Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 92
Filter
1.
Cell ; 186(23): 4985-4991, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37949054

ABSTRACT

Mexican, Puerto Rican, and Central American Ancestry (MPRCA) individuals represent 82% of US Latinos. An intergenerational group of MPRCA women and allies met to discuss persistent underrepresentation of MPRCA women in STEM, identifying multi-level challenges and solutions. Implementation of these solutions is important and will benefit MPRCA women and the entire academic community.


Subject(s)
Hispanic or Latino , Science , Female , Humans , United States , Science/education
2.
Mol Cell ; 83(21): 3766-3772, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37922871

ABSTRACT

Building a diverse laboratory that is equitable is critical for the retention of talent and the growth of trainees professionally and personally. Here, we outline several strategies including enhancing understanding of cultural competency and humility, establishing laboratory values, and developing equitable laboratory structures to create an inclusive laboratory environment to enable trainees to achieve their highest success.


Subject(s)
Diversity, Equity, Inclusion , Laboratories
3.
Trends Biochem Sci ; 47(9): 725-727, 2022 09.
Article in English | MEDLINE | ID: mdl-35606213

ABSTRACT

The coronavirus disease 2019 (COVID-19) pandemic has created unprecedented obstacles for new investigators to traverse. The pandemic's impact exacerbates inequities for groups historically excluded from science. We provide recommendations to support junior faculty, including women and faculty from groups historically excluded from science, in establishing laboratories during the pandemic and foreseeable future.


Subject(s)
COVID-19 , Pandemics , Female , Humans , Laboratories
4.
J Neurosci ; 43(21): 3970-3984, 2023 05 24.
Article in English | MEDLINE | ID: mdl-37019623

ABSTRACT

Endolysosomal defects in neurons are central to the pathogenesis of prion and other neurodegenerative disorders. In prion disease, prion oligomers traffic through the multivesicular body (MVB) and are routed for degradation in lysosomes or for release in exosomes, yet how prions impact proteostatic pathways is unclear. We found that prion-affected human and mouse brain showed a marked reduction in Hrs and STAM1 (ESCRT-0), which route ubiquitinated membrane proteins from early endosomes into MVBs. To determine how the reduction in ESCRT-0 impacts prion conversion and cellular toxicity in vivo, we prion-challenged conditional knockout mice (male and female) having Hrs deleted from neurons, astrocytes, or microglia. The neuronal, but not astrocytic or microglial, Hrs-depleted mice showed a shortened survival and an acceleration in synaptic derangements, including an accumulation of ubiquitinated proteins, deregulation of phosphorylated AMPA and metabotropic glutamate receptors, and profoundly altered synaptic structure, all of which occurred later in the prion-infected control mice. Finally, we found that neuronal Hrs (nHrs) depletion increased surface levels of the cellular prion protein, PrPC, which may contribute to the rapidly advancing disease through neurotoxic signaling. Taken together, the reduced Hrs in the prion-affected brain hampers ubiquitinated protein clearance at the synapse, exacerbates postsynaptic glutamate receptor deregulation, and accelerates neurodegeneration.SIGNIFICANCE STATEMENT Prion diseases are rapidly progressive neurodegenerative disorders characterized by prion aggregate spread through the central nervous system. Early disease features include ubiquitinated protein accumulation and synapse loss. Here, we investigate how prion aggregates alter ubiquitinated protein clearance pathways (ESCRT) in mouse and human prion-infected brain, discovering a marked reduction in Hrs. Using a prion-infection mouse model with neuronal Hrs (nHrs) depleted, we show that low neuronal Hrs is detrimental and markedly shortens survival time while accelerating synaptic derangements, including ubiquitinated protein accumulation, indicating that Hrs loss exacerbates prion disease progression. Additionally, Hrs depletion increases the surface distribution of prion protein (PrPC), linked to aggregate-induced neurotoxic signaling, suggesting that Hrs loss in prion disease accelerates disease through enhancing PrPC-mediated neurotoxic signaling.


Subject(s)
Neurodegenerative Diseases , Prion Diseases , Prions , Male , Female , Mice , Humans , Animals , Prions/metabolism , Prion Proteins/metabolism , Receptors, AMPA/metabolism , Neurons/metabolism , Prion Diseases/metabolism , Prion Diseases/pathology , Neurodegenerative Diseases/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism
5.
Am J Physiol Cell Physiol ; 326(6): C1605-C1610, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38646783

ABSTRACT

G protein-coupled receptors (GPCRs) are ubiquitously expressed cell surface receptors that mediate numerous physiological responses and are highly druggable. Upon activation, GPCRs rapidly couple to heterotrimeric G proteins and are then phosphorylated and internalized from the cell surface. Recent studies indicate that GPCRs not only localize at the plasma membrane but also exist in intracellular compartments where they are competent to signal. Intracellular signaling by GPCRs is best described to occur at endosomes. Several studies have elegantly documented endosomal GPCR-G protein and GPCR-ß-arrestin signaling. Besides phosphorylation, GPCRs are also posttranslationally modified with ubiquitin. GPCR ubiquitination has been studied mainly in the context of receptor endosomal-lysosomal trafficking. However, new studies indicate that ubiquitination of endogenous GPCRs expressed in endothelial cells initiates the assembly of an intracellular p38 mitogen-activated kinase signaling complex that promotes inflammatory responses from endosomes. In this mini-review, we discuss emerging discoveries that provide critical insights into the function of ubiquitination in regulating GPCR inflammatory signaling at endosomes.


