Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Mol Cancer ; 23(1): 21, 2024 01 23.
Article in English | MEDLINE | ID: mdl-38263180

ABSTRACT

BACKGROUND: The ATM kinase constitutes a master regulatory hub of DNA damage and activates the p53 response pathway by phosphorylating the MDM2 protein, which develops an affinity for the p53 mRNA secondary structure. Disruption of this interaction prevents the activation of the nascent p53. The link of the MDM2 protein-p53 mRNA interaction with the upstream DNA damage sensor ATM kinase and the role of the p53 mRNA in the DNA damage sensing mechanism, are still highly anticipated. METHODS: The proximity ligation assay (PLA) has been extensively used to reveal the sub-cellular localisation of the protein-mRNA and protein-protein interactions. ELISA and co-immunoprecipitation confirmed the interactions in vitro and in cells. RESULTS: This study provides a novel mechanism whereby the p53 mRNA interacts with the ATM kinase enzyme and shows that the L22L synonymous mutant, known to alter the secondary structure of the p53 mRNA, prevents the interaction. The relevant mechanistic roles in the DNA Damage Sensing pathway, which is linked to downstream DNA damage response, are explored. Following DNA damage (double-stranded DNA breaks activating ATM), activated MDMX protein competes the ATM-p53 mRNA interaction and prevents the association of the p53 mRNA with NBS1 (MRN complex). These data also reveal the binding domains and the phosphorylation events on ATM that regulate the interaction and the trafficking of the complex to the cytoplasm. CONCLUSION: The presented model shows a novel interaction of ATM with the p53 mRNA and describes the link between DNA Damage Sensing with the downstream p53 activation pathways; supporting the rising functional implications of synonymous mutations altering secondary mRNA structures.


Subject(s)
Polynucleotide 5'-Hydroxyl-Kinase , Proto-Oncogene Proteins c-mdm2 , Humans , Tumor Suppressor Protein p53 , DNA Damage , DNA Repair , Ataxia Telangiectasia Mutated Proteins
2.
Nucleic Acids Res ; 50(17): 10110-10122, 2022 09 23.
Article in English | MEDLINE | ID: mdl-36107769

ABSTRACT

Protein aggregates and abnormal proteins are toxic and associated with neurodegenerative diseases. There are several mechanisms to help cells get rid of aggregates but little is known on how cells prevent aggregate-prone proteins from being synthesised. The EBNA1 of the Epstein-Barr virus (EBV) evades the immune system by suppressing its own mRNA translation initiation in order to minimize the production of antigenic peptides for the major histocompatibility (MHC) class I pathway. Here we show that the emerging peptide of the disordered glycine-alanine repeat (GAr) within EBNA1 dislodges the nascent polypeptide-associated complex (NAC) from the ribosome. This results in the recruitment of nucleolin to the GAr-encoding mRNA and suppression of mRNA translation initiation in cis. Suppressing NAC alpha (NACA) expression prevents nucleolin from binding to the GAr mRNA and overcomes GAr-mediated translation inhibition. Taken together, these observations suggest that EBNA1 exploits a nascent protein quality control pathway to regulate its own rate of synthesis that is based on sensing the nascent GAr peptide by NAC followed by the recruitment of nucleolin to the GAr-encoding RNA sequence.


Subject(s)
Epstein-Barr Virus Infections , Herpesvirus 4, Human , RNA-Binding Proteins/metabolism , Alanine , Epstein-Barr Virus Nuclear Antigens/metabolism , Glycine , Herpesvirus 4, Human/genetics , Humans , Peptides/genetics , Phosphoproteins , Protein Aggregates , RNA, Messenger/genetics , RNA, Messenger/metabolism , Nucleolin
3.
Life Sci Alliance ; 5(2)2022 02.
Article in English | MEDLINE | ID: mdl-34785537

ABSTRACT

The role of G-quadruplex (G4) RNA structures is multifaceted and controversial. Here, we have used as a model the EBV-encoded EBNA1 and the Kaposi's sarcoma-associated herpesvirus (KSHV)-encoded LANA1 mRNAs. We have compared the G4s in these two messages in terms of nucleolin binding, nuclear mRNA retention, and mRNA translation inhibition and their effects on immune evasion. The G4s in the EBNA1 message are clustered in one repeat sequence and the G4 ligand PhenDH2 prevents all G4-associated activities. The RNA G4s in the LANA1 message take part in similar multiple mRNA functions but are spread throughout the message. The different G4 activities depend on flanking coding and non-coding sequences and, interestingly, can be separated individually. Together, the results illustrate the multifunctional, dynamic and context-dependent nature of G4 RNAs and highlight the possibility to develop ligands targeting specific RNA G4 functions. The data also suggest a common multifunctional repertoire of viral G4 RNA activities for immune evasion.


