1
|
Crecelius JM, Manz AR, Benzow S, Marchese A. Receptor Determinants for β-Arrestin Functional Specificity at C-X-C Chemokine Receptor 5. Mol Pharmacol 2024; 106:287-297. [PMID: 39472027 PMCID: PMC11585254 DOI: 10.1124/molpharm.124.000942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 10/07/2024] [Indexed: 11/20/2024] Open
Abstract
β-arrestins are multifaceted adaptor proteins that mediate G protein-coupled receptor (GPCR) desensitization, internalization, and signaling. It is emerging that receptor-specific determinants specify these divergent functions at GPCRs, yet this remains poorly understood. Here, we set out to identify the receptor determinants responsible for β-arrestin-mediated regulation of the chemokine receptor C-X-C motif chemokine receptor 5 (CXCR5). Using bioluminescence resonance energy transfer, we show that β-arrestin1 and β-arrestin2 are dose-dependently recruited to CXCR5 by its cognate ligand C-X-C motif chemokine ligand 13 (CXCL13). The carboxy-terminal tail of CXCR5 contains several serine/threonine residues that can be divided into three discrete phospho-site clusters based on their position relative to transmembrane domain 7. Mutagenesis experiments revealed that the distal and medial phospho-site clusters, but not the proximal, are required for agonist-stimulated β-arrestin1 or β-arrestin2 recruitment to CXCR5. Consistent with this, we provide evidence that the distal and medial, but not proximal, phospho-site clusters are required for receptor desensitization. Surprisingly, the individual phospho-site clusters are not required for agonist-stimulated internalization of CXCR5. Further, we show that CXCL13-stimulated CXCR5 internalization and ERK1/2 phosphorylation, but not desensitization, remain intact in human embryonic kidney 293 cells lacking β-arrestin1 and β-arrestin2. Our study provides evidence that β-arrestins are recruited to CXCR5 and are required for desensitization but are dispensable for internalization or signaling, suggesting that discrete receptor determinants specify the divergent functions of β-arrestins. SIGNIFICANCE STATEMENT: C-X-C motif ligand 13 (CXCL13) and C-X-C motif chemokine receptor 5 (CXCR5) are important in the immune system and are linked to diseases, yet regulation of CXCR5 signaling remains poorly understood. We provide evidence that a phospho-site cluster located at the extreme distal carboxyl-terminal tail of the receptor is responsible for β-arrestin recruitment and receptor desensitization. β-arrestins are not required for CXCL13-stimulated internalization or signaling, indicating that β-arrestins perform only one of their functions at CXCR5 and that discrete receptor determinants specify the divergent functions of β-arrestins.
Collapse
Affiliation(s)
- Joseph M Crecelius
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Aaren R Manz
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Sara Benzow
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
2
|
Robleto VL, Zhuo Y, Crecelius JM, Benzow S, Marchese A. SNX9 family mediates βarrestin-independent GPCR endocytosis. Commun Biol 2024; 7:1455. [PMID: 39511325 PMCID: PMC11544122 DOI: 10.1038/s42003-024-07157-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 10/28/2024] [Indexed: 11/15/2024] Open
Abstract
Agonist-stimulated GPCR endocytosis typically occurs via the multi-faceted adaptor proteins known as βarrestins. However, endocytosis of several GPCRs occurs independently of β-arrestins, suggesting an additional mode of GPCR endocytosis, but the mechanisms remain unknown. Here we provide evidence that sorting nexin 9 (SNX9), a previously described endocytic remodeling protein, functions as a novel cargo adaptor that promotes agonist-stimulated GPCR endocytosis. We show that SNX9 and SNX18, but not β-arrestins, are necessary for endocytosis of the chemokine receptor CXCR4. SNX9 is recruited to CXCR4 at the plasma membrane and interacts directly with the carboxyl-terminal tail of the receptor in a phosphorylation-dependent manner. We also provide evidence that some receptors do not require SNX9 and SNX18 nor β-arrestins for endocytosis, suggesting additional modes for GPCR endocytosis. These results provide novel insights into the mechanisms regulating GPCR trafficking and broaden our overall understanding of GPCR regulation.
Collapse
Affiliation(s)
- Valeria L Robleto
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Ya Zhuo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Joseph M Crecelius
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Sara Benzow
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, 53226, USA.
| |
Collapse
|
3
|
Gahlot P, Kravic B, Rota G, van den Boom J, Levantovsky S, Schulze N, Maspero E, Polo S, Behrends C, Meyer H. Lysosomal damage sensing and lysophagy initiation by SPG20-ITCH. Mol Cell 2024; 84:1556-1569.e10. [PMID: 38503285 DOI: 10.1016/j.molcel.2024.02.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/30/2024] [Accepted: 02/27/2024] [Indexed: 03/21/2024]
Abstract
Cells respond to lysosomal membrane permeabilization by membrane repair or selective macroautophagy of damaged lysosomes, termed lysophagy, but it is not fully understood how this decision is made. Here, we uncover a pathway in human cells that detects lipid bilayer perturbations in the limiting membrane of compromised lysosomes, which fail to be repaired, and then initiates ubiquitin-triggered lysophagy. We find that SPG20 binds the repair factor IST1 on damaged lysosomes and, importantly, integrates that with the detection of damage-associated lipid-packing defects of the lysosomal membrane. Detection occurs via sensory amphipathic helices in SPG20 before rupture of the membrane. If lipid-packing defects are extensive, such as during lipid peroxidation, SPG20 recruits and activates ITCH, which marks the damaged lysosome with lysine-63-linked ubiquitin chains to initiate lysophagy and thus triages the lysosome for destruction. With SPG20 being linked to neurodegeneration, these findings highlight the relevance of a coordinated lysosomal damage response for cellular homeostasis.
Collapse
Affiliation(s)
- Pinki Gahlot
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Bojana Kravic
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Giulia Rota
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Johannes van den Boom
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Sophie Levantovsky
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Nina Schulze
- Imaging Center Campus Essen, Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany
| | - Elena Maspero
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Simona Polo
- IFOM ETS, The AIRC Institute of Molecular Oncology, Milan, Italy
| | - Christian Behrends
- Munich Cluster for Systems Neurology, Medical Faculty, Ludwig-Maximilians-University München, Munich, Germany
| | - Hemmo Meyer
- Center of Medical Biotechnology, Faculty of Biology, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
4
|
DeNies MS, Liu AP, Schnell S. Seeing beyond the blot: A critical look at assumptions and raw data interpretation in Western blotting. Biomol Concepts 2024; 15:bmc-2022-0047. [PMID: 38557557 DOI: 10.1515/bmc-2022-0047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 03/08/2024] [Indexed: 04/04/2024] Open
Abstract
Rapid advancements in technology refine our understanding of intricate biological processes, but a crucial emphasis remains on understanding the assumptions and sources of uncertainty underlying biological measurements. This is particularly critical in cell signaling research, where a quantitative understanding of the fundamental mechanisms governing these transient events is essential for drug development, given their importance in both homeostatic and pathogenic processes. Western blotting, a technique developed decades ago, remains an indispensable tool for investigating cell signaling, protein expression, and protein-protein interactions. While improvements in statistical analysis and methodology reporting have undoubtedly enhanced data quality, understanding the underlying assumptions and limitations of visual inspection in Western blotting can provide valuable additional information for evaluating experimental conclusions. Using the example of agonist-induced receptor post-translational modification, we highlight the theoretical and experimental assumptions associated with Western blotting and demonstrate how raw blot data can offer clues to experimental variability that may not be fully captured by statistical analyses and reported methodologies. This article is not intended as a comprehensive technical review of Western blotting. Instead, we leverage an illustrative example to demonstrate how assumptions about experimental design and data normalization can be revealed within raw data and subsequently influence data interpretation.
Collapse
Affiliation(s)
- Maxwell S DeNies
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Allen P Liu
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Santiago Schnell
- Department of Biological Sciences, University of Notre Dame, Notre Dame, Indiana, United States of America
- Department of Applied & Computational Mathematics & Statistics, University of Notre Dame, Notre Dame, Indiana, United States of America
| |
Collapse
|
5
|
McElrath CJ, Benzow S, Zhuo Y, Marchese A. β-arrestin1 is an E3 ubiquitin ligase adaptor for substrate linear polyubiquitination. J Biol Chem 2023; 299:105474. [PMID: 37981209 PMCID: PMC10755771 DOI: 10.1016/j.jbc.2023.105474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/19/2023] [Accepted: 11/08/2023] [Indexed: 11/21/2023] Open
Abstract
G protein-coupled receptor (GPCR) signaling and trafficking are regulated by multiple mechanisms, including posttranslational modifications such as ubiquitination by E3 ubiquitin ligases. E3 ligases have been linked to agonist-stimulated ubiquitination of GPCRs via simultaneous binding to βarrestins. In addition, βarrestins have been suggested to assist E3 ligases for ubiquitination of key effector molecules, yet mechanistic insight is lacking. Here, we developed an in vitro reconstituted system and show that βarrestin1 (βarr1) serves as an adaptor between the effector protein signal-transducing adaptor molecule 1 (STAM1) and the E3 ligase atrophin-interacting protein 4. Via mass spectrometry, we identified seven lysine residues within STAM1 that are ubiquitinated and several types of ubiquitin linkages. We provide evidence that βarr1 facilitates the formation of linear polyubiquitin chains at lysine residue 136 on STAM1. This lysine residue is important for stabilizing the βarr1:STAM1 interaction in cells following GPCR activation. Our study identifies atrophin-interacting protein 4 as only the second E3 ligase known to conjugate linear polyubiquitin chains and a possible role for linear ubiquitin chains in GPCR signaling and trafficking.