Subject(s)
Endosomes , Inflammation , Receptors, G-Protein-Coupled , Signal Transduction , Ubiquitin , Ubiquitination , Endosomes/metabolism , Humans , Receptors, G-Protein-Coupled/metabolism , Animals , Inflammation/metabolism , Inflammation/pathology , Ubiquitin/metabolism , Phosphorylation
6.
J Biol Chem ; 299(12): 105370, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37865315

ABSTRACT

G protein-coupled receptors (GPCRs) are highly druggable and implicated in numerous diseases, including vascular inflammation. GPCR signals are transduced from the plasma membrane as well as from endosomes and controlled by posttranslational modifications. The thrombin-activated GPCR protease-activated receptor-1 is modified by ubiquitin. Ubiquitination of protease-activated receptor-1 drives recruitment of transforming growth factor-ß-activated kinase-1-binding protein 2 (TAB2) and coassociation of TAB1 on endosomes, which triggers p38 mitogen-activated protein kinase-dependent inflammatory responses in endothelial cells. Other endothelial GPCRs also induce p38 activation via a noncanonical TAB1-TAB2-dependent pathway. However, the regulatory processes that control GPCR ubiquitin-driven p38 inflammatory signaling remains poorly understood. We discovered mechanisms that turn on GPCR ubiquitin-dependent p38 signaling, however, the mechanisms that turn off the pathway are not known. We hypothesize that deubiquitination is an important step in regulating ubiquitin-driven p38 signaling. To identify specific deubiquitinating enzymes (DUBs) that control GPCR-p38 mitogen-activated protein kinase signaling, we conducted a siRNA library screen targeting 96 DUBs in endothelial cells and HeLa cells. We identified nine DUBs and validated the function two DUBs including cylindromatosis and ubiquitin-specific protease-34 that specifically regulate thrombin-induced p38 phosphorylation. Depletion of cylindromatosis expression by siRNA enhanced thrombin-stimulated p38 signaling, endothelial barrier permeability, and increased interleukin-6 cytokine expression. Conversely, siRNA knockdown of ubiquitin-specific protease-34 expression decreased thrombin-promoted interleukin-6 expression and had no effect on thrombin-induced endothelial barrier permeability. These studies suggest that specific DUBs distinctly regulate GPCR-induced p38-mediated inflammatory responses.


Subject(s)
Deubiquitinating Enzyme CYLD , Deubiquitinating Enzymes , Endothelial Cells , Thrombin , Humans , Adaptor Proteins, Signal Transducing/metabolism , Deubiquitinating Enzyme CYLD/metabolism , Deubiquitinating Enzymes/metabolism , Endothelial Cells/metabolism , HeLa Cells , Interleukin-6/metabolism , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Receptor, PAR-1/metabolism , RNA, Small Interfering/metabolism , Thrombin/pharmacology , Thrombin/metabolism , Ubiquitin/metabolism , Ubiquitin-Specific Proteases/metabolism , Cell Line , Gene Expression Regulation, Enzymologic , Phosphorylation/genetics
7.
Proc Natl Acad Sci U S A ; 118(49)2021 12 07.
Article in English | MEDLINE | ID: mdl-34873055

