Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
2.
Int J Mol Sci ; 19(7)2018 06 27.
Article in English | MEDLINE | ID: mdl-29954118

ABSTRACT

The H1069Q substitution is the most frequent mutation of the Cu transporter ATP7B that causes Wilson disease in the Caucasian population. ATP7B localizes to the Golgi complex in hepatocytes, but, in the presence of excessive Cu, it relocates to the endo-lysosomal compartment to excrete Cu via bile canaliculi. In contrast, ATP7B-H1069Q is strongly retained in the ER, does not reach the Golgi complex and fails to move to the endo-lysosomal compartment in the presence of excessive Cu, thus causing toxic Cu accumulation. We have previously shown that, in transfected cells, the small heat-shock protein αB-crystallin is able to correct the mislocalization of ATP7B-H1069Q and its trafficking in the presence of Cu overload. Here, we first show that the α-crystallin domain of αB-crystallin mimics the effect of the full-length protein, whereas the N- and C-terminal domains have no such effect. Next, and most importantly, we demonstrate that a twenty-residue peptide derived from the α-crystallin domain of αB-crystallin fully rescues Golgi localization and the trafficking response of ATP7B-H1069Q in the presence of Cu overload. In addition, we show that this peptide interacts with the mutant transporter in the live cell. These results open the way to attempt developing a pharmacologically active peptide to specifically contrast the Wilson disease form caused by the ATP7B-H1069Q mutant.


Subject(s)
Copper-Transporting ATPases/metabolism , Copper/metabolism , Hepatolenticular Degeneration/metabolism , Peptides/chemistry , Peptides/pharmacology , alpha-Crystallin B Chain/chemistry , Animals , COS Cells , Chlorocebus aethiops , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Protein Transport/drug effects , White People
3.
Sci Rep ; 8(1): 6247, 2018 04 19.
Article in English | MEDLINE | ID: mdl-29674751

ABSTRACT

H1069Q substitution represents the most frequent mutation of the copper transporter ATP7B causing Wilson disease in Caucasian population. ATP7B localizes to the Golgi complex in hepatocytes but moves in response to copper overload to the endo-lysosomal compartment to support copper excretion via bile canaliculi. In heterologous or hepatoma-derived cell lines, overexpressed ATP7B-H1069Q is strongly retained in the ER and fails to move to the post-Golgi sites, resulting in toxic copper accumulation. However, this pathogenic mechanism has never been tested in patients' hepatocytes, while animal models recapitulating this form of WD are still lacking. To reach this goal, we have reprogrammed skin fibroblasts of homozygous ATP7B-H1069Q patients into induced pluripotent stem cells and differentiated them into hepatocyte-like cells. Surprisingly, in HLCs we found one third of ATP7B-H1069Q localized in the Golgi complex and able to move to the endo-lysosomal compartment upon copper stimulation. However, despite normal mRNA levels, the expression of the mutant protein was only 20% compared to the control because of endoplasmic reticulum-associated degradation. These results pinpoint rapid degradation as the major cause for loss of ATP7B function in H1069Q patients, and thus as the primary target for designing therapeutic strategies to rescue ATP7B-H1069Q function.


Subject(s)
Copper-Transporting ATPases/genetics , Hepatocytes/pathology , Hepatolenticular Degeneration/genetics , Induced Pluripotent Stem Cells/cytology , Polymorphism, Single Nucleotide , Cell Line , Cellular Reprogramming , Copper/metabolism , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum-Associated Degradation , Fibroblasts/cytology , Golgi Apparatus/metabolism , Hepatolenticular Degeneration/pathology , Humans , Mutant Proteins/metabolism , Skin/cytology
4.
Biochem Biophys Res Commun ; 479(2): 325-330, 2016 Oct 14.
Article in English | MEDLINE | ID: mdl-27641668