Collapse
Affiliation(s)
- Chandler J McElrath
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Sara Benzow
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Ya Zhuo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.
| |
Collapse
|
6
|
Cheng N, Trejo J. An siRNA library screen identifies CYLD and USP34 as deubiquitinases that regulate GPCR-p38 MAPK signaling and distinct inflammatory responses. J Biol Chem 2023; 299:105370. [PMID: 37865315 PMCID: PMC10694601 DOI: 10.1016/j.jbc.2023.105370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 10/08/2023] [Accepted: 10/09/2023] [Indexed: 10/23/2023] Open
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-β-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.
Collapse
Affiliation(s)
- Norton Cheng
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California, USA; Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California, USA
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
7
|
Sarma P, Carino CMC, Seetharama D, Pandey S, Dwivedi-Agnihotri H, Rui X, Cao Y, Kawakami K, Kumari P, Chen YC, Luker KE, Yadav PN, Luker GD, Laporte SA, Chen X, Inoue A, Shukla AK. Molecular insights into intrinsic transducer-coupling bias in the CXCR4-CXCR7 system. Nat Commun 2023; 14:4808. [PMID: 37558722 PMCID: PMC10412580 DOI: 10.1038/s41467-023-40482-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 07/31/2023] [Indexed: 08/11/2023] Open
Abstract
Chemokine receptors constitute an important subfamily of G protein-coupled receptors (GPCRs), and they are critically involved in a broad range of immune response mechanisms. Ligand promiscuity among these receptors makes them an interesting target to explore multiple aspects of biased agonism. Here, we comprehensively characterize two chemokine receptors namely, CXCR4 and CXCR7, in terms of their transducer-coupling and downstream signaling upon their stimulation by a common chemokine agonist, CXCL12, and a small molecule agonist, VUF11207. We observe that CXCR7 lacks G-protein-coupling while maintaining robust βarr recruitment with a major contribution of GRK5/6. On the other hand, CXCR4 displays robust G-protein activation as expected but exhibits significantly reduced βarr-coupling compared to CXCR7. These two receptors induce distinct βarr conformations even when activated by the same agonist, and CXCR7, unlike CXCR4, fails to activate ERK1/2 MAP kinase. We also identify a key contribution of a single phosphorylation site in CXCR7 for βarr recruitment and endosomal localization. Our study provides molecular insights into intrinsic-bias encoded in the CXCR4-CXCR7 system with broad implications for drug discovery.
Collapse
Affiliation(s)
- Parishmita Sarma
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Carlo Marion C Carino
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Deeksha Seetharama
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Shubhi Pandey
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Hemlata Dwivedi-Agnihotri
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India
| | - Xue Rui
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Yubo Cao
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
| | - Kouki Kawakami
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Poonam Kumari
- Neuroscience and Ageing Biology Division, CSIR-Central Drug Research Institute Sector 10, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Yu-Chih Chen
- Department of Computational and Systems Biology, Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathryn E Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Prem N Yadav
- Neuroscience and Ageing Biology Division, CSIR-Central Drug Research Institute Sector 10, Sitapur Road, Lucknow, 226031, Uttar Pradesh, India
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA
| | - Stéphane A Laporte
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, H3G 1Y6, Canada
- Department of Medicine, McGill University Health Center, McGill University, Montréal, QC, H4A 3J1, Canada
| | - Xin Chen
- Department of Medicinal Chemistry, School of Pharmaceutical Engineering and Life Science, Changzhou University, Changzhou, Jiangsu, 213164, China
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi, 980-8578, Japan
| | - Arun K Shukla
- Department of Biological Sciences and Bioengineering, Indian Institute of Technology, Kanpur, 208016, India.
| |
Collapse
|
8
|
Yun Y, Yoon HJ, Jeong Y, Choi Y, Jang S, Chung KY, Lee HH. GPCR targeting of E3 ubiquitin ligase MDM2 by inactive β-arrestin. Proc Natl Acad Sci U S A 2023; 120:e2301934120. [PMID: 37399373 PMCID: PMC10334748 DOI: 10.1073/pnas.2301934120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/18/2023] [Indexed: 07/05/2023] Open
Abstract
E3 ubiquitin ligase Mdm2 facilitates β-arrestin ubiquitination, leading to the internalization of G protein-coupled receptors (GPCRs). In this process, β-arrestins bind to Mdm2 and recruit it to the receptor; however, the molecular architecture of the β-arrestin-Mdm2 complex has not been elucidated yet. Here, we identified the β-arrestin-binding region (ABR) on Mdm2 and solved the crystal structure of β-arrestin1 in complex with Mdm2ABR peptide. The acidic residues of Mdm2ABR bind to the positively charged concave side of the β-arrestin1 N-domain. The C-tail of β-arrestin1 is still bound to the N-domain, indicating that Mdm2 binds to the inactive state of β-arrestin1, whereas the phosphorylated C-terminal tail of GPCRs binds to activate β-arrestins. The overlapped binding site of Mdm2 and GPCR C-tails on β-arrestin1 suggests that the binding of GPCR C-tails might trigger the release of Mdm2. Moreover, hydrogen/deuterium exchange experiments further show that Mdm2ABR binding to β-arrestin1 induces the interdomain interface to be more dynamic and uncouples the IP6-induced oligomer of β-arrestin1. These results show how the E3 ligase, Mdm2, interacts with β-arrestins to promote the internalization of GPCRs.
Collapse
Affiliation(s)
- Yaejin Yun
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul08826, Republic of Korea
| | - Hye-Jin Yoon
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul08826, Republic of Korea
| | - Yejin Jeong
- School of Pharmacy, Sungkyunkwan University, Suwon16419, Republic of Korea
| | - Yuri Choi
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul08826, Republic of Korea
| | - Soonmin Jang
- Department of Chemistry, Sejong University, Seoul05006, Republic of Korea
| | - Ka Young Chung
- School of Pharmacy, Sungkyunkwan University, Suwon16419, Republic of Korea
| | - Hyung Ho Lee
- Department of Chemistry, College of Natural Sciences, Seoul National University, Seoul08826, Republic of Korea
| |
Collapse
|
9
|
Kotb RM, Ibrahim SS, Mostafa OM, Shahin NN. Potential role of CXCR4 in trastuzumab resistance in breast cancer patients. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166520. [PMID: 35985446 DOI: 10.1016/j.bbadis.2022.166520] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/07/2022] [Accepted: 08/10/2022] [Indexed: 11/16/2022]
Abstract
Despite the efficacy of trastuzumab in treating HER2-positive breast cancer patients, a significant proportion of patients relapse after treatment. The role of C-X-C chemokine receptor type 4 (CXCR4) in trastuzumab resistance was studied only in cell lines and the underlying mechanisms remain largely unclear. This study investigated the role of CXCR4 in trastuzumab resistance in breast cancer patients and explored the possible underlying mechanisms. The study was performed retrospectively on tissue samples from 62 breast cancer patients including 42 who were treated with trastuzumab and chemotherapy and 20 who received chemotherapy alone in adjuvant setting. Expression levels of CXCR4 and its regulators hypoxia-inducible factor 1-alpha (HIF-1α), tristetraprolin (TTP), human antigen R (HuR), itchy E3 ubiquitin protein ligase (ITCH), miR-302a and miR-494 were determined and their associations with tumor recurrence and disease-free survival were analyzed. In trastuzumab-treated patients, high CXCR4 expression was associated with recurrence and was an independent predictor of progression risk after therapy. CXCR4 correlated positively with its transcriptional regulator, HIF-1α, and negatively with its post-translational regulator, ITCH. HIF-1α, HuR and ITCH were significantly associated with clinical outcome. In chemotherapy-treated patients, neither CXCR4 nor any of its regulators were associated with recurrence or predicted disease progression risk after chemotherapy. In conclusion, this study suggests a potential role for CXCR4 in recurrence after trastuzumab-based therapy in human breast cancer that could be mediated, at least in part, by hypoxia and/or decreased ubiquitination. These findings highlight the potential utility of CXCR4 as a promising target for enhancing trastuzumab therapeutic outcome.
Collapse
Affiliation(s)
- Ranim M Kotb
- General Administration of Clinical Trials, Central Administration of Biological and Innovative Products and Clinical Studies, Egyptian Drug Authority, Giza, Egypt
| | - Safinaz S Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Osama M Mostafa
- Department of Pathology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Nancy N Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt.
| |
Collapse
|
10
|
Zhuo Y, Crecelius JM, Marchese A. G protein-coupled receptor kinase phosphorylation of distal C-tail sites specifies βarrestin1-mediated signaling by chemokine receptor CXCR4. J Biol Chem 2022; 298:102351. [PMID: 35940305 PMCID: PMC9465349 DOI: 10.1016/j.jbc.2022.102351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 10/25/2022] Open
|
11
|
Patwardhan A, Cheng N, Trejo J. Post-Translational Modifications of G Protein-Coupled Receptors Control Cellular Signaling Dynamics in Space and Time. Pharmacol Rev 2021; 73:120-151. [PMID: 33268549 PMCID: PMC7736832 DOI: 10.1124/pharmrev.120.000082] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
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.
