1
|
Castellan M, Zamperin G, Foiani G, Zorzan M, Priore MF, Drzewnioková P, Melchiotti E, Vascellari M, Monne I, Crovella S, Leopardi S, De Benedictis P. Immunological findings of West Caucasian bat virus in an accidental host. J Virol 2025; 99:e0191424. [PMID: 39846740 PMCID: PMC11853057 DOI: 10.1128/jvi.01914-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
The Lyssavirus genus includes seventeen viral species able to cause rabies, an acute and almost invariably fatal encephalomyelitis of mammals. Rabies virus (RABV), which represents the type species of the genus, is a multi-host pathogen that over the years has undergone multiple events of host-switching, thus occupying several geographical and ecological niches. In contrast, non-RABV lyssaviruses are mainly confined within a single natural host with rare spillover events. In this scenario, unveiling the mechanisms underlying the host immune response against a virus is crucial to understand the dynamics of infection and to predict the probability of colonization/adaptation to a new target species. Presently, the host response to lyssaviruses has only been partially explored, with the majority of data extrapolated from RABV infection. West Caucasian bat virus (WCBV), a divergent lyssavirus, has recently been associated with a spillover event to a domestic cat, raising concern about the risks to public health due to the circulation of the virus in its natural host. Through this study we have investigated the immune response determined by the WCBV versus two widely known lyssaviruses. We selected the Syrian hamster as representative of an accidental host, and chose the intramuscular route in order to mimic the natural infection. In hamsters, WCBV was highly pathogenic, determining 100% lethality and mild encephalitis. In comparison with Duvenhage virus (DUVV) and RABV, we found that WCBV displayed an intermediate ability to promote cellular antiviral response, produce pro-inflammatory cytokines, and recruit and activate lymphocytes in the hamsters' central nervous system. IMPORTANCE Although all lyssaviruses cause fatal encephalomyelitis in mammals, they display a different host tropism and pathogenicity, with the ecology of Rabies virus (RABV) continually evolving and adapting to new host species. In 2020, West Caucasian bat virus (WCBV) was identified as the causative agent of rabies in a domestic cat in Italy. This event raised concerns about its public health consequences, due to the absence of biologicals against the infection. Our study investigates the host immune response triggered by WCBV in comparison with a pathogenic strain of RABV and the low pathogenic Duvenhage lyssavirus (DUVV), as a proxy to understand the mechanisms leading to lyssavirus spillover and pathogenicity. We overall confirm that previous evidence indicating an inverse relationship between lyssavirus pathogenicity and immune response is applicable for WCBV as well. Importantly, this work represents the first transcriptomic analysis of the WCBV interaction in the central nervous system with an accidental host.
Collapse
Affiliation(s)
- Martina Castellan
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Gianpiero Zamperin
- Viral Genomics and Transcriptomics Laboratory, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Greta Foiani
- Laboratory of Histopathology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Maira Zorzan
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Maria Francesca Priore
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Petra Drzewnioková
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Erica Melchiotti
- Laboratory of Histopathology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Marta Vascellari
- Laboratory of Histopathology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Isabella Monne
- Viral Genomics and Transcriptomics Laboratory, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Sergio Crovella
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| | - Stefania Leopardi
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Paola De Benedictis
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| |
Collapse
|
2
|
Chen SJ, Rai CI, Wang SC, Chen YC. Infection and Prevention of Rabies Viruses. Microorganisms 2025; 13:380. [PMID: 40005749 PMCID: PMC11858514 DOI: 10.3390/microorganisms13020380] [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: 12/08/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Rabies is a fatal zoonotic disease and causes about 59,000 human deaths globally every year. Especially, its mortality is almost 100% in cases where the rabies virus has transmitted to the central nervous system. The special virus life cycle and pathogenic mechanism make it difficult for the host immune system to combat rabies viruses. Vaccination including pre-exposure and post-exposure prophylaxis is an effective strategy for rabies prevention. The pre-exposure vaccination is mainly applied for animals and the post-exposure vaccination is the most application for humans. Although rabies vaccines are widely used and seem to be safe and effective, there are some disadvantages, limitations, or challenges affecting vaccine promotion and distribution. Therefore, more effective, convenient, safer, and cheaper rabies vaccines have been developed or are being developed. The development of novel human rabies vaccine is mainly focusing on vaccines based on a purified Vero cell-cultured freeze-dried rabies vaccine (PVRV). PVRV has been demonstrated to be promising to make the rabies vaccine more effective and secure in animal studies or clinical trials. Moreover, mRNA-based vaccines have been shown to have the potential to enhance the safety and efficacy of rabies vaccines for both animal and human uses.
Collapse
Affiliation(s)
- Shiu-Jau Chen
- Department of Neurosurgery, Mackay Memorial Hospital, Taipei 10449, Taiwan;
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Chung-I Rai
- Department of Cosmetic Science, Vanung University, 1 Van Nung Road, Chung-Li City, Taoyuan 320676, Taiwan;
| | - Shao-Cheng Wang
- Department of Psychiatric, Taoyuan General Hospital, Ministry of Health and Welfare, Taoyuan 33004, Taiwan
- Department of Psychiatry, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung 833, Taiwan
- Department of Psychiatry, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung 801, Taiwan
| | - Yuan-Chuan Chen
- Department of Nursing, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 356006, Taiwan
- Department of Medical Technology, Jenteh Junior College of Medicine, Nursing and Management, Miaoli County 356006, Taiwan
| |
Collapse
|
3
|
Ya N, Auerswald H, Touch S, In S, Yun C, Thai P, Sann S, Heng B, Leng C, Duong V, Peng YS, Ly S, Cantaert T. Evaluation of one year immunity following rabies post-exposure prophylaxis in dog bite cases. NPJ Vaccines 2024; 9:237. [PMID: 39604401 PMCID: PMC11603308 DOI: 10.1038/s41541-024-01030-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
Rabies remains a global health threat despite being preventable with post-exposure prophylaxis (PEP). This study assessed one-year humoral and T cell immunity in PEP recipients of the Insitut Pasteur du Cambodge (IPC) regimen, recommended by WHO. We analyzed rabies virus (RABV) neutralizing antibodies (nAbs) and T cell responses at baseline, 7 and 14 days, 6 and 12 months after PEP. A total of 148 patients were included, with 78 bitten by confirmed RABV-positive dogs receiving PEP and equine rabies immunoglobulins (eRIG), and 70 bitten by RABV-negative dogs receiving only PEP. Fourteen days after PEP, all but two individuals seroconverted for nAbs ( ≥ 0.5 IU/mL) with 87% maintaining this response even after 12 months. Interleukin-4 (IL-4) and interferon-gamma (IFN-γ)-secreting T cells were significantly elevated after 14 days and sustained for one year. No differences were observed between the RABV-exposed and -unexposed groups. This study demonstrates robust one-year immunity after IPC PEP.
Collapse
Affiliation(s)
- Nisa Ya
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Heidi Auerswald
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- Virology Unit, Istituto Zooprofilattico Sperimentale dell'Abruzzo e del Molise 'G. Caporale' (IZSAM), Teramo, Italy
| | - Sothy Touch
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Saraden In
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Chanvannak Yun
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Pisey Thai
- Rabies Prevention Center, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Borita Heng
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Chanthy Leng
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Veasna Duong
- Virology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Yik Sing Peng
- Rabies Prevention Center, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Sowath Ly
- Epidemiology and Public Health Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia.
| |
Collapse
|
4
|
Liu Q, He Q, Tao X, Yu P, Liu S, Xie Y, Zhu W. Resveratrol inhibits rabies virus infection in N2a cells by activating the SIRT1/Nrf2/HO-1 pathway. Heliyon 2024; 10:e36494. [PMID: 39281556 PMCID: PMC11399676 DOI: 10.1016/j.heliyon.2024.e36494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/12/2024] [Accepted: 08/16/2024] [Indexed: 09/18/2024] Open
Abstract
Rabies is a highly lethal infectious disease with no existing treatment available, thus investigating effective antiviral compounds to control rabies virus (RABV) infection is of utmost importance. Resveratrol is a natural phenolic compound that, as a phytoalexin, exhibits several biological activities, including antiviral activity. In this study, we evaluated the inhibitory effect of resveratrol on RABV infection and investigated its molecular antiviral mechanism. We found that resveratrol significantly inhibited RABV infection, including the phases of adsorption, replication, and release, and also directly inactivated RABV and inhibited its infectivity. However, resveratrol had no significant effect on RABV internalization. Resveratrol also reduced RABV-induced oxidative stress, specifically reactive oxygen species and malondialdehyde levels. Western blotting analysis revealed that resveratrol enhanced antioxidant signaling via the SIRT1/Nrf2/HO-1 pathway and inhibited viral replication. Viral infection was enhanced after SIRT1 knockdown, which inhibited the SIRT1/Nrf2/HO-1 antioxidant signaling pathway, suggesting that this pathway plays an important role in RABV replication. Overall, resveratrol prevented the adsorption, replication, and release of RABV and directly inactivated RABV, but failed to inhibit RABV internalization. Furthermore, resveratrol activated the SIRT1/Nrf2/HO-1 pathway to inhibit RABV replication and suppressed RABV-induced oxidative stress. These findings highlight the therapeutic potential of resveratrol for fighting RABV infections.
Collapse
Affiliation(s)
- Qian Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Qing He
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Xiaoyan Tao
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Pengcheng Yu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Shuqing Liu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Yuan Xie
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Wuyang Zhu
- National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| |
Collapse
|
5
|
Aliakbari S, Hasanzadeh L, Sayyah M, Amini N, Pourbadie HG. Induced expression of rabies glycoprotein in the dorsal hippocampus enhances hippocampal dependent memory in a rat model of Alzheimer's disease. J Neurovirol 2024; 30:274-285. [PMID: 38943023 DOI: 10.1007/s13365-024-01221-y] [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: 03/24/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 06/30/2024]
Abstract
The Rabies virus is a neurotropic virus that manipulates the natural cell death processes of its host to ensure its own survival and replication. Studies have shown that the anti-apoptotic effect of the virus is mediated by one of its protein named, rabies glycoprotein (RVG). Alzheimer's disease (AD) is characterized by the loss of neural cells and memory impairment. We aim to examine whether expression of RVG in the hippocampal cells can shield the detrimental effects induced by Aβ. Oligomeric form of Aβ (oAβ) or vehicle was bilaterally microinjected into the dorsal hippocampus of male Wistar rats. One week later, two μl (108 T.U. /ml) of the lentiviral vector carrying RVG gene was injected into their dorsal hippocampus (post-treatment). In another experiment, the lentiviral vector was microinjected one week before Aβ injection (pre-treatment). One week later, the rat's brain was sliced into cross-sections, and the presence of RVG-expressing neuronal cells was confirmed using fluorescent microscopy. Rats were subjected to assessments of spatial learning and memory as well as passive avoidance using the Morris water maze (MWM) and the Shuttle box apparatuses, respectively. Protein expression of AMPA receptor subunit (GluA1) was determined using western blotting technique. In MWM, Aβ treated rats showed decelerated acquisition of the task and impairment of reference memory. RVG expression in the hippocampus prevented and restored the deficits in both pre- and post- treatment conditions, respectively. It also improved inhibitory memory in the oAβ treated rats. RVG increased the expression level of GluA1 level in the hippocampus. Based on our findings, the expression of RVG in the hippocampus has the potential to enhance both inhibitory and spatial learning abilities, ultimately improving memory performance in an AD rat model. This beneficial effect is likely attributed, at least in part, to the increased expression of GluA1-containing AMPA receptors.
Collapse
Affiliation(s)
- Shayan Aliakbari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Leila Hasanzadeh
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Niloufar Amini
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
6
|
Carrillo JFC, Boaretto AG, Santana DJ, Silva DB. Skin secretions of Leptodactylidae (Anura) and their potential applications. J Venom Anim Toxins Incl Trop Dis 2024; 30:e20230042. [PMID: 38374940 PMCID: PMC10876013 DOI: 10.1590/1678-9199-jvatitd-2023-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 12/04/2023] [Indexed: 02/21/2024] Open
Abstract
The skin of anuran species is a protective barrier against predators and pathogens, showing also chemical defense by substances that represent a potential source for bioactive substances. This review describes the current chemical and biological knowledge from the skin secretions of Leptodactylidae species, one of the most diverse neotropical frog families. These skin secretions reveal a variety of substances such as amines (12), neuropeptides (16), and antimicrobial peptides (72). The amines include histamine and its methylated derivatives, tryptamine derivatives and quaternary amines. The peptides of Leptodactylidae species show molecular weight up to 3364 Da and ocellatins are the most reported. The peptides exhibit commonly glycine (G) or glycine-valine (GV) as C-terminal amino acids, and the most common N-terminal amino acids are glutamic acid (E), lysine (K), and valine (V). The substances from Leptodactylidae species have been evaluated against pathogenic microorganisms, particularly Escherichia coli and Staphylococcus aureus, and the most active peptides showed MIC of 1-15 µM. Furthermore, some compounds showed also pharmacological properties such as immunomodulation, treatment of degenerative diseases, anticancer, and antioxidant. Currently, only 9% of the species in this family have been properly studied, highlighting a large number of unstudied species such as an entire subfamily (Paratelmatobiinae). The ecological context, functions, and evolution of peptides and amines in this family are poorly understood and represent a large field for further exploration.