Subject(s)
DNA, Intergenic/chemistry , DNA, Intergenic/genetics , G-Quadruplexes , RNA/chemistry , RNA/genetics , Epstein-Barr Virus Nuclear Antigens/chemistry , Epstein-Barr Virus Nuclear Antigens/genetics , Gene Expression Regulation , Humans , RNA Transport , RNA, Viral
4.
Nucleic Acids Res ; 48(12): 6775-6787, 2020 07 09.
Article in English | MEDLINE | ID: mdl-32453417

ABSTRACT

Cell growth requires a high level of protein synthesis and oncogenic pathways stimulate cell proliferation and ribosome biogenesis. Less is known about how cells respond to dysfunctional mRNA translation and how this feeds back into growth regulatory pathways. The Epstein-Barr virus (EBV)-encoded EBNA1 causes mRNA translation stress in cis that activates PI3Kδ. This leads to the stabilization of MDM2, induces MDM2's binding to the E2F1 mRNA and promotes E2F1 translation. The MDM2 serine 166 regulates the interaction with the E2F1 mRNA and deletion of MDM2 C-terminal RING domain results in a constitutive E2F1 mRNA binding. Phosphorylation on serine 395 following DNA damage instead regulates p53 mRNA binding to its RING domain and prevents the E2F1 mRNA interaction. The p14Arf tumour suppressor binds MDM2 and in addition to preventing degradation of the p53 protein it also prevents the E2F1 mRNA interaction. The data illustrate how two MDM2 domains selectively bind specific mRNAs in response to cellular conditions to promote, or suppress, cell growth and how p14Arf coordinates MDM2's activity towards p53 and E2F1. The data also show how EBV via EBNA1-induced mRNA translation stress targets the E2F1 and the MDM2 - p53 pathway.


Subject(s)
E2F1 Transcription Factor/genetics , Neoplasms/genetics , Proto-Oncogene Proteins c-mdm2/genetics , Tumor Suppressor Protein p53/genetics , Carcinogenesis/genetics , Cell Cycle/genetics , Cell Proliferation/genetics , DNA Damage/genetics , Genes, Tumor Suppressor , Herpesvirus 4, Human/genetics , Humans , Neoplasms/virology , Oncogenes/genetics , Phosphorylation/genetics , Protein Domains/genetics , RNA Recognition Motif Proteins/genetics , RNA, Messenger/genetics , Tumor Suppressor Protein p14ARF/genetics
5.
Nucleic Acids Res ; 47(6): 3086-3100, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30624716

ABSTRACT

Peptides presented on major histocompatibility (MHC) class I molecules form an essential part of the immune system's capacity to detect virus-infected or transformed cells. Earlier works have shown that pioneer translation peptides (PTPs) for the MHC class I pathway are as efficiently produced from introns as from exons, or from mRNAs targeted for the nonsense-mediated decay pathway. The production of PTPs is a target for viral immune evasion but the underlying molecular mechanisms that govern this non-canonical translation are unknown. Here, we have used different approaches to show how events taking place on the nascent transcript control the synthesis of PTPs and full-length proteins. By controlling the subcellular interaction between the G-quadruplex structure (G4) of a gly-ala encoding mRNA and nucleolin (NCL) and by interfering with mRNA maturation using multiple approaches, we demonstrate that antigenic peptides derive from a nuclear non-canonical translation event that is independently regulated from the synthesis of full-length proteins. Moreover, we show that G4 are exploited to control mRNA localization and translation by distinguishable mechanisms that are targets for viral immune evasion.