Collapse
Affiliation(s)
- Anand Patwardhan
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - Norton Cheng
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| | - JoAnn Trejo
- Department of Pharmacology and the Biomedical Sciences Graduate Program, School of Medicine, University of California, San Diego, La Jolla, California
| |
Collapse
|
12
|
Zhuo Y, Gurevich VV, Vishnivetskiy SA, Klug CS, Marchese A. A non-GPCR-binding partner interacts with a novel surface on β-arrestin1 to mediate GPCR signaling. J Biol Chem 2020; 295:14111-14124. [PMID: 32753481 PMCID: PMC7549033 DOI: 10.1074/jbc.ra120.015074] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/29/2020] [Indexed: 12/30/2022] Open
Abstract
The multifaceted adaptor protein β-arr1 (β-arrestin1) promotes activation of focal adhesion kinase (FAK) by the chemokine receptor CXCR4, facilitating chemotaxis. This function of β-arr1 requires the assistance of the adaptor protein STAM1 (signal-transducing adaptor molecule 1) because disruption of the interaction between STAM1 and β-arr1 reduces CXCR4-mediated activation of FAK and chemotaxis. To begin to understand the mechanism by which β-arr1 together with STAM1 activates FAK, we used site-directed spin-labeling EPR spectroscopy-based studies coupled with bioluminescence resonance energy transfer-based cellular studies to show that STAM1 is recruited to activated β-arr1 by binding to a novel surface on β-arr1 at the base of the finger loop, at a site that is distinct from the receptor-binding site. Expression of a STAM1-deficient binding β-arr1 mutant that is still able to bind to CXCR4 significantly reduced CXCL12-induced activation of FAK but had no impact on ERK-1/2 activation. We provide evidence of a novel surface at the base of the finger loop that dictates non-GPCR interactions specifying β-arrestin-dependent signaling by a GPCR. This surface might represent a previously unidentified switch region that engages with effector molecules to drive β-arrestin signaling.
Collapse
Affiliation(s)
- Ya Zhuo
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Vsevolod V Gurevich
- Department of Pharmacology, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Candice S Klug
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
13
|
Guillien M, le Maire A, Mouhand A, Bernadó P, Bourguet W, Banères JL, Sibille N. IDPs and their complexes in GPCR and nuclear receptor signaling. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2020; 174:105-155. [DOI: 10.1016/bs.pmbts.2020.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
14
|
Zenko D, Thompson D, Hislop JN. Endocytic sorting and downregulation of the M2 acetylcholine receptor is regulated by ubiquitin and the ESCRT complex. Neuropharmacology 2020; 162:107828. [PMID: 31654703 DOI: 10.1016/j.neuropharm.2019.107828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2019] [Revised: 10/16/2019] [Accepted: 10/21/2019] [Indexed: 01/14/2023]
Abstract
Cholinergic dysfunction plays a critical role in a number of disease states, and the loss of functional muscarinic acetylcholine receptors plays a key role in disease pathogenesis. Therefore, preventing receptor downregulation would maintain functional receptor number, and be predicted to alleviate symptoms. However, the molecular mechanism(s) underlying muscarinic receptor downregulation are currently unknown. Here we demonstrate that the M2 muscarinic receptor undergoes rapid lysosomal proteolysis, and this lysosomal trafficking is facilitated by ubiquitination of the receptor. Importantly, we show that this trafficking is driven specifically by ESCRT mediated involution. Critically, we provide evidence that disruption of this process leads to a re-routing of the trafficking of the M2 receptor away from the lysosome and into recycling pathway, and eventually back to the plasma membrane. This study is the first to identify the process by which the M2 muscarinic acetylcholine receptor undergoes endocytic sorting, and critically reveals a regulatory checkpoint that represents a target to pharmacologically increase the number of functional muscarinic receptors within the central nervous system.
Collapse
Affiliation(s)
- Dmitry Zenko
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, AB25 2ZD, UK
| | - Dawn Thompson
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, AB25 2ZD, UK
| | - James N Hislop
- Institute of Medical Sciences, School of Medicine, Medical Sciences and Nutrition, University of Aberdeen, AB25 2ZD, UK.
| |
Collapse
|
15
|
Patrussi L, Capitani N, Baldari CT. Abnormalities in chemokine receptor recycling in chronic lymphocytic leukemia. Cell Mol Life Sci 2019; 76:3249-3261. [PMID: 30830241 PMCID: PMC11105227 DOI: 10.1007/s00018-019-03058-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 02/12/2019] [Accepted: 02/28/2019] [Indexed: 12/16/2022]
Abstract
In addition to their modulation through de novo expression and degradation, surface levels of chemokine receptors are tuned by their ligand-dependent recycling to the plasma membrane, which ensures that engaged receptors become rapidly available for further rounds of signaling. Dysregulation of this process contributes to the pathogenesis of chronic lymphocytic leukemia (CLL) by enhancing surface expression of chemokine receptors, thereby favoring leukemic cell accumulation in the protective niche of lymphoid organs. In this review, we summarize our current understanding of the process of chemokine receptor recycling, focusing on the impact of its dysregulation in CLL.
Collapse
Affiliation(s)
- Laura Patrussi
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy.
| | - Nagaja Capitani
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| | - Cosima T Baldari
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100, Siena, Italy
| |
Collapse
|
16
|
Petrillo MG, Oakley RH, Cidlowski JA. β-Arrestin-1 inhibits glucocorticoid receptor turnover and alters glucocorticoid signaling. J Biol Chem 2019; 294:11225-11239. [PMID: 31167788 DOI: 10.1074/jbc.ra118.007150] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 05/30/2019] [Indexed: 01/14/2023] Open
Abstract
Glucocorticoids are among the most widely used drugs to treat many autoimmune and inflammatory diseases. Although much research has been focused on investigating glucocorticoid activity, it remains unclear how glucocorticoids regulate distinct processes in different cells. Glucocorticoids exert their effects through the glucocorticoid receptor (GR), which, upon glucocorticoid binding, interacts with regulatory proteins, affecting its activity and function. These protein-protein interactions are necessary for the resolution of glucocorticoid-dependent physiological and pharmacological processes. In this study, we discovered a novel protein interaction between the glucocorticoid receptor and β-arrestin-1, a scaffold protein with a well-established role in G protein-coupled receptor signaling. Using co-immunoprecipitation and in situ proximity ligation assays in A549 cells, we observed that β-arrestin-1 and unliganded GR interact in the cytoplasm and that, following glucocorticoid binding, the protein complex is found in the nucleus. We show that siRNA-mediated β-arrestin-1 knockdown alters GR protein turnover by up-regulating the E3 ubiquitin ligase Pellino-1, which catalyzes GR ubiquitination and thereby marks the receptor for proteasomal degradation. The enhanced GR turnover observed in β-arrestin-1-deficient cells limits the duration of the glucocorticoid response on GR target genes. These results demonstrate that β-arrestin-1 is a crucial player for the stability of the glucocorticoid receptor. The GR/β-arrestin-1 interaction uncovered here may help unravel mechanisms that contribute to the cell type-specific activities of glucocorticoids.
Collapse
Affiliation(s)
- Maria G Petrillo
- Signal Transduction Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - Robert H Oakley
- Signal Transduction Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| | - John A Cidlowski
- Signal Transduction Laboratory, NIEHS, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, North Carolina 27709
| |
Collapse
|
17
|
DeNies MS, Rosselli-Murai LK, Schnell S, Liu AP. Clathrin Heavy Chain Knockdown Impacts CXCR4 Signaling and Post-translational Modification. Front Cell Dev Biol 2019; 7:77. [PMID: 31139626 PMCID: PMC6518350 DOI: 10.3389/fcell.2019.00077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/23/2019] [Indexed: 01/22/2023] Open
Abstract
Recent research has implicated endocytic pathways as important regulators of receptor signaling. However, the role of endocytosis in regulating chemokine CXC receptor 4 (CXCR4) signaling remains largely unknown. In the present work we systematically investigate the impact of clathrin knockdown on CXCR4 internalization, signaling, and receptor post-translational modification. Inhibition of clathrin-mediated endocytosis (CME) significantly reduced CXCR4 internalization. In contrast to other receptors, clathrin knockdown increased CXCL12-dependent ERK1/2 signaling. Simultaneous inhibition of CME and lipid raft disruption abrogated this increase in ERK1/2 phosphorylation suggesting that endocytic pathway compensation can influence signaling outcomes. Interestingly, using an antibody sensitive to CXCR4 post-translational modification, we also found that our ability to detect CXCR4 was drastically reduced upon clathrin knockdown. We hypothesize that this effect was due to differences in receptor post-translational modification as total CXCR4 protein and mRNA levels were unchanged. Lastly, we show that clathrin knockdown reduced CXCL12-dependent cell migration irrespective of an observed increase in ERK1/2 phosphorylation. Altogether, this work supports a complex model by which modulation of endocytosis affects not only receptor signaling and internalization but also receptor post-translational modification.
Collapse
Affiliation(s)
- Maxwell S DeNies
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States
| | | | - Santiago Schnell
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Molecular and Integrative Physiology, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Allen P Liu
- Cellular and Molecular Biology Graduate Program, University of Michigan Medical School, Ann Arbor, MI, United States.,Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, United States.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, United States.,Department of Biophysics, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
18
|
Regulation of autoimmune disease by the E3 ubiquitin ligase Itch. Cell Immunol 2019; 340:103916. [PMID: 31126634 DOI: 10.1016/j.cellimm.2019.04.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 04/03/2019] [Indexed: 12/14/2022]
Abstract
Itch is a HECT type E3 ubiquitin ligase that is required to prevent the development of autoimmune disease in both mice and humans. Itch is expressed in most mammalian cell types, and, based on published data, it regulates many cellular pathways ranging from T cell differentiation to liver tumorigenesis. Since 1998, when Itch was first discovered, hundreds of publications have described mechanisms through which Itch controls various biologic activities in both immune and non-immune cells. Other studies have provided insight into how Itch catalytic activity is regulated. However, while autoimmunity is the primary clinical feature that occurs in both mice and humans lacking Itch, and Itch control of immune cell function has been well-studied, it remains unclear how Itch prevents the emergence of autoimmune disease. In this review, we explore recent discoveries that advance our understanding of how Itch regulates immune cell biology, and the extent to which these clarify how Itch prevents autoimmune disease. Additionally, we discuss how molecular regulators of Itch impact its ability to control these processes, as this may provide clues on how to therapeutically target Itch to treat patients with autoimmune disease.
