1
|
Hippee CE, Singh BK, Thurman AL, Cooney AL, Pezzulo AA, Cattaneo R, Sinn PL. Measles virus exits human airway epithelia within dislodged metabolically active infectious centers. PLoS Pathog 2021; 17:e1009458. [PMID: 34383863 PMCID: PMC8384213 DOI: 10.1371/journal.ppat.1009458] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 08/24/2021] [Accepted: 07/27/2021] [Indexed: 11/24/2022] Open
Abstract
Measles virus (MeV) is the most contagious human virus. Unlike most respiratory viruses, MeV does not directly infect epithelial cells upon entry in a new host. MeV traverses the epithelium within immune cells that carry it to lymphatic organs where amplification occurs. Infected immune cells then synchronously deliver large amounts of virus to the airways. However, our understanding of MeV replication in airway epithelia is limited. To model it, we use well-differentiated primary cultures of human airway epithelial cells (HAE) from lung donors. In HAE, MeV spreads directly cell-to-cell forming infectious centers that grow for ~3–5 days, are stable for a few days, and then disappear. Transepithelial electrical resistance remains intact during the entire course of HAE infection, thus we hypothesized that MeV infectious centers may dislodge while epithelial function is preserved. After documenting by confocal microscopy that infectious centers progressively detach from HAE, we recovered apical washes and separated cell-associated from cell-free virus by centrifugation. Virus titers were about 10 times higher in the cell-associated fraction than in the supernatant. In dislodged infectious centers, ciliary beating persisted, and apoptotic markers were not readily detected, suggesting that they retain functional metabolism. Cell-associated MeV infected primary human monocyte-derived macrophages, which models the first stage of infection in a new host. Single-cell RNA sequencing identified wound healing, cell growth, and cell differentiation as biological processes relevant for infectious center dislodging. 5-ethynyl-2’-deoxyuridine (EdU) staining located proliferating cells underneath infectious centers. Thus, cells located below infectious centers divide and differentiate to repair the dislodged infected epithelial patch. As an extension of these studies, we postulate that expulsion of infectious centers through coughing and sneezing could contribute to MeV’s strikingly high reproductive number by allowing the virus to survive longer in the environment and by delivering a high infectious dose to the next host. Measles virus (MeV) is a respiratory pathogen that infects millions worldwide each year. Although sometimes mischaracterized as an innocuous childhood disease, measles remains a leading cause of death for children under five. MeV is the most contagious human virus and requires vaccination rates above 90% to maintain herd immunity. Global decreases in vaccination rates over the past ten years contributed to recent, widespread MeV outbreaks. We uncover here a novel mechanism by which MeV exits the human airways that may explain why it is much more contagious than other viruses. We document that infected cells containing cell-associated virus detach en masse from the airway epithelial sheet. These dislodged infectious centers are metabolically active and can transmit infection to primary human monocyte-derived macrophages via cell-cell contact as efficiently as cell-free virus particles. Thus, cell-associated MeV could spread host-to-host and is a potentially vital strategy for efficient respiratory virus transmission.
Collapse
Affiliation(s)
- Camilla E. Hippee
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Brajesh K. Singh
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Andrew L. Thurman
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Ashley L. Cooney
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Alejandro A. Pezzulo
- Department of Internal Medicine, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota, United States of America
| | - Patrick L. Sinn
- Stead Family Department of Pediatrics, Carver College of Medicine, The University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
2
|
Arruda B, Shen H, Zheng Y, Li G. Novel Morbillivirus as Putative Cause of Fetal Death and Encephalitis among Swine. Emerg Infect Dis 2021; 27:1858-1866. [PMID: 34152961 PMCID: PMC8237871 DOI: 10.3201/eid2707.203971] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Morbilliviruses are highly contagious pathogens. The Morbillivirus genus includes measles virus, canine distemper virus (CDV), phocine distemper virus (PDV), peste des petits ruminants virus, rinderpest virus, and feline morbillivirus. We detected a novel porcine morbillivirus (PoMV) as a putative cause of fetal death, encephalitis, and placentitis among swine by using histopathology, metagenomic sequencing, and in situ hybridization. Phylogenetic analyses showed PoMV is most closely related to CDV (62.9% nt identities) and PDV (62.8% nt identities). We observed intranuclear inclusions in neurons and glial cells of swine fetuses with encephalitis. Cellular tropism is similar to other morbilliviruses, and PoMV viral RNA was detected in neurons, respiratory epithelium, and lymphocytes. This study provides fundamental knowledge concerning the pathology, genome composition, transmission, and cellular tropism of a novel pathogen within the genus Morbillivirus and opens the door to a new, applicable disease model to drive research forward.
Collapse
|
3
|
Lin WHW, Moran E, Adams RJ, Sievers RE, Hauer D, Godin S, Griffin DE. A durable protective immune response to wild-type measles virus infection of macaques is due to viral replication and spread in lymphoid tissues. Sci Transl Med 2021; 12:12/537/eaax7799. [PMID: 32238577 DOI: 10.1126/scitranslmed.aax7799] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 08/15/2019] [Accepted: 12/02/2019] [Indexed: 12/21/2022]
Abstract
Infection with wild-type (WT) measles virus (MeV) is an important cause of childhood mortality that leads to lifelong protective immunity in survivors. WT MeV and the live-attenuated MeV used in the measles vaccine (LAMV) are antigenically similar, but the determinants of attenuation are unknown, and protective immunity induced by LAMV is less robust than that induced by WT MeV. To identify factors that contribute to these differences, we compared virologic and immunologic responses after respiratory infection of rhesus macaques with WT MeV or LAMV. In infected macaques, WT MeV replicated efficiently in B and T lymphocytes with spreading throughout lymphoid tissues resulting in prolonged persistence of viral RNA. In contrast, LAMV replicated efficiently in the respiratory tract but displayed limited spread to lymphoid tissue or peripheral blood mononuclear cells. In vitro, WT MeV and LAMV replicated similarly in macaque primary respiratory epithelial cells and human lymphocytes, but LAMV-infected lymphocytes produced little virus. Plasma concentrations of interleukin-1β (IL-1β), IL-12, interferon-γ (IFN-γ), CCL2, CCL11, CXCL9, and CXCL11 increased in macaques after WT MeV but not LAMV infection. WT MeV infection induced more protective neutralizing, hemagglutinin-specific antibodies and bone marrow plasma cells than did LAMV infection, although numbers of MeV-specific IFN-γ- and IL-4-producing T cells were comparable. Therefore, MeV attenuation may involve altered viral replication in lymphoid tissue that limited spread and decreased the host antibody response, suggesting a link between lifelong protective immunity and the ability of WT MeV, but not LAMV, to spread in lymphocytes.
