1
|
Mavri M, Glišić S, Senćanski M, Vrecl M, Rosenkilde MM, Spiess K, Kubale V. Patterns of human and porcine gammaherpesvirus-encoded BILF1 receptor endocytosis. Cell Mol Biol Lett 2023; 28:14. [PMID: 36810008 PMCID: PMC9942385 DOI: 10.1186/s11658-023-00427-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 01/30/2023] [Indexed: 02/23/2023] Open
Abstract
BACKGROUND The viral G-protein-coupled receptor (vGPCR) BILF1 encoded by the Epstein-Barr virus (EBV) is an oncogene and immunoevasin and can downregulate MHC-I molecules at the surface of infected cells. MHC-I downregulation, which presumably occurs through co-internalization with EBV-BILF1, is preserved among BILF1 receptors, including the three BILF1 orthologs encoded by porcine lymphotropic herpesviruses (PLHV BILFs). This study aimed to understand the detailed mechanisms of BILF1 receptor constitutive internalization, to explore the translational potential of PLHV BILFs compared with EBV-BILF1. METHODS A novel real-time fluorescence resonance energy transfer (FRET)-based internalization assay combined with dominant-negative variants of dynamin-1 (Dyn K44A) and the chemical clathrin inhibitor Pitstop2 in HEK-293A cells was used to study the effect of specific endocytic proteins on BILF1 internalization. Bioluminescence resonance energy transfer (BRET)-saturation analysis was used to study BILF1 receptor interaction with β-arrestin2 and Rab7. In addition, a bioinformatics approach informational spectrum method (ISM) was used to investigate the interaction affinity of BILF1 receptors with β-arrestin2, AP-2, and caveolin-1. RESULTS We identified dynamin-dependent, clathrin-mediated constitutive endocytosis for all BILF1 receptors. The observed interaction affinity between BILF1 receptors and caveolin-1 and the decreased internalization in the presence of a dominant-negative variant of caveolin-1 (Cav S80E) indicated the involvement of caveolin-1 in BILF1 trafficking. Furthermore, after BILF1 internalization from the plasma membrane, both the recycling and degradation pathways are proposed for BILF1 receptors. CONCLUSIONS The similarity in the internalization mechanisms observed for EBV-BILF1 and PLHV1-2 BILF1 provide a foundation for further studies exploring a possible translational potential for PLHVs, as proposed previously, and provides new information about receptor trafficking.
Collapse
Affiliation(s)
- Maša Mavri
- Institute for preclinical sciences, Veterinary Faculty, Ljubljana, Slovenia
| | - Sanja Glišić
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Milan Senćanski
- Center for Multidisciplinary Research, Institute of Nuclear Sciences VINCA, University of Belgrade, Belgrade, Serbia
| | - Milka Vrecl
- Institute for preclinical sciences, Veterinary Faculty, Ljubljana, Slovenia
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Katja Spiess
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Virus and Microbiological Special Diagnostics, Statens Serum Institute, Copenhagen, Denmark
| | - Valentina Kubale
- Institute for preclinical sciences, Veterinary Faculty, Ljubljana, Slovenia.
| |
Collapse
|
2
|
XCL1, a serum biomarker in neurological diseases; HTLV-1-associated myelopathy and multiple sclerosis. Microb Pathog 2023; 174:105962. [PMID: 36572194 DOI: 10.1016/j.micpath.2022.105962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 08/14/2022] [Accepted: 12/22/2022] [Indexed: 12/24/2022]
Abstract
The XCL1-XCR1 axis has a potential role in the recruitment of immune cells to the site of inflammation. The present study aimed to examine the relation of XCL1 serum levels with Multiple sclerosis (MS) and HTLV-1-associated myelopathy (HAM), as chronic inflammatory diseases of the central nervous system (CNS). DNA was extracted to evaluate HTLV-1 proviral load (PVL) using real-time PCR. Serum levels of XCL1 was determined by using an ELISA assay. The serum level of XCL1 was significantly higher in patients with HAM than that of asymptomatic carriers (ACs) and healthy controls (HCs) (p < 0.001 and p < 0.0001, respectively) and was also higher in MS patients compared to HCs (p < 0.0001). Moreover, the concentration of XCL1 serum level was significantly different between the ACs and HCs group (p < 0.0001). In conclusion, increased expression of XCL1 might contribute to the migration of autoreactive T cells to the central nervous system and play a critical role in the development and pathogenesis of inflammatory neurological diseases including HAM and MS.
Collapse
|
3
|
Berg C, Wedemeyer MJ, Melynis M, Schlimgen RR, Hansen LH, Våbenø J, Peterson FC, Volkman BF, Rosenkilde MM, Lüttichau HR. The non-ELR CXC chemokine encoded by human cytomegalovirus UL146 genotype 5 contains a C-terminal β-hairpin and induces neutrophil migration as a selective CXCR2 agonist. PLoS Pathog 2022; 18:e1010355. [PMID: 35271688 PMCID: PMC8939814 DOI: 10.1371/journal.ppat.1010355] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 03/22/2022] [Accepted: 02/09/2022] [Indexed: 11/19/2022] Open
Abstract
Human cytomegalovirus (HCMV) is a major pathogen in immunocompromised patients. The UL146 gene exists as 14 diverse genotypes among clinical isolates, which encode 14 different CXC chemokines. One genotype (vCXCL1GT1) is a known agonist for CXCR1 and CXCR2, while two others (vCXCL1GT5 and vCXCL1GT6) lack the ELR motif considered crucial for CXCR1 and CXCR2 binding, thus suggesting another receptor targeting profile. To determine the receptor target for vCXCL1GT5, the chemokine was probed in a G protein signaling assay on all 18 classical human chemokine receptors, where CXCR2 was the only receptor being activated. In addition, vCXCL1GT5 recruited β-arrestin in a BRET-based assay and induced migration in a chemotaxis assay through CXCR2, but not CXCR1. In contrast, vCXCL1GT1 stimulated G protein signaling, recruited β-arrestin and induced migration through both CXCR1 and CXCR2. Both vCXCL1GT1 and vCXCL1GT5 induced equally potent and efficacious migration of neutrophils, and ELR vCXCL1GT4 and non-ELR vCXCL1GT6 activated only CXCR2. In contrast to most human chemokines, the 14 UL146 genotypes have remarkably long C-termini. Comparative modeling using Rosetta showed that each genotype could adopt the classic chemokine core structure, and predicted that the extended C-terminal tail of several genotypes (including vCXCL1GT1, vCXCL1GT4, vCXCL1GT5, and vCXCL1GT6) forms a novel β-hairpin not found in human chemokines. Secondary NMR shift and TALOS+ analysis of vCXCL1GT1 supported the existence of two stable β-strands. C-terminal deletion of vCXCL1GT1 resulted in a non-functional protein and in a shift to solvent exposure for tryptophan residues likely due to destabilization of the chemokine fold. The results demonstrate that non-ELR chemokines can activate CXCR2 and suggest that the UL146 chemokines have unique C-terminal structures that stabilize the chemokine fold. Increased knowledge of the structure and interaction partners of the chemokine variants encoded by UL146 is key to understanding why circulating HCMV strains sustain 14 stable genotypes. Human cytomegalovirus (HCMV) is a prevalent herpesvirus infecting an estimated 60% of the human population worldwide. It is commonly transmitted during early childhood and leads to life-long latency, where viral reactivation can cause severe complications in case of host immune suppression. Furthermore, HCMV is the leading cause of congenital infections. Circulating HCMV strains exhibit great genetic diversity unusual for DNA viruses. One of its most diverse genes is UL146, which encodes a chemokine that facilitates viral dissemination by exploiting the human immune system through mimicry of key immunity components. In this study, we investigate how the diversity of UL146 affects its signaling and structural properties to understand why its genetic diversity is maintained across human populations. We find that certain genotypes that lack key structural domains present in the human homologs nonetheless exert similar functions in the virus-host relationship. Furthermore, many of the UL146 genotypes contain novel structural elements critical for correct protein folding and with the potential to provide HCMV with additional immune modulatory and evasive features. Together, our data highlight a considerable degree of host-adaptation by HCMV and propose novel structural interactions with implications for the virus-host interplay.