Collapse
|
19
|
Hitchinson B, Eby JM, Gao X, Guite-Vinet F, Ziarek JJ, Abdelkarim H, Lee Y, Okamoto Y, Shikano S, Majetschak M, Heveker N, Volkman BF, Tarasova NI, Gaponenko V. Biased antagonism of CXCR4 avoids antagonist tolerance. Sci Signal 2018; 11:11/552/eaat2214. [PMID: 30327409 DOI: 10.1126/scisignal.aat2214] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Repeated dosing of drugs targeting G protein-coupled receptors can stimulate antagonist tolerance, which reduces their efficacy; thus, strategies to avoid tolerance are needed. The efficacy of AMD3100, a competitive antagonist of the chemokine receptor CXCR4 that mobilizes leukemic blasts from the bone marrow into the blood to sensitize them to chemotherapy, is reduced after prolonged treatment. Tolerance to AMD3100 increases the abundance of CXCR4 on the surface of leukemic blasts, which promotes their rehoming to the bone marrow. AMD3100 inhibits both G protein signaling by CXCR4 and β-arrestin1/2-dependent receptor endocytosis. We demonstrated that biased antagonists of G protein-dependent chemotaxis but not β-arrestin1/2 recruitment and subsequent receptor endocytosis avoided tolerance. The peptide antagonist X4-2-6, which is derived from transmembrane helix 2 and extracellular loop 1 of CXCR4, limited chemotaxis and signaling but did not promote CXCR4 accumulation on the cell surface or cause tolerance. The activity of X4-2-6 was due to its distinct mechanism of inhibition of CXCR4. The peptide formed a ternary complex with the receptor and its ligand, the chemokine CXCL12. Within this complex, X4-2-6 released the portion of CXCL12 critical for receptor-mediated activation of G proteins but enabled the rest of the chemokine to recruit β-arrestins to the receptor. In contrast, AMD3100 displaced all components of the chemokine responsible for CXCR4 activation. We further identified a small molecule with similar biased antagonist properties to those of X4-2-6, which may provide a viable alternative to patients when antagonist tolerance prevents drugs from reaching efficacy.
Collapse
Affiliation(s)
- Ben Hitchinson
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Jonathan M Eby
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, Chicago, IL, USA
| | - Xianlong Gao
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, Chicago, IL, USA.,Department of Surgery, Morsani College of Medicine, University of South Florida, College of Medicine, Tampa, FL, USA
| | - Francois Guite-Vinet
- Department of Biochemistry, Research Centre, Sainte-Justine Hospital, Montréal, Quebec, Canada
| | - Joshua J Ziarek
- Department of Molecular and Cellular Biochemistry, Indiana University, Bloomington, IN, USA.,Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Hazem Abdelkarim
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Youngshim Lee
- Division of Bioscience and Biotechnology, Biomolecular Informatics Center, Konkuk University, Seoul 05029, Republic of Korea
| | - Yukari Okamoto
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Sojin Shikano
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA
| | - Matthias Majetschak
- Department of Surgery, Burn and Shock Trauma Research Institute, Loyola University Chicago, Chicago, IL, USA.,Department of Surgery, Morsani College of Medicine, University of South Florida, College of Medicine, Tampa, FL, USA
| | - Nikolaus Heveker
- Department of Biochemistry, Research Centre, Sainte-Justine Hospital, Montréal, Quebec, Canada
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Nadya I Tarasova
- Cancer and Inflammation Program, National Cancer Institute, P.O. Box B, Frederick, MD, USA
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL, USA.
| |
Collapse
|
20
|
English EJ, Mahn SA, Marchese A. Endocytosis is required for C XC chemokine receptor type 4 (CXCR4)-mediated Akt activation and antiapoptotic signaling. J Biol Chem 2018; 293:11470-11480. [PMID: 29899118 DOI: 10.1074/jbc.ra118.001872] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/07/2018] [Indexed: 11/06/2022] Open
Abstract
Signaling activated by binding of the CXC motif chemokine ligand 12 (CXCL12) to its cognate G protein-coupled receptor (GPCR), chemokine CXC motif receptor 4 (CXCR4), is linked to metastatic disease. However, the mechanisms governing CXCR4 signaling remain poorly understood. Here, we show that endocytosis and early endosome antigen 1 (EEA1), which is part of the endosome fusion machinery, are required for CXCL12-mediated AKT Ser/Thr kinase (Akt) signaling selective for certain Akt substrates. Pharmacological inhibition of endocytosis partially attenuated CXCL12-induced phosphorylation of Akt, but not phosphorylation of ERK-1/2. Similarly, phosphorylation of Akt, but not ERK-1/2, stimulated by CXCL13, the cognate ligand for the chemokine receptor CXCR5, was also attenuated by inhibited endocytosis. Furthermore, siRNA-mediated depletion of the Rab5-effector EEA1, but not of adaptor protein, phosphotyrosine-interacting with PH domain and leucine zipper 1 (APPL1), partially attenuated Akt, but not ERK-1/2, phosphorylation promoted by CXCR4. Attenuation of Akt phosphorylation through inhibition of endocytosis or EEA1 depletion was associated with reduced signaling to Akt substrate forkhead box O1/3a but not the Akt substrates TSC complex subunit 2 or glycogen synthase kinase 3β. This suggested that endocytosis and endosomes govern discrete aspects of CXCR4- or CXCR5-mediated Akt signaling. Consistent with this hypothesis, depletion of EEA1 reduced the ability of CXCL12 to attenuate apoptosis in suspended, but not adherent, HeLa cells. Our results suggest a mechanism whereby compartmentalized chemokine-mediated Akt signaling from endosomes suppresses the cancer-related process known as anoikis. Targeting this signaling pathway may help inhibit metastatic cancer involving receptors such as CXCR4.
Collapse
Affiliation(s)
- Elizabeth J English
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Sarah A Mahn
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226.
| |
Collapse
|
21
|
Dergai O, Dergai M, Rynditch A. Ubiquitin-ligase AIP4 controls differential ubiquitination and stability of isoforms of the scaffold protein ITSN1. FEBS Lett 2018; 592:2259-2267. [PMID: 29851086 DOI: 10.1002/1873-3468.13118] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 05/12/2018] [Accepted: 05/17/2018] [Indexed: 11/07/2022]
Abstract
At present, the role of ubiquitination of cargoes internalized from the plasma membrane is better understood than the consequences of ubiquitination of proteins comprising the endocytic machinery. Here, we show that the E3 ubiquitin ligase AIP4/ITCH contributes to the differential ubiquitination of isoforms of the endocytic scaffold protein intersectin1 (ITSN1). The major isoform ITSN1-s is monoubiquitinated, whereas the minor one, ITSN1-22a undergoes a combination of mono- and oligoubiquitination. The monoubiquitination is required for ITSN1-s stability, whereas the oligoubiquitination of ITSN1-22a causes its proteasomal degradation. This explains the observed low abundance of the minor isoform in cells. Thus, different modes of ubiquitination regulated by AIP4 have opposite effects on ITSN1 isoform stability.
Collapse
Affiliation(s)
- Oleksandr Dergai
- Institute of Molecular Biology and Genetics, The National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Mykola Dergai
- Institute of Molecular Biology and Genetics, The National Academy of Science of Ukraine, Kyiv, Ukraine
| | - Alla Rynditch
- Institute of Molecular Biology and Genetics, The National Academy of Science of Ukraine, Kyiv, Ukraine
| |
Collapse
|
22
|
Abstract
G protein-coupled receptors (GPCRs) are cell surface receptors that relay extracellular signals to the inside of the cells. C-X-C chemokine receptor 4 (CXCR4) is a GPCR that undergoes receptor internalization and recycling upon stimulation with its cognate ligand, C-X-C chemokine 12 (CXCL12). Using this receptor/ligand pair we describe the use of two techniques, enzyme-linked immunosorbent assay (ELISA) and flow cytometry, widely used to quantify GPCR internalization from the plasma membrane and its return to the cell surface by recycling.
Collapse
Affiliation(s)
- Amanda M Nevins
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Adriano Marchese
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, USA.
| |
Collapse
|
23
|
Extracellular LDLR repeats modulate Wnt signaling activity by promoting LRP6 receptor endocytosis mediated by the Itch E3 ubiquitin ligase. Genes Cancer 2017; 8:613-627. [PMID: 28966723 PMCID: PMC5620007 DOI: 10.18632/genesandcancer.146] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The LOW-density lipoprotein related protein 6 (LRP6) receptor is an important effector of canonical Wnt signaling, a developmental pathway, whose dysregulation has been implicated in various diseases including cancer. The membrane proximal low-density lipoprotein (LDL) receptor repeats in LRP6 exhibit homology to ligand binding repeats in the LDL receptor (LDLR), but lack known function. We generated single amino acid substitutions of LRP6-LDLR repeat residues, which are highly conserved in the human LDLR and mutated in patients with Familial Hypercholesteremia (FH). These substitutions negatively impacted LRP6 internalization and activation of Wnt signaling. By mass spectrometry, we observed that the Itch E3 ubiquitin ligase associated with and ubiquitinated wild type LRP6 but not the LDLR repeat mutants. These findings establish the involvement of LRP6-LDLR repeats in the regulation of canonical Wnt signaling.