Collapse
Affiliation(s)
- Juan F. C. Carrillo
- Program in Ecology and Conservation, Institute of Biosciences,
Federal University of Mato Grosso do Sul, Campo Grande, MS, Brazil
- Laboratory of Systematics and Biogeography of Amphibians and
Reptiles (Mapinguari), Institute of Biosciences, Federal University of Mato Grosso
do Sul, Campo Grande, MS, Brazil
- Laboratory of Natural Products and Mass Spectrometry (LaPNEM),
Faculty of Pharmaceutical Sciences, Food and Nutrition (FACFAN), Federal University
of Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Amanda Galdi Boaretto
- Program in Ecology and Conservation, Institute of Biosciences,
Federal University of Mato Grosso do Sul, Campo Grande, MS, Brazil
- Laboratory of Natural Products and Mass Spectrometry (LaPNEM),
Faculty of Pharmaceutical Sciences, Food and Nutrition (FACFAN), Federal University
of Mato Grosso do Sul, Campo Grande, MS, Brazil
| | - Diego J. Santana
- Program in Ecology and Conservation, Institute of Biosciences,
Federal University of Mato Grosso do Sul, Campo Grande, MS, Brazil
- Laboratory of Systematics and Biogeography of Amphibians and
Reptiles (Mapinguari), Institute of Biosciences, Federal University of Mato Grosso
do Sul, Campo Grande, MS, Brazil
| | - Denise Brentan Silva
- Program in Ecology and Conservation, Institute of Biosciences,
Federal University of Mato Grosso do Sul, Campo Grande, MS, Brazil
| |
Collapse
|
7
|
Liu HY, Li X, Wang ZG, Liu SL. Virus-mimicking nanosystems: from design to biomedical applications. Chem Soc Rev 2023; 52:8481-8499. [PMID: 37929845 DOI: 10.1039/d3cs00138e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Nanomedicine, as an interdisciplinary discipline involving the development and application of nanoscale materials and technologies, is rapidly developing under the impetus of bionanotechnology and has attracted a great deal of attention from researchers. Especially, with the global outbreak of COVID-19, the in-depth investigation of the infection mechanism of the viruses has made the study of virus-mimicking nanosystems (VMNs) a popular research topic. In this review, we initiate with a brief historical perspective on the emergence and development of VMNs for providing a comprehensive view of the field. Next, we present emerging design principles and functionalization strategies for fabricating VMNs in light of viral infection mechanisms. Then, we describe recent advances in VMNs in biology, with a major emphasis on representative examples. Finally, we summarize the opportunities and challenges that exist in this field, hoping to provide new insights and inspiration to develop VMNs for disease diagnosis and treatment and to attract the interest of more researchers from different fields.
Collapse
Affiliation(s)
- Hao-Yang Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China.
| | - Xiao Li
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China.
| | - Zhi-Gang Wang
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China.
| | - Shu-Lin Liu
- State Key Laboratory of Medicinal Chemical Biology, Frontiers Science Centre for New Organic Matter, Tianjin Key Laboratory of Biosensing and Molecular Recognition, Research Centre for Analytical Sciences, College of Chemistry, School of Medicine and Frontiers Science Center for Cell Responses, Nankai University, Tianjin 300071, P. R. China.
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, P. R. China
| |
Collapse
|
8
|
Mastraccio KE, Huaman C, Coggins SA, Clouse C, Rader M, Yan L, Mandal P, Hussain I, Ahmed AE, Ho T, Feasley A, Vu BK, Smith IL, Markotter W, Weir DL, Laing ED, Broder CC, Schaefer BC. mAb therapy controls CNS-resident lyssavirus infection via a CD4 T cell-dependent mechanism. EMBO Mol Med 2023; 15:e16394. [PMID: 37767784 PMCID: PMC10565638 DOI: 10.15252/emmm.202216394] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Infections with rabies virus (RABV) and related lyssaviruses are uniformly fatal once virus accesses the central nervous system (CNS) and causes disease signs. Current immunotherapies are thus focused on the early, pre-symptomatic stage of disease, with the goal of peripheral neutralization of virus to prevent CNS infection. Here, we evaluated the therapeutic efficacy of F11, an anti-lyssavirus human monoclonal antibody (mAb), on established lyssavirus infections. We show that a single dose of F11 limits viral load in the brain and reverses disease signs following infection with a lethal dose of lyssavirus, even when administered after initiation of robust virus replication in the CNS. Importantly, we found that F11-dependent neutralization is not sufficient to protect animals from mortality, and a CD4 T cell-dependent adaptive immune response is required for successful control of infection. F11 significantly changes the spectrum of leukocyte populations in the brain, and the FcRγ-binding function of F11 contributes to therapeutic efficacy. Thus, mAb therapy can drive potent neutralization-independent T cell-mediated effects, even against an established CNS infection by a lethal neurotropic virus.
Collapse
Affiliation(s)
- Kate E Mastraccio
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
- Present address:
Wadsworth CenterNew York State Department of HealthAlbanyNYUSA
| | - Celeste Huaman
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
| | - Si'Ana A Coggins
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
| | - Caitlyn Clouse
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
| | - Madeline Rader
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
| | - Lianying Yan
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
| | - Pratyusha Mandal
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
| | - Imran Hussain
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
| | - Anwar E Ahmed
- Department of Preventive Medicine and BiostatisticsUniformed Services UniversityBethesdaMDUSA
| | - Trung Ho
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
| | - Austin Feasley
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc.MDBethesdaUSA
| | - Bang K Vu
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Present address:
Lentigen Technology, Inc.GaithersburgMDUSA
| | - Ina L Smith
- Risk Evaluation and Preparedness Program, Health and BiosecurityCSIROBlack MountainACTAustralia
| | - Wanda Markotter
- Centre for Viral Zoonoses, Department of Medical Virology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
- Centre for Emerging Zoonotic and Parasitic DiseasesNational Institute for Communicable Diseases, National Health Laboratory ServicePretoriaSouth Africa
| | - Dawn L Weir
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
- Present address:
The Center for Bio/Molecular Science and EngineeringU.S. Naval Research LaboratoryWashingtonDCUSA
| | - Eric D Laing
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
| | - Christopher C Broder
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
| | - Brian C Schaefer
- Department of Microbiology and ImmunologyUniformed Services UniversityBethesdaMDUSA
| |
Collapse
|
9
|
Bastos V, Pacheco V, Rodrigues ÉDL, Moraes CNS, Nóbile AL, Fonseca DLM, Souza KBS, do Vale FYN, Filgueiras IS, Schimke LF, Giil LM, Moll G, Cabral-Miranda G, Ochs HD, Vasconcelos PFDC, de Melo GD, Bourhy H, Casseb LMN, Cabral-Marques O. Neuroimmunology of rabies: New insights into an ancient disease. J Med Virol 2023; 95:e29042. [PMID: 37885152 DOI: 10.1002/jmv.29042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 09/28/2023] [Accepted: 09/30/2023] [Indexed: 10/28/2023]
Abstract
Rabies is an ancient neuroinvasive viral (genus Lyssavirus, family Rhabdoviridae) disease affecting approximately 59,000 people worldwide. The central nervous system (CNS) is targeted, and rabies has a case fatality rate of almost 100% in humans and animals. Rabies is entirely preventable through proper vaccination, and thus, the highest incidence is typically observed in developing countries, mainly in Africa and Asia. However, there are still cases in European countries and the United States. Recently, demographic, increasing income levels, and the coronavirus disease 2019 (COVID-19) pandemic have caused a massive raising in the animal population, enhancing the need for preventive measures (e.g., vaccination, surveillance, and animal control programs), postexposure prophylaxis, and a better understanding of rabies pathophysiology to identify therapeutic targets, since there is no effective treatment after the onset of clinical manifestations. Here, we review the neuroimmune biology and mechanisms of rabies. Its pathogenesis involves a complex and poorly understood modulation of immune and brain functions associated with metabolic, synaptic, and neuronal impairments, resulting in fatal outcomes without significant histopathological lesions in the CNS. In this context, the neuroimmunological and neurochemical aspects of excitatory/inhibitory signaling (e.g., GABA/glutamate crosstalk) are likely related to the clinical manifestations of rabies infection. Uncovering new links between immunopathological mechanisms and neurochemical imbalance will be essential to identify novel potential therapeutic targets to reduce rabies morbidity and mortality.
Collapse
Affiliation(s)
- Victor Bastos
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Vinicius Pacheco
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Érika D L Rodrigues
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Cássia N S Moraes
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Adriel L Nóbile
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
| | - Dennyson Leandro M Fonseca
- Interunit Postgraduate Program on Bioinformatics, Institute of Mathematics and Statistics (IME), University of São Paulo, São Paulo, Brazil
| | - Kamilla B S Souza
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Fernando Y N do Vale
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
| | - Igor S Filgueiras
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Lena F Schimke
- Department of Immunology, University of São Paulo, São Paulo, Brazil
| | - Lasse M Giil
- Department of Internal Medicine, Haraldsplass Deaconess Hospital, Bergen, Norway
| | - Guido Moll
- Department of Nephrology and Internal Intensive Care Medicine, Charité University Hospital, Berlin, Germany
| | | | - Hans D Ochs
- School of Medicine and Seattle Children's Research Institute, University of Washington, Seattle, Washington, USA
| | - Pedro F da Costa Vasconcelos
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
- Department of Pathology, University of the State of Pará, Belem, Brazil
| | - Guilherme D de Melo
- Lyssavirus Epidemiology and Neuropathology Unit, WHO Collaborating Centre for Reference and Research on Rabies, Institut Pasteur, Université Paris Cité, Paris, France
| | - Hervé Bourhy
- Lyssavirus Epidemiology and Neuropathology Unit, WHO Collaborating Centre for Reference and Research on Rabies, Institut Pasteur, Université Paris Cité, Paris, France
| | - Livia M N Casseb
- Department of Arbovirology and Hemorrhagic Fevers, PAHO Collaborating Centre for Emerging and Reemerging Arboviruses and other Zoonotic Viruses, Evandro Chagas Institute, Ananindeua, Brazil
| | - Otavio Cabral-Marques
- Department of Pharmaceutical Sciences, Postgraduate Program of Physiopathology and Toxicology, University of São Paulo, São Paulo, Brazil
- Department of Immunology, University of São Paulo, São Paulo, Brazil
- Network of Immunity in Infection, Malignancy, Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), São Paulo, Brazil
- Department of Medicine, Division of Molecular Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
- Laboratory of Medical Investigation 29, School of Medicine, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
10
|
Gowda V, Dinesh S, Sharma S. Manipulative neuroparasites: uncovering the intricacies of neurological host control. Arch Microbiol 2023; 205:314. [PMID: 37603130 DOI: 10.1007/s00203-023-03637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 07/11/2023] [Accepted: 07/13/2023] [Indexed: 08/22/2023]
Abstract
Manipulative neuroparasites are a fascinating group of organisms that possess the ability to hijack the nervous systems of their hosts, manipulating their behavior in order to enhance their own survival and reproductive success. This review provides an overview of the different strategies employed by manipulative neuroparasites, ranging from viruses to parasitic worms and fungi. By examining specific examples, such as Toxoplasma gondii, Leucochloridium paradoxum, and Ophiocordyceps unilateralis, we highlight the complex mechanisms employed by these parasites to manipulate their hosts' behavior. We explore the mechanisms through which these parasites alter the neural processes and behavior of their hosts, including the modulation of neurotransmitters, hormonal pathways, and neural circuits. This review focuses less on the diseases that neuroparasites induce and more on the process of their neurological manipulation. We also investigate the fundamental mechanisms of host manipulation in the developing field of neuroparasitology, which blends neuroscience and parasitology. Finally, understanding the complex interaction between manipulative neuroparasites and their hosts may help us to better understand the fundamentals of behavior, neurology, and host-parasite relationships.
