1
|
Zheng Y, Feng J, Ling M, Yu Y, Tao Y, Wang X. A comprehensive review on targeting cluster of differentiation: An attractive strategy for inhibiting viruses through host proteins. Int J Biol Macromol 2024; 269:132200. [PMID: 38723834 DOI: 10.1016/j.ijbiomac.2024.132200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/20/2024] [Accepted: 05/06/2024] [Indexed: 05/13/2024]
Abstract
Viral infections continue to pose a significant global public health threat. Targeting host proteins, such as cluster of differentiation (CD) macromolecules, may offer a promising alternative approach to developing antiviral treatments. CDs are cell-surface biological macromolecules mainly expressed on leukocytes that viruses can use to enter cells, thereby evading immune detection and promoting their replication. The manipulation of CDs by viruses may represent an effective and clever means of survival through the prolonged co-evolution of hosts and viruses. Targeting of CDs is anticipated to hinder the invasion of related viruses, modulate the body's immune system, and diminish the incidence of subsequent inflammation. They have become crucial for biomedical diagnosis, and some have been used as valuable tools for resisting viral infections. However, a summary of the structures and functions of CDs involved in viral infection is currently lacking. The development of drugs targeting these biological macromolecules is restricted both in terms of their availability and the number of compounds currently identified. This review provides a comprehensive analysis of the critical role of CD proteins in virus invasion and a list of relevant targeted antiviral agents, which will serve as a valuable reference for future research in this field.
Collapse
Affiliation(s)
- Youle Zheng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Jin Feng
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Min Ling
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yixin Yu
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Yanfei Tao
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China
| | - Xu Wang
- National Reference Laboratory of Veterinary Drug Residues (HZAU), MAO Key Laboratory for Detection of Veterinary Drug Residues, Huazhong Agricultural University, Wuhan, Hubei 430070, China; MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products, Huazhong Agricultural University, Wuhan, Hubei 430070, China.
| |
Collapse
|
2
|
Fan X, Kannan Villalan A, Hu Y, Wu X, Wang H, Wang X. Prediction of the Potential Host of Peste Des Petits Ruminants Virus by the Least Common Amino Acid Pattern in SLAM Receptor. Transbound Emerg Dis 2024; 2024:4374388. [PMID: 40303034 PMCID: PMC12017033 DOI: 10.1155/2024/4374388] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 03/17/2024] [Accepted: 03/28/2024] [Indexed: 05/02/2025]
Abstract
Peste-des-Petits Ruminants Virus (PPRV) causes a highly contagious and severe infectious disease known as Peste-des-Petits Ruminants (PPR), resulting in significant mortality in both domestic and wild ruminants. An in-depth understanding of the molecular relationship between PPRV and susceptible hosts is essential for the prevention of PPR. The signaling lymphocytic-activation molecule (SLAM) acts as a key receptor in susceptible host species, mediating interactions with PPRV and triggering PPR in ruminants. This study offers an in-depth analysis of PPRV-susceptible host species as well as the identified SLAM amino acid sequences to date. Investigation reveals that nine families-Bovidae, Camelidae, Cervidae, Elephantidae, Suidae, Felidae, Canidae, Muridae, and Ceratopogonidae-have been affected by PPRV infection. Furthermore, a bioinformatics-based approach was proposed to screen the least common amino acid patterns (LCAP) in important SLAM receptor regions of known PPRV-susceptible species. Research findings reveal that 14 least common amino acid sites (LCAS) in SLAM amino acid sequences (I61, I63, S60, S70, K76, K78, I79, S81, L82, E123, N125, S127, V128, and F131) exhibit a prevalent similarity to LCAP across all known susceptible species. Comparative analysis of these 14 LCAP with SLAM nucleotide sequences from unknown susceptible ruminants to identify species at heightened risk of PPRV. In the result, 48 species from 20 different families across six orders were at potential risk of being infected with PPRV. This exploration suggests the feasibility of assessing potential hosts at high risk of PPRV infection through the LCAS screening technique. Moreover, it offers a means to anticipate and issue warnings regarding the likelihood of interspecies transmission. In conclusion, this study integrates molecular biology and bioinformatics, shedding light on PPRV infection dynamics and paving the way for predictive strategies to prevent the spread of this devastating disease among ruminant populations.
Collapse
Affiliation(s)
- Xin Fan
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang Province, China
- Key Laboratory for Wildlife Diseases and Bio-Security Management of Heilongjiang Province, Harbin 150040, Heilongjiang Province, China
| | - Arivizhivendhan Kannan Villalan
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang Province, China
- Key Laboratory for Wildlife Diseases and Bio-Security Management of Heilongjiang Province, Harbin 150040, Heilongjiang Province, China
| | - YeZhi Hu
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang Province, China
- Key Laboratory for Wildlife Diseases and Bio-Security Management of Heilongjiang Province, Harbin 150040, Heilongjiang Province, China
| | - XiaoDong Wu
- China Animal Health and Epidemiology Center, Qingdao 266032, Shandong Province, China
| | - HaoNing Wang
- School of Geography and Tourism, Harbin University, Harbin 150086, Heilongjiang Province, China
| | - XiaoLong Wang
- College of Wildlife and Protected Area, Northeast Forestry University, Harbin 150040, Heilongjiang Province, China
- Key Laboratory for Wildlife Diseases and Bio-Security Management of Heilongjiang Province, Harbin 150040, Heilongjiang Province, China
| |
Collapse
|
3
|
Sakamoto K, Konami M, Kameda S, Satoh Y, Wakimoto H, Kitagawa Y, Gotoh B, Jiang DP, Hotta H, Itoh M. Suppression of viral RNA polymerase activity is necessary for persistent infection during the transformation of measles virus into SSPE virus. PLoS Pathog 2023; 19:e1011528. [PMID: 37494386 PMCID: PMC10406308 DOI: 10.1371/journal.ppat.1011528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 08/07/2023] [Accepted: 07/03/2023] [Indexed: 07/28/2023] Open
Abstract
Subacute sclerosing panencephalitis (SSPE) is a fatal neurodegenerative disease caused by measles virus (MV), which typically develops 7 to 10 years after acute measles. During the incubation period, MV establishes a persistent infection in the brain and accumulates mutations that generate neuropathogenic SSPE virus. The neuropathogenicity is closely associated with enhanced propagation mediated by cell-to-cell fusion in the brain, which is principally regulated by hyperfusogenic mutations of the viral F protein. The molecular mechanisms underlying establishment and maintenance of persistent infection are unclear because it is impractical to isolate viruses before the appearance of clinical signs. In this study, we found that the L and P proteins, components of viral RNA-dependent RNA polymerase (RdRp), of an SSPE virus Kobe-1 strain did not promote but rather attenuated viral neuropathogenicity. Viral RdRp activity corresponded to F protein expression; the suppression of RdRp activity in the Kobe-1 strain because of mutations in the L and P proteins led to restriction of the F protein level, thereby reducing cell-to-cell fusion mediated propagation in neuronal cells and decreasing neuropathogenicity. Therefore, the L and P proteins of Kobe-1 did not contribute to progression of SSPE. Three mutations in the L protein strongly suppressed RdRp activity. Recombinant MV harboring the three mutations limited viral spread in neuronal cells while preventing the release of infectious progeny particles; these changes could support persistent infection by enabling host immune escape and preventing host cell lysis. Therefore, the suppression of RdRp activity is necessary for the persistent infection of the parental MV on the way to transform into Kobe-1 SSPE virus. Because mutations in the genome of an SSPE virus reflect the process of SSPE development, mutation analysis will provide insight into the mechanisms underlying persistent infection.
Collapse
Affiliation(s)
- Kento Sakamoto
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Miho Konami
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Shinra Kameda
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Yuto Satoh
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Hiroshi Wakimoto
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Yoshinori Kitagawa
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Bin Gotoh
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Da-Peng Jiang
- Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Hak Hotta
- Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Masae Itoh
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| |
Collapse
|
4
|
Seki F, Takeda M. Novel and classical morbilliviruses: Current knowledge of three divergent morbillivirus groups. Microbiol Immunol 2022; 66:552-563. [PMID: 36151905 DOI: 10.1111/1348-0421.13030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 08/30/2022] [Accepted: 09/23/2022] [Indexed: 12/24/2022]
Abstract
Currently, seven species of morbillivirus have been classified. Six of these species (Measles morbillivirus, Rinderpest morbillivirus, Small ruminant morbillivirus, Canine morbillivirus, Phocine morbillivirus, and Cetacean morbillivirus) are highly infectious and cause serious systemic diseases in humans, livestock, domestic dogs, and wild animals. These species commonly use the host proteins signaling lymphocytic activation molecule (SLAM) and nectin-4 as receptors, and this usage contributes to their virulence. The seventh species (Feline morbillivirus: FeMV) is phylogenetically divergent from the six SLAM-using species. FeMV differs from the SLAM-using morbillivirus group in pathogenicity and infectivity, and is speculated to use non-SLAM receptors. Recently, novel species of morbilliviruses have been discovered in bats, rodents, and domestic pigs. Because the ability to use SLAM and nectin-4 is closely related to the infectivity and pathogenicity of morbilliviruses, investigation of the potential usage of these receptors is useful for estimating infectivity and pathogenicity. The SLAM-binding sites in the receptor-binding protein show high similarity among the SLAM-using morbilliviruses. This feature may help to estimate whether novel morbillivirus species can use SLAM as a receptor. A novel morbillivirus species isolated from wild mice diverged from the classified morbilliviruses in the phylogenetic tree, forming a third group separate from the SLAM-using morbillivirus group and FeMV. This suggests that the novel rodent morbillivirus may exhibit a different risk from the SLAM-using morbillivirus group, and analyses of its viral pathogenicity and infectivity toward humans are warranted.
Collapse
Affiliation(s)
- Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| | - Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Musashimurayama, Tokyo, Japan
| |
Collapse
|
5
|
Chen Y, Wang T, Yang Y, Fang Y, Zhao B, Zeng W, Lv D, Zhang L, Zhang Y, Xue Q, Chen X, Wang J, Qi X. Extracellular vesicles derived from PPRV-infected cells enhance signaling lymphocyte activation molecular (SLAM) receptor expression and facilitate virus infection. PLoS Pathog 2022; 18:e1010759. [PMID: 36084159 PMCID: PMC9491601 DOI: 10.1371/journal.ppat.1010759] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 09/21/2022] [Accepted: 07/22/2022] [Indexed: 11/18/2022] Open
Abstract
Peste des petits ruminants virus (PPRV) is an important pathogen that seriously influences the productivity of small ruminants worldwide. PPRV is lymphotropic in nature and SLAM was identified as the primary receptor for PPRV and other Morbilliviruses. Many viruses have been demonstrated to engage extracellular vesicles (EVs) to facilitate their replication and pathogenesis. Here, we provide evidence that PPRV infection significantly induced the secretion levels of EVs from goat PBMC, and that PPRV-H protein carried in EVs can enhance SLAM receptor expression in the recipient cells via suppressing miR-218, a negative miRNA directly targeting SLAM gene. Importantly, EVs-mediated increased SLAM expression enhances PPRV infectivity as well as the expression of various cytokines related to SLAM signaling pathway in the recipient cells. Moreover, our data reveal that PPRV associate EVs rapidly entry into the recipient cells mainly through macropinocytosis pathway and cooperated with caveolin- and clathrin-mediated endocytosis. Taken together, our findings identify a new strategy by PPRV to enhance virus infection and escape innate immunity by engaging EVs pathway. Peste des petitsruminants virus (PPRV) infection induces a transient but severe immunosuppression in the host, which threatens both small livestock and endangered susceptible wildlife populations in many countries. Despite extensive research, the mechanism underlying pathogenesis of PPRV infection remains elusive. Our data provide the first direct evidence that the EVs derived from PPRV-infected cells are involved in PPRV replication. In this study, the EVs derived from PPRV-infected goat PBMCs can enhance SLAM expression in the recipient cells, and more importantly, EVs-mediated increased SLAM expression enhances PPRV replication as well as the expression of various cytokines related to SLAM signaling pathway in the recipient cells. Taken together, our research has provided new insight into understanding the effect of EVs on PPRV replication and pathogenesis, and revealed a potential therapeutic target for antiviral intervention.