Collapse
Affiliation(s)
- Christian Berg
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
- Unit for Infectious Diseases, Department of Medicine, Herlev-Gentofte Hospital, University of Copenhagen, Herlev, Denmark
| | - Michael J. Wedemeyer
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Motiejus Melynis
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Roman R. Schlimgen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Lasse H. Hansen
- Department of Clinical Biochemistry, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen N, Denmark
| | - Jon Våbenø
- Helgeland Hospital Trust, Sandnessjøen, Norway
| | - Francis C. Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Brian F. Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Mette M. Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
- * E-mail: (MMR); (HRL)
| | - Hans R. Lüttichau
- Laboratory for Molecular Pharmacology, Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Copenhagen, Denmark
- Unit for Infectious Diseases, Department of Medicine, Herlev-Gentofte Hospital, University of Copenhagen, Herlev, Denmark
- * E-mail: (MMR); (HRL)
| |
Collapse
|
4
|
Mavri M, Kubale V, Depledge DP, Zuo J, Huang CA, Breuer J, Vrecl M, Jarvis MA, Jovičić EJ, Petan T, Ehlers B, Rosenkilde MM, Spiess K. Epstein-Barr Virus-Encoded BILF1 Orthologues From Porcine Lymphotropic Herpesviruses Display Common Molecular Functionality. Front Endocrinol (Lausanne) 2022; 13:862940. [PMID: 35721730 PMCID: PMC9204316 DOI: 10.3389/fendo.2022.862940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/19/2022] [Indexed: 11/13/2022] Open
Abstract
Infection of immunosuppressed transplant patients with the human γ-herpesvirus Epstein-Barr virus (EBV) is associated with post-transplant lymphoproliferative disease (PTLD), an often fatal complication. Immunosuppressed miniature pigs infected with γ-herpesvirus porcine lymphotropic herpesvirus 1 (PLHV1) develop a similar disease, identifying pigs as a potential preclinical model for PTLD in humans. BILF1 is a G protein-coupled receptor (GPCR) encoded by EBV with constitutive activity linked to tumorigenesis and immunoevasive function downregulating MHC-I. In the present study, we compared BILF1-orthologues encoded by the three known PLHVs (PLHV1-3) with EBV-BILF1 to determine pharmacological suitability of BILF1 orthologues as model system to study EBV-BILF1 druggability. Cell surface localization, constitutive internalization, and MHC-I downregulation as well as membrane proximal constitutive Gαi signaling patterns were conserved across all BILFs. Only subtle differences between the individual BILFs were observed in downstream transcription factor activation. Using Illumina sequencing, PLHV1 was observed in lymphatic tissue from PTLD-diseased, but not non-diseased pigs. Importantly, these tissues showed enhanced expression of PLHV1-BILF1 supporting its involvement in PTLD infection.
Collapse
Affiliation(s)
- Maša Mavri
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Valentina Kubale
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Daniel P. Depledge
- Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Jianmin Zuo
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Christene A. Huang
- Department of Surgery, Division of Plastic & Reconstructive Surgery, Division of Transplant Surgery, Anschutz Medical Campus, University of Colorado, Denver, CO, United States
| | - Judith Breuer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Milka Vrecl
- Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Michael A. Jarvis
- The Vaccine Group Ltd, Plymouth; and the University of Plymouth, Plymouth, United Kingdom
| | - Eva Jarc Jovičić
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Toni Petan
- Department of Molecular and Biomedical Sciences, Jožef Stefan Institute, Ljubljana, Slovenia
| | - Bernhard Ehlers
- Division 12, Measles, Mumps, Rubella, and Viruses Affecting Immunocompromised Patients, Robert Koch Institute, Berlin, Germany
| | - Mette M. Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Katja Spiess, ; ; Mette M. Rosenkilde,
| | - Katja Spiess
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- *Correspondence: Katja Spiess, ; ; Mette M. Rosenkilde,
| |
Collapse
|
5
|
Mavri M, Spiess K, Rosenkilde MM, Rutland CS, Vrecl M, Kubale V. Methods for Studying Endocytotic Pathways of Herpesvirus Encoded G Protein-Coupled Receptors. Molecules 2020; 25:E5710. [PMID: 33287269 PMCID: PMC7730005 DOI: 10.3390/molecules25235710] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 11/20/2020] [Accepted: 12/01/2020] [Indexed: 12/20/2022] Open
Abstract
Endocytosis is a fundamental process involved in trafficking of various extracellular and transmembrane molecules from the cell surface to its interior. This enables cells to communicate and respond to external environments, maintain cellular homeostasis, and transduce signals. G protein-coupled receptors (GPCRs) constitute a family of receptors with seven transmembrane alpha-helical domains (7TM receptors) expressed at the cell surface, where they regulate physiological and pathological cellular processes. Several herpesviruses encode receptors (vGPCRs) which benefits the virus by avoiding host immune surveillance, supporting viral dissemination, and thereby establishing widespread and lifelong infection, processes where receptor signaling and/or endocytosis seem central. vGPCRs are rising as potential drug targets as exemplified by the cytomegalovirus-encoded receptor US28, where its constitutive internalization has been exploited for selective drug delivery in virus infected cells. Therefore, studying GPCR trafficking is of great importance. This review provides an overview of the current knowledge of endocytic and cell localization properties of vGPCRs and methodological approaches used for studying receptor internalization. Using such novel approaches, we show constitutive internalization of the BILF1 receptor from human and porcine γ-1 herpesviruses and present motifs from the eukaryotic linear motif (ELM) resources with importance for vGPCR endocytosis.
Collapse
Affiliation(s)
- Maša Mavri
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (M.M.); (M.V.)