ABSTRACT

We have previously shown that αB-crystallin (CRYAB), a small heat shock protein (sHsp) that prevents irreversible aggregation of unfolded protein by an ATP-independent chaperone activity, plays a pivotal role in the biogenesis of multipass transmembrane proteins (TMPs) assisting their folding from the cytosolic side of the endoplasmic reticulum (ER) (D'Agostino et al., 2013). Here we present evidence, based on phosphomimetic substitutions, that the three phosphorytable serine residues at position 19, 45 and 59 of CRYAB play a different regulatory role in this novel chaperone activity: S19 and S45 have a strong inhibitory effect, either alone or in combination, while S59 has not and counteracts the inhibition caused by single phosphomimetic substitutions at S19 and S45. Interestingly, all phosphomimetic substitutions determine the formation of smaller oligomeric complexes containing CRYAB, indicating that the inhibitory effect seen for S19 and S45 cannot be ascribed to the reduction of oligomerization frequently associated to a decreased chaperone activity. These results indicate that phosphorylation finely regulates the chaperone activity of CRYAB with multipass TMPs and suggest a pivotal role for S59 in this process.


Subject(s)
Molecular Chaperones/metabolism , alpha-Crystallin B Chain/metabolism , Cell Line, Tumor , DNA, Complementary/metabolism , Green Fluorescent Proteins/metabolism , Humans , Membrane Proteins/metabolism , Mutagenesis , Mutation , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Folding , Serine/chemistry , Signal Transduction
5.
Nat Chem Biol ; 11(4): 280-6, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25751279

ABSTRACT

Upon binding, ligands can chaperone their protein targets by preventing them from misfolding and aggregating. Thus, an organic molecule that works as folding chaperone for a protein might be its specific ligand, and, similarly, the chaperone potential could represent an alternative readout in a molecular screening campaign toward the identification of new hits. Here we show that small molecules selected for acting as pharmacological chaperones on a misfolded mutant of the Frizzled4 (Fz4) receptor bind and modulate wild-type Fz4, representing what are to our knowledge the first organic ligands of this until-now-undruggable GPCR. The novelty and the advantages of the screening platform, the allosteric binding site addressed by these new ligands and the mechanism they use to modulate Fz4 suggest new avenues for development of inhibitors of the Wnt-ß-catenin pathway and for drug discovery.


Subject(s)
Frizzled Receptors/chemistry , Molecular Chaperones/chemistry , Allosteric Site , Amino Acid Motifs , Base Sequence , Cell Line, Tumor , Chemistry, Pharmaceutical/methods , Dose-Response Relationship, Drug , Drug Design , Glycerol/chemistry , HEK293 Cells , HeLa Cells , Humans , Ligands , Microscopy, Fluorescence , Molecular Sequence Data , Mutagenesis , Protein Binding , Protein Folding , Receptors, G-Protein-Coupled/chemistry
6.
Cell Host Microbe ; 16(3): 338-50, 2014 Sep 10.
Article in English | MEDLINE | ID: mdl-25211076

ABSTRACT

Dugesia japonica planarian flatworms are naturally exposed to various microbes but typically survive this challenge. We show that planarians eliminate bacteria pathogenic to Homo sapiens, Caenorhabditis elegans, and/or Drosophila melanogaster and thus represent a model to identify innate resistance mechanisms. Whole-transcriptome analysis coupled with RNAi screening of worms infected with Staphylococcus aureus or Legionella pneumophila identified 18 resistance genes with nine human orthologs, of which we examined the function of MORN2. Human MORN2 facilitates phagocytosis-mediated restriction of Mycobacterium tuberculosis, L. pneumophila, and S. aureus in macrophages. MORN2 promotes the recruitment of LC3, an autophagy protein also involved in phagocytosis, to M. tuberculosis-containing phagosomes and subsequent maturation to degradative phagolysosomes. MORN2-driven trafficking of M. tuberculosis to single-membrane, LC3-positive compartments requires autophagy-related proteins Atg5 and Beclin-1, but not Ulk-1 and Atg13, highlighting the importance of MORN2 in LC3-associated phagocytosis. These findings underscore the value of studying planarian defenses to identify immune factors.


