1
|
Rosenkilde MM, Tsutsumi N, Knerr JM, Kildedal DF, Garcia KC. Viral G Protein-Coupled Receptors Encoded by β- and γ-Herpesviruses. Annu Rev Virol 2022; 9:329-351. [PMID: 35671566 PMCID: PMC9584139 DOI: 10.1146/annurev-virology-100220-113942] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Herpesviruses are ancient large DNA viruses that have exploited gene capture as part of their strategy to escape immune surveillance, promote virus spreading, or reprogram host cells to benefit their survival. Most acquired genes are transmembrane proteins and cytokines, such as viral G protein-coupled receptors (vGPCRs), chemokines, and chemokine-binding proteins. This review focuses on the vGPCRs encoded by the human β- and γ-herpesviruses. These include receptors from human cytomegalovirus, which encodes four vGPCRs: US27, US28, UL33, and UL78; human herpesvirus 6 and 7 with two receptors: U12 and U51; Epstein-Barr virus with one: BILF1; and Kaposi's sarcoma-associated herpesvirus with one: open reading frame 74. We discuss ligand binding, signaling, and structures of the vGPCRs in light of robust differences from endogenous receptors. Finally, we briefly discuss the therapeutic targeting of vGPCRs as future treatment of acute and chronic herpesvirus infections. Expected final online publication date for the Annual Review of Virology, Volume 9 is September 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Mette M Rosenkilde
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | - Naotaka Tsutsumi
- Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| | - Julius M Knerr
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;
| | | | - K Christopher Garcia
- Departments of Molecular and Cellular Physiology, and Structural Biology, and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California, USA;
| |
Collapse
|
2
|
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
|
3
|
The Chemokine System in Oncogenic Pathways Driven by Viruses: Perspectives for Cancer Immunotherapy. Cancers (Basel) 2022; 14:cancers14030848. [PMID: 35159113 PMCID: PMC8834488 DOI: 10.3390/cancers14030848] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/03/2022] [Accepted: 02/05/2022] [Indexed: 12/18/2022] Open
Abstract
Simple Summary Oncoviruses are viruses with oncogenic potential, responsible for almost 20% of human cancers worldwide. They are from various families, some of which belong to the microbial communities that inhabit several sites in the body of healthy humans. As a result, they most often establish latent infections controlled by the arsenal of human host responses that include the chemokine system playing key roles at the interface between tissue homeostasis and immune surveillance. Yet, chemokines and their receptors also contribute to oncogenic processes as they are targeted by the virus-induced deregulations of host responses and/or directly encoded by viruses. Thus, the chemokine system offers a strong rationale for therapeutic options, some few already approved or in trials, and future ones that we are discussing in view of the pharmacological approaches targeting the different functions of chemokines operating in both cancer cells and the tumor microenvironment. Abstract Chemokines interact with glycosaminoglycans of the extracellular matrix and activate heptahelical cellular receptors that mainly consist of G Protein-Coupled Receptors and a few atypical receptors also with decoy activity. They are well-described targets of oncogenic pathways and key players in cancer development, invasiveness, and metastasis acting both at the level of cancer cells and cells of the tumor microenvironment. Hence, they can regulate cancer cell proliferation and survival and promote immune or endothelial cell migration into the tumor microenvironment. Additionally, oncogenic viruses display the potential of jeopardizing the chemokine system by encoding mimics of chemokines and receptors as well as several products such as oncogenic proteins or microRNAs that deregulate their human host transcriptome. Conversely, the chemokine system participates in the host responses that control the virus life cycle, knowing that most oncoviruses establish asymptomatic latent infections. Therefore, the deregulated expression and function of chemokines and receptors as a consequence of acquired or inherited mutations could bias oncovirus infection toward pro-oncogenic pathways. We here review these different processes and discuss the anticancer therapeutic potential of targeting chemokine availability or receptor activation, from signaling to decoy-associated functions, in combination with immunotherapies.
Collapse
|
4
|
Pontejo SM, Murphy PM. Chemokines act as phosphatidylserine-bound "find-me" signals in apoptotic cell clearance. PLoS Biol 2021; 19:e3001259. [PMID: 34038417 PMCID: PMC8213124 DOI: 10.1371/journal.pbio.3001259] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 06/18/2021] [Accepted: 05/05/2021] [Indexed: 12/14/2022] Open
Abstract
Removal of apoptotic cells is essential for maintenance of tissue homeostasis. Chemotactic cues termed "find-me" signals attract phagocytes toward apoptotic cells, which selectively expose the anionic phospholipid phosphatidylserine (PS) and other "eat-me" signals to distinguish healthy from apoptotic cells for phagocytosis. Blebs released by apoptotic cells can deliver find-me signals; however, the mechanism is poorly understood. Here, we demonstrate that apoptotic blebs generated in vivo from mouse thymus attract phagocytes using endogenous chemokines bound to the bleb surface. We show that chemokine binding to apoptotic cells is mediated by PS and that high affinity binding of PS and other anionic phospholipids is a general property of many but not all chemokines. Chemokines are positively charged proteins that also bind to anionic glycosaminoglycans (GAGs) on cell surfaces for presentation to leukocyte G protein-coupled receptors (GPCRs). We found that apoptotic cells down-regulate GAGs as they up-regulate PS on the cell surface and that PS-bound chemokines, unlike GAG-bound chemokines, are able to directly activate chemokine receptors. Thus, we conclude that PS-bound chemokines may serve as find-me signals on apoptotic vesicles acting at cognate chemokine receptors on leukocytes.