Collapse
Affiliation(s)
- Yan Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Ting Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yang Yang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yuan Fang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Bao Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- Shaanxi Animal Disease Control Center, Xi’an, China
| | - Wei Zeng
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Daiyue Lv
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Leyan Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Yanming Zhang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Qinghong Xue
- China Institute of Veterinary Drug Control, Beijing, China
| | - Xiwen Chen
- Animal Disease Prevention and Control & Healthy Breeding Engineering Technology Research Center, Mianyang Normal University, Mianyang, Sichuan, China
| | - Jingyu Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (JW); (XQ)
| | - Xuefeng Qi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
- * E-mail: (JW); (XQ)
| |
Collapse
|
6
|
Altamirano-Samaniego F, Enciso-Benavides J, Rojas N, Iglesias-Pedraz JM, Enciso N, Fossatti M, Enciso J. First report of canine morbillivirus infection of adipose tissue-derived stem cells from dogs with distemper. Vet World 2022; 15:1835-1842. [PMID: 36185532 PMCID: PMC9394121 DOI: 10.14202/vetworld.2022.1835-1842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 06/15/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Ribonucleic acid viruses remain latent in different cell types, including mesenchymal stem cells; however, the distemper virus remains undetected in these cells. This study aimed to determine whether adipose stem cells (ASCs) from dogs with distemper disease are infected with the canine morbillivirus (CM). Materials and Methods: Twelve dogs with the neurological phase of the disease and who were positive for CM by reverse transcription polymerase chain reaction (RT-PCR), were studied. ASCs from adipose tissue of the lesser omentum of these infected dogs were isolated and characterized. Direct fluorescence was used to detect the viral antigen in cell cultures. Flow cytometry and RT-PCR identified detectable quantities of the virus in two cultures, while electron microscopy confirmed the CM particles within ASCs. Results: This study revealed that ASCs of the omentum of dogs with distemper disease can be infected with CM, indicating their possible involvement in this virus latency and persistence. This suggests that its detection should be considered within the quality control process of stem cells intended for regenerative medicine. Conclusion: To the best of our knowledge, this is the first study that demonstrates that omentum ASCs from dogs with distemper disease can be infected with CM and may be involved in viral latency or persistence. Our study also suggests that the detection of CM should be considered within the quality control process of stem cells intended for regenerative medicine.
Collapse
Affiliation(s)
| | | | - Nancy Rojas
- Laboratorio de Microscopía Electrónica, Facultad de Medicina, Universidad Nacional Mayor de San Marcos, Lima, Perú
| | | | - Nathaly Enciso
- Grupo de Medicina Regenerativa, Universidad Científica del Sur, Lima, Perú
| | - Matia Fossatti
- Laboratorio de Bioquímica y Genética Molecular, Universidad Científica del Sur, Lima, Perú
| | - Javier Enciso
- Grupo de Medicina Regenerativa, Universidad Científica del Sur, Lima, Perú
| |
Collapse
|
7
|
Sakamoto K, Satoh Y, Takahashi KI, Wakimoto H, Kitagawa Y, Gotoh B, Ayata M, Itoh M. Upregulation of viral RNA polymerase activity promotes adaptation of SSPE virus to neuronal cells. Virology 2022; 573:1-11. [DOI: 10.1016/j.virol.2022.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 05/13/2022] [Accepted: 05/24/2022] [Indexed: 11/26/2022]
|
8
|
Ogawa H, Fujikura D, Namba H, Yamashita N, Honda T, Yamada M. Nectin-2 Acts as a Viral Entry Mediated Molecule That Binds to Human Herpesvirus 6B Glycoprotein B. Viruses 2022; 14:v14010160. [PMID: 35062364 PMCID: PMC8779676 DOI: 10.3390/v14010160] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 01/06/2022] [Accepted: 01/13/2022] [Indexed: 02/01/2023] Open
Abstract
Human herpesvirus 6B (HHV-6B) is a T-lymphotropic virus and the etiological agent of exanthem subitum. HHV-6B is present in a latent or persistent form after primary infection and is produced in the salivary glands or transmitted to this organ. Infected individuals continue to secrete the virus in their saliva, which is thus considered a source for virus transmission. HHV-6B primarily propagates in T cells because its entry receptor, CD134, is mainly expressed by activated T cells. The virus then spreads to the host’s organs, including the salivary glands, nervous system, and liver. However, CD134 expression is not detected in these organs. Therefore, HHV-6B may be entering cells via a currently unidentified cell surface molecule, but the mechanisms for this have not yet been investigated. In this study, we investigated a CD134-independent virus entry mechanism in the parotid-derived cell line HSY. First, we confirmed viral infection in CD134-membrane unanchored HSY cells. We then determined that nectin cell adhesion molecule 2 (nectin-2) mediated virus entry and that HHV-6B-insensitive T-cells transduced with nectin-2 were transformed into virus-permissive cells. We also found that virus entry was significantly reduced in nectin-2 knockout parotid-derived cells. Furthermore, we showed that HHV-6B glycoprotein B (gB) interacted with the nectin-2 V-set domain. The results suggest that nectin-2 acts as an HHV-6B entry-mediated protein.
Collapse
Affiliation(s)
- Hirohito Ogawa
- Department of Virology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (H.N.); (N.Y.); (T.H.)
- Correspondence: (H.O.); (M.Y.)
| | - Daisuke Fujikura
- School of Veterinary Medicine, Kitasato University, Higashi 23-35-1, Towada 034-8628, Japan;
| | - Hikaru Namba
- Department of Virology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (H.N.); (N.Y.); (T.H.)
| | - Nobuko Yamashita
- Department of Virology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (H.N.); (N.Y.); (T.H.)
| | - Tomoyuki Honda
- Department of Virology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (H.N.); (N.Y.); (T.H.)
| | - Masao Yamada
- Department of Virology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan; (H.N.); (N.Y.); (T.H.)
- Correspondence: (H.O.); (M.Y.)
| |
Collapse
|
9
|
Satoh Y, Higuchi K, Nishikawa D, Wakimoto H, Konami M, Sakamoto K, Kitagawa Y, Gotoh B, Jiang DP, Hotta H, Itoh M. M protein of subacute sclerosing panencephalitis virus, synergistically with the F protein, plays a crucial role in viral neuropathogenicity. J Gen Virol 2021; 102. [PMID: 34643483 PMCID: PMC8604190 DOI: 10.1099/jgv.0.001682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Subacute sclerosing panencephalitis (SSPE) is a rare fatal neurodegenerative disease caused by a measles virus (MV) variant, SSPE virus, that accumulates mutations during long-term persistent infection of the central nervous system (CNS). Clusters of mutations identified around the matrix (M) protein in many SSPE viruses suppress productive infectious particle release and accelerate cell–cell fusion, which are features of SSPE viruses. It was reported, however, that these defects of M protein function might not be correlated directly with promotion of neurovirulence, although they might enable establishment of persistent infection. Neuropathogenicity is closely related to the character of the viral fusion (F) protein, and amino acid substitution(s) in the F protein of some SSPE viruses confers F protein hyperfusogenicity, facilitating viral propagation in the CNS through cell–cell fusion and leading to neurovirulence. The F protein of an SSPE virus Kobe-1 strain, however, displayed only moderately enhanced fusion activity and required additional mutations in the M protein for neuropathogenicity in mice. We demonstrated here the mechanism for the M protein of the Kobe-1 strain supporting the fusion activity of the F protein and cooperatively inducing neurovirulence, even though each protein, independently, has no effect on virulence. The occurrence of SSPE has been estimated recently as one in several thousand in children who acquired measles under the age of 5 years, markedly higher than reported previously. The probability of a specific mutation (or mutations) occurring in the F protein conferring hyperfusogenicity and neuropathogenicity might not be sufficient to explain the high frequency of SSPE. The induction of neurovirulence by M protein synergistically with moderately fusogenic F protein could account for the high frequency of SSPE.
Collapse
Affiliation(s)
- Yuto Satoh
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Kurara Higuchi
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Daichi Nishikawa
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Hiroshi Wakimoto
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Miho Konami
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Kento Sakamoto
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
| | - Yoshinori Kitagawa
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Bin Gotoh
- Division of Microbiology and Infectious Diseases, Department of Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | - Da-Peng Jiang
- Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
| | - Hak Hotta
- Graduate School of Medicine, Kobe University, Kobe, Hyogo, Japan
- Present address: Faculty of Clinical Nutrition and Dietetics, Konan Women’s University, Kobe, Hyogo, Japan
| | - Masae Itoh
- Department of Microbiology, Faculty of Bio-Science, Nagahama Institute of Bio-Science and Technology, Nagahama, Shiga, Japan
- *Correspondence: Masae Itoh,
| |
Collapse
|
10
|
Yentür SP, Demirbilek V, Gurses C, Baris S, Kuru U, Ayta S, Yapici Z, Adin-Cinar S, Uysal S, Celik Yilmaz G, Onal E, Cokar O, Saruhan-Direskeneli G. Immune alterations in subacute sclerosing panencephalitis reflect an incompetent response to eliminate the measles virus. PLoS One 2021; 16:e0245077. [PMID: 33411786 PMCID: PMC7790413 DOI: 10.1371/journal.pone.0245077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 12/21/2020] [Indexed: 11/19/2022] Open
Abstract
In subacute sclerosing panencephalitis (SSPE) the persistence of measles virus (MeV) may be related to the altered immune response. In this study, cytokine responses of lymphocytes and monocytes were evaluated in SSPE compared to controls with non-inflammatory (NICON) and inflammatory (ICON) diseases. Patients with SSPE (n = 120), 78 patients with ICON and 63 patients with NICON were included in this study. Phenotypes of peripheral blood mononuclear cells (PBMC) have been analyzed by flow cytometry. CD3 and CD28, and S. aureus Cowan strain I (SAC) stimulated and unstimulated cells were cultured and IL-2, IL-10, IFN-γ, IL-12p40, IL-12p70 and IL-23 were detected in supernatants by ELISA. MeV peptides were used for MeV-specific stimulation and IFN-γ secretion of PBMC was measured by ELISPOT. Spontaneous and stimulated secretions of IL-10 were lower in SSPE compared to both control groups. T cell stimulation induced lower IFN-γ production than ICON group, but higher IL-2 than NICON group in SSPE. Stimulated PBMC produced lower IL-12p70 in SSPE and had decreased CD46 on the cell surface, suggesting the interaction with the virus. IFN-γ responses against MeV peptides were not prominent and similar to NICON patients. The immune response did not reveal an inflammatory activity to eliminate the virus in SSPE patients. Even IL-10 production was diminished implicating that the response is self-limited in controlling the disease.
