Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Mol Ther Nucleic Acids ; 32: 937-948, 2023 Jun 13.
Article in English | MEDLINE | ID: mdl-37346979

ABSTRACT

Dominant missense mutations in DNAJB6, a co-chaperone of HSP70, cause limb girdle muscular dystrophy (LGMD) D1. No treatments are currently available. Two isoforms exist, DNAJB6a and DNAJB6b, each with distinct localizations in muscle. Mutations reside in both isoforms, yet evidence suggests that DNAJB6b is primarily responsible for disease pathogenesis. Knockdown treatment strategies involving both isoforms carry risk, as DNAJB6 knockout is embryonic lethal. We therefore developed an isoform-specific knockdown approach using morpholinos. Selective reduction of each isoform was achieved in vitro in primary mouse myotubes and human LGMDD1 myoblasts, as well as in vivo in mouse skeletal muscle. To assess isoform specific knockdown in LGMDD1, we created primary myotube cultures from a knockin LGMDD1 mouse model. Using mass spectrometry, we identified an LGMDD1 protein signature related to protein homeostasis and myofibril structure. Selective reduction of DNAJB6b levels in LGMDD1 myotubes corrected much of the proteomic disease signature toward wild type levels. Additional in vivo functional data is required to determine if selective reduction of DNAJB6b is a viable therapeutic target for LGMDD1.

2.
Infect Immun ; 87(4)2019 04.
Article in English | MEDLINE | ID: mdl-30642904

ABSTRACT

Plasmodium falciparum erythrocyte-binding antigen 140 (EBA-140) plays a role in tight junction formation during parasite invasion of red blood cells and is a potential vaccine candidate for malaria. Individuals in areas where malaria is endemic possess EBA-140-specific antibodies, and individuals with high antibody titers to this protein have a lower rate of reinfection by parasites. The red blood cell binding segment of EBA-140 is comprised of two Duffy-binding-like domains, called F1 and F2, that together create region II. The sialic acid-binding pocket of F1 is essential for binding, whereas the sialic acid-binding pocket in F2 appears dispensable. Here, we show that immunization of mice with the complete region II results in poorly neutralizing antibodies. In contrast, immunization of mice with the functionally relevant F1 domain of region II results in antibodies that confer a 2-fold increase in parasite neutralization compared to that of the F2 domain. Epitope mapping of diverse F1 and F2 monoclonal antibodies revealed that the functionally relevant F1 sialic acid-binding pocket is a privileged site inaccessible to antibodies, that the F2 sialic acid-binding pocket contains a nonneutralizing epitope, and that two additional epitopes reside in F1 on the opposite face from the sialic acid-binding pocket. These studies indicate that focusing the immune response to the functionally important F1 sialic acid binding pocket improves the protective immune response of EBA-140. These results have implications for improving future vaccine designs and emphasize the importance of structural vaccinology for malaria.


Subject(s)
Antibodies, Protozoan/immunology , Carrier Proteins/immunology , Epitopes/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Neutralizing/immunology , Antibody Formation , Carrier Proteins/chemistry , Carrier Proteins/genetics , Epitope Mapping , Epitopes/chemistry , Epitopes/genetics , Humans , Malaria, Falciparum/parasitology , Male , Membrane Proteins , Mice, Inbred BALB C , Plasmodium falciparum/genetics , Protein Domains , Protozoan Proteins/chemistry , Protozoan Proteins/genetics
3.
Elife ; 72018 12 17.
Article in English | MEDLINE | ID: mdl-30556808

ABSTRACT

Erythrocyte Binding Antigen of 175 kDa (EBA-175) has a well-defined role in binding to glycophorin A (GpA) during Plasmodium falciparum invasion of erythrocytes. However, EBA-175 is shed post invasion and a role for this shed protein has not been defined. We show that EBA-175 shed from parasites promotes clustering of RBCs, and EBA-175-dependent clusters occur in parasite culture. Region II of EBA-175 is sufficient for clustering RBCs in a GpA-dependent manner. These clusters are capable of forming under physiological flow conditions and across a range of concentrations. EBA-175-dependent RBC clustering provides daughter merozoites ready access to uninfected RBCs enhancing parasite growth. Clustering provides a general method to protect the invasion machinery from immune recognition and disruption as exemplified by protection from neutralizing antibodies that target AMA-1 and RH5. These findings provide a mechanistic framework for the role of shed proteins in RBC clustering, immune evasion, and malaria.