Collapse
Affiliation(s)
- Wen-Hsuan W Lin
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Eileen Moran
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Robert J Adams
- Department of Molecular and Comparative Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Robert E Sievers
- Department of Chemistry, University of Colorado, Boulder, CO 80309, USA
| | - Debra Hauer
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | | | - Diane E Griffin
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
4
|
Thépaut M, Luczkowiak J, Vivès C, Labiod N, Bally I, Lasala F, Grimoire Y, Fenel D, Sattin S, Thielens N, Schoehn G, Bernardi A, Delgado R, Fieschi F. DC/L-SIGN recognition of spike glycoprotein promotes SARS-CoV-2 trans-infection and can be inhibited by a glycomimetic antagonist. PLoS Pathog 2021; 17:e1009576. [PMID: 34015061 PMCID: PMC8136665 DOI: 10.1371/journal.ppat.1009576] [Citation(s) in RCA: 124] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/20/2021] [Indexed: 12/26/2022] Open
Abstract
The efficient spread of SARS-CoV-2 resulted in a unique pandemic in modern history. Despite early identification of ACE2 as the receptor for viral spike protein, much remains to be understood about the molecular events behind viral dissemination. We evaluated the contribution of C-type lectin receptors (CLRS) of antigen-presenting cells, widely present in respiratory mucosa and lung tissue. DC-SIGN, L-SIGN, Langerin and MGL bind to diverse glycans of the spike using multiple interaction areas. Using pseudovirus and cells derived from monocytes or T-lymphocytes, we demonstrate that while virus capture by the CLRs examined does not allow direct cell infection, DC/L-SIGN, among these receptors, promote virus transfer to permissive ACE2+ Vero E6 cells. A glycomimetic compound designed against DC-SIGN, enable inhibition of this process. These data have been then confirmed using authentic SARS-CoV-2 virus and human respiratory cell lines. Thus, we described a mechanism potentiating viral spreading of infection.
Collapse
Affiliation(s)
- Michel Thépaut
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Joanna Luczkowiak
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Corinne Vivès
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Nuria Labiod
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Isabelle Bally
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Fátima Lasala
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Yasmina Grimoire
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Daphna Fenel
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Sara Sattin
- Universita`degli Studi di Milano, Dipartimento di Chimica, Milano, Italy
| | - Nicole Thielens
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Guy Schoehn
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| | - Anna Bernardi
- Universita`degli Studi di Milano, Dipartimento di Chimica, Milano, Italy
| | - Rafael Delgado
- Instituto de Investigación Hospital Universitario 12 de Octubre (imas12), Universidad Complutense School of Medicine, Madrid, Spain
| | - Franck Fieschi
- Univ. Grenoble Alpes, CNRS, CEA, Institut de Biologie Structurale, Grenoble, France
| |
Collapse
|
5
|
Haeryfar SMM. On invariant T cells and measles: A theory of "innate immune amnesia". PLoS Pathog 2020; 16:e1009071. [PMID: 33332470 PMCID: PMC7745983 DOI: 10.1371/journal.ppat.1009071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- S. M. Mansour Haeryfar
- Department of Microbiology and Immunology, Western University, London, Ontario, Canada
- Department of Medicine, Division of Clinical Immunology & Allergy, Western University, London, Ontario, Canada
- Department of Surgery, Division of General Surgery, Western University, London, Ontario, Canada
- Centre for Human Immunology, Western University, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
6
|
Ojeda N, Salazar C, Cárdenas C, Marshall SH. Expression of DC-SIGN-like C-Type Lectin Receptors in Salmo salar. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2020; 113:103806. [PMID: 32739503 PMCID: PMC7392198 DOI: 10.1016/j.dci.2020.103806] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 07/20/2020] [Accepted: 07/20/2020] [Indexed: 05/05/2023]
Abstract
C-Type Lectin Receptors (CTLR) are involved in the activation of innate and adaptative immune responses. Among these receptors, the Dendritic Cell-Specific ICAM-3-Grabbing nonintegrin (DC-SIGN/CD209) has become a hot topic due to its ability to bind and facilitate the infections processes of several pathogens. Although well characterized in mammals, little documentation exists about the receptor in salmonid fishes. Here, we report the sequence and expression analysis of eight DC-SIGN-like genes in Salmo salar. Each receptor displays structural similarities to DC-SIGN molecules described in mammals, including internalization motifs, a neck region with heptad repeats, and a Ca+2-dependent carbohydrate recognition domain. The receptors are expressed in multiple tissues of fish, and fish cell lines, with differential expression upon infection with viral and bacterial pathogens. The identification of DC-SIGN-like receptors in Salmo salar provides new information regarding the structure of the immune system of salmon, potential markers for cell subsets, as well as insights into DC-SIGN conservation across species.
Collapse
Affiliation(s)
- Nicolás Ojeda
- Instituto de Biologia, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Carolina Salazar
- Instituto de Biologia, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Constanza Cárdenas
- Instituto de Biologia, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile
| | - Sergio H Marshall
- Instituto de Biologia, Pontificia Universidad Catolica de Valparaiso, Valparaiso, Chile.