Subject(s)
Helminth Proteins/immunology , Legionella pneumophila/physiology , Microtubule-Associated Proteins/immunology , Phagocytosis , Planarians/immunology , Planarians/microbiology , Staphylococcus aureus/physiology , Animals , Bacterial Infections/genetics , Bacterial Infections/immunology , Bacterial Infections/microbiology , Disease Models, Animal , Helminth Proteins/genetics , Humans , Legionella pneumophila/immunology , Microtubule-Associated Proteins/genetics , Planarians/genetics , Staphylococcus aureus/immunology
7.
J Membr Biol ; 247(11): 1149-59, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25086772

ABSTRACT

The newly synthesized mutant L501fsX533 Frizzled-4 form and the alpha3beta4 nicotinic acetylcholine receptor expressed in the absence of nicotine accumulate in the endoplasmic reticulum of COS-7 cells and induce the formation of large areas of smooth and highly convoluted cisternae. This results in a generalized block of the transport to the Golgi complex of newly synthesized proteins. Intriguingly, both effects happen peculiarly in COS-7 cells; HeLa, Huh-7, and HEK293 cells expressing the two receptors at similar level than COS-7 cells show normal ER and normal transport toward the plasma membrane. These results question the conclusion that a dominant-negative mechanism would explain the dominance of the mutant L501fsX533 Fz4 allele in the transmission of a form of Familial exudative vitreoretinopathy. Moreover, they indicate that the coordination of endoplasmic reticulum homeostasis in COS-7 cells is particularly error prone. This finding suggests that COS-7 cells may be extremely useful to study the molecular mechanisms regulating endoplasmic reticulum size and architecture.


Subject(s)
Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/ultrastructure , Frizzled Receptors/metabolism , Receptors, Nicotinic/metabolism , Animals , COS Cells , Chlorocebus aethiops , Frizzled Receptors/genetics , HEK293 Cells , HeLa Cells , Humans , Mutation/genetics , Receptors, Nicotinic/genetics
8.
Ophthalmic Genet ; 35(3): 184-6, 2014 Sep.
Article in English | MEDLINE | ID: mdl-23834558

ABSTRACT

Familial exudative vitreoretinopathy (FEVR) is a genetic disease affecting the vascularization of the peripheral retina. The clinical manifestations are very heterogeneous, ranging from mildly affected patients, who could present no visual defects, to severe conditions which can also cause complete blindness at birth or in the first decade. FEVR can be inherited in all the three genetic forms: dominant, recessive and X-linked. To date, four genes have been associated with the condition: TSPAN12. NDP. FDZ4 and LRP5. Interestingly, mutations in TSPAN12 have been considered causative of both a dominant and recessive inheritance and a FEVR phenotype sensitive to the number of TSPAN12 mutations has been supposed. Here we describe a case of a female infant affected by cystic fibrosis and by a severe form of exudative vitreoretinopathy. In particular, we have detected the homozygous missense mutation c.668 T > C in TSPAN12. Neither of the heterozygous parents has ocular manifestations of the disease, suggesting a classic recessive mendelian pattern of inheritance.


Subject(s)
Cystic Fibrosis/genetics , Genes, Recessive , Mutation, Missense , Tetraspanins/genetics , Consanguinity , Eye Diseases, Hereditary , Familial Exudative Vitreoretinopathies , Female , Homozygote , Humans , Infant , Pedigree , Point Mutation , Retinal Diseases/genetics
9.
Sci Rep ; 3: 2659, 2013.
Article in English | MEDLINE | ID: mdl-24036468

ABSTRACT

Frizzled 4 belongs to the superfamily of G protein coupled receptors. The unstructured cytosolic tail of the receptor is essential for its activity. The mutation L501fsX533 in the fz4 gene results in a new COOH-tail of the receptor and causes a form of Familial exudative vitreoretinopathy. Here we show that the mutated tail is structured. Two amphipathic helices, displaying affinity for membranes and resembling the structure of Influenza Hemagglutinin fusion peptide, constitute the new fold. This tail induces the aggregation of the receptor in the Endoplasmic Reticulum and it is sufficient to block the export to the Golgi of a chimeric VSVG protein containing the mutated tail. Affecting the tail's structure, net charge or amphipathicity relocates the mutated Fz4 receptor to the Plasma Membrane. Such disorder-to-order structural transition was never described in GPCRs and opens a new scenario on the possible effect of mutations on unstructured regions of proteins.