Collapse
Affiliation(s)
- Vishvas Gowda
- Department of Bioinformatics, BioNome, Bangalore, 560043, India
| | - Susha Dinesh
- Department of Bioinformatics, BioNome, Bangalore, 560043, India
| | - Sameer Sharma
- Department of Bioinformatics, BioNome, Bangalore, 560043, India.
| |
Collapse
|
11
|
Wu D, Wang J, Huang C, Zhao J, Fu ZF, Zhao L, Zhou M. Interleukin-1β suppresses rabies virus infection by activating cGAS-STING pathway and compromising the blood-brain barrier integrity in mice. Vet Microbiol 2023; 280:109708. [PMID: 36857805 DOI: 10.1016/j.vetmic.2023.109708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 02/20/2023] [Accepted: 02/24/2023] [Indexed: 02/27/2023]
Abstract
Rabies, caused by rabies virus (RABV), is an ancient zoonotic disease that severely threatens the public health throughout the world. Previous study indicated that interleukin-1β (IL-1β) plays an important role in RABV infection. However, the mechanism how IL-1β affects RABV pathogenicity is still unknown yet. In this study, we confirmed that IL-1β was able to reduce viral titers of RABV in different cells, and the recombinant RABV expressing IL-1β, designated as rCVS-IL1β, could be suppressed in different cells due to the expression of IL-1β. Furthermore, the survival rates of mice infected with rCVS-IL1β by intramuscular route was significantly higher than those of mice infected with parent virus rCVS, which is associated with the less viral loads for entry into the central nervous system (CNS). We further characterized that the cGAS-STING pathway was activated in rCVS-IL1β infected bone marrow derived dendritic cells (BMDC), which could contribute to the decreased viral loads of RABV after intramuscular infection. Moreover, we also observed that the expression of IL-1β by rCVS-IL1β could compromise the blood-brain barrier (BBB) integrity by degrading the tight junction proteins, which allowing peripheral inflammatory cytokines, chemokines, and CD4+T cells to enter into the brain for the clearance of RABV in the CNS. Together, our study suggests that IL-1β could attenuate RABV pathogenicity through activating cGAS-STING pathway in to decrease the viral entry into the CNS and enhance the BBB permeability to promote RABV clearance in the CNS as well, which provides new insight into developing effective therapeutics for rabies.
Collapse
Affiliation(s)
- Di Wu
- Key Laboratory of Preventive Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jinxiao Wang
- Key Laboratory of Preventive Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
| | - Chengli Huang
- Key Laboratory of Preventive Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
| | - Jianqing Zhao
- Key Laboratory of Preventive Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhen F Fu
- Key Laboratory of Preventive Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
| | - Ling Zhao
- Key Laboratory of Preventive Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China.
| | - Ming Zhou
- Key Laboratory of Preventive Veterinary Medicine, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
12
|
Liu Q, He Q, Zhu W. Deoxynivalenol Mycotoxin Inhibits Rabies Virus Replication In Vitro. Int J Mol Sci 2023; 24:ijms24097793. [PMID: 37175500 PMCID: PMC10178062 DOI: 10.3390/ijms24097793] [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: 03/06/2023] [Revised: 03/31/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Rabies is a highly fatal disease, and it is vital to find effective ways to manage and control infection. There is a need for new effective antiviral drugs that are particularly effective treatments for rabies. Deoxynivalenol (DON) is known mainly for its toxicity, but at the molecular level, it can inhibit RNA and DNA replication, and there is increasing evidence that different doses of DON have a positive effect on inhibiting virus replication. Based on this, we evaluated the effect of DON on inhibiting the rabies virus in vitro. The inhibitory effect of DON on rabies virus activity was dose- and time-dependent, and 0.25 μg/mL of DON could inhibit 99% of rabies virus activity within 24 h. Furthermore, DON could inhibit the adsorption, entry, replication, and release of rabies virus but could not inactivate the virus. The inhibitory effect of DON on rabies virus may be achieved by promoting apoptosis. Our study provides a new perspective for the study of anti-rabies virus and expands the direction of action of mycotoxins.
Collapse
Affiliation(s)
- Qian Liu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qing He
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Wuyang Zhu
- NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
13
|
Ren M, Zhou Y, Tu T, Jiang D, Pang M, Li Y, Luo Y, Yao X, Yang Z, Wang Y. RVG Peptide-Functionalized Favipiravir Nanoparticle Delivery System Facilitates Antiviral Therapy of Neurotropic Virus Infection in a Mouse Model. Int J Mol Sci 2023; 24:ijms24065851. [PMID: 36982925 PMCID: PMC10058582 DOI: 10.3390/ijms24065851] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 03/14/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023] Open
Abstract
Neurotropic viruses severely damage the central nervous system (CNS) and human health. Common neurotropic viruses include rabies virus (RABV), Zika virus, and poliovirus. When treating neurotropic virus infection, obstruction of the blood-brain barrier (BBB) reduces the efficiency of drug delivery to the CNS. An efficient intracerebral delivery system can significantly increase intracerebral delivery efficiency and facilitate antiviral therapy. In this study, a rabies virus glycopeptide (RVG) functionalized mesoporous silica nanoparticle (MSN) packaging favipiravir (T-705) was developed to generate T-705@MSN-RVG. It was further evaluated for drug delivery and antiviral treatment in a VSV-infected mouse model. The RVG, a polypeptide consisting of 29 amino acids, was conjugated on the nanoparticle to enhance CNS delivery. The T-705@MSN-RVG caused a significant decrease in virus titers and virus proliferation without inducing substantial cell damage in vitro. By releasing T-705, the nanoparticle promoted viral inhibition in the brain during infection. At 21 days post-infection (dpi), a significantly enhanced survival ratio (77%) was observed in the group inoculated with nanoparticle compared with the non-treated group (23%). The viral RNA levels were also decreased in the therapy group at 4 and 6 dpi compared with that of the control group. The T-705@MSN-RVG could be considered a promising system for CNS delivery for treating neurotropic virus infection.
Collapse
Affiliation(s)
- Meishen Ren
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases (TMBJ), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
- Guangdong-Hong Kong-Macao Greater Bay Area International Research Platform for Aptamer-Based Translational Medicine and Drug Discovery (HKAP), Hong Kong SAR, China
- Institute of Integrated Bioinformedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - You Zhou
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Teng Tu
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Dike Jiang
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Maonan Pang
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yanwei Li
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yan Luo
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Xueping Yao
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Zexiao Yang
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| | - Yin Wang
- Key Laboratory of Animal Diseases and Human Health of Sichuan Province, Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu 611130, China
| |
Collapse
|
14
|
Zhang W, Liu Y, Li M, Zhu J, Li X, Luo TR, Liang J. Host Desmin Interacts with RABV Matrix Protein and Facilitates Virus Propagation. Viruses 2023; 15:v15020434. [PMID: 36851648 PMCID: PMC9964581 DOI: 10.3390/v15020434] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/28/2023] [Accepted: 01/31/2023] [Indexed: 02/09/2023] Open
Abstract
Microfilaments and microtubules, two crucial structures of cytoskeletal networks, are usurped by various viruses for their entry, egress, and/or intracellular trafficking, including the Rabies virus (RABV). Intermediate filaments (IFs) are the third major component of cytoskeletal filaments; however, little is known about the role of IFs during the RABV infection. Here, we identified the IF protein desmin as a novel host interactor with the RABV matrix protein, and we show that this physical interaction has a functional impact on the virus lifecycle. We found that the overexpression of desmin facilitates the RABV infection by increasing the progeny virus yield, and the suppression of endogenous desmin inhibits virus replication. Furthermore, we used confocal microscopy to observe that the RABV-M co-localizes with desmin in IF bundles in the BHK-21 cells. Lastly, we found that mice challenged with RABV displayed an enhanced expression of desmin in the brains of infected animals. These findings reveal a desmin/RABV-M interaction that positively regulates the virus infection and suggests that the RABV may utilize cellular IFs as tracks for the intracellular transport of viral components and efficient budding.
Collapse
Affiliation(s)
- Wen Zhang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Yuming Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Mengru Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Jian Zhu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Xiaoning Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Correspondence: (X.L.); (T.R.L.); (J.L.)
| | - Ting Rong Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Correspondence: (X.L.); (T.R.L.); (J.L.)
| | - Jingjing Liang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- Laboratory of Animal Infectious Diseases, College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Correspondence: (X.L.); (T.R.L.); (J.L.)
| |
Collapse
|
15
|
Embregts CW, Wentzel AS, den Dekker AT, van IJcken WF, Stadhouders R, GeurtsvanKessel CH. Rabies virus uniquely reprograms the transcriptome of human monocyte-derived macrophages. Front Cell Infect Microbiol 2023; 13:1013842. [PMID: 36798087 PMCID: PMC9927221 DOI: 10.3389/fcimb.2023.1013842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
Macrophages are amongst the first immune cells that encounter rabies virus (RABV) at virus entry sites. Activation of macrophages is essential for the onset of a potent immune response, but insights into the effects of RABV on macrophage activation are scarce. In this study we performed high-throughput sequencing on RNA extracted from macrophages that were exposed to RABV for 48 hours, and compared their transcriptional profiles to that of non-polarized macrophages (M0), and macrophages polarized towards the canonical M1, M2a and M2c phenotypes. Our analysis revealed that RABV-stimulated macrophages show high expression of several M1, M2a and M2c signature genes. Apart from their partial resemblance to these phenotypes, unbiased clustering analysis revealed that RABV induces a unique and distinct polarization program. Closer examination revealed that RABV induced multiple pathways related to the interferon- and antiviral response, which were not induced under other classical polarization strategies. Surprisingly, our data show that RABV induces an activated rather than a fully suppressed macrophage phenotype, triggering virus-induced activation and polarization. This includes multiple genes with known antiviral (e.g. APOBEC3A, IFIT/OAS/TRIM genes), which may play a role in anti-RABV immunity.
Collapse
Affiliation(s)
- Carmen W.E. Embregts
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands,*Correspondence: Carmen W.E. Embregts,
| | - Annelieke S. Wentzel
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | | | | | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands,Department of Cell Biology, Erasmus Medical Center, Rotterdam, Netherlands
| | | |
Collapse
|
16
|
Feige L, Kozaki T, Dias de Melo G, Guillemot V, Larrous F, Ginhoux F, Bourhy H. Susceptibilities of CNS Cells towards Rabies Virus Infection Is Linked to Cellular Innate Immune Responses. Viruses 2022; 15:88. [PMID: 36680128 PMCID: PMC9860954 DOI: 10.3390/v15010088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 12/31/2022] Open
Abstract
Rabies is caused by neurotropic rabies virus (RABV), contributing to 60,000 human deaths annually. Even though rabies leads to major public health concerns worldwide, we still do not fully understand factors determining RABV tropism and why glial cells are unable to clear RABV from the infected brain. Here, we compare susceptibilities and immune responses of CNS cell types to infection with two RABV strains, Tha and its attenuated variant Th2P-4M, mutated on phospho- (P-protein) and matrix protein (M-protein). We demonstrate that RABV replicates in human stem cell-derived neurons and astrocytes but fails to infect human iPSC-derived microglia. Additionally, we observed major differences in transcription profiles and quantification of intracellular protein levels between antiviral immune responses mediated by neurons, astrocytes (IFNB1, CCL5, CXCL10, IL1B, IL6, and LIF), and microglia (CCL5, CXCL10, ISG15, MX1, and IL6) upon Tha infection. We also show that P- and M-proteins of Tha mediate evasion of NF-κB- and JAK-STAT-controlled antiviral host responses in neuronal cell types in contrast to glial cells, potentially explaining the strong neuron-specific tropism of RABV. Further, Tha-infected astrocytes and microglia protect neurons from Tha infection via a filtrable and transferable agent. Overall, our study provides novel insights into RABV tropism, showing the interest in studying the interplay of CNS cell types during RABV infection.