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (K.S.); (M.M.R.)
| | - Katja Spiess
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (K.S.); (M.M.R.)
| | - Mette Marie Rosenkilde
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark; (K.S.); (M.M.R.)
| | - Catrin Sian Rutland
- School of Veterinary Medicine and Science, Medical Faculty, Sutton, Bonington Campus, University of Nottingham, Sutton Bonington LE12 5RD, UK;
| | - Milka Vrecl
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (M.M.); (M.V.)
| | - Valentina Kubale
- Department of Anatomy, Histology with Embryology and Cytology, Institute of Preclinical Sciences, Veterinary Faculty, University of Ljubljana, Gerbičeva 60, 1000 Ljubljana, Slovenia; (M.M.); (M.V.)
| |
Collapse
|
6
|
Bauer A, Madela J, Berg C, Daugvilaite V, Gurka S, Mages HW, Kroczek RA, Rosenkilde MM, Voigt S. Rat cytomegalovirus-encoded γ-chemokine vXCL1 is a highly adapted, species-specific agonist for rat XCR1-positive dendritic cells. J Cell Sci 2019; 133:jcs.236190. [PMID: 31649144 DOI: 10.1242/jcs.236190] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 10/22/2019] [Indexed: 12/22/2022] Open
Abstract
Dendritic cells (DCs) expressing the chemokine receptor XCR1 are specialized in antigen cross-presentation to control infections with intracellular pathogens. XCR1-positive (XCR1+) DCs are attracted by XCL1, a γ-chemokine secreted by activated CD8+ T cells and natural killer cells. Rat cytomegalovirus (RCMV) is the only virus known to encode a viral XCL1 analog (vXCL1) that competes for XCR1 binding with the endogenous chemokine. Here we show that vXCL1 from two different RCMV strains, as well as endogenous rat XCL1 (rXCL1) bind to and induce chemotaxis exclusively in rat XCR1+ DCs. Whereas rXCL1 activates the XCR1 Gi signaling pathway in rats and humans, both of the vXCL1s function as species-specific agonists for rat XCR1. In addition, we demonstrate constitutive internalization of XCR1 in XCR1-transfected HEK293A cells and in splenic XCR1+ DCs. This internalization was independent of β-arrestin 1 and 2 and was enhanced after binding of vXCL1 and rXCL1; however, vXCL1 appeared to be a stronger agonist. These findings suggest a decreased surface expression of XCR1 during DC cultivation at 37°C, and subsequent impairment of chemotactic activity and XCR1+ DC function.
Collapse
Affiliation(s)
- Agnieszka Bauer
- Department of Infectious Diseases, Robert Koch Institute, 13353 Berlin, Germany
| | - Julia Madela
- Department of Infectious Diseases, Robert Koch Institute, 13353 Berlin, Germany
| | - Christian Berg
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark.,Infectious Diseases Unit, Department of Medicine, Herlev-Gentofte Hospital, University of Copenhagen, 2730 Herlev, Denmark
| | - Viktorija Daugvilaite
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Stephanie Gurka
- Molecular Immunology, Robert Koch Institute, 13353 Berlin, Germany
| | - Hans Werner Mages
- Centre for biological threats and special pathogens, Robert Koch Institute, 13353 Berlin, Germany
| | | | - Mette M Rosenkilde
- Faculty of Health and Medical Sciences, Department of Biomedical Sciences, The Panum Institute, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sebastian Voigt
- Department of Infectious Diseases, Robert Koch Institute, 13353 Berlin, Germany .,Department of Pediatric Oncology/Hematology/Stem Cell Transplantation, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
7
|
Abraham M, Wald H, Vaizel-Ohayon D, Grabovsky V, Oren Z, Karni A, Weiss L, Galun E, Peled A, Eizenberg O. Development of Novel Promiscuous Anti-Chemokine Peptibodies for Treating Autoimmunity and Inflammation. Front Immunol 2017; 8:1432. [PMID: 29218043 PMCID: PMC5703867 DOI: 10.3389/fimmu.2017.01432] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 10/13/2017] [Indexed: 12/27/2022] Open
Abstract
Chemokines and their receptors play critical roles in the progression of autoimmunity and inflammation. Typically, multiple chemokines are involved in the development of these pathologies. Indeed, targeting single chemokines or chemokine receptors has failed to achieve significant clinical benefits in treating autoimmunity and inflammation. Moreover, the binding of host atypical chemokine receptors to multiple chemokines as well as the binding of chemokine-binding proteins secreted by various pathogens can serve as a strategy for controlling inflammation. In this work, promiscuous chemokine-binding peptides that could bind and inhibit multiple inflammatory chemokines, such as CCL2, CCL5, and CXCL9/10/11, were selected from phage display libraries. These peptides were cloned into human mutated immunoglobulin Fc-protein fusions (peptibodies). The peptibodies BKT120Fc and BKT130Fc inhibited the ability of inflammatory chemokines to induce the adhesion and migration of immune cells. Furthermore, BKT120Fc and BKT130Fc also showed a significant inhibition of disease progression in a variety of animal models for autoimmunity and inflammation. Developing a novel class of antagonists that can control the courses of diseases by selectively blocking multiple chemokines could be a novel way of generating effective therapeutics.
Collapse
Affiliation(s)
| | - Hanna Wald
- Biokine Therapeutics Ltd, Ness Ziona, Israel
| | | | | | - Zohar Oren
- Biokine Therapeutics Ltd, Ness Ziona, Israel
| | - Arnon Karni
- Department of Neurology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Lola Weiss
- Goldyne Savad Institute of Gene Therapy, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Eithan Galun
- Goldyne Savad Institute of Gene Therapy, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Amnon Peled
- Biokine Therapeutics Ltd, Ness Ziona, Israel.,Goldyne Savad Institute of Gene Therapy, Hebrew University of Jerusalem, Jerusalem, Israel
| | | |
Collapse
|
8
|
Varicella zoster virus glycoprotein C increases chemokine-mediated leukocyte migration. PLoS Pathog 2017; 13:e1006346. [PMID: 28542541 PMCID: PMC5444840 DOI: 10.1371/journal.ppat.1006346] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Accepted: 04/11/2017] [Indexed: 02/07/2023] Open
Abstract
Varicella zoster virus (VZV) is a highly prevalent human pathogen that establishes latency in neurons of the peripheral nervous system. Primary infection causes varicella whereas reactivation results in zoster, which is often followed by chronic pain in adults. Following infection of epithelial cells in the respiratory tract, VZV spreads within the host by hijacking leukocytes, including T cells, in the tonsils and other regional lymph nodes, and modifying their activity. In spite of its importance in pathogenesis, the mechanism of dissemination remains poorly understood. Here we addressed the influence of VZV on leukocyte migration and found that the purified recombinant soluble ectodomain of VZV glycoprotein C (rSgC) binds chemokines with high affinity. Functional experiments show that VZV rSgC potentiates chemokine activity, enhancing the migration of monocyte and T cell lines and, most importantly, human tonsillar leukocytes at low chemokine concentrations. Binding and potentiation of chemokine activity occurs through the C-terminal part of gC ectodomain, containing predicted immunoglobulin-like domains. The mechanism of action of VZV rSgC requires interaction with the chemokine and signalling through the chemokine receptor. Finally, we show that VZV viral particles enhance chemokine-dependent T cell migration and that gC is partially required for this activity. We propose that VZV gC activity facilitates the recruitment and subsequent infection of leukocytes and thereby enhances VZV systemic dissemination in humans. Varicella zoster virus (VZV) causes two main pathologies in humans, chickenpox during primary infection, and shingles following reactivation. The latter is a painful condition that is often followed by chronic pain in a large numbers of shingles patients. Despite the existence of a vaccine, shingles-related complications cause expenses of more than $1 billion per year in the USA alone. Following primary infection, the virus infects leukocytes including T cells, spreading to the skin causing chickenpox. Direct infection of neurons from leukocytes has also been postulated. Given the relevance of leukocytes in VZV biology and the importance of chemokines in directing their migration, we investigated whether VZV modulates the function of chemokines. Our results show that VZV glycoprotein C potentiates the activity of chemokines, inducing higher migration of human leukocytes at low chemokine concentration. This may attract additional susceptible leukocytes to the site of infection enhancing virus spread and pathogenesis.