| |
Collapse
|
7
|
Measles skin rash: Infection of lymphoid and myeloid cells in the dermis precedes viral dissemination to the epidermis. PLoS Pathog 2020; 16:e1008253. [PMID: 33031460 PMCID: PMC7575069 DOI: 10.1371/journal.ppat.1008253] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 10/20/2020] [Accepted: 09/11/2020] [Indexed: 12/22/2022] Open
Abstract
Measles is characterized by fever and a maculopapular skin rash, which is accompanied by immune clearance of measles virus (MV)-infected cells. Histopathological analyses of skin biopsies from humans and non-human primates (NHPs) with measles rash have identified MV-infected keratinocytes and mononuclear cells in the epidermis, around hair follicles and near sebaceous glands. Here, we address the pathogenesis of measles skin rash by combining data from experimentally infected NHPs, ex vivo infection of human skin sheets and in vitro infection of primary human keratinocytes. Analysis of NHP skin samples collected at different time points following MV inoculation demonstrated that infection in the skin precedes onset of rash by several days. MV infection was detected in lymphoid and myeloid cells in the dermis before dissemination to the epidermal leukocytes and keratinocytes. These data were in good concordance with ex vivo MV infections of human skin sheets, in which dermal cells were more targeted than the epidermal cells. To address viral dissemination to the epidermis and to determine whether the dissemination is receptor-dependent, we performed experimental infections of primary keratinocytes collected from healthy donors. These experiments demonstrated that MV infection of keratinocytes is mainly nectin-4-dependent, and differentiated keratinocytes, which express higher levels of nectin-4, are more susceptible to MV infection than proliferating keratinocytes. Based on these data, we propose a model to explain measles skin rash: migrating MV-infected lymphocytes initiate the infection of dermal skin-resident CD150+ immune cells. The infection is subsequently disseminated from the dermal papillae to nectin-4+ keratinocytes in the basal epidermis. Lateral spread of MV infection is observed in the superficial epidermis, most likely due to the higher level of nectin-4 expression on differentiated keratinocytes. Finally, MV-infected cells are cleared by infiltrating immune cells, causing hyperemia and edema, which give the appearance of morbilliform skin rash. Several viral infections are associated with skin rash, including parvovirus B19, human herpesvirus type 6, dengue virus and rubella virus. However, the archetype virus infection that leads to skin rash is measles. Although all of these viral exanthemata often appear similar, their pathogenesis is different. In the case of measles, the appearance of skin rash is a sign that the immune system is clearing MV-infected cells from the skin. How the virus reaches the skin and is locally disseminated remains unknown. Here, we combine observations and expertise from pathologists, dermatologists, virologists and immunologists to delineate the pathogenesis of measles skin rash. We show that MV infection of dermal myeloid and lymphoid cells precedes viral dissemination to the epidermal leukocytes and keratinocytes. We speculate that immune-mediated clearance of these infected cells results in hyperemia and edema, explaining the redness of the skin and the slightly elevated spots of the morbilliform rash.
Collapse
|
8
|
Measles pathogenesis, immune suppression and animal models. Curr Opin Virol 2020; 41:31-37. [PMID: 32339942 DOI: 10.1016/j.coviro.2020.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 03/05/2020] [Accepted: 03/06/2020] [Indexed: 02/09/2023]
Abstract
Measles virus causes a disease with seemingly innocent symptoms, such as fever and rash. However, measles immune suppression causes increased susceptibility to opportunistic infections that are responsible for the majority of over 100000 yearly fatalities. The pathogenesis of measles is complex, because measles virus uses multiple receptors to infect different cell types in different phases of the disease. Experimental morbillivirus infections with wild-type viruses in natural host species have demonstrated that direct infection and depletion of memory immune cells causes immune amnesia. This was confirmed in studies of a measles outbreak in unvaccinated children and provides an explanation for epidemiological observations of long-term increases in morbidity and mortality after measles.
Collapse
|
9
|
Griffin DE. Measles virus persistence and its consequences. Curr Opin Virol 2020; 41:46-51. [PMID: 32387998 PMCID: PMC7492426 DOI: 10.1016/j.coviro.2020.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 03/13/2020] [Accepted: 03/19/2020] [Indexed: 11/16/2022]
Abstract
Clearance of measles virus is complex. Infectious virus is cleared by the adaptive immune response manifested by the characteristic maculopapular rash. CD8+ T cells are major effectors of infectious virus clearance, a process that may fail in individuals with compromised cellular immune responses leading to progressive giant cell pneumonia and/or measles inclusion body encephalitis. In contrast to the usual rapid clearance of infectious virus, clearance of viral RNA is slow with persistence in lymphoid tissue for many months. Persistence of MeV RNA may contribute to the late development of the slowly progressive disease subacute sclerosing panencephalitis in children infected at a young age and to measles-associated immune suppression but also to maturation of the immune response and development of life-long immunity.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
10
|
Muñoz-Fontela C, McElroy AK. Ebola Virus Disease in Humans: Pathophysiology and Immunity. Curr Top Microbiol Immunol 2019; 411:141-169. [PMID: 28653186 PMCID: PMC7122202 DOI: 10.1007/82_2017_11] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Viruses of the Ebolavirus genus cause sporadic epidemics of severe and systemic febrile disease that are fueled by human-to-human transmission. Despite the notoriety of ebolaviruses, particularly Ebola virus (EBOV), as prominent viral hemorrhagic fever agents, and the international concern regarding Ebola virus disease (EVD) outbreaks, very little is known about the pathophysiology of EVD in humans and, in particular, about the human immune correlates of survival and immune memory. This lack of basic knowledge about physiological characteristics of EVD is probably attributable to the dearth of clinical and laboratory data gathered from past outbreaks. The unprecedented magnitude of the EVD epidemic that occurred in West Africa from 2013 to 2016 has allowed, for the first time, evaluation of clinical, epidemiological, and immunological parameters in a significant number of patients using state-of-the-art laboratory equipment. This review will summarize the data from the literature regarding human pathophysiologic and immunologic responses to filoviral infection.
Collapse
Affiliation(s)
- César Muñoz-Fontela
- Laboratory of Emerging Viruses, Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistraße 52, 20251, Hamburg, Germany.
| | - Anita K McElroy
- Infectious Diseases, Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Drive NE, Atlanta, GA, 30322, USA.
| |
Collapse
|
11
|
Studies into the mechanism of measles-associated immune suppression during a measles outbreak in the Netherlands. Nat Commun 2018; 9:4944. [PMID: 30470742 PMCID: PMC6251901 DOI: 10.1038/s41467-018-07515-0] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 11/07/2018] [Indexed: 01/08/2023] Open
Abstract
Measles causes a transient immune suppression, leading to increased susceptibility to opportunistic infections. In experimentally infected non-human primates (NHPs) measles virus (MV) infects and depletes pre-existing memory lymphocytes, causing immune amnesia. A measles outbreak in the Dutch Orthodox Protestant community provided a unique opportunity to study the pathogenesis of measles immune suppression in unvaccinated children. In peripheral blood mononuclear cells (PBMC) of prodromal measles patients, we detected MV-infected memory CD4+ and CD8+ T cells and naive and memory B cells at similar levels as those observed in NHPs. In paired PBMC collected before and after measles we found reduced frequencies of circulating memory B cells and increased frequencies of regulatory T cells and transitional B cells after measles. These data support our immune amnesia hypothesis and offer an explanation for the previously observed long-term effects of measles on host resistance. This study emphasises the importance of maintaining high measles vaccination coverage. The mechanisms by which measles virus infection induces transient immune suppression in humans are poorly understood. Here, Laksono and colleagues characterise the pathogenesis of measles-associated immune suppression in unvaccinated children, and shed new light on the long-term effects of measles on the host.