Subject(s)
Frizzled Receptors/chemistry , Protein Folding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Familial Exudative Vitreoretinopathies , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Genetic Diseases, X-Linked/genetics , Humans , Molecular Sequence Data , Mutation , Protein Binding , Protein Conformation , Protein Transport , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Sequence Alignment , Solutions , Vitreoretinopathy, Proliferative/genetics
10.
J Cell Sci ; 126(Pt 18): 4160-72, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23843626

ABSTRACT

The α-crystallin B chain (CRYAB or HspB5) is a cytosolic chaperone belonging to the small heat shock protein family, which is known to help in the folding of cytosolic proteins. Here we show that CRYAB binds the mutant form of at least two multispan transmembrane proteins (TMPs), exerting an anti-aggregation activity. It rescues the folding of mutant Frizzled4, which is responsible for a rare autosomal dominant form of familial exudative vitreoretinopathy (Fz4-FEVR), and the mutant ATP7B Cu transporter (ATP7B-H1069Q) associated with a common form of Wilson's disease. In the case of Fz4-FEVR, CRYAB prevents the formation of inter-chain disulfide bridges between the lumenal ectodomains of the aggregated mutant chains, which enables correct folding and promotes appropriate compartmentalization on the plasma membrane. ATP7B-H1069Q, with help from CRYAB, folds into the proper conformation, moves to the Golgi complex, and responds to copper overload in the same manner as wild-type ATP7B. These findings strongly suggest that CRYAB plays a pivotal role, previously undetected, in the folding of multispan TMPs and, from the cytosol, is able to orchestrate folding events that take place in the lumen of the ER. Our results contribute to the explanation of the complex scenario behind multispan TMP folding; additionally, they serve to expose interesting avenues for novel therapeutic approaches.


Subject(s)
Heat-Shock Proteins/genetics , Membrane Proteins/genetics , Molecular Chaperones/chemistry , alpha-Crystallin B Chain/chemistry , Heat-Shock Proteins/metabolism , Molecular Chaperones/metabolism , Protein Conformation , Protein Folding , Transfection , alpha-Crystallin B Chain/physiology
11.
Cell Host Microbe ; 12(6): 751-63, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23245320

ABSTRACT

Variations in lipopolysaccharide (LPS), a bacterial outer membrane component, determine virulence of the obligate intracellular bacterium Coxiella burnetii, but the underlying mechanisms are unknown. We find that while avirulent C. burnetii LPS (avLPS) stimulates host p38α-MAPK signaling required for proper trafficking of bacteria containing compartments to lysosomes for destruction, pathogenic C. burnetii LPS (vLPS) does not. The defect in vLPS and pathogenic C. burnetii targeting to degradative compartments involves an antagonistic engagement of TLR4 by vLPS, lack of p38α-MAPK-driven phosphorylation, and block in recruitment of the homotypic fusion and protein-sorting complex component Vps41 to vLPS-containing vesicles. An upstream activator of p38α-MAPK or phosphomimetic mutant Vps41-S796E expression overrides the inhibition, allowing vLPS and pathogenic C. burnetii targeting to phagolysosomes. Thus, p38α-MAPK and its crosstalk with Vps41 play a central role in trafficking bacteria to phagolysosomes. Pathogenic C. burnetii has evolved LPS variations to evade this host response and thrive intracellularly.


Subject(s)
Coxiella burnetii/immunology , Immune Evasion , Lipopolysaccharides/immunology , Phagosomes/immunology , Toll-Like Receptor 4/antagonists & inhibitors , Vesicular Transport Proteins/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Coxiella burnetii/pathogenicity , Host-Pathogen Interactions , Lipopolysaccharides/metabolism , Phagosomes/microbiology , Vesicular Transport Proteins/immunology , p38 Mitogen-Activated Protein Kinases/immunology
12.
FEMS Immunol Med Microbiol ; 64(1): 98-100, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22066909

ABSTRACT

Q fever is a disease caused by Coxiella burnetii, an obligate intracellular bacterium. Acute Q fever is spontaneously resolutive and is characterized by an efficient immune response. In contrast, chronic Q fever is characterized by dysregulated immune response, as demonstrated by the failure of C. burnetii to induce lymphoproliferation and the lack of granulomas. Recently, it has been demonstrated that when co-expressed in heterologous mammalian cell lines, the ligands of Numb proteins X1 and X2 (LNX1 and LNX2) regulate the level of the T-cell co-receptor CD8, which plays an essential role in T-cell-mediated immune response. We decided to investigate the expression of LNX1 and LNX2 genes in patients with acute or chronic Q fever. Interestingly, we found a high level of LNX1 and LNX2 mRNAs in endocarditis, the principal manifestation of chronic Q fever, but not in acute Q fever. Our data suggest that LNXs may be used as complementary biomarkers to follow the prognosis of chronic Q fever.