Collapse
|
24
|
Regulation of G Protein-Coupled Receptors by Ubiquitination. Int J Mol Sci 2017; 18:ijms18050923. [PMID: 28448471 PMCID: PMC5454836 DOI: 10.3390/ijms18050923] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Revised: 04/20/2017] [Accepted: 04/23/2017] [Indexed: 02/07/2023] Open
Abstract
G protein-coupled receptors (GPCRs) comprise the largest family of membrane receptors that control many cellular processes and consequently often serve as drug targets. These receptors undergo a strict regulation by mechanisms such as internalization and desensitization, which are strongly influenced by posttranslational modifications. Ubiquitination is a posttranslational modification with a broad range of functions that is currently gaining increased appreciation as a regulator of GPCR activity. The role of ubiquitination in directing GPCRs for lysosomal degradation has already been well-established. Furthermore, this modification can also play a role in targeting membrane and endoplasmic reticulum-associated receptors to the proteasome. Most recently, ubiquitination was also shown to be involved in GPCR signaling. In this review, we present current knowledge on the molecular basis of GPCR regulation by ubiquitination, and highlight the importance of E3 ubiquitin ligases, deubiquitinating enzymes and β-arrestins. Finally, we discuss classical and newly-discovered functions of ubiquitination in controlling GPCR activity.
Collapse
|
25
|
Desrochers G, Cappadocia L, Lussier-Price M, Ton AT, Ayoubi R, Serohijos A, Omichinski JG, Angers A. Molecular basis of interactions between SH3 domain-containing proteins and the proline-rich region of the ubiquitin ligase Itch. J Biol Chem 2017; 292:6325-6338. [PMID: 28235806 DOI: 10.1074/jbc.m116.754440] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 02/07/2017] [Indexed: 11/06/2022] Open
Abstract
The ligase Itch plays major roles in signaling pathways by inducing ubiquitylation-dependent degradation of several substrates. Substrate recognition and binding are critical for the regulation of this reaction. Like closely related ligases, Itch can interact with proteins containing a PPXY motif via its WW domains. In addition to these WW domains, Itch possesses a proline-rich region (PRR) that has been shown to interact with several Src homology 3 (SH3) domain-containing proteins. We have previously established that despite the apparent surface uniformity and conserved fold of SH3 domains, they display different binding mechanisms and affinities for their interaction with the PRR of Itch. Here, we attempt to determine the molecular bases underlying the wide range of binding properties of the Itch PRR. Using pulldown assays combined with mass spectrometry analysis, we show that the Itch PRR preferentially forms complexes with endophilins, amphyphisins, and pacsins but can also target a variety of other SH3 domain-containing proteins. In addition, we map the binding sites of these proteins using a combination of PRR sub-sequences and mutants. We find that different SH3 domains target distinct proline-rich sequences overlapping significantly. We also structurally analyze these protein complexes using crystallography and molecular modeling. These structures depict the position of Itch PRR engaged in a 1:2 protein complex with β-PIX and a 1:1 complex with the other SH3 domain-containing proteins. Taken together, these results reveal the binding preferences of the Itch PRR toward its most common SH3 domain-containing partners and demonstrate that the PRR region is sufficient for binding.
Collapse
Affiliation(s)
| | - Laurent Cappadocia
- Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Mathieu Lussier-Price
- Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - Anh-Tien Ton
- Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | | | - Adrian Serohijos
- Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | - James G Omichinski
- Biochemistry and Molecular Medicine, University of Montreal, Montreal, Quebec H3C 3J7, Canada
| | | |
Collapse
|
26
|
Alekhina O, Marchese A. β-Arrestin1 and Signal-transducing Adaptor Molecule 1 (STAM1) Cooperate to Promote Focal Adhesion Kinase Autophosphorylation and Chemotaxis via the Chemokine Receptor CXCR4. J Biol Chem 2016; 291:26083-26097. [PMID: 27789711 DOI: 10.1074/jbc.m116.757138] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/26/2016] [Indexed: 01/14/2023] Open
Abstract
The chemokine receptor CXCR4 and its chemokine ligand CXCL12 mediate directed cell migration during organogenesis, immune responses, and metastatic disease. However, the mechanisms governing CXCL12/CXCR4-dependent chemotaxis remain poorly understood. Here, we show that the β-arrestin1·signal-transducing adaptor molecule 1 (STAM1) complex, initially identified to govern lysosomal trafficking of CXCR4, also mediates CXCR4-dependent chemotaxis. Expression of minigene fragments from β-arrestin1 or STAM1, known to disrupt the β-arrestin1·STAM1 complex, and RNAi against β-arrestin1 or STAM1, attenuates CXCL12-induced chemotaxis. The β-arrestin1·STAM1 complex is necessary for promoting autophosphorylation of focal adhesion kinase (FAK). FAK is necessary for CXCL12-induced chemotaxis and associates with and localizes with β-arrestin1 and STAM1 in a CXCL12-dependent manner. Our data reveal previously unknown roles in CXCR4-dependent chemotaxis for β-arrestin1 and STAM1, which we propose act in concert to regulate FAK signaling. The β-arrestin1·STAM1 complex is a promising target for blocking CXCR4-promoted FAK autophosphorylation and chemotaxis.
Collapse
Affiliation(s)
- Olga Alekhina
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| | - Adriano Marchese
- From the Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin 53226
| |
Collapse
|
27
|
Abstract
Itch or itchy E3 ubiquitin ligase was initially discovered by genetic studies on the mouse coat color changes, and its deletion results in an itchy phenotype with constant skin scratching and multi-organ inflammation. It is a member of the homologous to E6-associated protein C-terminus (HECT)-type family of E3 ligases, with the protein-interacting WW-domains for the recruitment of substrate and the HECT domain for the transfer of ubiquitin to the substrate. Since its discovery, numerous studies have demonstrated that Itch is involved in the control of many aspects of immune responses including T-cell activation and tolerance and T-helper cell differentiation. Itch is also implicated in other biological contexts such as tumorigenesis, development, and stress responses. Many signaling pathways are regulated by Itch-promoted ubiquitylation of diverse target proteins. Itch is also involved in human diseases. Here, we discuss the major progress in understanding the biological significance of Itch-promoted protein ubiquitylation in the immune and other systems and in Itch-mediated regulation of signal transduction.
Collapse
Affiliation(s)
- Daisuke Aki
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.,Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Wen Zhang
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
| | - Yun-Cai Liu
- Institute for Immunology, Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.,Division of Cell Biology, La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| |
Collapse
|
28
|
Le Clorennec C, Lazrek Y, Dubreuil O, Larbouret C, Poul MA, Mondon P, Melino G, Pèlegrin A, Chardès T. The anti-HER3 (ErbB3) therapeutic antibody 9F7-F11 induces HER3 ubiquitination and degradation in tumors through JNK1/2- dependent ITCH/AIP4 activation. Oncotarget 2016; 7:37013-37029. [PMID: 27203743 PMCID: PMC5095055 DOI: 10.18632/oncotarget.9455] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/16/2016] [Indexed: 01/28/2023] Open
Abstract
We characterized the mechanism of action of the neuregulin-non-competitive anti-HER3 therapeutic antibody 9F7-F11 that blocks the PI3K/AKT pathway, leading to cell cycle arrest and apoptosis in vitro and regression of pancreatic and breast cancer in vivo. We found that 9F7-F11 induces rapid HER3 down-regulation. Specifically, 9F7-F11-induced HER3 ubiquitination and degradation in pancreatic, breast and prostate cancer cell lines was driven mainly by the itchy E3 ubiquitin ligase (ITCH/AIP4). Overexpression of the ITCH/AIP4 inhibitor N4BP1 or small-interfering RNA-mediated knockdown of ITCH/AIP4 inhibited HER3 ubiquitination/degradation and PI3K/AKT signaling blockade induced by 9F7-F11. Moreover, 9F7-F11-mediated JNK1/2 phosphorylation led to ITCH/AIP4 activation and recruitment to HER3 for receptor ubiquitination and degradation. ITCH/AIP4 activity was activated by the deubiquitinases USP8 and USP9X, as demonstrated by RNA interference. Taken together, our results suggest that 9F7-F11-induced HER3 ubiquitination and degradation in cancer cells mainly occurs through JNK1/2-dependent ITCH/AIP4 activation.