Subject(s)
Antigens, Protozoan/immunology , Glycophorins/immunology , Immune Evasion , Merozoites/immunology , Plasmodium falciparum/immunology , Protozoan Proteins/immunology , Antibodies, Neutralizing/pharmacology , Antigens, Protozoan/genetics , Antigens, Protozoan/metabolism , Carrier Proteins/genetics , Carrier Proteins/immunology , Cell Aggregation/immunology , Cells, Cultured , Culture Media, Conditioned/chemistry , Erythrocytes/drug effects , Erythrocytes/parasitology , Gene Expression , Glycophorins/metabolism , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Merozoites/genetics , Merozoites/growth & development , Plasmodium falciparum/drug effects , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Protein Binding , Protozoan Proteins/genetics , Protozoan Proteins/metabolism
4.
mBio ; 5(5): e01606-14, 2014 Sep 09.
Article in English | MEDLINE | ID: mdl-25205096

ABSTRACT

UNLABELLED: Erythrocyte invasion is an essential step in the pathogenesis of malaria. The erythrocyte binding-like (EBL) family of Plasmodium falciparum proteins recognizes glycophorins (Gp) on erythrocytes and plays a critical role in attachment during invasion. However, the molecular basis for specific receptor recognition by each parasite ligand has remained elusive, as is the case with the ligand/receptor pair P. falciparum EBA-175 (PfEBA-175)/GpA. This is due largely to difficulties in producing properly glycosylated and functional receptors. Here, we developed an expression system to produce recombinant glycosylated and functional GpA, as well as mutations and truncations. We identified the essential binding region and determinants for PfEBA-175 engagement, demonstrated that these determinants are required for the inhibition of parasite growth, and identified the glycans important in mediating the PfEBA-175-GpA interaction. The results suggest that PfEBA-175 engages multiple glycans of GpA encoded by exon 3 and that the presentation of glycans is likely required for high-avidity binding. The absence of exon 3 in GpB and GpE due to a splice site mutation confers specific recognition of GpA by PfEBA-175. We speculate that GpB and GpE may have arisen due to selective pressure to lose the PfEBA-175 binding site in GpA. The expression system described here has wider application for examining other EBL members important in parasite invasion, as well as additional pathogens that recognize glycophorins. The ability to define critical binding determinants in receptor-ligand interactions, as well as a system to genetically manipulate glycosylated receptors, opens new avenues for the design of interventions that disrupt parasite invasion. IMPORTANCE: Plasmodium falciparum uses distinct ligands that bind host cell receptors for invasion of red blood cells (RBCs) during malaria infection. A key entry pathway involves P. falciparum EBA-175 (PfEBA-175) recognizing glycophorin A (GpA) on RBCs. Despite knowledge of this protein-protein interaction, the complete mechanism for specific receptor engagement is not known. PfEBA-175 recognizes GpA but is unable to engage the related RBC receptor GpB or GpE. Understanding the necessary elements that enable PfEBA-175 to specifically recognize GpA is critical in developing specific and potent inhibitors of PfEBA-175 that disrupt host cell invasion and aid in malaria control. Here, we describe a novel system to produce and manipulate the host receptor GpA. Using this system, we probed the elements in GpA necessary for engagement and thus for host cell invasion. These studies have important implications for understanding how ligands and receptors interact and for the future development of malaria interventions.


Subject(s)
Antigens, Protozoan/metabolism , Erythrocytes/parasitology , Exons , Glycophorins/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/metabolism , Amino Acid Sequence , Antigens, Protozoan/genetics , Binding Sites , Glycophorins/metabolism , Glycosylation , HEK293 Cells , Humans , Molecular Sequence Data , Mutation , Plasmodium falciparum/growth & development , Protein Binding/genetics , Protein Interaction Domains and Motifs , Protozoan Proteins/genetics , RNA Splice Sites , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
6.
PLoS Pathog ; 9(5): e1003390, 2013.
Article in English | MEDLINE | ID: mdl-23717209