Collapse
Affiliation(s)
- Sibel P. Yentür
- Department of Physiology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Veysi Demirbilek
- Department of Neurology, Cerrahpasa Medical Faculty, Istanbul University Cerrahpasa, Istanbul, Turkey
| | - Candan Gurses
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Safa Baris
- Department of Pediatrics, Marmara Medical Faculty, Marmara University, Istanbul, Turkey
| | - Umit Kuru
- Department of Pediatrics, Bayrampasa State Hospital, Istanbul, Turkey
| | - Semih Ayta
- Department of Neurology, Haseki State Hospital, Istanbul, Turkey
| | - Zuhal Yapici
- Department of Neurology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Suzan Adin-Cinar
- Department of Immunology, Aziz Sancar Institute of Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Serap Uysal
- Department of Pediatrics, Cerrahpasa Medical Faculty, Istanbul University Cerrahpasa, Istanbul, Turkey
| | - Gulden Celik Yilmaz
- Department of Microbiology, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Emel Onal
- Department of Public Health, Istanbul Medical Faculty, Istanbul University, Istanbul, Turkey
| | - Ozlem Cokar
- Department of Neurology, Haseki State Hospital, Istanbul, Turkey
| | | |
Collapse
|
11
|
Mühlebach MD. Measles virus in cancer therapy. Curr Opin Virol 2020; 41:85-97. [PMID: 32861945 DOI: 10.1016/j.coviro.2020.07.016] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023]
Abstract
Over the last years, the development of viruses to treat cancer patients has re-gained considerable attention. A genetically modified herpesvirus, Talimogene laherparepvec, has already been authorized for the treatment of melanoma patients. Also recombinant measles virus (MeV) is developed as an oncolytic virus. Because of its high genetic flexibility, a number of different MeV strains have been the basis for the generation of targeted, armed, or shielded viruses that are highly specific for a given tumor target, more effective, or protected against serum neutralization. Such MeV have been extensively tested in vitro and in vivo, whereby remarkable oncolytic potency is accompanied by safety also in non-human primates. Therefore, MeV has been introduced into 19 different clinical trials and has reached phase II against two different tumor entities, multiple myeloma and ovarian carcinoma. Remarkably, one patient with advanced stage myeloma experienced long-term remission after treatment, visualizing the potency of this approach.
Collapse
Affiliation(s)
- Michael D Mühlebach
- Division of Veterinary Medicine, Paul-Ehrlich-Institut, Paul-Ehrlich-Strasse 51-59, D-63225 Langen, Germany.
| |
Collapse
|
12
|
Seki F, Yamamoto Y, Fukuhara H, Ohishi K, Maruyama T, Maenaka K, Tokiwa H, Takeda M. Measles Virus Hemagglutinin Protein Establishes a Specific Interaction With the Extreme N-Terminal Region of Human Signaling Lymphocytic Activation Molecule to Enhance Infection. Front Microbiol 2020; 11:1830. [PMID: 32922371 PMCID: PMC7457132 DOI: 10.3389/fmicb.2020.01830] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 07/13/2020] [Indexed: 11/26/2022] Open
Abstract
Measles virus (MV) is a human pathogen that is classified in the genus Morbillivirus in the family Paramyxoviridae together with several non-human animal morbilliviruses. They cause severe systemic infections by using signaling lymphocytic activation molecule (SLAM) and poliovirus receptor-like 4 expressed on immune and epithelial cells, respectively, as receptors. The viral hemagglutinin (H) protein is responsible for the receptor-binding. Previously determined structures of MV-H and SLAM complexes revealed a major binding interface between the SLAM V domain and MV-H with four binding components (sites 1–4) in the interface. We studied the MV-H and human SLAM (hSLAM) complex structure in further detail by in silico analyses and determined missing regions or residues in the previously determined complex structures. These analyses showed that, in addition to sites 1–4, MV-H establishes a unique interaction with the extreme N-terminal region (ExNTR) of hSLAM. The first principles calculation-based fragment molecular orbital computation method revealed that methionine at position 29 (hSLAM-Met29) is the key residue for the interaction. hSLAM-Met29 was predicted to establish a CH-π interaction with phenylalanine at position 549 of MV-H (MVH-Phe549). A cell-cell fusion assay showed that the hSLAM-Met29 and MVH-Phe549 interaction is important for hSLAM-dependent MV membrane fusion. Furthermore, Jurkat cell lines expressing hSLAM with or without Met29 and recombinant MV possessing the H protein with or without Phe549 showed that the hSLAM-Met29 and MVH-Phe549 interaction enhanced hSLAM-dependent MV infection by ~10-fold. We speculate that in the evolutionary history of morbilliviruses, this interaction may have contributed to MV adaptation to humans because this interaction is unique for MV and only MV uses hSLAM efficiently among morbilliviruses.
Collapse
Affiliation(s)
- Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Tokyo, Japan
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Tokyo, Japan
| | - Hideo Fukuhara
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Kazue Ohishi
- Faculty of Engineering, Tokyo Polytechnic University, Atsugi, Japan
| | | | - Katsumi Maenaka
- Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Hiroaki Tokiwa
- Department of Chemistry, Rikkyo University, Tokyo, Japan
| | - Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Tokyo, Japan
| |
Collapse
|
13
|
Takeda M, Seki F, Yamamoto Y, Nao N, Tokiwa H. Animal morbilliviruses and their cross-species transmission potential. Curr Opin Virol 2020; 41:38-45. [PMID: 32344228 DOI: 10.1016/j.coviro.2020.03.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/21/2020] [Accepted: 03/23/2020] [Indexed: 02/01/2023]
Abstract
Like measles virus (MV), whose primary hosts are humans, non-human animal morbilliviruses use SLAM (signaling lymphocytic activation molecule) and PVRL4 (nectin-4) expressed on immune and epithelial cells, respectively, as receptors. PVRL4's amino acid sequence is highly conserved across species, while that of SLAM varies significantly. However, non-host animal SLAMs often function as receptors for different morbilliviruses. Uniquely, human SLAM is somewhat specific for MV, but canine distemper virus, which shows the widest host range among morbilliviruses, readily gains the ability to use human SLAM. The host range for morbilliviruses is also modulated by their ability to counteract the host's innate immunity, but the risk of cross-species transmission of non-human animal morbilliviruses to humans could occur if MV is successfully eradicated.
Collapse
Affiliation(s)
- Makoto Takeda
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan.
| | - Fumio Seki
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| | - Yuta Yamamoto
- Department of Chemistry, Rikkyo University, Nishi-Ikebukuro 3-34-1, Toshima-ku, Tokyo 171-8501, Japan
| | - Naganori Nao
- Department of Virology 3, National Institute of Infectious Diseases, Gakuen 4-7-1, Musashimurayama, Tokyo 208-0011, Japan
| | - Hiroaki Tokiwa
- Department of Chemistry, Rikkyo University, Nishi-Ikebukuro 3-34-1, Toshima-ku, Tokyo 171-8501, Japan
| |
Collapse
|
14
|
Dai X, Zhang X, Ostrikov K, Abrahamyan L. Host receptors: the key to establishing cells with broad viral tropism for vaccine production. Crit Rev Microbiol 2020; 46:147-168. [PMID: 32202955 PMCID: PMC7113910 DOI: 10.1080/1040841x.2020.1735992] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Cell culture-based vaccine technology is a flexible and convenient approach for vaccine production that requires adaptation of the vaccine strains to the new cells. Driven by the motivation to develop a broadly permissive cell line for infection with a wide range of viruses, we identified a set of the most relevant host receptors involved in viral attachment and entry. This identification was done through a review of different viral entry pathways and host cell lines, and in the context of the Baltimore classification of viruses. In addition, we indicated the potential technical problems and proposed some solutions regarding how to modify the host cell genome in order to meet industrial requirements for mass production of antiviral vaccines. Our work contributes to a finer understanding of the importance of breaking the host–virus recognition specificities for the possibility of creating a cell line feasible for the production of vaccines against a broad spectrum of viruses.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Xuanhao Zhang
- School of Biotechnology, Jiangnan University, Wuxi, China
| | - Kostya Ostrikov
- School of Chemistry and Physics and Institute for Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Australia
| | - Levon Abrahamyan
- Faculty of Veterinary Medicine, Swine and Poultry Infectious Diseases Research Center (CRIPA), Research Group on Infectious Diseases in Production Animals (GREMIP), Université de Montréal, Saint-Hyacinthe, Canada
| |
Collapse
|
15
|
meng X, Zhu X, Alfred N, Zhang Z. Identification of amino acid residues involved in the interaction between peste-des-petits-ruminants virus haemagglutinin protein and cellular receptors. J Gen Virol 2020; 101:242-251. [PMID: 31859612 PMCID: PMC7416607 DOI: 10.1099/jgv.0.001368] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 11/19/2019] [Indexed: 02/04/2023] Open
Abstract
Peste-des-petits-ruminants virus (PPRV) haemagglutinin (H) protein mediates binding to cellular receptors and then initiates virus entry. To identify the key residues of PPRV H (Hv) protein of the Nigeria 75/1 strain involved in binding to receptors, interaction of the Hv and mutated Hv (mHv) proteins with receptors (SLAM and Nectin 4) and their mutants (mSLAM1, mSLAM2, mSLAM3 and mNectin 4) was investigated using surface plasmon resonance imaging (SPRi) and coimmunoprecipitation (co-IP) assays. The results showed that the Hv protein failed to interact with mSLAM3, but interacted at a strong or medium intensity with SLAM, mSLAM2, Nectin 4 and mNectin 4, and at a low level with mSLAM1. The mHv protein was unable to interact with SLAM and its mutants, but bound to Nectin 4 and mNectin 4 with medium and weak intensity, respectively. Further analysis showed that the Hv protein could precipitate mSLAM1, mSLAM2 and mNectin 4, but not mSLAM3. The mHv protein failed to coprecipitate with SLAM and its mutants. The binding activities of mNectin 4 and Nectin 4 to mHv were less than 30.36 and 51.94 % of the wild-type levels, respectively. Based on the results obtained, amino acids at positions R389, L464, I498, R503, R533, Y541, Y543, F552 and Y553 of H protein and I61, H62, L64, K76, K78, E123, H130, I210, A211, S226 and R227 in SLAM were identified to be essential for the specificity of H-SLAM interaction, while the critical residues of H-Nectin 4 interaction require further study. These findings would improve our understanding of the invasive mechanisms of PPRV.
Collapse
Affiliation(s)
- Xuelian meng
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| | - Xueliang Zhu
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| | - Niyokwishimira Alfred
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| | - Zhidong Zhang
- State Key Laboratory of Veterinary Etiological Biology, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Xujiaping 1, Yanchangpu, Chengguan District, Lanzhou 730046, PR China
| |
Collapse
|
16
|
A structure-based rationale for sialic acid independent host-cell entry of Sosuga virus. Proc Natl Acad Sci U S A 2019; 116:21514-21520. [PMID: 31591233 PMCID: PMC6815108 DOI: 10.1073/pnas.1906717116] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The bat-borne paramyxovirus, Sosuga virus (SosV), is one of many paramyxoviruses recently identified and classified within the newly established genus Pararubulavirus, family Paramyxoviridae The envelope surface of SosV presents a receptor-binding protein (RBP), SosV-RBP, which facilitates host-cell attachment and entry. Unlike closely related hemagglutinin neuraminidase RBPs from other genera of the Paramyxoviridae, SosV-RBP and other pararubulavirus RBPs lack many of the stringently conserved residues required for sialic acid recognition and hydrolysis. We determined the crystal structure of the globular head region of SosV-RBP, revealing that while the glycoprotein presents a classical paramyxoviral six-bladed β-propeller fold and structurally classifies in close proximity to paramyxoviral RBPs with hemagglutinin-neuraminidase (HN) functionality, it presents a receptor-binding face incongruent with sialic acid recognition. Hemadsorption and neuraminidase activity analysis confirms the limited capacity of SosV-RBP to interact with sialic acid in vitro and indicates that SosV-RBP undergoes a nonclassical route of host-cell entry. The close overall structural conservation of SosV-RBP with other classical HN RBPs supports a model by which pararubulaviruses only recently diverged from sialic acid binding functionality.
Collapse
|
17
|
Host Cellular Receptors for the Peste des Petits Ruminant Virus. Viruses 2019; 11:v11080729. [PMID: 31398809 PMCID: PMC6723671 DOI: 10.3390/v11080729] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 08/05/2019] [Accepted: 08/06/2019] [Indexed: 12/17/2022] Open
Abstract
Peste des Petits Ruminant (PPR) is an important transboundary, OIE-listed contagious viral disease of primarily sheep and goats caused by the PPR virus (PPRV), which belongs to the genus Morbillivirus of the family Paramyxoviridae. The mortality rate is 90–100%, and the morbidity rate may reach up to 100%. PPR is considered economically important as it decreases the production and productivity of livestock. In many endemic poor countries, it has remained an obstacle to the development of sustainable agriculture. Hence, proper control measures have become a necessity to prevent its rapid spread across the world. For this, detailed information on the pathogenesis of the virus and the virus host interaction through cellular receptors needs to be understood clearly. Presently, two cellular receptors; signaling lymphocyte activation molecule (SLAM) and Nectin-4 are known for PPRV. However, extensive information on virus interactions with these receptors and their impact on host immune response is still required. Hence, a thorough understanding of PPRV receptors and the mechanism involved in the induction of immunosuppression is crucial for controlling PPR. In this review, we discuss PPRV cellular receptors, viral host interaction with cellular receptors, and immunosuppression induced by the virus with reference to other Morbilliviruses.