Collapse
|
9
|
Frank T, Reichel A, Larsen O, Stilp AC, Rosenkilde MM, Stamminger T, Ozawa T, Tschammer N. Attenuation of chemokine receptor function and surface expression as an immunomodulatory strategy employed by human cytomegalovirus is linked to vGPCR US28. Cell Commun Signal 2016; 14:31. [PMID: 27955674 PMCID: PMC5153698 DOI: 10.1186/s12964-016-0154-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 12/02/2016] [Indexed: 01/08/2023] Open
Abstract
Background Some herpesviruses like human cytomegalovirus (HCMV) encode viral G protein-coupled receptors that cause reprogramming of cell signaling to facilitate dissemination of the virus, prevent immune surveillance and establish life-long latency. Human GPCRs are known to function in complex signaling networks involving direct physical interactions as well as indirect crosstalk of orthogonal signaling networks. The human chemokine receptor CXCR4 is expressed on hematopoietic stem cells, leukocytes, endothelial and epithelial cells, which are infected by HCMV or display reservoirs of latency. Results We investigated the potential heteromerization of US28 with CXCR4 as well as the influence of US28 on CXCR4 signaling. Using Bioluminescence Resonance Energy Transfer and luciferase-complementation based methods we show that US28 expression exhibits negative effects on CXCR4 signaling and constitutive surface expression in HEK293T cells. Furthermore, we demonstrate that this effect is not mediated by receptor heteromerization but via signaling crosstalk. Additionally, we show that in HCMV, strain TB40E, infected HUVEC the surface expression of CXCR4 is strongly downregulated, whereas in TB40E-delUS28 infected cells, CXCR4 surface expression is not altered in particular at late time points of infection. Conclusions We show that the vGPCR US28 is leading to severely disturbed signaling and surface expression of the chemokine receptor CXCR4 thereby representing an effective mechanism used by vGPCRs to reprogram host cell signaling. In contrast to other studies, we demonstrate that these effects are not mediated via heteromerization. Electronic supplementary material The online version of this article (doi:10.1186/s12964-016-0154-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Theresa Frank
- Department of Chemistry and Pharmacy, Emil Fischer Center, University of Erlangen-Nuremberg, Erlangen, Germany.,Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Anna Reichel
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Olav Larsen
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Anne-Charlotte Stilp
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Mette M Rosenkilde
- Department of Neuroscience and Pharmacology, Laboratory for Molecular Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Stamminger
- Institute for Clinical and Molecular Virology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Takeaki Ozawa
- Department of Chemistry, School of Science, The University of Tokyo, Tokyo, Japan
| | - Nuska Tschammer
- Department of Chemistry and Pharmacy, Emil Fischer Center, University of Erlangen-Nuremberg, Erlangen, Germany. .,Present Address: NanoTemper Technologies GmbH, Floessergasse 4, 81069, Munich, Germany.
| |
Collapse
|
10
|
Spiess K, Jakobsen MH, Kledal TN, Rosenkilde MM. The future of antiviral immunotoxins. J Leukoc Biol 2016; 99:911-25. [PMID: 26729815 DOI: 10.1189/jlb.2mr1015-468r] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/03/2015] [Indexed: 01/06/2023] Open
Abstract
There is a constant need for new therapeutic interventions in a wide range of infectious diseases. Over the past few years, the immunotoxins have entered the stage as promising antiviral treatments. Immunotoxins have been extensively explored in cancer treatment and have achieved FDA approval in several cases. Indeed, the design of new anticancer immunotoxins is a rapidly developing field. However, at present, several immunotoxins have been developed targeting a variety of different viruses with high specificity and efficacy. Rather than blocking a viral or cellular pathway needed for virus replication and dissemination, immunotoxins exert their effect by killing and eradicating the pool of infected cells. By targeting a virus-encoded target molecule, it is possible to obtain superior selectivity and drastically limit the side effects, which is an immunotoxin-related challenge that has hindered the success of immunotoxins in cancer treatment. Therefore, it seems beneficial to use immunotoxins for the treatment of virus infections. One recent example showed that targeting of virus-encoded 7 transmembrane (7TM) receptors by immunotoxins could be a future strategy for designing ultraspecific antiviral treatment, ensuring efficient internalization and hence efficient eradication of the pool of infected cells, both in vitro and in vivo. In this review, we provide an overview of the mechanisms of action of immunotoxins and highlight the advantages of immunotoxins as future anti-viral therapies.
Collapse
Affiliation(s)
- Katja Spiess
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Denmark; and
| | - Mette Høy Jakobsen
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Denmark; and
| | - Thomas N Kledal
- Section for Virology, Veterinary Institute, The Danish Technical University, Denmark
| | - Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Science, University of Copenhagen, Denmark; and
| |
Collapse
|
11
|
Amarandi RM, Hjortø GM, Rosenkilde MM, Karlshøj S. Probing Biased Signaling in Chemokine Receptors. Methods Enzymol 2015; 570:155-86. [PMID: 26921946 DOI: 10.1016/bs.mie.2015.09.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The chemokine system mediates leukocyte migration during homeostatic and inflammatory processes. Traditionally, it is described as redundant and promiscuous, with a single chemokine ligand binding to different receptors and a single receptor having several ligands. Signaling of chemokine receptors occurs via two major routes, G protein- and β-arrestin-dependent, which can be preferentially modulated depending on the ligands or receptors involved, as well as the cell types or tissues in which the signaling event occurs. The preferential activation of a certain signaling pathway to the detriment of others has been termed signaling bias and can accordingly be grouped into ligand bias, receptor bias, and tissue bias. Bias has so far been broadly overlooked in the process of drug development. The low number of currently approved drugs targeting the chemokine system, as well as the broad range of failed clinical trials, reflects the need for a better understanding of the chemokine system. Thus, understanding the character, direction, and consequence of biased signaling in the chemokine system may aid the development of new therapeutics. This review describes experiments to assess G protein-dependent and -independent signaling in order to quantify chemokine system bias.
Collapse
Affiliation(s)
- Roxana-Maria Amarandi
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark; Faculty of Chemistry, Alexandru Ioan Cuza University of Iaşi, Iaşi, Romania
| | - Gertrud Malene Hjortø
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Mette Marie Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | - Stefanie Karlshøj
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
12
|
El-Tholoth M, El-Kenawy AA. G-Protein-Coupled Chemokine Receptor Gene in Lumpy Skin Disease Virus Isolates from Cattle and Water Buffalo (Bubalus bubalis) in Egypt. Transbound Emerg Dis 2015; 63:e288-e295. [PMID: 25754131 DOI: 10.1111/tbed.12344] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Indexed: 12/01/2022]
Abstract
Lumpy skin disease virus (LSDV), sheep poxvirus (SPV) and goat poxvirus (GPV) are the most serious poxviruses of ruminants. In this study, we analysed the G-protein-coupled chemokine receptor (GPCR) genes of LSDV isolates from cattle and water buffalo (Bubalus bubalis) in Egypt during the summer of 2011. Multiple alignments of the nucleotide sequences revealed that the water buffalo LSDV isolate differed from the cattle isolate at four nucleotide positions, and both isolates had nine nucleotide mutations from the reference strain, Egyptian tissue culture-adapted cattle LSDV/Ismailyia88 strain. Compared with the GPCR sequences of SPV and GPV strains, a 21 nucleotide insertion and a 12 nucleotide deletion were identified in the GPCR genes of our used isolates and other LSDVs. The amino acid sequences of GPCR genes of our isolates contained the unique signature of LSDV (A11 , T12 , T34 , S99 and P199 ). Phylogenetic analyses showed that the GPCR genes of cattle and water buffalo LSDVs were closest genetically, indicating a potential transmission of cattle LSDV to water buffalo.