ABSTRACT

Disrupting erythrocyte invasion by Plasmodium falciparum is an attractive approach to combat malaria. P. falciparum EBA-175 (PfEBA-175) engages the host receptor Glycophorin A (GpA) during invasion and is a leading vaccine candidate. Antibodies that recognize PfEBA-175 can prevent parasite growth, although not all antibodies are inhibitory. Here, using x-ray crystallography, small-angle x-ray scattering and functional studies, we report the structural basis and mechanism for inhibition by two PfEBA-175 antibodies. Structures of each antibody in complex with the PfEBA-175 receptor binding domain reveal that the most potent inhibitory antibody, R217, engages critical GpA binding residues and the proposed dimer interface of PfEBA-175. A second weakly inhibitory antibody, R218, binds to an asparagine-rich surface loop. We show that the epitopes identified by structural studies are critical for antibody binding. Together, the structural and mapping studies reveal distinct mechanisms of action, with R217 directly preventing receptor binding while R218 allows for receptor binding. Using a direct receptor binding assay we show R217 directly blocks GpA engagement while R218 does not. Our studies elaborate on the complex interaction between PfEBA-175 and GpA and highlight new approaches to targeting the molecular mechanism of P. falciparum invasion of erythrocytes. The results suggest studies aiming to improve the efficacy of blood-stage vaccines, either by selecting single or combining multiple parasite antigens, should assess the antibody response to defined inhibitory epitopes as well as the response to the whole protein antigen. Finally, this work demonstrates the importance of identifying inhibitory-epitopes and avoiding decoy-epitopes in antibody-based therapies, vaccines and diagnostics.


Subject(s)
Antibodies, Neutralizing/chemistry , Antibodies, Protozoan/chemistry , Antigens, Protozoan/chemistry , Glycophorins/chemistry , Plasmodium falciparum/chemistry , Protozoan Proteins/chemistry , Antibodies, Neutralizing/immunology , Antibodies, Protozoan/immunology , Antigens, Protozoan/genetics , Antigens, Protozoan/immunology , Epitopes/chemistry , Epitopes/genetics , Epitopes/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Glycophorins/genetics , Glycophorins/immunology , Glycophorins/metabolism , Humans , Plasmodium falciparum/genetics , Plasmodium falciparum/immunology , Protein Structure, Quaternary , Protein Structure, Tertiary , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Protozoan Proteins/immunology , Structure-Activity Relationship
7.
Mol Biol Cell ; 23(18): 3612-23, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22833563

ABSTRACT

The sorting of signaling receptors within the endocytic system is important for appropriate cellular responses. After activation, receptors are trafficked to early endosomes and either recycled or sorted to lysosomes and degraded. Most receptors trafficked to lysosomes are modified with ubiquitin and recruited into an endosomal subdomain enriched in hepatocyte growth factor-regulated tyrosine kinase substrate (HRS), a ubiquitin-binding component of the endosomal-sorting complex required for transport (ESCRT) machinery, and then sorted into intraluminal vesicles (ILVs) of multivesicular bodies (MVBs)/lysosomes. However, not all receptors use ubiquitin or the canonical ESCRT machinery to sort to MVBs/lysosomes. This is exemplified by protease-activated receptor-1 (PAR1), a G protein-coupled receptor for thrombin, which sorts to lysosomes independent of ubiquitination and HRS. We recently showed that the adaptor protein ALIX binds to PAR1, recruits ESCRT-III, and mediates receptor sorting to ILVs of MVBs. However, the mechanism that initiates PAR1 sorting at the early endosome is not known. We now report that the adaptor protein complex-3 (AP-3) regulates PAR1 ubiquitin-independent sorting to MVBs through an ALIX-dependent pathway. AP-3 binds to a PAR1 cytoplasmic tail-localized tyrosine-based motif and mediates PAR1 lysosomal degradation independent of ubiquitination. Moreover, AP-3 facilitates PAR1 interaction with ALIX, suggesting that AP-3 functions before PAR1 engagement of ALIX and MVB/lysosomal sorting.