Collapse
|
18
|
Ohishi K, Maruyama T, Seki F, Takeda M. Marine Morbilliviruses: Diversity and Interaction with Signaling Lymphocyte Activation Molecules. Viruses 2019; 11:E606. [PMID: 31277275 PMCID: PMC6669707 DOI: 10.3390/v11070606] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 06/27/2019] [Accepted: 06/29/2019] [Indexed: 01/08/2023] Open
Abstract
Epidemiological reports of phocine distemper virus (PDV) and cetacean morbillivirus (CeMV) have accumulated since their discovery nearly 30 years ago. In this review, we focus on the interaction between these marine morbilliviruses and their major cellular receptor, the signaling lymphocyte activation molecule (SLAM). The three-dimensional crystal structure and homology models of SLAMs have demonstrated that 35 residues are important for binding to the morbillivirus hemagglutinin (H) protein and contribute to viral tropism. These 35 residues are essentially conserved among pinnipeds and highly conserved among the Caniformia, suggesting that PDV can infect these animals, but are less conserved among cetaceans. Because CeMV can infect various cetacean species, including toothed and baleen whales, the CeMV-H protein is postulated to have broader specificity to accommodate more divergent SLAM interfaces and may enable the virus to infect seals. In silico analysis of viral H protein and SLAM indicates that each residue of the H protein interacts with multiple residues of SLAM and vice versa. The integration of epidemiological, virological, structural, and computational studies should provide deeper insight into host specificity and switching of marine morbilliviruses.
Collapse
Affiliation(s)
- Kazue Ohishi
- Faculty of Engineering, Tokyo Polytechnic University, 1583, Iiyama, Atsugi, Kanagawa 243-0297, Japan.
| | - Tadashi Maruyama
- School of Marine Biosciences, Kitasato University, 1-15-1, Kitazato, Minami, Sagamihara, Kanagawa 252-0373, Japan
| | - Fumio Seki
- Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| | - Makoto Takeda
- Department of Virology III, National Institute of Infectious Diseases, 4-7-1, Gakuen, Musashimurayama, Tokyo 208-0011, Japan
| |
Collapse
|
19
|
Naaman H, Rabinski T, Yizhak A, Mizrahi S, Avni YS, Taube R, Rager B, Weinstein Y, Rall G, Gopas J, Ofir R. Measles Virus Persistent Infection of Human Induced Pluripotent Stem Cells. Cell Reprogram 2019; 20:17-26. [PMID: 29412740 DOI: 10.1089/cell.2017.0034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
In this study, we found that the measles virus (MV) can infect human-induced pluripotent stem cells (hiPSCs). Wild-type MV strains generally use human signaling lymphocyte activation molecule (SLAM; CD150) as a cellular receptor, while vaccine strains such as the Edmonston strain can use both CD150 and CD46 as receptors. It is not yet known how early in the embryonal differentiation stages these receptors are expressed. We established two hiPSCs (BGU-iPSCs and EMF-iPSCs) which express CD46 and CD150. Both cell types can be infected by MV to form persistent, noncytopathic cell lines that release infectious MV particles. Following MV persistent infection, BGU-iPSCs and EMF-iPSCs remain pluripotent and can differentiate in vitro into the three germ layers. This includes cells expressing the neuronal differentiation markers: NF68 and miRNA-124. Since the MV does not integrate into the cell's genome, it can be utilized as a vehicle to systematically introduce genes into iPSC, to dissect and to define factors regulating lineage differentiation.
Collapse
Affiliation(s)
- Hila Naaman
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel
| | - Tatiana Rabinski
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel .,2 Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev , Beer Sheva, Israel
| | - Avi Yizhak
- 3 Department of Surgery A, Soroka University Medical Center , Beer Sheva, Israel
| | - Solly Mizrahi
- 3 Department of Surgery A, Soroka University Medical Center , Beer Sheva, Israel
| | - Yonat Shemer Avni
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel .,4 Department of Clinical Virology, Soroka University Medical Center , Beer Sheva, Israel
| | - Ran Taube
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel
| | - Bracha Rager
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel
| | - Yacov Weinstein
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel
| | - Glenn Rall
- 5 Fox Chase Cancer Center , Blood Cell Development and Function, Philadelphia, Pennsylvania, USA
| | - Jacob Gopas
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel .,6 Department of Oncology, Soroka University Medical Center , Beer Sheva, Israel
| | - Rivka Ofir
- 1 The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev , Beer Sheva, Israel .,2 Regenerative Medicine & Stem Cell Research Center, Ben-Gurion University of the Negev , Beer Sheva, Israel
| |
Collapse
|
20
|
Rendon-Marin S, da Fontoura Budaszewski R, Canal CW, Ruiz-Saenz J. Tropism and molecular pathogenesis of canine distemper virus. Virol J 2019; 16:30. [PMID: 30845967 PMCID: PMC6407191 DOI: 10.1186/s12985-019-1136-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 02/25/2019] [Indexed: 11/10/2022] Open
Abstract
Background Canine distemper virus (CDV), currently termed Canine morbillivirus, is an extremely contagious disease that affects dogs. It is identified as a multiple cell tropism pathogen, and its host range includes a vast array of species. As a member of Mononegavirales, CDV has a negative, single-stranded RNA genome, which encodes eight proteins. Main body Regarding the molecular pathogenesis, the hemagglutinin protein (H) plays a crucial role both in the antigenic recognition and the viral interaction with SLAM and nectin-4, the host cells’ receptors. These cellular receptors have been studied widely as CDV receptors in vitro in different cellular models. The SLAM receptor is located in lymphoid cells; therefore, the infection of these cells by CDV leads to immunosuppression, the severity of which can lead to variability in the clinical disease with the potential of secondary bacterial infection, up to and including the development of neurological signs in its later stage. Conclusion Improving the understanding of the CDV molecules implicated in the determination of infection, especially the H protein, can help to enhance the biochemical comprehension of the difference between a wide range of CDV variants, their tropism, and different steps in viral infection. The regions of interaction between the viral proteins and the identified host cell receptors have been elucidated to facilitate this understanding. Hence, this review describes the significant molecular and cellular characteristics of CDV that contribute to viral pathogenesis. Electronic supplementary material The online version of this article (10.1186/s12985-019-1136-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Santiago Rendon-Marin
- Grupo de Investigación en Ciencias Animales - GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga, Colombia
| | - Renata da Fontoura Budaszewski
- Laboratório de Virologia, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Cláudio Wageck Canal
- Laboratório de Virologia, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Julian Ruiz-Saenz
- Grupo de Investigación en Ciencias Animales - GRICA, Facultad de Medicina Veterinaria y Zootecnia, Universidad Cooperativa de Colombia, Bucaramanga, Colombia.
| |
Collapse
|
21
|
Gordiienko I, Shlapatska L, Kovalevska L, Sidorenko SP. SLAMF1/CD150 in hematologic malignancies: Silent marker or active player? Clin Immunol 2018; 204:14-22. [PMID: 30616923 DOI: 10.1016/j.clim.2018.10.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 10/23/2018] [Accepted: 10/23/2018] [Indexed: 12/12/2022]
Abstract
SLAMF1/CD150 receptor is a founder of signaling lymphocyte activation molecule (SLAM) family of cell-surface receptors. It is widely expressed on cells within hematopoietic system. In hematologic malignancies CD150 cell surface expression is restricted to cutaneous T-cell lymphomas, few types of B-cell non-Hodgkin's lymphoma, near half of cases of chronic lymphocytic leukemia, Hodgkin's lymphoma, and multiple myeloma. Differential expression among various types of hematological malignancies allows considering CD150 as diagnostical and potential prognostic marker. Moreover, CD150 may be a target for antibody-based or measles virus oncolytic therapy. Due to CD150 signaling properties it is involved in regulation of malignant cell fate decision and tumor microenvironment in Hodgkin's lymphoma and chronic lymphocytic leukemia. This review summarizes evidence for the important role of CD150 in pathogenesis of hematologic malignancies.
Collapse
Affiliation(s)
- Inna Gordiienko
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine.
| | - Larysa Shlapatska
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Larysa Kovalevska
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| | - Svetlana P Sidorenko
- Department of Molecular and Cellular Pathobiology, R.E. Kavetsky Institute of Experimental Pathology, Oncology and Radiobiology National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
22
|
Antigen-specific oncolytic MV-based tumor vaccines through presentation of selected tumor-associated antigens on infected cells or virus-like particles. Sci Rep 2017; 7:16892. [PMID: 29203786 PMCID: PMC5715114 DOI: 10.1038/s41598-017-16928-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 11/19/2017] [Indexed: 12/24/2022] Open
Abstract
Recombinant vaccine strain-derived measles virus (MV) is clinically tested both as vaccine platform to protect against other pathogens and as oncolytic virus for tumor treatment. To investigate the potential synergism in anti-tumoral efficacy of oncolytic and vaccine properties, we chose Ovalbumin and an ideal tumor antigen, claudin-6, for pre-clinical proof of concept. To enhance immunogenicity, both antigens were presented by retroviral virus-like particle produced in situ during MV-infection. All recombinant MV revealed normal growths, genetic stability, and proper expression and presentation of both antigens. Potent antigen-specific humoral and cellular immunity were found in immunized MV-susceptible IFNAR-/--CD46Ge mice. These immune responses significantly inhibited metastasis formation or increased therapeutic efficacy compared to control MV in respective novel in vivo tumor models using syngeneic B16-hCD46/mCLDN6 murine melanoma cells. These data indicate the potential of MV to trigger selected tumor antigen-specific immune responses on top of direct tumor lysis for enhanced efficacy.
Collapse
|
23
|
Suzuki Y. Predicting receptor functionality of signaling lymphocyte activation molecule for measles virus hemagglutinin by docking simulation. Microbiol Immunol 2017; 61:185-189. [PMID: 28419512 PMCID: PMC7168510 DOI: 10.1111/1348-0421.12484] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2017] [Revised: 03/11/2017] [Accepted: 04/13/2017] [Indexed: 11/27/2022]
Abstract
Predicting susceptibility of various species to a virus assists assessment of risk of interspecies transmission. Evaluation of receptor functionality may be useful in screening for susceptibility. In this study, docking simulation was conducted for measles virus hemagglutinin (MV‐H) and immunoglobulin‐like variable domain of signaling lymphocyte activation molecule (SLAM‐V). It was observed that the docking scores for MV‐H and SLAM‐V correlated with the activity of SLAM as an MV receptor. These results suggest that the receptor functionality may be predicted from the docking scores of virion surface proteins and cellular receptor molecules.
Collapse
Affiliation(s)
- Yoshiyuki Suzuki
- Graduate School of Natural Sciences, Nagoya City University, 1 Yamanohata, Mizuho-cho, Mizuho-ku, Nagoya-shi, Aichi-ken 467-8501, Japan
| |
Collapse
|
24
|
Loots AK, Mitchell E, Dalton DL, Kotzé A, Venter EH. Advances in canine distemper virus pathogenesis research: a wildlife perspective. J Gen Virol 2017; 98:311-321. [PMID: 27902345 DOI: 10.1099/jgv.0.000666] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Canine distemper virus (CDV) has emerged as a significant disease of wildlife, which is highly contagious and readily transmitted between susceptible hosts. Initially described as an infectious disease of domestic dogs, it is now recognized as a global multi-host pathogen, infecting and causing mass mortalities in a wide range of carnivore species. The last decade has seen the effect of numerous CDV outbreaks in various wildlife populations. Prevention of CDV requires a clear understanding of the potential hosts in danger of infection as well as the dynamic pathways CDV uses to gain entry to its host cells and its ability to initiate viral shedding and disease transmission. We review recent research conducted on CDV infections in wildlife, including the latest findings on the causes of host specificity and cellular receptors involved in distemper pathogenesis.