Collapse
Affiliation(s)
- Lena Feige
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 75015 Paris, France
| | - Tatsuya Kozaki
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, Immunos Building, Level 3, Singapore 138648, Singapore
| | - Guilherme Dias de Melo
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 75015 Paris, France
| | - Vincent Guillemot
- Hub de Bioinformatique et Biostatistique, Département Biologie Computationnelle, Institut Pasteur, 75015 Paris, France
| | - Florence Larrous
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 75015 Paris, France
| | - Florent Ginhoux
- Singapore Immunology Network, Agency for Science, Technology and Research, 8A Biomedical Grove, Immunos Building, Level 3, Singapore 138648, Singapore
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, 280 South Chongqing Road, Shanghai 200025, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Center, 20 College Road, Discovery Tower Level 8, Singapore 169856, Singapore
- Inserm U1015, Gustave Roussy, Bâtiment de Médecine Moléculaire, 114 Rue Edouard Vaillant, 94800 Villejuif, France
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 75015 Paris, France
| |
Collapse
|
17
|
Ghassemi S, Asgari T, Mirzapour-Delavar H, Aliakbari S, Pourbadie HG, Prehaud C, Lafon M, Gholami A, Azadmanesh K, Naderi N, Sayyah M. Lentiviral Expression of Rabies Virus Glycoprotein in the Rat Hippocampus Strengthens Synaptic Plasticity. Cell Mol Neurobiol 2022; 42:1429-1440. [PMID: 33462779 PMCID: PMC11421698 DOI: 10.1007/s10571-020-01032-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 12/16/2020] [Indexed: 11/30/2022]
Abstract
Rabies virus (RABV) is a neurotropic virus exclusively infecting neurons in the central nervous system. RABV encodes five proteins. Among them, the viral glycoprotein (RVG) plays a key role in viral entry into neurons and rabies pathogenesis. It was shown that the nature of the C-terminus of the RABV G protein, which possesses a PDZ-binding motif (PBM), modulates the virulence of the RABV strain. The neuronal protein partners recruited by this PBM may alter host cell function. This study was conducted to investigate the effect of RVG on synaptic function in the hippocampal dentate gyrus (DG) of rat. Two μl (108 T.U./ml) of the lentiviral vector containing RVG gene was injected into the DG of rat hippocampus. After 2 weeks, the rat's brain was cross-sectioned and RVG-expressing cells were detected by fluorescent microscopy. Hippocampal synaptic activity of the infected rats was then examined by recording the local field potentials from DG after stimulation of the perforant pathway. Short-term synaptic plasticity was also assessed by double pulse stimulation. Expression of RVG in DG increased long-term potentiation population spikes (LTP-PS), whereas no facilitation of LTP-PS was found in neurons expressing δRVG (deleted PBM). Furthermore, RVG and δRVG strengthened paired-pulse facilitation. Heterosynaptic long-term depression (LTD) in the DG was significantly blocked in RVG-expressing group compared to the control group. This blockade was dependent to PBM motif as rats expressing δRVG in the DG-expressed LTD comparable to the RVG group. Our data demonstrate that RVG expression facilitates both short- and long-term synaptic plasticity in the DG indicating that it may involve both pre- and postsynaptic mechanisms to alter synaptic function. Further studies are needed to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Soheil Ghassemi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Tara Asgari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Shayan Aliakbari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Christophe Prehaud
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris, France
| | - Monique Lafon
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris, France
| | - Alireza Gholami
- WHO Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | | | - Nima Naderi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
18
|
Role of the glycoprotein thorns in anxious effects of rabies virus: Evidence from an animal study. Brain Res Bull 2022; 185:107-116. [DOI: 10.1016/j.brainresbull.2022.05.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 04/11/2022] [Accepted: 05/03/2022] [Indexed: 12/17/2022]
|
19
|
Feige L, Sáenz-de-Santa-María I, Regnault B, Lavenir R, Lepelletier A, Halacu A, Rajerison R, Diop S, Nareth C, Reynes JM, Buchy P, Bourhy H, Dacheux L. Transcriptome Profile During Rabies Virus Infection: Identification of Human CXCL16 as a Potential New Viral Target. Front Cell Infect Microbiol 2021; 11:761074. [PMID: 34804996 PMCID: PMC8602097 DOI: 10.3389/fcimb.2021.761074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022] Open
Abstract
Rabies virus (RABV), the causative agent for rabies disease is still presenting a major public health concern causing approximately 60,000 deaths annually. This neurotropic virus (genus Lyssavirus, family Rhabdoviridae) induces an acute and almost always fatal form of encephalomyelitis in humans. Despite the lethal consequences associated with clinical symptoms of rabies, RABV limits neuro-inflammation without causing major histopathological lesions in humans. Nevertheless, information about the mechanisms of infection and cellular response in the central nervous system (CNS) remain scarce. Here, we investigated the expression of inflammatory genes involved in immune response to RABV (dog-adapted strain Tha) in mice, the most common animal model used to study rabies. To better elucidate the pathophysiological mechanisms during natural RABV infection, we compared the inflammatory transcriptome profile observed at the late stage of infection in the mouse brain (cortex and brain stem/cerebellum) with the ortholog gene expression in post-mortem brain biopsies of rabid patients. Our data indicate that the inflammatory response associated with rabies is more pronounced in the murine brain compared to the human brain. In contrast to murine transcription profiles, we identified CXC motif chemokine ligand 16 (CXCL16) as the only significant differentially expressed gene in post-mortem brains of rabid patients. This result was confirmed in vitro, in which Tha suppressed interferon alpha (IFN-α)-induced CXCL16 expression in human CNS cell lines but induced CXCL16 expression in IFN-α-stimulated murine astrocytes. We hypothesize that RABV-induced modulation of the CXCL16 pathway in the brain possibly affects neurotransmission, natural killer (NK) and T cell recruitment and activation. Overall, we show species-specific differences in the inflammatory response of the brain, highlighted the importance of understanding the potential limitations of extrapolating data from animal models to humans.
Collapse
Affiliation(s)
- Lena Feige
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | | | | | - Rachel Lavenir
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | - Anthony Lepelletier
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | - Ala Halacu
- National Agency for Public Health, Chișinău, Moldova
| | | | - Sylvie Diop
- Infectious Diseases Department, National and University Hospital Center of Fann-Dakar, Dakar, Senegal
| | | | - Jean-Marc Reynes
- Virology Unit, Institut Pasteur de Madagascar, Tananarive, Madagascar
| | - Philippe Buchy
- Virology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | - Laurent Dacheux
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| |
Collapse
|
20
|
Ren M, Wang Y, Luo Y, Yao X, Yang Z, Zhang P, Zhao W, Jiang D. Functionalized Nanoparticles in Prevention and Targeted Therapy of Viral Diseases With Neurotropism Properties, Special Insight on COVID-19. Front Microbiol 2021; 12:767104. [PMID: 34867899 PMCID: PMC8634613 DOI: 10.3389/fmicb.2021.767104] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 12/15/2022] Open
Abstract
Neurotropic viruses have neural-invasive and neurovirulent properties to damage the central nervous system (CNS), leading to humans' fatal symptoms. Neurotropic viruses comprise a lot of viruses, such as Zika virus (ZIKV), herpes simplex virus (HSV), rabies virus (RABV), and severe acute respiratory syndrome coronavirus type 2 (SARS-CoV-2). Effective therapy is needed to prevent infection by these viruses in vivo and in vitro. However, the blood-brain barrier (BBB) usually prevents macromolecules from entering the CNS, which challenges the usage of the traditional probes, antiviral drugs, or neutralizing antibodies in the CNS. Functionalized nanoparticles (NPs) have been increasingly reported in the targeted therapy of neurotropic viruses due to their sensitivity and targeting characteristics. Therefore, the present review outlines efficient functionalized NPs to further understand the recent trends, challenges, and prospects of these materials.
Collapse
Affiliation(s)
| | - Yin Wang
- Animal Quarantine Laboratory, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Beier KT. The Serendipity of Viral Trans-Neuronal Specificity: More Than Meets the Eye. Front Cell Neurosci 2021; 15:720807. [PMID: 34671244 PMCID: PMC8521040 DOI: 10.3389/fncel.2021.720807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Accepted: 09/07/2021] [Indexed: 12/25/2022] Open
Abstract
Trans-neuronal viruses are frequently used as neuroanatomical tools for mapping neuronal circuits. Specifically, recombinant one-step rabies viruses (RABV) have been instrumental in the widespread application of viral circuit mapping, as these viruses have enabled labs to map the direct inputs onto defined cell populations. Within the neuroscience community, it is widely believed that RABV spreads directly between neurons via synaptic connections, a hypothesis based principally on two observations. First, the virus labels neurons in a pattern consistent with known anatomical connectivity. Second, few glial cells appear to be infected following RABV injections, despite the fact that glial cells are abundant in the brain. However, there is no direct evidence that RABV can actually be transmitted through synaptic connections. Here we review the immunosubversive mechanisms that are critical to RABV’s success for infiltration of the central nervous system (CNS). These include interfering with and ultimately killing migratory T cells while maintaining levels of interferon (IFN) signaling in the brain parenchyma. Finally, we critically evaluate studies that support or are against synaptically-restricted RABV transmission and the implications of viral-host immune responses for RABV transmission in the brain.
Collapse
Affiliation(s)
- Kevin Thomas Beier
- Department of Physiology & Biophysics, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
22
|
Zhao P, Hou K, Zhong Z, Guo S, Yang S, Xia X. Quantitative characterization of the T cell receptor repertoires of human immunized by rabies virus vaccine. Hum Vaccin Immunother 2021; 17:2530-2537. [PMID: 33823121 PMCID: PMC8475554 DOI: 10.1080/21645515.2021.1893575] [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: 07/25/2020] [Revised: 02/03/2021] [Accepted: 02/16/2021] [Indexed: 02/05/2023] Open
Abstract
Cellular immunity is crucial for an efficient host immune response against rabies virus (RABV) infection. But the T cell receptor (TCR) repertoire in human after RABV vaccine immunization remained unclear. In this study, we conducted high-throughput sequencing of TCR β chain complementarity determining region 3(CDR3) repertoires in 4 healthy volunteers before and after immunization with RABV vaccine. Our data showed that RABV vaccination changed the TCR diversity and the usage of V/J gene segments, as well as V-J pairing. The high-frequency clonotypes that altered after vaccination were identified. These results may provide us with new insights into T cell receptor condition after RABV vaccination.
Collapse
Affiliation(s)
- Pingsen Zhao
- Department of Laboratory Medicine, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
- Shaoguan Municipal Quality Control Center for Laboratory Medicine, Yuebei People’s Hospital, Shaoguan, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
- CONTACT Pingsen Zhao ; Head & Professor, Department of Laboratory Medicine, Yuebei People’s Hospital, Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan512025, P. R. China
| | - Kaijian Hou
- Department of Endocrine and Metabolic Diseases, Longhu Hospital, The First Affiliated Hospital of Shantou University Medical College, Shantou, China
| | - Zhixiong Zhong
- Center for Precision Medicine, Meizhou People’s Hospital, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China
| | - Sharula Guo
- Department of Infection Control, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Songtao Yang
- Academy of Military Medical Sciences, Institute of Military Veterinary, Changchun, China
| | - Xianzhu Xia
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
- Academy of Military Medical Sciences, Institute of Military Veterinary, Changchun, China
| |
Collapse
|
23
|
Zhao P, Guo S, Zhong Z, Yang S, Xia X. Quantitative characterization of the B cell receptor repertoires of human immunized with commercial rabies virus vaccine. Hum Vaccin Immunother 2021; 17:2538-2546. [PMID: 34559619 PMCID: PMC8475592 DOI: 10.1080/21645515.2021.1893576] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 02/03/2021] [Accepted: 02/16/2021] [Indexed: 02/05/2023] Open
Abstract
Humoral immunity is crucial for an efficient host immune response against rabies virus (RABV) infection. But the B cell receptor (BCR) repertoire in human after RABV vaccine immunization remained unclear. To study the BCR repertoires in peripheral blood mononuclear cells (PBMCs) of human immunized with rabies virus vaccine. In this study, we conducted BCR complementarity determining region 3 (CDR3) repertoires in 4 healthy volunteers before and after immunization with RABV vaccine by high-throughput sequencing. The bioinformatics analysis process was performed. The results showed that RABV vaccination changed the BCR diversity and the usage of V/J gene segments, as well as V-J pairing. B cell clone expansion was induced by the vaccination and sequences of high expand CDR3 aa clones were identified. To the best of our knowledge, we firstly quantitative characterized B cell receptor repertoire of human immunized with c rabies virus vaccine. It might provide us with new insights into B cell receptor condition after RABV vaccination.
Collapse
Affiliation(s)
- Pingsen Zhao
- Department of Laboratory Medicine, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
- Yuebei People’s Hospital, Shaoguan Municipal Quality Control Center for Laboratory Medicine, Shaoguan, China
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
- CONTACT Pingsen Zhao ; Department of Laboratory Medicine, Yuebei People’s Hospital, Shantou University Medical College, No 133, Huimin Road South, Wujiang District, Shaoguan512025, P. R. China
| | - Sharula Guo
- Department of Infection Control, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
| | - Zhixiong Zhong
- Center for Precision Medicine, Meizhou People’s Hospital, Meizhou Hospital Affiliated to Sun Yat-sen University, Meizhou, China
| | - Songtao Yang
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| | - Xianzhu Xia
- Laboratory for Diagnosis of Clinical Microbiology and Infection, Yuebei People’s Hospital, Shantou University Medical College, Shaoguan, China
- Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, China
| |
Collapse
|
24
|
Farahtaj F, Gholami A, Khosravy MS, Gharibzadeh S, Niknam HM, Ghaemi A. Enhancement of immune responses by co-stimulation of TLR3 - TLR7 agonists as a potential therapeutics against rabies in mouse model. Microb Pathog 2021; 157:104971. [PMID: 34029660 DOI: 10.1016/j.micpath.2021.104971] [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: 10/06/2020] [Revised: 04/18/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
Rabies is always fatal, when post-exposure prophylaxis is administered after the onset of clinical symptoms. To date, there is no effective treatment of rabies once clinical symptoms has initiated. Therefore, we aimed to provide evidences which indicate the promising effects of combination treatment with TLR agonists following rabies infection. Four groups of rabies infected-mice (10-mice/group) were treated with PolyI:C 50 μg (a TLR3 agonist), Imiquimod50 μg (a TLR7 agonist), (Poly + Imi)25 μg and (Poly + Imi)50 μg respectively. The immune responses in each experimental groups were investigated in the brain through evaluation of GFAP, MAP2, CD4, HSP70, TLR3, TLR7 and apoptotic cell expression as well as determination of IFN-γ, TNF-α and IL-4, levels. The treatment with combination of agonists (Poly + Imi)50 μg/mouse resulted a 75% decrease of mortality rate and better extended survival time following street rabies virus infection. Higher number of CD4+T cells, TLR3 and TLR7 expression in the brain parenchyma observed in the groups receiving both combined agonist therapies at the levels of 25 μg and 50 μg. In spite of decreased number of neuronal cell, significant higher number of astrocytes was shown in the group given (Poly + Imi)25 μg. The obtained results also pointed to the dramatic decrease of HSP70 expression in all groups of infected mice whereas higher number of apoptotic cells and Caspase 8 expression were recorded in (Poly + Imi)25 μg treated group. Furthermore, the cytokine profile consisting the increased levels of TNF-α, IFN-γ and IL-4 revealed that both humoral and cellular responses were highly modulated in combination therapy of 50 μg of Imiquimod and Poly I:C. Reduced viral load as quantified by real-time PCR of rabies N gene expression in the brain also correlated with the better survival of agonist-treated groups of mice. Based on obtained results, we have presented evidences of beneficial utilization of combined agonist therapy composed of TLR3/TLR7 ligands. This treatment regimen extended survival of infected mice and decreased significantly their mortality rate. We believe that the results of synergy-inducing protection of both TLR3/TLR7 agonists lead to the enhancement of innate immune responses cells residing in the CNS which warrant the studies to further understanding of crosstalk mechanisms in cellular immunity against rabies in the future.