Subject(s)
Antigens/genetics , Histocompatibility Antigens Class I/genetics , Peptides/genetics , Protein Biosynthesis/genetics , Antigens/immunology , Cell Nucleus/genetics , Cell Nucleus/immunology , G-Quadruplexes , Histocompatibility Antigens Class I/immunology , Humans , Immune Evasion/genetics , Immune Evasion/immunology , Nonsense Mediated mRNA Decay/genetics , Nonsense Mediated mRNA Decay/immunology , Peptides/immunology , Protein Biosynthesis/immunology , RNA, Messenger/genetics , RNA, Messenger/immunology
6.
Cell Death Differ ; 24(10): 1717-1729, 2017 10.
Article in English | MEDLINE | ID: mdl-28622297

ABSTRACT

Physiological and pathological conditions that affect the folding capacity of the endoplasmic reticulum (ER) provoke ER stress and trigger the unfolded protein response (UPR). The UPR aims to either restore the balance between newly synthesized and misfolded proteins or if the damage is severe, to trigger cell death. However, the molecular events underlying the switch between repair and cell death are not well understood. The ER-resident chaperone BiP governs the UPR by sensing misfolded proteins and thereby releasing and activating the three mediators of the UPR: PERK, IRE1 and ATF6. PERK promotes G2 cell cycle arrest and cellular repair by inducing the alternative translated p53 isoform p53ΔN40 (p53/47), which activates 14-3-3σ via suppression of p21CDKN1A. Here we show that prolonged ER stress promotes apoptosis via a p53-dependent inhibition of BiP expression. This leads to the release of the pro-apoptotic BH3-only BIK from BiP and activation of apoptosis. Suppression of bip mRNA translation is mediated via the specific binding of p53 to the first 346-nt of the bip mRNA and via a p53 trans-suppression domain located within the first seven N-terminal amino acids of p53ΔN40. This work shows how p53 targets BiP to promote apoptosis during severe ER stress and further illustrates how regulation of mRNA translation has a key role in p53-mediated regulation of gene expression during the UPR.


Subject(s)
Apoptosis/physiology , Endoplasmic Reticulum Stress/physiology , Heat-Shock Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/metabolism , Humans , Mitochondrial Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Tumor Suppressor Protein p53/genetics , Unfolded Protein Response/physiology
7.
Cell Cycle ; 14(3): 449-58, 2015.
Article in English | MEDLINE | ID: mdl-25659040

ABSTRACT

Isoforms derived from alternative splicing, mRNA translation initiation or promoter usage extend the functional repertoire of the p53, p63 and p73 genes family and of their regulators MDM2 and MDMX. Here we show cap-independent translation of an N-terminal truncated isoform of hMDMX, hMDMX(p60), which is initiated at the 7th AUG codon downstream of the initiation site for full length hMDMX(FL) at position +384. hMDMX(p60) lacks the p53 binding motif but retains the RING domain and interacts with hMDM2 and hMDMX(FL). hMDMX(p60) shows higher affinity for hMDM2, as compared to hMDMX(FL). In vitro data reveal a positive cooperative interaction between hMDMX(p60) and hMDM2 and in cellulo data show that low levels of hMDMX(p60) promote degradation of hMDM2 whereas higher levels stabilize hMDM2 and prevent hMDM2-mediated degradation of hMDMX(FL). These results describe a novel alternatively translated hMDMX isoform that exhibits unique regulatory activity toward hMDM2 autoubiquitination. The data illustrate how the N-terminus of hMDMX regulates its C-terminal RING domain and the hMDM2 activity.


Subject(s)
Alternative Splicing , Nuclear Proteins/metabolism , Protein Biosynthesis , Proto-Oncogene Proteins c-mdm2/metabolism , Proto-Oncogene Proteins/metabolism , Cell Cycle Proteins , Cell Line , Codon/genetics , Humans , Protein Binding , Protein Isoforms/metabolism , Protein Stability , Proteolysis , RNA Caps/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
8.
Mol Cell ; 54(3): 500-11, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24813712