Collapse
|
12
|
Canine Distemper Virus Spread and Transmission to Naive Ferrets: Selective Pressure on Signaling Lymphocyte Activation Molecule-Dependent Entry. J Virol 2018; 92:JVI.00669-18. [PMID: 29793948 DOI: 10.1128/jvi.00669-18] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 05/14/2018] [Indexed: 12/12/2022] Open
Abstract
Upon infection, morbilliviruses such as measles virus, rinderpest virus, and canine distemper virus (CDV) initially target immune cells via the signaling lymphocyte activation molecule (SLAM) before spreading to respiratory epithelia through the adherens junction protein nectin-4. However, the roles of these receptors in transmission from infected to naive hosts have not yet been formally tested. To experimentally addressing this question, we established a model of CDV contact transmission between ferrets. We show here that transmission of wild-type CDV sometimes precedes the onset of clinical disease. In contrast, transmission was not observed in most animals infected with SLAM- or nectin-4-blind CDVs, even though all animals infected with the nectin-4-blind virus developed sustained viremia. There was an unexpected case of transmission of a nectin-4-blind virus, possibly due to biting. Another unprecedented event was transient viremia in an infection with a SLAM-blind virus. We identified three compensatory mutations within or near the SLAM-binding surface of the attachment protein. A recombinant CDV expressing the mutated attachment protein regained the ability to infect ferret lymphocytes in vitro, but its replication was not as efficient as that of wild-type CDV. Ferrets infected with this virus developed transient viremia and fever, but there was no transmission to naive contacts. Our study supports the importance of epithelial cell infection and of sequential CDV H protein interactions first with SLAM and then nectin-4 receptors for transmission to naive hosts. It also highlights the in vivo selection pressure on the H protein interactions with SLAM.IMPORTANCE Morbilliviruses such as measles virus, rinderpest virus, and canine distemper virus (CDV) are highly contagious. Despite extensive knowledge of how morbilliviruses interact with their receptors, little is known about how those interactions influence viral transmission to naive hosts. In a ferret model of CDV contact transmission, we showed that sequential use of the signaling lymphocytic activation molecule (SLAM) and nectin-4 receptors is essential for transmission. In one animal infected with a SLAM-blind CDV, we documented mild viremia due to the acquisition of three compensatory mutations within or near the SLAM-binding surface. The interaction, however, was not sufficient to cause disease or sustain transmission to naive contacts. This work confirms the sequential roles of SLAM and nectin-4 in morbillivirus transmission and highlights the selective pressure directed toward productive interactions with SLAM.
Collapse
|
13
|
Mascia C, Pozzetto I, Kertusha B, Marocco R, Del Borgo C, Tieghi T, Vita S, Savinelli S, Iannetta M, Vullo V, Lichtner M, Mastroianni CM. Persistent high plasma levels of sCD163 and sCD14 in adult patients with measles virus infection. PLoS One 2018; 13:e0198174. [PMID: 29795672 PMCID: PMC5967820 DOI: 10.1371/journal.pone.0198174] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2018] [Accepted: 05/15/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND AND AIMS Measles is an infectious disease that represents a serious public health problem worldwide, being associated with increased susceptibility to secondary infections, especially in the respiratory and gastrointestinal tracts. The aim of this study was to evaluate sCD163 and sCD14 levels in measles virus (MV) infected patients, as markers of immune activation, in order to better understand their role in the pathogenesis of the disease. TNF-α plasma levels were also evaluated. METHODS sCD163, sCD14 and TNF-α were measured by ELISA in plasma samples of 27 MV infected patients and 27 healthy donors (HD) included as controls. RESULTS At the time of hospital admission, sCD163 and sCD14 levels were significantly higher in MV infected patients than in HD, while a decrease in TNF-α levels were found even if without statistical significance. sCD163 and sCD14 levels were significantly decreased after two months from acute infection compared to hospital admission although they remained significantly higher compared to HD. TNF-α levels increased significantly during the follow-up period. Considering clinical parameters, sCD163 levels positively correlated with aspartate aminotransferase, white blood cell count and neutrophils rate, while negatively correlated with the lymphocyte percentage. sCD14 levels positively correlated with the neutrophil and lymphocyte percentages. CONCLUSIONS These results indicate that, despite the resolution of symptoms, an important macrophage/monocyte activation persists in measles patients, even after two months from infection.
Collapse
Affiliation(s)
- Claudia Mascia
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Irene Pozzetto
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- Infectious Diseases Unit, Sapienza University, S. M. Goretti Hospital, Latina, Italy
| | - Blerta Kertusha
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- Infectious Diseases Unit, Sapienza University, S. M. Goretti Hospital, Latina, Italy
| | - Raffaella Marocco
- Infectious Diseases Unit, Sapienza University, S. M. Goretti Hospital, Latina, Italy
| | - Cosmo Del Borgo
- Infectious Diseases Unit, Sapienza University, S. M. Goretti Hospital, Latina, Italy
| | - Tiziana Tieghi
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- Infectious Diseases Unit, Sapienza University, S. M. Goretti Hospital, Latina, Italy
| | - Serena Vita
- Infectious Diseases Unit, Sapienza University, S. M. Goretti Hospital, Latina, Italy
| | - Stefano Savinelli
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Marco Iannetta
- National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Rome, Italy
| | - Vincenzo Vullo
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
| | - Miriam Lichtner
- Department of Public Health and Infectious Diseases, Sapienza University, Rome, Italy
- Infectious Diseases Unit, Sapienza University, S. M. Goretti Hospital, Latina, Italy
| | | |
Collapse
|
14
|
De Vries RD, Rennick LJ, Duprex WP, De Swart RL. Paramyxovirus Infections in Ex Vivo Lung Slice Cultures of Different Host Species. Methods Protoc 2018; 1:E12. [PMID: 31164557 PMCID: PMC6526457 DOI: 10.3390/mps1020012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 03/11/2018] [Accepted: 03/21/2018] [Indexed: 11/17/2022] Open
Abstract
In vivo experiments in animal models of disease are of crucial importance for viral tropism and pathogenesis studies. However, these experiments must be complemented with in vitro and ex vivo experiments. Here, we describe a protocol for the preparation and ex vivo infection of lung slices from different mammalian host species with various respiratory paramyxoviruses expressing fluorescent reporter proteins, and suggest follow-up experiments including immunohistochemistry, flow cytometry and confocal microscopy.
