Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Biol Chem ; 295(49): 16630-16642, 2020 12 04.
Article in English | MEDLINE | ID: mdl-32967969

ABSTRACT

The glucagon receptor (GCGR) activated by the peptide hormone glucagon is a seven-transmembrane G protein-coupled receptor (GPCR) that regulates blood glucose levels. Ubiquitination influences trafficking and signaling of many GPCRs, but its characterization for the GCGR is lacking. Using endocytic colocalization and ubiquitination assays, we have identified a correlation between the ubiquitination profile and recycling of the GCGR. Our experiments revealed that GCGRs are constitutively ubiquitinated at the cell surface. Glucagon stimulation not only promoted GCGR endocytic trafficking through Rab5a early endosomes and Rab4a recycling endosomes, but also induced rapid deubiquitination of GCGRs. Inhibiting GCGR internalization or disrupting endocytic trafficking prevented agonist-induced deubiquitination of the GCGR. Furthermore, a Rab4a dominant negative (DN) that blocks trafficking at recycling endosomes enabled GCGR deubiquitination, whereas a Rab5a DN that blocks trafficking at early endosomes eliminated agonist-induced GCGR deubiquitination. By down-regulating candidate deubiquitinases that are either linked with GPCR trafficking or localized on endosomes, we identified signal-transducing adaptor molecule-binding protein (STAMBP) and ubiquitin-specific protease 33 (USP33) as cognate deubiquitinases for the GCGR. Our data suggest that USP33 constitutively deubiquitinates the GCGR, whereas both STAMBP and USP33 deubiquitinate agonist-activated GCGRs at early endosomes. A mutant GCGR with all five intracellular lysines altered to arginines remains deubiquitinated and shows augmented trafficking to Rab4a recycling endosomes compared with the WT, thus affirming the role of deubiquitination in GCGR recycling. We conclude that the GCGRs are rapidly deubiquitinated after agonist-activation to facilitate Rab4a-dependent recycling and that USP33 and STAMBP activities are critical for the endocytic recycling of the GCGR.


Subject(s)
Deubiquitinating Enzymes/metabolism , Endosomes/metabolism , Receptors, Glucagon/metabolism , rab4 GTP-Binding Proteins/metabolism , Cell Line , Down-Regulation , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/genetics , Endosomal Sorting Complexes Required for Transport/metabolism , Glucagon/pharmacology , Humans , Monensin/pharmacology , Mutagenesis , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Glucagon/agonists , Receptors, Glucagon/genetics , Ubiquitin Thiolesterase/antagonists & inhibitors , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitination/drug effects , rab4 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
2.
Anim Genet ; 52(3): 275-283, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33709423

ABSTRACT

The Wagyu breed of taurine cattle possess favourable genetics for intramuscular fat (IMF) but genomic loci associated with the trait remain under characterised. Here, we report the identification of a previously unidentified genomic region possessing a particular haplotype structure in Wagyu. Through deployment of a genome-wide haplotype detection analysis that captures regions conserved in a target population but not other populations we screened 100 individual Wagyu and contrasted them with 100 individuals from two independent comparison breeds, Charolais and Angus, using high-density SNPs. An extreme level of Wagyu conservation was assigned to a single genomic window (spanning genomic coordinates BTA28:41 088-300 265 bp). In fact, a five-SNP region spanning 27 096 bp is almost perfectly conserved among the 100 Wagyu individuals assayed and partially overlaps RAB4A. Focussing in, two consecutive SNPs (genomic coordinates 236 949 and 239 950) are apparently fixed within the Wagyu (BB and AA respectively), but at mixed frequencies in the other two breeds. These SNPs are located in the two introns straddling exon 7. In a separate analysis using the 1000 Bulls database, we found that, coincident with exon 7 of RAB4A first allele frequencies were highest in the high IMF Japanese Native (Wagyu) breeds (0.78) and lowest in the low IMF indicine breeds (Nelore and Brahman), with intermediate marbling breeds (Angus and Charolais) assigned intermediate rankings (0.42). RAB4A is known to encode a protein that regulates intracellular trafficking of the insulin-regulated glucose transporter GLUT4. RAB4A can be considered an attractive new positional candidate for IMF development.


Subject(s)
Adipose Tissue/metabolism , Cattle/genetics , Glucose/metabolism , Muscle, Skeletal/metabolism , rab4 GTP-Binding Proteins/genetics , Animals , Breeding , Gene Frequency , Haplotypes , Lipogenesis/genetics , Polymorphism, Single Nucleotide , Red Meat , Selection, Genetic
3.
Hum Mol Genet ; 27(2): 396-405, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29092026