ABSTRACT

Regulated protein synthesis via changes in mRNA structures forms an important part of how prokaryotic cells adapt protein expression in response to changes in the environment. Little is known regarding how this concept has adapted to regulate mRNA translation via signaling pathways in mammalian cells. Here, we show that following phosphorylation by the ataxia telangiectasia mutated (ATM) kinase at serine 403, the C-terminal RING domain of HDMX binds the nascent p53 mRNA to promote a conformation that supports the p53 mRNA-HDM2 interaction and the induction of p53 synthesis. HDMX and its homolog HDM2 bind the same p53 internal ribosome entry sequences (IRES) structure but with different specificity and function. The results show how HDMX and HDM2 act as nonredundant IRES trans-acting factors (ITAFs) to bring a positive synergistic effect on p53 expression during genotoxic stress by first altering the structure of the newly synthesized p53 mRNA followed by stimulation of translation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , Nuclear Proteins/physiology , Proto-Oncogene Proteins/physiology , RNA, Messenger/genetics , Tumor Suppressor Protein p53/genetics , Animals , Base Sequence , Cell Cycle Proteins , DNA Damage , Gene Expression Regulation , Humans , Inverted Repeat Sequences , Molecular Sequence Data , Nuclear Proteins/chemistry , Phosphorylation , Protein Binding , Protein Biosynthesis , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins c-mdm2/metabolism , RNA Folding , RNA Processing, Post-Transcriptional , RNA, Messenger/chemistry , RNA, Messenger/metabolism , Sf9 Cells , Spodoptera , Substrate Specificity , Tumor Suppressor Protein p53/metabolism
9.
Cancer Cell ; 21(1): 25-35, 2012 Jan 17.
Article in English | MEDLINE | ID: mdl-22264786

ABSTRACT

The ATM kinase and p53 are key tumor suppressor factors that control the genotoxic stress response pathway. The ATM substrate Mdm2 controls p53 activity by either targeting p53 for degradation or promoting its synthesis by binding the p53 mRNA. The physiological role and regulation of Mdm2's dual function toward p53 is not known. Here we show that ATM-dependent phosphorylation of Mdm2 at Ser395 is required for the p53 mRNA-Mdm2 interaction. This event also promotes SUMO-conjugation of Mdm2 and its nucleoli accumulation. Interfering with the p53 mRNA-Mdm2 interaction prevents p53 stabilization and activation following DNA damage. These results demonstrate how ATM activity switches Mdm2 from a negative to a positive regulator of p53 via the p53 mRNA.


Subject(s)
DNA Damage , Gene Expression Regulation , Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/physiology , Active Transport, Cell Nucleus , Cell Line, Tumor , Humans , Phosphorylation , RNA, Messenger/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
10.
J Virol ; 83(3): 1289-98, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19019958

ABSTRACT

The glycine-alanine repeat (GAr) sequence of the Epstein-Barr virus-encoded EBNA-1 prevents presentation of antigenic peptides to major histocompatibility complex class I molecules. This has been attributed to its capacity to suppress mRNA translation in cis. However, the underlying mechanism of this function remains largely unknown. Here, we have further investigated the effect of the GAr as a regulator of mRNA translation. Introduction of silent mutations in each codon of a 30-amino-acid GAr sequence does not significantly affect the translation-inhibitory capacity, whereas minimal alterations in the amino acid composition have strong effects, which underscores the observation that the amino acid sequence and not the mRNA sequence mediates GAr-dependent translation suppression. The capacity of the GAr to repress translation is dose and position dependent and leads to a relative accumulation of preinitiation complexes on the mRNA. Taken together with the surprising observation that fusion of the 5' untranslated region (UTR) of the c-myc mRNA to the 5' UTR of GAr-carrying mRNAs specifically inactivates the effect of the GAr, these results indicate that the GAr targets components of the translation initiation process. We propose a model in which the nascent GAr peptide delays the assembly of the initiation complex on its own mRNA.