Subject(s)
Carrier Proteins/biosynthesis , Coxiella burnetii/pathogenicity , Q Fever/diagnosis , Q Fever/pathology , Ubiquitin-Protein Ligases/biosynthesis , Biomarkers , CD8-Positive T-Lymphocytes/immunology , Gene Expression Profiling , Humans , Prognosis , RNA, Messenger/biosynthesis
13.
J Cell Sci ; 124(Pt 21): 3545-56, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-22045731

ABSTRACT

E3 ubiquitin ligases give specificity to the ubiquitylation process by selectively binding substrates. Recently, their function has emerged as a crucial modulator of T-cell tolerance and immunity. However, substrates, partners and mechanism of action for most E3 ligases remain largely unknown. In this study, we identified the human T-cell co-receptor CD8 α-chain as binding partner of the ligand of Numb proteins X1 (LNX1p80 isoform) and X2 (LNX2). Both LNX mRNAs were found expressed in T cells purified from human blood, and both proteins interacted with CD8α in human HPB-ALL T cells. By using an in vitro assay and a heterologous expression system we showed that the interaction is mediated by the PDZ (PSD95-DlgA-ZO-1) domains of LNX proteins and the cytosolic C-terminal valine motif of CD8α. Moreover, CD8α redistributed LNX1 or LNX2 from the cytosol to the plasma membrane, whereas, remarkably, LNX1 or LNX2 promoted CD8α ubiquitylation, downregulation from the plasma membrane, transport to the lysosomes, and degradation. Our findings highlight the function of LNX proteins as E3 ligases and suggest a mechanism of regulation for CD8α localization at the plasma membrane by ubiquitylation and endocytosis.


Subject(s)
CD8 Antigens/metabolism , Carrier Proteins/metabolism , Endocytosis , Ubiquitin-Protein Ligases/metabolism , CD8 Antigens/chemistry , CD8 Antigens/genetics , Carrier Proteins/chemistry , Carrier Proteins/genetics , Cell Line , Cell Membrane/chemistry , Cell Membrane/genetics , Cell Membrane/metabolism , Humans , PDZ Domains , Protein Binding , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/genetics , Ubiquitination
14.
BMC Res Notes ; 3: 24, 2010 Jan 27.
Article in English | MEDLINE | ID: mdl-20181027

ABSTRACT

BACKGROUND: The importance of non-coding RNAs (ncRNAs) as fine regulators of eukaryotic gene expression has emerged by several studies focusing on microRNAs (miRNAs). miRNAs represent a newly discovered family of non coding-RNAs. They are thought to be crucial players of human hematopoiesis and related tumorigenesis and to represent a potential tool to detect the early stages of cancer. More recently, the expression regulation of numerous long ncRNAs has been linked to cell growth, differentiation and cancer although the molecular mechanism of their function is still unknown.NB4 cells are promyelocytic cells that can be induced to differentiation upon retinoic acid (ATRA) treatment and represent a feasible model to study changes of non coding RNAs expression between cancer cells and their terminally differentiated counterpart. FINDINGS: we screened, by microarray analysis, the expression of 243 miRNAs and 492 human genes transcribing for putative long ncRNAs different from miRNAs in NB4 cells before and after ATRA induced differentiation. Our data show that 8 miRNAs, and 58 long ncRNAs were deregulated by ATRA induced NB4 differentiation. CONCLUSION: our data suggest that ATRA-induced differentiation lead to deregulation of a large number of the ncRNAs that can play regulatory roles in both tumorigenesis and differentiation.