Subject(s)
Adaptor Protein Complex 3/metabolism , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Lysosomes/metabolism , Multivesicular Bodies/metabolism , Receptor, PAR-1/metabolism , Adaptor Protein Complex 3/genetics , Adaptor Protein Complex delta Subunits/genetics , Adaptor Protein Complex delta Subunits/metabolism , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/genetics , Endosomal Sorting Complexes Required for Transport/genetics , HeLa Cells , Humans , Immunoblotting , Microscopy, Confocal , Models, Biological , Protein Binding , Protein Transport , RNA Interference , Receptor, PAR-1/genetics , Signal Transduction , Ubiquitin/metabolism , Ubiquitination
8.
J Cell Biol ; 197(3): 407-19, 2012 Apr 30.
Article in English | MEDLINE | ID: mdl-22547407

ABSTRACT

The sorting of signaling receptors to lysosomes is an essential regulatory process in mammalian cells. During degradation, receptors are modified with ubiquitin and sorted by endosomal sorting complex required for transport (ESCRT)-0, -I, -II, and -III complexes into intraluminal vesicles (ILVs) of multivesicular bodies (MVBs). However, it remains unclear whether a single universal mechanism mediates MVB sorting of all receptors. We previously showed that protease-activated receptor 1 (PAR1), a G protein-coupled receptor (GPCR) for thrombin, is internalized after activation and sorted to lysosomes independent of ubiquitination and the ubiquitin-binding ESCRT components hepatocyte growth factor-regulated tyrosine kinase substrate and Tsg101. In this paper, we report that PAR1 sorted to ILVs of MVBs through an ESCRT-III-dependent pathway independent of ubiquitination. We further demonstrate that ALIX, a charged MVB protein 4-ESCRT-III interacting protein, bound to a YPX(3)L motif of PAR1 via its central V domain to mediate lysosomal degradation. This study reveals a novel MVB/lysosomal sorting pathway for signaling receptors that bypasses the requirement for ubiquitination and ubiquitin-binding ESCRTs and may be applicable to a subset of GPCRs containing YPX(n)L motifs.


Subject(s)
Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Multivesicular Bodies/metabolism , Receptor, PAR-1/metabolism , Ubiquitin/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/genetics , Adenosine Triphosphatases/metabolism , Amino Acid Motifs , Blotting, Western , Calcium-Binding Proteins/antagonists & inhibitors , Calcium-Binding Proteins/genetics , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Endosomal Sorting Complexes Required for Transport/antagonists & inhibitors , Endosomal Sorting Complexes Required for Transport/genetics , Endosomes/metabolism , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , HeLa Cells , Humans , Immunoprecipitation , Lysosomes/metabolism , Protein Binding , Protein Transport , RNA, Messenger/genetics , RNA, Small Interfering/genetics , Real-Time Polymerase Chain Reaction , Receptor, PAR-1/genetics , Ubiquitination , Vacuolar Proton-Translocating ATPases/antagonists & inhibitors , Vacuolar Proton-Translocating ATPases/genetics , Vacuolar Proton-Translocating ATPases/metabolism
9.
Proc Natl Acad Sci U S A ; 106(15): 6393-7, 2009 Apr 14.
Article in English | MEDLINE | ID: mdl-19332793

ABSTRACT

Protease-activated receptor-1 (PAR(1)) is a G-protein-coupled receptor uniquely activated by proteolysis. Thrombin, a coagulant protease, induces inflammatory responses and endothelial barrier permeability through the activation of PAR(1). Activated protein C (APC), an anti-coagulant protease, also activates PAR(1). However, unlike thrombin, APC elicits anti-inflammatory responses and protects against endothelial barrier dysfunction induced by thrombin. We found that thrombin and APC signaling were lost in PAR(1)-deficient endothelial cells, indicating that PAR(1) is the major effector of protease signaling. To delineate the mechanism responsible for protease-selective signaling by PAR(1), we examined the effect of APC and thrombin on the activation of RhoA and Rac1, small GTPases that differentially regulate endothelial barrier permeability. Thrombin caused robust RhoA signaling but not Rac1 activation, whereas APC stimulated a marked increase in Rac1 activation but not RhoA signaling, consistent with the opposing functions of these proteases on endothelial barrier integrity. Strikingly, APC signaling and endothelial barrier protection effects were abolished in cells lacking caveolin-1, whereas thrombin signaling remained intact. These findings suggest that compartmentalization of PAR(1) in caveolae is critical for APC selective signaling to Rac1 activation and endothelial barrier protection. We further report that APC induces PAR(1) phosphorylation and desensitizes endothelial cells to thrombin signaling but promotes limited receptor cleavage and negligible internalization and degradation even after prolonged APC exposure. Thus, APC selective signaling and endothelial barrier protective effects are mediated through compartmentalization of PAR(1) in caveolae and a novel mechanism of PAR(1) signal regulation.