Subject(s)
Alanine/chemistry , Epstein-Barr Virus Nuclear Antigens/chemistry , Glycine/chemistry , Protein Biosynthesis , RNA, Messenger/genetics , Repetitive Sequences, Amino Acid , 5' Untranslated Regions , Base Sequence , Cell Line, Tumor , DNA Primers , Epstein-Barr Virus Nuclear Antigens/genetics , Humans
11.
Cell Cycle ; 8(1): 31-4, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19106616

ABSTRACT

The E3 ligase Mdm2 is a key regulator of p53 activity via a complex regulatory feedback system that involves all levels of expression control including transcription, mRNA translation and protein degradation. Best known is the effect of p53 on Mdm2 transcription and the capacity of Mdm2 to target p53 for degradation, but more recently the role of Mdm2 as a positive regulator of p53 activity has also started to emerge. Mdm2 stimulates p53 mRNA translation by binding the p53 mRNA and, interestingly, this interaction also suppresses Mdm2's capacity to promote p53 polyubiquitination and degradation. Another interesting aspect of the p53 mRNA-Mdm2 interaction is that the p53 mRNA sequence encoding the amino acids which bind the N-terminus of Mdm2 is the same that interacts with the Mdm2 RING domain. Indeed, the regulatory elements for controlling Mdm2-dependent expression of p53 are derived from the same p53 genomic sequence. In addition, the RNA binding and the E3 ligase domain of Mdm2 overlap, indicati that the two functions of Mdm2 to control p53 synthesis and degradation have co-evolved in parallel in both p53 and Mdm2. Here we illustrate how the p53-Mdm2 protein-protein and p53 mRNA-Mdm2 interactions affect Mdm2-mediated control of p53 expression using the Phe19Ala p53 mutant. We discuss how the new insights into the regulation of p53 expression levels can help to shed light on the origin of this elegant feedback system and on the function of Mdm2 isoforms.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Base Sequence , Cell Line, Tumor , Humans , Models, Biological , Molecular Sequence Data , Mutation/genetics , Nucleic Acid Conformation , Protein Binding , Protein Biosynthesis , RNA, Messenger/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Nat Cell Biol ; 10(9): 1098-105, 2008 Sep.
Article in English | MEDLINE | ID: mdl-19160491

ABSTRACT

The E3 ubiquitin ligase Mdm2 is a focal regulator of p53 tumour suppressor activity. It binds p53, promoting its polyubiquitination and degradation, and also controls p53 synthesis. However, it is not known how this dual function of Mdm2 on p53 synthesis and degradation is achieved. Here we show that the p53 mRNA region encoding the Mdm2-binding site interacts directly with the RING domain of Mdm2. This impairs the E3 ligase activity of Mdm2 and promotes p53 mRNA translation. We also show that introduction of cancer-derived single silent point-mutations in the p53 mRNA weakens its binding to Mdm2 and results in reduced p53 activity. These data are consistent with a mechanism by which changes in silent nucleotides can affect the function of the encoded protein, and indicate that Mdm2-mediated control of p53 synthesis and degradation has evolved in the p53 mRNA sequence and its encoded amino acids.


Subject(s)
Proto-Oncogene Proteins c-mdm2/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Base Sequence , Gene Expression Regulation , Humans , Models, Biological , Protein Binding , Protein Biosynthesis , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Ubiquitin-Protein Ligases/metabolism
13.
Am J Respir Cell Mol Biol ; 35(5): 519-27, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16741163

ABSTRACT

Alveolar hypoxia, a common feature of many respiratory disorders, has been previously reported to induce functional changes, particularly a decrease of transepithelial Na and fluid transport. In polarized epithelia, cytoskeleton plays a regulatory role in transcellular and paracellular transport of ions and fluid. We hypothesized that exposure to hypoxia could damage cytoskeleton organization, which in turn, may adversely affect ion and fluid transport. Primary rat alveolar epithelial cells (AEC) were exposed to either mild (3% O(2)) or severe (0.5% O(2)) hypoxia for 18 h or to normoxia (21% O(2)). First, mild and severe hypoxia induced a disorganization of actin, a major protein of the cytoskeleton, reflected by disruption of F-actin filaments. Second, alpha-spectrin, an apical cytoskeleton protein, which binds to actin cytoskeleton and Na transport proteins, was cleaved by hypoxia. Pretreatment of AEC by a caspase inhibitor (z-VAD-fmk; 90 microM) blunted hypoxia-induced spectrin cleavage as well as hypoxia-induced decrease in surface membrane alpha-ENaC and concomitantly induced a partial recovery of hypoxia-induced decrease of amiloride-sensitive Na transport at 3% O(2). Finally, tight junctions (TJs) proteins, which are linked to actin and are a determinant of paracellular permeability, were altered by mild and severe hypoxia: hypoxia induced a mislocalization of occludin from the TJ to cytoplasm and a decrease in zonula occludens-1 protein level. These modifications were associated with modest changes in paracellular permeability at 0.5% O(2,) as assessed by small 4-kD dextran flux and transepithelial resistance measurements. Together, these findings indicate that hypoxia disrupted cytoskeleton and TJ organization in AEC and may participate, at least in part, to hypoxia-induced decrease in Na transport.