ABSTRACT

Chronic obstructive pulmonary disease (COPD) is among the major health burdens in adults. While cigarette smoking is the leading risk factor, a growing number of genetic variations have been discovered to influence disease susceptibility. Epigenetic modifications may mediate the response of the genome to smoking and regulate gene expression. Chromosome 19q13.2 region is associated with both smoking and COPD, yet its functional role is unclear. Our study aimed to determine whether rs7937 (RAB4B, EGLN2), a top genetic variant in 19q13.2 region identified in genome-wide association studies of COPD, is associated with differential DNA methylation in blood (N = 1490) and gene expression in blood (N = 721) and lungs (N = 1087). We combined genetic and epigenetic data from the Rotterdam Study (RS) to perform the epigenome-wide association analysis of rs7937. Further, we used genetic and transcriptomic data from blood (RS) and from lung tissue (Lung expression quantitative trait loci mapping study), to perform the transcriptome-wide association study of rs7937. Rs7937 was significantly (FDR < 0.05) and consistently associated with differential DNA methylation in blood at 4 CpG sites in cis, independent of smoking. One methylation site (cg11298343-EGLN2) was also associated with COPD (P = 0.001). Additionally, rs7937 was associated with gene expression levels in blood in cis (EGLN2), 42% mediated through cg11298343, and in lung tissue, in cis and trans (NUMBL, EGLN2, DNMT3A, LOC101929709 and PAK2). Our results suggest that changes of DNA methylation and gene expression may be intermediate steps between genetic variants and COPD, but further causal studies in lung tissue should confirm this hypothesis.


Subject(s)
Chromosomes, Human, Pair 19 , DNA Methylation , Pulmonary Disease, Chronic Obstructive/genetics , Adult , Aged , Chromosome Mapping , Epigenesis, Genetic , Female , Gene Expression , Genetic Predisposition to Disease , Genetic Variation , Genome-Wide Association Study , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Male , Middle Aged , Polymorphism, Single Nucleotide , Pulmonary Disease, Chronic Obstructive/metabolism , Quantitative Trait Loci , Smoking/genetics , rab4 GTP-Binding Proteins/genetics
4.
Cell Mol Life Sci ; 76(24): 4995-5009, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31139847

ABSTRACT

Protein interacting with Amyloid Precursor Protein (APP) tail 1 (PAT1) also called APPBP2 or Ara 67 has different targets such as APP or androgen receptor and is expressed in several tissues. PAT1 is known to be involved in the subcellular trafficking of its targets. We previously observed in primary neurons that PAT1 is poorly associated with APP at the cell surface. Here we show that PAT1 colocalizes with vesicles close to the cell surface labeled with Rab5, Rab4, EEA1 and Rabaptin-5 but not with Rab11 and Rab7. Moreover, PAT1 expression regulates the number of EEA1 and Rab5 vesicles, and endocytosis/recycling of the transferrin receptor. In addition, low levels of PAT1 decrease the size of transferrin-colocalized EEA1 vesicles with time following transferrin uptake. Finally, overexpression of the APP binding domain to PAT1 is sufficient to compromise endocytosis. Altogether, these data suggest that PAT1 is a new actor in transferrin early endocytosis. Whether this new function of PAT1 may have consequences in pathology remains to be determined.


Subject(s)
Amino Acid Transport Systems/genetics , Symporters/genetics , Amyloid beta-Protein Precursor/genetics , Animals , Cell Membrane/genetics , Cell Membrane/metabolism , Endocytosis/genetics , Endosomes/genetics , Endosomes/metabolism , Gene Expression Regulation , Humans , Mice , Neurons/metabolism , Protein Transport , Receptors, Androgen/genetics , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/genetics , rab7 GTP-Binding Proteins
5.
J Biol Chem ; 293(13): 4805-4817, 2018 03 30.
Article in English | MEDLINE | ID: mdl-29425100

ABSTRACT

As a master regulator of endothelial cell function, vascular endothelial growth factor receptor-2 (VEGFR2) activates multiple downstream signaling pathways that are critical for vascular development and normal vessel function. VEGFR2 trafficking through various endosomal compartments modulates its signaling output. Accordingly, proteins that regulate the speed and direction by which VEGFR2 traffics through endosomes have been demonstrated to be particularly important for arteriogenesis. However, little is known about how these proteins control VEGFR2 trafficking and about the implications of this control for endothelial cell function. Here, we show that Rab GTPase-binding effector protein 2 (RABEP2), a Rab-effector protein implicated in arteriogenesis, modulates VEGFR2 trafficking. By employing high-resolution microscopy and biochemical assays, we demonstrate that RABEP2 interacts with the small GTPase Rab4 and regulates VEGFR2 endosomal trafficking to maintain cell-surface expression of VEGFR2 and VEGF signaling. Lack of RABEP2 also led to prolonged retention of VEGFR2 in Rab5-positive sorting endosomes, which increased VEGFR2's exposure to phosphotyrosine phosphatase 1b (PTP1b), causing diminished VEGFR2 signaling. Finally, the loss of RABEP2 increased VEGFR2 degradation by diverting VEGFR2 to Rab7-positive endosomes destined for the lysosome. These results implicate RABEP2 as a key modulator of VEGFR2 endosomal trafficking, and demonstrate the importance of RABEP2 and Rab4 for VEGFR2 signaling in endothelial cells.