Collapse
Affiliation(s)
- M El-Tholoth
- Department of Virology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt.
| | - A A El-Kenawy
- Department of Virology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
13
|
Steen A, Sparre-Ulrich AH, Thiele S, Guo D, Frimurer TM, Rosenkilde MM. Gating function of isoleucine-116 in TM-3 (position III:16/3.40) for the activity state of the CC-chemokine receptor 5 (CCR5). Br J Pharmacol 2014; 171:1566-79. [PMID: 24328926 DOI: 10.1111/bph.12553] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 11/25/2013] [Accepted: 12/02/2013] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND PURPOSE A conserved amino acid within a protein family indicates a significance of the residue. In the centre of transmembrane helix (TM)-5, position V:13/5.47, an aromatic amino acid is conserved among class A 7TM receptors. However, in 37% of chemokine receptors - a subgroup of 7TM receptors - it is a leucine indicating an altered function. Here, we describe the significance of this position and its possible interaction with TM-3 for CCR5 activity. EXPERIMENTAL APPROACH The effects of [L203F]-CCR5 in TM-5 (position V:13/5.47), [I116A]-CCR5 in TM-3 (III:16/3.40) and [L203F;G286F]-CCR5 (V:13/5.47;VII:09/7.42) were determined in G-protein- and β-arrestin-coupled signalling. Computational modelling monitored changes in amino acid conformation. KEY RESULTS [L203F]-CCR5 increased the basal level of G-protein coupling (20-70% of Emax ) and β-arrestin recruitment (50% of Emax ) with a threefold increase in agonist potency. In silico, [I116A]-CCR5 switched χ1-angle in [L203F]-CCR5. Furthermore, [I116A]-CCR5 was constitutively active to a similar degree as [L203F]-CCR5. Tyr(244) in TM-6 (VI:09/6.44) moved towards TM-5 in silico, consistent with its previously shown function for CCR5 activation. On [L203F;G286F]-CCR5 the antagonist aplaviroc was converted to a superagonist. CONCLUSIONS AND IMPLICATIONS The results imply that an aromatic amino acid in the centre of TM-5 controls the level of receptor activity. Furthermore, Ile(116) acts as a gate for the movement of Tyr(244) towards TM-5 in the active state, a mechanism proposed previously for the β2 -adrenoceptor. The results provide an understanding of chemokine receptor function and thereby information for the development of biased and non-biased antagonists and inverse agonists.
Collapse
Affiliation(s)
- A Steen
- Department of Neuroscience and Pharmacology, Faculty of Health and Medical Sciences, The Panum Institute, University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | |
Collapse
|
14
|
Abstract
Chemokines play important roles in atherosclerotic vascular disease. Expressed by not only cells of the vessel wall but also emigrated leukocytes, chemokines were initially discovered to direct leukocytes to sites of inflammation. However, chemokines can also exert multiple functions beyond cell recruitment. Here, we discuss novel and recently emerging aspects of chemokines and their involvement in atherosclerosis. While reviewing newly identified roles of chemokines and their receptors in monocyte and neutrophil recruitment during atherogenesis and atheroregression, we also revisit homeostatic functions of chemokines, including their roles in cell homeostasis and foam cell formation. The functional diversity of chemokines in atherosclerosis warrants a clear-cut mechanistic dissection and stage-specific assessment to better appreciate the full scope of their actions in vascular inflammation and to identify pathways that harbor the potential for a therapeutic targeting of chemokines in atherosclerosis.
Collapse
Affiliation(s)
- Alma Zernecke
- From the Institute of Clinical Biochemistry and Pathobiochemistry, University Hospital Würzburg, Würzburg, Germany (A.Z.); Department of Vascular Surgery, Klinikum rechts der Isar, Technical University, Munich, Germany (A.Z.); DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany (A.Z., C.W.); and Institute for Cardiovascular Prevention, Ludwig-Maximilians-University, Munich, Germany (C.W.)
| | | |
Collapse
|
15
|
Structural Diversity in Conserved Regions Like the DRY-Motif among Viral 7TM Receptors-A Consequence of Evolutionary Pressure? Adv Virol 2012; 2012:231813. [PMID: 22899926 PMCID: PMC3414077 DOI: 10.1155/2012/231813] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 05/31/2012] [Indexed: 01/31/2023] Open
Abstract
Several herpes- and poxviruses have captured chemokine receptors from their hosts and modified these to their own benefit. The human and viral chemokine receptors belong to class A 7 transmembrane (TM) receptors which are characterized by several structural motifs like the DRY-motif in TM3 and the C-terminal tail. In the DRY-motif, the arginine residue serves important purposes by being directly involved in G protein coupling. Interestingly, among the viral receptors there is a greater diversity in the DRY-motif compared to their endogenous receptor homologous. The C-terminal receptor tail constitutes another regulatory region that through a number of phosphorylation sites is involved in signaling, desensitization, and internalization. Also this region is more variable among virus-encoded 7TM receptors compared to human class A receptors. In this review we will focus on these two structural motifs and discuss their role in viral 7TM receptor signaling compared to their endogenous counterparts.
Collapse
|
16
|
Castiello L, Stroncek DF, Finn MW, Wang E, Marincola FM, Clayberger C, Krensky AM, Sabatino M. 15 kDa Granulysin versus GM-CSF for monocytes differentiation: analogies and differences at the transcriptome level. J Transl Med 2011; 9:41. [PMID: 21501511 PMCID: PMC3094223 DOI: 10.1186/1479-5876-9-41] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2011] [Accepted: 04/18/2011] [Indexed: 02/06/2023] Open
Abstract
Background Granulysin is an antimicrobial and proinflammatory protein with several isoforms. While the 9 kDa isoform is a well described cytolytic molecule with pro-inflammatory activity, the functions of the 15 kDa isoform is less well understood. Recently it was shown that 15 kDa Granulysin can act as an alarmin that is able to activate monocytes and immature dendritic cells. Granulocyte Macrophage Colony Stimulating Factor (GM-CSF) is a growth factor widely used in immunotherapy both for in vivo and ex vivo applications, especially for its proliferative effects. Methods We analyzed gene expression profiles of monocytes cultured with 15 kDa Granulysin or GM-CSF for 4, 12, 24 and 48 hours to unravel both similarities and differences between the effects of these stimulators. Results The analysis revealed a common signature induced by both factors at each time point, but over time, a more specific signature for each factor became evident. At all time points, 15 kDa Granulysin induced immune response, chemotaxis and cell adhesion genes. In addition, only 15 kDa Granulsyin induced the activation of pathways related to fundamental dendritic cell functions, such as co-stimulation of T-cell activation and Th1 development. GM-CSF specifically down-regulated genes related to cell cycle arrest and the immune response. More specifically, cytokine production, lymphocyte mediated immunity and humoral immune response were down-regulated at late time points. Conclusion This study provides important insights on the effects of a novel agent, 15 kDa granulysin, that holds promise for therapeutic applications aimed at the activation of the immune response.