Collapse
Affiliation(s)
- Rory D De Vries
- Department of Viroscience, Erasmus MC, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| | - Linda J Rennick
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - W Paul Duprex
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Rik L De Swart
- Department of Viroscience, Erasmus MC, P.O. Box 2040, 3000 CA Rotterdam, The Netherlands.
| |
Collapse
|
15
|
Abstract
Measles is an acute systemic viral disease with initial amplification of infection in lymphoid tissue and subsequent spread over 10–14 days to multiple organs. Failure of the innate response to control initial measles virus (MeV) replication is associated with the ability of MeV to inhibit the induction of type I interferon and interferon-stimulated antiviral genes. Rather, the innate response is characterized by the expression of proteins regulated by nuclear factor kappa B and the inflammasome. With eventual development of the adaptive response, the rash appears with immune cell infiltration into sites of virus replication to initiate the clearance of infectious virus. However, MeV RNA is cleared much more slowly than recoverable infectious virus and remains present in lymphoid tissue for at least 6 months after infection. Persistence of viral RNA and protein suggests persistent low-level replication in lymphoid tissue that may facilitate maturation of the immune response, resulting in lifelong protection from reinfection, while persistence in other tissues (for example, the nervous system) may predispose to development of late disease such as subacute sclerosing panencephalitis. Further studies are needed to identify mechanisms of viral clearance and to understand the relationship between persistence and development of lifelong immunity.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Wen-Hsuan W Lin
- Department of Pathology, Columbia University School of Medicine, New York, NY, 10032, USA
| | - Ashley N Nelson
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| |
Collapse
|
16
|
Pfeffermann K, Dörr M, Zirkel F, von Messling V. Morbillivirus Pathogenesis and Virus-Host Interactions. Adv Virus Res 2018; 100:75-98. [PMID: 29551144 DOI: 10.1016/bs.aivir.2017.12.003] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Despite the availability of safe and effective vaccines against measles and several animal morbilliviruses, they continue to cause regular outbreaks and epidemics in susceptible populations. Morbilliviruses are highly contagious and share a similar pathogenesis in their respective hosts. This review provides an overview of morbillivirus history and the general replication cycle and recapitulates Morbillivirus pathogenesis focusing on common and unique aspects seen in different hosts. It also summarizes the state of knowledge regarding virus-host interactions on the cellular level with an emphasis on viral interference with innate immune response activation, and highlights remaining knowledge gaps.
Collapse
|
17
|
Cytomegalovirus Virions Shed in Urine Have a Reversible Block to Epithelial Cell Entry and Are Highly Resistant to Antibody Neutralization. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2017; 24:CVI.00024-17. [PMID: 28404573 DOI: 10.1128/cvi.00024-17] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Accepted: 04/04/2017] [Indexed: 12/31/2022]
Abstract
Cytomegalovirus (CMV) causes sensorineural hearing loss and developmental disabilities in newborns when infections are acquired in utero Pregnant women may acquire CMV from oral exposure to CMV in urine or saliva from young children. Neutralizing antibodies in maternal saliva have the potential to prevent maternal infection and, in turn, fetal infection. As CMV uses different viral glycoprotein complexes to enter different cell types, the first cells to be infected in the oral cavity could determine the type of antibodies needed to disrupt oral transmission. Antibodies targeting the pentameric complex (PC) should block CMV entry into epithelial cells but not into fibroblasts or Langerhans cells (which do not require the PC for entry), while antibodies targeting glycoprotein complexes gB or gH/gL would be needed to block entry into fibroblasts, Langerhans cells, or other cell types. To assess the potential for antibodies to disrupt oral acquisition, CMV from culture-positive urine samples (uCMV) was used to study cell tropisms and sensitivity to antibody neutralization. uCMV entered epithelial cells poorly compared with the entry into fibroblasts. CMV-hyperimmune globulin or monoclonal antibodies targeting gB, gH/gL, or the PC were incapable of blocking the entry of uCMV into either fibroblasts or epithelial cells. Both phenotypes were lost after one passage in cultured fibroblasts, suggestive of a nongenetic mechanism. These results suggest that uCMV virions have a reversible block to epithelial cell entry. Antibodies may be ineffective in preventing maternal oral CMV acquisition but may limit viral spread in blood or tissues, thereby reducing or preventing fetal infection and disease.
Collapse
|
18
|
El-Awady AR, Arce RM, Cutler CW. Dendritic cells: microbial clearance via autophagy and potential immunobiological consequences for periodontal disease. Periodontol 2000 2017; 69:160-80. [PMID: 26252408 PMCID: PMC4530502 DOI: 10.1111/prd.12096] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2015] [Indexed: 12/15/2022]
Abstract
Dendritic cells are potent antigen‐capture and antigen‐presenting cells that play a key role in the initiation and regulation of the adaptive immune response. This process of immune homeostasis, as maintained by dendritic cells, is susceptible to dysregulation by certain pathogens during chronic infections. Such dysregulation may lead to disease perpetuation with potentially severe systemic consequences. Here we discuss in detail how intracellular pathogens exploit dendritic cells and escape degradation by altering or evading autophagy. This novel mechanism explains, in part, the chronic, persistent nature observed in several immuno‐inflammatory diseases, including periodontal disease. We also propose a hypothetical model of the plausible role of autophagy in the context of periodontal disease. Promotion of autophagy may open new therapeutic strategies in the search of a ‘cure’ for periodontal disease in humans.
Collapse
|
19
|
|
20
|
Griffin DE. The Immune Response in Measles: Virus Control, Clearance and Protective Immunity. Viruses 2016; 8:v8100282. [PMID: 27754341 PMCID: PMC5086614 DOI: 10.3390/v8100282] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 12/25/2022] Open
Abstract
Measles is an acute systemic viral infection with immune system interactions that play essential roles in multiple stages of infection and disease. Measles virus (MeV) infection does not induce type 1 interferons, but leads to production of cytokines and chemokines associated with nuclear factor kappa-light-chain-enhancer of activated B cells (NFκB) signaling and activation of the NACHT, LRR and PYD domains-containing protein (NLRP3) inflammasome. This restricted response allows extensive virus replication and spread during a clinically silent latent period of 10–14 days. The first appearance of the disease is a 2–3 day prodrome of fever, runny nose, cough, and conjunctivitis that is followed by a characteristic maculopapular rash that spreads from the face and trunk to the extremities. The rash is a manifestation of the MeV-specific type 1 CD4+ and CD8+ T cell adaptive immune response with lymphocyte infiltration into tissue sites of MeV replication and coincides with clearance of infectious virus. However, clearance of viral RNA from blood and tissues occurs over weeks to months after resolution of the rash and is associated with a period of immunosuppression. However, during viral RNA clearance, MeV-specific antibody also matures in type and avidity and T cell functions evolve from type 1 to type 2 and 17 responses that promote B cell development. Recovery is associated with sustained levels of neutralizing antibody and life-long protective immunity.