ABSTRACT

Endothelial dysfunction is associated with vascular disease and results in disruption of endothelial barrier function and increased sensitivity to apoptosis. Currently, there are limited treatments for improving endothelial dysfunction. Activated protein C (aPC), a promising therapeutic, signals via protease-activated receptor-1 (PAR1) and mediates several cytoprotective responses, including endothelial barrier stabilization and anti-apoptotic responses. We showed that aPC-activated PAR1 signals preferentially via ß-arrestin-2 (ß-arr2) and dishevelled-2 (Dvl2) scaffolds rather than G proteins to promote Rac1 activation and barrier protection. However, the signaling pathways utilized by aPC/PAR1 to mediate anti-apoptotic activities are not known. aPC/PAR1 cytoprotective responses also require coreceptors; however, it is not clear how coreceptors impact different aPC/PAR1 signaling pathways to drive distinct cytoprotective responses. Here, we define a ß-arr2-mediated sphingosine kinase-1 (SphK1)-sphingosine-1-phosphate receptor-1 (S1PR1)-Akt signaling axis that confers aPC/PAR1-mediated protection against cell death. Using human cultured endothelial cells, we found that endogenous PAR1 and S1PR1 coexist in caveolin-1 (Cav1)-rich microdomains and that S1PR1 coassociation with Cav1 is increased by aPC activation of PAR1. Our study further shows that aPC stimulates ß-arr2-dependent SphK1 activation independent of Dvl2 and is required for transactivation of S1PR1-Akt signaling and protection against cell death. While aPC/PAR1-induced, extracellular signal-regulated kinase 1/2 (ERK1/2) activation is also dependent on ß-arr2, neither SphK1 nor S1PR1 are integrated into the ERK1/2 pathway. Finally, aPC activation of PAR1-ß-arr2-mediated protection against apoptosis is dependent on Cav1, the principal structural protein of endothelial caveolae. These studies reveal that different aPC/PAR1 cytoprotective responses are mediated by discrete, ß-arr2-driven signaling pathways in caveolae.


Subject(s)
Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein C/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptor, PAR-1/metabolism , Sphingosine-1-Phosphate Receptors/metabolism , beta-Arrestin 2/metabolism , Anilides/pharmacology , Apoptosis/physiology , Endothelial Cells/physiology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Heterocyclic Compounds, 3-Ring/pharmacology , Humans , Lactones/pharmacology , Methanol/pharmacology , Organophosphonates/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Platelet Aggregation Inhibitors/pharmacology , Protein C/genetics , Proto-Oncogene Proteins c-akt/genetics , Pyridines/pharmacology , Pyrrolidines/pharmacology , Receptor, PAR-1/genetics , Sphingosine-1-Phosphate Receptors/genetics , Sulfones/pharmacology , beta-Arrestin 2/genetics
8.
Pharmacol Rev ; 73(1): 120-151, 2021 01.
Article in English | MEDLINE | ID: mdl-33268549

ABSTRACT

G protein-coupled receptors (GPCRs) are a large family comprising >800 signaling receptors that regulate numerous cellular and physiologic responses. GPCRs have been implicated in numerous diseases and represent the largest class of drug targets. Although advances in GPCR structure and pharmacology have improved drug discovery, the regulation of GPCR function by diverse post-translational modifications (PTMs) has received minimal attention. Over 200 PTMs are known to exist in mammalian cells, yet only a few have been reported for GPCRs. Early studies revealed phosphorylation as a major regulator of GPCR signaling, whereas later reports implicated a function for ubiquitination, glycosylation, and palmitoylation in GPCR biology. Although our knowledge of GPCR phosphorylation is extensive, our knowledge of the modifying enzymes, regulation, and function of other GPCR PTMs is limited. In this review we provide a comprehensive overview of GPCR post-translational modifications with a greater focus on new discoveries. We discuss the subcellular location and regulatory mechanisms that control post-translational modifications of GPCRs. The functional implications of newly discovered GPCR PTMs on receptor folding, biosynthesis, endocytic trafficking, dimerization, compartmentalized signaling, and biased signaling are also provided. Methods to detect and study GPCR PTMs as well as PTM crosstalk are further highlighted. Finally, we conclude with a discussion of the implications of GPCR PTMs in human disease and their importance for drug discovery. SIGNIFICANCE STATEMENT: Post-translational modification of G protein-coupled receptors (GPCRs) controls all aspects of receptor function; however, the detection and study of diverse types of GPCR modifications are limited. A thorough understanding of the role and mechanisms by which diverse post-translational modifications regulate GPCR signaling and trafficking is essential for understanding dysregulated mechanisms in disease and for improving and refining drug development for GPCRs.


Subject(s)
Protein Processing, Post-Translational , Receptors, G-Protein-Coupled , Animals , Humans , Phosphorylation , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Ubiquitination
9.
J Biol Chem ; 298(4): 101801, 2022 04.
Article in English | MEDLINE | ID: mdl-35257745