Collapse
Affiliation(s)
- Firouzeh Farahtaj
- Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Gholami
- Viral Vaccine Production, Pasteur Institute of Iran, Karaj, Iran
| | | | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics, Research Center for Emerging and Reemerging of Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | | | - Amir Ghaemi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
25
|
Rabies virus glycoprotein enhances spatial memory via the PDZ binding motif. J Neurovirol 2021; 27:434-443. [PMID: 33788140 DOI: 10.1007/s13365-021-00972-2] [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: 07/06/2020] [Revised: 03/09/2021] [Accepted: 03/17/2021] [Indexed: 10/21/2022]
Abstract
Rabies is a life-threatening viral infection of the brain. Rabies virus (RABV) merely infects excitable cells including neurons provoking drastic behaviors including negative emotional memories. RABV glycoprotein (RVG) plays a critical role in RABV pathogenesis. RVG interacts with various cytoplasmic PDZ (PSD-95/Dlg/ZO-1) containing proteins through its PDZ binding motif (PBM). PTZ domains have crucial role in formation and function of signal transduction. Hippocampus is one of the cerebral regions that contain high load of viral antigens. We examined impact of RVG expression in the dorsal hippocampus on aversive as well as spatial learning and memory performance in rats. Two microliter of the lentiviral vector (~108 T.U./ml) encoding RVG or ∆RVG (deleted PBM) genomes was microinjected into the hippocampal CA1. After 1 week, rat's brain was cross-sectioned and RVG/∆RVG-expressing neuronal cells were confirmed by fluorescent microscopy. Passive avoidance and spatial learning and memory were assessed in rats by Shuttle box and Morris water maze (MWM). In the shuttle box, both RVG and ∆RVG decreased the time spent in the dark compartment compared to control (p < 0.05). In MWM, RVG and ∆RVG did not affect the acquisition of spatial task. In the probe test, RVG-expressing rats spent more time in the target quadrant, and also reached the platform position sooner than control group (p < 0.05). Rats expressing ∆RVG significantly swam farther from the hidden platform than RVG group (p < 0.05). Our data indicate RVG expression in the hippocampus strengthens aversive and spatial learning and memory performance. The boosting effect on spatial but not avoidance memory is mediated through PBM.
Collapse
|
26
|
Sui B, Chen D, Liu W, Tian B, Lv L, Pei J, Wu Q, Zhou M, Fu ZF, Zhang Y, Zhao L. Comparison of lncRNA and mRNA expression in mouse brains infected by a wild-type and a lab-attenuated Rabies lyssavirus. J Gen Virol 2020; 102. [PMID: 33284098 DOI: 10.1099/jgv.0.001538] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rabies is a lethal disease caused by Rabies lyssavirus, commonly known as rabies virus (RABV), and results in nearly 100 % death once clinical symptoms occur in human and animals. Long non-coding RNAs (lncRNAs) have been reported to be associated with viral infection. But the role of lncRNAs involved in RABV infection is still elusive. In this study, we performed global transcriptome analysis of both of lncRNA and mRNA expression profiles in wild-type (WT) and lab-attenuated RABV-infected mouse brains by using next-generation sequencing. The differentially expressed lncRNAs and mRNAs were analysed by using the edgeR package. We identified 1422 differentially expressed lncRNAs and 4475 differentially expressed mRNAs by comparing WT and lab-attenuated RABV-infected brains. Then we predicted the enriched biological pathways by the Gene Ontology (GO) and Kyoto Encyclopaedia of Genes and Genomes (KEGG) database based on the differentially expressed lncRNAs and mRNAs. Our analysis revealed the relationships between lncRNAs and RABV-infection-associated immune response and ion transport-related pathways, which provide a fresh insight into the potential role of lncRNA in immune evasion and neuron injury induced by WT RABV.
Collapse
Affiliation(s)
- Baokun Sui
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Dong Chen
- ABLife BioBigData Institute, Wuhan, 430075, PR China
| | - Wei Liu
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Bin Tian
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Lei Lv
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Jie Pei
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Qiong Wu
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Ming Zhou
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Zhen F Fu
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| | - Yi Zhang
- ABLife BioBigData Institute, Wuhan, 430075, PR China
| | - Ling Zhao
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, 430070, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, 430070, PR China
| |
Collapse
|
27
|
Wang C, Liu H, Li K, Wu ZZ, Wu C, Yu JY, Gong Q, Fang P, Wang XX, Duan SM, Wang H, Gu Y, Hu J, Pan BX, Schmidt MV, Liu YJ, Wang XD. Tactile modulation of memory and anxiety requires dentate granule cells along the dorsoventral axis. Nat Commun 2020; 11:6045. [PMID: 33247136 PMCID: PMC7695841 DOI: 10.1038/s41467-020-19874-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 11/02/2020] [Indexed: 12/21/2022] Open
Abstract
Touch can positively influence cognition and emotion, but the underlying mechanisms remain unclear. Here, we report that tactile experience enrichment improves memory and alleviates anxiety by remodeling neurons along the dorsoventral axis of the dentate gyrus (DG) in adult mice. Tactile enrichment induces differential activation and structural modification of neurons in the dorsal and ventral DG, and increases the presynaptic input from the lateral entorhinal cortex (LEC), which is reciprocally connected with the primary somatosensory cortex (S1), to tactile experience-activated DG neurons. Chemogenetic activation of tactile experience-tagged dorsal and ventral DG neurons enhances memory and reduces anxiety respectively, whereas inactivation of these neurons or S1-innervated LEC neurons abolishes the beneficial effects of tactile enrichment. Moreover, adulthood tactile enrichment attenuates early-life stress-induced memory deficits and anxiety-related behavior. Our findings demonstrate that enriched tactile experience retunes the pathway from S1 to DG and enhances DG neuronal plasticity to modulate cognition and emotion. Touch can positively modulate cognitive performance and emotional response. Here the authors demonstrate that enriched tactile experience improves memory and reduces anxiety in adult mice by remodelling the pathway from the primary somatosensory cortex to the dentate gyrus.
Collapse
Affiliation(s)
- Chi Wang
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Hui Liu
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Kun Li
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Zhen-Zhen Wu
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Chen Wu
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China.,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Jing-Ying Yu
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Qian Gong
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Ping Fang
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Xing-Xing Wang
- Department of Anesthesiology, Technische Universität München/Klinikum Rechts der Isar, 81675, Munich, Germany
| | - Shu-Min Duan
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Hao Wang
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, 310058, Hangzhou, China
| | - Yan Gu
- Center of Stem Cell and Regenerative Medicine, and Department of Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Ji Hu
- School of Life Science and Technology, ShanghaiTech University, 201210, Shanghai, China
| | - Bing-Xing Pan
- Laboratory of Fear and Anxiety Disorders, Institute of Life Science, Nanchang University, 330031, Nanchang, China
| | | | - Yi-Jun Liu
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China
| | - Xiao-Dong Wang
- Department of Neurobiology and Department of Psychiatry of Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310058, Hangzhou, China. .,NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, 310058, Hangzhou, China.
| |
Collapse
|
28
|
Rabies virus infection in mice up-regulates B7-H1 via epigenetic modifications. Virusdisease 2020; 31:388-394. [DOI: 10.1007/s13337-020-00588-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 04/24/2020] [Indexed: 10/24/2022] Open
|
29
|
Castro BS, Guedes F, Fernandes ER, Koike G, Katz ISS, Chaves LB, Silva SR. Development of biotinylated polyclonal anti-ribonucleoprotein IgG for detection of rabies virus antigen by direct rapid immunohistochemical test. Biologicals 2020; 68:74-78. [PMID: 32859463 DOI: 10.1016/j.biologicals.2020.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/22/2020] [Accepted: 08/07/2020] [Indexed: 11/30/2022] Open
Abstract
The direct rapid immunohistochemical test (dRIT) has been recommended for laboratorial diagnosis of rabies, especially in developing countries. The absence of commercial primary antibodies, however, still represents a major limitation to its wider use in testing. We describe here the development of a biotinylated polyclonal antibody against Rabies lyssavirus (RABV) ribonucleoprotein (RNP) and its use as a primary reagent in dRIT. Anti-RNP polyclonal horse IgG was purified by ionic exchange chromatography followed by immunoaffinity column chromatography, and its affinity, diagnostic sensitivity, and specificity were evaluated. CNS samples (120) of suspected rabies cases in different animal species were tested by dRIT, with the positive (n = 14) and negative (n = 106) results confirmed by direct fluorescence antibody testing (dFAT). Comparing the results of dRIT and dFAT, we found that the biotinylated anti-RNP IgG delivered 100% diagnostic specificity and sensibility for rabies diagnosis. Our findings show that the biotinylated anti-RNP polyclonal IgG can be produced with the quality required for application in dRIT. This work represents an important step in efforts to diagnose rabies in developing countries.
Collapse
|
30
|
Sundaramoorthy V, Godde N, J. Farr R, Green D, M. Haynes J, Bingham J, O’Brien CM, Dearnley M. Modelling Lyssavirus Infections in Human Stem Cell-Derived Neural Cultures. Viruses 2020; 12:E359. [PMID: 32218146 PMCID: PMC7232326 DOI: 10.3390/v12040359] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/02/2020] [Accepted: 03/20/2020] [Indexed: 12/20/2022] Open
Abstract
Rabies is a zoonotic neurological infection caused by lyssavirus that continues to result in devastating loss of human life. Many aspects of rabies pathogenesis in human neurons are not well understood. Lack of appropriate ex-vivo models for studying rabies infection in human neurons has contributed to this knowledge gap. In this study, we utilize advances in stem cell technology to characterize rabies infection in human stem cell-derived neurons. We show key cellular features of rabies infection in our human neural cultures, including upregulation of inflammatory chemokines, lack of neuronal apoptosis, and axonal transmission of viruses in neuronal networks. In addition, we highlight specific differences in cellular pathogenesis between laboratory-adapted and field strain lyssavirus. This study therefore defines the first stem cell-derived ex-vivo model system to study rabies pathogenesis in human neurons. This new model system demonstrates the potential for enabling an increased understanding of molecular mechanisms in human rabies, which could lead to improved control methods.