Collapse
Affiliation(s)
- Christophe Le Clorennec
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France
- INSERM, U1194 Montpellier, Montpellier, F-34298, France
- Université de Montpellier, Montpellier, F-34298, France
- ICM, Institut Régional du Cancer Montpellier, Montpellier, F-34298, France
| | - Yassamine Lazrek
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France
- INSERM, U1194 Montpellier, Montpellier, F-34298, France
- Université de Montpellier, Montpellier, F-34298, France
- ICM, Institut Régional du Cancer Montpellier, Montpellier, F-34298, France
- Millegen SA, Labège, F-31670, France
- Institut Pasteur de Guyane, BP 6010, 97306, Cayenne Cedex, France
| | - Olivier Dubreuil
- Millegen SA, Labège, F-31670, France
- GamaMabs Pharma SA, Centre Pierre Potier, ONCOPOLE, BP 50624, France
| | - Christel Larbouret
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France
- INSERM, U1194 Montpellier, Montpellier, F-34298, France
- Université de Montpellier, Montpellier, F-34298, France
- ICM, Institut Régional du Cancer Montpellier, Montpellier, F-34298, France
| | - Marie-Alix Poul
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France
- INSERM, U1194 Montpellier, Montpellier, F-34298, France
- Université de Montpellier, Montpellier, F-34298, France
- ICM, Institut Régional du Cancer Montpellier, Montpellier, F-34298, France
| | - Philippe Mondon
- Millegen SA, Labège, F-31670, France
- LFB Biotechnologies, 59000, Lille, France
| | - Gerry Melino
- Biochemistry Laboratory, Instituto Dermopatico Dell'Immacolata, Department of Experimental Medicine and Surgery, University of Rome “Tor Vergata,” 00133 Rome, Italy
- Toxicology Unit, Medical Research Council, Leicester University, Leicester LE1 9HN, United Kingdom
| | - André Pèlegrin
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France
- INSERM, U1194 Montpellier, Montpellier, F-34298, France
- Université de Montpellier, Montpellier, F-34298, France
- ICM, Institut Régional du Cancer Montpellier, Montpellier, F-34298, France
| | - Thierry Chardès
- IRCM, Institut de Recherche en Cancérologie de Montpellier, Montpellier, F-34298, France
- INSERM, U1194 Montpellier, Montpellier, F-34298, France
- Université de Montpellier, Montpellier, F-34298, France
- ICM, Institut Régional du Cancer Montpellier, Montpellier, F-34298, France
| |
Collapse
|
29
|
Bamidele AO, Kremer KN, Hirsova P, Clift IC, Gores GJ, Billadeau DD, Hedin KE. IQGAP1 promotes CXCR4 chemokine receptor function and trafficking via EEA-1+ endosomes. J Cell Biol 2016. [PMID: 26195666 PMCID: PMC4508899 DOI: 10.1083/jcb.201411045] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
IQGAP1 mediates CXCR4 cell surface expression and signaling by regulating EEA-1+ endosome interactions with microtubules during CXCR4 trafficking and recycling. IQ motif–containing GTPase-activating protein 1 (IQGAP1) is a cytoskeleton-interacting scaffold protein. CXCR4 is a chemokine receptor that binds stromal cell–derived factor-1 (SDF-1; also known as CXCL12). Both IQGAP1 and CXCR4 are overexpressed in cancer cell types, yet it was unclear whether these molecules functionally interact. Here, we show that depleting IQGAP1 in Jurkat T leukemic cells reduced CXCR4 expression, disrupted trafficking of endocytosed CXCR4 via EEA-1+ endosomes, and decreased efficiency of CXCR4 recycling. SDF-1–induced cell migration and activation of extracellular signal-regulated kinases 1 and 2 (ERK) MAPK were strongly inhibited, even when forced overexpression restored CXCR4 levels. Similar results were seen in KMBC and HEK293 cells. Exploring the mechanism, we found that SDF-1 treatment induced IQGAP1 binding to α-tubulin and localization to CXCR4-containing endosomes and that CXCR4-containing EEA-1+ endosomes were abnormally located distal from the microtubule (MT)-organizing center (MTOC) in IQGAP1-deficient cells. Thus, IQGAP1 critically mediates CXCR4 cell surface expression and signaling, evidently by regulating EEA-1+ endosome interactions with MTs during CXCR4 trafficking and recycling. IQGAP1 may similarly promote CXCR4 functions in other cancer cell types.
Collapse
Affiliation(s)
- Adebowale O Bamidele
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905 Department of Immunology, Mayo Clinic, Rochester, MN 55905
| | | | - Petra Hirsova
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Ian C Clift
- Department of Immunology, Mayo Clinic, Rochester, MN 55905 Neurobiology of Disease Research Program, Mayo Clinic, Rochester, MN 55905
| | - Gregory J Gores
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905
| | - Daniel D Billadeau
- Department of Immunology, Mayo Clinic, Rochester, MN 55905 Division of Oncology Research, Mayo Clinic, Rochester, MN 55905
| | - Karen E Hedin
- Department of Immunology, Mayo Clinic, Rochester, MN 55905
| |
Collapse
|
30
|
Jean-Charles PY, Snyder JC, Shenoy SK. Chapter One - Ubiquitination and Deubiquitination of G Protein-Coupled Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 141:1-55. [PMID: 27378754 DOI: 10.1016/bs.pmbts.2016.05.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The seven-transmembrane containing G protein-coupled receptors (GPCRs) constitute the largest family of cell-surface receptors. Transmembrane signaling by GPCRs is fundamental to many aspects of physiology including vision, olfaction, cardiovascular, and reproductive functions as well as pain, behavior and psychomotor responses. The duration and magnitude of signal transduction is tightly controlled by a series of coordinated trafficking events that regulate the cell-surface expression of GPCRs at the plasma membrane. Moreover, the intracellular trafficking profiles of GPCRs can correlate with the signaling efficacy and efficiency triggered by the extracellular stimuli that activate GPCRs. Of the various molecular mechanisms that impart selectivity, sensitivity and strength of transmembrane signaling, ubiquitination of the receptor protein plays an important role because it defines both trafficking and signaling properties of the activated GPCR. Ubiquitination of proteins was originally discovered in the context of lysosome-independent degradation of cytosolic proteins by the 26S proteasome; however a large body of work suggests that ubiquitination also orchestrates the downregulation of membrane proteins in the lysosomes. In the case of GPCRs, such ubiquitin-mediated lysosomal degradation engenders long-term desensitization of transmembrane signaling. To date about 40 GPCRs are known to be ubiquitinated. For many GPCRs, ubiquitination plays a major role in postendocytic trafficking and sorting to the lysosomes. This chapter will focus on the patterns and functional roles of GPCR ubiquitination, and will describe various molecular mechanisms involved in GPCR ubiquitination.
Collapse
Affiliation(s)
- P-Y Jean-Charles
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC, United States
| | - J C Snyder
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States
| | - S K Shenoy
- Department of Medicine (Cardiology), Duke University Medical Center, Durham, NC, United States; Department of Cell Biology, Duke University Medical Center, Durham, NC, United States.
| |
Collapse
|
31
|
Desrochers G, Lussier-Price M, Omichinski JG, Angers A. Multiple Src Homology 3 Binding to the Ubiquitin Ligase Itch Conserved Proline-Rich Region. Biochemistry 2015; 54:7345-54. [PMID: 26613292 DOI: 10.1021/acs.biochem.5b01131] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Itch is a member of the C2-WW-HECT (CWH) family of ubiquitin ligases involved in the control of inflammatory signaling pathways, several transcription factors, and sorting of surface receptors to the degradative pathway. In addition to these common domains, Itch also contains a conserved proline-rich region (PRR) allowing its interaction with Src homology 3 (SH3) domain-containing proteins. This region is composed of 20 amino acids and contains one consensus class I and three class II SH3-binding motifs. Several SH3 domain-containing partners have been shown to recognize the Itch PRR, but their binding properties have been poorly defined. Here we compare a subset of endocytic SH3 domain-containing proteins using bioluminescence resonance energy transfer, isothermal titration calorimetry, and pull-down assays. Results indicate that Endophilin is a high-affinity binding partner of Itch both in vivo and in vitro, with a calculated KD placing this complex among the highest-affinity SH3 domain-mediated interactions reported to date. All of the SH3 domains tested here bind to Itch with a 1:1 stoichiometry, except for β-PIX that binds with a 2:1 stoichiometry. Together, these results indicate that Itch PRR is a versatile binding module that can accommodate several different SH3 domain-containing proteins but has a preference for Endophilin. Interestingly, the catalytic activity of Itch toward different SH3 domain-containing proteins was similar, except for β-PIX that was not readily ubiquitylated even though it could interact with an affinity comparable to those of other substrates tested.
Collapse
Affiliation(s)
- Guillaume Desrochers
- Department of Biological Sciences and ‡Department of Biochemistry and Molecular Medicine, University of Montreal , Montreal, Quebec H3C 3J7, Canada
| | - Mathieu Lussier-Price
- Department of Biological Sciences and ‡Department of Biochemistry and Molecular Medicine, University of Montreal , Montreal, Quebec H3C 3J7, Canada
| | - James G Omichinski
- Department of Biological Sciences and ‡Department of Biochemistry and Molecular Medicine, University of Montreal , Montreal, Quebec H3C 3J7, Canada
| | - Annie Angers
- Department of Biological Sciences and ‡Department of Biochemistry and Molecular Medicine, University of Montreal , Montreal, Quebec H3C 3J7, Canada
| |
Collapse
|
32
|
Tripathi A, Vana PG, Chavan TS, Brueggemann LI, Byron KL, Tarasova NI, Volkman BF, Gaponenko V, Majetschak M. Heteromerization of chemokine (C-X-C motif) receptor 4 with α1A/B-adrenergic receptors controls α1-adrenergic receptor function. Proc Natl Acad Sci U S A 2015; 112:E1659-68. [PMID: 25775528 PMCID: PMC4386352 DOI: 10.1073/pnas.1417564112] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recent evidence suggests that chemokine (C-X-C motif) receptor 4 (CXCR4) contributes to the regulation of blood pressure through interactions with α1-adrenergic receptors (ARs) in vascular smooth muscle. The underlying molecular mechanisms, however, are unknown. Using proximity ligation assays to visualize single-molecule interactions, we detected that α1A/B-ARs associate with CXCR4 on the cell surface of rat and human vascular smooth muscle cells (VSMC). Furthermore, α1A/B-AR could be coimmunoprecipitated with CXCR4 in a HeLa expression system and in human VSMC. A peptide derived from the second transmembrane helix of CXCR4 induced chemical shift changes in the NMR spectrum of CXCR4 in membranes, disturbed the association between α1A/B-AR and CXCR4, and inhibited Ca(2+) mobilization, myosin light chain (MLC) 2 phosphorylation, and contraction of VSMC upon α1-AR activation. CXCR4 silencing reduced α1A/B-AR:CXCR4 heteromeric complexes in VSMC and abolished phenylephrine-induced Ca(2+) fluxes and MLC2 phosphorylation. Treatment of rats with CXCR4 agonists (CXCL12, ubiquitin) reduced the EC50 of the phenylephrine-induced blood pressure response three- to fourfold. These observations suggest that disruption of the quaternary structure of α1A/B-AR:CXCR4 heteromeric complexes by targeting transmembrane helix 2 of CXCR4 and depletion of the heteromeric receptor complexes by CXCR4 knockdown inhibit α1-AR-mediated function in VSMC and that activation of CXCR4 enhances the potency of α1-AR agonists. Our findings extend the current understanding of the molecular mechanisms regulating α1-AR and provide an example of the importance of G protein-coupled receptor (GPCR) heteromerization for GPCR function. Compounds targeting the α1A/B-AR:CXCR4 interaction could provide an alternative pharmacological approach to modulate blood pressure.