Collapse
Affiliation(s)
- Sergio M Pontejo
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Philip M Murphy
- Molecular Signaling Section, Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
5
|
Baba O, Huang LH, Elvington A, Szpakowska M, Sultan D, Heo GS, Zhang X, Luehmann H, Detering L, Chevigné A, Liu Y, Randolph GJ. CXCR4-Binding Positron Emission Tomography Tracers Link Monocyte Recruitment and Endothelial Injury in Murine Atherosclerosis. Arterioscler Thromb Vasc Biol 2021; 41:822-836. [PMID: 33327748 PMCID: PMC8105279 DOI: 10.1161/atvbaha.120.315053] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE vMIP-II (viral macrophage inflammatory protein 2)/vCCL2 (viral chemotactic cytokine ligand 2) binds to multiple chemokine receptors, and vMIP-II-based positron emission tomography tracer (64Cu-DOTA-vMIP-II: vMIP-II tracer) accumulates at atherosclerotic lesions in mice. Given that it would be expected to react with multiple chemokine receptors on monocytes and macrophages, we wondered if its accumulation in atherosclerosis lesion-bearing mice might correlate with overall macrophage burden or, alternatively, the pace of monocyte recruitment. Approach and Results: We employed a mouse model of atherosclerosis regression involving adenoassociated virus 8 vector encoding murine Apoe (AAV-mApoE) treatment of Apoe-/- mice where the pace of monocyte recruitment slows before macrophage burden subsequently declines. Accumulation of 64Cu-DOTA-vMIP-II at Apoe-/- plaque sites was strong but declined with AAV-mApoE-induced decline in monocyte recruitment, before macrophage burden reduced. Monocyte depletion indicated that monocytes and macrophages themselves were not the only target of the 64Cu-DOTA-vMIP-II tracer. Using fluorescence-tagged vMIP-II tracer, competitive receptor blocking with CXCR4 antagonists, endothelial-specific Cre-mediated deletion of CXCR4, CXCR4-specific tracer 64Cu-DOTA-FC131, and CXCR4 staining during disease progression and regression, we show endothelial cell expression of CXCR4 is a key target of 64Cu-DOTA-vMIP-II imaging. Expression of CXCR4 was low in nonplaque areas but strongly detected on endothelium of progressing plaques, especially on proliferating endothelium, where vascular permeability was increased and monocyte recruitment was the strongest. CONCLUSIONS Endothelial injury status of plaques is marked by CXCR4 expression and this injury correlates with the tendency of such plaques to recruit monocytes. Furthermore, our findings suggest positron emission tomography tracers that mark CXCR4 can be used translationally to monitor the state of plaque injury and monocyte recruitment.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/diagnostic imaging
- Aorta, Thoracic/immunology
- Aorta, Thoracic/metabolism
- Aorta, Thoracic/pathology
- Atherosclerosis/diagnostic imaging
- Atherosclerosis/immunology
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Biomarkers/metabolism
- Cell Line
- Chemokines/administration & dosage
- Chemokines/pharmacokinetics
- Disease Models, Animal
- Endothelial Cells/immunology
- Endothelial Cells/metabolism
- Endothelial Cells/pathology
- Endothelium, Vascular/diagnostic imaging
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/pathology
- Injections, Intravenous
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Knockout, ApoE
- Molecular Imaging
- Monocytes/immunology
- Monocytes/metabolism
- Monocytes/pathology
- Organometallic Compounds/administration & dosage
- Organometallic Compounds/pharmacokinetics
- Plaque, Atherosclerotic
- Positron-Emission Tomography
- Predictive Value of Tests
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/pharmacokinetics
- Receptors, CXCR4/genetics
- Receptors, CXCR4/metabolism
- Mice
Collapse
Affiliation(s)
- Osamu Baba
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis
| | - Li-Hao Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis
| | - Andrew Elvington
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Deborah Sultan
- Department of Radiology, Washington University School of Medicine, St. Louis
| | - Gyu Seong Heo
- Department of Radiology, Washington University School of Medicine, St. Louis
| | - Xiaohui Zhang
- Department of Radiology, Washington University School of Medicine, St. Louis
| | - Hannah Luehmann
- Department of Radiology, Washington University School of Medicine, St. Louis
| | - Lisa Detering
- Department of Radiology, Washington University School of Medicine, St. Louis
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg
| | - Yongjian Liu
- Department of Radiology, Washington University School of Medicine, St. Louis
| | - Gwendalyn J. Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis
| |
Collapse
|
6
|
Choi YB, Cousins E, Nicholas J. Novel Functions and Virus-Host Interactions Implicated in Pathogenesis and Replication of Human Herpesvirus 8. Recent Results Cancer Res 2021; 217:245-301. [PMID: 33200369 DOI: 10.1007/978-3-030-57362-1_11] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Human herpesvirus 8 (HHV-8) is classified as a γ2-herpesvirus and is related to Epstein-Barr virus (EBV), a γ1-herpesvirus. One important aspect of the γ-herpesviruses is their association with neoplasia, either naturally or in animal model systems. HHV-8 is associated with B-cell-derived primary effusion lymphoma (PEL) and multicentric Castleman's disease (MCD), endothelial-derived Kaposi's sarcoma (KS), and KSHV inflammatory cytokine syndrome (KICS). EBV is also associated with a number of B-cell malignancies, such as Burkitt's lymphoma, Hodgkin's lymphoma, and posttransplant lymphoproliferative disease, in addition to epithelial nasopharyngeal and gastric carcinomas. Despite the similarities between these viruses and their associated malignancies, the particular protein functions and activities involved in key aspects of virus biology and neoplastic transformation appear to be quite distinct. Indeed, HHV-8 specifies a number of proteins for which counterparts had not previously been identified in EBV, other herpesviruses, or even viruses in general, and these proteins are believed to play vital functions in virus biology and to be involved centrally in viral pathogenesis. Additionally, a set of microRNAs encoded by HHV-8 appears to modulate the expression of multiple host proteins to provide conditions conductive to virus persistence within the host and possibly contributing to HHV-8-induced neoplasia. Here, we review the molecular biology underlying these novel virus-host interactions and their potential roles in both virus biology and virus-associated disease.