Collapse
Affiliation(s)
- Vinod Sundaramoorthy
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Australian Animal Health Laboratory (AAHL), East Geelong, VIC 3219, Australia; (V.S.); (N.G.); (R.J.F.); (D.G.); (J.B.)
| | - Nathan Godde
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Australian Animal Health Laboratory (AAHL), East Geelong, VIC 3219, Australia; (V.S.); (N.G.); (R.J.F.); (D.G.); (J.B.)
| | - Ryan J. Farr
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Australian Animal Health Laboratory (AAHL), East Geelong, VIC 3219, Australia; (V.S.); (N.G.); (R.J.F.); (D.G.); (J.B.)
| | - Diane Green
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Australian Animal Health Laboratory (AAHL), East Geelong, VIC 3219, Australia; (V.S.); (N.G.); (R.J.F.); (D.G.); (J.B.)
| | - John M. Haynes
- Monash Institute of Pharmaceutical Sciences, 399 Royal Parade, Parkville, VIC 3052, Australia;
| | - John Bingham
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Australian Animal Health Laboratory (AAHL), East Geelong, VIC 3219, Australia; (V.S.); (N.G.); (R.J.F.); (D.G.); (J.B.)
| | - Carmel M. O’Brien
- CSIRO Manufacturing, Research Way, Clayton, VIC 3168, Australia
- Australian Regenerative Medicine Institute, Monash University, Clayton, VIC 3168, Australia
| | - Megan Dearnley
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Australian Animal Health Laboratory (AAHL), East Geelong, VIC 3219, Australia; (V.S.); (N.G.); (R.J.F.); (D.G.); (J.B.)
| |
Collapse
|
31
|
Yang F, Lin S, Ye F, Yang J, Qi J, Chen Z, Lin X, Wang J, Yue D, Cheng Y, Chen Z, Chen H, You Y, Zhang Z, Yang Y, Yang M, Sun H, Li Y, Cao Y, Yang S, Wei Y, Gao GF, Lu G. Structural Analysis of Rabies Virus Glycoprotein Reveals pH-Dependent Conformational Changes and Interactions with a Neutralizing Antibody. Cell Host Microbe 2020; 27:441-453.e7. [DOI: 10.1016/j.chom.2019.12.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/06/2019] [Accepted: 12/30/2019] [Indexed: 12/21/2022]
|
32
|
A 1-week intradermal dose-sparing regimen for rabies post-exposure prophylaxis (RESIST-2): an observational cohort study. THE LANCET. INFECTIOUS DISEASES 2019; 19:1355-1362. [PMID: 31570311 DOI: 10.1016/s1473-3099(19)30311-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Revised: 04/30/2019] [Accepted: 06/05/2019] [Indexed: 11/23/2022]
Abstract
BACKGROUND The international health authorities are backing an effort to eliminate canine-mediated rabies in humans by 2030. This effort will require improving access to adequate and timely rabies post-exposure prophylaxis as compliance is low with WHO-recommended regimens (given in four to five visits over 1 month). Access could be substantially improved by an abridged regimen to reduce doses, direct and indirect costs, and improve vaccine equity by better sharing of available vaccine. We aimed to compare rabies virus neutralising antibody titres before and after the fourth visit to determine whether that session was needed or the current regimen could be abridged. METHODS In this observational cohort study, we measured rabies virus neutralising antibody titres using rapid fluorescent focus inhibition tests in 116 people bitten by dogs with laboratory-confirmed rabies and 20 control individuals. Percentages of circulating plasmablasts were determined by flow cytometry. All individuals had been referred to the rabies prevention clinic at Institut Pasteur in Cambodia and received two intradermal injections of post-exposure prophylaxis on days 0, 3, 7, and 28 (Thai Red Cross regimen) with or without equine rabies immunoglobulin, as per 2010 WHO recommendations. FINDINGS All individuals had rabies virus neutralising antibody titres considered protective (≥0·5 IU/mL) and plasmablast activation on day 28 before the last injection. The median rabies virus neutralising antibody concentration in the group of individuals bitten by rabies virus-positive dogs was 1·08 IU/mL (IQR 0·37-3·09) on day 7, 26·86 (22·68-49·50) on day 28, and 26·74 (11·78-49·06) on day 42. No significant differences were observed in titres between days 28 and 42, after titres reached a plateau. These titres were reached notwithstanding equine rabies immunoglobulin use, age, sex, nutrition status as indicated by upper-arm circumference in children or BMI in adults, or dog infection status. Titres or plasmablast percentages did not increase between the day of the last injection and 2 weeks later. All patients were alive 1 year after post-exposure prophylaxis. INTERPRETATION The fourth vaccine session on day 28 provides no additional benefit. Rabies post-exposure prophylaxis can be abridged to a two-dose, three-session, 1 week regimen to improve post-exposure prophylaxis coverage and equity at no risk to patients. FUNDING Institut Pasteur.
Collapse
|
33
|
Mannitol treatment is not effective in therapy of rabies virus infection in mice. Vaccine 2019; 37:4710-4714. [DOI: 10.1016/j.vaccine.2017.12.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 11/29/2017] [Accepted: 12/11/2017] [Indexed: 11/23/2022]
|
34
|
Khan Z, Terrien E, Delhommel F, Lefebvre-Omar C, Bohl D, Vitry S, Bernard C, Ramirez J, Chaffotte A, Ricquier K, Vincentelli R, Buc H, Prehaud C, Wolff N, Lafon M. Structure-based optimization of a PDZ-binding motif within a viral peptide stimulates neurite outgrowth. J Biol Chem 2019; 294:13755-13768. [PMID: 31346033 DOI: 10.1074/jbc.ra119.008238] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/11/2019] [Indexed: 11/06/2022] Open
Abstract
Protection of neuronal homeostasis is a major goal in the management of neurodegenerative diseases. Microtubule-associated Ser/Thr kinase 2 (MAST2) inhibits neurite outgrowth, and its inhibition therefore represents a potential therapeutic strategy. We previously reported that a viral protein (G-protein from rabies virus) capable of interfering with protein-protein interactions between the PDZ domain of MAST2 and the C-terminal moieties of its cellular partners counteracts MAST2-mediated suppression of neurite outgrowth. Here, we designed peptides derived from the native viral protein to increase the affinity of these peptides for the MAST2-PDZ domain. Our strategy involved modifying the length and flexibility of the noninteracting sequence linking the two subsites anchoring the peptide to the PDZ domain. Three peptides, Neurovita1 (NV1), NV2, and NV3, were selected, and we found that they all had increased affinities for the MAST2-PDZ domain, with Kd values decreasing from 1300 to 60 nm, while target selectivity was maintained. A parallel biological assay evaluating neurite extension and branching in cell cultures revealed that the NV peptides gradually improved neural activity, with the efficacies of these peptides for stimulating neurite outgrowth mirroring their affinities for MAST2-PDZ. We also show that NVs can be delivered into the cytoplasm of neurons as a gene or peptide. In summary, our findings indicate that virus-derived peptides targeted to MAST2-PDZ stimulate neurite outgrowth in several neuron types, opening up promising avenues for potentially using NVs in the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Zakir Khan
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris 75015, France
| | - Elouan Terrien
- Institut Pasteur, Unité de RMN des Biomolécules, UMR 3528, CNRS, Paris 75015, France
| | - Florent Delhommel
- Institut Pasteur, Unité de RMN des Biomolécules, UMR 3528, CNRS, Paris 75015, France
| | - Cynthia Lefebvre-Omar
- Institut du Cerveau et de la Moelle Epinière, ICM, U1127 INSERM, UMR 7225 CNRS, Sorbonne Université, Paris 75013, France
| | - Delphine Bohl
- Institut du Cerveau et de la Moelle Epinière, ICM, U1127 INSERM, UMR 7225 CNRS, Sorbonne Université, Paris 75013, France
| | - Sandrine Vitry
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris 75015, France
| | - Clara Bernard
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris 75015, France
| | - Juan Ramirez
- Institut Pasteur, Unité de RMN des Biomolécules, UMR 3528, CNRS, Paris 75015, France
| | - Alain Chaffotte
- Institut Pasteur, Unité de RMN des Biomolécules, UMR 3528, CNRS, Paris 75015, France
| | - Kevin Ricquier
- Institut Pasteur, Unité de RMN des Biomolécules, UMR 3528, CNRS, Paris 75015, France
| | - Renaud Vincentelli
- Unité Mixte de Recherche 7257, CNRS Aix-Marseille Université, Architecture et Fonction des Macromolécules Biologiques (AFMB), Marseille 13009, France
| | - Henri Buc
- Institut Pasteur, Paris 75015, France
| | - Christophe Prehaud
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris 75015, France
| | - Nicolas Wolff
- Institut Pasteur, Unité de RMN des Biomolécules, UMR 3528, CNRS, Paris 75015, France
| | - Monique Lafon
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris 75015, France
| |
Collapse
|
35
|
Status of antiviral therapeutics against rabies virus and related emerging lyssaviruses. Curr Opin Virol 2019; 35:1-13. [PMID: 30753961 DOI: 10.1016/j.coviro.2018.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/19/2022]
Abstract
Rabies virus (RABV) constitutes a major social and economic burden associated with 60 000 deaths annually worldwide. Although pre-exposure and post-exposure treatment options are available, they are efficacious only when initiated before the onset of clinical symptoms. Aggravating the problem, the current RABV vaccine does not cross-protect against the emerging zoonotic phylogroup II lyssaviruses. A requirement for an uninterrupted cold chain and high cost of the immunoglobulin component of rabies prophylaxis generate an unmet need for the development of RABV-specific antivirals. We discuss desirable anti-RABV drug profiles, past efforts to address the problem and inhibitor candidates identified, and examine how the rapidly expanding structural insight into RABV protein organization has illuminated novel druggable target candidates and paved the way to structure-aided drug optimization. Special emphasis is given to the viral RNA-dependent RNA polymerase complex as a promising target for direct-acting broad-spectrum RABV inhibitors.
Collapse
|
36
|
Inhibition of MALT1 Decreases Neuroinflammation and Pathogenicity of Virulent Rabies Virus in Mice. J Virol 2018; 92:JVI.00720-18. [PMID: 30158289 DOI: 10.1128/jvi.00720-18] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Accepted: 08/05/2018] [Indexed: 12/15/2022] Open
Abstract
Rabies virus is a neurovirulent RNA virus, which causes about 59,000 human deaths each year. Treatment for rabies does not exist due to incomplete understanding of the pathogenesis. MALT1 mediates activation of several immune cell types and is involved in the proliferation and survival of cancer cells. MALT1 acts as a scaffold protein for NF-κB signaling and a cysteine protease that cleaves substrates, leading to the expression of immunoregulatory genes. Here, we examined the impact of genetic or pharmacological MALT1 inhibition in mice on disease development after infection with the virulent rabies virus strain CVS-11. Morbidity and mortality were significantly delayed in Malt1 -/- compared to Malt1 +/+ mice, and this effect was associated with lower viral load, proinflammatory gene expression, and infiltration and activation of immune cells in the brain. Specific deletion of Malt1 in T cells also delayed disease development, while deletion in myeloid cells, neuronal cells, or NK cells had no effect. Disease development was also delayed in mice treated with the MALT1 protease inhibitor mepazine and in knock-in mice expressing a catalytically inactive MALT1 mutant protein, showing an important role of MALT1 proteolytic activity. The described protective effect of MALT1 inhibition against infection with a virulent rabies virus is the precise opposite of the sensitizing effect of MALT1 inhibition that we previously observed in the case of infection with an attenuated rabies virus strain. Together, these data demonstrate that the role of immunoregulatory responses in rabies pathogenicity is dependent on virus virulence and reveal the potential of MALT1 inhibition for therapeutic intervention.IMPORTANCE Rabies virus is a neurotropic RNA virus that causes encephalitis and still poses an enormous challenge to animal and public health. Efforts to establish reliable therapeutic strategies have been unsuccessful and are hampered by gaps in the understanding of virus pathogenicity. MALT1 is an intracellular protease that mediates the activation of several innate and adaptive immune cells in response to multiple receptors, and therapeutic MALT1 targeting is believed to be a valid approach for autoimmunity and MALT1-addicted cancers. Here, we study the impact of MALT1 deficiency on brain inflammation and disease development in response to infection of mice with the highly virulent CVS-11 rabies virus. We demonstrate that pharmacological or genetic MALT1 inhibition decreases neuroinflammation and extends the survival of CVS-11-infected mice, providing new insights in the biology of MALT1 and rabies virus infection.
Collapse
|
37
|
Analysis of expression profiles of long noncoding RNAs and mRNAs in brains of mice infected by rabies virus by RNA sequencing. Sci Rep 2018; 8:11858. [PMID: 30089776 PMCID: PMC6082909 DOI: 10.1038/s41598-018-30359-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 07/20/2018] [Indexed: 02/06/2023] Open
Abstract
Rabies, caused by rabies virus (RABV), is still the deadliest infectious disease. Mechanism of host immune response upon RABV infection is not yet fully understood. Accumulating evidences suggest that long noncoding RNAs (lncRNAs) plays key roles in host antiviral responses. However, expression profile and function of lncRNAs in RABV infection remain unclear. In the present study, expression profile of lncRNAs and mRNAs profiles were investigated in RABV-infected brain tissues of mice by RNA sequencing. A total of 140 lncRNAs and 3,807 mRNAs were differentially expressed in RABV-infected animals. The functional annotation and enrichment analysis using Gene Oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed that differentially expressed transcripts were predominantly involved in signaling pathways related to host immune response. The expression profiles of the selected lncRNAs in brains of mice during RABV infections were verified by quantitative real time polymerase chain reaction (qRT-PCR). To our knowledge, this is the first report to profile the lncRNA expression in RABV infected mice. Our findings provide insights into understanding the role of lncRNAs in host immune response against RABV infection.
Collapse
|
38
|
El-Sayed A. Advances in rabies prophylaxis and treatment with emphasis on immunoresponse mechanisms. Int J Vet Sci Med 2018; 6:8-15. [PMID: 30255072 PMCID: PMC6149183 DOI: 10.1016/j.ijvsm.2018.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Revised: 05/08/2018] [Accepted: 05/08/2018] [Indexed: 12/25/2022] Open
Abstract
Rabies is a vaccine-preventable fatal disease in man and most mammals. Although rabies is recorded in 150 territories and is responsible for at least 60,000 human deaths every year worldwide, it is a neglected tropical problem. Most of the rabies free countries are considered to be fragile free as the disease may re-emerge easily through wild mammals. For the performance of effective rabies eradication programs, a complex set of strategies and activities is required. At the time, a joint project of WHO-OIE-FAO which was announced in 2015, plans to control animal-human-ecosystems rabies interface. For effective rabies control, prophylactic policies must be applied. These include various educational outreaches for farmers and people living in endemic areas, enforced legislation for responsible dog ownership, control programs for the free-ranging stray dog and cat populations, field large-scale vaccination campaigns, and the development of new vaccine delivery strategies for both humans and animals. The present work presents the advances in the development of new safe, effective and economic vaccines for domestic dogs, and oral vaccines for the control of the disease in wild animals. It presents also some therapeutic protocols used for the treatment of patients.