Collapse
Affiliation(s)
- Angelika K Loots
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa.,National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa
| | - Emily Mitchell
- National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa
| | - Desiré L Dalton
- National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa.,Genetics Department, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Antoinette Kotzé
- National Zoological Gardens of South Africa, P.O. Box 754, Pretoria, 0001, South Africa.,Genetics Department, University of the Free State, P.O. Box 339, Bloemfontein 9300, South Africa
| | - Estelle H Venter
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| |
Collapse
|
25
|
Morbillivirus Experimental Animal Models: Measles Virus Pathogenesis Insights from Canine Distemper Virus. Viruses 2016; 8:v8100274. [PMID: 27727184 PMCID: PMC5086610 DOI: 10.3390/v8100274] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Revised: 09/29/2016] [Accepted: 09/30/2016] [Indexed: 12/19/2022] Open
Abstract
Morbilliviruses share considerable structural and functional similarities. Even though disease severity varies among the respective host species, the underlying pathogenesis and the clinical signs are comparable. Thus, insights gained with one morbillivirus often apply to the other members of the genus. Since the Canine distemper virus (CDV) causes severe and often lethal disease in dogs and ferrets, it is an attractive model to characterize morbillivirus pathogenesis mechanisms and to evaluate the efficacy of new prophylactic and therapeutic approaches. This review compares the cellular tropism, pathogenesis, mechanisms of persistence and immunosuppression of the Measles virus (MeV) and CDV. It then summarizes the contributions made by studies on the CDV in dogs and ferrets to our understanding of MeV pathogenesis and to vaccine and drugs development.
Collapse
|
26
|
Canine Distemper Virus Fusion Activation: Critical Role of Residue E123 of CD150/SLAM. J Virol 2015; 90:1622-37. [PMID: 26608324 DOI: 10.1128/jvi.02405-15] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/18/2015] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED Measles virus (MeV) and canine distemper virus (CDV) possess tetrameric attachment proteins (H) and trimeric fusion proteins, which cooperate with either SLAM or nectin 4 receptors to trigger membrane fusion for cell entry. While the MeV H-SLAM cocrystal structure revealed the binding interface, two distinct oligomeric H assemblies were also determined. In one of the conformations, two SLAM units were sandwiched between two discrete H head domains, thus spotlighting two binding interfaces ("front" and "back"). Here, we investigated the functional relevance of both interfaces in activating the CDV membrane fusion machinery. While alanine-scanning mutagenesis identified five critical regulatory residues in the front H-binding site of SLAM, the replacement of a conserved glutamate residue (E at position 123, replaced with A [E123A]) led to the most pronounced impact on fusion promotion. Intriguingly, while determination of the interaction of H with the receptor using soluble constructs revealed reduced binding for the identified SLAM mutants, no effect was recorded when physical interaction was investigated with the full-length counterparts of both molecules. Conversely, although mutagenesis of three strategically selected residues within the back H-binding site of SLAM did not substantially affect fusion triggering, nevertheless, the mutants weakened the H-SLAM interaction recorded with the membrane-anchored protein constructs. Collectively, our findings support a mode of binding between the attachment protein and the V domain of SLAM that is common to all morbilliviruses and suggest a major role of the SLAM residue E123, located at the front H-binding site, in triggering the fusion machinery. However, our data additionally support the hypothesis that other microdomain(s) of both glycoproteins (including the back H-binding site) might be required to achieve fully productive H-SLAM interactions. IMPORTANCE A complete understanding of the measles virus and canine distemper virus (CDV) cell entry molecular framework is still lacking, thus impeding the rational design of antivirals. Both viruses share many biological features that partially rely on the use of analogous Ig-like host cell receptors, namely, SLAM and nectin 4, for entering immune and epithelial cells, respectively. Here, we provide evidence that the mode of binding between the membrane-distal V domain of SLAM and the attachment protein (H) of morbilliviruses is very likely conserved. Moreover, although structural information revealed two discrete conformational states of H, one of the structures displayed two H-SLAM binding interfaces ("front" and "back"). Our data not only spotlight the front H-binding site of SLAM as the main determinant of membrane fusion promotion but suggest that the triggering efficiency of the viral entry machinery may rely on a local conformational change within the front H-SLAM interactive site rather than the binding affinity.
Collapse
|
27
|
Van Bressem MF, Duignan PJ, Banyard A, Barbieri M, Colegrove KM, De Guise S, Di Guardo G, Dobson A, Domingo M, Fauquier D, Fernandez A, Goldstein T, Grenfell B, Groch KR, Gulland F, Jensen BA, Jepson PD, Hall A, Kuiken T, Mazzariol S, Morris SE, Nielsen O, Raga JA, Rowles TK, Saliki J, Sierra E, Stephens N, Stone B, Tomo I, Wang J, Waltzek T, Wellehan JFX. Cetacean morbillivirus: current knowledge and future directions. Viruses 2014; 6:5145-81. [PMID: 25533660 PMCID: PMC4276946 DOI: 10.3390/v6125145] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2014] [Revised: 12/02/2014] [Accepted: 12/16/2014] [Indexed: 12/19/2022] Open
Abstract
We review the molecular and epidemiological characteristics of cetacean morbillivirus (CeMV) and the diagnosis and pathogenesis of associated disease, with six different strains detected in cetaceans worldwide. CeMV has caused epidemics with high mortality in odontocetes in Europe, the USA and Australia. It represents a distinct species within the Morbillivirus genus. Although most CeMV strains are phylogenetically closely related, recent data indicate that morbilliviruses recovered from Indo-Pacific bottlenose dolphins (Tursiops aduncus), from Western Australia, and a Guiana dolphin (Sotalia guianensis), from Brazil, are divergent. The signaling lymphocyte activation molecule (SLAM) cell receptor for CeMV has been characterized in cetaceans. It shares higher amino acid identity with the ruminant SLAM than with the receptors of carnivores or humans, reflecting the evolutionary history of these mammalian taxa. In Delphinidae, three amino acid substitutions may result in a higher affinity for the virus. Infection is diagnosed by histology, immunohistochemistry, virus isolation, RT-PCR, and serology. Classical CeMV-associated lesions include bronchointerstitial pneumonia, encephalitis, syncytia, and lymphoid depletion associated with immunosuppression. Cetaceans that survive the acute disease may develop fatal secondary infections and chronic encephalitis. Endemically infected, gregarious odontocetes probably serve as reservoirs and vectors. Transmission likely occurs through the inhalation of aerosolized virus but mother to fetus transmission was also reported.
Collapse
Affiliation(s)
- Marie-Françoise Van Bressem
- Cetacean Conservation Medicine Group (CMED), Peruvian Centre for Cetacean Research (CEPEC), Pucusana, Lima 20, Peru
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-30-53051397
| | - Pádraig J. Duignan
- Department of Ecosystem and Public Health, University of Calgary, Calgary, AL T2N 4Z6, Canada; E-Mail:
| | - Ashley Banyard
- Wildlife Zoonoses and Vector Borne Disease Research Group, Animal and Plant Health Agency (APHA), Weybridge, Surrey KT15 3NB, UK; E-Mail:
| | - Michelle Barbieri
- The Marine Mammal Centre, Sausalito, CA 94965, USA; E-Mails: (M.B.); (F.G.)
| | - Kathleen M Colegrove
- Zoological Pathology Program, College of Veterinary Medicine, University of Illinois at Maywood, IL 60153 , USA; E-Mail:
| | - Sylvain De Guise
- Department of Pathobiology and Veterinary Science, and Connecticut Sea Grant College Program, University of Connecticut, Storrs, CT 06269, USA; E-Mail:
| | - Giovanni Di Guardo
- Faculty of Veterinary Medicine, University of Teramo, 64100 Teramo, Italy; E-Mail:
| | - Andrew Dobson
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA; E-Mails: (A.D.); (B.G.); (S.E.M.)
| | - Mariano Domingo
- Centre de Recerca en Sanitat Animal (CReSA), Autonomous University of Barcelona, Bellaterra, Barcelona 08193, Spain; E-Mail:
| | - Deborah Fauquier
- National Marine Fisheries Service, Marine Mammal Health and Stranding Response Program, Silver Spring, MD 20910, USA; E-Mails: (D.F.); (T.K.R.)
| | - Antonio Fernandez
- Department of Veterinary Pathology, Institute of Animal Health, Veterinary School, Universidad de Las Palmas de Gran Canaria, Las Palmas 35413, Spain; E-Mails: (A.F.); (E.S.)
| | - Tracey Goldstein
- One Health Institute School of Veterinary Medicine University of California, Davis, CA 95616, USA; E-Mail:
| | - Bryan Grenfell
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA; E-Mails: (A.D.); (B.G.); (S.E.M.)
- Fogarty International Center, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kátia R. Groch
- Department of Pathology, School of Veterinary Medicine and Animal Sciences, University of São Paulo, São Paulo 05508-207, Brazil; E-Mail:
- Instituto Baleia Jubarte (Humpback Whale Institute), Caravelas, Bahia 45900-000, Brazil
| | - Frances Gulland
- The Marine Mammal Centre, Sausalito, CA 94965, USA; E-Mails: (M.B.); (F.G.)
- Marine Mammal Commission, 4340 East-West Highway, Bethesda, MD 20814, USA
| | - Brenda A Jensen
- Department of Natural Sciences, Hawai`i Pacific University, Kaneohe, HI 96744, USA; E-Mail:
| | - Paul D Jepson
- Institute of Zoology, Regent’s Park, London NW1 4RY, UK; E-Mail:
| | - Ailsa Hall
- Sea Mammal Research Unit, Scottish Oceans Institute, University of St. Andrews, St. Andrews KY16 8LB, UK; E-Mail:
| | - Thijs Kuiken
- Department of Viroscience, Erasmus MC, Rotterdam 3015 CN, The Netherlands; E-Mail:
| | - Sandro Mazzariol
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy; E-Mail:
| | - Sinead E Morris
- Department of Ecology and Evolutionary Biology, Princeton University, Princeton, NJ 08544, USA; E-Mails: (A.D.); (B.G.); (S.E.M.)
| | - Ole Nielsen
- Department of Fisheries and Oceans Canada, Central and Arctic Region, 501 University Crescent, Winnipeg, MB R3T 2N6 , Canada; E-Mail:
| | - Juan A Raga
- Marine Zoology Unit, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Valencia 22085, Spain; E-Mail:
| | - Teresa K Rowles
- National Marine Fisheries Service, Marine Mammal Health and Stranding Response Program, Silver Spring, MD 20910, USA; E-Mails: (D.F.); (T.K.R.)
| | - Jeremy Saliki
- Athens Veterinary Diagnostic Laboratory, College of Veterinary Medicine, University of Georgia, Athens, GA GA 30602 , USA; E-Mail:
| | - Eva Sierra
- Department of Veterinary Pathology, Institute of Animal Health, Veterinary School, Universidad de Las Palmas de Gran Canaria, Las Palmas 35413, Spain; E-Mails: (A.F.); (E.S.)