Collapse
Affiliation(s)
- Young Bong Choi
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA.
| | - Emily Cousins
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| | - John Nicholas
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins University School of Medicine, 1650 Orleans Street, Baltimore, MD, 21287, USA
| |
Collapse
|
7
|
Alomari N, Totonchy J. Cytokine-Targeted Therapeutics for KSHV-Associated Disease. Viruses 2020; 12:E1097. [PMID: 32998419 PMCID: PMC7600567 DOI: 10.3390/v12101097] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/23/2020] [Accepted: 09/25/2020] [Indexed: 12/15/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) also known as human herpesvirus 8 (HHV-8), is linked to several human malignancies including Kaposi sarcoma (KS), primary effusion lymphoma (PEL), multicentric Castleman's disease (MCD) and recently KSHV inflammatory cytokine syndrome (KICS). As with other diseases that have a significant inflammatory component, current therapy for KSHV-associated disease is associated with significant off-target effects. However, recent advances in our understanding of the pathogenesis of KSHV have produced new insight into the use of cytokines as potential therapeutic targets. Better understanding of the role of cytokines during KSHV infection and tumorigenesis may lead to new preventive or therapeutic strategies to limit KSHV spread and improve clinical outcomes. The cytokines that appear to be promising candidates as KSHV antiviral therapies include interleukins 6, 10, and 12 as well as interferons and tumor necrosis factor-family cytokines. This review explores our current understanding of the roles that cytokines play in promoting KSHV infection and tumorigenesis, and summarizes the current use of cytokines as therapeutic targets in KSHV-associated diseases.
Collapse
Affiliation(s)
| | - Jennifer Totonchy
- Department of Biomedical and Pharmaceutical Sciences, Chapman University School of Pharmacy, Irvine, CA 92618, USA;
| |
Collapse
|
8
|
Broussard G, Damania B. KSHV: Immune Modulation and Immunotherapy. Front Immunol 2020; 10:3084. [PMID: 32117196 PMCID: PMC7025529 DOI: 10.3389/fimmu.2019.03084] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2019] [Accepted: 12/17/2019] [Indexed: 12/21/2022] Open
Abstract
Kaposi's sarcoma (KS)-associated herpesvirus (KSHV) is associated with KS, primary effusion lymphoma (PEL), and multicentric Castleman disease (MCD). To ensure its own survival and propagation, KSHV employs an extensive network of viral proteins to subvert the host immune system, resulting in lifelong latent infection. Modulation of cellular and systemic immune defenses allows KSHV to persist in the host, which may eventually lead to the progression of KSHV-associated cancers. Due to KSHV's reliance on modifying immune responses to efficiently infect its host, immunotherapy is an attractive option for treating KSHV-associated malignancies. In this review, we will focus on the mechanisms by which KSHV evades the immune system and the current immune-related clinical strategies to treat KSHV-associated disease.
Collapse
Affiliation(s)
- Grant Broussard
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Blossom Damania
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.,Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| |
Collapse
|
9
|
Münz C. Tumor Microenvironment Conditioning by Abortive Lytic Replication of Oncogenic γ-Herpesviruses. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1225:127-135. [PMID: 32030652 DOI: 10.1007/978-3-030-35727-6_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Epstein Barr virus (EBV) and Kaposi sarcoma-associated herpesvirus (KSHV) constitute the human γ-herpesviruses and two of the seven human tumor viruses. In addition to their viral oncogenes that primarily belong to the latent infection programs of these viruses, they encode proteins that condition the microenvironment. Many of these are early lytic gene products and are only expressed in a subset of infected cells of the tumor mass. In this chapter I will describe their function and the evidence that targeting them in addition to the latent oncogenes could be beneficial for the treatment of EBV- and KSHV-associated malignancies.
Collapse
Affiliation(s)
- Christian Münz
- Viral Immunobiology, Institute of Experimental Immunology, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
10
|
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
|
11
|
Quinn KE, Mackie DI, Caron KM. Emerging roles of atypical chemokine receptor 3 (ACKR3) in normal development and physiology. Cytokine 2019; 109:17-23. [PMID: 29903572 DOI: 10.1016/j.cyto.2018.02.024] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Revised: 02/21/2018] [Accepted: 02/21/2018] [Indexed: 01/16/2023]
Abstract
The discovery that atypical chemokine receptors (ACKRs) can initiate alternative signaling pathways rather than classical G-protein coupled receptor (GPCR) signaling has changed the paradigm of chemokine receptors and their roles in modulating chemotactic responses. The ACKR family has grown over the years, with discovery of new functions and roles in a variety of pathophysiological conditions. However, the extent to which these receptors regulate normal physiology is still continuously expanding. In particular, atypical chemokine receptor 3 (ACKR3) has proven to be an important receptor in mediating normal biological functions, including cardiac development and migration of cortical neurons. In this review, we illustrate the versatile and intriguing role of ACKR3 in physiology.
Collapse
Affiliation(s)
- K E Quinn
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | - D I Mackie
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA
| | - K M Caron
- Department of Cell Biology and Physiology, 111 MasonFarm Rd., 6312B MBRB CB# 7545, The University of North Carolina, Chapel Hill, NC 27599-7545, USA.
| |
Collapse
|
12
|
Pontejo SM, Murphy PM, Pease JE. Chemokine Subversion by Human Herpesviruses. J Innate Immun 2018; 10:465-478. [PMID: 30165356 DOI: 10.1159/000492161] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/11/2018] [Indexed: 12/30/2022] Open
Abstract
Viruses use diverse molecular mechanisms to exploit and evade the immune response. Herpesviruses, in particular, encode functional chemokine and chemokine receptor homologs pirated from the host, as well as secreted chemokine-binding proteins with unique structures. Multiple functions have been described for herpesvirus chemokine components, including attraction of target cells, blockade of leukocyte migration, and modulation of gene expression and cell entry by the virus. Here we review current concepts about how human herpesvirus chemokines, chemokine receptors, and chemokine-binding proteins may be used to shape a proviral state in the host.
Collapse
Affiliation(s)
- Sergio M Pontejo
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - James E Pease
- Inflammation, Repair and Development Section, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United
| |
Collapse
|
13
|
Wang C, Chen W, Shen J. CXCR7 Targeting and Its Major Disease Relevance. Front Pharmacol 2018; 9:641. [PMID: 29977203 PMCID: PMC6021539 DOI: 10.3389/fphar.2018.00641] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 05/29/2018] [Indexed: 12/25/2022] Open
Abstract
Chemokine receptors are the target of small peptide chemokines. They play various important roles in physiological and pathological processes. CXCR7, later renamed ACKR3, is a non-classical seven transmembrane-spanning receptor whose function as a signaling or non-signaling scavenger/decoy receptor is currently under debate. Even for cell signaling mechanisms, there has been inconsistency on whether CXCR7 couples to G-proteins or β-arrestins. Several reasons may contribute to this uncertainty or controversy. In one hand, it has been neglected that CXCR7 has more than five natural ligands and unfortunately, most of the prior research only studied SDF-1 (CXCL12) and/or I-TAC (CXCL11); on the other hand, there are mounting evidence supporting ligand and tissue bias for receptor signaling, but limited such information is available for CXCR7. In this review we focus on summarizing the endogenous and exogenous ligands of CXCR7, the main diseases related to CXCR7 and the biased signaling events happening on CXCR7. These three aspects of CXCR7 pharmacologic properties may explain why the contradicting opinions of whether CXCR7 is a signaling or non-signaling receptor exist. Further, potential new direction and perspective for the study of CXCR7 biology and pharmacology are highlighted.