Subject(s)
Endosomes/metabolism , Endothelial Cells/metabolism , Signal Transduction , Vascular Endothelial Growth Factor Receptor-2/metabolism , Vesicular Transport Proteins/metabolism , Animals , Endosomes/genetics , Endothelial Cells/cytology , Mice , Mice, Inbred BALB C , Protein Transport , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vesicular Transport Proteins/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
6.
Nicotine Tob Res ; 21(6): 714-722, 2019 05 21.
Article in English | MEDLINE | ID: mdl-29767774

ABSTRACT

INTRODUCTION: Cigarette smoking is a major environmental risk factor for many diseases, including chronic obstructive pulmonary disease (COPD). There are shared genetic influences on cigarette smoking and COPD. Genetic risk factors for cigarette smoking in cohorts enriched for COPD are largely unknown. METHODS: We performed genome-wide association analyses for average cigarettes per day (CPD) across the Genetic Epidemiology of COPD (COPDGene) non-Hispanic white (NHW) (n = 6659) and African American (AA) (n = 3260), GenKOLS (the Genetics of Chronic Obstructive Lung Disease) (n = 1671), and ECLIPSE (the Evaluation of COPD Longitudinally to Identify Predictive Surrogate Endpoints) (n = 1942) cohorts. In addition, we performed exome array association analyses across the COPDGene NHW and AA cohorts. We considered analyses across the entire cohort and stratified by COPD case-control status. RESULTS: We identified genome-wide significant associations for CPD on chromosome 15q25 across all cohorts (lowest p = 1.78 × 10-15), except in the COPDGene AA cohort alone. Previously reported associations on chromosome 19 had suggestive and directionally consistent associations (RAB4, p = 1.95 × 10-6; CYP2A7, p = 7.50 × 10-5; CYP2B6, p = 4.04 × 10-4). When we stratified by COPD case-control status, single nucleotide polymorphisms on chromosome 15q25 were nominally associated with both NHW COPD cases (ß = 0.11, p = 5.58 × 10-4) and controls (ß = 0.12, p = 3.86 × 10-5) For the gene-based exome array association analysis of rare variants, there were no exome-wide significant associations. For these previously replicated associations, the most significant results were among COPDGene NHW subjects for CYP2A7 (p = 5.2 × 10-4). CONCLUSIONS: In a large genome-wide association study of both common variants and a gene-based association of rare coding variants in ever-smokers, we found genome-wide significant associations on chromosome 15q25 with CPD for common variants, but not for rare coding variants. These results were directionally consistent among COPD cases and controls. IMPLICATIONS: We examined both common and rare coding variants associated with CPD in a large population of heavy smokers with and without COPD of NHW and AA descent. We replicated genome-wide significant associations on chromosome 15q25 with CPD for common variants among NHW subjects, but not for rare variants. We demonstrated for the first time that common variants on chromosome 15q25 associated with CPD are similar among COPD cases and controls. Previously reported associations on chromosome 19 showed suggestive and directionally consistent associations among common variants (RAB4, CYP2A7, and CYP2B6) and for rare variants (CYP2A7) among COPDGene NHW subjects. Although the genetic effect sizes for these single nucleotide polymorphisms on chromosome 15q25 are modest, we show that this creates a substantial smoking burden over the lifetime of a smoker.


Subject(s)
Ethnicity/genetics , Genetic Markers , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , Pulmonary Disease, Chronic Obstructive/etiology , Smokers/statistics & numerical data , Smoking/genetics , Adult , Aged , Aged, 80 and over , Aryl Hydrocarbon Hydroxylases/genetics , Case-Control Studies , Cytochrome P-450 CYP2B6/genetics , Cytochrome P450 Family 2/genetics , Europe/epidemiology , Female , Genome-Wide Association Study/methods , Humans , Longitudinal Studies , Male , Middle Aged , Prevalence , Prognosis , Pulmonary Disease, Chronic Obstructive/epidemiology , Pulmonary Disease, Chronic Obstructive/pathology , Smoking/adverse effects , Smoking/epidemiology , United States/epidemiology , rab4 GTP-Binding Proteins/genetics
7.
Biochim Biophys Acta ; 1863(12): 2892-2904, 2016 12.
Article in English | MEDLINE | ID: mdl-27627840

ABSTRACT

The formation of tight cell-cell junctions is essential in the epidermis for its barrier properties. In this tissue, keratinocytes follow a differentiation program tightly associated with their movement from the innermost basal to the outer suprabasal layers, and with changes in their cell-cell adhesion profile. Intercellular adhesion in keratinocytes is mediated through cell-cell contacts, including E-cadherin-based adherens junctions. Although the mechanisms that mediate E-cadherin delivery to the plasma membrane have been widely studied in simple epithelia, this process is less well understood in the stratified epidermis. In this study, we have investigated the role of Engulfment and Cell Motility 2 (ELMO2) and integrin-linked kinase (ILK) in the positioning of E-cadherin-containing recycling endosomes during establishment of cell-cell contacts in differentiating keratinocytes. We now show that induction of keratinocyte differentiation by Ca2+ is accompanied by localization of ELMO2 and ILK to Rab4- and Rab11a-containing recycling endosomes. The positioning of long-loop Rab11a-positive endosomes at areas adjacent to cell-cell contacts is disrupted in ELMO2- or ILK-deficient keratinocytes, and is associated with impaired localization of E-cadherin to cell borders. Our studies show a previously unrecognized role for ELMO2 and ILK in modulation of endosomal positioning, which may play key roles in epidermal sheet maintenance and permeability barrier function.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Adherens Junctions/metabolism , Cadherins/genetics , Cytoskeletal Proteins/genetics , Endosomes/metabolism , Keratinocytes/metabolism , Protein Serine-Threonine Kinases/genetics , Adaptor Proteins, Signal Transducing/deficiency , Adherens Junctions/ultrastructure , Animals , Animals, Newborn , Cadherins/metabolism , Calcium/metabolism , Cell Adhesion , Cell Differentiation , Cell Membrane/metabolism , Cell Membrane/ultrastructure , Cytoskeletal Proteins/deficiency , Endosomes/ultrastructure , Epidermal Cells , Epidermis/metabolism , Gene Expression , Keratinocytes/cytology , Mice , Mice, Transgenic , Primary Cell Culture , Protein Serine-Threonine Kinases/deficiency , Protein Transport , Signal Transduction , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism
8.
Cell Microbiol ; 18(7): 928-48, 2016 07.
Article in English | MEDLINE | ID: mdl-26617273