Subject(s)
Cytoskeleton/metabolism , Epithelial Cells , Hypoxia , Pulmonary Alveoli/cytology , Actins/metabolism , Amino Acid Chloromethyl Ketones/metabolism , Animals , Calpain/antagonists & inhibitors , Cell Membrane/metabolism , Cell Polarity , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/metabolism , Epithelial Sodium Channels/metabolism , Male , Membrane Proteins/metabolism , Occludin , Patch-Clamp Techniques , Permeability , Phosphoproteins/metabolism , Rats , Rats, Sprague-Dawley , Spectrin/metabolism , Tight Junctions/metabolism , Zonula Occludens-1 Protein
14.
Am J Physiol Renal Physiol ; 287(3): F550-61, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15140763

ABSTRACT

The epithelial Na(+) channel (ENaC) is regulated by the ubiquitin-protein ligase Nedd4-2 via interaction with ENaC PY-motifs. These PY-motifs are mutated/deleted in Liddle's syndrome, resulting in elevated Na(+) reabsorption and hypertension explained partly by impaired ENaC-Nedd4-2 interaction. We hypothesized that Nedd4-2 is a susceptibility gene for hypertension and screened 856 renal patients and healthy controls for mutations in a subset of exons of the human Nedd4-2 gene that are relevant for ENaC regulation by PCR/single-strand conformational polymorphism. Several variants were identified, and one nonsynonymous mutation (Nedd4-2-P355L) was further characterized. This mutation next to the 3' donor site of exon 15 does not affect in vitro splicing of Nedd4-2 mRNA. However, in the Xenopus oocyte expression system, Nedd4-2-P355L-dependent ENaC inhibition was weaker compared with the wild type (Nedd4-2-WT), and this difference depended on the presence of intact PY-motifs on ENaC. This could not be explained by the amount of wild type or mutant Nedd4-2 coimmunoprecipitating with ENaC. When the phosphorylation level of human Nedd4-2 Ser(448) (known to be phosphorylated by the Sgk1 kinase) was determined with a specific anti-pSer(448) antibody, we observed stronger basal phosphorylation of Nedd4-2-P355L. Both the phosphorylation level and the accompanying amiloride-sensitive Na(+) currents could be further enhanced to approximately the same levels by coexpressing Sgk1. In addition, the role of the two other putative Sgk1 phosphorylation sites (S342 and T367) appears also to be affected by the P355L mutation. The differential phosphorylation status between wild-type and mutant Nedd4-2 provides an explanation for the different potential to inhibit ENaC activity.


Subject(s)
Kidney Failure, Chronic/genetics , Nuclear Proteins , Sodium Channels/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Animals , Down-Regulation , Endosomal Sorting Complexes Required for Transport , Epithelial Sodium Channels , Female , Homeostasis , Humans , Immediate-Early Proteins , Kidney Failure, Chronic/metabolism , Male , Middle Aged , Nedd4 Ubiquitin Protein Ligases , Oocytes , Phosphorylation , Point Mutation , Polymorphism, Single-Stranded Conformational , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary , RNA Splicing , Serine/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases/chemistry , Xenopus Proteins , Xenopus laevis
15.
C R Biol ; 326(7): 615-24, 2003 Jul.
Article in English | MEDLINE | ID: mdl-14556380

ABSTRACT

A fine regulation of the amiloride-sensitive Epithelial Sodium Channel (ENaC), made of alpha, beta and gamma subunits, is crucial for maintenance of Na+ balance and blood pressure. Both beta- and gamma-ENaC participate in negative regulation by interacting with Nedd4-2, an E3 ubiquitin-ligase. Disruption of this interaction results in increased ENaC activity (Liddle syndrome). By two-hybrid screenings, we identified new potential partners of alpha-ENaC: WWP1 (E3 ubiquitin-ligase protein), UBC9 and TSG101 (E2 ubiquitin/SUMO-conjugating enzymes) and confirmed these interactions in GST pull-down assays. All these partners are implicated in protein trafficking and could be involved in the regulation of ENaC activity.