Subject(s)
Caveolae/metabolism , Receptor, PAR-1/metabolism , Signal Transduction , Cells, Cultured , Enzyme Activation , Humans , Phosphorylation , Protein C/metabolism , Receptor, PAR-1/genetics , Substrate Specificity , Thrombin/metabolism , rac1 GTP-Binding Protein/metabolism , rho GTP-Binding Proteins/metabolism
10.
Annu Rev Pharmacol Toxicol ; 48: 601-29, 2008.
Article in English | MEDLINE | ID: mdl-17995450

ABSTRACT

The heptahelical G protein-coupled receptors (GPCRs) belong to the largest family of cell surface signaling receptors encoded in the human genome. GPCRs signal to diverse extracellular stimuli and control a vast number of physiological responses, making this receptor class the target of nearly half the drugs currently in use. In addition to rapid desensitization, receptor trafficking is crucial for the temporal and spatial control of GPCR signaling. Sorting signals present in the intracytosolic domains of GPCRs regulate trafficking through the endosomal-lysosomal system. GPCR internalization is mediated by serine and threonine phosphorylation and arrestin binding. Short, linear peptide sequences including tyrosine- and dileucine-based motifs, and PDZ ligands that are recognized by distinct endocytic adaptor proteins also mediate internalization and endosomal sorting of GPCRs. We present new data from bioinformatic searches that reveal the presence of these types of sorting signals in the cytoplasmic tails of many known GPCRs. Several recent studies also indicate that the covalent modification of GPCRs with ubiquitin serves as a signal for internalization and lysosomal sorting, expanding the diversity of mechanisms that control trafficking of mammalian GPCRs.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Receptors, G-Protein-Coupled/metabolism , Arrestins/metabolism , Humans , Ligands , Phosphorylation , Protein Sorting Signals/physiology , Ubiquitin/metabolism
11.
Mol Cell Biol ; 26(8): 3231-42, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16581796

ABSTRACT

Protease-activated receptor 1 (PAR1), a G protein-coupled receptor for the coagulant protease thrombin, is irreversibly activated by proteolysis. Unactivated PAR1 cycles constitutively between the plasma membrane and intracellular stores, thereby providing a protected receptor pool that replenishes the cell surface after thrombin exposure and leads to rapid resensitization to thrombin signaling independent of de novo receptor synthesis. Here, we show that AP2, a clathrin adaptor, binds directly to a tyrosine-based motif in the cytoplasmic tail of PAR1 and is essential for constitutive receptor internalization and cellular recovery of thrombin signaling. Expression of a PAR1 tyrosine mutant or depletion of AP2 by RNA interference leads to significant inhibition of PAR1 constitutive internalization, loss of intracellular uncleaved PAR1, and failure of endothelial cells and other cell types to regain thrombin responsiveness. Our findings establish a novel role for AP2 in direct regulation of PAR1 trafficking, a process critically important to the temporal and spatial aspects of thrombin signaling.


Subject(s)
Adaptor Protein Complex 2/metabolism , Endothelium, Vascular/metabolism , Receptors, Thrombin/metabolism , Adaptor Protein Complex 2/chemistry , Adaptor Protein Complex 2/genetics , Amino Acid Motifs , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Calcium/analysis , Cell Line , Endothelium, Vascular/cytology , Fluorescent Antibody Technique , Glutathione Transferase/metabolism , HeLa Cells , Histidine/chemistry , Humans , Immunoblotting , Microscopy, Confocal , Molecular Sequence Data , Protein Subunits/chemistry , Protein Subunits/metabolism , RNA, Small Interfering/metabolism , Receptor, PAR-1/chemistry , Receptor, PAR-1/metabolism , Recombinant Fusion Proteins/metabolism , Surface Plasmon Resonance , Tyrosine/chemistry , Umbilical Veins/cytology
12.
Mol Pharmacol ; 67(1): 78-87, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15475570