ABSTRACT

Although human gingival epithelium prevents intrusions by periodontal bacteria, Porphyromonas gingivalis, the most well-known periodontal pathogen, is able to invade gingival epithelial cells and pass through the epithelial barrier into deeper tissues. We previously reported that intracellular P. gingivalis exits from gingival epithelial cells via a recycling pathway. However, the underlying molecular process remains unknown. In the present study, we found that the pathogen localized in early endosomes recruits VAMP2 and Rab4A. VAMP2 was found to be specifically localized in early endosomes, although its localization remained unclear in mammalian cells. A single transmembrane domain of VAMP2 was found to be necessary and sufficient for localizing in early endosomes containing P. gingivalis in gingival epithelial cells. VAMP2 forms a complex with EXOC2/Sec5 and EXOC3/Sec6, whereas Rab4A mediates dissociation of the EXOC complex followed by recruitment of RUFY1/Rabip4, Rab4A effector, and Rab14. Depletion of VAMP2 or Rab4A resulted in accumulation of bacteria in early endosomes and disturbed bacterial exit from infected cells. It is suggested that these novel dynamics allow P. gingivalis to exploit fast recycling pathways promoting further bacterial penetration of gingival tissues.


Subject(s)
Gingiva/microbiology , Host-Pathogen Interactions/physiology , Porphyromonas gingivalis/pathogenicity , Vesicle-Associated Membrane Protein 2/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Biological Transport , Endosomes/metabolism , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Gingiva/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Domains , Vesicle-Associated Membrane Protein 2/genetics , Vesicular Transport Proteins/metabolism , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism
9.
J Biol Chem ; 290(34): 21101-21113, 2015 Aug 21.
Article in English | MEDLINE | ID: mdl-26152716

ABSTRACT

The human ether-a-go-go-related gene (hERG) encodes the pore-forming subunit of the rapidly activating delayed rectifier potassium channel (IKr). A reduction in the hERG current causes long QT syndrome, which predisposes affected individuals to ventricular arrhythmias and sudden death. We reported previously that hERG channels in the plasma membrane undergo vigorous internalization under low K(+) conditions. In the present study, we addressed whether hERG internalization occurs under normal K(+) conditions and whether/how internalized channels are recycled back to the plasma membrane. Using patch clamp, Western blot, and confocal imaging analyses, we demonstrated that internalized hERG channels can effectively recycle back to the plasma membrane. Low K(+)-enhanced hERG internalization is accompanied by an increased rate of hERG recovery in the plasma membrane upon reculture following proteinase K-mediated clearance of cell-surface proteins. The increased recovery rate is not due to enhanced protein synthesis, as hERG mRNA expression was not altered by low K(+) exposure, and the increased recovery was observed in the presence of the protein biosynthesis inhibitor cycloheximide. GTPase Rab11, but not Rab4, is involved in the recycling of hERG channels. Interfering with Rab11 function not only delayed hERG recovery in cells after exposure to low K(+) medium but also decreased hERG expression and function in cells under normal culture conditions. We concluded that the recycling pathway plays an important role in the homeostasis of plasma membrane-bound hERG channels.


Subject(s)
Cell Membrane/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Potassium/metabolism , RNA, Messenger/metabolism , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/metabolism , Amino Acid Sequence , Cell Membrane/chemistry , Cell Membrane/drug effects , Cycloheximide/pharmacology , ERG1 Potassium Channel , Endopeptidase K/chemistry , Ether-A-Go-Go Potassium Channels/chemistry , Ether-A-Go-Go Potassium Channels/genetics , Gene Expression , HEK293 Cells , Humans , Ion Transport , Molecular Sequence Data , Patch-Clamp Techniques , Protein Transport , RNA, Messenger/genetics , rab GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/genetics
10.
Biochim Biophys Acta ; 1853(10 Pt A): 2709-21, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26232624

