, high cholesterol (HFC) diet, which results in hepatic lipid accumulation accompanied by liver inflammation. Depletion of hepatocyte Commd1 resulted in elevated levels of the NF-κB transactivation subunit p65 (RelA) but, surprisingly, the level of liver inflammation was not aggravated. In contrast, deficiency of myeloid Commd1 exacerbated diet-induced liver inflammation. Unexpectedly we observed that hepatic and myeloid Commd1 deficiency in the mice both augmented hepatic lipid accumulation. The elevated levels of proinflammatory cytokines in myeloid Commd1-deficient mice might be responsible for the increased level of steatosis. This increase was not seen in hepatocyte Commd1-deficient mice, in which increased lipid accumulation appeared to be independent of inflammation. Our mouse models demonstrate a cell-type-specific role for Commd1 in suppressing liver inflammation and in the progression of NAFLD.

9.
Ann N Y Acad Sci ; 1314: 6-14, 2014 May.
Article in English | MEDLINE | ID: mdl-24697840

ABSTRACT

Copper is an important cofactor in numerous biological processes in all living organisms. However, excessive copper can be extremely toxic, so it is vital that the copper level within a cell is tightly regulated. The damaging effect of copper is seen in several hereditary forms of copper toxicity in humans and animals. At present, Wilson's disease is the best-described and best-studied copper-storage disorder in humans; it is caused by mutations in the ATP7B gene. In dogs, a mutation in the COMMD1 gene has been found to be associated with copper toxicosis. Using a liver-specific Commd1 knockout mouse, the biological role of Commd1 in copper homeostasis has been confirmed. Yet, the exact mechanism by which COMMD1 regulates copper homeostasis is still unknown. Here, we give an overview of the current knowledge and perspectives on the molecular function of COMMD1 in copper homeostasis.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Copper/metabolism , Copper/toxicity , Hepatolenticular Degeneration/metabolism , Liver Cirrhosis/metabolism , Metal Metabolism, Inborn Errors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Animals , Biological Transport , Cation Transport Proteins/genetics , Cation Transport Proteins/metabolism , Copper/blood , Copper-Transporting ATPases , Dogs , Hepatolenticular Degeneration/genetics , Homeostasis/genetics , Humans , Liver Cirrhosis/genetics , Metal Metabolism, Inborn Errors/genetics , Mice , Mice, Knockout
SELECTION OF CITATIONS
SEARCH DETAIL
...