| | - Nahiid Stephens
- School of Veterinary and Life Sciences, Murdoch University, Perth 6150, Western Australia, Australia; E-Mail:
| | - Brett Stone
- QML Vetnostics, Metroplex on Gateway, Murarrie, Queensland 4172, Australia; E-Mail:
| | - Ikuko Tomo
- South Australian Museum, North Terrace, Adelaide 5000, South Australia, Australia; E-Mail:
| | - Jianning Wang
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), East Geelong, Victoria 3220, Australia; E-Mail:
| | - Thomas Waltzek
- Department of Infectious Diseases and Pathology, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; E-Mail:
| | - James FX Wellehan
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32611, USA; E-Mail:
| |
Collapse
|
28
|
Marty RR, Knuchel MC, Morin TNA, Naim HY. An immune competent mouse model for the characterization of recombinant measles vaccines. Hum Vaccin Immunother 2014; 11:83-90. [PMID: 25483519 DOI: 10.4161/hv.34358] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Today, immune compromised interferon-α-receptor deficient mice expressing hCD46 (IFNARCD46tg) are usually used for measles virus (MV) based vaccine characterization. However, for the development of MV-based recombinant vaccine candidates (rMV), an immune competent mouse model is desirable in order to induce and evaluate meaningful immune response. In this study, humoral and cellular immune response induced by rMV in immune competent mice expressing human MV receptor CD46 (hCD46tg) were compared with those induced in wild-type black/6, and IFNARCD46tg mice. All three strains developed humoral and cellular response against MV, whereas only hCD46tg and IFNARCD46tg mice developed a humoral response against the transgene. Differences were observed in the magnitude of the response, where the IFNARCD46tg mice displayed the strongest immune responses, followed by the hCD46tg mice and the black/6 mice. Interestingly, hCD46tg and wt black/6 mice showed a predominant CD4(+) T-cell response against MV-N, whereas IFNARCD46tg mice developed both, CD4(+) and CD8(+) T-cell response against MV-N. Analysis of the cytokine profile of MV-N specific CD4(+) T-cells and transgene (SIVgag) specific CD8(+) T-cells revealed qualitative differences of the T-cell responses; noticeably a significant reduction of the frequency of CD4(+)IL-2(+) expressing cells in IFNARCD46tg mice as compared with hCD46tg or wt black/6 mice. We show in this study significant quantitative and qualitative differences in immune responses between immune competent and immune-compromised mice. Our results therefore highlight the importance of the animal model and support the use of hCD46tg mice as mouse model for the characterization of the immunological profile induced by recombinant measles virus vaccine candidates.
Collapse
|
29
|
Carsillo T, Huey D, Levinsky A, Obojes K, Schneider-Schaulies J, Niewiesk S. Cotton rat (Sigmodon hispidus) signaling lymphocyte activation molecule (CD150) is an entry receptor for measles virus. PLoS One 2014; 9:e110120. [PMID: 25295727 PMCID: PMC4190324 DOI: 10.1371/journal.pone.0110120] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/16/2014] [Indexed: 11/18/2022] Open
Abstract
Cotton rats (Sigmodon hispidus) replicate measles virus (MV) after intranasal infection in the respiratory tract and lymphoid tissue. We have cloned the cotton rat signaling lymphocytic activation molecule (CD150, SLAM) in order to investigate its role as a potential receptor for MV. Cotton rat CD150 displays 58% and 78% amino acid homology with human and mouse CD150, respectively. By staining with a newly generated cotton rat CD150 specific monoclonal antibody expression of CD150 was confirmed in cotton rat lymphoid cells and in tissues with a pattern of expression similar to mouse and humans. Previously, binding of MV hemagglutinin has been shown to be dependent on amino acids 60, 61 and 63 in the V region of CD150. The human molecule contains isoleucine, histidine and valine at these positions and binds to MV-H whereas the mouse molecule contains valine, arginine and leucine and does not function as a receptor for MV. In the cotton rat molecule, amino acids 61 and 63 are identical with the mouse molecule and amino acid 60 with the human molecule. After transfection with cotton rat CD150 HEK 293 T cells became susceptible to infection with single cycle VSV pseudotype virus expressing wild type MV glycoproteins and with a MV wildtype virus. After infection, cells expressing cotton rat CD150 replicated virus to lower levels than cells expressing the human molecule and formed smaller plaques. These data might explain why the cotton rat is a semipermissive model for measles virus infection.
Collapse
Affiliation(s)
- Thomas Carsillo
- Department of Veterinary Biosciences and Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Devra Huey
- Department of Veterinary Biosciences and Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Amy Levinsky
- Department of Veterinary Biosciences and Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
| | - Karola Obojes
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | | - Stefan Niewiesk
- Department of Veterinary Biosciences and Center for Microbial Interface Biology, The Ohio State University, Columbus, Ohio, United States of America
- * E-mail:
| |
Collapse
|
30
|
Delpeut S, Noyce RS, Richardson CD. The tumor-associated marker, PVRL4 (nectin-4), is the epithelial receptor for morbilliviruses. Viruses 2014; 6:2268-86. [PMID: 24892636 PMCID: PMC4074928 DOI: 10.3390/v6062268] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Revised: 05/14/2014] [Accepted: 05/15/2014] [Indexed: 01/25/2023] Open
Abstract
PVRL4 (nectin-4) was recently identified as the epithelial receptor for members of the Morbillivirus genus, including measles virus, canine distemper virus and peste des petits ruminants virus. Here, we describe the role of PVRL4 in morbillivirus pathogenesis and its promising use in cancer therapies. This discovery establishes a new paradigm for the spread of virus from lymphocytes to airway epithelial cells and its subsequent release into the environment. Measles virus vaccine strains have emerged as a promising oncolytic platform for cancer therapy in the last ten years. Given that PVRL4 is a well-known tumor-associated marker for several adenocarcinoma (lung, breast and ovary), the measles virus could potentially be used to specifically target, infect and destroy cancers expressing PVRL4.
Collapse
Affiliation(s)
- Sebastien Delpeut
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| | - Ryan S Noyce
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| | - Christopher D Richardson
- The Department of Microbiology and Immunology, Dalhousie University, Halifax, B3H 1X5 NS, Canada.
| |
Collapse
|
31
|
Recent host range expansion of canine distemper virus and variation in its receptor, the signaling lymphocyte activation molecule, in carnivores. J Wildl Dis 2014; 50:596-606. [PMID: 24807184 DOI: 10.7589/2013-09-228] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The signaling lymphocyte activation molecule (SLAM) is a receptor for morbilliviruses. To understand the recent host range expansion of canine distemper virus (CDV) in carnivores, we determined the nucleotide sequences of SLAMs of various carnivores and generated three-dimensional homology SLAM models. Thirty-four amino acid residues were found for the candidates binding to CDV on the interface of the carnivore SLAMs. SLAM of the domestic dog (Canis lupus familiaris) were similar to those of other members of the suborder Caniformia, indicating that the animals in this group have similar sensitivity to dog CDV. However, they were different at nine positions from those of felids. Among the nine residues, four of domestic cat (Felis catus) SLAM (72, 76, 82, and 129) and three of lion (Panthera leo persica) SLAM (72, 82, and 129) were associated with charge alterations, suggesting that the felid interfaces have lower affinities to dog CDV. Only the residue at 76 was different between domestic cat and lion SLAM interfaces. The domestic cat SLAM had threonine at 76, whereas the lion SLAM had arginine, a positively charged residue like that of the dog SLAM. The cat SLAM with threonine is likely to have lower affinity to CDV-H and to confer higher resistance against dog CDV. Thus, the four residues (72, 76, 82, and 129) on carnivore SLAMs are important for the determination of affinity and sensitivity with CDV. Additionally, the CDV-H protein of felid strains had a substitution of histidine for tyrosine at 549 of dog CDV-H and may have higher affinity to lion SLAM. Three-dimensional model construction is a new risk assessment method of morbillivirus infectivity. Because the method is applicable to animals that have no information about virus infection, it is especially useful for morbillivirus risk assessment and wildlife conservation.
Collapse
|
32
|
Namuwulya P, Abernathy E, Bukenya H, Bwogi J, Tushabe P, Birungi M, Seguya R, Kabaliisa T, Alibu VP, Kayondo JK, Rivailler P, Icenogle J, Bakamutumaho B. Phylogenetic analysis of rubella viruses identified in Uganda, 2003-2012. J Med Virol 2014; 86:2107-13. [PMID: 24700073 DOI: 10.1002/jmv.23935] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2014] [Indexed: 11/07/2022]
Abstract
Molecular data on rubella viruses are limited in Uganda despite the importance of congenital rubella syndrome (CRS). Routine rubella vaccination, while not administered currently in Uganda, is expected to begin by 2015. The World Health Organization recommends that countries without rubella vaccination programs assess the burden of rubella and CRS before starting a routine vaccination program. Uganda is already involved in integrated case-based surveillance, including laboratory testing to confirm measles and rubella, but molecular epidemiologic aspects of rubella circulation have so far not been documented in Uganda. Twenty throat swab or oral fluid samples collected from 12 districts during routine rash and fever surveillance between 2003 and 2012 were identified as rubella virus RNA positive and PCR products encompassing the region used for genotyping were sequenced. Phylogenetic analysis of the 20 sequences identified 19 genotype 1G viruses and 1 genotype 1E virus. Genotype-specific trees showed that the Uganda viruses belonged to specific clusters for both genotypes 1G and 1E and grouped with similar sequences from neighboring countries. Genotype 1G was predominant in Uganda. More epidemiological and molecular epidemiological data are required to determine if genotype 1E is also endemic in Uganda. The information obtained in this study will assist the immunization program in monitoring changes in circulating genotypes.
Collapse
|
33
|
Mateo M, Navaratnarajah CK, Cattaneo R. Structural basis of efficient contagion: measles variations on a theme by parainfluenza viruses. Curr Opin Virol 2014; 5:16-23. [PMID: 24492202 PMCID: PMC4028398 DOI: 10.1016/j.coviro.2014.01.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 11/26/2013] [Accepted: 01/08/2014] [Indexed: 11/19/2022]
Abstract
A quartet of attachment proteins and a trio of fusion protein subunits play the cell entry concert of parainfluenza viruses. While many of these viruses bind sialic acid to enter cells, wild type measles binds exclusively two tissue-specific proteins, the lymphatic receptor signaling lymphocytic activation molecule (SLAM), and the epithelial receptor nectin-4. SLAM binds near the stalk-head junction of the hemagglutinin. Nectin-4 binds a hydrophobic groove located between blades 4 and 5 of the hemagglutinin β-propeller head. The mutated vaccine strain hemagglutinin binds in addition the ubiquitous protein CD46, which explains attenuation. The measles virus entry concert has four movements. Andante misterioso: the virus takes over the immune system. Allegro con brio: it rapidly spreads in the upper airway's epithelia. 'Targeting' fugue: the versatile orchestra takes off. Presto furioso: the virus exits the host with thunder. Be careful: music is contagious.