15.
J Biol Chem ; 284(50): 34849-60, 2009 Dec 11.
Article in English | MEDLINE | ID: mdl-19840934

ABSTRACT

The Golgi matrix proteins GRASP65 and GRASP55 have recognized roles in maintaining the architecture of the Golgi complex, in mitotic progression and in unconventional protein secretion whereas, surprisingly, they have been shown to be dispensable for the transport of commonly used reporter cargo proteins along the secretory pathway. However, it is becoming increasingly clear that many trafficking machineries operate in a cargo-specific manner, thus we have investigated whether GRASPs may control the trafficking of selected classes of cargo. We have taken into consideration the C-terminal valine-bearing receptors CD8alpha and Frizzled4 that we show bind directly to the PSD95-DlgA-zo-1 (PDZ) domains of GRASP65 and GRASP55. We demonstrate that both GRASPs are needed sequentially for the efficient transport to and through the Golgi complex of these receptors, thus highlighting a novel role for the GRASPs in membrane trafficking. Our results open new perspectives for our understanding of the regulation of surface expression of a class of membrane proteins, and suggests the causal mechanisms of a dominant form of autosomal human familial exudative vitreoretinopathy that arises from the Frizzled4 mutation involving its C-terminal valine.


Subject(s)
Golgi Apparatus/metabolism , Membrane Proteins/metabolism , Valine/metabolism , Animals , CD8 Antigens/genetics , CD8 Antigens/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Golgi Apparatus/ultrastructure , Golgi Matrix Proteins , Humans , Membrane Proteins/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport/physiology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
16.
Int J Biochem Cell Biol ; 41(12): 2511-21, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19695339

ABSTRACT

In eukaryotic cells several physiologic and pathologic conditions generate the accumulation of unfolded proteins in the endoplasmic reticulum (ER), leading to ER stress. To restore normal function, some ER transmembrane proteins sense the ER stress and activate coordinated signalling pathways collectively called the Unfolded Protein Response (UPR). Little is known on how the UPR relates to post-ER compartments and to the export from the ER of newly synthesized proteins. Here, we report that the ER stress response induced by either thapsigargin or nitric oxide modifies the dynamics of the intracellular distribution of ERGIC-53 and GM130, two markers of the ER Golgi Intermediate Compartment and of the cis-Golgi, respectively. In addition, induction of ER stress alters the morphology of the ERGIC and the Golgi complex and interferes with the reformation of both compartments. Moreover, ER stress rapidly reduces the transport to the Golgi complex of the temperature sensitive mutant of the Vesicular Stomatitis Virus G Glycoprotein (VSV-G) fused with the Green Fluorescent Protein (ts045G), without apparently decreasing the amount of the protein competent for export. Interestingly, a parallel rapid reduction of the number of Sec31 labelled fluorescent puncta on the ER membranes does occur, thus suggesting that the ER stress alters the ER export and the dynamic of post-ER compartments by rapidly targeting the formation of COPII-coated transport intermediates.


Subject(s)
Autoantigens/metabolism , Biomarkers/metabolism , Calcium-Transporting ATPases/antagonists & inhibitors , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Thapsigargin/pharmacology , COP-Coated Vesicles/drug effects , COP-Coated Vesicles/metabolism , Cell Line , Cellular Structures/drug effects , Cellular Structures/metabolism , Cellular Structures/ultrastructure , Endoplasmic Reticulum , Golgi Apparatus , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/ultrastructure , Humans , Membrane Glycoproteins/metabolism , Protein Engineering , Protein Transport/drug effects , Recombinant Fusion Proteins/metabolism , Signal Transduction , Unfolded Protein Response , Viral Envelope Proteins/metabolism
17.
Nucleic Acids Res ; 36(20): 6608-19, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18940871