ABSTRACT

The irreversible proteolytic nature of protease-activated receptor-2 (PAR2) activation suggests that mechanism(s) responsible for termination of receptor signaling are critical determinants of the magnitude and duration of PAR2-elicited cellular responses. Rapid desensitization of activated G-protein-coupled receptors (GPCRs) involves both phosphorylation and binding of arrestins. Arrestins also function as scaffolds and transducers of mitogen-activated protein (MAP) kinase signaling cascades. The PAR2 cytoplasmic tail (C-tail) contains multiple sites of phosphorylation and may be an important determinant for arrestin interaction. Desensitization and internalization of activated PAR2 were markedly impaired in arrestin-deficient cells compared with wild-type control cells. PAR2 C-tail truncation mutants displayed normal agonist-induced internalization, caused rapid distribution of betaarr2-GFP to the plasma membrane, and desensitized in an arrestin-dependent manner similar to that of wild-type PAR2. It is interesting that PAR2 C-tail mutants lost the capacity to stably associate with arrestins and consequently, redistributed to endocytic vesicles without betaarr2-GFP, whereas internalized wild-type PAR2 remained stably associated with betaarr2-GFP in endosomes. Moreover, activated PAR2 caused rapid and prolonged activation of endogenous extracellular signal-regulated kinase (ERK1/2). It was striking that in arrestin-deficient cells, activated PAR2 induced an initial peak in ERK1/2 activity that rapidly declined. The inability of internalized PAR2 C-tail mutants to stably associate with arrestins also resulted in loss of prolonged ERK2 activation. Thus, the PAR2 C-tail regulates the stability of arrestin interaction and kinetics of ERK1/2 activation but is not essential for desensitization or internalization. These findings further suggest that the diverse functions of arrestins in regulating PAR2 signaling and trafficking are controlled by multiple independent interactions involving both the intracellular loops and the C-tail.


Subject(s)
Arrestins/pharmacology , Receptor, PAR-2/physiology , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cell Line , Cell Membrane/physiology , Chlorocebus aethiops , HeLa Cells , Humans , Kinetics , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligopeptides/pharmacology , Phosphatidylinositols/metabolism , RNA, Small Interfering/genetics , Receptor, PAR-2/agonists , Receptor, PAR-2/drug effects , Recombinant Proteins/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Transfection
13.
J Biol Chem ; 279(21): 21938-47, 2004 May 21.
Article in English | MEDLINE | ID: mdl-15023990

ABSTRACT

Protease-activated receptor-1 (PAR1), a G protein-coupled receptor (GPCR) for thrombin, is irreversibly proteolytically activated, internalized, and then sorted to lysosomes and degraded. Internalization and lysosomal sorting of activated PAR1 is critical for termination of receptor signaling. We previously demonstrated that activated PAR1 is rapidly phosphorylated and internalized via a clathrin- and dynamin-dependent pathway that is independent of arrestins. Toward understanding the mechanisms responsible for activated PAR1 internalization through clathrin-coated pits we examined the function of a highly conserved tyrosine-based motif, YXXL, localized in the cytoplasmic carboxyl tail of the receptor. A mutant PAR1 in which tyrosine 383 and leucine 386 were replaced with alanines (Y383A/L386A) was significantly impaired in agonist-triggered internalization and degradation compared with wild-type receptor. In contrast, constitutive internalization, and recycling of unactivated PAR1 Y383A/L386A mutant was not affected, suggesting that tonic cycling of the mutant receptor remained intact. Strikingly, a PAR1 C387Z truncation mutant in which the YXXL motif was exposed at the C terminus constitutively internalized and degraded in an agonist-independent manner, whereas C387Z truncation mutant in which the critical tyrosine and leucine were mutated to alanine (C387Z-Y383A/L386A) failed to internalize. Inhibition of PAR1 C387Z mutant constitutive internalization with dominant-negative K44A dynamin blocked agonist-independent degradation of the mutant receptor. Together these findings strongly suggest that internalization of activated PAR1 is controlled by multiple regulatory mechanisms involving phosphorylation and a highly conserved tyrosine-based motif, YXXL. This study is the first to describe a function for a tyrosine-based motif, YXX, in GPCR internalization and reveal novel complexities in the regulation of GPCR trafficking.