Collapse
Affiliation(s)
- A El-Sayed
- Faculty of Veterinary Medicine, Department of Medicine and Infectious Diseases, Cairo University, Giza, Egypt
| |
Collapse
|
39
|
Singh R, Singh KP, Cherian S, Saminathan M, Kapoor S, Manjunatha Reddy GB, Panda S, Dhama K. Rabies - epidemiology, pathogenesis, public health concerns and advances in diagnosis and control: a comprehensive review. Vet Q 2017. [PMID: 28643547 DOI: 10.1080/01652176.2017.1343516] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Rabies is a zoonotic, fatal and progressive neurological infection caused by rabies virus of the genus Lyssavirus and family Rhabdoviridae. It affects all warm-blooded animals and the disease is prevalent throughout the world and endemic in many countries except in Islands like Australia and Antarctica. Over 60,000 peoples die every year due to rabies, while approximately 15 million people receive rabies post-exposure prophylaxis (PEP) annually. Bite of rabid animals and saliva of infected host are mainly responsible for transmission and wildlife like raccoons, skunks, bats and foxes are main reservoirs for rabies. The incubation period is highly variable from 2 weeks to 6 years (avg. 2-3 months). Though severe neurologic signs and fatal outcome, neuropathological lesions are relatively mild. Rabies virus exploits various mechanisms to evade the host immune responses. Being a major zoonosis, precise and rapid diagnosis is important for early treatment and effective prevention and control measures. Traditional rapid Seller's staining and histopathological methods are still in use for diagnosis of rabies. Direct immunofluoroscent test (dFAT) is gold standard test and most commonly recommended for diagnosis of rabies in fresh brain tissues of dogs by both OIE and WHO. Mouse inoculation test (MIT) and polymerase chain reaction (PCR) are superior and used for routine diagnosis. Vaccination with live attenuated or inactivated viruses, DNA and recombinant vaccines can be done in endemic areas. This review describes in detail about epidemiology, transmission, pathogenesis, advances in diagnosis, vaccination and therapeutic approaches along with appropriate prevention and control strategies.
Collapse
Affiliation(s)
- Rajendra Singh
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Karam Pal Singh
- b Centre for Animal Disease Research and Diagnosis (CADRAD) , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Susan Cherian
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Mani Saminathan
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Sanjay Kapoor
- c Department of Veterinary Microbiology , LLR University of Veterinary and Animal Sciences , Hisar , Haryana , India
| | - G B Manjunatha Reddy
- d ICAR-National Institute of Veterinary Epidemiology and Disease Informatics , Bengaluru , Karnataka , India
| | - Shibani Panda
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| | - Kuldeep Dhama
- a Division of Pathology , ICAR-Indian Veterinary Research Institute , Bareilly , Uttar Pradesh , India
| |
Collapse
|
40
|
Fooks AR, Cliquet F, Finke S, Freuling C, Hemachudha T, Mani RS, Müller T, Nadin-Davis S, Picard-Meyer E, Wilde H, Banyard AC. Rabies. Nat Rev Dis Primers 2017; 3:17091. [PMID: 29188797 DOI: 10.1038/nrdp.2017.91] [Citation(s) in RCA: 196] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Rabies is a life-threatening neglected tropical disease: tens of thousands of cases are reported annually in endemic countries (mainly in Africa and Asia), although the actual numbers are most likely underestimated. Rabies is a zoonotic disease that is caused by infection with viruses of the Lyssavirus genus, which are transmitted via the saliva of an infected animal. Dogs are the most important reservoir for rabies viruses, and dog bites account for >99% of human cases. The virus first infects peripheral motor neurons, and symptoms occur after the virus reaches the central nervous system. Once clinical disease develops, it is almost certainly fatal. Primary prevention involves dog vaccination campaigns to reduce the virus reservoir. If exposure occurs, timely post-exposure prophylaxis can prevent the progression to clinical disease and involves appropriate wound care, the administration of rabies immunoglobulin and vaccination. A multifaceted approach for human rabies eradication that involves government support, disease awareness, vaccination of at-risk human populations and, most importantly, dog rabies control is necessary to achieve the WHO goal of reducing the number of cases of dog-mediated human rabies to zero by 2030.
Collapse
Affiliation(s)
- Anthony R Fooks
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector Borne Diseases Research Group, (WHO Collaborating Centre for the Characterisation of Rabies and Rabies-Related Viruses, World Organisation for Animal Health (OIE) Reference Laboratory for Rabies), Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK.,Institute of Infection &Global Health, University of Liverpool, Liverpool, UK.,Institute for Infection and Immunity, St. George's Hospital Medical School, University of London, London, UK
| | - Florence Cliquet
- French Agency for Food, Environmental and Occupational Health &Safety (ANSES)-Nancy Laboratory for Rabies and Wildlife (European Union Reference Laboratory for Rabies, WHO Collaborating Centre for Research and Management in Zoonoses Control, OIE Reference Laboratory for Rabies, European Union Reference Institute for Rabies Serology), Technopôle Agricole et Vétérinaire de Pixérécourt, Malzéville, France
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology (WHO Collaborating Centre for Rabies Surveillance and Research, OIE Reference Laboratory for Rabies), Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Conrad Freuling
- Institute of Molecular Virology and Cell Biology (WHO Collaborating Centre for Rabies Surveillance and Research, OIE Reference Laboratory for Rabies), Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Thiravat Hemachudha
- Department of Medicine (Neurology) and (WHO Collaborating Centre for Research and Training on Viral Zoonoses), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Thai Red Cross Emerging Infectious Disease-Health Science Centre, Thai Red Cross Society, Bangkok, Thailand
| | - Reeta S Mani
- Department of Neurovirology (WHO Collaborating Centre for Reference and Research in Rabies), National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Thomas Müller
- Institute of Molecular Virology and Cell Biology (WHO Collaborating Centre for Rabies Surveillance and Research, OIE Reference Laboratory for Rabies), Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - Susan Nadin-Davis
- Ottawa Laboratory Fallowfield, Canadian Food Inspection Agency (WHO Collaborating Centre for Control, Pathogenesis and Epidemiology of Rabies in Carnivores), Ottawa, Ontario, Canada
| | - Evelyne Picard-Meyer
- French Agency for Food, Environmental and Occupational Health &Safety (ANSES)-Nancy Laboratory for Rabies and Wildlife (European Union Reference Laboratory for Rabies, WHO Collaborating Centre for Research and Management in Zoonoses Control, OIE Reference Laboratory for Rabies, European Union Reference Institute for Rabies Serology), Technopôle Agricole et Vétérinaire de Pixérécourt, Malzéville, France
| | - Henry Wilde
- Department of Medicine (Neurology) and (WHO Collaborating Centre for Research and Training on Viral Zoonoses), Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Ashley C Banyard
- Animal and Plant Health Agency (APHA), Wildlife Zoonoses and Vector Borne Diseases Research Group, (WHO Collaborating Centre for the Characterisation of Rabies and Rabies-Related Viruses, World Organisation for Animal Health (OIE) Reference Laboratory for Rabies), Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK
| |
Collapse
|
41
|
The phenotype of the RABV glycoprotein determines cellular and global virus load in the brain and is decisive for the pace of the disease. Virology 2017; 511:82-94. [DOI: 10.1016/j.virol.2017.08.019] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/21/2017] [Accepted: 08/14/2017] [Indexed: 11/18/2022]
|
42
|
Manjunatha V, Singh KP, Saminathan M, Singh R, Shivasharanappa N, Umeshappa CS, Dhama K, Manjunathareddy GB. Inhibition of MEK-ERK1/2-MAP kinase signalling pathway reduces rabies virus induced pathologies in mouse model. Microb Pathog 2017; 112:38-49. [PMID: 28939254 DOI: 10.1016/j.micpath.2017.09.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 09/18/2017] [Accepted: 09/18/2017] [Indexed: 12/25/2022]
Abstract
The extracellular signal-regulated kinase (ERK) pathway has been shown to regulate pathogenesis of many viral infections, but its role during rabies virus (RV) infection in vivo is not clear. In the present study, we investigated the potential role of MEK-ERK1/2 signalling pathway in the pathogenesis of rabies in mouse model and its regulatory effects on pro-inflammatory cytokines and other mediators of immunity, and kinetics of immune cells. Mice were infected with 25 LD50 of challenge virus standard (CVS) strain of RV by intracerebral (i.c.) inoculation and were treated i.c. with U0126 (specific inhibitor of MEK1/2) at 10 μM/mouse at 0, 2, 4 and 6 days post-infection. Treatment with U0126 resulted in delayed disease development and clinical signs, increased survival time with lesser mortality than untreated mice. The better survival of inhibitor-treated and RV infected mice was positively correlated with reduced viral load and reduced viral spread in the brain as quantified by real-time PCR, direct fluorescent antibody test and immunohistochemistry. CVS-infected/mock-treated mice developed severe histopathological lesions with increased Fluoro-Jade B positive degenerating neurons in brain, which were associated with higher levels of serum nitric oxide, iNOS, TNF-α, and CXCL10 mRNA. Also CVS-infected/U0126-treated mice revealed significant decrease in caspase 3 but increase in Bcl-2 mRNA levels and less TUNEL positive apoptotic cells. CVS-infected/U0126-treated group also showed significant increase in CD4+, CD8+ T lymphocytes and NK cells in blood and spleen possibly due to less apoptosis of these cells. In conclusion, these data suggest that MEK-ERK1/2 signalling pathway play critical role in the pathogenesis of RV infection in vivo and opens up new avenues of therapeutics.
Collapse
Affiliation(s)
- Venkataravanappa Manjunatha
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India; Centre for Animal Disease Research and Diagnosis, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Karam Pal Singh
- Centre for Animal Disease Research and Diagnosis, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India.
| | - Mani Saminathan
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | - Rajendra Singh
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | | | | | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar, Bareilly, Uttar Pradesh, India
| | | |
Collapse
|
43
|
Rabies Virus Antibodies from Oral Vaccination as a Correlate of Protection against Lethal Infection in Wildlife. Trop Med Infect Dis 2017; 2:tropicalmed2030031. [PMID: 30270888 PMCID: PMC6082110 DOI: 10.3390/tropicalmed2030031] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 07/06/2017] [Accepted: 07/08/2017] [Indexed: 12/24/2022] Open
Abstract
Both cell-mediated and humoral immune effectors are important in combating rabies infection, although the humoral response receives greater attention regarding rabies prevention. The principle of preventive vaccination has been adopted for strategies of oral rabies vaccination (ORV) of wildlife reservoir populations for decades to control circulation of rabies virus in free-ranging hosts. There remains much debate about the levels of rabies antibodies (and the assays to measure them) that confer resistance to rabies virus. In this paper, data from published literature and our own unpublished animal studies on the induction of rabies binding and neutralizing antibodies following oral immunization of animals with live attenuated or recombinant rabies vaccines, are examined as correlates of protection against lethal rabies infection in captive challenge settings. Analysis of our studies suggests that, though serum neutralization test results are expected to reflect in vivo protection, the blocking enzyme linked immunosorbent assay (ELISA) result at Day 28 was a better predictor of survival. ELISA kits may have an advantage of greater precision and ability to compare results among different studies and laboratories based on the inherent standardization of the kit format. This paper examines current knowledge and study findings to guide meaningful interpretation of serology results in oral baiting monitoring.
Collapse
|
44
|
Immunological aspects of rabies: a literature review. Arch Virol 2017; 162:3251-3268. [PMID: 28726129 DOI: 10.1007/s00705-017-3484-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/27/2017] [Indexed: 02/08/2023]
Abstract
Rabies is a lethal disease caused by the neurotropic virus rabies virus (RABV), and it remains an important public health problem globally. It is known that the host immune response is important for control of viral infection and promoting viral clearance. In this context, it is well documented that, in addition to RABV neutralizing antibody, interferons and cell-mediated immunity also have an important role in preventing the establishment of disease. On the other hand, RABV suppresses host immunity through different mechanisms, for example, direct inhibition of host gene expression, sequestration of pathogen-associated molecular patterns, or modification of cytokine signalling pathways, which hinder the protective host immune responses to RABV infection. Here, we review the immunological aspects of rabies, highlighting innate and adaptive immunity, as well as the host evasion immune mechanisms used by the virus. Finally, we briefly discuss how this knowledge can direct new research and be harnessed for future therapeutic strategies.