Collapse
Affiliation(s)
- Chuan Wang
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| | - Weilin Chen
- Department of Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Jianzhong Shen
- Department of Drug Discovery and Development, Harrison School of Pharmacy, Auburn University, Auburn, AL, United States
| |
Collapse
|
14
|
Pontejo SM, Murphy PM. Chemokines encoded by herpesviruses. J Leukoc Biol 2017; 102:1199-1217. [PMID: 28848041 DOI: 10.1189/jlb.4ru0417-145rr] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Revised: 07/25/2017] [Accepted: 07/26/2017] [Indexed: 12/15/2022] Open
Abstract
Viruses use diverse strategies to elude the immune system, including copying and repurposing host cytokine and cytokine receptor genes. For herpesviruses, the chemokine system of chemotactic cytokines and receptors is a common source of copied genes. Here, we review the current state of knowledge about herpesvirus-encoded chemokines and discuss their possible roles in viral pathogenesis, as well as their clinical potential as novel anti-inflammatory agents or targets for new antiviral strategies.
Collapse
Affiliation(s)
- Sergio M Pontejo
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Philip M Murphy
- Laboratory of Molecular Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
15
|
Novel Chemokine-Based Immunotoxins for Potent and Selective Targeting of Cytomegalovirus Infected Cells. J Immunol Res 2017; 2017:4069260. [PMID: 28251165 PMCID: PMC5303859 DOI: 10.1155/2017/4069260] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 12/27/2016] [Indexed: 11/17/2022] Open
Abstract
Immunotoxins as antiviral therapeutics are largely unexplored but have promising prospective due to their high selectivity potential and their unparalleled efficiency. One recent example targeted the virus-encoded G protein-coupled receptor US28 as a strategy for specific and efficient treatment of human cytomegalovirus (HCMV) infections. US28 is expressed on virus-infected cells and scavenge chemokines by rapid internalization. The chemokine-based fusion-toxin protein (FTP) consisted of a variant (F49A) of CX3CL1 specifically targeting US28 linked to the catalytic domain of Pseudomonas exotoxin A (PE). Here, we systematically seek to improve F49A-FTP by modifications in its three structural domains; we generated variants with (1) altered chemokine sequence (K14A, F49L, and F49E), (2) shortened and elongated linker region, and (3) modified toxin domain. Only F49L-FTP displayed higher selectivity in its binding to US28 versus CX3CR1, the endogenous receptor for CX3CL1, but this was not matched by a more selective killing of US28-expressing cells. A longer linker and different toxin variants decreased US28 affinity and selective killing. Thereby, F49A-FTP represents the best candidate for HCMV treatment. Many viruses encode internalizing receptors suggesting that not only HCMV but also, for instance, Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus may be targeted by FTPs.
Collapse
|
16
|
Szpakowska M, Dupuis N, Baragli A, Counson M, Hanson J, Piette J, Chevigné A. Human herpesvirus 8-encoded chemokine vCCL2/vMIP-II is an agonist of the atypical chemokine receptor ACKR3/CXCR7. Biochem Pharmacol 2016; 114:14-21. [DOI: 10.1016/j.bcp.2016.05.012] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/25/2016] [Indexed: 10/21/2022]
|
17
|
Kleist AB, Getschman AE, Ziarek JJ, Nevins AM, Gauthier PA, Chevigné A, Szpakowska M, Volkman BF. New paradigms in chemokine receptor signal transduction: Moving beyond the two-site model. Biochem Pharmacol 2016; 114:53-68. [PMID: 27106080 DOI: 10.1016/j.bcp.2016.04.007] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022]
Abstract
Chemokine receptor (CKR) signaling forms the basis of essential immune cellular functions, and dysregulated CKR signaling underpins numerous disease processes of the immune system and beyond. CKRs, which belong to the seven transmembrane domain receptor (7TMR) superfamily, initiate signaling upon binding of endogenous, secreted chemokine ligands. Chemokine-CKR interactions are traditionally described by a two-step/two-site mechanism, in which the CKR N-terminus recognizes the chemokine globular core (i.e. site 1 interaction), followed by activation when the unstructured chemokine N-terminus is inserted into the receptor TM bundle (i.e. site 2 interaction). Several recent studies challenge the structural independence of sites 1 and 2 by demonstrating physical and allosteric links between these supposedly separate sites. Others contest the functional independence of these sites, identifying nuanced roles for site 1 and other interactions in CKR activation. These developments emerge within a rapidly changing landscape in which CKR signaling is influenced by receptor PTMs, chemokine and CKR dimerization, and endogenous non-chemokine ligands. Simultaneous advances in the structural and functional characterization of 7TMR biased signaling have altered how we understand promiscuous chemokine-CKR interactions. In this review, we explore new paradigms in CKR signal transduction by considering studies that depict a more intricate architecture governing the consequences of chemokine-CKR interactions.
Collapse
Affiliation(s)
- Andrew B Kleist
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Anthony E Getschman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Joshua J Ziarek
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Ave, Boston, MA 02115, USA.
| | - Amanda M Nevins
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| | - Pierre-Arnaud Gauthier
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Andy Chevigné
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Martyna Szpakowska
- Department of Infection and Immunity, Luxembourg Institute of Health, L-4354 Esch-sur-Alzette, Luxembourg.