Subject(s)
Receptor, PAR-1/metabolism , Receptors, G-Protein-Coupled/metabolism , Tyrosine/chemistry , Alanine/chemistry , Amino Acid Motifs , Amino Acid Sequence , Cell Membrane/metabolism , Clathrin/chemistry , Cytoplasm/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Genes, Dominant , HeLa Cells , Humans , Hydrolysis , Immunoblotting , Leucine/chemistry , Microscopy, Fluorescence , Molecular Sequence Data , Mutation , Phosphatidylinositols/chemistry , Phosphorylation , Protein Structure, Tertiary , Sequence Homology, Amino Acid , Signal Transduction , Time Factors , Transfection
14.
Mol Biol Cell ; 15(5): 2143-55, 2004 May.
Article in English | MEDLINE | ID: mdl-14978220

ABSTRACT

Sorting nexin 1 (SNX1) and SNX2, homologues of the yeast vacuolar protein-sorting (Vps)5p, contain a phospholipid-binding motif termed the phox homology (PX) domain and a carboxyl terminal coiled-coil region. A role for SNX1 in trafficking of cell surface receptors from endosomes to lysosomes has been proposed; however, the function of SNX2 remains unknown. Toward understanding the function of SNX2, we first examined the distribution of endogenous protein in HeLa cells. We show that SNX2 resides primarily in early endosomes, whereas SNX1 is found partially in early endosomes and in tubulovesicular-like structures distributed throughout the cytoplasm. We also demonstrate that SNX1 interacts with the mammalian retromer complex through its amino terminal domain, whereas SNX2 does not. Moreover, activated endogenous epidermal growth factor receptor (EGFR) colocalizes markedly with SNX2-positive endosomes, but minimally with SNX1-containing vesicles. To assess SNX2 function, we examined the effect of a PX domain-mutated SNX2 that is defective in vesicle localization on EGFR trafficking. Mutant SNX2 markedly inhibited agonist-induced EGFR degradation, whereas internalization remained intact. In contrast, SNX1 PX domain mutants failed to effect EGFR degradation, whereas a SNX1 deletion mutant significantly inhibited receptor down-regulation. Interestingly, knockdown of SNX1 and SNX2 expression by RNA interference failed to alter agonist-induced EGFR down-regulation. Together, these findings suggest that both SNX1 and SNX2 are involved in regulating lysosomal sorting of internalized EGFR, but neither protein is essential for this process. These studies are the first to demonstrate a function for SNX2 in protein trafficking.


Subject(s)
Down-Regulation , ErbB Receptors/metabolism , Vesicular Transport Proteins/physiology , Endosomes/metabolism , ErbB Receptors/genetics , Gene Deletion , Gene Expression , Gene Silencing , HeLa Cells , Humans , Immunoprecipitation , Lysosomes/metabolism , Microscopy, Confocal , Nuclear Proteins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Point Mutation/genetics , Protein Binding , Protein Interaction Mapping , Protein Structure, Tertiary , Protein Transport/genetics , RNA, Small Interfering/genetics , Sorting Nexins , Transcription Factor TFIIIB , rab5 GTP-Binding Proteins/metabolism
15.
J Biol Chem ; 279(11): 10020-31, 2004 Mar 12.
Article in English | MEDLINE | ID: mdl-14699102