Collapse
MESH Headings
- Animals
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cell Adhesion Molecules/chemistry
- Cell Adhesion Molecules/genetics
- Cell Adhesion Molecules/metabolism
- Hemagglutinins, Viral/chemistry
- Hemagglutinins, Viral/genetics
- Hemagglutinins, Viral/metabolism
- Humans
- Measles/genetics
- Measles/metabolism
- Measles/virology
- Measles virus/chemistry
- Measles virus/genetics
- Measles virus/metabolism
- Protein Binding
- Receptors, Cell Surface/chemistry
- Receptors, Cell Surface/genetics
- Receptors, Cell Surface/metabolism
- Receptors, Virus/chemistry
- Receptors, Virus/genetics
- Receptors, Virus/metabolism
- Signaling Lymphocytic Activation Molecule Family Member 1
Collapse
Affiliation(s)
- Mathieu Mateo
- Department of Molecular Medicine, Mayo Clinic, and Virology and Gene Therapy Track, Mayo Graduate School, 200 First Street SW, Rochester, MN 55905, USA
| | - Chanakha K Navaratnarajah
- Department of Molecular Medicine, Mayo Clinic, and Virology and Gene Therapy Track, Mayo Graduate School, 200 First Street SW, Rochester, MN 55905, USA
| | - Roberto Cattaneo
- Department of Molecular Medicine, Mayo Clinic, and Virology and Gene Therapy Track, Mayo Graduate School, 200 First Street SW, Rochester, MN 55905, USA.
| |
Collapse
|
34
|
Canine distemper virus epithelial cell infection is required for clinical disease but not for immunosuppression. J Virol 2012; 86:3658-66. [PMID: 22278252 DOI: 10.1128/jvi.06414-11] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To characterize the importance of infection of epithelial cells for morbillivirus pathogenesis, we took advantage of the severe disease caused by canine distemper virus (CDV) in ferrets. To obtain a CDV that was unable to enter epithelial cells but retained the ability to enter immune cells, we transferred to its attachment (H) protein two mutations shown to interfere with the interaction of measles virus H with its epithelial receptor, human nectin-4. As expected for an epithelial receptor (EpR)-blind CDV, this virus infected dog and ferret epithelial cells inefficiently and did not cause cell fusion or syncytium formation. On the other hand, the EpR-blind CDV replicated in cells expressing canine signaling lymphocyte activation molecule (SLAM), the morbillivirus immune cell receptor, with similar kinetics to those of wild-type CDV. While ferrets infected with wild-type CDV died within 12 days after infection, after developing severe rash and fever, animals infected with the EpR-blind virus showed no clinical signs of disease. Nevertheless, both viruses spread rapidly and efficiently in immune cells, causing similar levels of leukopenia and inhibition of lymphocyte proliferation activity, two indicators of morbillivirus immunosuppression. Infection was documented for airway epithelia of ferrets infected with wild-type CDV but not for those of animals infected with the EpR-blind virus, and only animals infected with wild-type CDV shed virus. Thus, epithelial cell infection is necessary for clinical disease and efficient virus shedding but not for immunosuppression.
Collapse
|
35
|
Hashiguchi T, Maenaka K, Yanagi Y. Measles virus hemagglutinin: structural insights into cell entry and measles vaccine. Front Microbiol 2011; 2:247. [PMID: 22319511 PMCID: PMC3267179 DOI: 10.3389/fmicb.2011.00247] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Accepted: 11/23/2011] [Indexed: 12/17/2022] Open
Abstract
Measles is one of the most contagious viral diseases, and remains a major cause of childhood morbidity and mortality worldwide. The measles virus (MV), a member of the family Paramyxoviridae, enters cells through a cellular receptor, the signaling lymphocyte activation molecule (SLAM), CD46 or nectin-4. Entry is mediated by two MV envelope glycoproteins, the hemagglutinin (H) and the fusion (F) protein. The H protein mediates receptor attachment, while the F protein causes membrane fusion. The interaction between the H and F proteins is essential to initiate the cell entry process. Recently determined crystal structures of the MV-H protein unbound and bound to SLAM or CD46 have provided insights into paramyxovirus entry and the effectiveness of measles vaccine.
Collapse
Affiliation(s)
- Takao Hashiguchi
- Department of Virology, Faculty of Medicine, Kyushu University Fukuoka, Japan
| | | | | |
Collapse
|
36
|
Ovsyannikova IG, Haralambieva IH, Vierkant RA, O'Byrne MM, Jacobson RM, Poland GA. The association of CD46, SLAM and CD209 cellular receptor gene SNPs with variations in measles vaccine-induced immune responses: a replication study and examination of novel polymorphisms. Hum Hered 2011; 72:206-23. [PMID: 22086389 DOI: 10.1159/000331585] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Accepted: 08/09/2011] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND The measles virus (MV) interacts with two known cellular receptors: CD46 and SLAM. The transmembrane receptor CD209 interacts with MV and augments dendritic cell infection. METHODS 764 subjects previously immunized with measles-mumps-rubella vaccine were genotyped for 66 candidate SNPs in the CD46, SLAM and CD209 genes as part of a larger study. RESULTS A previously detected association of the CD46 SNP rs2724384 with measles-specific antibodies was successfully replicated in this study. Increased representation of the minor allele G for an intronic CD46 SNP was associated with an allele dose-related decrease (978 vs. 522 mIU/ml, p = 0.0007) in antibody levels. This polymorphism rs2724384 also demonstrated associations with IL-6 (p = 0.02), IFN-α (p = 0.007) and TNF-α (p = 0.0007) responses. Two polymorphisms (coding rs164288 and intronic rs11265452) in the SLAM gene that were associated with measles antibody levels in our previous study were associated with IFN-γ Elispot (p = 0.04) and IL-10 responses (p = 0.0008), respectively, in this study. We found associations between haplotypes, AACGGAATGGAAAG (p = 0.009) and GGCCGAGAGGAGAG (p < 0.001), in the CD46 gene and TNF-α secretion. CONCLUSION Understanding the functional and mechanistic consequences of these genetic polymorphisms on immune response variations could assist in directing new measles and potentially other viral vaccine design, and in better understanding measles immunogenetics.
Collapse
|
37
|
Canine distemper virus infects canine keratinocytes and immune cells by using overlapping and distinct regions located on one side of the attachment protein. J Virol 2011; 85:11242-54. [PMID: 21849439 DOI: 10.1128/jvi.05340-11] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The morbilliviruses measles virus (MeV) and canine distemper virus (CDV) both rely on two surface glycoproteins, the attachment (H) and fusion proteins, to promote fusion activity for viral cell entry. Growing evidence suggests that morbilliviruses infect multiple cell types by binding to distinct host cell surface receptors. Currently, the only known in vivo receptor used by morbilliviruses is CD150/SLAM, a molecule expressed in certain immune cells. Here we investigated the usage of multiple receptors by the highly virulent and demyelinating CDV strain A75/17. We based our study on the assumption that CDV-H may interact with receptors similar to those for MeV, and we conducted systematic alanine-scanning mutagenesis on CDV-H throughout one side of the β-propeller documented in MeV-H to contain multiple receptor-binding sites. Functional and biochemical assays performed with SLAM-expressing cells and primary canine epithelial keratinocytes identified 11 residues mutation of which selectively abrogated fusion in keratinocytes. Among these, four were identical to amino acids identified in MeV-H as residues contacting a putative receptor expressed in polarized epithelial cells. Strikingly, when mapped on a CDV-H structural model, all residues clustered in or around a recessed groove located on one side of CDV-H. In contrast, reported CDV-H mutants with SLAM-dependent fusion deficiencies were characterized by additional impairments to the promotion of fusion in keratinocytes. Furthermore, upon transfer of residues that selectively impaired fusion induction in keratinocytes into the CDV-H of the vaccine strain, fusion remained largely unaltered. Taken together, our results suggest that a restricted region on one side of CDV-H contains distinct and overlapping sites that control functional interaction with multiple receptors.
Collapse
|
38
|
Navaratnarajah CK, Oezguen N, Rupp L, Kay L, Leonard VHJ, Braun W, Cattaneo R. The heads of the measles virus attachment protein move to transmit the fusion-triggering signal. Nat Struct Mol Biol 2011; 18:128-34. [PMID: 21217701 PMCID: PMC3059746 DOI: 10.1038/nsmb.1967] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2010] [Accepted: 10/25/2010] [Indexed: 12/21/2022]
Abstract
The measles virus entry system, consisting of attachment (hemagglutinin, H) and fusion proteins, operates by means of a variety of natural and targeted receptors; however, the mechanism that triggers fusion of the viral envelope with the plasma membrane is not understood. Here, we tested a model proposing that the two heads of an H dimer, which are covalently linked at their base, after binding two receptor molecules, move relative to each other to transmit the fusion-triggering signal. Indeed, stabilizing the H-dimer interface with additional intermolecular disulfide bonds prevented membrane fusion, an effect that was reversed by a reducing agent. Moreover, a membrane-anchored designated receptor efficiently triggered fusion, provided that it engaged the H dimer at locations proximal to where the natural receptors bind and distal to the H-dimer interface. We discuss how receptors may force H-protein heads to switch partners and transmit the fusion-triggering signal.
Collapse
|
39
|
Structure of the measles virus hemagglutinin bound to its cellular receptor SLAM. Nat Struct Mol Biol 2011; 18:135-41. [DOI: 10.1038/nsmb.1969] [Citation(s) in RCA: 197] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 10/29/2010] [Indexed: 12/26/2022]
|
40
|
Bowden T, Crispin M, Jones E, Stuart D. Shared paramyxoviral glycoprotein architecture is adapted for diverse attachment strategies. Biochem Soc Trans 2010; 38:1349-55. [PMID: 20863312 PMCID: PMC3433257 DOI: 10.1042/bst0381349] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2010] [Indexed: 11/17/2022]
Abstract
Members within the paramyxovirus subfamily Paramyxovirinae constitute a large number of highly virulent human and animal pathogens. The glycoproteins present on these viruses are responsible for mediating host cell attachment and fusion and are key targets for the design of antiviral entry inhibitors. In the present review, we discuss recent structural studies which have led to a better understanding of the various mechanisms by which different paramyxoviruses use their attachment glycoproteins to hijack specific protein and glycan cell-surface receptors to facilitate viral entry. It is observed that the paramyxovirus attachment glycoprotein consists of a conserved overall structure which includes an N-terminal six-bladed β-propeller domain which is responsible for cell receptor binding. Crystal structures of this domain from different biomedically important paramyxoviruses, including measles, Nipah, Hendra, Newcastle disease and parainfluenza viruses, alone and in complex with their functional cell-surface receptors, demonstrate three contrasting mechanisms of receptor engagement that paramyxoviruses have evolved to confer discreet protein- and glycan-receptor specificity. This structural information highlights the adaptability of the paramyxovirus attachment glycoprotein surface and the potential for the emergence of new and potentially harmful viruses in human hosts.
Collapse
Key Words
- paramyxovirus
- protein crystallography
- structural virology
- viral glycoprotein
- virus entry
- dana, 2,3-dehydro-2-deoxy-n-acetylneuraminic acid
- f, fusion glycoprotein
- g, attachment glycoprotein
- h, haemagglutinin
- hev, hendra virus
- hn, haemagglutinin-neuraminidase
- hnv, henipavirus
- mv, measles virus
- ndv, newcastle disease virus
- piv, parainfluenza virus
- niv, nipah virus
- r.m.s.d., root mean square deviation
- scr, short consensus repeat
- slam, signalling lymphocyte activation molecule
Collapse
Affiliation(s)
- Thomas A. Bowden
- *Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K
| | - Max Crispin
- †Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, South Parks Road, Oxford OX1 3QU, U.K
| | - E. Yvonne Jones
- *Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K
| | - David I. Stuart
- *Division of Structural Biology, Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, U.K
| |
Collapse
|
41
|
Santiago C, Gutiérrez-Rodríguez A, Tucker PA, Stehle T, Casasnovas JM. Crystallization and preliminary crystallographic analysis of the measles virus hemagglutinin in complex with the CD46 receptor. Acta Crystallogr Sect F Struct Biol Cryst Commun 2010; 66:91-4. [PMID: 20057080 PMCID: PMC2805546 DOI: 10.1107/s1744309109050593] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 11/24/2009] [Indexed: 05/28/2023]
Abstract
The measles virus (MV) hemagglutinin (MV-H) mediates the attachment of MV particles to cell-surface receptors for entry into host cells. MV uses two receptors for attachment to host cells: the complement-control protein CD46 and the signalling lymphocyte activation molecule (SLAM). The MV-H glycoprotein from an Edmonston MV variant and the MV-binding fragment of the CD46 receptor were overproduced in mammalian cells and used to crystallize an MV-H-CD46 complex. Well diffracting crystals containing two complexes in the asymmetric unit were obtained and the structure of the complex was solved by the molecular-replacement method.