ABSTRACT

Stromal interaction molecule 1 (STIM1) senses depletion of ER-Ca2+ store and clusters in ER-PM junctions where it associates with and gates Ca2+ influx channels, Orai1 and TRPC1. Clustering of TRPC1 with STIM1 and Orai1 in these junctions is critical since Orai1-mediated Ca2+ entry triggers surface expression of TRPC1 while STIM1 gates the channel. Thus, plasma membrane function of TRPC1 depends on the delivery of the channel to the sites where STIM1 puncta are formed. This study examines intracellular trafficking mechanism(s) that determine plasma membrane expression and function of TRPC1 in cells where Orai1 and TRPC1 are endogenously expressed and contribute to Ca2+ entry. We report that TRPC1 is internalized by Arf6-dependent pathway, sorted to Rab5-containing early endosomes, and trafficked to ER-PM junctions by Rab4-dependent fast recycling. Overexpression of Arf6, or Rab5, but not the respective dominant negative mutants, induced retention of TRPC1 in early endosomes and suppressed TRPC1 function. Notably, cells expressing Arf6 or Rab5 displayed an inwardly rectifying ICRAC current that is mediated by Orai1 instead of TRPC1-associated ISOC, demonstrating that Orai1 function was not altered. Importantly, expression of Rab4, but not STIM1, with Rab5 rescued surface expression and function of TRPC1, restoring generation of ISOC. Together, these data demonstrate that trafficking via fast recycling endosomes determines TRPC1-STIM1 clustering within ER-PM junctions following ER-Ca2+ store depletion which is critical for the surface expression and function of the channel. Ca2+ influx mediated by TRPC1 modifies Ca2+-dependent physiological response of cells.


Subject(s)
Calcium Channels/metabolism , Cell Membrane/metabolism , Endocytosis/physiology , Endoplasmic Reticulum/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , TRPC Cation Channels/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Calcium/metabolism , Calcium Channels/genetics , Cell Membrane/genetics , Endoplasmic Reticulum/genetics , HEK293 Cells , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , ORAI1 Protein , Stromal Interaction Molecule 1 , TRPC Cation Channels/genetics , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism , rab5 GTP-Binding Proteins/genetics , rab5 GTP-Binding Proteins/metabolism
11.
J Cell Sci ; 127(Pt 1): 60-71, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24163434

ABSTRACT

p0071 is an intercellular junction protein of the p120 catenin family. We have identified Rab11a as a novel interaction partner of p0071. p0071 interacted preferentially with active Rab11a. Knockdown experiments revealed an interdependent regulation of both proteins. On the one hand, p0071 depletion induced a perinuclear accumulation of Rab11, suggesting a role of p0071 in the anterograde transport of Rab11 from the pericentrosomal region to the plasma membrane but not in retrograde transport. p0071 as well as Rab11 depletion increased transferrin receptor recycling indicating that p0071-induced Rab11 mislocalization interfered with Rab11 function and shifted recycling from the slow Rab11-dependent pathway to the fast Rab4-dependent pathway. When p0071 or Rab11 depletion was combined with a Rab4 knockdown the effect was reversed. On the other hand, Rab11a depletion increased p0071 recycling to cell contacts thereby identifying p0071 as a Rab11 cargo protein. This correlated with increased intercellular adhesion. Thus, we propose that p0071 has a key role in regulating recycling through the Rab11-dependent perinuclear recycling compartment, and links the regulation of adherens junctions to recycling to allow dynamic modulation of intercellular adhesion.


Subject(s)
Adherens Junctions/metabolism , Cell Membrane/metabolism , Cell Nucleus/metabolism , Plakophilins/metabolism , rab GTP-Binding Proteins/metabolism , Cell Adhesion , Cell Line, Tumor , Endosomes/metabolism , Gene Expression Regulation , Humans , Plakophilins/antagonists & inhibitors , Plakophilins/genetics , Protein Binding , Protein Transport , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Transferrin/genetics , Receptors, Transferrin/metabolism , Signal Transduction , rab GTP-Binding Proteins/antagonists & inhibitors , rab GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 309(7): L700-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26254426

ABSTRACT

Neovascularization, the formation of new blood vessels, requires multiple processes including vascular leak, migration, and adhesion. Endosomal proteins, such as Rabs, regulate trafficking of key signaling proteins involved in neovascularization. The novel endosome protein, p18, enhances vascular endothelial (VE)-cadherin recycling from early endosome to cell junction to improve pulmonary endothelial barrier function. Since endothelial barrier integrity is vital in neovascularization, we sought to elucidate the role for endosome proteins p18 and Rab4, Rab7, and Rab9 in the process of vessel formation within the pulmonary vasculature. Overexpression of wild-type p18 (p18(wt)), but not the nonendosomal-binding mutant (p18(N39)), significantly increased lung microvascular endothelial cell migration, adhesion, and both in vitro and in vivo tube formation. Chemical inhibition of mTOR or p38 attenuated the proneovascularization role of p18(wt). Similar to the effect of p18(wt), overexpression of prorecycling wild-type (Rab4(WT)) and endosome-anchored (Rab4(Q67L)) Rab4 enhanced neovascularization processes, whereas molecular inhibition of Rab4, by using the nonendosomal-binding mutant (Rab4(S22N)) attenuated VEGF-induced neovascularization. Unlike p18, Rab4-induced neovascularization was independent of mTOR or p38 inhibition but was dependent on p18 expression. This study shows for the first time that neovascularization within the pulmonary vasculature is dependent on the prorecycling endocytic proteins Rab4 and p18.