Collapse
Affiliation(s)
- Diane E Griffin
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA.
| |
Collapse
|
21
|
Laksono BM, de Vries RD, McQuaid S, Duprex WP, de Swart RL. Measles Virus Host Invasion and Pathogenesis. Viruses 2016; 8:E210. [PMID: 27483301 PMCID: PMC4997572 DOI: 10.3390/v8080210] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/20/2016] [Accepted: 07/21/2016] [Indexed: 01/05/2023] Open
Abstract
Measles virus is a highly contagious negative strand RNA virus that is transmitted via the respiratory route and causes systemic disease in previously unexposed humans and non-human primates. Measles is characterised by fever and skin rash and usually associated with cough, coryza and conjunctivitis. A hallmark of measles is the transient immune suppression, leading to increased susceptibility to opportunistic infections. At the same time, the disease is paradoxically associated with induction of a robust virus-specific immune response, resulting in lifelong immunity to measles. Identification of CD150 and nectin-4 as cellular receptors for measles virus has led to new perspectives on tropism and pathogenesis. In vivo studies in non-human primates have shown that the virus initially infects CD150⁺ lymphocytes and dendritic cells, both in circulation and in lymphoid tissues, followed by virus transmission to nectin-4 expressing epithelial cells. The abilities of the virus to cause systemic infection, to transmit to numerous new hosts via droplets or aerosols and to suppress the host immune response for several months or even years after infection make measles a remarkable disease. This review briefly highlights current topics in studies of measles virus host invasion and pathogenesis.
Collapse
Affiliation(s)
- Brigitta M Laksono
- Department of Viroscience, Erasmus MC, 3015CN Rotterdam, The Netherlands.
| | - Rory D de Vries
- Department of Viroscience, Erasmus MC, 3015CN Rotterdam, The Netherlands.
| | - Stephen McQuaid
- Centre for Cancer Research and Cell Biology, Queen's University of Belfast, BT7 1NN Belfast, UK.
| | - W Paul Duprex
- Department of Microbiology, Boston University School of Medicine, Boston, MA 02118, USA.
| | - Rik L de Swart
- Department of Viroscience, Erasmus MC, 3015CN Rotterdam, The Netherlands.
| |
Collapse
|
22
|
Loke I, Kolarich D, Packer NH, Thaysen-Andersen M. Emerging roles of protein mannosylation in inflammation and infection. Mol Aspects Med 2016; 51:31-55. [PMID: 27086127 DOI: 10.1016/j.mam.2016.04.004] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 04/05/2016] [Accepted: 04/10/2016] [Indexed: 02/07/2023]
Abstract
Proteins are frequently modified by complex carbohydrates (glycans) that play central roles in maintaining the structural and functional integrity of cells and tissues in humans and lower organisms. Mannose forms an essential building block of protein glycosylation, and its functional involvement as components of larger and diverse α-mannosidic glycoepitopes in important intra- and intercellular glycoimmunological processes is gaining recognition. With a focus on the mannose-rich asparagine (N-linked) glycosylation type, this review summarises the increasing volume of literature covering human and non-human protein mannosylation, including their structures, biosynthesis and spatiotemporal expression. The review also covers their known interactions with specialised host and microbial mannose-recognising C-type lectin receptors (mrCLRs) and antibodies (mrAbs) during inflammation and pathogen infection. Advances in molecular mapping technologies have recently revealed novel immuno-centric mannose-terminating truncated N-glycans, termed paucimannosylation, on human proteins. The cellular presentation of α-mannosidic glycoepitopes on N-glycoproteins appears tightly regulated; α-mannose determinants are relative rare glycoepitopes in physiological extracellular environments, but may be actively secreted or leaked from cells to transmit potent signals when required. Simultaneously, our understanding of the molecular basis on the recognition of mannosidic epitopes by mrCLRs including DC-SIGN, mannose receptor, mannose binding lectin and mrAb is rapidly advancing, together with the functional implications of these interactions in facilitating an effective immune response during physiological and pathophysiological conditions. Ultimately, deciphering these complex mannose-based receptor-ligand interactions at the detailed molecular level will significantly advance our understanding of immunological disorders and infectious diseases, promoting the development of future therapeutics to improve patient clinical outcomes.
Collapse
Affiliation(s)
- Ian Loke
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Daniel Kolarich
- Department of Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, 14424 Potsdam, Germany
| | - Nicolle H Packer
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Morten Thaysen-Andersen
- Department of Chemistry and Biomolecular Sciences, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
23
|
Yan X, Li W, Pan L, Fu E, Xie Y, Chen M, Mu D. Lewis Lung Cancer Cells Promote SIGNR1(CD209b)-Mediated Macrophages Polarization Induced by IL-4 to Facilitate Immune Evasion. J Cell Biochem 2015; 117:1158-66. [PMID: 26447454 DOI: 10.1002/jcb.25399] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 10/06/2015] [Indexed: 01/16/2023]
Abstract
Tumor-associated macrophages are a prominent component of lung cancer and contribute to tumor progression by facilitating the immune evasion of cancer cells. DC-SIGN (CD209) assists in the immune evasion of a broad spectrum of pathogens and neoplasms by inhibiting the maturation of DCs and subsequent cytokines production. However, the expression of DC-SIGN in macrophages and its role in mediating immune evasion in lung cancer and the underlying mechanism remain unclear. Our study aimed to identify the immunosuppressive role of SIGNR1 in murine macrophage differentiation and lung cancer progression. We found that SIGNR1-positive RAW264.7 macrophages were enriched in mixed cultures with Lewis lung cancer cells (LLC) (ratio of RAW 264.7 to LLC being 1:1) after stimulation with IL-4. Moreover, LLC-educated macrophages exhibited significantly higher levels of IL-10 but lower IL-12 in response to IL-4 treatment as determined by RT-PCR and ELISA. However, inhibition of SIGNR1 markedly hampered the production of IL-10, indicating that SIGNR1 was indispensable for IL-4+LLC induced macrophage polarization towards the M2 subtype. Furthermore, polarized M2 cells immersed in a tumor microenvironment promoted the migration of LLCs, as measured by transwell assays, but migration was suppressed after blockade of SIGNR1 using CD209b antibody. In addition, IL-4+LLC-educated macrophages reduced the proliferation of the activated T cells and reduced IFN-γ-mediated Th1 response in T cells, while SIGNR1 inhibition rescued Th1 cell functions. In conclusion, murine SIGNR1 expressed in LLC-educated macrophages appears to mediate IL-4-induced RAW264.7 macrophage polarization and thus facilitate lung cancer evasion.