Collapse
|
45
|
Seo W, Servat A, Cliquet F, Akinbowale J, Prehaud C, Lafon M, Sabeta C. Comparison of G protein sequences of South African street rabies viruses showing distinct progression of the disease in a mouse model of experimental rabies. Microbes Infect 2017. [PMID: 28627433 DOI: 10.1016/j.micinf.2017.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Rabies is a fatal zoonotic disease and infections generally lead to a fatal encephalomyelitis in both humans and animals. In South Africa, domestic (dogs) and the wildlife (yellow mongoose) host species maintain the canid and mongoose rabies variants respectively. In this study, pathogenicity differences of South African canid and mongoose rabies viruses were investigated in a murine model, by assessing the progression of clinical signs and survivorship. Comparison of glycoprotein gene sequences revealed amino acid differences that may underpin the observed pathogenicity differences. Cumulatively, our results suggest that the canid rabies virus may be more neurovirulent in mice than the mongoose rabies variant.
Collapse
Affiliation(s)
- Wonhyo Seo
- OIE Rabies Reference Laboratory, ARC-Onderstepoort Veterinary Institute (ARC-OVI), Onderstepoort, South Africa; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, South Africa
| | - Alexandre Servat
- ANSES, Nancy Laboratory for Rabies and Wildlife, OIE and EU Rabies Reference Laboratory, WHO Collaborative Center for Research and Management in Zoonoses Control, Malzéville, France
| | - Florence Cliquet
- ANSES, Nancy Laboratory for Rabies and Wildlife, OIE and EU Rabies Reference Laboratory, WHO Collaborative Center for Research and Management in Zoonoses Control, Malzéville, France
| | - Jenkins Akinbowale
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, South Africa
| | - Christophe Prehaud
- Institut Pasteur, CNRS, Unité de Neuroimmunologie Virale, Département de Virologie Paris, France
| | - Monique Lafon
- Institut Pasteur, CNRS, Unité de Neuroimmunologie Virale, Département de Virologie Paris, France
| | - Claude Sabeta
- OIE Rabies Reference Laboratory, ARC-Onderstepoort Veterinary Institute (ARC-OVI), Onderstepoort, South Africa; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, South Africa.
| |
Collapse
|
46
|
Seo W, Prehaud C, Khan Z, Sabeta C, Lafon M. Investigation of rabies virus glycoprotein carboxyl terminus as an in vitro predictive tool of neurovirulence. A 3R approach. Microbes Infect 2017; 19:476-484. [PMID: 28602914 DOI: 10.1016/j.micinf.2017.05.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 03/10/2017] [Accepted: 05/30/2017] [Indexed: 01/29/2023]
Abstract
In the field of live viral vaccines production, there is an unmet need for in vitro tests complying a 3R approach (Refine, Replace and Reduce the use of animal experimentation) to replace the post-licensing safety tests currently assayed in animals. Here, we performed a pilot study evaluating whether virulence of rabies virus, RABV, can be forecast by an in vitro test of neurite outgrowth. The rationale to use neurite outgrowth as a read-out for this test is based on the salient property of the cytoplasmic domain of the G-protein (Cyto-G) of virulent RABV strains - not of attenuated RABV strains - to stimulate neurite outgrowth in vitro. We observed that neurite elongation triggered by the Cyto-Gs encoded by different RABV field isolates correlate with the distinct virulence scores obtained in a mouse model of experimental rabies. Our results cast the idea that it could be feasible to predict RABV virulence by testing the in vitro property of a RABV strain to promote neurite outgrowth without the use of animal experimentation.
Collapse
Affiliation(s)
- Wonhyo Seo
- OIE Rabies Reference Laboratory, ARC-Onderstepoort Veterinary Institute (ARC-OVI), Onderstepoort, South Africa; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, South Africa; Institut Pasteur, CNRS, Unité de Neuroimmunologie Virale, Département de Virologie, Paris, France
| | - Christophe Prehaud
- Institut Pasteur, CNRS, Unité de Neuroimmunologie Virale, Département de Virologie, Paris, France
| | - Zakir Khan
- Institut Pasteur, CNRS, Unité de Neuroimmunologie Virale, Département de Virologie, Paris, France
| | - Claude Sabeta
- OIE Rabies Reference Laboratory, ARC-Onderstepoort Veterinary Institute (ARC-OVI), Onderstepoort, South Africa; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, South Africa
| | - Monique Lafon
- Institut Pasteur, CNRS, Unité de Neuroimmunologie Virale, Département de Virologie, Paris, France.
| |
Collapse
|
47
|
Impact of caspase-1/11, -3, -7, or IL-1 β/IL-18 deficiency on rabies virus-induced macrophage cell death and onset of disease. Cell Death Discov 2017; 3:17012. [PMID: 28280602 PMCID: PMC5339016 DOI: 10.1038/cddiscovery.2017.12] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 01/27/2017] [Indexed: 12/17/2022] Open
Abstract
Rabies virus is a highly neurovirulent RNA virus, which causes about 59000 deaths in humans each year. Previously, we described macrophage cytotoxicity upon infection with rabies virus. Here we examined the type of cell death and the role of specific caspases in cell death and disease development upon infection with two laboratory strains of rabies virus: Challenge Virus Standard strain-11 (CVS-11) is highly neurotropic and lethal for mice, while the attenuated Evelyn–Rotnycki–Abelseth (ERA) strain has a broader cell tropism, is non-lethal and has been used as an oral vaccine for animals. Infection of Mf4/4 macrophages with both strains led to caspase-1 activation and IL-1β and IL-18 production, as well as activation of caspases-3, -7, -8, and -9. Moreover, absence of caspase-3, but not of caspase-1 and -11 or -7, partially inhibited virus-induced cell death of bone marrow-derived macrophages. Intranasal inoculation with CVS-11 of mice deficient for either caspase-1 and -11 or -7 or both IL-1β and IL-18 led to general brain infection and lethal disease similar to wild-type mice. Deficiency of caspase-3, on the other hand, significantly delayed the onset of disease, but did not prevent final lethal outcome. Interestingly, deficiency of caspase-1/11, the key executioner of pyroptosis, aggravated disease severity caused by ERA virus, whereas wild-type mice or mice deficient for either caspase-3, -7, or both IL-1β and IL-18 presented the typical mild symptoms associated with ERA virus. In conclusion, rabies virus infection of macrophages induces caspase-1- and caspase-3-dependent cell death. In vivo caspase-1/11 and caspase-3 differently affect disease development in response to infection with the attenuated ERA strain or the virulent CVS-11 strain, respectively. Inflammatory caspases seem to control attenuated rabies virus infection, while caspase-3 aggravates virulent rabies virus infection.
Collapse
|
48
|
Miao FM, Zhang SF, Wang SC, Liu Y, Zhang F, Hu RL. Comparison of immune responses to attenuated rabies virus and street virus in mouse brain. Arch Virol 2016; 162:247-257. [PMID: 27722994 DOI: 10.1007/s00705-016-3081-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2016] [Accepted: 09/19/2016] [Indexed: 12/25/2022]
Abstract
Rabies is a lethal neurological disease caused by the neurotropic rabies virus (RABV). To investigate the innate immune response in the brain during rabies infection, key gene transcripts indicative of innate immunity in a mouse model system were measured using real-time RT-PCR. Mice were infected via the intracerebral or intramuscular route with either attenuated rabies virus (SRV9) or pathogenic rabies virus (BD06). Infection with SRV9 resulted in the early detection of viral replication and the rapid induction of innate immune response gene expression in the brain. BD06 infection elicited innate immune response gene expression during only the late stage of infection. We measured Na-fluorescein uptake to assess blood-brain barrier (BBB) permeability, which was enhanced during the early stage of SRV9 infection and significantly enhanced during the late stage of BD06 infection. Furthermore, early SRV9 replication increased the maturation and differentiation of dendritic cells (DCs) and B cells in the inguinal lymph nodes and initiated the generation of virus-neutralizing antibodies (VNAs), which cooperate with the innate immune response to eliminate virus from the CNS. However, BD06 infection did not stimulate VNA production; thus, the virus was able to evade the host immune response and cause encephalitis. The rabies virus phosphoprotein has been reported to counteract IFN activation. In an in vitro study of the relationship between IFN antagonism and RABV pathogenicity, we demonstrated that SRV9 more strongly antagonized IFN activity than did BD06. Therefore, there is no positive relationship between the IFN antagonist activity of the virus and its pathogenicity.
Collapse
Affiliation(s)
- Fa-Ming Miao
- Laboratory of Epidemiology, Institute of Military Veterinary, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, 666 Liuying West Road, Changchun, 130122, Jilin, China
| | - Shou-Feng Zhang
- Laboratory of Epidemiology, Institute of Military Veterinary, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, 666 Liuying West Road, Changchun, 130122, Jilin, China
| | - Shu-Chao Wang
- Laboratory of Epidemiology, Institute of Military Veterinary, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, 666 Liuying West Road, Changchun, 130122, Jilin, China
| | - Ye Liu
- Laboratory of Epidemiology, Institute of Military Veterinary, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, 666 Liuying West Road, Changchun, 130122, Jilin, China
| | - Fei Zhang
- Laboratory of Epidemiology, Institute of Military Veterinary, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, 666 Liuying West Road, Changchun, 130122, Jilin, China
| | - Rong-Liang Hu
- Laboratory of Epidemiology, Institute of Military Veterinary, Academy of Military Medical Sciences, Key Laboratory of Jilin Province for Zoonosis Prevention and Control, 666 Liuying West Road, Changchun, 130122, Jilin, China.
| |
Collapse
|
49
|
Scott TP, Nel LH. Subversion of the Immune Response by Rabies Virus. Viruses 2016; 8:v8080231. [PMID: 27548204 PMCID: PMC4997593 DOI: 10.3390/v8080231] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/24/2022] Open
Abstract
Rabies has affected mankind for several centuries and is one of the oldest known zoonoses. It is peculiar how little is known regarding the means by which rabies virus (RABV) evades the immune response and kills its host. This review investigates the complex interplay between RABV and the immune system, including the various means by which RABV evades, or advantageously utilizes, the host immune response in order to ensure successful replication and spread to another host. Different factors that influence immune responses—including age, sex, cerebral lateralization and temperature—are discussed, with specific reference to RABV and the effects on host morbidity and mortality. We also investigate the role of apoptosis and discuss whether it is a detrimental or beneficial mechanism of the host’s response to infection. The various RABV proteins and their roles in immune evasion are examined in depth with reference to important domains and the downstream effects of these interactions. Lastly, an overview of the means by which RABV evades important immune responses is provided. The research discussed in this review will be important in determining the roles of the immune response during RABV infections as well as to highlight important therapeutic target regions and potential strategies for rabies treatment.
Collapse
Affiliation(s)
- Terence P Scott
- Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0002, South Africa.
| | - Louis H Nel
- Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0002, South Africa.
| |
Collapse
|
50
|
Ruigrok TJH, van Touw S, Coulon P. Caveats in Transneuronal Tracing with Unmodified Rabies Virus: An Evaluation of Aberrant Results Using a Nearly Perfect Tracing Technique. Front Neural Circuits 2016; 10:46. [PMID: 27462206 PMCID: PMC4939302 DOI: 10.3389/fncir.2016.00046] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Accepted: 06/15/2016] [Indexed: 12/23/2022] Open
Abstract
Apart from the genetically engineered, modified, strains of rabies virus (RABV), unmodified ‘fixed’ virus strains of RABV, such as the ‘French’ subtype of CVS11, are used to examine synaptically connected networks in the brain. This technique has been shown to have all the prerequisite characteristics for ideal tracing as it does not metabolically affect infected neurons within the time span of the experiment, it is transferred transneuronally in one direction only and to all types of neurons presynaptic to the infected neuron, number of transneuronal steps can be precisely controlled by survival time and it is easily detectable with a sensitive technique. Here, using the ‘French’ CVS 11 subtype of RABV in Wistar rats, we show that some of these characteristics may not be as perfect as previously indicated. Using injection of RABV in hind limb muscles, we show that RABV-infected spinal motoneurons may already show lysis 1 or 2 days after infection. Using longer survival times we were able to establish that Purkinje cells may succumb approximately 3 days after infection. In addition, some neurons seem to resist infection, as we noted that the number of RABV-infected inferior olivary neurons did not progress in the same rate as other infected neurons. Furthermore, in our hands, we noted that infection of Purkinje cells did not result in expected transneuronal labeling of cell types that are presynaptic to Purkinje cells such as molecular layer interneurons and granule cells. However, these cell types were readily infected when RABV was injected directly in the cerebellar cortex. Conversely, neurons in the cerebellar nuclei that project to the inferior olive did not take up RABV when this was injected in the inferior olive, whereas these cells could be infected with RABV via a transneuronal route. These results suggest that viral entry from the extracellular space depends on other factors or mechanisms than those used for retrograde transneuronal transfer. We conclude that transneuronal tracing with RABV may result in unexpected results, as not all properties of RABV seem to be ubiquitously valid.
Collapse
Affiliation(s)
- Tom J H Ruigrok
- Department of Neuroscience, Erasmus Medical Center Rotterdam, Netherlands
| | - Sven van Touw
- Department of Neuroscience, Erasmus Medical Center Rotterdam, Netherlands
| | - Patrice Coulon
- Equipe P3M - UMR 7298, Institut de Neurosciences de la Timone, Aix-Marseille Université, CNRS Marseille, France
| |
Collapse
|