Collapse
Affiliation(s)
- César Santiago
- Centro Nacional de Biotecnologia, CSIC, Campus Universidad Autonoma, 28049 Madrid, Spain
- Karolinska Institute, Department of Biosciences at NOVUM, Center for Biotechnology, 141 57 Huddinge, Sweden
| | - Angel Gutiérrez-Rodríguez
- Karolinska Institute, Department of Biosciences at NOVUM, Center for Biotechnology, 141 57 Huddinge, Sweden
| | - Paul A. Tucker
- European Molecular Biology Laboratory, Hamburg Outstation, Notkestrasse 85, D-22603 Hamburg, Germany
| | - Thilo Stehle
- Interfaculty Institute for Biochemistry, University of Tübingen, D-72076 Tübingen, Germany
- Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | - José M. Casasnovas
- Centro Nacional de Biotecnologia, CSIC, Campus Universidad Autonoma, 28049 Madrid, Spain
- Karolinska Institute, Department of Biosciences at NOVUM, Center for Biotechnology, 141 57 Huddinge, Sweden
| |
Collapse
|
42
|
Measles virus infection of alveolar macrophages and dendritic cells precedes spread to lymphatic organs in transgenic mice expressing human signaling lymphocytic activation molecule (SLAM, CD150). J Virol 2009; 84:3033-42. [PMID: 20042501 DOI: 10.1128/jvi.01559-09] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Recent studies of primate models suggest that wild-type measles virus (MV) infects immune cells located in the airways before spreading systemically, but the identity of these cells is unknown. To identify cells supporting primary MV infection, we took advantage of mice expressing the MV receptor human signaling lymphocyte activation molecule (SLAM, CD150) with human-like tissue specificity. We infected these mice intranasally (IN) with a wild-type MV expressing green fluorescent protein. One, two, or three days after inoculation, nasal-associated lymphoid tissue (NALT), the lungs, several lymph nodes (LNs), the spleen, and the thymus were collected and analyzed by microscopy and flow cytometry, and virus isolation was attempted. One day after inoculation, MV replication was documented only in the airways, in about 2.5% of alveolar macrophages (AM) and 0.5% of dendritic cells (DC). These cells expressed human SLAM, and it was observed that MV infection temporarily enhanced SLAM expression. Later, MV infected other immune cell types, including B and T lymphocytes. Virus was isolated from lymphatic tissue as early as 2 days post-IN inoculation; the mediastinal lymph node was an early site of replication and supported high levels of infection. Three days after intraperitoneal inoculation, 1 to 8% of the mediastinal LN cells were infected. Thus, MV infection of alveolar macrophages and subepithelial dendritic cells in the airways precedes infection of lymphocytes in lymphatic organs of mice expressing human SLAM with human-like tissue specificity.
Collapse
|
43
|
Santiago C, Celma ML, Stehle T, Casasnovas JM. Structure of the measles virus hemagglutinin bound to the CD46 receptor. Nat Struct Mol Biol 2009; 17:124-9. [DOI: 10.1038/nsmb.1726] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Accepted: 09/29/2009] [Indexed: 12/25/2022]
|
44
|
Dermody TS, Kirchner E, Guglielmi KM, Stehle T. Immunoglobulin superfamily virus receptors and the evolution of adaptive immunity. PLoS Pathog 2009; 5:e1000481. [PMID: 19956667 PMCID: PMC2777377 DOI: 10.1371/journal.ppat.1000481] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Terence S. Dermody
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- * E-mail: (TSD); (TS)
| | - Eva Kirchner
- Interfaculty Institute for Biochemistry, University of Tuebingen, Tuebingen, Germany
| | - Kristen M. Guglielmi
- Department of Microbiology and Immunology, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Elizabeth B. Lamb Center for Pediatric Research, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
| | - Thilo Stehle
- Department of Pediatrics, Vanderbilt University School of Medicine, Nashville, Tennessee, United States of America
- Interfaculty Institute for Biochemistry, University of Tuebingen, Tuebingen, Germany
- * E-mail: (TSD); (TS)
| |
Collapse
|
45
|
Sarkar J, Balamurugan V, Sen A, Saravanan P, Sahay B, Rajak KK, Rasool TJ, Bhanuprakash V, Singh RK. Sequence analysis of morbillivirus CD150 receptor-Signaling Lymphocyte Activation Molecule (SLAM) of different animal species. Virus Genes 2009; 39:335-41. [PMID: 19669672 DOI: 10.1007/s11262-009-0391-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2009] [Accepted: 07/31/2009] [Indexed: 10/20/2022]
Abstract
Signaling Lymphocyte Activation Molecule-SLAM (CD150) molecule has been reported as a putative receptor for most morbilliviruses for their respective host species. In this study, we determined the complete nucleotide sequence of the gene coding for the morbillivirus receptor-SLAM from the four species, namely, goat (Capra hircus), sheep (Ovis aries), Indian cattle (Bos indicus), and buffalo (Bubalus bubalis). The nucleotide (nt) open reading frame sequence of SLAM gene in all the four species studied was 1017 nucleotides in length encoding a polypeptide of 339 amino acids (aa), similar to Bos taurus, but different from canine, human, marmoset, and mouse SLAM, which were 1029, 1008, 1011, and 1032 nts, respectively, in length, and coding for 343, 336, 337, and 344 aa, respectively. Sequence analysis revealed 96.3-98.5% and 92.9-96.8% identities among the four species at the nt and aa level, respectively. Sequence diversity at aa level between various species revealed that the critical functional region of SLAM protein among different species is relatively conserved, thereby facilitating this molecule to act as a receptor for morbillivirus. Phylogenetic relationship based on the aa sequences of SLAM protein revealed that caprine, ovine, cattle, and buffalo fall under a defined cluster but caprine SLAM is more closely related to ovine, followed by bovine.
Collapse
Affiliation(s)
- J Sarkar
- National Morbillivirus Referral Laboratory, Division of Virology, Indian Veterinary Research Institute, Uttarakhand, India
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Griffin DE, Oldstone MBA. Measles virus glycoprotein complex assembly, receptor attachment, and cell entry. Curr Top Microbiol Immunol 2009; 329:59-76. [PMID: 19198562 PMCID: PMC7121846 DOI: 10.1007/978-3-540-70523-9_4] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Measles virus (MV) enters cells by membrane fusion at the cell surface at neutral pH. Two glycoproteins mediate this process: the hemagglutinin (H) and fusion (F) proteins. The H-protein binds to receptors, while the F-protein mediates fusion of the viral and cellular membranes. H naturally interacts with at least three different receptors. The wild-type virus primarily uses the signaling lymphocyte activation molecule (SLAM, CD150) expressed on certain lymphatic cells, while the vaccine strain has gained the ability to also use the ubiquitous membrane cofactor protein (MCP, CD46), a regulator of complement activation. Additionally, MV infects polarized epithelial cells through an unidentified receptor (EpR). The footprints of the three receptors on H have been characterized, and the focus of research is shifting to the characterization of receptor-specific conformational changes that occur in the H-protein dimer and how these are transmitted to the F-protein trimer. It was also shown that MV attachment and cell entry can be readily targeted to designated receptors by adding specificity determinants to the H-protein. These studies have contributed to our understanding of membrane fusion by the glycoprotein complex of paramyxoviruses in general.
Collapse
Affiliation(s)
- Diane E. Griffin
- Department of Molecular Microbiology, Johns Hopkins University School of Hygiene and Public Health, 615 N. Wolfe Street, Baltimore, MD 21205 USA
| | - Michael B. A. Oldstone
- Department of Immunology and Microbial Science, The Scripps Research Institute, 10550 N. Torrey Pines, La Jolla, CA 92037 USA
| |
Collapse
|
47
|
Yanagi Y, Takeda M, Ohno S, Hashiguchi T. Measles virus receptors. Curr Top Microbiol Immunol 2009; 329:13-30. [PMID: 19198560 DOI: 10.1007/978-3-540-70523-9_2] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Measles virus (MV) has two envelope glycoproteins, the hemagglutinin (H) and fusion protein, which are responsible for attachment and membrane fusion, respectively. Signaling lymphocyte activation molecule (SLAM, also called CD150), a membrane glycoprotein expressed on immune cells, acts as the principal cellular receptor for MV, accounting for its lymphotropism and immunosuppressive nature. MV also infects polarized epithelial cells via an as yet unknown receptor molecule, thereby presumably facilitating transmission via aerosol droplets. Vaccine and laboratory-adapted strains of MV use ubiquitously expressed CD46 as an alternate receptor through amino acid substitutions in the H protein. The crystal structure of the H protein indicates that the putative binding sites for SLAM, CD46, and the epithelial cell receptor are strategically located in different positions of the H protein. Other molecules have also been implicated in MV infection, although their relevance remains to be determined. The identification of MV receptors has advanced our understanding of MV tropism and pathogenesis.
Collapse
Affiliation(s)
- Y Yanagi
- Department of Virology, Faculty of Medicine, Kyushu University, 812-8582, Fukuoka, Japan.
| | | | | | | |
Collapse
|
48
|
Abstract
X-ray crystallographic analyses, together with nuclear magnetic resonance, have revealed three-dimensional structures of many important viral proteins, thereby allowing us to better understand the interactions between viral and host cell molecules. In this review, we summarize the recently determined crystal structure of the measles virus (MV) attachment protein hemagglutinin. Based on this structural information, we also discuss how the MV hemagglutinin interacts with various cellular receptors and why MV vaccines have been effective for many years without inducing escape mutant viruses. Other topics discussed are a putative MV receptor present on polarized epithelial cells and the protein expression system using a cultured human cell line 293SGnTI(-), which is suitable for X-ray crystallographic analyses.
Collapse
|
49
|
Abstract
Measles virus (MV) is one of the most transmissible microorganisms known, continuing to result in extensive morbidity and mortality worldwide. While rare, MV can infect the human central nervous system, triggering fatal CNS diseases weeks to years after exposure. The advent of crucial laboratory tools to dissect MV neuropathogenesis, including permissive transgenic mouse models, the capacity to manipulate the viral genome using reverse genetics, and cell biology advances in understanding the processes that govern intracellular trafficking of viral components, have substantially clarified how MV infects, spreads, and persists in this unique cell population. This review highlights some of these technical advances, followed by a discussion of our present understanding of MV neuronal infection and transport. Because some of these processes may be shared among diverse viruses, comparisons are made to parallel studies with other neurotropic viruses. While a crystallized view of how the unique environment of the neuron affects MV replication, spread, and, ultimately, neuropathogenesis is not fully realized, the tools and ideas are in place for exciting advances in the coming years.
Collapse
Affiliation(s)
| | - Glenn F. Rall
- corresponding author Division of Basic Science Fox Chase Cancer Center 333 Cottman Avenue Philadelphia, PA 19111 USA Phone: 215-728-3617 Fax: 215-728-2412
| |
Collapse
|
50
|
Ohishi K, Ando A, Suzuki R, Takishita K, Kawato M, Katsumata E, Ohtsu D, Okutsu K, Tokutake K, Miyahara H, Nakamura H, Murayama T, Maruyama T. Host-virus specificity of morbilliviruses predicted by structural modeling of the marine mammal SLAM, a receptor. Comp Immunol Microbiol Infect Dis 2008; 33:227-41. [PMID: 19027953 DOI: 10.1016/j.cimid.2008.10.003] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/04/2008] [Indexed: 10/21/2022]
Abstract
Signaling lymphocyte activation molecule (SLAM) is thought to be a major cellular receptor for high-host specificity morbilliviruses, which cause devastating and highly infectious diseases in mammals. We determined the sequences of SLAM cDNA from five species of marine mammal, including two cetaceans, two pinnipeds and one sirenian, and generated three-dimensional models to understand the receptor-virus interaction. Twenty-one amino acid residues in the immunoglobulin-like V domains of the SLAMs were shown to bind the viral protein. Notably, the sequences from pinnipeds and dogs were highly homologous, which is consistent with the fact that canine distemper virus was previously shown to cause a mass die-off of seals. Among these twenty-one residues, eight (63, 66, 68, 72, 84, 119, 121 and 130) were shared by animal groups susceptible to a particular morbillivirus species. This set of residues appears to determine host-virus specificity and may be useful for risk estimation for morbilliviruses.
Collapse
Affiliation(s)
- Kazue Ohishi
- Japan Agency for Marine-Earth Science and Technology (JAMSTEC), Yokosuka, Kanagawa, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|