Collapse
Affiliation(s)
- Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Wenhai Li
- Department of Thoracic Surgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Lei Pan
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Enqing Fu
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Yonghong Xie
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Min Chen
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| | - Deguang Mu
- Department of Respiration Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710038, Shaanxi Province, China
| |
Collapse
|
24
|
Melia MM, Earle JP, Abdullah H, Reaney K, Tangy F, Cosby SL. Use of SLAM and PVRL4 and identification of pro-HB-EGF as cell entry receptors for wild type phocine distemper virus. PLoS One 2014; 9:e106281. [PMID: 25171206 PMCID: PMC4149546 DOI: 10.1371/journal.pone.0106281] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 07/31/2014] [Indexed: 12/13/2022] Open
Abstract
Signalling lymphocyte activation molecule (SLAM) has been identified as an immune cell receptor for the morbilliviruses, measles (MV), canine distemper (CDV), rinderpest and peste des petits ruminants (PPRV) viruses, while CD46 is a receptor for vaccine strains of MV. More recently poliovirus like receptor 4 (PVRL4), also known as nectin 4, has been identified as a receptor for MV, CDV and PPRV on the basolateral surface of polarised epithelial cells. PVRL4 is also up-regulated by MV in human brain endothelial cells. Utilisation of PVRL4 as a receptor by phocine distemper virus (PDV) remains to be demonstrated as well as confirmation of use of SLAM. We have observed that unlike wild type (wt) MV or wtCDV, wtPDV strains replicate in African green monkey kidney Vero cells without prior adaptation, suggesting the use of a further receptor. We therefore examined candidate molecules, glycosaminoglycans (GAG) and the tetraspan proteins, integrin β and the membrane bound form of heparin binding epithelial growth factor (proHB-EGF),for receptor usage by wtPDV in Vero cells. We show that wtPDV replicates in Chinese hamster ovary (CHO) cells expressing SLAM and PVRL4. Similar wtPDV titres are produced in Vero and VeroSLAM cells but more limited fusion occurs in the latter. Infection of Vero cells was not inhibited by anti-CD46 antibody. Removal/disruption of GAG decreased fusion but not the titre of virus. Treatment with anti-integrin β antibody increased rather than decreased infection of Vero cells by wtPDV. However, infection was inhibited by antibody to HB-EGF and the virus replicated in CHO-proHB-EGF cells, indicating use of this molecule as a receptor. Common use of SLAM and PVRL4 by morbilliviruses increases the possibility of cross-species infection. Lack of a requirement for wtPDV adaptation to Vero cells raises the possibility of usage of proHB-EGF as a receptor in vivo but requires further investigation.
Collapse
Affiliation(s)
- Mary M. Melia
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - John Philip Earle
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Haniah Abdullah
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Katherine Reaney
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| | - Frederic Tangy
- Viral Genomics and Vaccination Laboratory, Institut Pasteur, CNRS-URA3015, Paris, France
| | - Sara Louise Cosby
- Centre for Infection and Immunity, School of Medicine, Dentistry and Biomedical Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
25
|
Mesman AW, Zijlstra-Willems EM, Kaptein TM, de Swart RL, Davis ME, Ludlow M, Duprex WP, Gack MU, Gringhuis SI, Geijtenbeek TBH. Measles virus suppresses RIG-I-like receptor activation in dendritic cells via DC-SIGN-mediated inhibition of PP1 phosphatases. Cell Host Microbe 2014; 16:31-42. [PMID: 25011106 PMCID: PMC4159752 DOI: 10.1016/j.chom.2014.06.008] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/02/2014] [Accepted: 05/21/2014] [Indexed: 12/12/2022]
Abstract
Dendritic cells (DCs) are targets of measles virus (MV) and play central roles in viral dissemination. However, DCs express the RIG-I-like receptors (RLRs) RIG-I and Mda5 that sense MV and induce type I interferon (IFN) production. Given the potency of this antiviral response, RLRs are tightly regulated at various steps, including dephosphorylation by PP1 phosphatases, which induces their activation. We demonstrate that MV suppresses RIG-I and Mda5 by activating the C-type lectin DC-SIGN and inducing signaling that prevents RLR dephosphorylation. MV binding to DC-SIGN leads to activation of the kinase Raf-1, which induces the association of PP1 inhibitor I-1 with GADD34-PP1 holoenzymes, thereby inhibiting phosphatase activity. Consequently, GADD34-PP1 holoenzymes are unable to dephosphorylate RIG-I and Mda5, hence suppressing type I IFN responses and enhancing MV replication. Blocking DC-SIGN signaling allows RLR activation and suppresses MV infection of DCs. Thus, MV subverts DC-SIGN to control RLR activation and escape antiviral responses.
Collapse
Affiliation(s)
- Annelies W Mesman
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Esther M Zijlstra-Willems
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Tanja M Kaptein
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Rik L de Swart
- Department of Virology, Erasmus MC, 's-Gravendijkwal 230, 3015 CA Rotterdam, the Netherlands
| | - Meredith E Davis
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Martin Ludlow
- Department of Microbiology, Boston University, School of Medicine, Boston, MA 02118, USA
| | - W Paul Duprex
- Department of Microbiology, Boston University, School of Medicine, Boston, MA 02118, USA
| | - Michaela U Gack
- Department of Microbiology and Immunobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Sonja I Gringhuis
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Teunis B H Geijtenbeek
- Department of Experimental Immunology, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| |
Collapse
|
26
|
|
27
|
Miles B, Abdel-Ghaffar KA, Gamal AY, Baban B, Cutler CW. Blood dendritic cells: "canary in the coal mine" to predict chronic inflammatory disease? Front Microbiol 2014; 5:6. [PMID: 24478766 PMCID: PMC3902297 DOI: 10.3389/fmicb.2014.00006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2013] [Accepted: 01/07/2014] [Indexed: 12/19/2022] Open
Abstract
The majority of risk factors for chronic inflammatory diseases are unknown. This makes personalized medicine for assessment, prognosis, and choice of therapy very difficult. It is becoming increasingly clear, however, that low-grade subclinical infections may be an underlying cause of many chronic inflammatory diseases and thus may contribute to secondary outcomes (e.g., cancer). Many diseases are now categorized as inflammatory-mediated diseases that stem from a dysregulation in host immunity. There is a growing need to study the links between low-grade infections, the immune responses they elicit, and how this impacts overall health. One such link explored in detail here is the extreme sensitivity of myeloid dendritic cells (mDCs) in peripheral blood to chronic low-grade infections and the role that these mDCs play in arbitrating the resulting immune responses. We find that emerging evidence supports a role for pathogen-induced mDCs in chronic inflammation leading to increased risk of secondary clinical disease. The mDCs that are elevated in the blood as a result of low-grade bacteremia often do not trigger a productive immune response, but can disseminate the pathogen throughout the host. This aberrant trafficking of mDCs can accelerate systemic inflammatory disease progression. Conversely, restoration of dendritic cell homeostasis may aid in pathogen elimination and minimize dissemination. Thus it would seem prudent when assessing chronic inflammatory disease risk to consider blood mDC numbers, and the microbial content (microbiome) and activation state of these mDCs. These may provide important clues (“the canary in the coal mine”) of high inflammatory disease risk. This will facilitate development of novel immunotherapies to eliminate such smoldering infections in atherosclerosis, cancer, rheumatoid arthritis, and pre-eclampsia.