Collapse
Affiliation(s)
- Luciano Castiello
- Cell Processing Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Panaro MA, Calvello R, Mitolo CI, Sisto M, Cianciulli A. Evidence for endogenous retroviruses in human chemokine receptor gene introns: possible evolutionary inferences and biological roles. Immunopharmacol Immunotoxicol 2010; 33:291-301. [PMID: 21054205 DOI: 10.3109/08923973.2010.503243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The human chemokine receptor (CKR) genes CCR2, CCR6, CCR7, CCR9, CCR10, CXCR4, and CXCR5 harbor one or two introns. CCR7, CCR9, CCR10, and CXCR5 introns, (but not CCR2, CCR6, and CXCR4 introns) encompass retrovirus-like inserts with the characteristics of SINEs (short interspersed nuclear elements) up to 300 nucleotides (nt) long. Other characteristic elements of the retroviral genome, such as long terminal repeats and gag, pol, and env genes, are lacking. The inserts likely derived from one (or more) of the following retroviruses: XA34 (NCBI GenBank Nucleotides, U29659), HERV-P-T47D (AF087913), ERV FTD (U27241), HERV-K (Y17832), HML6p (U86698), HERV-H/env60 (AJ289710), XA38 (U37066). Virus-like inserts are remarkably homogeneous in all CKR introns, with nt identities of about 80%. Percentages of nt identities between the CKR inserts and the corresponding viral sequences are also about 80%. With reference to the CKR sequence, the viral sequence aligns in some instances Plus/Plus (XA34, HML6p, HERV-H/env60, and XA38) and in other instances Plus/Minus (HERV-P-T47D, ERV FTD, and HERV-K). Some aspects of the evolution of retroviruses and CKRs as well as hypotheses on the biological significance of the SINE inserts are discussed.
Collapse
|
18
|
Galzi JL, Hachet-Haas M, Bonnet D, Daubeuf F, Lecat S, Hibert M, Haiech J, Frossard N. Neutralizing endogenous chemokines with small molecules. Principles and potential therapeutic applications. Pharmacol Ther 2010; 126:39-55. [PMID: 20117133 PMCID: PMC7112609 DOI: 10.1016/j.pharmthera.2009.12.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2009] [Accepted: 12/24/2009] [Indexed: 02/08/2023]
Abstract
Regulation of cellular responses to external stimuli such as hormones, neurotransmitters, or cytokines is achieved through the control of all steps of the complex cascade starting with synthesis, going through maturation steps, release, distribution, degradation and/or uptake of the signalling molecule interacting with the target protein. One possible way of regulation, referred to as scavenging or neutralization of the ligand, has been increasingly studied, especially for small protein ligands. It shows innovative potential in chemical biology approaches as well as in disease treatment. Neutralization of protein ligands, as for example cytokines or chemokines can lead to the validation of signalling pathways under physiological or pathophysiological conditions, and in certain cases, to the development of therapeutic molecules now used in autoimmune diseases, chronic inflammation and cancer treatment. This review explores the field of ligand neutralization and tries to determine to what extent small chemical molecules could substitute for neutralizing antibodies in therapeutic approaches.
Collapse
Affiliation(s)
- Jean-Luc Galzi
- IREBS, FRE3211, Ecole Supérieure de Biotechnologie de Strasbourg, Boulevard Sébastien Brant, 67412 Illkirch, France.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Le Goff C, Lamien CE, Fakhfakh E, Chadeyras A, Aba-Adulugba E, Libeau G, Tuppurainen E, Wallace DB, Adam T, Silber R, Gulyaz V, Madani H, Caufour P, Hammami S, Diallo A, Albina E. Capripoxvirus G-protein-coupled chemokine receptor: a host-range gene suitable for virus animal origin discrimination. J Gen Virol 2009; 90:1967-1977. [PMID: 19339476 DOI: 10.1099/vir.0.010686-0] [Citation(s) in RCA: 99] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
The genus Capripoxvirus within the family Poxviridae comprises three closely related viruses, namely goat pox, sheep pox and lumpy skin disease viruses. This nomenclature is based on the animal species from which the virus was first isolated, respectively, goat, sheep and cattle. Since capripoxviruses are serologically identical, their specific identification relies exclusively on the use of molecular tools. We describe here the suitability of the G-protein-coupled chemokine receptor (GPCR) gene for use in host-range grouping of capripoxviruses. The analysis of 58 capripoxviruses showed three tight genetic clusters consisting of goat pox, sheep pox and lumpy skin disease viruses. However, a few discrepancies exist with the classical virus-host origin nomenclature: a virus isolated from sheep is grouped in the goat poxvirus clade and vice versa. Intra-group diversity was further observed for the goat pox and lumpy skin disease virus isolates. Despite the presence of nine vaccine strains, no genetic determinants of virulence were identified on the GPCR gene. For sheep poxviruses, the addition or deletion of 21 nucleic acids (7 aa) was consistently observed in the 5' terminal part of the gene. Specific signatures for each cluster were also identified. Prediction of the capripoxvirus GPCR topology, and its comparison with other known mammalian GPCRs and viral homologues, revealed not only a classical GPCR profile in the last three-quarters of the protein but also unique features such as a longer N-terminal end with a proximal hydrophobic alpha-helix and a shorter serine-rich C-tail.
Collapse
Affiliation(s)
| | - Charles Euloge Lamien
- Animal Production Unit, FAO/IAEA Agriculture and Biotechnology Laboratory, IAEA Laboratories Seibersdorf, International Atomic Energy Agency, Wagramer Strasse 5, PO Box 100, A-1400 Vienna, Austria
| | | | | | | | | | - Eeva Tuppurainen
- Institute of Animal Health, Pirbright Laboratory, Woking, Surrey GU24 ONF, UK
| | - David B Wallace
- Department of Veterinary Tropical Diseases, University of Pretoria, Faculty of Veterinary Science, Private Bag X4, Onderstepoort 0110, South Africa.,Biotechnology Division, ARC-Onderstepoort Veterinary Institute, Private Bag X5, Onderstepoort 0110, South Africa
| | - Tajelser Adam
- Department of Viral Vaccines Production, Central Veterinary Research Laboratories Centre, Animal Resources Research Corporation, Ministry of Science and Technology, Khartoum, Sudan
| | - Roland Silber
- Institute for Veterinary Disease Control, Austrian Agency for Health and Food Security, Robert Koch Gasse 17, A-2340 Mödling, Austria
| | - Velý Gulyaz
- Pendik Veterinary Control and Research Institute, Pendik, Istanbul, Turkey
| | - Hafsa Madani
- Institut National de la Médecine Vétérinaire, Laboratoire Central Vétérinaire d'Alger, BP 205 Hacen Badi, El Harrach, Alger, Algeria
| | | | | | - Adama Diallo
- Animal Production Unit, FAO/IAEA Agriculture and Biotechnology Laboratory, IAEA Laboratories Seibersdorf, International Atomic Energy Agency, Wagramer Strasse 5, PO Box 100, A-1400 Vienna, Austria
| | - Emmanuel Albina
- CIRAD, UMR Contrôle des Maladies, F-34398 Montpellier, France
| |
Collapse
|
20
|
Glatt S, Halbauer D, Heindl S, Wernitznig A, Kozina D, Su KC, Puri C, Garin-Chesa P, Sommergruber W. hGPR87 contributes to viability of human tumor cells. Int J Cancer 2008; 122:2008-16. [PMID: 18183596 DOI: 10.1002/ijc.23349] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Emerging in vitro and in vivo data underline the crucial role of G-protein-coupled receptors (GPCRs) in tumorigenesis. Here, we report the contribution of hGPR87, a predicted member of the P2Y subfamily of GPCRs, to proliferation and survival of human tumor cell lines. hGPR87 mRNA transcript was found to be preferentially overexpressed in squamous cell carcinomas (SCCs) of different locations and in their lymph node metastases. Up-regulation of both, transcript and protein, was detected in samples of SCC of the lung, cervix, skin and head and neck (pharynx, larynx and epiglottis). In addition to the expression of hGPR87 in tumors which originate from stratified epithelia, we identified other hGPR87-positive tumor types including subsets of large cell and adenocarcinomas of the lung and transitional cell carcinomas of the urinary bladder. Loss of function studies using siRNA in human cancer cell lines lead to antiproliferative effects and induction of apoptosis. Like other known P2Y receptors, hGPR87 was found to be mainly located on the cell surface. The overexpression of hGPR87 preferentially in SCCs together with our functional data suggests a common molecular mechanism for SCC tumorigenesis and may provide a novel intervention site for mechanism-based antitumor therapies.