Subject(s)
Sodium Channels/analysis , Amino Acid Sequence , Epithelial Sodium Channels , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Sodium Channels/physiology
16.
Pflugers Arch ; 447(1): 35-43, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12908109

ABSTRACT

Ubiquitination of proteins such as ion transporters appears to be an important process in the regulation of their membrane expression. Recently, using two-hybrid screening, we have selected a potential partner for the alpha-subunit of the amiloride-sensitive epithelial sodium channel (ENaC): the WWP1 protein, a ubiquitin ligase belonging to the Nedd4 family. To establish whether WWP1 is co-expressed with ENaC, we employed in situ hybridisation, immunohistochemistry and Western blotting to determine the expression of WWP1 in various tissues and cell lines, including those known to express ENaC. As expected, WWP1 was expressed, like ENaC, in the bronchiolar epithelium. However it was also present in the proximal colon and the proximal part of the nephron (where ENaC is not expressed) and absent in the distal parts of the nephron (where ENaC is expressed abundantly). These results suggest that other channels or transport proteins, containing specific domains, such as PY motifs, could be the targets for regulation by WWP1.


Subject(s)
Epithelial Cells/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Animals , Base Sequence , COS Cells , Cell Line , Chlorocebus aethiops , Endosomal Sorting Complexes Required for Transport , Female , Gene Expression Regulation/physiology , Humans , LLC-PK1 Cells , Male , Molecular Sequence Data , Nedd4 Ubiquitin Protein Ligases , Rats , Rats, Sprague-Dawley , Swine , Ubiquitin-Protein Ligases/genetics , Xenopus , Xenopus Proteins
17.
Mol Cell Biol ; 22(10): 3527-36, 2002 May.
Article in English | MEDLINE | ID: mdl-11971983

ABSTRACT

Spectrins, components of the membrane skeleton, are implicated in various cellular functions. Understanding the diversity of these functions requires better characterization of the interacting domains of spectrins, such as the SH3 domain. Yeast two-hybrid screening of a kidney cDNA library revealed that the SH3 domain of alpha II-spectrin binds specifically isoform A of low-molecular-weight phosphotyrosine phosphatase (LMW-PTP). The alpha II-spectrin SH3 domain does not interact with LMW-PTP B or C nor does LMW-PTP A interact with the alpha I-spectrin SH3 domain. The interaction of spectrin with LMW-PTP A led us to look for a tyrosine-phosphorylated residue in alpha II-spectrin. Western blotting showed that alpha II-spectrin is tyrosine phosphorylated in vivo. Using mutagenesis on recombinant peptides, we identified the residue Y1176 located in the calpain cleavage site of alpha II-spectrin, near the SH3 domain, as an in vitro substrate for Src kinase and LMW-PTP A. This Y1176 residue is also an in vivo target for kinases and phosphatases in COS cells. Phosphorylation of this residue decreases spectrin sensitivity to calpain in vitro. Similarly, the presence of phosphatase inhibitors in cell culture is associated with the absence of spectrin cleavage products. This suggests that the Y1176 phosphorylation state could modulate spectrin cleavage by calpain and may play an important role during membrane skeleton remodeling.


Subject(s)
Calpain/metabolism , Protein Tyrosine Phosphatases/metabolism , Spectrin/metabolism , Tyrosine/metabolism , Amino Acid Sequence , Animals , Cell Fractionation , Cell Line , Cytoskeleton/metabolism , Male , Molecular Sequence Data , Phosphorylation , Protein Isoforms , Protein Structure, Tertiary , Protein Tyrosine Phosphatases/chemistry , Protein Tyrosine Phosphatases/genetics , Rats , Rats, Sprague-Dawley , Sequence Alignment , Two-Hybrid System Techniques , Yeasts/genetics , Yeasts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...