ABSTRACT

The Epstein-Barr virus (EBV)-encoded latent membrane protein-1 (LMP1), a functional homologue of the tumor necrosis factor receptor family, substantially contributes to EBV's oncogenic potential by activating nuclear factor-kappaB (NF-kappaB). miR-155 is an oncogenic miRNA critical for B-cell maturation and immunoglobulin production in response to antigen. We report that miR-155 expression is much higher in EBV-immortalized B cells than in EBV-negative B cells. LMP1, but not LMP2, up-regulated the expression of miR-155, when transfected in EBV-negative B cells. We analyzed two putative NF-kappaB binding sites in the miR-155 promoter; both sites recruited NF-kappaB complex, in nuclear extract from EBV-immortalized cells. The exogenous expression of LMP1, in EBV-negative background, is temporally correlated to induction of p65 with binding on both NF-kappaB sites and with miR-155 overexpression. The induction of p65 binding together with increased RNA polymerase II binding, confirms that LMP1-mediated activation of miR-155 occurs transcriptionally. In reporter assays, miR-155 promoter lacking NF-kappaB binding sites was no longer activated by LMP1 expression and an intact AP1 site is needed to attain maximum activation. Finally, we demonstrate that LMP1-mediated activation of miR-155 in an EBV-negative background correlates with reduction of protein PU.1, which is a possible miR target.


Subject(s)
B-Lymphocytes/virology , MicroRNAs/genetics , NF-kappa B/metabolism , Transcriptional Activation , Viral Matrix Proteins/metabolism , Animals , Binding Sites , Cell Line , Cells, Cultured , Herpesvirus 4, Human/physiology , Humans , Mice , MicroRNAs/biosynthesis , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transcription Factor RelA/metabolism
18.
J Biol Chem ; 282(31): 22499-512, 2007 Aug 03.
Article in English | MEDLINE | ID: mdl-17535801

ABSTRACT

Accumulation of unfolded proteins within the endoplasmic reticulum (ER) activates the unfolded protein response, also known as the ER stress response. We previously demonstrated that ER stress induces transcription of the ER Golgi intermediate compartment protein ERGIC-53. To investigate the molecular events that regulate unfolded protein response-mediated induction of the gene, we have analyzed the transcriptional regulation of ERGIC-53. We found that the ERGIC-53 promoter contains a single cis-acting element that mediates induction of the gene by thapsigargin and other ER stress-causing agents. This ER stress response element proved to retain a novel structure and to be highly conserved in mammalian ERGIC-53 genes. The ER stress response element identified contains a 5'-end CCAAT sequence that constitutively binds NFY/CBF and, 9 nucleotides away, a 3'-end region (5'-CCCTGTTGGCCATC-3') that is equally important for ER stress-mediated induction of the gene. This sequence is the binding site for endogenous YY1 at the 5'-CCCTGTTGG-3' part and for undefined factors at the CCATC 3'-end. ATF6 alpha-YY1, but not XBP1, interacted with the ERGIC-53 regulatory region and activated ERGIC-53 ER stress response element-dependent transcription. A molecular model for the transcriptional regulation of the ERGIC-53 gene is proposed.


Subject(s)
Endoplasmic Reticulum/metabolism , Mannose-Binding Lectins/physiology , Membrane Proteins/physiology , Transcription, Genetic , Animals , Base Sequence , Binding Sites , HeLa Cells , Humans , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Promoter Regions, Genetic , Protein Denaturation , Protein Folding , Transcriptional Activation
19.
Int J Biochem Cell Biol ; 38(12): 2040-8, 2006.
Article in English | MEDLINE | ID: mdl-16899390

ABSTRACT

The endoplasmic reticulum Golgi intermediate compartment 53 protein recycles continuously between the endoplasmic reticulum and the Golgi complex and ensures the anterograde transport of specific glycoproteins with the assistance of the Multiple Clotting Factor Deficiency adaptor protein. Therefore, to analyze the effect of the endoplasmic reticulum stress on the secretory pathway beyond the endoplasmic reticulum, we analyzed the expression of both proteins in J774 macrophages incubated with the nitric oxide donor DETA NONOate or with thapsigargin. Both proteins accumulated progressively, by a transcriptional mechanism, in response to these inducers. Nitric oxide also induced a higher level of calreticulin and glucose regulated 78 protein, two endoplasmic reticulum proteins controlled by the unfolded protein response. Interestingly, nitric oxide induced the processing of the activating transcription factor 6alpha of the unfolded protein response, while thapsigargin also induced the activation of the transcription factor X-box Binding Protein 1. In addition, we showed that the accumulation of both transporters occurred simultaneously with the activation of endoplasmic reticulum-stress-dependent apoptosis, suggesting that these proteins may participate in the events that will eventually decide the fate of the cell. Induction of endoplasmic reticulum stress affected the rate of anterograde transport of a reporter glycoprotein, indicating that the endoplasmic reticulum to Golgi transport is remarkably impaired. Our results indicate that increased levels of cargo receptor proteins might have a function either in the quality control of protein folding in the endoplasmic reticulum or in the homeostasis of the intermediate compartment and Golgi complex during cell stress.