Collapse
Affiliation(s)
- Abhishek Tripathi
- Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153
| | - P Geoff Vana
- Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153
| | - Tanmay S Chavan
- Department of Medicinal Chemistry, University of Illinois, Chicago, IL 60607
| | - Lioubov I Brueggemann
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153
| | - Kenneth L Byron
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153
| | - Nadya I Tarasova
- Cancer and Inflammation Program, National Cancer Institute, Frederick, MD 21702-1201
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226; and
| | - Vadim Gaponenko
- Department of Biochemistry and Molecular Genetics, University of Illinois, Chicago, IL 60607
| | - Matthias Majetschak
- Burn and Shock Trauma Research Institute, Department of Surgery, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153; Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago Stritch School of Medicine, Maywood, IL 60153;
| |
Collapse
|
33
|
Kennedy JE, Marchese A. Regulation of GPCR Trafficking by Ubiquitin. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:15-38. [PMID: 26055053 DOI: 10.1016/bs.pmbts.2015.02.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
G protein-coupled receptor (GPCR)-promoted signaling mediates cellular responses to a variety of stimuli involved in diverse physiological processes. In addition, GPCRs are also the largest class of target for many drugs used to treat a variety of diseases. Despite the role of GPCR signaling in health and disease, the molecular mechanisms governing GPCR signaling remain poorly understanding. Classically, GPCR signaling is tightly regulated by GPCR kinases and β-arrestins, which act in a concerted fashion to govern GPCR desensitization and also GPCR trafficking. Ubiquitination has now emerged as an important posttranslational modification that has multiple roles, either directly or indirectly, in governing GPCR trafficking. Recent studies have revealed a mechanistic link between GPCR phosphorylation, β-arrestins, and ubiquitination. Here, we review recent developments in our understanding of how ubiquitin regulates GPCR trafficking within the endocytic pathway.
Collapse
Affiliation(s)
- Justine E Kennedy
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Health Sciences Division, Maywood, Illinois, USA
| | - Adriano Marchese
- Department of Molecular Pharmacology and Therapeutics, Loyola University Chicago, Health Sciences Division, Maywood, Illinois, USA.
| |
Collapse
|
34
|
Verma R, Marchese A. The endosomal sorting complex required for transport pathway mediates chemokine receptor CXCR4-promoted lysosomal degradation of the mammalian target of rapamycin antagonist DEPTOR. J Biol Chem 2015; 290:6810-24. [PMID: 25605718 DOI: 10.1074/jbc.m114.606699] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
G protein-coupled receptor (GPCR) signaling mediates many cellular functions, including cell survival, proliferation, and cell motility. Many of these processes are mediated by GPCR-promoted activation of Akt signaling by mammalian target of rapamycin complex 2 (mTORC2) and the phosphatidylinositol 3-kinase (PI3K)/phosphoinositide-dependent kinase 1 (PDK1) pathway. However, the molecular mechanisms by which GPCRs govern Akt activation by these kinases remain poorly understood. Here, we show that the endosomal sorting complex required for transport (ESCRT) pathway mediates Akt signaling promoted by the chemokine receptor CXCR4. Pharmacological inhibition of heterotrimeric G protein Gαi or PI3K signaling and siRNA targeting ESCRTs blocks CXCR4-promoted degradation of DEPTOR, an endogenous antagonist of mTORC2 activity. Depletion of ESCRTs by siRNA leads to increased levels of DEPTOR and attenuated CXCR4-promoted Akt activation and signaling, consistent with decreased mTORC2 activity. In addition, ESCRTs likely have a broad role in Akt signaling because ESCRT depletion also attenuates receptor tyrosine kinase-promoted Akt activation and signaling. Our data reveal a novel role for the ESCRT pathway in promoting intracellular signaling, which may begin to identify the signal transduction pathways that are important in the physiological roles of ESCRTs and Akt.
Collapse
Affiliation(s)
- Rita Verma
- From the Biochemistry and Molecular Biology Program, and
| | - Adriano Marchese
- From the Biochemistry and Molecular Biology Program, and Department of Molecular Pharmacology and Therapeutics, Health Sciences Division, Loyola University Chicago, Maywood, Illinois 60153
| |
Collapse
|
35
|
Itch is required for lateral line development in zebrafish. PLoS One 2014; 9:e111799. [PMID: 25369329 PMCID: PMC4219781 DOI: 10.1371/journal.pone.0111799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2014] [Accepted: 10/03/2014] [Indexed: 11/20/2022] Open
Abstract
The zebrafish posterior lateral line is formed during early development by the deposition of neuromasts from a migrating primordium. The molecular mechanisms regulating the regional organization and migration of the primordium involve interactions between Fgf and Wnt/β-catenin signaling and the establishment of specific cxcr4b and cxcr7b cytokine receptor expression domains. Itch has been identified as a regulator in several different signaling pathways, including Wnt and Cxcr4 signaling. We identified two homologous itch genes in zebrafish, itcha and itchb, with generalized expression patterns. By reducing itchb expression in particular upon morpholino knockdown, we demonstrated the importance of Itch in regulating lateral line development by perturbing the patterns of cxcr4b and cxcr7b expression. Itch knockdown results in a failure to down-regulate Wnt signaling and overexpression of cxcr4b in the primordium, slowing migration of the posterior lateral line primordium and resulting in abnormal development of the lateral line.
Collapse
|
36
|
Holleman J, Marchese A. The ubiquitin ligase deltex-3l regulates endosomal sorting of the G protein-coupled receptor CXCR4. Mol Biol Cell 2014; 25:1892-904. [PMID: 24790097 PMCID: PMC4055268 DOI: 10.1091/mbc.e13-10-0612] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
G protein-coupled receptor (GPCR) sorting into the degradative pathway is important for limiting the duration and magnitude of signaling. Agonist activation of the GPCR CXCR4 induces its rapid ubiquitination and sorting to lysosomes via the endosomal sorting complex required for transport (ESCRT) pathway. We recently reported that ESCRT-0 ubiquitination is linked to the efficiency with which CXCR4 is sorted for lysosomal degradation; however mechanistic insight is lacking. Here we define a novel role for the really interesting new gene-domain E3 ubiquitin ligase deltex-3-like (DTX3L) in regulating CXCR4 sorting from endosomes to lysosomes. We show that DTX3L localizes to early endosomes upon CXCR4 activation and interacts directly with and inhibits the activity of the E3 ubiquitin ligase atrophin-1 interacting protein 4. This serves to limit the extent to which ESCRT-0 is ubiquitinated and is able to sort CXCR4 for lysosomal degradation. Therefore we define a novel role for DTX3L in GPCR endosomal sorting and reveal an unprecedented link between two distinct E3 ubiquitin ligases to control the activity of the ESCRT machinery.
Collapse
Affiliation(s)
- Justine Holleman
- Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153
| | - Adriano Marchese
- Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, Maywood, IL 60153
| |
Collapse
|
37
|
Raehal KM, Bohn LM. β-arrestins: regulatory role and therapeutic potential in opioid and cannabinoid receptor-mediated analgesia. Handb Exp Pharmacol 2014; 219:427-43. [PMID: 24292843 PMCID: PMC4804701 DOI: 10.1007/978-3-642-41199-1_22] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Pain is a complex disorder with neurochemical and psychological components contributing to the severity, the persistence, and the difficulty in adequately treating the condition. Opioid and cannabinoids are two classes of analgesics that have been used to treat pain for centuries and are arguably the oldest of "pharmacological" interventions used by man. Unfortunately, they also produce several adverse side effects that can complicate pain management. Opioids and cannabinoids act at G protein-coupled receptors (GPCRs), and much of their effects are mediated by the mu-opioid receptor (MOR) and cannabinoid CB1 receptor (CB1R), respectively. These receptors couple to intracellular second messengers and regulatory proteins to impart their biological effects. In this chapter, we review the role of the intracellular regulatory proteins, β-arrestins, in modulating MOR and CB1R and how they influence the analgesic and side-effect profiles of opioid and cannabinoid drugs in vivo. This review of the literature suggests that the development of opioid and cannabinoid agonists that bias MOR and CB1R toward G protein signaling cascades and away from β-arrestin interactions may provide a novel mechanism by which to produce analgesia with less severe adverse effects.