ABSTRACT

Endothelial dysfunction is a hallmark of inflammation and is mediated by inflammatory factors that signal through G protein-coupled receptors including protease-activated receptor-1 (PAR1). PAR1, a receptor for thrombin, signals via the small GTPase RhoA and myosin light chain intermediates to facilitate endothelial barrier permeability. PAR1 also induces endothelial barrier disruption through a p38 mitogen-activated protein kinase-dependent pathway, which does not integrate into the RhoA/MLC pathway; however, the PAR1-p38 signaling pathways that promote endothelial dysfunction remain poorly defined. To identify effectors of this pathway, we performed a global phosphoproteome analysis of thrombin signaling regulated by p38 in human cultured endothelial cells using multiplexed quantitative mass spectrometry. We identified 5491 unique phosphopeptides and 2317 phosphoproteins, four distinct dynamic phosphoproteome profiles of thrombin-p38 signaling, and an enrichment of biological functions associated with endothelial dysfunction, including modulators of endothelial barrier disruption and a subset of kinases predicted to regulate p38-dependent thrombin signaling. Using available antibodies to detect identified phosphosites of key p38-regulated proteins, we discovered that inhibition of p38 activity and siRNA-targeted depletion of the p38α isoform increased basal phosphorylation of extracellular signal-regulated protein kinase 1/2, resulting in amplified thrombin-stimulated extracellular signal-regulated protein kinase 1/2 phosphorylation that was dependent on PAR1. We also discovered a role for p38 in the phosphorylation of α-catenin, a component of adherens junctions, suggesting that this phosphorylation may function as an important regulatory process. Taken together, these studies define a rich array of thrombin- and p38-regulated candidate proteins that may serve important roles in endothelial dysfunction.


Subject(s)
Endothelial Cells , Thrombin , p38 Mitogen-Activated Protein Kinases , Cells, Cultured , Endothelial Cells/metabolism , Humans , MAP Kinase Signaling System , Phosphorylation , Proteomics , Receptor, PAR-1/metabolism , Thrombin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
10.
J Cell Sci ; 134(8)2021 04 15.
Article in English | MEDLINE | ID: mdl-33722977

ABSTRACT

The α-arrestin domain containing protein 3 (ARRDC3) is a tumor suppressor in triple-negative breast carcinoma (TNBC), a highly metastatic subtype of breast cancer that lacks targeted therapies. Thus, understanding the mechanisms and targets of ARRDC3 in TNBC is important. ARRDC3 regulates trafficking of protease-activated receptor 1 (PAR1, also known as F2R), a G-protein-coupled receptor (GPCR) implicated in breast cancer metastasis. Loss of ARRDC3 causes overexpression of PAR1 and aberrant signaling. Moreover, dysregulation of GPCR-induced Hippo signaling is associated with breast cancer progression. However, the mechanisms responsible for Hippo dysregulation remain unknown. Here, we report that the Hippo pathway transcriptional co-activator TAZ (also known as WWTR1) is the major effector of GPCR signaling and is required for TNBC migration and invasion. Additionally, ARRDC3 suppresses PAR1-induced Hippo signaling via sequestration of TAZ, which occurs independently of ARRDC3-regulated PAR1 trafficking. The ARRDC3 C-terminal PPXY motifs and TAZ WW domain are crucial for this interaction and are required for suppression of TNBC migration and lung metastasis in vivo. These studies are the first to demonstrate a role for ARRDC3 in regulating GPCR-induced TAZ activity in TNBC and reveal multi-faceted tumor suppressor functions of ARRDC3. This article has an associated First Person interview with the first author of the paper.


Subject(s)
Breast Neoplasms , Arrestins/metabolism , Breast Neoplasms/genetics , Female , Humans , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Signal Transduction , Transcription Factors
11.
Proc Natl Acad Sci U S A ; 117(9): 5039-5048, 2020 03 03.
Article in English | MEDLINE | ID: mdl-32071217

ABSTRACT

Thrombin, a procoagulant protease, cleaves and activates protease-activated receptor-1 (PAR1) to promote inflammatory responses and endothelial dysfunction. In contrast, activated protein C (APC), an anticoagulant protease, activates PAR1 through a distinct cleavage site and promotes anti-inflammatory responses, prosurvival, and endothelial barrier stabilization. The distinct tethered ligands formed through cleavage of PAR1 by thrombin versus APC result in unique active receptor conformations that bias PAR1 signaling. Despite progress in understanding PAR1 biased signaling, the proteins and pathways utilized by thrombin versus APC signaling to induce opposing cellular functions are largely unknown. Here, we report the global phosphoproteome induced by thrombin and APC signaling in endothelial cells with the quantification of 11,266 unique phosphopeptides using multiplexed quantitative mass spectrometry. Our results reveal unique dynamic phosphoproteome profiles of thrombin and APC signaling, an enrichment of associated biological functions, including key modulators of endothelial barrier function, regulators of gene transcription, and specific kinases predicted to mediate PAR1 biased signaling. Using small interfering RNA to deplete a subset of phosphorylated proteins not previously linked to thrombin or APC signaling, a function for afadin and adducin-1 actin binding proteins in thrombin-induced endothelial barrier disruption is unveiled. Afadin depletion resulted in enhanced thrombin-promoted barrier permeability, whereas adducin-1 depletion completely ablated thrombin-induced barrier disruption without compromising p38 signaling. However, loss of adducin-1 blocked APC-induced Akt signaling. These studies define distinct thrombin and APC dynamic signaling profiles and a rich array of proteins and biological pathways that engender PAR1 biased signaling in endothelial cells.