Subject(s)
Carrier Proteins/metabolism , Endosomes/metabolism , Endothelium/metabolism , Lung/metabolism , Neovascularization, Physiologic/physiology , rab4 GTP-Binding Proteins/metabolism , Animals , Carrier Proteins/genetics , Endosomes/genetics , Endothelium/cytology , Mutation , Rats , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , rab4 GTP-Binding Proteins/genetics
13.
J Cell Sci ; 126(Pt 21): 4950-62, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-24006255

ABSTRACT

The endocytic pathway is essential for cell homeostasis and numerous small Rab GTPases are involved in its control. The endocytic trafficking step controlled by Rab4b has not been elucidated, although recent data suggested it could be important for glucose homeostasis, synaptic homeostasis or adaptive immunity. Here, we show that Rab4b is required for early endosome sorting of transferrin receptors (TfRs) to the recycling endosomes, and we identified the AP1γ subunit of the clathrin adaptor AP-1 as a Rab4b effector and key component of the machinery of early endosome sorting. We show that internalised transferrin (Tf) does not reach Vamp3/Rab11 recycling endosomes in the absence of Rab4b, whereas it is rapidly recycled back to the plasma membrane. By contrast, overexpression of Rab4b leads to the accumulation of internalised Tf within AP-1- and clathrin-coated vesicles. These vesicles are poor in early and recycling endocytic markers except for TfR and require AP1γ for their formation. Furthermore, the targeted overexpression of the Rab4b-binding domain of AP1γ to early endosome upon its fusion with FYVE domains inhibited the interaction between Rab4b and endogenous AP1γ, and perturbed Tf traffic. We thus proposed that the interaction between early endocytic Rab4b and AP1γ could allow the budding of clathrin-coated vesicles for subsequent traffic to recycling endosomes. The data also uncover a novel type of endosomes, characterised by low abundance of either early or recycling endocytic markers, which could potentially be generated in cell types that naturally express high level of Rab4b.


Subject(s)
Adaptor Protein Complex gamma Subunits/metabolism , Endosomes/metabolism , rab4 GTP-Binding Proteins/metabolism , Adaptor Protein Complex gamma Subunits/genetics , Biological Transport , Cell Membrane/enzymology , Cell Membrane/genetics , Cell Membrane/metabolism , Clathrin-Coated Vesicles/metabolism , Endocytosis , Endosomes/enzymology , Endosomes/genetics , HeLa Cells , Humans , Protein Binding , Protein Transport , Transferrin/genetics , Transferrin/metabolism , rab4 GTP-Binding Proteins/genetics
14.
Proc Natl Acad Sci U S A ; 109(39): 15787-92, 2012 Sep 25.
Article in English | MEDLINE | ID: mdl-23019362

ABSTRACT

Rab4A is a master regulator of receptor recycling from endocytic compartments to the plasma membrane. The protein TBC1D16 is up-regulated in melanoma, and TBC1D16-overexpressing melanoma cells are dependent on TBC1D16. We show here that TBC1D16 enhances the intrinsic rate of GTP hydrolysis by Rab4A. TBC1D16 is both cytosolic and membrane associated; the membrane-associated pool colocalizes with transferrin and EGF receptors (EGFRs) and early endosome antigen 1, but not with LAMP1 protein. Expression of two TBC1D16 isoforms, but not the inactive R494A mutant, reduces transferrin receptor recycling but has no effect on transferrin receptor internalization. Expression of TBC1D16 alters GFP-Rab4A membrane localization. In HeLa cells, overexpression of TBC1D16 enhances EGF-stimulated EGFR degradation, concomitant with decreased EGFR levels and signaling. Thus, TBC1D16 is a GTPase activating protein for Rab4A that regulates transferrin receptor recycling and EGFR trafficking and signaling.


Subject(s)
ErbB Receptors/metabolism , GTPase-Activating Proteins/metabolism , Signal Transduction/physiology , rab4 GTP-Binding Proteins/metabolism , Epidermal Growth Factor/metabolism , ErbB Receptors/genetics , GTPase-Activating Proteins/genetics , Guanosine Triphosphate/genetics , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Mutation, Missense , Protein Isoforms/genetics , Protein Isoforms/metabolism , Protein Transport/physiology , Proteolysis , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , rab4 GTP-Binding Proteins/genetics
15.
J Biol Chem ; 288(30): 21876-86, 2013 Jul 26.
Article in English | MEDLINE | ID: mdl-23792956

ABSTRACT

The human ether-a-go-go-related gene (hERG) encodes the pore-forming α-subunit of the rapidly activating delayed rectifier K(+) channel in the heart, which plays a critical role in cardiac action potential repolarization. Dysfunction of IKr causes long QT syndrome, a cardiac electrical disorder that predisposes affected individuals to fatal arrhythmias and sudden death. The homeostasis of hERG channels in the plasma membrane depends on a balance between protein synthesis and degradation. Our recent data indicate that hERG channels undergo enhanced endocytic degradation under low potassium (hypokalemia) conditions. The GTPase Rab4 is known to mediate rapid recycling of various internalized proteins to the plasma membrane. In the present study, we investigated the effect of Rab4 on the expression level of hERG channels. Our data revealed that overexpression of Rab4 decreases the expression level of hERG in the plasma membrane. Rab4 does not affect the expression level of the Kv1.5 or EAG K(+) channels. Mechanistically, our data demonstrate that overexpression of Rab4 increases the expression level of endogenous Nedd4-2, a ubiquitin ligase that targets hERG but not Kv1.5 or EAG channels for ubiquitination and degradation. Nedd4-2 undergoes self- ubiquitination and degradation. Rab4 interferes with Nedd4-2 degradation, resulting in an increased expression level of Nedd4-2, which targets hERG. In summary, the present study demonstrates a novel pathway for hERG regulation; Rab4 decreases the hERG density at the plasma membrane by increasing the endogenous Nedd4-2 expression.