Subject(s)
Calcium-Binding Proteins/metabolism , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Glycoproteins/metabolism , Golgi Apparatus/drug effects , Mannose-Binding Lectins/metabolism , Membrane Proteins/metabolism , Nitric Oxide/pharmacology , Receptors, Cytoplasmic and Nuclear/metabolism , Vesicular Transport Proteins/metabolism , Activating Transcription Factor 6/metabolism , Animals , Calcium-Binding Proteins/genetics , Cell Death/drug effects , Cells, Cultured , DNA-Binding Proteins/metabolism , Gallic Acid/analogs & derivatives , Gallic Acid/pharmacology , Gene Expression Regulation/drug effects , Humans , Macrophages/drug effects , Mice , Nitric Oxide Donors/pharmacology , Nitroso Compounds/pharmacology , Nuclear Proteins/metabolism , Protein Transport/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Regulatory Factor X Transcription Factors , Thapsigargin/pharmacology , Time Factors , Transcription Factors , Vesicular Transport Proteins/genetics
20.
J Biol Chem ; 281(1): 418-28, 2006 Jan 06.
Article in English | MEDLINE | ID: mdl-16260777

ABSTRACT

KCNQ2 and KCNQ3 K+ channel subunits underlie the muscarinic-regulated K+ current (I(KM)), a widespread regulator of neuronal excitability. Mutations in KCNQ2- or KCNQ3-encoding genes cause benign familiar neonatal convulsions (BFNCs), a rare autosomal-dominant idiopathic epilepsy of the newborn. In the present study, we have investigated, by means of electrophysiological, biochemical, and immunocytochemical techniques in transiently transfected cells, the consequences prompted by a BFNC-causing 1-bp deletion (2043deltaT) in the KCNQ2 gene; this frameshift mutation caused the substitution of the last 163 amino acids of the KCNQ2 C terminus and the extension of the subunit by additional 56 residues. The 2043deltaT mutation abolished voltage-gated K+ currents produced upon homomeric expression of KCNQ2 subunits, dramatically reduced the steady-state cellular levels of KCNQ2 subunits, and prevented their delivery to the plasma membrane. Metabolic labeling experiments revealed that mutant KCNQ2 subunits underwent faster degradation; 10-h treatment with the proteasomal inhibitor MG132 (20 microm) at least partially reversed such enhanced degradation. Co-expression with KCNQ3 subunits reduced the degradation rate of mutant KCNQ2 subunits and led to their expression on the plasma membrane. Finally, co-expression of KCNQ2 2043deltaT together with KCNQ3 subunits generated functional voltage-gated K+ currents having pharmacological and biophysical properties of heteromeric channels. Collectively, the present results suggest that mutation-induced reduced stability of KCNQ2 subunits may cause epilepsy in neonates.


Subject(s)
Epilepsy, Benign Neonatal/genetics , KCNQ2 Potassium Channel/genetics , KCNQ2 Potassium Channel/physiology , Protein Subunits/physiology , Animals , CHO Cells , Carcinoma, Hepatocellular , Cell Line, Tumor , Cell Membrane/physiology , Cricetinae , Epilepsy, Benign Neonatal/physiopathology , Frameshift Mutation , Green Fluorescent Proteins/genetics , Humans , Infant, Newborn , KCNQ2 Potassium Channel/chemistry , KCNQ3 Potassium Channel/chemistry , KCNQ3 Potassium Channel/genetics , KCNQ3 Potassium Channel/physiology , Liver Neoplasms , Mutagenesis , Patch-Clamp Techniques , Protein Subunits/chemistry , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...