Subject(s)
Proteomics , Receptor, PAR-1/metabolism , Signal Transduction , Thrombin/metabolism , Calmodulin-Binding Proteins , Carrier Proteins , Endothelial Cells/metabolism , Humans , Microfilament Proteins , Phosphorylation , Protein C Inhibitor/metabolism
12.
Traffic ; 20(2): 101-109, 2019 02.
Article in English | MEDLINE | ID: mdl-30353650

ABSTRACT

Ubiquitin is covalently attached to substrate proteins in the form of a single ubiquitin moiety or polyubiquitin chains and has been generally linked to protein degradation, however, distinct types of ubiquitin linkages are also used to control other critical cellular processes like cell signaling. Over forty mammalian G protein-coupled receptors (GPCRs) have been reported to be ubiquitinated, but despite the diverse and rich complexity of GPCR signaling, ubiquitin has been largely ascribed to receptor degradation. Indeed, GPCR ubiquitination targets the receptors for degradation by lysosome, which is mediated by the Endosomal Sorting Complexes Required for Transport (ESCRT) machinery, and the proteasome. This has led to the view that ubiquitin and ESCRTs primarily function as the signal to target GPCRs for destruction. Contrary to this conventional view, studies indicate that ubiquitination of certain GPCRs and canonical ubiquitin-binding ESCRTs are not required for receptor degradation and revealed that diverse and complex pathways exist to regulate endo-lysosomal sorting of GPCRs. In other studies, GPCR ubiquitination has been shown to drive signaling and not receptor degradation and further revealed novel insight into the mechanisms by which GPCRs trigger the activity of the ubiquitination machinery. Here, we discuss the diverse pathways by which ubiquitin controls GPCR endo-lysosomal sorting and beyond.


Subject(s)
Endosomes/metabolism , Lysosomes/metabolism , Receptors, G-Protein-Coupled/metabolism , Ubiquitination , Animals , Endosomal Sorting Complexes Required for Transport/metabolism , Humans , Protein Transport
13.
J Biol Chem ; 294(15): 5867-5878, 2019 04 12.
Article in English | MEDLINE | ID: mdl-30760523

ABSTRACT

Endothelial dysfunction is induced by inflammatory mediators including multiple G protein-coupled receptor (GPCR) agonists. However, the GPCR signaling pathways that promote endothelial dysfunction are incompletely understood. We previously showed that thrombin promotes endothelial barrier disruption through autophosphorylation and activation of p38 mitogen-activated protein kinase (MAPK) via a non-canonical transforming growth factor-ß-activated protein kinase-1-binding protein-1 (TAB1) and TAB2-dependent pathway rather than the canonical three-tiered kinase cascade. Here, we sought to determine whether other GPCR agonists stimulate p38 MAPK activation via this non-canonical pathway in human endothelial cells derived from different vascular beds. Using primary human umbilical vein endothelial cells (HUVECs), HUVEC-derived EA.hy926 cells, and human dermal microvascular endothelial cells (HDMECs), we found that both non-canonical and canonical p38 activation pathways components are expressed in these various endothelial cell types, including TAB3, a structurally-related TAB2 homolog. Moreover, multiple GPCRs agonists, including thrombin, histamine, prostaglandin E2, and ADP, stimulated robust p38 autophosphorylation, whereas phosphorylation of the upstream MAPKs MAP kinase kinase 3 (MKK3) and MKK6, was virtually undetectable, indicating that non-canonical p38 activation may exist for other GPCRs. Indeed, in EA.hy926 cells, thrombin- and histamine-stimulated p38 activation depended on TAB1-TAB2, whereas in primary HUVECs, both TAB1-TAB2 and TAB1-TAB3 were required for p38 activation. In HDMECs, thrombin-induced p38 activation depended on TAB1-TAB3, but histamine-induced p38 activation required TAB1-TAB2. Moreover, thrombin- and histamine-stimulated interleukin-6 production required both TAB1-TAB2 and TAB1-TAB3 in HUVEC. We conclude that multiple GPCR agonists utilize non-canonical TAB1-TAB2 and TAB1-TAB3-dependent p38 activation to promote endothelial inflammatory responses.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , MAP Kinase Signaling System , p38 Mitogen-Activated Protein Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adenosine Diphosphate/genetics , Adenosine Diphosphate/metabolism , Cell Line , Dinoprostone/genetics , Dinoprostone/metabolism , Histamine/genetics , Histamine/metabolism , Human Umbilical Vein Endothelial Cells/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Interleukin-6/biosynthesis , Interleukin-6/genetics , MAP Kinase Kinase 3/genetics , MAP Kinase Kinase 3/metabolism , Phosphorylation/genetics , Thrombin/genetics , Thrombin/metabolism , p38 Mitogen-Activated Protein Kinases/genetics
14.
Annu Rev Pharmacol Toxicol ; 57: 349-373, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27618736