ABSTRACT

Protease-activated receptor 1 (PAR1), a G protein-coupled receptor (GPCR) for thrombin, is the prototypic member of a family of protease-activated receptors. PAR1 is irreversibly proteolytically activated; thus, the magnitude and duration of thrombin cellular responses are determined primarily by mechanisms responsible for termination of receptor signaling. Both phosphorylation and beta-arrestins contribute to rapid desensitization of PAR1 signaling. However, the relative contribution of each of these pathways to the termination of PAR1 signaling is not known. Co-expression of PAR1 with beta-arrestin 1 (betaarr1) in COS-7 cells resulted in a marked inhibition of PAR1 signaling, whereas beta-arrestin 2 (betaarr2) was essentially inactive. Strikingly, signaling by a PAR1 cytoplasmic tail mutant defective in agonist-induced phosphorylation was also attenuated more effectively by betaarr1 compared with betaarr2. In contrast, both beta-arrestin isoforms were equally effective at desensitizing the substance P receptor, a classic reversibly activated GPCR. PAR1 coimmunoprecipitated betaarr1 in an agonist-dependent manner, whereas betaarr2 association was virtually undetectable. Remarkably, betaarr1 also interacted with phosphorylation defective PAR1 mutant, whereas betaarr2 did not. Moreover, constitutively active beta-arrestin mutants, betaarr1 R169E and betaarr2 R170E, that bind to activated receptor independent of phosphorylation failed to enhance either wild type or mutant PAR1 desensitization compared with normal versions of these proteins. In contrast, beta-arrestin mutants displayed enhanced activity at desensitizing the serotonin 5-hydroxytryptamine(2A) receptor. Taken together, these results suggest that, in addition to PAR1 cytoplasmic tail phosphorylation itself, beta-arrestin binding independent of phosphorylation promotes desensitization of PAR1 signaling. These findings reveal a new level of complexity in the regulation of protease-activated GPCR signaling.


Subject(s)
Arrestins/metabolism , Receptor, PAR-1/metabolism , Animals , Arrestins/chemistry , COS Cells , Cytoplasm/metabolism , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation , Humans , Hydrolysis , Immunoblotting , Microscopy, Confocal , Microscopy, Fluorescence , Mutation , Phosphorylation , Plasmids/metabolism , Precipitin Tests , Protein Isoforms , Protein Structure, Tertiary , Serotonin/metabolism , Signal Transduction , Thrombin/chemistry , Time Factors , Transfection , beta-Arrestin 1 , beta-Arrestin 2 , beta-Arrestins
16.
J Biol Chem ; 277(2): 1292-300, 2002 Jan 11.
Article in English | MEDLINE | ID: mdl-11694535

ABSTRACT

The widely expressed beta-arrestin isoforms 1 and 2 bind phosphorylated G protein-coupled receptors (GPCRs) and mediate desensitization and internalization. Phosphorylation of protease-activated receptor-1 (PAR1), a GPCR for thrombin, is important for desensitization and internalization, however, the role of beta-arrestins in signaling and trafficking of PAR1 remains unknown. To assess beta-arrestin function we examined signaling and trafficking of PAR1 in mouse embryonic fibroblasts (MEFs) derived from beta-arrestin (betaarr) knockouts. Desensitization of PAR1 signaling was markedly impaired in MEFs lacking both betaarr1 and betaarr2 isoforms compared with wild-type cells. Strikingly, in cells lacking only betaarr1 PAR1 desensitization was also significantly impaired compared with betaarr2-lacking or wild-type cells. In wild-type MEFs, activated PAR1 was internalized through a dynamin- and clathrin-dependent pathway and degraded. Surprisingly, in cells lacking both betaarr1 and betaarr2 activated PAR1 was similarly internalized through a dynamin- and clathrin-dependent pathway and degraded, whereas the beta(2)-adrenergic receptor (beta(2)-AR) failed to internalize. A PAR1 cytoplasmic tail mutant defective in agonist-induced phosphorylation failed to internalize in both wild-type and beta-arrestin knockout cells. Thus, PAR1 appears to utilize a distinct phosphorylation-dependent but beta-arrestin-independent pathway for internalization through clathrin-coated pits. Together, these findings strongly suggest that the individual beta-arrestin isoforms can differentially regulate GPCR desensitization and further reveal a novel mechanism by which GPCRs can internalize through a dynamin- and clathrin-dependent pathway that is independent of arrestins.


Subject(s)
Arrestins/metabolism , Protein Transport , Receptors, Thrombin/metabolism , Signal Transduction , Animals , Arrestins/genetics , Cells, Cultured , Clathrin/antagonists & inhibitors , Clathrin/metabolism , Down-Regulation , Dynamins , Embryo, Mammalian/cytology , Fibroblasts/cytology , Fibroblasts/metabolism , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Genes, Reporter , Mice , Mice, Knockout , Microscopy, Fluorescence , Protein Isoforms/metabolism , Receptor, PAR-1 , Receptors, Thrombin/genetics , Recombinant Fusion Proteins/metabolism , beta-Arrestins
SELECTION OF CITATIONS
SEARCH DETAIL
...