Collapse
Affiliation(s)
- Brodie Miles
- Department of Periodontics, College of Dental Medicine, Georgia Regents University Augusta, GA, USA
| | | | | | - Babak Baban
- Department of Oral Biology, Georgia Regents University Augusta, GA, USA
| | - Christopher W Cutler
- Department of Periodontics, College of Dental Medicine, Georgia Regents University Augusta, GA, USA
| |
Collapse
|
28
|
Hoving JC, Wilson GJ, Brown GD. Signalling C-type lectin receptors, microbial recognition and immunity. Cell Microbiol 2014; 16:185-94. [PMID: 24330199 PMCID: PMC4016756 DOI: 10.1111/cmi.12249] [Citation(s) in RCA: 176] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Revised: 12/06/2013] [Accepted: 12/09/2013] [Indexed: 12/16/2022]
Abstract
Signalling C‐type lectin receptors (CLRs) are crucial in shaping the immune response to fungal pathogens, but comparably little is known about the role of these receptors in bacterial, viral and parasitic infections. CLRs have many diverse functions depending on the signalling motifs in their cytoplasmic domains, and can induce endocytic, phagocytic, antimicrobial, pro‐inflammatory or anti‐inflammatory responses which are either protective or not during an infection. Understanding the role of CLRs in shaping anti‐microbial immunity offers great potential for the future development of therapeutics for disease intervention. In this review we will focus on the recognition of bacterial, viral and parasitic pathogens by CLRs, and how these receptors influence the outcome of infection. We will also provide a brief update on the role of CLRs in antifungal immunity.
Collapse
Affiliation(s)
- J Claire Hoving
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Observatory, Cape Town, South Africa
| | | | | |
Collapse
|
29
|
Paramyxovirus infections in ex vivo lung slice cultures of different host species. J Virol Methods 2013; 193:159-65. [DOI: 10.1016/j.jviromet.2013.06.016] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 05/31/2013] [Accepted: 06/08/2013] [Indexed: 11/18/2022]
|
30
|
Takaki H, Takeda M, Tahara M, Shingai M, Oshiumi H, Matsumoto M, Seya T. The MyD88 pathway in plasmacytoid and CD4+ dendritic cells primarily triggers type I IFN production against measles virus in a mouse infection model. THE JOURNAL OF IMMUNOLOGY 2013; 191:4740-7. [PMID: 24078691 DOI: 10.4049/jimmunol.1301744] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Infection by measles virus (MV) induces type I IFN via the retinoic acid-inducible gene I/melanoma differentiation-associated gene 5/mitochondrial antiviral signaling protein (MAVS) pathway in human cells. However, the in vivo role of the MAVS pathway in host defense against MV infection remains undetermined. CD150 transgenic (Tg) mice, which express human CD150, an entry receptor for MV, with the disrupting IFNR gene (Ifnar(-/-)), are susceptible to MV and serve as a model for MV infection. In this study, we generated CD150Tg/Mavs(-/-) mice and examined MV permissiveness compared with that in CD150Tg/Ifnar(-/-) mice. MV replicated mostly in the spleen of i.p.-infected CD150Tg/Ifnar(-/-) mice. Strikingly, CD150Tg/Mavs(-/-) mice were not permissive to MV in vivo because of substantial type I IFN induction. MV barely replicated in any other organs tested. When T cells, B cells, and dendritic cells (DCs) isolated from CD150Tg/Mavs(-/-) splenocytes were cultured with MV in vitro, only the DCs produced type I IFN. In vitro infection analysis using CD150Tg/Mavs(-/-) DC subsets revealed that CD4(+) and plasmacytoid DCs, but not CD8α(+) and CD8α(-)CD4(-) double negative DCs, were exclusively involved in type I IFN production in response to MV infection. Because CD150Tg/Mavs(-/-) mice turned permissive to MV by anti-IFNAR Ab, type I IFN produced by CD4(+) DCs and plasmacytoid DCs plays a critical role in antiviral protection for neighboring cells expressing IFNAR. Induction of type I IFN in these DC subsets was abolished by the MyD88 inhibitory peptide. Thus, production of type I IFN occurs via the MyD88-dependent and MAVS-independent signaling pathway during MV infection.
Collapse
Affiliation(s)
- Hiromi Takaki
- Department of Microbiology and Immunology, Graduate School of Medicine, Hokkaido University, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | | | | | |
Collapse
|
31
|
Measles virus infection of epithelial cells in the macaque upper respiratory tract is mediated by subepithelial immune cells. J Virol 2013; 87:4033-42. [PMID: 23365435 DOI: 10.1128/jvi.03258-12] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Measles virus (MV), one of the most contagious viruses infecting humans, causes a systemic infection leading to fever, immune suppression, and a characteristic maculopapular rash. However, the specific mechanism or mechanisms responsible for the spread of MV into the respiratory epithelium in the late stages of the disease are unknown. Here we show the crucial role of PVRL4 in mediating the spread of MV from immune to epithelial cells by generating a PVRL4 "blind" recombinant wild-type MV and developing a novel in vitro coculture model of B cells with primary differentiated normal human bronchial epithelial cells. We utilized the macaque model of measles to analyze virus distribution in the respiratory tract prior to and at the peak of MV replication. Expression of PVRL4 was widespread in both the lower and upper respiratory tract (URT) of macaques, indicating MV transmission can be facilitated by more than only epithelial cells of the trachea. Analysis of tissues collected at early time points after experimental MV infection demonstrated the presence of MV-infected lymphoid and myeloid cells contacting respiratory tract epithelium in the absence of infected epithelial cells, suggesting that these immune cells seed the infection in vivo. Thereafter, lateral cell-to-cell spread of MV led to the formation of large foci of infected cells in the trachea and high levels of MV infection in the URT, particularly in the nasal cavity. These novel findings have important implications for our understanding of the high transmissibility of measles.
Collapse
|