ABSTRACT

Protease-activated receptors (PARs) are a unique class of G protein-coupled receptors (GPCRs) that transduce cellular responses to extracellular proteases. PARs have important functions in the vasculature, inflammation, and cancer and are important drug targets. A unique feature of PARs is their irreversible proteolytic mechanism of activation that results in the generation of a tethered ligand that cannot diffuse away. Despite the fact that GPCRs have proved to be the most successful class of druggable targets, the development of agents that target PARs specifically has been challenging. As a consequence, researchers have taken a remarkable diversity of approaches to develop pharmacological entities that modulate PAR function. Here, we present an overview of the diversity of therapeutic agents that have been developed against PARs. We further discuss PAR biased signaling and the influence of receptor compartmentalization, posttranslational modifications, and dimerization, which are important considerations for drug development.


Subject(s)
Drug Discovery/methods , Receptors, Proteinase-Activated/antagonists & inhibitors , Receptors, Proteinase-Activated/metabolism , Animals , Drug Discovery/trends , Humans , Oligopeptides/metabolism , Oligopeptides/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology
15.
J Biol Chem ; 293(9): 3350-3362, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29348172

ABSTRACT

Aberrant G protein-coupled receptor (GPCR) expression and activation has been linked to tumor initiation, progression, invasion, and metastasis. However, compared with other cancer drivers, the exploitation of GPCRs as potential therapeutic targets has been largely ignored, despite the fact that GPCRs are highly druggable. Therefore, to advance the potential status of GPCRs as therapeutic targets, it is important to understand how GPCRs function together with other cancer drivers during tumor progression. We now report that the α-arrestin domain-containing protein-3 (ARRDC3) acts as a tumor suppressor in part by controlling signaling and trafficking of the GPCR, protease-activated receptor-1 (PAR1). In a series of highly invasive basal-like breast carcinomas, we found that expression of ARRDC3 is suppressed whereas PAR1 is aberrantly overexpressed because of defective lysosomal sorting that results in persistent signaling. Using a lentiviral doxycycline-inducible system, we demonstrate that re-expression of ARRDC3 in invasive breast carcinoma is sufficient to restore normal PAR1 trafficking through the ALG-interacting protein X (ALIX)-dependent lysosomal degradative pathway. We also show that ARRDC3 re-expression attenuates PAR1-stimulated persistent signaling of c-Jun N-terminal kinase (JNK) in invasive breast cancer. Remarkably, restoration of ARRDC3 expression significantly reduced activated PAR1-induced breast carcinoma invasion, which was also dependent on JNK signaling. These findings are the first to identify a critical link between the tumor suppressor ARRDC3 and regulation of GPCR trafficking and signaling in breast cancer.


Subject(s)
Arrestins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Lysosomes/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Endosomal Sorting Complexes Required for Transport/metabolism , Enzyme Activation , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Proteolysis , Receptor, PAR-1/metabolism
16.
J Natl Med Assoc ; 111(1): 46-53, 2019 Feb.
Article in English | MEDLINE | ID: mdl-30129483

ABSTRACT

There is a national call for academic medicine to use evidence-based initiatives to improve its culture and climate. The authors report data-driven policy and programmatic interventions that were associated with increased faculty diversity, equity, respectful behavior and improved faculty climate, at UC San Diego Health Sciences. METHODS: Based on demographic and survey data, interventions were designed to improve the climate between 2005 and 2015. Interventions included routine measuring and dissemination of demographic data, changes and dissemination of policy and procedures, and new and improved faculty development programming. Impact was measured using demographic data over time, salary equity studies, and school-wide climate surveys in 2005, 2011, and 2015. Specific outcomes included measures of diversity, salary equity, behavior, and climate. RESULTS: Over the ten-year period, the proportion of women increased from 16% to 23% of tenure/tenure-track faculty and 31%-40% of all faculty. Underrepresented minority faculty increased from less than 1%-7% of tenure/tenure-track faculty and from 5% to 8% of all faculty. While women continued to be paid less than men, the adjusted difference dropped from 23% to 12%. Reports of inappropriate behavior by faculty decreased significantly, while satisfaction and knowledge about institutional mentoring and resources improved. CONCLUSION: Multiple interventions including new faculty development programs, changes in policy, and measuring demographics/climate supported diverse faculty recruitment, enhanced a culture of respect and improved faculty morale. Cultural changes in policy, periodic faculty data collection with dissemination, and increased faculty development, improve the climate in academic medicine.