Subject(s)
Endosomal Sorting Complexes Required for Transport/metabolism , Ether-A-Go-Go Potassium Channels/metabolism , Ubiquitin-Protein Ligases/metabolism , rab4 GTP-Binding Proteins/metabolism , Animals , Animals, Newborn , Blotting, Western , Cell Membrane/metabolism , Cells, Cultured , ERG1 Potassium Channel , Endosomal Sorting Complexes Required for Transport/genetics , Ether-A-Go-Go Potassium Channels/genetics , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HEK293 Cells , Humans , Male , Membrane Potentials/physiology , Microscopy, Fluorescence , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/physiology , Nedd4 Ubiquitin Protein Ligases , Protein Binding , Rats , Rats, Sprague-Dawley , Ubiquitin-Protein Ligases/genetics , Ubiquitination , rab4 GTP-Binding Proteins/genetics
16.
Am J Physiol Gastrointest Liver Physiol ; 307(8): G863-70, 2014 Oct 15.
Article in English | MEDLINE | ID: mdl-25190474

ABSTRACT

Rab proteins (Ras homologous for brain) play an important role in vesicle trafficking. Rab4 and Rab11 are involved in vesicular trafficking to the plasma membrane from early endosomes and recycling endosomes, respectively. Tauroursodeoxycholate (TUDC) and cAMP increase bile formation, in part, by increasing plasma membrane localization of multidrug resistance-associated protein 2 (MRP2). The goal of the present study was to determine the role of these Rab proteins in the trafficking of MRP2 by testing the hypothesis that Rab11 and/or Rab4 facilitate cAMP- and TUDC-induced MRP2 translocation to the plasma membrane. Studies were conducted in HuH-NTCP cells (HuH7 cells stably transfected with human NTCP), which constitutively express MRP2. HuH-NTCP cells were transfected with Rab11-WT and GDP-locked dominant inactive Rab11-GDP or with Rab4-GDP to study the role of Rab11 and Rab4. A biotinylation method and a GTP overlay assay were used to determine plasma membrane MRP2 and activation of Rab proteins (Rab11 and Rab4), respectively. Cyclic AMP and TUDC increased plasma membrane MRP2 and stimulated Rab11 activity. Plasma membrane translocation of MRP2 by cAMP and TUDC was increased and inhibited in cells transfected with Rab11-WT and Rab11-GDP, respectively. Cyclic AMP (previous study) and TUDC increased Rab4 activity. However, cAMP- and TUDC-induced increases in MRP2 were not inhibited by Rab4-GDP. Taken together, these results suggest that Rab11 is involved in cAMP- and TUDC-induced MRP2 translocation to the plasma membrane.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Cell Membrane/metabolism , Cyclic AMP/pharmacology , Taurochenodeoxycholic Acid/pharmacology , rab GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/metabolism , ATP Binding Cassette Transporter, Subfamily B/genetics , Cell Line , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Organic Anion Transporters, Sodium-Dependent/genetics , Organic Anion Transporters, Sodium-Dependent/metabolism , Protein Transport , Symporters/genetics , Symporters/metabolism , rab GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/genetics , ATP-Binding Cassette Sub-Family B Member 4
17.
J Cell Sci ; 125(Pt 20): 4841-52, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22825871

ABSTRACT

Protein tyrosine phosphatases (PTPs) are a group of tightly regulated enzymes that coordinate with protein tyrosine kinases to control protein phosphorylation during various cellular processes. Using genetic analysis in Drosophila non-transmembrane PTPs, we identified one role that Myopic (Mop), the Drosophila homolog of the human His domain phosphotyrosine phosphatase (HDPTP), plays in cell adhesion. Depletion of Mop results in aberrant integrin distribution and border cell dissociation during Drosophila oogenesis. Interestingly, Mop phosphatase activity is not required for its role in maintaining border cell cluster integrity. We further identified Rab4 GTPase as a Mop interactor in a yeast two-hybrid screen. Expression of the Rab4 dominant-negative mutant leads to border cell dissociation and suppression of Mop-induced wing-blade adhesion defects, suggesting a critical role of Rab4 in Mop-mediated signaling. In mammals, it has been shown that Rab4-dependent recycling of integrins is necessary for cell adhesion and migration. We found that human HDPTP regulates the spatial distribution of Rab4 and integrin trafficking. Depletion of HDPTP resulted in actin reorganization and increased cell motility. Together, our findings suggest an evolutionarily conserved function of HDPTP-Rab4 in the regulation of endocytic trafficking, cell adhesion and migration.


Subject(s)
Cell Adhesion , Cell Movement , Drosophila Proteins , Protein Tyrosine Phosphatases , rab4 GTP-Binding Proteins , Actins/metabolism , Animals , Cell Adhesion/genetics , Cell Movement/genetics , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Mutation , Oogenesis/genetics , Phosphorylation , Protein Transport , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/metabolism , Signal Transduction , Wings, Animal/growth & development , Wings, Animal/pathology , rab4 GTP-Binding Proteins/genetics , rab4 GTP-Binding Proteins/metabolism
18.
Toxins (Basel) ; 16(9)2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39330865

ABSTRACT

Glaesserella parasuis cytolethal distending toxin (GpCDT) can induce cell cycle arrest and apoptosis. Our laboratory's previous work demonstrated that GTPase 4b (Rab4b) is a key host protein implicated in GpCDT-induced cytotoxicity. This study investigated the probable involvement of Rab4b in the process. Our study used CRISPR/Cas9 technology to create a Rab4b-knockout cell line. The results showed greater resistance to GpCDT-induced cell cytotoxicity. In contrast, forced Rab4b overexpression increased GpCDT-induced cytotoxicity. Further immunoprecipitation study reveals that GpCDT may bind with Rab4b. In PK-15 cells, GpCDT is transported to the early endosomes and late endosomes, while after knocking out Rab4b, GpCDT cannot be transported to the early endosome via vesicles. Rab4b appears essential for GpCDT-induced cytotoxicity in PK-15 cells.


Subject(s)
Bacterial Toxins , rab4 GTP-Binding Proteins , Animals , Cell Line , Bacterial Toxins/toxicity , rab4 GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/genetics , Swine , Endosomes/metabolism , Cell Survival/drug effects , Apoptosis/drug effects
19.
J Cell Biol ; 223(11)2024 Nov 04.
Article in English | MEDLINE | ID: mdl-39325073

ABSTRACT

Deleterious mutations in the lipopolysaccharide responsive beige-like anchor protein (LRBA) gene cause severe childhood immune dysregulation. The complexity of the symptoms involving multiple organs and the broad range of unpredictable clinical manifestations of LRBA deficiency complicate the choice of therapeutic interventions. Although LRBA has been linked to Rab11-dependent trafficking of the immune checkpoint protein CTLA-4, its precise cellular role remains elusive. We show that LRBA, however, only slightly colocalizes with Rab11. Instead, LRBA is recruited by members of the small GTPase Arf protein family to the TGN and to Rab4+ endosomes, where it controls intracellular traffic. In patient-derived fibroblasts, loss of LRBA led to defects in the endosomal pathway promoting the accumulation of enlarged endolysosomes and lysosome secretion. Thus, LRBA appears to regulate flow through the endosomal system on Rab4+ endosomes. Our data strongly suggest functions of LRBA beyond CTLA-4 trafficking and provide a conceptual framework to develop new therapies for LRBA deficiency.


Subject(s)
Adaptor Proteins, Signal Transducing , Endosomes , Homeostasis , Lysosomes , rab GTP-Binding Proteins , rab4 GTP-Binding Proteins , Humans , Endosomes/metabolism , Lysosomes/metabolism , rab4 GTP-Binding Proteins/metabolism , rab4 GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Protein Transport , Fibroblasts/metabolism , ADP-Ribosylation Factors/metabolism , ADP-Ribosylation Factors/genetics , trans-Golgi Network/metabolism , HeLa Cells , HEK293 Cells , ADP-Ribosylation Factor 1
20.
Traffic ; 12(3): 313-29, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21134079

ABSTRACT

The trans Golgi network (TGN) of plant cells sorts and packages Golgi products into secretory (SV) and clathrin-coated (CCV) vesicles. We have analyzed of TGN cisternae in Arabidopsis root meristem cells by cell fractionation and electron microscopy/tomography to establish reliable criteria for identifying TGN cisternae in plant cells, and to define their functional attributes. Transformation of a trans Golgi cisterna into a Golgi-associated TGN cisterna begins with cisternal peeling, the formation of SV buds outside the plane of the cisterna and a 30-35% reduction in cisternal membrane area. Free TGN compartments are defined as cisternae that have detached from the Golgi to become independent organelles. Golgi-associated and free TGN compartments, but not trans Golgi cisternae, bind anti-RabA4b and anti-phosphatidylinositol-4 kinase (PI-4K) antibodies. RabA4b and PI-4Kß1 localize to budding SVs in the TGN and to SVs en route to the cell surface. SV and CCV release occurs simultaneously via cisternal fragmentation, which typically yields ∼30 vesicles and one to four residual cisternal fragments. Early endosomal markers, VHA-a1-green fluorescent protein (GFP) and SYP61-cyan fluorescent protein (CFP), colocalized with RabA4b in TGN cisternae, suggesting that the secretory and endocytic pathways converge at the TGN. pi4k1/pi4k2 knockout mutant plants produce SVs with highly variable sizes indicating that PI-4Kß1/2 regulates SV size.


Subject(s)
1-Phosphatidylinositol 4-Kinase/metabolism , Arabidopsis Proteins/metabolism , Arabidopsis , Electron Microscope Tomography , rab4 GTP-Binding Proteins/metabolism , 1-Phosphatidylinositol 4-Kinase/genetics , Arabidopsis/enzymology , Arabidopsis/ultrastructure , Arabidopsis Proteins/genetics , Cell Membrane/ultrastructure , Cell Wall/chemistry , Cell Wall/metabolism , Cell Wall/ultrastructure , Cytokinesis/physiology , Gene Knockout Techniques , Immunohistochemistry , Mutation , Plant Roots/enzymology , Plant Roots/ultrastructure , Polysaccharides/analysis , Secretory Vesicles/ultrastructure , rab4 GTP-Binding Proteins/genetics , trans-Golgi Network/chemistry , trans-Golgi Network/ultrastructure
SELECTION OF CITATIONS
SEARCH DETAIL