Collapse
MESH Headings
- Analgesics/adverse effects
- Analgesics/pharmacology
- Analgesics, Opioid/adverse effects
- Analgesics, Opioid/pharmacology
- Animals
- Arrestins/metabolism
- Cannabinoids/adverse effects
- Cannabinoids/pharmacology
- Drug Design
- Humans
- Pain/drug therapy
- Pain/physiopathology
- Receptor, Cannabinoid, CB1/drug effects
- Receptor, Cannabinoid, CB1/metabolism
- Receptors, G-Protein-Coupled/drug effects
- Receptors, G-Protein-Coupled/metabolism
- Receptors, Opioid, mu/drug effects
- Receptors, Opioid, mu/metabolism
- beta-Arrestins
Collapse
Affiliation(s)
- Kirsten M Raehal
- The Scripps Research Institute, 130 Scripps Way #2A2, Jupiter, FL, 33458, USA,
| | | |
Collapse
|
38
|
Endocytic trafficking of chemokine receptors. Curr Opin Cell Biol 2013; 27:72-7. [PMID: 24680433 DOI: 10.1016/j.ceb.2013.11.011] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Revised: 11/15/2013] [Accepted: 11/24/2013] [Indexed: 12/23/2022]
Abstract
Chemokine receptors belong to the super family of G protein-coupled receptors (GPCRs). The cognate ligands for chemokine receptors are small circulating proteins known as chemokines. Upon binding to their cognate chemokines, receptors are rapidly desensitized, internalized onto early endosomes and sorted either into a recycling pathway or degradative pathway. Chemokine receptor trafficking is essential because it limits the magnitude and duration of signaling by removing receptors from the cell surface thereby limiting access to their ligands, but it also delivers bound chemokines to lysosomes for degradation. Receptor sorting into the recycling pathway contributes to resensitization of receptor signaling, whereas sorting into the degradative pathway leads to long-term attenuation of signaling. Recent studies have revealed some key information regarding the molecular determinants mediating chemokine receptor internalization and have shed light on the mechanisms dictating sorting into either the recycling or degradative pathways. Here I discuss our current understanding of the mechanisms mediating chemokine receptor trafficking with a focus primarily on recent findings for the chemokine receptor CXCR4.
Collapse
|
39
|
Shu L, Zhang H, Boyce B, Xing L. Ubiquitin E3 ligase Wwp1 negatively regulates osteoblast function by inhibiting osteoblast differentiation and migration. J Bone Miner Res 2013; 28:1925-35. [PMID: 23553732 PMCID: PMC3749248 DOI: 10.1002/jbmr.1938] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 03/01/2013] [Accepted: 03/20/2013] [Indexed: 01/09/2023]
Abstract
Ubiquitin E3 ligase-mediated protein degradation promotes proteasomal degradation of key positive regulators of osteoblast functions. For example, the E3 ligases--SMAD-specific E3 ubiquitin protein ligase 1 (Smurf1), Itch, and WW domain-containing E3 ubiquitin protein ligase 1 (Wwp1)--promote degradation of Runt-related transcription factor 2 (Runx2), transcription factor jun-B (JunB), and chemokine (C-X-C) receptor type 4 (CXCR-4) proteins to inhibit their functions. However, the role of E3 ligases in age-associated bone loss is unknown. We found that the expression level of Wwp1, but not Smurf1 or Itch, was significantly increased in CD45-negative (CD45(-)) bone marrow-derived mesenchymal stem cells from 6-month-old and 12-month-old wild-type (WT) mice. Wwp1 knockout (Wwp1(-/-)) mice developed increased bone mass as they aged, associated with increased bone formation rates and normal bone resorption parameters. Bone marrow stromal cells (BMSCs) from Wwp1(-/-) mice formed increased numbers and areas of alkaline phosphatase(+) and Alizarin red(+) nodules and had increased migration potential toward chemokine (C-X-C motif) ligand 12 (CXCL12) gradients. Runx2, JunB, and CXCR-4 protein levels were significantly increased in Wwp1(-/-) BMSCs. Wwp1(-/-) BMSCs had increased amount of ubiquitinated JunB protein, but Runx2 ubiquitination was no change. Knocking down JunB in Wwp1(-/-) BMSCs returned Runx2 protein levels to that in WT cells. Thus, Wwp1 negatively regulates osteoblast functions by affecting both their migration and differentiation. Mechanisms designed to decrease Wwp1 levels in BMSCs may represent a new approach to prevent the decrease in osteoblastic bone formation associated with aging.
Collapse
Affiliation(s)
| | | | | | - Lianping Xing
- Correspondence to: Lianping Xing, Department of Pathology and Laboratory Medicine, 601 Elmwood Ave, Box 626, Rochester, NY 14642, USA. Phone (585) 273-4090, Fax (585) 756-4468,
| |
Collapse
|
40
|
Cottrell GS. Roles of proteolysis in regulation of GPCR function. Br J Pharmacol 2013; 168:576-90. [PMID: 23043558 DOI: 10.1111/j.1476-5381.2012.02234.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Revised: 09/03/2012] [Accepted: 09/24/2012] [Indexed: 12/18/2022] Open
Abstract
The enzymatic activity of peptidases must be tightly regulated to prevent uncontrolled hydrolysis of peptide bonds, which could have devastating effects on biological systems. Peptidases are often generated as inactive propeptidases, secreted with endogenous inhibitors, or they are compartmentalized. Propeptidases become active after proteolytic removal of N-terminal activation peptides by other peptidases. Some peptidases only become active towards substrates only at certain pHs, thus confining activity to specific compartments or conditions. This review discusses the different roles proteolysis plays in regulating GPCRs. At the cell-surface, certain GPCRs are regulated by the hydrolytic inactivation of bioactive peptides by membrane-anchored peptidases, which prevent signalling. Conversely, cell-surface peptidases can also generate bioactive peptides, which directly activate GPCRs. Alternatively, cell-surface peptidases activated by GPCRs, can generate bioactive peptides to cause transactivation of receptor tyrosine kinases, thereby promoting signalling. Certain peptidases can signal directly to cells, by cleaving GPCR to initiate intracellular signalling cascades. Intracellular peptidases also regulate GPCRs; lysosomal peptidases destroy GPCRs in lysosomes to permanently terminate signalling and mediate down-regulation; endosomal peptidases cleave internalized peptide agonists to regulate GPCR recycling, resensitization and signalling; and soluble intracellular peptidases also participate in GPCR function by regulating the ubiquitination state of GPCRs, thereby altering GPCR signalling and fate. Although the use of peptidase inhibitors has already brought success in the treatment of diseases such as hypertension, the discovery of new regulatory mechanisms involving proteolysis that control GPCRs may provide additional targets to modulate dysregulated GPCR signalling in disease.
Collapse
Affiliation(s)
- G S Cottrell
- Reading School of Pharmacy, University of Reading, Reading, UK.
| |
Collapse
|
41
|
Marchese A, Trejo J. Ubiquitin-dependent regulation of G protein-coupled receptor trafficking and signaling. Cell Signal 2013; 25:707-16. [PMID: 23201781 PMCID: PMC3593103 DOI: 10.1016/j.cellsig.2012.11.024] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 11/25/2012] [Indexed: 01/07/2023]
Abstract
G protein-coupled receptors (GPCRs) belong to one of the largest family of signaling receptors in the mammalian genome [1]. GPCRs elicit cellular responses to multiple diverse stimuli and play essential roles in human health and disease. GPCRs have important clinical implications in various diseases and are the targets of approximately 25-50% of all marketed drugs [2,3]. Understanding how GPCRs are regulated is essential to delineating their role in normal physiology and in the pathophysiology of several diseases. Given the vast number and diversity of GPCRs, it is likely that multiple mechanisms exist to regulate GPCR function. While GPCR signaling is typically regulated by desensitization and endocytosis mediated by phosphorylation and β-arrestins, it can also be modulated by ubiquitination. Ubiquitination is emerging an important regulatory process that may have unique roles in governing GPCR trafficking and signaling. Recent studies have revealed a mechanistic link between GPCR phosphorylation, β-arrestins and ubiquitination that may be applicable to some GPCRs but not others. While the function of ubiquitination is generally thought to promote receptor endocytosis and endosomal sorting, recent studies have revealed that ubiquitination also plays an important role in positive regulation of GPCR signaling. Here, we will review recent developments in our understanding of how ubiquitin regulates GPCR endocytic trafficking and how it contributes to signal transduction induced by GPCR activation.
Collapse
Affiliation(s)
- Adriano Marchese
- Department of Molecular Pharmacology and Therapeutics, Stritch School of Medicine, Loyola University Chicago, 2160 S. 1 Ave., Building 101; Room 2721, Maywood, IL 60153
| | - JoAnn Trejo
- Department of Pharmacology, School of Medicine, University of California, San Diego, 9500 Gilman Drive, Biomedical Sciences Building, Room 3044A, La Jolla, CA 92093
| |
Collapse
|
42
|
Kommaddi RP, Shenoy SK. Arrestins and protein ubiquitination. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 118:175-204. [PMID: 23764054 DOI: 10.1016/b978-0-12-394440-5.00007-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The adaptor proteins, β-arrestins 1 and 2, were originally identified as inhibitors of G protein signaling at the seven-transmembrane receptors (7TMRs, also called G protein-coupled receptors or GPCRs). Subsequent studies have established β-arrestins as critical multifunctional 7TMR adaptors that mediate receptor trafficking and activate G protein-independent signaling pathways. 7TMR activation leads not only to the recruitment of arrestin proteins upon phosphorylation by GPCR kinases but also to β-arrestin ubiquitination. This posttranslational modification of β-arrestin is appended by specific E3 ubiquitin ligases and reversed by deubiquitinases, which are also recruited in a receptor- and agonist-specific manner. β-Arrestin ubiquitination allows it to form protein complexes with activated 7TMRs, endocytic proteins such as clathrin, and phosphorylated ERK1/2. β-Arrestin ubiquitination is dependent on its activated conformation and likely regulates timing and subcellular localization of various protein interactions during receptor trafficking and signaling. β-Arrestins also serve as adaptors that escort E3 ubiquitin ligases to mediate ubiquitination of a wide list of substrate proteins including 7TMRs and provide an added layer of regulation for defining substrate specificity in the cellular ubiquitination pathway.
Collapse
Affiliation(s)
- Reddy Peera Kommaddi
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | | |
Collapse
|