Subject(s)
Cultural Diversity , Faculty, Medical/organization & administration , Organizational Culture , Schools, Medical/organization & administration , California , Faculty, Medical/psychology , Faculty, Medical/statistics & numerical data , Female , Humans , Interpersonal Relations , Longitudinal Studies , Male , Minority Groups/statistics & numerical data , Organizational Innovation , Organizational Policy , Physicians, Women/statistics & numerical data , Salaries and Fringe Benefits/statistics & numerical data , Schools, Medical/statistics & numerical data , Sexism/economics , Sexism/psychology , Sexism/statistics & numerical data , Sexual Harassment/statistics & numerical data
17.
J Biol Chem ; 292(33): 13867-13878, 2017 08 18.
Article in English | MEDLINE | ID: mdl-28652403

ABSTRACT

Vascular inflammation and thrombosis require the concerted actions of several different agonists, many of which act on G protein-coupled receptors (GPCRs). GPCR dimerization is a well-established phenomenon that can alter protomer function. In platelets and other cell types, protease-activated receptor-4 (PAR4) has been shown to dimerize with the purinergic receptor P2Y12 to coordinate ß-arrestin-mediated Akt signaling, an important mediator of integrin activation. However, the mechanism by which the PAR4-P2Y12 dimer controls ß-arrestin-dependent Akt signaling is not known. We now report that PAR4 and P2Y12 heterodimer internalization is required for ß-arrestin recruitment to endosomes and Akt signaling. Using bioluminescence resonance energy transfer, immunofluorescence microscopy, and co-immunoprecipitation in cells expressing receptors exogenously and endogenously, we demonstrate that PAR4 and P2Y12 specifically interact and form dimers expressed at the cell surface. We also found that activation of PAR4 but not of P2Y12 drives internalization of the PAR4-P2Y12 heterodimer. Remarkably, activated PAR4 internalization was required for recruitment of ß-arrestin to endocytic vesicles, which was dependent on co-expression of P2Y12. Interestingly, stimulation of the PAR4-P2Y12 heterodimer promotes ß-arrestin and Akt co-localization to intracellular vesicles. Moreover, activated PAR4-P2Y12 internalization is required for sustained Akt activation. Thus, internalization of the PAR4-P2Y12 heterodimer is necessary for ß-arrestin recruitment to endosomes and Akt signaling and lays the foundation for examining whether blockade of PAR4 internalization reduces integrin and platelet activation.


Subject(s)
Endocytosis , Proto-Oncogene Proteins c-akt/agonists , Receptors, Purinergic P2Y12/metabolism , Receptors, Thrombin/agonists , Signal Transduction , beta-Arrestin 2/metabolism , Amino Acid Substitution , Animals , Bioluminescence Resonance Energy Transfer Techniques , COS Cells , Cell Line, Tumor , Chlorocebus aethiops , Endosomes/metabolism , Humans , Immunoprecipitation , Microscopy, Fluorescence , Protein Multimerization , Protein Transport , Proto-Oncogene Proteins c-akt/metabolism , Receptor, PAR-1/agonists , Receptor, PAR-1/chemistry , Receptor, PAR-1/genetics , Receptor, PAR-1/metabolism , Receptors, Purinergic P2Y12/chemistry , Receptors, Purinergic P2Y12/genetics , Receptors, Thrombin/chemistry , Receptors, Thrombin/genetics , Receptors, Thrombin/metabolism , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , beta-Arrestin 2/chemistry
18.
Proc Natl Acad Sci U S A ; 112(27): E3600-8, 2015 Jul 07.
Article in English | MEDLINE | ID: mdl-26100877

ABSTRACT

Protease-activated receptor-1 (PAR1) is a G-protein-coupled receptor (GPCR) for the coagulant protease thrombin. Similar to other GPCRs, PAR1 is promiscuous and couples to multiple heterotrimeric G-protein subtypes in the same cell and promotes diverse cellular responses. The molecular mechanism by which activation of a given GPCR with the same ligand permits coupling to multiple G-protein subtypes is unclear. Here, we report that N-linked glycosylation of PAR1 at extracellular loop 2 (ECL2) controls G12/13 versus Gq coupling specificity in response to thrombin stimulation. A PAR1 mutant deficient in glycosylation at ECL2 was more effective at stimulating Gq-mediated phosphoinositide signaling compared with glycosylated wildtype receptor. In contrast, wildtype PAR1 displayed a greater efficacy at G12/13-dependent RhoA activation compared with mutant receptor lacking glycosylation at ECL2. Endogenous PAR1 rendered deficient in glycosylation using tunicamycin, a glycoprotein synthesis inhibitor, also exhibited increased PI signaling and diminished RhoA activation opposite to native receptor. Remarkably, PAR1 wildtype and glycosylation-deficient mutant were equally effective at coupling to Gi and ß-arrestin-1. Consistent with preferential G12/13 coupling, thrombin-stimulated PAR1 wildtype strongly induced RhoA-mediated stress fiber formation compared with mutant receptor. In striking contrast, glycosylation-deficient PAR1 was more effective at increasing cellular proliferation, associated with Gq signaling, than wildtype receptor. These studies suggest that N-linked glycosylation at ECL2 contributes to the stabilization of an active PAR1 state that preferentially couples to G12/13 versus Gq and defines a previously unidentified function for N-linked glycosylation of GPCRs in regulating G-protein signaling bias.