Collapse
Affiliation(s)
- Sebastian Glatt
- Boehringer-Ingelheim Austria GmbH, Dr. Boehringer-Gassse 5-11, Vienna, Austria
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Structure, function and physiological consequences of virally encoded chemokine seven transmembrane receptors. Br J Pharmacol 2008; 153 Suppl 1:S154-66. [PMID: 18204488 DOI: 10.1038/sj.bjp.0707660] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
A number of human and animal herpes viruses encode G-protein coupled receptors with seven transmembrane (7TM) segments-most of which are clearly related to human chemokine receptors. It appears, that these receptors are used by the virus for immune evasion, cellular transformation, tissue targeting, and possibly for cell entry. In addition, many virally-encoded chemokine 7TM receptors have been suggested to be causally involved in pathogenic phenotypes like Kaposi sarcoma, atherosclerosis, HIV-infection and tumour development. The role of these receptors during the viral life cycle and in viral pathogenesis is still poorly understood. Here we focus on the current knowledge of structure, function and trafficking patterns of virally encoded chemokine receptors and further address the putative roles of these receptors in virus survival and host -cell and/or -immune system modulation. Finally, we highlight the emerging impact of these receptor on virus-mediated diseases.
Collapse
|
22
|
Boudry C, Markine-Goriaynoff N, Delforge C, Springael JY, de Leval L, Drion P, Russell G, Haig DM, Vanderplasschen AF, Dewals B. The A5 gene of alcelaphine herpesvirus 1 encodes a constitutively active G-protein-coupled receptor that is non-essential for the induction of malignant catarrhal fever in rabbits. J Gen Virol 2008; 88:3224-3233. [PMID: 18024890 DOI: 10.1099/vir.0.83153-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Many gammaherpesviruses encode G-protein-coupled receptors (GPCRs). Several in vivo studies have revealed that gammaherpesvirus GPCRs are important for viral replication and for virus-induced pathogenesis. The gammaherpesvirus alcelaphine herpesvirus 1 (AlHV-1) is carried asymptomatically by wildebeest, but causes malignant catarrhal fever (MCF) following cross-species transmission to a variety of susceptible species. The A5 ORF of the AlHV-1 genome encodes a putative GPCR. In the present study, we investigated whether A5 encodes a functional GPCR and addressed its role in viral replication and in the pathogenesis of MCF. In silico analysis supported the hypothesis that A5 could encode a functional GPCR as its expression product contained several hallmark features of GPCRs. Expression of A5 as tagged proteins in various cell lines revealed that A5 localizes in cell membranes, including the plasma membrane. Using [35S]GTPgammaS and reporter gene assays, we found that A5 is able to constitutively couple to alpha i-type G-proteins in transfected cells, and that this interaction is able to inhibit forskolin-triggered cAMP response element-binding protein (CREB) activation. Finally, using an AlHV-1 BAC clone, we produced a strain deleted for A5 and a revertant strain. Interestingly, the strain deleted for A5 replicated comparably to the wild-type parental strain and induced MCF in rabbits that was indistinguishable from that of the parental strain. The present study is the first to investigate the role of an individual gene of AlHV-1 in MCF pathogenesis.
Collapse
Affiliation(s)
- C Boudry
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| | - N Markine-Goriaynoff
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| | - C Delforge
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| | - J-Y Springael
- Institute of Interdisciplinary Research in Human and Molecular Biology (IRIBHM), Free University of Brussels, Erasme, 808 Route de Lennik, B-1070 Brussels, Belgium
| | - L de Leval
- Department of Pathology (B23), Faculty of Medicine, University of Liège, B-4000 Liège, Belgium
| | - P Drion
- Animal facility (B23), University of Liège, B-4000 Liège, Belgium
| | - G Russell
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian EH26 0PZ, UK
| | - D M Haig
- Moredun Research Institute, Pentlands Science Park, Penicuik, Midlothian EH26 0PZ, UK
| | - A F Vanderplasschen
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| | - B Dewals
- Immunology-Vaccinology (B43b), Department of Infectious and Parasitic Diseases, Faculty of Veterinary Medicine, University of Liège, B-4000 Liège, Belgium
| |
Collapse
|
23
|
Tadagaki K, Yamanishi K, Mori Y. Reciprocal roles of cellular chemokine receptors and human herpesvirus 7-encoded chemokine receptors, U12 and U51. J Gen Virol 2007; 88:1423-1428. [PMID: 17412969 DOI: 10.1099/vir.0.82665-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Human herpesvirus 7 (HHV-7) is a member of the subfamilyBetaherpesvirinaethat exhibits a restricted cell tropism, preferentially infecting CD4+T cellsin vitro. HHV-7 encodes two functional chemokine receptors, U12 and U51. The human chemokines that act as ligands for these receptors have been identified as CCL22 (the natural ligand for CCR4) and CCL19 (the natural ligand for CCR7). It was found that murine L1.2 cells co-expressing CCR4 or CCR7 and U12 responded to both CCL22 and CCL19 in calcium-mobilization assays, but migrated in response only to the appropriate ligand for the expressed cellular receptor. Similar results were obtained with L1.2 cells co-expressing CCR4 or CCR7 with U51. These results suggest that the HHV-7 U12 and U51 receptors can function in concert with CCR4 and CCR7 in host-cell signalling pathways.
Collapse
Affiliation(s)
- Kenjiro Tadagaki
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Koichi Yamanishi
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Yasuko Mori
- Laboratory of Virology and Vaccinology, Division of Biomedical Research, National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| |
Collapse
|
24
|
Abstract
G-protein-coupled receptors (GPCRs), the largest family of cell-surface molecules involved in signal transmission, have recently emerged as crucial players in tumour growth and metastasis. Malignant cells often hijack the normal physiological functions of GPCRs to survive, proliferate autonomously, evade the immune system, increase their blood supply, invade their surrounding tissues and disseminate to other organs. This Review will address our current understanding of the many roles of GPCRs and their signalling circuitry in tumour progression and metastasis. We will also discuss how interfering with GPCRs might provide unique opportunities for cancer prevention and treatment.
Collapse
Affiliation(s)
- Robert T Dorsam
- Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland 20892-4330, USA
| | | |
Collapse
|
25
|
Rosenkilde MM, Benned-Jensen T, Andersen H, Holst PJ, Kledal TN, Lüttichau HR, Larsen JK, Christensen JP, Schwartz TW. Molecular Pharmacological Phenotyping of EBI2. J Biol Chem 2006; 281:13199-13208. [PMID: 16540462 DOI: 10.1074/jbc.m602245200] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Epstein-Barr virus (EBV)-induced receptor 2 (EBI2) is an orphan seven-transmembrane (7TM) receptor originally identified as the most up-regulated gene (>200-fold) in EBV-infected cells. Here we show that EBI2 signals with constitutive activity through Galpha(i) as determined by a receptor-mediated inhibition of forskolin-induced cAMP production and an induction of the serum response element-driven transcriptional activity in a pertussis toxin-sensitive manner. Galpha(s) and Galpha(q) were not activated constitutively as determined by the lack of cAMP production, the lack of inositol phosphate turnover, and the lack of activities of the transcription factors: cAMP response element-binding protein and nuclear factor-kappaB. Immunohistochemistry and confocal microscopy of FLAG- and green fluorescent protein-tagged EBI2 revealed cell-surface expression. A putative N-terminal truncated version of EBI2, delta4-EBI2, showed similar expression and signaling through Galpha(i) as full-length EBI2. By using a 32P-labeled EBI2 probe we found a very high expression in lymphoid tissue (spleen and lymph node) and peripheral blood mononuclear cells and a high expression in lung tissue. Real-time PCR of EBV-infected cells showed high expression of EBI2 during latent and lytic infection, in contrast to the EBV-encoded 7TM receptor BILF1, which was induced during lytic infection. EBI2 clustered with the orphan GPR18 by alignment analysis as well as by close proximity in the chromosomal region 13q32.3. Based on the constitutive signaling and cellular expression pattern of EBI2, it is suggested that it may function in conjunction with BILF1 in the reprogramming of the cell during EBV infection.
Collapse
Affiliation(s)
- Mette M Rosenkilde
- Laboratory for Molecular Pharmacology, Department of Pharmacology, University of Copenhagen, 2200 Copenhagen N, Copenhagen, Denmark.
| | - Tau Benned-Jensen
- Laboratory for Molecular Pharmacology, Department of Pharmacology, University of Copenhagen, 2200 Copenhagen N, Copenhagen, Denmark
| | - Helene Andersen
- Clinical Research Unit, Copenhagen University Hospital, 2650 Hvidovre, Denmark
| | - Peter J Holst
- Institute for Medical Microbiology and Immunology, The Panum Institute, Building 18.6, Blegdamsvej 3, 2200 Copenhagen N, Copenhagen, Denmark
| | - Thomas N Kledal
- Clinical Research Unit, Copenhagen University Hospital, 2650 Hvidovre, Denmark
| | - Hans R Lüttichau
- Laboratory for Molecular Pharmacology, Department of Pharmacology, University of Copenhagen, 2200 Copenhagen N, Copenhagen, Denmark; Department for Infectious Diseases, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Jørgen K Larsen
- Finsen Laboratory, The Finsen Center, Copenhagen University Hospital, 2200 Copenhagen, Denmark
| | - Jan P Christensen
- Institute for Medical Microbiology and Immunology, The Panum Institute, Building 18.6, Blegdamsvej 3, 2200 Copenhagen N, Copenhagen, Denmark
| | - Thue W Schwartz
- Laboratory for Molecular Pharmacology, Department of Pharmacology, University of Copenhagen, 2200 Copenhagen N, Copenhagen, Denmark
| |
Collapse
|
26
|
Abstract
Multicellular organisms possess very sophisticated defense mechanisms that are designed to effectively counter the continual microbial insult of the environment within the vertebrate host. However, successful microbial pathogens have in turn evolved complex and efficient methods to overcome innate and adaptive immune mechanisms, which can result in disease or chronic infections. Although the various virulence strategies used by viral and bacterial pathogens are numerous, there are several general mechanisms that are used to subvert and exploit immune systems that are shared between these diverse microbial pathogens. The success of each pathogen is directly dependant on its ability to mount an effective anti-immune response within the infected host, which can ultimately result in acute disease, chronic infection, or pathogen clearance. In this review, we highlight and compare some of the many molecular mechanisms that bacterial and viral pathogens use to evade host immune defenses.
Collapse
Affiliation(s)
- B Brett Finlay
- Michael Smith Laboratories, University of British Columbia, Vancouver, B.C. V6T 1Z4 Canada.
| | | |
Collapse
|
27
|
Taylor RT, Bresnahan WA. Human cytomegalovirus immediate-early 2 protein IE86 blocks virus-induced chemokine expression. J Virol 2006; 80:920-8. [PMID: 16378994 PMCID: PMC1346867 DOI: 10.1128/jvi.80.2.920-928.2006] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
The effect of human cytomegalovirus (HCMV) gene expression on cytokine (beta interferon) and chemokine (RANTES, MIG, MCP-2, MIP-1alpha, and interleukin-8) expression was examined. We demonstrate that HCMV gene expression is required to suppress the transcriptional induction of these cytokines and that the HCMV immediate-early 2 gene product IE86 can effectively block the expression of cytokines and proinflammatory chemokines during HCMV and Sendai virus infection. Additionally, we present data on viral mutants and ectopic protein expression which demonstrate that pp65, another identified HCMV cytokine antagonist, is not involved in regulating these proinflammatory cytokines. This is the first report to demonstrate that IE86 can act to suppress virus-induced proinflammatory cytokine transcript expression, extending the antiviral properties of this multifunctional viral protein.
Collapse
Affiliation(s)
- R Travis Taylor
- Department of Microbiology, University of Minnesota, 1060 Mayo Building, MMC196, Minneapolis, MN 55455, USA
| | | |
Collapse
|
28
|
Piqueras B, Connolly J, Freitas H, Palucka AK, Banchereau J. Upon viral exposure, myeloid and plasmacytoid dendritic cells produce 3 waves of distinct chemokines to recruit immune effectors. Blood 2005; 107:2613-8. [PMID: 16317096 PMCID: PMC1895384 DOI: 10.1182/blood-2005-07-2965] [Citation(s) in RCA: 179] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Host response to viral infection involves distinct effectors of innate and adaptive immunity, whose mobilization needs to be coordinated to ensure protection. Here we show that influenza virus triggers, in human blood dendritic-cell (DC) subsets (ie, plasmacytoid and myeloid DCs), a coordinated chemokine (CK) secretion program with 3 successive waves. The first one, occurring at early time points (2 to 4 hours), includes CKs potentially attracting effector cells such as neutrophils, cytotoxic T cells, and natural killer (NK) cells (CXCL16, CXCL1, CXCL2, and CXCL3). The second one occurs within 8 to 12 hours and includes CKs attracting effector memory T cells (CXCL8, CCL3, CCL4, CCL5, CXCL9, CXCL10, and CXCL11). The third wave, which occurs after 24 to 48 hours, when DCs have reached the lymphoid organs, includes CCL19, CCL22, and CXCL13, which attract naive T and B lymphocytes. Thus, human blood DC subsets carry a common program of CK production, which allows for a coordinated attraction of the different immune effectors in response to viral infection.
Collapse
Affiliation(s)
- Bernard Piqueras
- Baylor NIAID (National Institute of Allergy and Infectious Disease) Cooperative Center for Translational Research on Human Immunology and Biodefense, Dallas, TX 75204, USA
| | | | | | | | | |
Collapse
|