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
18
|
Szpakowska M, Chevigné A. vCCL2/vMIP-II, the viral master KEYmokine. J Leukoc Biol 2015; 99:893-900. [DOI: 10.1189/jlb.2mr0815-383r] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 11/20/2015] [Indexed: 11/24/2022] Open
|
19
|
Cornaby C, Tanner A, Stutz EW, Poole BD, Berges BK. Piracy on the molecular level: human herpesviruses manipulate cellular chemotaxis. J Gen Virol 2015; 97:543-560. [PMID: 26669819 DOI: 10.1099/jgv.0.000370] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cellular chemotaxis is important to tissue homeostasis and proper development. Human herpesvirus species influence cellular chemotaxis by regulating cellular chemokines and chemokine receptors. Herpesviruses also express various viral chemokines and chemokine receptors during infection. These changes to chemokine concentrations and receptor availability assist in the pathogenesis of herpesviruses and contribute to a variety of diseases and malignancies. By interfering with the positioning of host cells during herpesvirus infection, viral spread is assisted, latency can be established and the immune system is prevented from eradicating viral infection.
Collapse
Affiliation(s)
- Caleb Cornaby
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Anne Tanner
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Eric W Stutz
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Brian D Poole
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| | - Bradford K Berges
- Department of Microbiology and Molecular Biology, Brigham Young University, Provo, UT 84602, USA
| |
Collapse
|
20
|
Rivas-Fuentes S, Salgado-Aguayo A, Pertuz Belloso S, Gorocica Rosete P, Alvarado-Vásquez N, Aquino-Jarquin G. Role of Chemokines in Non-Small Cell Lung Cancer: Angiogenesis and Inflammation. J Cancer 2015; 6:938-52. [PMID: 26316890 PMCID: PMC4543754 DOI: 10.7150/jca.12286] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2015] [Accepted: 06/23/2015] [Indexed: 12/12/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of aggressive cancer. The tumor tissue, which shows an active angiogenesis, is composed of neoplastic and stromal cells, and an abundant inflammatory infiltrate. Angiogenesis is important to support tumor growth, while infiltrating cells contribute to the tumor microenvironment through the secretion of growth factors, cytokines and chemokines, important molecules in the progression of the disease. Chemokines are important in development, activation of the immune response, and physiological angiogenesis. Chemokines have emerged as important regulators in the pathophysiology of cancer. These molecules are involved in the angiogenesis/angiostasis balance and in the recruitment of tumor infiltrating hematopoietic cells. In addition, chemokines promote tumor cell survival, as well as the directing and establishment of tumor cells to metastasis sites. The findings summarized here emphasize the central role of chemokines as modulators of tumor angiogenesis and their potential role as therapeutic targets in the inflammatory process of NSCLC angiogenesis.
Collapse
Affiliation(s)
- Selma Rivas-Fuentes
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Alfonso Salgado-Aguayo
- 2. Laboratory of Research on Rheumatic Diseases, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Silvana Pertuz Belloso
- 3. Department of Comparative Biology, Faculty of Sciences, National Autonomous University of Mexico, Mexico City, Mexico
| | - Patricia Gorocica Rosete
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Noé Alvarado-Vásquez
- 1. Department of Biochemistry Research, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Guillermo Aquino-Jarquin
- 4. Laboratory of Research on Genomics, Genetics and Bioinformatics. Tower of Haemato-oncology, Children´s Hospital of Mexico “Federico Gomez”, Mexico City, Mexico
| |
Collapse
|
21
|
Abstract
ABSTRACT Viruses have evolved to subvert host cell pathways to enable their replication and persistence. In particular, virus-encoded gene products target the host's immune system to evade elimination by antiviral immune defenses. Cytokines are soluble, secreted proteins, which regulate many aspects of immune responses, by providing signals through cell surface receptors on target cells. Cytokine pathways are therefore attractive targets for modulation by viruses during their replication cycle. This review deals with modulation of cytokine pathways by the human herpesvirus, a family of viruses that are capable of life-long persistence in the host and cause severe disease particularly in immunocompromised individuals.
Collapse
|
22
|
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
|
23
|
Cousins E, Nicholas J. Molecular biology of human herpesvirus 8: novel functions and virus-host interactions implicated in viral pathogenesis and replication. Recent Results Cancer Res 2014; 193:227-68. [PMID: 24008302 PMCID: PMC4124616 DOI: 10.1007/978-3-642-38965-8_13] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human herpesvirus 8 (HHV-8), also known as Kaposi's sarcoma-associated herpesvirus (KSHV), is the second identified human gammaherpesvirus. Like its relative Epstein-Barr virus, HHV-8 is linked to B-cell tumors, specifically primary effusion lymphoma and multicentric Castleman's disease, in addition to endothelial-derived KS. HHV-8 is unusual in its possession of a plethora of "accessory" genes and encoded proteins in addition to the core, conserved herpesvirus and gammaherpesvirus genes that are necessary for basic biological functions of these viruses. The HHV-8 accessory proteins specify not only activities deducible from their cellular protein homologies but also novel, unsuspected activities that have revealed new mechanisms of virus-host interaction that serve virus replication or latency and may contribute to the development and progression of virus-associated neoplasia. These proteins include viral interleukin-6 (vIL-6), viral chemokines (vCCLs), viral G protein-coupled receptor (vGPCR), viral interferon regulatory factors (vIRFs), and viral antiapoptotic proteins homologous to FLICE (FADD-like IL-1β converting enzyme)-inhibitory protein (FLIP) and survivin. Other HHV-8 proteins, such as signaling membrane receptors encoded by open reading frames K1 and K15, also interact with host mechanisms in unique ways and have been implicated in viral pathogenesis. Additionally, a set of micro-RNAs encoded by HHV-8 appear to modulate expression of multiple host proteins to provide conditions conducive to virus persistence within the host and could also contribute to HHV-8-induced neoplasia. Here, we review the molecular biology underlying these novel virus-host interactions and their potential roles in both virus biology and virus-associated disease.
Collapse
Affiliation(s)
- Emily Cousins
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans Street, Baltimore, MD, 21287, USA,
| | | |
Collapse
|
24
|
Bachelerie F, Ben-Baruch A, Burkhardt AM, Combadiere C, Farber JM, Graham GJ, Horuk R, Sparre-Ulrich AH, Locati M, Luster AD, Mantovani A, Matsushima K, Murphy PM, Nibbs R, Nomiyama H, Power CA, Proudfoot AEI, Rosenkilde MM, Rot A, Sozzani S, Thelen M, Yoshie O, Zlotnik A. International Union of Basic and Clinical Pharmacology. [corrected]. LXXXIX. Update on the extended family of chemokine receptors and introducing a new nomenclature for atypical chemokine receptors. Pharmacol Rev 2013; 66:1-79. [PMID: 24218476 DOI: 10.1124/pr.113.007724] [Citation(s) in RCA: 668] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Sixteen years ago, the Nomenclature Committee of the International Union of Pharmacology approved a system for naming human seven-transmembrane (7TM) G protein-coupled chemokine receptors, the large family of leukocyte chemoattractant receptors that regulates immune system development and function, in large part by mediating leukocyte trafficking. This was announced in Pharmacological Reviews in a major overview of the first decade of research in this field [Murphy PM, Baggiolini M, Charo IF, Hébert CA, Horuk R, Matsushima K, Miller LH, Oppenheim JJ, and Power CA (2000) Pharmacol Rev 52:145-176]. Since then, several new receptors have been discovered, and major advances have been made for the others in many areas, including structural biology, signal transduction mechanisms, biology, and pharmacology. New and diverse roles have been identified in infection, immunity, inflammation, development, cancer, and other areas. The first two drugs acting at chemokine receptors have been approved by the U.S. Food and Drug Administration (FDA), maraviroc targeting CCR5 in human immunodeficiency virus (HIV)/AIDS, and plerixafor targeting CXCR4 for stem cell mobilization for transplantation in cancer, and other candidates are now undergoing pivotal clinical trials for diverse disease indications. In addition, a subfamily of atypical chemokine receptors has emerged that may signal through arrestins instead of G proteins to act as chemokine scavengers, and many microbial and invertebrate G protein-coupled chemokine receptors and soluble chemokine-binding proteins have been described. Here, we review this extended family of chemokine receptors and chemokine-binding proteins at the basic, translational, and clinical levels, including an update on drug development. We also introduce a new nomenclature for atypical chemokine receptors with the stem ACKR (atypical chemokine receptor) approved by the Nomenclature Committee of the International Union of Pharmacology and the Human Genome Nomenclature Committee.
Collapse
Affiliation(s)
- Francoise Bachelerie
- Chair, Subcommittee on Chemokine Receptors, Nomenclature Committee-International Union of Pharmacology, Bldg. 10, Room 11N113, NIH, Bethesda, MD 20892.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Cytomegalovirus expresses the chemokine homologue vXCL1 capable of attracting XCR1+ CD4- dendritic cells. J Virol 2013; 88:292-302. [PMID: 24155383 DOI: 10.1128/jvi.02330-13] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Cytomegaloviruses (CMV) have developed various strategies to escape the immune system of the host. One strategy involves the expression of virus-encoded chemokines to modulate the host chemokine network. We have identified in the English isolate of rat CMV (murid herpesvirus 8 [MuHV8]) an open reading frame encoding a protein homologous to the chemokine XCL1, the only known C chemokine. Viral XCL1 (vXCL1), a glycosylated protein of 96 amino acids, can be detected 13 h postinfection in the supernatant of MuHV8-infected rat embryo fibroblasts. vXCL1 exclusively binds to CD4(-) rat dendritic cells (DC), a subset of DC that express the corresponding chemokine receptor XCR1. Like endogenous rat XCL1, vXCL1 selectively chemoattracts XCR1(+) CD4(-) DC. Since XCR1(+) DC in mice and humans have been shown to excel in antigen cross-presentation and thus in the induction of cytotoxic CD8(+) T lymphocytes, the virus has apparently hijacked this gene to subvert cytotoxic immune responses. The biology of vXCL1 offers an interesting opportunity to study the role of XCL1 and XCR1(+) DC in the cross-presentation of viral antigens.
Collapse
|
26
|
Liu Y, Pierce R, Luehmann HP, Sharp TL, Welch MJ. PET imaging of chemokine receptors in vascular injury-accelerated atherosclerosis. J Nucl Med 2013; 54:1135-41. [PMID: 23658218 PMCID: PMC4251467 DOI: 10.2967/jnumed.112.114777] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
UNLABELLED Atherosclerosis is the pathophysiologic process behind lethal cardiovascular diseases. It is a chronic inflammatory progression. Chemokines can strongly affect the initiation and progression of atherosclerosis by controlling the trafficking of inflammatory cells in vivo through interaction with their receptors. Some chemokine receptors have been reported to play an important role in plaque development and stability. However, the diagnostic potential of chemokine receptors has not yet been explored. The purpose of this study was to develop a positron emitter-radiolabeled probe to image the upregulation of chemokine receptor in a wire-injury-accelerated apolipoprotein E knockout (ApoE(-/-)) mouse model of atherosclerosis. METHODS A viral macrophage inflammatory protein II (vMIP-II) was used to image the upregulation of multiple chemokine receptors through conjugation with DOTA for (64)Cu radiolabeling and PET. Imaging studies were performed at 2 and 4 wk after injury in both wire-injured ApoE(-/-) and wild-type C57BL/6 mice. Competitive PET blocking studies with nonradiolabeled vMIP-II were performed to confirm the imaging specificity. Specific PET blocking with individual chemokine receptor antagonists was also performed to verify the upregulation of a particular chemokine receptor. In contrast, (18)F-FDG PET imaging was performed in both models to evaluate tracer uptake. Immunohistochemistry on the injury and sham tissues was performed to assess the upregulation of chemokine receptors. RESULTS (15)O-CO PET showed decreased blood volume in the femoral artery after the injury. (64)Cu-DOTA-vMIP-II exhibited fast in vivo pharmacokinetics with major renal clearance. PET images showed specific accumulation around the injury site, with consistent expression during the study period. Quantitative analysis of tracer uptake at the injury lesion in the ApoE(-/-) model showed a 3-fold increase over the sham-operated site and the sites in the injured wild-type mouse. (18)F-FDG PET showed significantly less tracer accumulation than (64)Cu-DOTA-vMIP-II, with no difference observed between injury and sham sites. PET blocking studies identified chemokine receptor-mediated (64)Cu-DOTA-vMIP-II uptake and verified the presence of 8 chemokine receptors, and this finding was confirmed by immunohistochemistry. CONCLUSION (64)Cu-DOTA-vMIP-II was proven a sensitive and useful PET imaging probe for the detection of 8 up-regulated chemokine receptors in a model of injury-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Yongjian Liu
- Department of Radiology, Washington University, St. Louis, Missouri 63110, USA.
| | | | | | | | | |
Collapse
|
27
|
Nomiyama H, Osada N, Yoshie O. Systematic classification of vertebrate chemokines based on conserved synteny and evolutionary history. Genes Cells 2012; 18:1-16. [PMID: 23145839 PMCID: PMC3568907 DOI: 10.1111/gtc.12013] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2012] [Accepted: 08/22/2012] [Indexed: 02/04/2023]
Abstract
The genes involved in host defences are known to undergo rapid evolution. Therefore, it is often difficult to assign orthologs in multigene families among various vertebrate species. Chemokines are a large family of small cytokines that orchestrate cell migration in health and disease. Herein, we have surveyed the genomes of 18 representative vertebrate species for chemokine genes and identified a total of 553 genes. We have determined their orthologous relationships and classified them in accordance with the current systematic chemokine nomenclature system. Our study reveals an interesting evolutionary history that gave origin and diversification to the vertebrate chemokine superfamily.
Collapse
Affiliation(s)
- Hisayuki Nomiyama
- Department of Molecular Enzymology, Kumamoto University Graduate School of Medical Sciences, Honjo, Kumamoto, 860-8556, Japan.
| | | | | |
Collapse
|
28
|
Lei Y, Takahama Y. XCL1 and XCR1 in the immune system. Microbes Infect 2011; 14:262-7. [PMID: 22100876 DOI: 10.1016/j.micinf.2011.10.003] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Accepted: 10/18/2011] [Indexed: 12/21/2022]
Abstract
XCL1, a C class chemokine also known as lymphotactin, is produced by T, NK, and NKT cells during infectious and inflammatory responses, whereas XCR1, the receptor of XCL1, is expressed by a dendritic cell subpopulation. The XCL1-XCR1 axis plays an important role in dendritic-cell-mediated cytotoxic immune response. It has been also shown that XCL1 and XCR1 are constitutively expressed in the thymus and regulate the thymic establishment of self-tolerance and the generation of regulatory T cells. This review summarizes the expression and function of XCL1 and XCR1 in the immune system.
Collapse
Affiliation(s)
- Yu Lei
- Key Laboratory of Molecular Biology for Infectious Disease, People's Republic of China Ministry of Education, Institute for Viral Hepatitis, The Second Affiliated Hospital, Chongqing Medical University, Chongqing 400010, China
| | | |
Collapse
|
29
|
Lüttichau HR. The cytomegalovirus UL146 gene product vCXCL1 targets both CXCR1 and CXCR2 as an agonist. J Biol Chem 2010; 285:9137-46. [PMID: 20044480 PMCID: PMC2838333 DOI: 10.1074/jbc.m109.002774] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Large DNA viruses, such as herpesvirus and poxvirus, encode proteins that target and exploit the chemokine system of their host. UL146 and UL147 in the cytomegalovirus (CMV) genome encode the two CXC chemokines vCXCL1 and vCXCL2. In this study, vCXCL1 was probed against a panel of the 18 classified human chemokine receptors. In calcium mobilization assays vCXCL1 acted as an agonist on both CXCR1 and CXCR2 but did not activate or block any of the other 16 chemokine receptors. vCXCL1 was characterized and compared with CXCL1/GROalpha, CXCL2/GRObeta, CXCL3/GROgamma, CXCL5/ENA-78, CXCL6/GCP-2, CXCL7/NAP-2 and CXCL8/IL-8 in competition binding, calcium mobilization, inositol triphosphate turnover, and chemotaxis assays using CXCR1- and CXCR2-expressing Chinese hamster ovary, 300.19, COS7, and L1.2 cells. The affinities of vCXCL1 for the CXCR1 and CXCR2 receptors were 44 and 5.6 nm, respectively, as determined in competition binding against radioactively labeled CXCL8. In calcium mobilization, phosphatidylinositol turnover, and chemotaxis assays, vCXCL1 acted as a highly efficacious activator of both receptors, with a rather low potency for the CXCR1 receptor but comparable with CXCL5 and CXCL7. It is suggested that CMV uses the UL146 gene product expressed in infected endothelial cells to attract neutrophils by activating their CXCR1 and CXCR2 receptors, whereby neutrophils can act as carriers of the virus to uninfected endothelial cells. In that way a lasting pool of CMV-infected endothelial cells could be maintained.
Collapse
Affiliation(s)
- Hans R Lüttichau
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Panum Institute, University of Copenhagen, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
30
|
Abstract
Human herpesvirus (HHV)-8, also called Kaposi's sarcoma-associated herpesvirus, was discovered in 1994 and was rapidly sequenced, revealing several unique and surprising features of its genetic makeup. Among these discoveries was the identification of the first viral homolog of IL-6 and three CC/beta-chemokine ligands (viral CCL-1, -2 and -3), not previously found in gamma-herpesviruses. Viral IL-6 was immediately recognized as a potential contributor to HHV-8 pathogenesis, specifically endothelial-derived Kaposi's sarcoma and the B-cell malignancy multicentric Castleman's disease with which IL-6, a proangiogenic and B-cell growth factor, had previously been implicated. The roles of the viral chemokines were speculated to involve immune evasion; however, like viral IL-6, the viral chemokines have the potential to contribute to pathogenesis through their shared angiogenic activities, known to be important for Kaposi's sarcoma and HHV-8-associated primary effusion lymphoma, and also via direct prosurvival activities. This article will discuss the molecular properties, activities and functions of viral IL-6 and the viral CCLs, proteins that could provide appropriate targets for antiviral and therapeutic strategies.
Collapse
Affiliation(s)
- John Nicholas
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Department of Oncology, Johns Hopkins School of Medicine, 1650 Orleans Street, Room 309, Baltimore, MD 21287, USA, Tel.: +1 410 502 6801, ,
| |
Collapse
|
31
|
Neospora caninum excreted/secreted antigens trigger CC-chemokine receptor 5-dependent cell migration. Int J Parasitol 2010; 40:797-805. [PMID: 20060395 DOI: 10.1016/j.ijpara.2009.12.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2009] [Revised: 12/04/2009] [Accepted: 12/06/2009] [Indexed: 01/10/2023]
Abstract
Neospora caninum, the causative agent of neosporosis, is an obligate intracellular parasite considered to be a major cause of abortion in cattle throughout the world. Most studies concerning N. caninum have focused on life cycle, seroepidemiology, pathology and vaccination, while data on host-parasite interaction, such as host cell migration, mechanisms of evasion and dissemination of this parasite during the early phase of infection are still poorly understood. Here we show the ability of excreted/secreted antigens from N. caninum (NcESAs) to attract monocytic cells to the site of primary infection in both in vitro and in vivo assays. Molecules from the family of cyclophilins present on the NcESAs were shown to work as chemokine-like proteins and NcESA-induced chemoattraction involved G(i) protein signaling and participation of CC-chemokine receptor 5 (CCR5). Additionally, we demonstrate the ability of NcESAs to enhance the expression of CCR5 on monocytic cells and this increase occurred in parallel with the chemotactic activity of NcESAs by increasing cell migration. These results suggest that during the first days of infection, N. caninum produces molecules capable of inducing monocytic cell migration to the sites of infection, which will consequently enhance initial parasite invasion and proliferation. Altogether, these results help to clarify some key features involved in the process of cell migration and may reveal virulence factors and therapeutic targets to control neosporosis.
Collapse
|
32
|
Autocrine and paracrine promotion of cell survival and virus replication by human herpesvirus 8 chemokines. J Virol 2008; 82:6501-13. [PMID: 18434408 DOI: 10.1128/jvi.02396-07] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Human herpesvirus 8 (HHV-8), which is associated with the endothelial tumor Kaposi's sarcoma, encodes three CC/beta-chemokines. These are expressed early during productive (lytic) infection and are believed to be involved in immune evasion, in addition to viral pathogenesis via induction of angiogenic cytokines. Here we report that two of the HHV-8 chemokines, CCR8 agonists vCCL-1 and vCCL-2, have direct effects on endothelial survival and virus replication. The v-chemokines stimulated virus replication when added to infected cultures exogenously, and CCR8 knockdown absent v-chemokine supplementation inhibited virus production, indicative of autocrine effects of endogenously produced vCCLs. This was verified and proreplication functions of each chemokine were demonstrated via shRNA-mediated vCCL depletion. The v-chemokines inhibited expression of lytic cycle-induced proapoptotic protein Bim, RNA interference-mediated suppression of which mimicked v-chemokine proreplication functions. Our data show for the first time that the v-chemokines have direct effects on virus biology, independently of their postulated immune evasion functions, and suggest that in vivo the v-chemokines might play direct roles in Kaposi's sarcomagenesis via paracrine prosurvival signaling.
Collapse
|
33
|
Lüttichau HR. The herpesvirus 8 encoded chemokines vCCL2 (vMIP-II) and vCCL3 (vMIP-III) target the human but not the murine lymphotactin receptor. Virol J 2008; 5:50. [PMID: 18426556 PMCID: PMC2359738 DOI: 10.1186/1743-422x-5-50] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2007] [Accepted: 04/21/2008] [Indexed: 11/24/2022] Open
Abstract
Background Large DNA-viruses such as herpesvirus and poxvirus encode proteins that target and exploit the chemokine system of their host. The Kaposi sarcoma- associated herpes virus (KSHV) encodes three chemokines. Two of these, vCCL2 and vCCL3, target the human lymphotactin receptor as an antagonist and a selective agonist, respectively. Therefore these virally endcoded chemokines have the potential to be used as tools in the study of lymphotactin receptor pathways in murine models. Results The activities of vCCL2, vCCL3, human lymphotactin (XCL1) and murine lymphotactin (mXCL1) were probed in parallel on the human and murine lymphotactin receptor (XCR1 and mXCR1) using a phosphatidyl-inositol assay. On the human XCR1, vCCL3, mXCL1 and XCL1 acted as agonists. In contrast, only mXCL1 was able to activate the murine lymphotactin receptor. Using the same assay, vCCL2 was able to block the response using any of the three agonists on the humane lymphotactin receptor with IC50s of 2–3 nM. However, vCCL2 was unable to block the response of mXCL1 through the murine lymphotactin receptor. Conclusion This study shows that vCCL2 and vCCL3 cannot be used to investigate lymphotactin receptor pathways in murine models. These results also add vCCL2 and vCCL3 to a growing list of viral chemokines with known human chemokine receptor targets, which do not target the corresponding murine receptors. This fits with the observation that viral and endogenous ligands for the same human chemokine receptor tend to have relatively divergent amino-acid sequences, suggesting that these viruses have fine-tuned the design of their chemokines such that the action of the viral encoded chemokines cannot be expected to cross species barriers.
Collapse
Affiliation(s)
- Hans R Lüttichau
- Laboratory for Molecular Pharmacology, Department of Neuroscience and Pharmacology, Panum Institute, DK-2200 Copenhagen, Denmark.
| |
Collapse
|
34
|
Garrote JA, Gómez E, León AJ, Bernardo D, Calvo C, Fernández-Salazar L, Blanco-Quirós A, Arranz E. Cytokine, Chemokine and Immune Activation Pathway Profiles in Celiac Disease: An Immune System Activity Screening by Expression Macroarrays. Drug Target Insights 2008. [DOI: 10.4137/dti.s399] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Affiliation(s)
- José A. Garrote
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
- Research Unit. (Spain)
| | - Emma Gómez
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | - Alberto J. León
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | - David Bernardo
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | | | - Luis Fernández-Salazar
- Adults Digestive Diseases Services. Hospital Clinico Universitario of Valladolid. (Spain)
| | - Alfredo Blanco-Quirós
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| | - Eduardo Arranz
- Group of Mucosal Immunology. Pediatrics and Immunology Areas- Instituto de Biologia y Genética Molecular (IBGM). University of Valladolid. (Spain)
| |
Collapse
|