Subject(s)
GTP-Binding Protein alpha Subunits, G12-G13/metabolism , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Receptor, PAR-1/metabolism , Signal Transduction , Algorithms , Animals , Binding Sites/genetics , COS Cells , Chlorocebus aethiops , Fibroblasts/drug effects , Fibroblasts/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/genetics , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Glycosylation , HeLa Cells , Humans , Immunoblotting , Mice, Knockout , Mutation , Protein Binding/drug effects , RNA Interference , Receptor, PAR-1/genetics , Thrombin/pharmacology , Thymidine/metabolism , rhoA GTP-Binding Protein/metabolism
19.
Int J Mol Sci ; 19(7)2018 Jun 27.
Article in English | MEDLINE | ID: mdl-29954076

ABSTRACT

G protein-coupled receptors (GPCRs) are a large diverse family of cell surface signaling receptors implicated in various types of cancers. Several studies indicate that GPCRs control many aspects of cancer progression including tumor growth, invasion, migration, survival and metastasis. While it is known that GPCR activity can be altered in cancer through aberrant overexpression, gain-of-function activating mutations, and increased production and secretion of agonists, the precise mechanisms of how GPCRs contribute to cancer progression remains elusive. Protease-activated receptors (PARs) are a unique class of GPCRs implicated in cancer. PARs are a subfamily of GPCRs comprised of four members that are irreversibly activated by proteolytic cleavage induced by various proteases generated in the tumor microenvironment. Given the unusual proteolytic irreversible activation of PARs, expression of receptors at the cell surface is a key feature that influences signaling responses and is exquisitely controlled by endocytic adaptor proteins. Here, we discuss new survey data from the Cancer Genome Atlas and the Genotype-Tissue Expression projects analysis of expression of all PAR family member expression in human tumor samples as well as the role and function of the endocytic sorting machinery that controls PAR expression and signaling of PARs in normal cells and in cancer.


Subject(s)
Neoplasms/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Proteinase-Activated/metabolism , Animals , Arrestins/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Humans , Neoplasms/genetics , Receptors, G-Protein-Coupled/genetics , Receptors, Proteinase-Activated/genetics , Signal Transduction/genetics , Signal Transduction/physiology
20.
J Biol Chem ; 291(5): 2223-36, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26635365

ABSTRACT

Protease-activated receptor-1 (PAR1) is a G protein-coupled receptor that undergoes proteolytic irreversible activation by coagulant and anti-coagulant proteases. Given the irreversible activation of PAR1, signaling by the receptor is tightly regulated through desensitization and intracellular trafficking. PAR1 displays both constitutive and agonist-induced internalization. Constitutive internalization of PAR1 is important for generating an internal pool of naïve receptors that replenish the cell surface and facilitate resensitization, whereas agonist-induced internalization of PAR1 is critical for terminating G protein signaling. We showed that PAR1 constitutive internalization is mediated by the adaptor protein complex-2 (AP-2), whereas AP-2 and epsin control agonist-induced PAR1 internalization. However, the mechanisms that regulate PAR1 recycling are not known. In the present study we screened a siRNA library of 140 different membrane trafficking proteins to identify key regulators of PAR1 intracellular trafficking. In addition to known mediators of PAR1 endocytosis, we identified Rab11B as a critical regulator of PAR1 trafficking. We found that siRNA-mediated depletion of Rab11B and not Rab11A blocks PAR1 recycling, which enhanced receptor lysosomal degradation. Although Rab11A is not required for PAR1 recycling, depletion of Rab11A resulted in intracellular accumulation of PAR1 through disruption of basal lysosomal degradation of the receptor. Moreover, enhanced degradation of PAR1 observed in Rab11B-deficient cells is blocked by depletion of Rab11A and the autophagy related-5 protein, suggesting that PAR1 is shuttled to an autophagic degradation pathway in the absence of Rab11B recycling. Together these findings suggest that Rab11A and Rab11B differentially regulate intracellular trafficking of PAR1 through distinct endosomal sorting mechanisms.


Subject(s)
Endosomes/metabolism , Fatty Acid-Binding Proteins/metabolism , Gene Expression Regulation , Receptor, PAR-1/metabolism , rab GTP-Binding Proteins/metabolism , Autophagy , Biotinylation , Cell Membrane/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Library , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Lysosomes/metabolism , Microscopy, Fluorescence , Phagosomes/metabolism , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/metabolism , Thrombin/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL