1
|
Wang S, Zhou H, Zhang X, Cao J, Lei T, Chen M, Zhang Y, Xu Y. Dynamic bidirectional regulation between Stk38 and rabies virus M protein coordinates apoptosis progression during neurotropic infection. Int J Biol Macromol 2025; 315:144398. [PMID: 40398756 DOI: 10.1016/j.ijbiomac.2025.144398] [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: 03/02/2025] [Revised: 04/26/2025] [Accepted: 05/18/2025] [Indexed: 05/23/2025]
Abstract
Rabies virus (RABV) causes fatal encephalitis in untreated humans, representing a critical global zoonotic threat. Although the viral matrix protein (M) is established as a mediator of mitochondrial apoptosis and enhancer of viral replication, its regulatory mechanisms remain incompletely characterized. This study reveals a reciprocal regulatory axis between RABV M protein and the host kinase Stk38. We demonstrate that Stk38 stabilizes M protein by blocking its ubiquitin-independent proteasomal degradation, thereby potentiating late-stage viral replication. Structural analysis identifies residues 1-88 and 383-465 of Stk38 as non-essential for M binding or stability maintenance, yet crucial for orchestrating M's mitochondrial localization and pro-apoptotic function. Conversely, RABV utilizes residues 154-202 of M protein to induce ubiquitin-dependent degradation of Stk38 during late infection. This mutual regulation establishes domain-specific control mechanisms governing protein stability, facilitating temporal coordination between viral proliferation and apoptotic progression. Our findings elucidate a pathogenic mechanism wherein viral and host factors reciprocally modulate each other's stability through distinct structural domains, positioning the Stk38-M regulatory axis as a promising therapeutic target for rabies intervention.
Collapse
Affiliation(s)
- Shujie Wang
- Key Laboratory of Infectious Diseases and Biosafety, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China; Institute of Life Sciences, Zunyi Medical University, Zunyi 563000, Guizhou, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Hao Zhou
- Key Laboratory of Infectious Diseases and Biosafety, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China; Institute of Life Sciences, Zunyi Medical University, Zunyi 563000, Guizhou, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Xi Zhang
- Key Laboratory of Infectious Diseases and Biosafety, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China; Institute of Life Sciences, Zunyi Medical University, Zunyi 563000, Guizhou, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Jingjing Cao
- State Key Laboratory of Microbial Technology, Microbial Technology Institute, Shandong University, Qingdao 266237, Shandong, China
| | - Taohong Lei
- Key Laboratory of Infectious Diseases and Biosafety, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China; Institute of Life Sciences, Zunyi Medical University, Zunyi 563000, Guizhou, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Ming Chen
- Key Laboratory of Infectious Diseases and Biosafety, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China; Institute of Life Sciences, Zunyi Medical University, Zunyi 563000, Guizhou, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Yulin Zhang
- Key Laboratory of Infectious Diseases and Biosafety, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China; Institute of Life Sciences, Zunyi Medical University, Zunyi 563000, Guizhou, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Yunbin Xu
- Key Laboratory of Infectious Diseases and Biosafety, Guizhou Provincial Department of Education, Zunyi Medical University, Zunyi 563000, Guizhou, China; Institute of Life Sciences, Zunyi Medical University, Zunyi 563000, Guizhou, China; School of Preclinical Medicine, Zunyi Medical University, Zunyi 563000, Guizhou, China.
| |
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
|
Harada M, Matsuu A, Park ES, Inoue Y, Uda A, Kaku Y, Okutani A, Posadas-Herrera G, Ishijima K, Inoue S, Maeda K. Construction of Vero cell-adapted rabies vaccine strain by five amino acid substitutions in HEP-Flury strain. Sci Rep 2024; 14:12559. [PMID: 38822013 PMCID: PMC11143356 DOI: 10.1038/s41598-024-63337-9] [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: 01/13/2024] [Accepted: 05/28/2024] [Indexed: 06/02/2024] Open
Abstract
Rabies virus (RABV) causes fatal neurological disease. Pre-exposure prophylaxis (PrEP) and post-exposure prophylaxis (PEP) using inactivated-virus vaccines are the most effective measures to prevent rabies. In Japan, HEP-Flury, the viral strain, used as a human rabies vaccine, has historically been propagated in primary fibroblast cells derived from chicken embryos. In the present study, to reduce the cost and labor of vaccine production, we sought to adapt the original HEP-Flury (HEP) to Vero cells. HEP was repeatedly passaged in Vero cells to generate ten- (HEP-10V) and thirty-passaged (HEP-30V) strains. Both HEP-10V and HEP-30V grew significantly better than HEP in Vero cells, with virulence and antigenicity similar to HEP. Comparison of the complete genomes with HEP revealed three non-synonymous mutations in HEP-10V and four additional non-synonymous mutations in HEP-30V. Comparison among 18 recombinant HEP strains constructed by reverse genetics and vesicular stomatitis viruses pseudotyped with RABV glycoproteins indicated that the substitution P(L115H) in the phosphoprotein and G(S15R) in the glycoprotein improved viral propagation in HEP-10V, while in HEP-30V, G(V164E), G(L183P), and G(A286V) in the glycoprotein enhanced entry into Vero cells. The obtained recombinant RABV strain, rHEP-PG4 strain, with these five substitutions, is a strong candidate for production of human rabies vaccine.
Collapse
Affiliation(s)
- Michiko Harada
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Aya Matsuu
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Eun-Sil Park
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yusuke Inoue
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Akihiko Uda
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yoshihiro Kaku
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Akiko Okutani
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Guillermo Posadas-Herrera
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Keita Ishijima
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Satoshi Inoue
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Ken Maeda
- Joint Graduate School of Veterinary Medicine, Yamaguchi University, 1677-1 Yoshida, Yamaguchi, 753-8515, Japan.
- Department of Veterinary Science, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| |
Collapse
|
4
|
Akter F, Shamimuzzaman M. Potency, immunogenicity, and efficacy of rabies vaccine: In vitro and in vivo approach. Immun Inflamm Dis 2024; 12:e1198. [PMID: 38411335 PMCID: PMC10898210 DOI: 10.1002/iid3.1198] [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: 10/20/2023] [Revised: 01/12/2024] [Accepted: 02/07/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Rabies, a potentially lethal virus, affects more than 150 countries. Although the rabies vaccine and immunoglobulin have been available since 1908, Bangladesh is new to vaccine manufacturing. We checked the quality of the local manufacturing rabies vaccine for substandard. METHODS The potency and immunogenicity of 20 vaccines were analyzed by three in vivo and in vitro methods from March 2020 to May 2023. Single radial immunodiffusion, fluorescent antibody virus neutralization, and national institutes of health tests were carried out to evaluate the vaccine's efficacy to provide sufficient protection against the rabies virus. RESULTS The potency of the rabies vaccine was determined by the in vitro SRID method by measuring glycoprotein content. An average of 16 articles from each batch was calculated. The minimum and maximum average mean values of the 20 batches were 5.058 and 5.346, respectively. The variance was calculated at 0.00566. We found a coefficient of variation (CV) between 9.36% and 14.80%. The 100% sample was satisfactory, as these samples had a potency of over 2.5 IU/mL. To observe immunogenicity, we applied the FAVN method for determining antibody titers. An average of 16 articles from every batch were counted to quantify antibody titers. The mean quantity of antibody titers ranged from 2.389 to 3.3875. The CV was slightly lower because of the dispersion of the data. At last, we performed an in vivo method, the NIH test method, to determine potency based on mortality rate. We found a mean value of 4.777 IU/SHD with a standard deviation of 1.13 IU/SHD. All 20 batches were found 100% satisfactory in the NIH test. CONCLUSION The study implies that the rabies human vaccines manufactured in Bangladesh are potent enough to provide sufficient immunogenicity. Our research is warranted testimony for healthcare providers who work to extirpate rabies.
Collapse
Affiliation(s)
- Fahima Akter
- Department of Microbiology and HygieneBangladesh Agricultural UniversityMymensinghBangladesh
| | - Md. Shamimuzzaman
- Department of MicrobiologyJashore University of Science and TechnologyJashoreBangladesh
- Directorate General of Drug AdministrationMinistry of Health and Family WelfareDhakaBangladesh
| |
Collapse
|
5
|
Yin J, Wang S, Ren S, Liang Z, Ge J, Sun Y, Yin X, Wang X. TMP269, a small molecule inhibitor of class IIa HDAC, suppresses RABV replication in vitro. Front Microbiol 2023; 14:1284439. [PMID: 38107853 PMCID: PMC10722228 DOI: 10.3389/fmicb.2023.1284439] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 12/19/2023] Open
Abstract
TMP269, a small molecular inhibitor of IIa histone deacetylase, plays a vital role in cancer therapeutic. However, the effect of TMP269 on the regulation of viral replication has not been studied. In the present study, we found that TMP269 treatment significantly inhibited RABV replication at concentrations without significant cytotoxicity in a dose-dependent manner. In addition, TMP269 can reduce the viral titers and protein levels of RABV at an early stage in the viral life cycle. RNA sequencing data revealed that immune-related pathways and autophagy-related genes were significantly downregulated after RABV infection treated with TMP269. Further exploration shows that autophagy enhances RABV replication in HEK-293T cells, while TMP269 can inhibit autophagy to decrease RABV replication. Together, these results provide a novel treatment strategy for rabies.
Collapse
Affiliation(s)
- Juanbin Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Shasha Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Shanhui Ren
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Zhengji Liang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Junwei Ge
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, Heilongjiang, China
| | - Yuefeng Sun
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangping Yin
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Xiangwei Wang
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
6
|
Almasoud I, Charlton FW, Finke S, Barr JN, Mankouri J. Internalization of rabies virus glycoprotein differs between pathogenic and attenuated virus strains. J Gen Virol 2023; 104:001935. [PMID: 38063294 PMCID: PMC10770930 DOI: 10.1099/jgv.0.001935] [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: 08/08/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The zoonotic rabies virus (RABV) is a non-segmented negative-sense RNA virus classified within the family Rhabdoviridae, and is the most common aetiological agent responsible for fatal rabies disease. The RABV glycoprotein (G) forms trimeric spikes that protrude from RABV virions and mediate virus attachment, entry and spread, and is a major determinant of RABV pathogenesis. A range of RABV strains exist that are highly pathogenic in part due to their ability to evade host immune detection. However, some strains are disease-attenuated and can be cleared by host defences. A detailed molecular understanding of how strain variation relates to pathogenesis is currently lacking. Here, we reveal key differences in the trafficking profiles of RABV-G proteins from the challenge virus standard strain (CVS-11) and a highly attenuated vaccine strain SAD-B19 (SAD). We show that CVS-G traffics to the cell surface and undergoes rapid internalization through both clathrin- and cholesterol-dependent endocytic pathways. In contrast, SAD-G remains resident at the plasma membrane and internalizes at a significantly slower rate. Through engineering hybrids of CVS-G and SAD-G, we show that the cytoplasmic tail of CVS-G is the key determinant of these different internalization profiles. Alanine scanning further revealed that mutation of Y497 in CVS-G (H497 in SAD-G) could reduce the rate of internalization to SAD-G levels. Together, these data reveal new phenotypic differences between CVS-G and SAD-G proteins that may contribute to altered in vivo pathogenicity.
Collapse
Affiliation(s)
- Ibrahim Almasoud
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
- Present address: Department of Biological Sciences, Faculty of Science, Kuwait University, Kuwait City, Kuwait
| | - Frank W. Charlton
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Greifswald-Insel Riems, Germany
| | - John N. Barr
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| | - Jamel Mankouri
- School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, UK
| |
Collapse
|
7
|
Salazar S, Luong KTY, Koyuncu OO. Cell Intrinsic Determinants of Alpha Herpesvirus Latency and Pathogenesis in the Nervous System. Viruses 2023; 15:2284. [PMID: 38140525 PMCID: PMC10747186 DOI: 10.3390/v15122284] [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: 10/20/2023] [Revised: 11/10/2023] [Accepted: 11/19/2023] [Indexed: 12/24/2023] Open
Abstract
Alpha herpesvirus infections (α-HVs) are widespread, affecting more than 70% of the adult human population. Typically, the infections start in the mucosal epithelia, from which the viral particles invade the axons of the peripheral nervous system. In the nuclei of the peripheral ganglia, α-HVs establish a lifelong latency and eventually undergo multiple reactivation cycles. Upon reactivation, viral progeny can move into the nerves, back out toward the periphery where they entered the organism, or they can move toward the central nervous system (CNS). This latency-reactivation cycle is remarkably well controlled by the intricate actions of the intrinsic and innate immune responses of the host, and finely counteracted by the viral proteins in an effort to co-exist in the population. If this yin-yang- or Nash-equilibrium-like balance state is broken due to immune suppression or genetic mutations in the host response factors particularly in the CNS, or the presence of other pathogenic stimuli, α-HV reactivations might lead to life-threatening pathologies. In this review, we will summarize the molecular virus-host interactions starting from mucosal epithelia infections leading to the establishment of latency in the PNS and to possible CNS invasion by α-HVs, highlighting the pathologies associated with uncontrolled virus replication in the NS.
Collapse
Affiliation(s)
| | | | - Orkide O. Koyuncu
- Department of Microbiology & Molecular Genetics, School of Medicine and Center for Virus Research, University of California, Irvine, CA 92697, USA; (S.S.); (K.T.Y.L.)
| |
Collapse
|
8
|
Jin L, Sullivan HA, Zhu M, Lea NE, Lavin TK, Fu X, Matsuyama M, Hou Y, Feng G, Wickersham IR. Third-generation rabies viral vectors allow nontoxic retrograde targeting of projection neurons with greatly increased efficiency. CELL REPORTS METHODS 2023; 3:100644. [PMID: 37989085 PMCID: PMC10694603 DOI: 10.1016/j.crmeth.2023.100644] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 08/16/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023]
Abstract
Rabies viral vectors have become important components of the systems neuroscience toolkit, allowing both direct retrograde targeting of projection neurons and monosynaptic tracing of inputs to defined postsynaptic populations, but the rapid cytotoxicity of first-generation (ΔG) vectors limits their use to short-term experiments. We recently introduced second-generation, double-deletion-mutant (ΔGL) rabies viral vectors, showing that they efficiently retrogradely infect projection neurons and express recombinases effectively but with little to no detectable toxicity; more recently, we have shown that ΔGL viruses can be used for monosynaptic tracing with far lower cytotoxicity than the first-generation system. Here, we introduce third-generation (ΔL) rabies viral vectors, which appear to be as nontoxic as second-generation ones but have the major advantage of growing to much higher titers, resulting in significantly increased numbers of retrogradely labeled neurons in vivo.
Collapse
Affiliation(s)
- Lei Jin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Heather A Sullivan
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mulangma Zhu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Nicholas E Lea
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Thomas K Lavin
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Xin Fu
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Makoto Matsuyama
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - YuanYuan Hou
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA; Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ian R Wickersham
- McGovern Institute for Brain Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
9
|
Konopka G, Bhaduri A. Functional genomics and systems biology in human neuroscience. Nature 2023; 623:274-282. [PMID: 37938705 PMCID: PMC11465930 DOI: 10.1038/s41586-023-06686-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/27/2023] [Indexed: 11/09/2023]
Abstract
Neuroscience research has entered a phase of key discoveries in the realm of neurogenomics owing to strong financial and intellectual support for resource building and tool development. The previous challenge of tissue heterogeneity has been met with the application of techniques that can profile individual cells at scale. Moreover, the ability to perturb genes, gene regulatory elements and neuronal activity in a cell-type-specific manner has been integrated with gene expression studies to uncover the functional underpinnings of the genome at a systems level. Although these insights have necessarily been grounded in model systems, we now have the opportunity to apply these approaches in humans and in human tissue, thanks to advances in human genetics, brain imaging and tissue collection. We acknowledge that there will probably always be limits to the extent to which we can apply the genomic tools developed in model systems to human neuroscience; however, as we describe in this Perspective, the neuroscience field is now primed with an optimal foundation for tackling this ambitious challenge. The application of systems-level network analyses to these datasets will facilitate a deeper appreciation of human neurogenomics that cannot otherwise be achieved from directly observable phenomena.
Collapse
Affiliation(s)
- Genevieve Konopka
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX, USA.
- Peter O'Donnell Jr Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Aparna Bhaduri
- Department of Biological Chemistry, University of California, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Wagner A, Mutschler H. Design principles and applications of synthetic self-replicating RNAs. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1803. [PMID: 37264531 DOI: 10.1002/wrna.1803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 04/24/2023] [Accepted: 05/11/2023] [Indexed: 06/03/2023]
Abstract
With the advent of ever more sophisticated methods for the in vitro synthesis and the in vivo delivery of RNAs, synthetic mRNAs have gained substantial interest both for medical applications, as well as for biotechnology. However, in most biological systems exogeneous mRNAs possess only a limited half-life, especially in fast dividing cells. In contrast, viral RNAs can extend their lifetime by actively replicating inside their host. As such they may serve as scaffolds for the design of synthetic self-replicating RNAs (srRNA), which can be used to increase both the half-life and intracellular concentration of coding RNAs. Synthetic srRNAs may be used to enhance recombinant protein expression or induce the reprogramming of differentiated cells into pluripotent stem cells but also to create cell-free systems for research based on experimental evolution. In this article, we discuss the applications and design principles of srRNAs used for cellular reprogramming, mRNA-based vaccines and tools for synthetic biology. This article is categorized under: RNA in Disease and Development > RNA in Disease RNA in Disease and Development > RNA in Development RNA Evolution and Genomics > RNA and Ribonucleoprotein Evolution.
Collapse
Affiliation(s)
- Alexander Wagner
- Biomimetic Chemistry, Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| | - Hannes Mutschler
- Biomimetic Chemistry, Department of Chemistry and Chemical Biology, TU Dortmund University, Dortmund, Germany
| |
Collapse
|
11
|
Wang C, Chen Y, Hu S, Liu X. Insights into the function of ESCRT and its role in enveloped virus infection. Front Microbiol 2023; 14:1261651. [PMID: 37869652 PMCID: PMC10587442 DOI: 10.3389/fmicb.2023.1261651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/20/2023] [Indexed: 10/24/2023] Open
Abstract
The endosomal sorting complex required for transport (ESCRT) is an essential molecular machinery in eukaryotic cells that facilitates the invagination of endosomal membranes, leading to the formation of multivesicular bodies (MVBs). It participates in various cellular processes, including lipid bilayer remodeling, cytoplasmic separation, autophagy, membrane fission and re-modeling, plasma membrane repair, as well as the invasion, budding, and release of certain enveloped viruses. The ESCRT complex consists of five complexes, ESCRT-0 to ESCRT-III and VPS4, along with several accessory proteins. ESCRT-0 to ESCRT-II form soluble complexes that shuttle between the cytoplasm and membranes, mainly responsible for recruiting and transporting membrane proteins and viral particles, as well as recruiting ESCRT-III for membrane neck scission. ESCRT-III, a soluble monomer, directly participates in vesicle scission and release, while VPS4 hydrolyzes ATP to provide energy for ESCRT-III complex disassembly, enabling recycling. Studies have confirmed the hijacking of ESCRT complexes by enveloped viruses to facilitate their entry, replication, and budding. Recent research has focused on the interaction between various components of the ESCRT complex and different viruses. In this review, we discuss how different viruses hijack specific ESCRT regulatory proteins to impact the viral life cycle, aiming to explore commonalities in the interaction between viruses and the ESCRT system.
Collapse
Affiliation(s)
- Chunxuan Wang
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yu Chen
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Shunlin Hu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiufan Liu
- Animal Infectious Disease Laboratory, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonosis, Yangzhou University, Yangzhou, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, China
| |
Collapse
|
12
|
Zhang B, Cai T, He H, Huang X, Luo Y, Huang S, Luo J, Guo X. TRIM25 Suppresses Rabies Virus Fixed HEP-Flury Strain Production by Activating RIG-1-Mediated Type I Interferons. Genes (Basel) 2023; 14:1555. [PMID: 37628607 PMCID: PMC10454932 DOI: 10.3390/genes14081555] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 07/25/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
Rabies remains a great threat to public health worldwide. So far, the mechanism of rabies virus (RABV) infection is not fully understood, and there is no effective treatment for rabies. Identifying more host restriction factors of RABV will spur the development of novel therapeutic interventions against rabies. Accumulating studies suggest that tripartite motif-containing (TRIM) proteins have great effects on virus replication. TRIMs control the antiviral responses through either direct interaction with viral proteins or indirect regulation of innate immune signaling molecules in the host. The role of TRIM25 in rabies virus (RABV) infection is poorly understood. Using next-generation sequencing, we found that TRIM25 is upregulated during HEP-Flury infection. Knockdown of TRIM25 enhances HEP-Flury production, while overexpression of TRIM25 suppresses HEP-Flury replication. Knockdown of interferon α and interferon β weakens the anti-RABV response induced by TRIM25 overexpression, and potentiates RABV production. Furthermore, we found that TRIM25 regulates type-I interferon response by targeting retinoic acid-inducible gene I (RIG-I) during HEP-Flury infection. Knockdown of RIG-I weakens the anti-HEP-Flury response induced by TRIM25 overexpression, indicating that TRIM25 regulates RABV production via the RIG-I-IFN axis. In addition, we observed that TRIM25 does not directly interact with HEP-Flury structural proteins, suggesting that TRIM25 regulates HEP-Flury production indirectly. Taken together, our work identifies TRIM25 as a new host factor involved in HEP-Flury infection, which may be a potential target for the development of antiviral drugs against RABV.
Collapse
Affiliation(s)
- Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Ting Cai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Hongling He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Xuezhe Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510651, China; (B.Z.); (T.C.); (H.H.); (X.H.); (Y.L.); (J.L.)
| |
Collapse
|
13
|
Zhang B, Cai T, He H, Huang X, Chen G, Lai Y, Luo Y, Huang S, Luo J, Guo X. TRIM21 Promotes Rabies Virus Production by Degrading IRF7 through Ubiquitination. Int J Mol Sci 2023; 24:10892. [PMID: 37446070 PMCID: PMC10341556 DOI: 10.3390/ijms241310892] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Rabies, a highly fatal zoonotic disease, is a significant global public health threat. Currently, the pathogenic mechanism of rabies has not been fully elucidated, and no effective treatment for rabies is available. Increasing evidence shows that the tripartite-motif protein (TRIM) family of proteins participates in the host's regulation of viral replication. Studies have demonstrated the upregulated expression of tripartite-motif protein 21 (TRIM21) in the brain tissue of mice infected with the rabies virus. Related studies have shown that TRIM21 knockdown inhibits RABV replication, while overexpression of TRIM21 exerted the opposite effect. Knockdown of interferon-alpha and interferon-beta modulates the inhibition of RABV replication caused by TRIM21 knockdown and promotes the replication of the virus. Furthermore, our previous study revealed that TRIM21 regulates the secretion of type I interferon during RABV infection by targeting interferon regulatory factor 7 (IRF7). IRF7 knockdown reduced the inhibition of RABV replication caused by the knockdown of TRIM21 and promoted viral replication. TRIM21 regulates RABV replication via the IRF7-IFN axis. Our study identified TRIM21 as a novel host factor required by RABV for replication. Thus, TRIM21 is a potential target for rabies treatment or management.
Collapse
Affiliation(s)
- Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Ting Cai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Hongling He
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Xuezhe Huang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Guie Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Yanqin Lai
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510000, China; (B.Z.); (T.C.); (H.H.); (X.H.); (G.C.); (Y.L.); (Y.L.)
| |
Collapse
|
14
|
Liu J, Li W, Yu D, Jin R, Hou H, Ling X, Kiflu AB, Wei X, Yang X, Li X, He Y, Luo TR. Transcriptomic Analysis of mRNA Expression Profiles in the Microglia of Mouse Brains Infected with Rabies Viruses of Varying Virulence. Viruses 2023; 15:1223. [PMID: 37376523 DOI: 10.3390/v15061223] [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: 04/13/2023] [Revised: 05/09/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
Rabies is a lethal encephalitis caused by the rabies virus (RABV) with a fatality rate near 100% after the onset of clinical symptoms in humans and animals. Microglia are resident immune cells in the central nervous system. Few studies have been conducted on the functional role of microglia in RABV infection. Here, we performed a transcriptomic analysis of mRNA expression profiles in the microglia of mouse brains intracerebrally infected with RABV. We successfully isolated single microglial cells from the mouse brains. The survival rate of dissociated microglial cells was 81.91%-96.7%, and the purity was 88.3%. Transcriptomic analysis revealed 22,079 differentially expressed mRNAs identified in the microglia of mouse brains infected with RABV strains (rRC-HL, GX074, and CVS-24) of varying virulence at 4 and 7 days post-infection (dpi) compared to the control group. The numbers of DEGs versus the control at 4 and 7 dpi in mice infected with rRC-HL, GX074, and CVS-24 were 3622 and 4590, 265 and 4901, and 4079 and 6337. The GO enrichment analysis showed that response to stress, response to external stimulus, regulation of response to stimulus, and immune system process were abundant during RABV infection. The KEGG analysis indicated that the Tlr, Tnf, RIG-I, NOD, NF-κB, MAPK, and Jak-STAT signaling pathways were involved in RABV infection at both 4 and 7 dpi. However, some phagocytosis and cell signal transduction processes, such as endocytosis, p53, phospholipase D, and oxidative phosphorylation signaling pathways, were only expressed at 7 dpi. The involvement of the Tnf and Tlr signaling pathways prompted us to construct a protein-protein interaction (PPI) network of these pathways. The PPI revealed 8 DEGs, including Mmp9, Jun, Pik3r1, and Mapk12. Notably, Il-1b interacted with Tnf and Il-6 with combined scores of 0.973 and 0.981, respectively. RABV causes significant changes in mRNA expression profiles in the microglia in mice. 22,079 differentially expressed mRNAs were identified in the microglia of mice infected with RABV strains of varying virulence at 4 and 7 dpi. The DEGs were evaluated using GO, KEGG, and PPI network analysis. Many immune pathways were up-regulated in RABV-infected groups. The findings will help elucidate the microglial molecular mechanisms of cellular metabolism dysregulated by RABV and may provide important information for investigating RABV pathogenesis and therapeutic methods.
Collapse
Affiliation(s)
- Jundan Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Wangchang Li
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Dongling Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Rong Jin
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Hualin Hou
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Xiaoqing Ling
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Abraha Bahlbi Kiflu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
| | - Xiankai Wei
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
| | - Xiaogan Yang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- 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
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
| | - Yongming He
- School of Life Science and Engineering, Foshan University, Foshan 528225, China
| | - Ting Rong Luo
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangxi University, Nanning 530004, China
- College of Animal Sciences and Veterinary Medicine, Guangxi University, Nanning 530004, China
- Guangxi Key Laboratory of Animal Breeding, Disease Control and Prevention, Nanning 530004, China
- Guangxi Zhuang Autonomous Region Engineering Research Center of Veterinary Biologics, Nanning 530004, China
| |
Collapse
|
15
|
The Amino Acid at Position 95 in the Matrix Protein of Rabies Virus Is Involved in Antiviral Stress Granule Formation in Infected Cells. J Virol 2022; 96:e0081022. [PMID: 36069552 PMCID: PMC9517722 DOI: 10.1128/jvi.00810-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Stress granules (SGs) are dynamic structures that store cytosolic messenger ribonucleoproteins. SGs have recently been shown to serve as a platform for activating antiviral innate immunity; however, several pathogenic viruses suppress SG formation to evade innate immunity. In this study, we investigated the relationship between rabies virus (RABV) virulence and SG formation, using viral strains with different levels of virulence. We found that the virulent Nishigahara strain did not induce SG formation, but its avirulent offshoot, the Ni-CE strain, strongly induced SG formation. Furthermore, we demonstrated that the amino acid at position 95 in the RABV matrix protein (M95), a pathogenic determinant for the Nishigahara strain, plays a key role in inhibiting SG formation, followed by protein kinase R (PKR)-dependent phosphorylation of the α subunit of eukaryotic initiation factor 2α (eIF2α). M95 was also implicated in the accumulation of RIG-I, a viral RNA sensor protein, in SGs and in the subsequent acceleration of interferon induction. Taken together, our findings strongly suggest that M95-related inhibition of SG formation contributes to the pathogenesis of RABV by allowing the virus to evade the innate immune responses of the host. IMPORTANCE Rabies virus (RABV) is a neglected zoonotic pathogen that causes lethal infections in almost all mammalian hosts, including humans. Recently, RABV has been reported to induce intracellular formation of stress granules (SGs), also known as platforms that activate innate immune responses. However, the relationship between SG formation capacity and pathogenicity of RABV has remained unclear. In this study, by comparing two RABV strains with completely different levels of virulence, we found that the amino acid mutation from valine to alanine at position 95 of matrix protein (M95), which is known to be one of the amino acid mutations that determine the difference in virulence between the strains, plays a major role in SG formation. Importantly, M95 was involved in the accumulation of RIG-I in SGs and in promoting interferon induction. These findings are the first report of the effect of a single amino acid substitution associated with SGs on viral virulence.
Collapse
|
16
|
Cartwright HN, Barbeau DJ, Doyle JD, Klein E, Heise MT, Ferris MT, McElroy AK. Genetic diversity of collaborative cross mice enables identification of novel rift valley fever virus encephalitis model. PLoS Pathog 2022; 18:e1010649. [PMID: 35834486 PMCID: PMC9282606 DOI: 10.1371/journal.ppat.1010649] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 06/07/2022] [Indexed: 11/18/2022] Open
Abstract
Rift Valley fever (RVF) is an arboviral disease of humans and livestock responsible for severe economic and human health impacts. In humans, RVF spans a variety of clinical manifestations, ranging from an acute flu-like illness to severe forms of disease, including late-onset encephalitis. The large variations in human RVF disease are inadequately represented by current murine models, which overwhelmingly die of early-onset hepatitis. Existing mouse models of RVF encephalitis are either immunosuppressed, display an inconsistent phenotype, or develop encephalitis only when challenged via intranasal or aerosol exposure. In this study, the genetically defined recombinant inbred mouse resource known as the Collaborative Cross (CC) was used to identify mice with additional RVF disease phenotypes when challenged via a peripheral foot-pad route to mimic mosquito-bite exposure. Wild-type Rift Valley fever virus (RVFV) challenge of 20 CC strains revealed three distinct disease phenotypes: early-onset hepatitis, mixed phenotype, and late-onset encephalitis. Strain CC057/Unc, with the most divergent phenotype, which died of late-onset encephalitis at a median of 11 days post-infection, is the first mouse strain to develop consistent encephalitis following peripheral challenge. CC057/Unc mice were directly compared to C57BL/6 mice, which uniformly succumb to hepatitis within 2–4 days of infection. Encephalitic disease in CC057/Unc mice was characterized by high viral RNA loads in brain tissue, accompanied by clearance of viral RNA from the periphery, low ALT levels, lymphopenia, and neutrophilia. In contrast, C57BL/6 mice succumbed from hepatitis at 3 days post-infection with high viral RNA loads in the liver, viremia, high ALT levels, lymphopenia, and thrombocytopenia. The identification of a strain of CC mice as an RVFV encephalitis model will allow for future investigation into the pathogenesis and treatment of RVF encephalitic disease and indicates that genetic background makes a major contribution to RVF disease variation.
Collapse
Affiliation(s)
- Haley N. Cartwright
- University of Pittsburgh, School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Disease, and Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America
| | - Dominique J. Barbeau
- University of Pittsburgh, School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Disease, and Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America
| | - Joshua D. Doyle
- University of Pittsburgh, School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Disease, and Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America
| | - Ed Klein
- University of Pittsburgh, Division of Laboratory Animal Resources, Pittsburgh, Pennsylvania, United States of America
| | - Mark T. Heise
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Martin T. Ferris
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Anita K. McElroy
- University of Pittsburgh, School of Medicine, Department of Pediatrics, Division of Pediatric Infectious Disease, and Center for Vaccine Research, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
17
|
Schöler L, Le-Trilling VTK, Dittmer U, Fiedler M, Trilling M. Establishment and clinical validation of an in-cell-ELISA-based assay for the rapid quantification of Rabies lyssavirus neutralizing antibodies. PLoS Negl Trop Dis 2022; 16:e0010425. [PMID: 35536867 PMCID: PMC9159627 DOI: 10.1371/journal.pntd.0010425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 06/01/2022] [Accepted: 04/18/2022] [Indexed: 11/19/2022] Open
Abstract
Neutralizing antibodies (nAbs) prevent the entry of viruses into permissive cells. Since nAbs represent correlates of protection against the Rabies lyssavirus, the presence of sufficient nAbs indicates effective vaccination. Accordingly, Rabies lyssavirus-specific nAb titers need to be determined in routine diagnostics to identify individuals being at risk of Rabies lyssavirus infections due to insufficient immunity. The current gold standard for the quantification of Rabies lyssavirus-specific nAbs is the rapid fluorescent focus inhibition test (RFFIT). However, RFFITs are expensive and labor-intensive since multiple microplate wells must be evaluated one-by-one by trained personnel through microscopic inspection, which limits the number of samples that can be processed. To overcome this disadvantage, we established a novel assay for Rabies lyssavirus-specific nAbs relying on an in-cell-ELISA (icELISA)-based neutralization test (icNT). The icNT differs from the RFFIT in the readout phase, and can be automatically quantified in minutes using broadly available microplate readers. During the establishment, icNT parameters such as antibody concentrations, permeabilization procedures, blocking reagents, infectious doses, and the duration of infection were optimized. Afterwards, a dose-dependent detection of Rabies lyssavirus neutralization was demonstrated using the WHO Standard Rabies Immunoglobulin reference. A panel of 200 sera with known RFFIT titers revealed very good sensitivity and specificity of the icNT. Furthermore, the icNT showed very good intra- and inter-assay precision. By recognizing Rabies lyssavirus-specific antigens, the assay can be applied immediately to automatically quantify the concentration of Rabies lyssavirus nAbs in routine diagnostics or for various basic research questions such as screening for antiviral compounds.
Collapse
Affiliation(s)
- Lara Schöler
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | | | - Ulf Dittmer
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Melanie Fiedler
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Mirko Trilling
- Institute for Virology, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
18
|
Itakura Y, Tabata K, Morimoto K, Ito N, Chambaro HM, Eguchi R, Otsuguro KI, Hall WW, Orba Y, Sawa H, Sasaki M. Glu333 in rabies virus glycoprotein is involved in virus attenuation through astrocyte infection and interferon responses. iScience 2022; 25:104122. [PMID: 35402872 PMCID: PMC8983343 DOI: 10.1016/j.isci.2022.104122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/10/2022] [Accepted: 03/16/2022] [Indexed: 11/29/2022] Open
Abstract
The amino acid residue at position 333 of the rabies virus (RABV) glycoprotein (G333) is a major determinant of RABV pathogenicity. Virulent RABV strains possess Arg333, whereas the attenuated strain HEP-Flury (HEP) possesses Glu333. To investigate the potential attenuation mechanism dependent on a single amino acid at G333, comparative analysis was performed between HEP and HEP333R mutant with Arg333. We examined their respective tropism for astrocytes and the subsequent immune responses in astrocytes. Virus replication and subsequent interferon (IFN) responses in astrocytes infected with HEP were increased compared with HEP333R both in vitro and in vivo. Furthermore, involvement of IFN in the avirulency of HEP was demonstrated in IFN-receptor knockout mice. These results indicate that Glu333 contributes to RABV attenuation by determining the ability of the virus to infect astrocytes and stimulate subsequent IFN responses. Glu333 in G protein is responsible for astrocyte infection with RABV HEP strain Arg333 mutation in G protein decreases astrocyte tropism of RABV HEP RABV HEP evokes higher IFN responses in astrocytes than HEP with Arg333 mutation Glu333-dependent astrocyte infection is involved in the attenuation of RABV HEP
Collapse
Affiliation(s)
- Yukari Itakura
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Koshiro Tabata
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Kohei Morimoto
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Naoto Ito
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, Gifu, Gifu 501-1193, Japan
| | - Herman M. Chambaro
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Ryota Eguchi
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - Ken-ichi Otsuguro
- Laboratory of Pharmacology, Department of Basic Veterinary Sciences, Faculty of Veterinary Medicine, Hokkaido University, Sapporo, Hokkaido 060-0818, Japan
| | - William W. Hall
- National Virus Reference Laboratory, School of Medicine, University College of Dublin, Dublin 4, Ireland
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Global Virus Network, Baltimore, MD 21201, USA
| | - Yasuko Orba
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Hirofumi Sawa
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Global Virus Network, Baltimore, MD 21201, USA
- One Health Research Center, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
| | - Michihito Sasaki
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Hokkaido 001-0020, Japan
- Corresponding author
| |
Collapse
|
19
|
Naqvi AAT, Anjum F, Shafie A, Badar S, Elasbali AM, Yadav DK, Hassan MI. Investigating host-virus interaction mechanism and phylogenetic analysis of viral proteins involved in the pathogenesis. PLoS One 2021; 16:e0261497. [PMID: 34914801 PMCID: PMC8675761 DOI: 10.1371/journal.pone.0261497] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/02/2021] [Indexed: 02/07/2023] Open
Abstract
Since the emergence of yellow fever in the Americas and the devastating 1918 influenza pandemic, biologists and clinicians have been drawn to human infecting viruses to understand their mechanisms of infection better and develop effective therapeutics against them. However, the complex molecular and cellular processes that these viruses use to infect and multiply in human cells have been a source of great concern for the scientific community since the discovery of the first human infecting virus. Viral disease outbreaks, such as the recent COVID-19 pandemic caused by a novel coronavirus, have claimed millions of lives and caused significant economic damage worldwide. In this study, we investigated the mechanisms of host-virus interaction and the molecular machinery involved in the pathogenesis of some common human viruses. We also performed a phylogenetic analysis of viral proteins involved in host-virus interaction to understand the changes in the sequence organization of these proteins during evolution for various strains of viruses to gain insights into the viral origin's evolutionary perspectives.
Collapse
Affiliation(s)
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Sufian Badar
- Department of Computer Science, Jamia Millia Islamia, New Delhi, India
| | - Abdelbaset Mohamed Elasbali
- Clinical Laboratory Science, College of Applied Medical Sciences-Qurayyat, Jouf University, Sakakah, Saudi Arabia
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsu-gu, Incheon City, South Korea
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| |
Collapse
|
20
|
Research Advances on the Interactions between Rabies Virus Structural Proteins and Host Target Cells: Accrued Knowledge from the Application of Reverse Genetics Systems. Viruses 2021; 13:v13112288. [PMID: 34835093 PMCID: PMC8617671 DOI: 10.3390/v13112288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 11/17/2022] Open
Abstract
Rabies is a lethal zoonotic disease caused by lyssaviruses, such as rabies virus (RABV), that results in nearly 100% mortality once clinical symptoms appear. There are no curable drugs available yet. RABV contains five structural proteins that play an important role in viral replication, transcription, infection, and immune escape mechanisms. In the past decade, progress has been made in research on the pathogenicity of RABV, which plays an important role in the creation of new recombinant RABV vaccines by reverse genetic manipulation. Here, we review the latest advances on the interaction between RABV proteins in the infected host and the applied development of rabies vaccines by using a fully operational RABV reverse genetics system. This article provides a background for more in-depth research on the pathogenic mechanism of RABV and the development of therapeutic drugs and new biologics.
Collapse
|
21
|
Nitschel S, Zaeck LM, Potratz M, Nolden T, te Kamp V, Franzke K, Höper D, Pfaff F, Finke S. Point Mutations in the Glycoprotein Ectodomain of Field Rabies Viruses Mediate Cell Culture Adaptation through Improved Virus Release in a Host Cell Dependent and Independent Manner. Viruses 2021; 13:v13101989. [PMID: 34696419 PMCID: PMC8538267 DOI: 10.3390/v13101989] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/21/2021] [Accepted: 09/29/2021] [Indexed: 11/29/2022] Open
Abstract
Molecular details of field rabies virus (RABV) adaptation to cell culture replication are insufficiently understood. A better understanding of adaptation may not only reveal requirements for efficient RABV replication in cell lines, but may also provide novel insights into RABV biology and adaptation-related loss of virulence and pathogenicity. Using two recombinant field rabies virus clones (rRABV Dog and rRABV Fox), we performed virus passages in three different cell lines to identify cell culture adaptive mutations. Ten passages were sufficient for the acquisition of adaptive mutations in the glycoprotein G and in the C-terminus of phosphoprotein P. Apart from the insertion of a glycosylation sequon via the mutation D247N in either virus, both acquired additional and cell line-specific mutations after passages on BHK (K425N) and MDCK-II (R346S or R350G) cells. As determined by virus replication kinetics, complementation, and immunofluorescence analysis, the major bottleneck in cell culture replication was the intracellular accumulation of field virus G protein, which was overcome after the acquisition of the adaptive mutations. Our data indicate that limited release of extracellular infectious virus at the plasma membrane is a defined characteristic of highly virulent field rabies viruses and we hypothesize that the observed suboptimal release of infectious virions is due to the inverse correlation of virus release and virulence in vivo.
Collapse
Affiliation(s)
- Sabine Nitschel
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology (IMVZ), 17493 Greifswald-Insel Riems, Germany; (S.N.); (L.M.Z.); (M.P.); (T.N.); (V.t.K.)
| | - Luca M. Zaeck
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology (IMVZ), 17493 Greifswald-Insel Riems, Germany; (S.N.); (L.M.Z.); (M.P.); (T.N.); (V.t.K.)
| | - Madlin Potratz
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology (IMVZ), 17493 Greifswald-Insel Riems, Germany; (S.N.); (L.M.Z.); (M.P.); (T.N.); (V.t.K.)
| | - Tobias Nolden
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology (IMVZ), 17493 Greifswald-Insel Riems, Germany; (S.N.); (L.M.Z.); (M.P.); (T.N.); (V.t.K.)
| | - Verena te Kamp
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology (IMVZ), 17493 Greifswald-Insel Riems, Germany; (S.N.); (L.M.Z.); (M.P.); (T.N.); (V.t.K.)
| | - Kati Franzke
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Infectiology (IMED), 17493 Greifswald-Insel Riems, Germany;
| | - Dirk Höper
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Diagnostic Virology (IVD), 17493 Greifswald-Insel Riems, Germany; (D.H.); (F.P.)
| | - Florian Pfaff
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Diagnostic Virology (IVD), 17493 Greifswald-Insel Riems, Germany; (D.H.); (F.P.)
| | - Stefan Finke
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology (IMVZ), 17493 Greifswald-Insel Riems, Germany; (S.N.); (L.M.Z.); (M.P.); (T.N.); (V.t.K.)
- Correspondence: ; Tel.: +49-38351-71283
| |
Collapse
|
22
|
Kojima I, Izumi F, Ozawa M, Fujimoto Y, Okajima M, Ito N, Sugiyama M, Masatani T. Analyses of cell death mechanisms related to amino acid substitution at position 95 in the rabies virus matrix protein. J Gen Virol 2021; 102. [PMID: 33891533 DOI: 10.1099/jgv.0.001594] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We previously reported that the avirulent fixed rabies virus strain Ni-CE induces a clear cytopathic effect in mouse neuroblastoma cells, whereas its virulent progenitor, the Nishigahara strain, does not. Infection with Nishigahara and Ni-CE mutants containing a single amino acid substitution in the matrix protein (M) demonstrated that the amino acid at position 95 of M (M95) is a cytopathic determinant. The characteristics of cell death induced by Ni-CE infection resemble those of apoptosis (rounded and shrunken cells, DNA fragmentation), but the intracellular signalling pathway for this process has not been fully investigated. In this study, we aimed to elucidate the mechanism by which M95 affects cell death induced by human neuroblastoma cell infection with the Nishigahara, Ni-CE and M95-mutated strains. We demonstrated that the Ni-CE strain induced DNA fragmentation, cell membrane disruption, exposure of phosphatidylserine (PS), activation of caspase-3/7 and anti-poly (ADP-ribose) polymerase 1 (PARP-1) cleavage, an early apoptosis indicator, whereas the Nishigahara strain did not induce DNA fragmentation, caspase-3/7 activation, cell membrane disruption, or PARP-1 cleavage, but did induce PS exposure. We also demonstrated that these characteristics were associated with M95 using M95-mutated strains. However, we found that Ni-CE induced cell death despite the presence of a caspase inhibitor, Z-VAD-FMK. In conclusion, our data suggest that M95 mutation-related cell death is caused by both the caspase-dependent and -independent pathways.
Collapse
Affiliation(s)
- Isshu Kojima
- Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Fumiki Izumi
- Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Makoto Ozawa
- Laboratory of Animal Hygiene, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Yoshikazu Fujimoto
- Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Misuzu Okajima
- Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| | - Naoto Ito
- Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.,Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Makoto Sugiyama
- Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.,Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan
| | - Tatsunori Masatani
- Transboundary Animal Diseases Research Center, Joint Faculty of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan.,Laboratory of Zoonotic Diseases, Faculty of Applied Biological Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.,Joint Graduate School of Veterinary Sciences, Gifu University, 1-1 Yanagido, Gifu 501-1193, Japan.,Joint Graduate School of Veterinary Medicine, Kagoshima University, 1-21-24 Korimoto, Kagoshima 890-0065, Japan
| |
Collapse
|
23
|
Moore SM. Challenges of Rabies Serology: Defining Context of Interpretation. Viruses 2021; 13:1516. [PMID: 34452381 PMCID: PMC8402924 DOI: 10.3390/v13081516] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 12/25/2022] Open
Abstract
The case fatality rate of rabies, nearly 100%, is one of the most unique characteristic of this ancient virus infection. The crucial role rabies virus neutralizing antibody plays in protection is both well established and explanation of why rabies serology is important. Various laboratory methods can and have been used but serum neutralization methods have long been the gold standard due to the ability to measure function (neutralization), however these methods can be difficult to perform for several reasons. Assays such as enzyme linked absorbance assays (ELISA), indirect fluorescence antibody (IFA) and more recently lateral flow methods are in use. Interpretation of results can be problematic, not only between methods but also due to modifications of the same method that can lead to misinterpretations. A common assumption in review of laboratory test results is that different methods for the same component produce comparable results under all conditions or circumstances. Assumptions and misinterpretations provide the potential for detrimental decisions, ranging from regulatory to clinically related, and most importantly what 'level' is protective. Review of the common challenges in performance and interpretation of rabies serology and specific examples illuminate critical issues to consider when reviewing and applying results of rabies serological testing.
Collapse
Affiliation(s)
- Susan M Moore
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
| |
Collapse
|
24
|
Kiriwan D, Choowongkomon K. In silico structural elucidation of the rabies RNA-dependent RNA polymerase (RdRp) toward the identification of potential rabies virus inhibitors. J Mol Model 2021; 27:183. [PMID: 34031746 PMCID: PMC8143072 DOI: 10.1007/s00894-021-04798-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 05/17/2021] [Indexed: 11/25/2022]
Abstract
The rabies virus (RABV) is a non-segmented, negative single-stranded RNA virus which causes acute infection of the central nervous system in humans. Once symptoms appear, the result is nearly always death, and to date, post-exposure prophylaxis (PEP) is the only treatment applicable only immediately after an exposure. Previous studies have identified viral RNA-dependent RNA polymerase (RdRp) as a potential drug target due to its significant role in viral replication and transcription. Herein we generated an energy-minimized homology model of RABIES-RdRp and used it for virtual screening against 2045 NCI Diversity Set III library. The best five ligand-RdRp complexes were picked for further energy minimization via molecular dynamics (MDs) where the complex with ligand Z01690699 shows a minimum score characterized with stable hydrogen bonds and hydrophobic interactions with the catalytic site residues. Our study identified an important ligand for development of remedial approach for treatment of rabies infection.
Collapse
Affiliation(s)
- Duangnapa Kiriwan
- Interdisciplinary Program in Genetic Engineering, Graduate School, Kasetsart University, Chatuchak, Bangkok, Thailand
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand. .,Center for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand.
| |
Collapse
|
25
|
Udayantha HMV, Liyanage DS, Nadarajapillai K, Omeka WKM, Yang H, Jeong T, Lee J. Molecular characterization, immune and xenobiotic responses of glutathione S-transferase omega 1 from the big-belly seahorse: Novel insights into antiviral defense. FISH & SHELLFISH IMMUNOLOGY 2021; 109:62-70. [PMID: 33348035 DOI: 10.1016/j.fsi.2020.12.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/10/2020] [Accepted: 12/16/2020] [Indexed: 06/12/2023]
Abstract
Glutathione S-transferases (GSTs) are important enzymes involved in phase II detoxification and function by conjugating with the thiol group of glutathione. In this study, we isolated an omega class GST from the big-belly seahorse (Hippocampus abdominalis; HaGSTO1) to study the putative xenobiotic responses and defense ability against viral and bacterial infections in this animal. The isolated HaGSTO1 gene, with a cording sequence of 720 bp, encodes a peptide of 239 amino acids. The predicted molecular mass and theoretical isoelectric point of HaGSTO1 was 27.47 kDa and 8.13, respectively. In-silico analysis of HaGSTO1 revealed a characteristic N-terminal thioredoxin-like domain and a C-terminal domain. Unlike other GSTs, the C-terminal of HaGSTO1 reached up to the N-terminal, and the N-terminal functional group was cysteine rather than tyrosine or serine, as observed in other GSTs. Phylogenetic analysis showed the evolutionary proximity of HaGSTO1 with other identified vertebrate and invertebrate GST orthologs. For the first time, we demonstrated the viral defense capability of HaGSTO1 against viral hemorrhagic septicemia virus (VHSV) infection. All six nucleoproteins of VHSV were significantly downregulated in HaGSTO1-overexpressing FHM cells at 24 h after infection compared with those in the control. Moreover, arsenic toxicity was significantly reduced in HaGSTO1-overexpressing FHM cells, and cell viability increased. Real-time polymerase chain reaction analysis showed that HaGSTO1 transcripts were highly expressed in the pouch and gill when compared with those in other tissues. Blood HaGSTO1 transcripts were significantly upregulated after Edwardsiella tarda, Streptococcus iniae, lipopolysaccharide, and polyinosinic:polycytidylic acid challenge experiments. Collectively, these findings suggest the involvement of HaGSTO1 in the host defense mechanism of seahorses.
Collapse
Affiliation(s)
- H M V Udayantha
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - D S Liyanage
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Kishanthini Nadarajapillai
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - W K M Omeka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Hyerim Yang
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea
| | - Taehyug Jeong
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju Self-Governing Province, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju Self-Governing Province, 63333, Republic of Korea.
| |
Collapse
|
26
|
Katiyar N, Raju G, Madhusudanan P, Gopalakrishnan-Prema V, Shankarappa SA. Neuronal delivery of nanoparticles via nerve fibres in the skin. Sci Rep 2021; 11:2566. [PMID: 33510229 PMCID: PMC7844288 DOI: 10.1038/s41598-021-81995-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 01/13/2021] [Indexed: 11/13/2022] Open
Abstract
Accessing the peripheral nervous system (PNS) by topically applied nanoparticles is a simple and novel approach with clinical applications in several PNS disorders. Skin is richly innervated by long peripheral axons that arise from cell bodies located distally within ganglia. In this study we attempt to target dorsal root ganglia (DRG) neurons, via their axons by topical application of lectin-functionalized gold nanoparticles (IB4-AuNP). In vitro, 140.2 ± 1.9 nm IB4-AuNP were found to bind both axons and cell bodies of DRG neurons, and AuNP applied at the axonal terminals were found to translocate to the cell bodies. Topical application of IB4-AuNP on rat hind-paw resulted in accumulation of three to fourfold higher AuNP in lumbar DRG than in contralateral control DRGs. Results from this study clearly suggest that topically applied nanoparticles with neurotropic targeting ligands can be utilized for delivering nanoparticles to neuronal cell bodies via axonal transport mechanisms.
Collapse
Affiliation(s)
- Neeraj Katiyar
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Gayathri Raju
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Pallavi Madhusudanan
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Vignesh Gopalakrishnan-Prema
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India
| | - Sahadev A Shankarappa
- Center for Nanosciences and Molecular Medicine, Amrita Institute of Medical Sciences and Research Center, Amrita Vishwa Vidyapeetham University, Kochi, 682041, Kerala, India.
| |
Collapse
|
27
|
Potratz M, Zaeck LM, Weigel C, Klein A, Freuling CM, Müller T, Finke S. Neuroglia infection by rabies virus after anterograde virus spread in peripheral neurons. Acta Neuropathol Commun 2020; 8:199. [PMID: 33228789 PMCID: PMC7684951 DOI: 10.1186/s40478-020-01074-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2020] [Accepted: 11/07/2020] [Indexed: 12/17/2022] Open
Abstract
The highly neurotropic rabies virus (RABV) enters peripheral neurons at axon termini and requires long distance axonal transport and trans-synaptic spread between neurons for the infection of the central nervous system (CNS). Recent 3D imaging of field RABV-infected brains revealed a remarkably high proportion of infected astroglia, indicating that highly virulent field viruses are able to suppress astrocyte-mediated innate immune responses and virus elimination pathways. While fundamental for CNS invasion, in vivo field RABV spread and tropism in peripheral tissues is understudied. Here, we used three-dimensional light sheet and confocal laser scanning microscopy to investigate the in vivo distribution patterns of a field RABV clone in cleared high-volume tissue samples after infection via a natural (intramuscular; hind leg) and an artificial (intracranial) inoculation route. Immunostaining of virus and host markers provided a comprehensive overview of RABV infection in the CNS and peripheral nerves after centripetal and centrifugal virus spread. Importantly, we identified non-neuronal, axon-ensheathing neuroglia (Schwann cells, SCs) in peripheral nerves of the hind leg and facial regions as a target cell population of field RABV. This suggests that virus release from axons and infected SCs is part of the RABV in vivo cycle and may affect RABV-related demyelination of peripheral neurons and local innate immune responses. Detection of RABV in axon-surrounding myelinating SCs after i.c. infection further provided evidence for anterograde spread of RABV, highlighting that RABV axonal transport and spread of infectious virus in peripheral nerves is not exclusively retrograde. Our data support a new model in which, comparable to CNS neuroglia, SC infection in peripheral nerves suppresses glia-mediated innate immunity and delays antiviral host responses required for successful transport from the peripheral infection sites to the brain.
Collapse
|
28
|
Luo J, Zhang Y, Wang Y, Liu Q, Chen L, Zhang B, Luo Y, Huang S, Guo X. Rhabdovirus Infection Is Dependent on Serine/Threonine Kinase AP2-Associated Kinase 1. Life (Basel) 2020; 10:E170. [PMID: 32872567 PMCID: PMC7554979 DOI: 10.3390/life10090170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/24/2020] [Accepted: 08/28/2020] [Indexed: 01/01/2023] Open
Abstract
Rabies virus (RABV) causes a fatal neurological disease in both humans and animals. Understanding the mechanism of RABV infection is vital for prevention and therapy of virulent rabies infection. Our previous proteomics analysis based on isobaric tags for relative and absolute quantitation to identify factors revealed that RABV infection enhanced AP-2-associated protein kinase 1 (AAK1) in N2a cells. In this study, to further confirm the role of AAK1, we showed that RABV infection increased the transcription and expression of AAK1 in N2a cells. AAK1 knockdown significantly decreased RABV infection in both N2a and BHK-21 cells. AAK1 knockout inhibited RABV infection in N2a cells. Furthermore, inhibition of AAK1 kinase activity using sunitinib decreased RABV infection. However, AAK1 overexpression did not change RABV infection in vitro. Therapeutic administration of sunitinib did not significantly improve the survival rate of mice following lethal RABV challenge. In addition, AAK1 knockdown decreased infection in N2a cells by vesicular stomatitis virus, which is another rhabdovirus. These results indicate that rhabdovirus infection is dependent on AAK1 and inhibition of AAK1 is a potential strategy for the prevention and therapy of rabies.
Collapse
Affiliation(s)
- Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yang Wang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Qing Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Luman Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Yongwen Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA;
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130-3932, USA
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China; (J.L.); (Y.Z.); (Y.W.); (Q.L.); (L.C.); (B.Z.); (Y.L.)
| |
Collapse
|
29
|
Viruses in connectomics: Viral transneuronal tracers and genetically modified recombinants as neuroscience research tools. J Neurosci Methods 2020; 346:108917. [PMID: 32835704 DOI: 10.1016/j.jneumeth.2020.108917] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/25/2022]
Abstract
Connectomic studies have become 'viral', as viral pathogens have been turned into irreplaceable neuroscience research tools. Highly sensitive viral transneuronal tracing technologies are available, based on the use of alpha-herpesviruses and a rhabdovirus (rabies virus), which function as self-amplifying markers by replicating in recipient neurons. These viruses highly differ with regard to host range, cellular receptors, peripheral uptake, replication, transport direction and specificity. Their characteristics, that make them useful for different purposes, will be highlighted and contrasted. Only transneuronal tracing with rabies virus is entirely specific. The neuroscientist toolbox currently include wild-type alpha-herpesviruses and rabies virus strains enabling polysynaptic tracing of neuronal networks across multiple synapses, as well as genetically modified viral tracers for dual transneuronal tracing, and complementary viral tools including defective and chimeric recombinants that function as single step or monosynaptically restricted tracers, or serve for monitoring and manipulating neuronal activity and gene expression. Methodological issues that are crucial for appropriate use of these technologies will be summarized. Among wild-type and genetically engineered viral tools, rabies virus and chimeric recombinants based on rabies virus as virus backbone are the most powerful, because of the ability of rabies virus to propagate exclusively among connected neurons unidirectionally (retrogradely), without affecting neuronal function. Understanding in depth viral properties is essential for neuroscientists who intend to exploit alpha-herpesviruses, rhabdoviruses or derived recombinants as research tools. Key knowledge will be summarized regarding their cellular receptors, intracellular trafficking and strategies to contrast host defense that explain their different pathophysiology and properties as research tools.
Collapse
|
30
|
Astrocyte Infection during Rabies Encephalitis Depends on the Virus Strain and Infection Route as Demonstrated by Novel Quantitative 3D Analysis of Cell Tropism. Cells 2020; 9:cells9020412. [PMID: 32053954 PMCID: PMC7072253 DOI: 10.3390/cells9020412] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 12/25/2022] Open
Abstract
Although conventional immunohistochemistry for neurotropic rabies virus (RABV) usually shows high preference for neurons, non-neuronal cells are also potential targets, and abortive astrocyte infection is considered a main trigger of innate immunity in the CNS. While in vitro studies indicated differences between field and less virulent lab-adapted RABVs, a systematic, quantitative comparison of astrocyte tropism in vivo is lacking. Here, solvent-based tissue clearing was used to measure RABV cell tropism in infected brains. Immunofluorescence analysis of 1 mm-thick tissue slices enabled 3D-segmentation and quantification of astrocyte and neuron infection frequencies. Comparison of three highly virulent field virus clones from fox, dog, and raccoon with three lab-adapted strains revealed remarkable differences in the ability to infect astrocytes in vivo. While all viruses and infection routes led to neuron infection frequencies between 7–19%, striking differences appeared for astrocytes. Whereas astrocyte infection by field viruses was detected independent of the inoculation route (8–27%), only one lab-adapted strain infected astrocytes route-dependently [0% after intramuscular (i.m.) and 13% after intracerebral (i.c.) inoculation]. Two lab-adapted vaccine viruses lacked astrocyte infection altogether (0%, i.c. and i.m.). This suggests a model in which the ability to establish productive astrocyte infection in vivo functionally distinguishes field and attenuated lab RABV strains.
Collapse
|
31
|
Vigerelli H, Sciani JM, Pereira PMC, Lavezo AA, Silva ACR, Collaço RCO, Rocha T, Bueno TC, Pimenta DC. Bufotenine, a tryptophan-derived alkaloid, suppresses the symptoms and increases the survival rate of rabies-infected mice: the development of a pharmacological approach for rabies treatment. J Venom Anim Toxins Incl Trop Dis 2020; 26:e20190050. [PMID: 32071597 PMCID: PMC6996410 DOI: 10.1590/1678-9199-jvatitd-2019-0050] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Accepted: 12/19/2019] [Indexed: 12/25/2022] Open
Abstract
Background: Between 40,000-70,000 people die yearly of rabies, an incurable disease.
Besides post-bite vaccination, no treatment is available for it. Methods: First, virus dilution for antiviral effects in mice was determined. Then,
animals were treated as follows: control (NaCl 250 µL/animal/day);
bufotenine (0.63, 1.05 and 2.1 mg in 250 µL of NaCl/animal/day); rabies
(10-6,82CVS dilution); and test (10-6,82 CVS
dilution and bufotenine, in the above-mentioned doses). Animals were
observed daily for 21 days or until the 3rd stage of rabies
infection. Twitch-tension and liposome studies were applied to understand
the possible interaction of bufotenine with receptors, particularly
acetylcholine. Results: Bufotenine was able to increase the survival rate of intracerebrally
virus-infected mice from 15 to 40%. Bufotenine did not seem to interfere
with the acetylcholine response in the skeletal muscle, indicating that its
mechanism of action is not blocking the virus entrance due to nAChR
antagonism. By analyzing liposomes, we could observe that bufotenine did not
passively penetrates cell membranes, indicating the necessity of
complementary structures to cell penetration. Conclusions: Bufotenine is a promising candidate for drug development. After further
chemical modification, it might be possible to dissociate minor side
effects, increase efficiency, efficacy and pharmacokinetics, yielding a true
anti-rabies drug.
Collapse
Affiliation(s)
- Hugo Vigerelli
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São Paulo, SP, Brazil
| | - Juliana M Sciani
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São Paulo, SP, Brazil.,Multidisciplinary Research Laboratory, São Francisco University, Bragança Paulista, SP, Brazil
| | - Patricia M C Pereira
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São Paulo, SP, Brazil
| | - Aline A Lavezo
- Laboratory of Biological Quality Control in vivo, Butantan Institute, São Paulo, SP, Brazil
| | - Andrea C R Silva
- Laboratory of Rabies Diagnostic, Serology, Pasteur Institute, São Paulo, Brazil
| | - Rita C O Collaço
- Department of Pharmacology, State University of Campinas (Unicamp), Campinas, SP, Brazil
| | - Thalita Rocha
- Multidisciplinary Research Laboratory, São Francisco University, Bragança Paulista, SP, Brazil
| | - Thais C Bueno
- Multidisciplinary Research Laboratory, São Francisco University, Bragança Paulista, SP, Brazil
| | - Daniel C Pimenta
- Laboratory of Biochemistry and Biophysics, Butantan Institute, São Paulo, SP, Brazil
| |
Collapse
|
32
|
Klein A, Fahrion A, Finke S, Eyngor M, Novak S, Yakobson B, Ngoepe E, Phahladira B, Sabeta C, De Benedictis P, Gourlaouen M, Orciari LA, Yager PA, Gigante CM, Knowles MK, Fehlner-Gardiner C, Servat A, Cliquet F, Marston D, McElhinney LM, Johnson T, Fooks AR, Müller T, Freuling CM. Further Evidence of Inadequate Quality in Lateral Flow Devices Commercially Offered for the Diagnosis of Rabies. Trop Med Infect Dis 2020; 5:tropicalmed5010013. [PMID: 31963635 PMCID: PMC7157750 DOI: 10.3390/tropicalmed5010013] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 11/16/2022] Open
Abstract
As a neglected zoonotic disease, rabies causes approximately 5.9 × 104 human deaths annually, primarily affecting low- and middle-income countries in Asia and Africa. In those regions, insufficient surveillance is hampering adequate medical intervention and is driving the vicious cycle of neglect. Where resources to provide laboratory disease confirmation are limited, there is a need for user-friendly and low-cost reliable diagnostic tools that do not rely on specialized laboratory facilities. Lateral flow devices (LFD) offer an alternative to conventional diagnostic methods and may strengthen control efforts in low-resource settings. Five different commercially available LFDs were compared in a multi-centered study with respect to their diagnostic sensitivity and their agreement with standard rabies diagnostic techniques. Our evaluation was conducted by several international reference laboratories using a broad panel of samples. The overall sensitivities ranged from 0% up to 62%, depending on the LFD manufacturer, with substantial variation between the different laboratories. Samples with high antigen content and high relative viral load tended to test positive more often in the Anigen/Bionote test, the latter being the one with the best performance. Still, the overall unsatisfactory findings corroborate a previous study and indicate a persistent lack of appropriate test validation and quality control. At present, the tested kits are not suitable for in-field use for rabies diagnosis, especially not for suspect animals where human contact has been identified, as an incorrect negative diagnosis may result in human casualties. This study points out the discrepancy between the enormous need for such a diagnostic tool on the one hand, and on the other hand, a number of already existing tests that are not yet ready for use.
Collapse
Affiliation(s)
- Antonia Klein
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology, 17493 Greifswald-Insel Riems, Germany; (A.K.); (A.F.); (S.F.); (T.M.)
| | - Anna Fahrion
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology, 17493 Greifswald-Insel Riems, Germany; (A.K.); (A.F.); (S.F.); (T.M.)
| | - Stefan Finke
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology, 17493 Greifswald-Insel Riems, Germany; (A.K.); (A.F.); (S.F.); (T.M.)
| | - Marina Eyngor
- Kimron Veterinary Institute (KVI), Veterinary Services and Animal Health, P.O. Box 12, Beit Dagan 50250, Israel; (M.E.); (S.N.); (B.Y.)
| | - Shiri Novak
- Kimron Veterinary Institute (KVI), Veterinary Services and Animal Health, P.O. Box 12, Beit Dagan 50250, Israel; (M.E.); (S.N.); (B.Y.)
| | - Boris Yakobson
- Kimron Veterinary Institute (KVI), Veterinary Services and Animal Health, P.O. Box 12, Beit Dagan 50250, Israel; (M.E.); (S.N.); (B.Y.)
| | - Ernest Ngoepe
- Onderstepoort Veterinary Institute (OVI), Rabies Unit, Private Bag X05, Onderstepoort 0110, South Africa; (E.N.); (B.P.); (C.S.)
| | - Baby Phahladira
- Onderstepoort Veterinary Institute (OVI), Rabies Unit, Private Bag X05, Onderstepoort 0110, South Africa; (E.N.); (B.P.); (C.S.)
| | - Claude Sabeta
- Onderstepoort Veterinary Institute (OVI), Rabies Unit, Private Bag X05, Onderstepoort 0110, South Africa; (E.N.); (B.P.); (C.S.)
| | - Paola De Benedictis
- Istituto Zooprofilattico Sperimentale delle Venezie, FAO Reference Centre for Rabies, Viale dell’Università, 10, 35020-Legnaro (PD), Italy; (P.D.B.); (M.G.)
| | - Morgane Gourlaouen
- Istituto Zooprofilattico Sperimentale delle Venezie, FAO Reference Centre for Rabies, Viale dell’Università, 10, 35020-Legnaro (PD), Italy; (P.D.B.); (M.G.)
| | - Lillian A. Orciari
- Centers for Disease Control and Prevention (CDC), Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Atlanta, GA 30329, USA; (L.A.O.); (P.A.Y.); (C.M.G.)
| | - Pamela A. Yager
- Centers for Disease Control and Prevention (CDC), Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Atlanta, GA 30329, USA; (L.A.O.); (P.A.Y.); (C.M.G.)
| | - Crystal M. Gigante
- Centers for Disease Control and Prevention (CDC), Poxvirus and Rabies Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Atlanta, GA 30329, USA; (L.A.O.); (P.A.Y.); (C.M.G.)
| | - M. Kimberly Knowles
- Canadian Food Inspection Agency, Centre of Expertise for Rabies, Ottawa Laboratory Fallowfield, 3851 Fallowfield Road, Nepean, ON K2H 8P9, Canada; (M.K.K.); (C.F.-G.)
| | - Christine Fehlner-Gardiner
- Canadian Food Inspection Agency, Centre of Expertise for Rabies, Ottawa Laboratory Fallowfield, 3851 Fallowfield Road, Nepean, ON K2H 8P9, Canada; (M.K.K.); (C.F.-G.)
| | - Alexandre Servat
- French Agency for Food, Environmental and Occupational Health and Safety (Anses), Laboratory for Rabies and Wildlife, Domaine de Pixérécourt, 54220 Malzéville CEDEX, France; (A.S.); (F.C.)
| | - Florence Cliquet
- French Agency for Food, Environmental and Occupational Health and Safety (Anses), Laboratory for Rabies and Wildlife, Domaine de Pixérécourt, 54220 Malzéville CEDEX, France; (A.S.); (F.C.)
| | - Denise Marston
- Animal and Plant Health Agency (APHA), Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK; (D.M.); (L.M.M.); (T.J.); (A.R.F.)
| | - Lorraine M. McElhinney
- Animal and Plant Health Agency (APHA), Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK; (D.M.); (L.M.M.); (T.J.); (A.R.F.)
| | - Trudy Johnson
- Animal and Plant Health Agency (APHA), Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK; (D.M.); (L.M.M.); (T.J.); (A.R.F.)
| | - Anthony R. Fooks
- Animal and Plant Health Agency (APHA), Weybridge, New Haw, Addlestone, Surrey KT15 3NB, UK; (D.M.); (L.M.M.); (T.J.); (A.R.F.)
| | - Thomas Müller
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology, 17493 Greifswald-Insel Riems, Germany; (A.K.); (A.F.); (S.F.); (T.M.)
| | - Conrad M. Freuling
- Friedrich-Loeffler-Institut (FLI), Federal Research Institute for Animal Health, Institute of Molecular Virology and Cell Biology, 17493 Greifswald-Insel Riems, Germany; (A.K.); (A.F.); (S.F.); (T.M.)
- Correspondence: ; Tel.: +49-3835171660
| |
Collapse
|
33
|
Luo J, Zhang Y, Zhang Q, Wu Y, Zhang B, Mo M, Tian Q, Zhao J, Mei M, Guo X. The Deoptimization of Rabies Virus Matrix Protein Impacts Viral Transcription and Replication. Viruses 2019; 12:v12010004. [PMID: 31861477 PMCID: PMC7019236 DOI: 10.3390/v12010004] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 12/06/2019] [Accepted: 12/16/2019] [Indexed: 12/17/2022] Open
Abstract
Rabies virus (RABV) matrix (M) protein plays several important roles during RABV infection. Although previous studies have assessed the functions of M through gene rearrangements, this interferes with the position of other viral proteins. In this study, we attenuated M expression through deoptimizing its codon usage based on codon pair bias in RABV. This strategy more objectively clarifies the role of M during virus infection. Codon-deoptimized M inhibited RABV replication during the early stages of infection, but enhanced viral titers at later stages. Codon-deoptimized M also inhibited genome synthesis at early stage of infection and increased the RABV transcription rates. Attenuated M through codon deoptimization enhanced RABV glycoprotein expression following RABV infection in neuronal cells, but had no influence on the cell-to-cell spread of RABV. In addition, codon-deoptimized M virus induced higher levels of apoptosis compared to the parental RABV. These results indicate that codon-deoptimized M increases glycoprotein expression, providing a foundation for further investigation of the role of M during RABV infection.
Collapse
|
34
|
Doyle BM, Turner SM, Sunshine MD, Doerfler PA, Poirier AE, Vaught LA, Jorgensen ML, Falk DJ, Byrne BJ, Fuller DD. AAV Gene Therapy Utilizing Glycosylation-Independent Lysosomal Targeting Tagged GAA in the Hypoglossal Motor System of Pompe Mice. Mol Ther Methods Clin Dev 2019; 15:194-203. [PMID: 31660421 PMCID: PMC6807287 DOI: 10.1016/j.omtm.2019.08.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/23/2019] [Indexed: 12/17/2022]
Abstract
Pompe disease is caused by mutations in the gene encoding the lysosomal glycogen-metabolizing enzyme, acid-alpha glucosidase (GAA). Tongue myofibers and hypoglossal motoneurons appear to be particularly susceptible in Pompe disease. Here we used intramuscular delivery of adeno-associated virus serotype 9 (AAV9) for targeted delivery of an enhanced form of GAA to tongue myofibers and motoneurons in 6-month-old Pompe (Gaa -/- ) mice. We hypothesized that addition of a glycosylation-independent lysosomal targeting tag to the protein would result in enhanced expression in tongue (hypoglossal) motoneurons when compared to the untagged GAA. Mice received an injection into the base of the tongue with AAV9 encoding either the tagged or untagged enzyme; tissues were harvested 4 months later. Both AAV9 constructs effectively drove GAA expression in lingual myofibers and hypoglossal motoneurons. However, mice treated with the AAV9 construct encoding the modified GAA enzyme had a >200% increase in the number of GAA-positive motoneurons as compared to the untagged GAA (p < 0.008). Our results confirm that tongue delivery of AAV9-encoding GAA can effectively target tongue myofibers and associated motoneurons in Pompe mice and indicate that the effectiveness of this approach can be improved by addition of the glycosylation-independent lysosomal targeting tag.
Collapse
Affiliation(s)
- Brendan M. Doyle
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| | - Sara M.F. Turner
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| | - Michael D. Sunshine
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| | - Phillip A. Doerfler
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
- Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
| | - Amy E. Poirier
- Department of Neuroscience, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| | - Lauren A. Vaught
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
- Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| | - Marda L. Jorgensen
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
| | - Darin J. Falk
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
- Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| | - Barry J. Byrne
- Department of Pediatrics, University of Florida, Gainesville, FL 32610, USA
- Powell Gene Therapy Center, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| | - David D. Fuller
- Department of Physical Therapy, University of Florida, Gainesville, FL 32610, USA
- Mcknight Brain Institute, University of Florida, Gainesville, FL 32610, USA
- Center for Respiratory Research and Rehabilitation, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
35
|
Smith SP, Wu G, Fooks AR, Ma J, Banyard AC. Trying to treat the untreatable: experimental approaches to clear rabies virus infection from the CNS. J Gen Virol 2019; 100:1171-1186. [PMID: 31237530 DOI: 10.1099/jgv.0.001269] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Rabies virus causes an invariably fatal encephalitis following the onset of clinical disease. Despite the availability of safe and effective vaccines, the clinical stages of rabies encephalitis remain untreatable, with few survivors being documented. A principal obstacle to the treatment of rabies is the neurotropic nature of the virus, with the blood-brain barrier size exclusion limit rendering the delivery of antiviral drugs and molecules to the central nervous system inherently problematic. This review focuses on efforts to try and overcome barriers to molecule delivery to treat clinical rabies and overviews current progress in the development of experimental live rabies virus vaccines that may have future applications in the treatment of clinical rabies, including the attenuation of rabies virus vectors through either the duplication or mutation of existing genes or the incorporation of non-viral elements within the genome. Rabies post-infection treatment (PIT) remains the holy grail of rabies research.
Collapse
Affiliation(s)
- Samuel P Smith
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK.,Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK
| | - Guanghui Wu
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK
| | - Anthony R Fooks
- Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK.,Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK.,Department of Clinical Infection, Microbiology and Immunology, Institute of Infection and Global Health, University of Liverpool, Liverpool, UK
| | - Julian Ma
- Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK
| | - Ashley C Banyard
- Institute for Infection and Immunity, St George's Hospital Medical School, University of London, London, UK.,School of Life Sciences, University of West Sussex, Falmer, West Sussex, UK.,Wildlife Zoonoses and Vector-borne Diseases Research Group, Animal and Plant Health Agency (APHA), Addlestone, Surrey, KT15 3NB, UK
| |
Collapse
|
36
|
Wang L, Wu X, Bao J, Song C, Du J. Phylodynamic and transmission pattern of rabies virus in China and its neighboring countries. Arch Virol 2019; 164:2119-2129. [PMID: 31147766 DOI: 10.1007/s00705-019-04297-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 04/30/2019] [Indexed: 11/25/2022]
Abstract
Rabies is a fatal disease caused by infection with rabies virus (RABV), and human rabies is still a critical public-health concern in China. Although there have been some phylogenetic studies about RABV transmission patterns, with the accumulation of more rabies sequences in recent years, there is an urgent need to update and clarify the spatial and temporal patterns of RABV circulating in China on a national scale. In this study, we collected all available RABV nucleoprotein gene sequences from China and its neighboring countries and performed comparative analysis. We identified six significant subclades of RABV circulating in China and found that each of them has a specific geographical distribution, reflecting possible physical barriers to gene flow. The phylogeographic analysis revealed minimal viral movement among different geographical locations. An analysis using Bayesian coalescent methods indicated that the current RABV strains in China may come from a common ancestor about 400 years ago, and currently, China is amid the second event of increasing RABV population since the 1950s, but the population has decreased gradually. We did not detect any evidence of recombination in the sequence dataset, nor did we find any evidence for positive selection during the expansion of RABV. Overall, geographic location and neutral genetic drift may be the main factors in shaping the phylogeography of RABV transmission in China.
Collapse
Affiliation(s)
- Lina Wang
- Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Neurology, Xi'an Ninth Hospital Affiliated to Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xiaoming Wu
- Department of Neurology, Xi'an Ninth Hospital Affiliated to Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Junpeng Bao
- Department of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Changxin Song
- Department of Computer, Qinghai Normal University, Xining, Qinghai, China
| | - Jianqiang Du
- Key Laboratory of Biomedical Information Engineering of Education Ministry, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
37
|
Mei M, Long T, Zhang Q, Zhao J, Tian Q, Peng J, Luo J, Jiang H, Lin Y, Lin Z, Guo X. Phenotypic Consequence of Rearranging the N Gene of RABV HEP-Flury. Viruses 2019; 11:v11050402. [PMID: 31035728 PMCID: PMC6563252 DOI: 10.3390/v11050402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/20/2019] [Accepted: 04/25/2019] [Indexed: 02/08/2023] Open
Abstract
Nucleoprotein (N) is a key element in rabies virus (RABV) replication. To further investigate the effect of N on RABV, we manipulated an infectious cDNA clone of the RABV HEP-Flury to rearrange the N gene from its wild-type position of 1 (N-P-M-G-L) to 2 (P-N-M-G-L), 3 (P-M-N-G-L), or 4 (P-M-G-N-L), using an approach that left the viral nucleotide sequence unaltered. Subsequently, viable viruses were recovered from each of the rearranged cDNA and examined for their gene expression levels, growth kinetics in cell culture, pathogenicity in suckling mice and protection in mice. The results showed that gene rearrangement decreased N mRNA transcription and vRNA replication. As a result, all viruses with rearranged genomes showed worse replication than that of rHEP-Flury in NA cells at a MOI of 0.01, but equivalent or slightly better replication levels at a MOI of 3. Consequently, the lethality in suckling mice infected with N4 was clearly attenuated compared with rHEP-Flury. However, the protection to mice was not enhanced. This study not only gives us insight into the understanding of the phenotype of RABV N gene rearrangement, but also helps with rabies vaccine candidate construction.
Collapse
Affiliation(s)
- Mingzhu Mei
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
- Guangdong Inspection and Quarantine Technology Center, Guangzhou 510623, China.
| | - Teng Long
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Qiong Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jing Zhao
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Qin Tian
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jiaojiao Peng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - He Jiang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Yingyi Lin
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| | - Zhixiong Lin
- Guangdong Inspection and Quarantine Technology Center, Guangzhou 510623, China.
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
38
|
Guo Y, Duan M, Wang X, Gao J, Guan Z, Zhang M. Early events in rabies virus infection—Attachment, entry, and intracellular trafficking. Virus Res 2019; 263:217-225. [DOI: 10.1016/j.virusres.2019.02.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 01/28/2019] [Accepted: 02/13/2019] [Indexed: 12/20/2022]
|
39
|
Qin S, Volokhov D, Rodionova E, Wirblich C, Schnell MJ, Chizhikov V, Dabrazhynetskaya A. A new recombinant rabies virus expressing a green fluorescent protein: A novel and fast approach to quantify virus neutralizing antibodies. Biologicals 2019; 59:56-61. [PMID: 30898479 DOI: 10.1016/j.biologicals.2019.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Revised: 02/27/2019] [Accepted: 03/02/2019] [Indexed: 12/25/2022] Open
Abstract
The Rapid Fluorescent Focus Inhibition Test (RFFIT) is a standard assay used to detect and assess the titers of rabies virus neutralizing antibodies (RVNA) in blood sera. To simplify the multistep RFFIT procedure by eliminating the immunostaining step, we generated a new recombinant RV expressing a green fluorescent protein (rRV-GFP) and assess its suitability for quantifying RVNA. We rescued the rRV-GFP virus from plasmid DNA carrying a full-length genome of the CVS-N2c strain of RV in which the eGFP gene was inserted between the glycoprotein and RNA-polymerase genes. The recombinant virus was genetically stable and grew efficiently in appropriate cells expressing sufficient GFP fluorescence to detect directly 20 h post infection (hpi). We evaluated the feasibility of using rRV-GFP in RFFIT by comparing RVNA titers in 27 serum samples measured by conventional RFFIT and RFFIT-GFP. A linear regression analysis of the data demonstrated a good agreement between these two methods (r = 0.9776) including results with samples having RVNA titers close to the minimally acceptable vaccine potency threshold (0.5 IU/ml). Study results showed that the rRV-GFP virus could replace the CVS-11 challenge virus currently used in the conventional RFFIT and enabling more rapid, simpler, and less expensive detection and quantitation of RVNA.
Collapse
Affiliation(s)
- Shuyun Qin
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Dmitriy Volokhov
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Elvira Rodionova
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA
| | - Matthias J Schnell
- Department of Microbiology and Immunology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA, USA; Jefferson Vaccine Center at Thomas Jefferson University, Philadelphia, PA, USA
| | - Vladimir Chizhikov
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA
| | - Alena Dabrazhynetskaya
- Laboratory of Method Development, Division of Viral Products, Center for Biologics Evaluation, Food and Drug Administration, Silver Spring, MD, USA.
| |
Collapse
|
40
|
Okoh GR, Kazeem HM, Kia GS, Ponfa ZN. Heat induced epitope retrieval for rabies virus detection by direct fluorescent antibody test in formalin-fixed dog brain tissues. Open Vet J 2018; 8:313-317. [PMID: 30237978 PMCID: PMC6140382 DOI: 10.4314/ovj.v8i3.10] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/02/2018] [Indexed: 11/17/2022] Open
Abstract
There is a great need for a chemical method of tissue preservation that would allow sample storage for extended periods at room temperature. This study aimed at retrieving and detecting rabies virus antigen by direct fluorescent antibody test (DFAT) in formalin-fixed dog brain tissues. Forty fresh dog brain specimens were collected as paired samples from rabies suspected cases that were received for postmortem detection of rabies in the Central Diagnostic Laboratory, National Veterinary Research Institute, Vom. One portion of each paired sample was prepared for fresh fluorescent antibody testing and the other portion was prepared for epitope retrieval and florescent antibody testing following fixation in 10% neutral buffered formalin. DFAT on formalin-fixed tissue exhibited a sensitivity of 100% in comparison to DFAT on fresh-tissue. No false positive result was obtained in formalin-fixed DFAT procedure, demonstrating 100% specificity. There was no apparent difference in the intensity of fluorescence in DFAT on fresh sample and formalin-fixed DFAT following heat induced epitope retrieval (concordance = 98%; 95% C.I. 0.9660 to 0.9903). The strength of agreement between DFAT on formalin-fixed and DFAT on fresh tissue was very good (Cohen's kappa coefficient value= 1.000; 95% C.I. 1.000-1.000). This study provides new information on the retrieval of rabies antigen by heat induced epitope retrieval for DFAT on formalinized tissues. Formalin could therefore, be used henceforth to fix tissues of rabies suspected cases for routine diagnosis, transportation or archival purposes. The heat induced epitope retrieval can be routinely used to retrieve rabies virus antigen for DFAT in cases where only formalin-fixed tissues are available or when preservation by freezing is difficult.
Collapse
Affiliation(s)
- God’spower R. Okoh
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, University of Abuja, P.M.B. 117, Nigeria
| | - Haruna M. Kazeem
- Department of Veterinary Microbiology, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, P.M.B. 1045, Nigeria
| | - Grace S.N. Kia
- Department of Veterinary Public Health and Preventive Medicine, Faculty of Veterinary Medicine, Ahmadu Bello University, Zaria, P.M.B. 1045, Nigeria
- Center of Excellence in Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Zaria, Nigeria
| | - Zhakum N. Ponfa
- Central Diagnostic Laboratory, National Veterinary Research Institute, Vom, P.M.B. 01, Nigeria
| |
Collapse
|
41
|
Comprehensive Analysis of Codon Usage on Rabies Virus and Other Lyssaviruses. Int J Mol Sci 2018; 19:ijms19082397. [PMID: 30110957 PMCID: PMC6121662 DOI: 10.3390/ijms19082397] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2018] [Revised: 08/08/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
Rabies virus (RABV) and other lyssaviruses can cause rabies and rabies-like diseases, which are a persistent public health threat to humans and other mammals. Lyssaviruses exhibit distinct characteristics in terms of geographical distribution and host specificity, indicative of a long-standing diversification to adapt to the environment. However, the evolutionary diversity of lyssaviruses, in terms of codon usage, is still unclear. We found that RABV has the lowest codon usage bias among lyssaviruses strains, evidenced by its high mean effective number of codons (ENC) (53.84 ± 0.35). Moreover, natural selection is the driving force in shaping the codon usage pattern of these strains. In summary, our study sheds light on the codon usage patterns of lyssaviruses, which can aid in the development of control strategies and experimental research.
Collapse
|
42
|
Biological Effects and Biodistribution of Bufotenine on Mice. BIOMED RESEARCH INTERNATIONAL 2018; 2018:1032638. [PMID: 29955598 PMCID: PMC6000854 DOI: 10.1155/2018/1032638] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 05/02/2018] [Indexed: 12/25/2022]
Abstract
Bufotenine is an alkaloid derived from serotonin, structurally similar to LSD and psilocin. This molecule is able to inhibit the rabies virus infection in in vitro and in vivo models, increasing the survival rate of infected animals. Being a very promising molecule for an incurable disease and because of the fact that there is no consensus regarding its neurological effects, this study aimed to evaluate chronic treatment of bufotenine on behavior, pathophysiology, and pharmacokinetics of mice. Animals were daily treated for 21 consecutive days with 0.63, 1.05, and 2.1 mg/animal/day bufotenine and evaluated by open field test and physiological parameters during all the experiment. After this period, organs were collected for histopathological and biodistribution analysis. Animals treated with bufotenine had mild behavioral alterations compared to the control group, being dose-response relationship. On the other hand, animals showed normal physiological functions and no histological alterations in the organs. With high doses, an inflammatory reaction was observed in the site of injection, but with no cellular damage. The alkaloid could be found in the heart and kidney with all doses and in the lungs and brain with higher doses. These results show that the effective dose, 0.63 mg/day, is safe to be administered in mice, since it did not cause significant effects on the animals' physiology and on the CNS. Higher doses were well tolerated, causing only mild behavioral effects. Thus, bufotenine might be a drug prototype for rabies treatment, an incurable disease.
Collapse
|
43
|
Shafaghi M, Maktoobian S, Rasouli R, Howaizi N, Ofoghi H, Ehsani P. Transient Expression of Biologically Active Anti-rabies Virus Monoclonal Antibody in Tobacco Leaves. IRANIAN JOURNAL OF BIOTECHNOLOGY 2018; 16:e1774. [PMID: 30555840 PMCID: PMC6217261 DOI: 10.21859/ijb.1774] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Revised: 10/02/2017] [Accepted: 03/02/2018] [Indexed: 12/16/2022]
Abstract
Background Rabies virus is a neurotropic virus that causes fatal, but, a preventable disease in mammals. Administration of rabies immunoglobulin (RIG) is essential for the post-exposure of the prophylaxis to prevent the disease. However, replacement of polyclonal RIGs with alternative monoclonal antibodies (MAbs) that are capable of neutralizing rabies virus has been recommended. Objectives Here, we have investigated the transient expression of the full-size human MAb against rabies virus glycoprotein; the MAb SO57 in the tobacco plants using vacuum agro-infiltration. Previously, stably transformed plants expressing the MAb have been reported. Materials and Methods In this study three vectors carrying the codon-optimized genes for the heavy or light chain and p19 silencing-suppressor were constructed. These vectors were co-infiltrated into Nicotiana tabacum leaves and the transgenes were expressed. Results Dot blot, Western blotting, ELISA, and in vitro neutralization assays of the plant extracts showed that the human MAb could assemble in tobacco leaves and was able to neutralize rabies virus. Conclusions This study is the first report of transient expression of human MAb SO57 gene in tobacco plant within a few days after vacuum agro-infiltration.
Collapse
Affiliation(s)
- Mona Shafaghi
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Somayeh Maktoobian
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| | - Rahimeh Rasouli
- Department of Medical Nanotechnology, School of Medicine, International Campus, Tehran University of Medical Sciences, Tehran, Iran
| | - Nader Howaizi
- WHO Collaborating Centre for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Hamideh Ofoghi
- Department of Biotechnology, Iranian Research Organization for Science and Technology (IROST), Tehran, Iran
| | - Parastoo Ehsani
- Department of Molecular Biology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
44
|
Zhang Y, Zhou M, Li Y, Luo Z, Chen H, Cui M, Fu ZF, Zhao L. Recombinant rabies virus with the glycoprotein fused with a DC-binding peptide is an efficacious rabies vaccine. Oncotarget 2018; 9:831-841. [PMID: 29416659 PMCID: PMC5787516 DOI: 10.18632/oncotarget.23160] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/15/2017] [Indexed: 02/07/2023] Open
Abstract
Our previous studies demonstrated that recruiting and/or activating dendritic cells (DCs) enhanced the immunogenicity of recombinant rabies viruses (rRABV). In this study, rRABV LBNSE with a small DC-binding peptide (designated as rLBNSE-DCBp) or a negative control peptide (designated as rLBNSE-DCCp) fused to the glycoprotein (G) was constructed and rescued. As expected, significantly more activated DCs were detected in rLBNSE-DCBp-immunized mice than those immunized with rLBNSE or rLBNSE-DCCp. Subsequently, significantly more generation of TFH and GC B cells were observed in rLBNSE-DCBp immunized mice than those in rLBNSE or rLBNSE-DCCp-immunized mice. In addition, significantly higher levels of virus neutralizing antibodies (VNAs) were observed in mice immunized with rLBNSE-DCBp than those immunized with rLBNSE or rLBNSE-DCCp, resulting in a better protection of rLBNSE-DCBp immunized mice against the lethal challenge. Taken together, our results suggest that rRABV with G fused with DCBp is a promising rabies vaccine candidate.
Collapse
Affiliation(s)
- Yachun Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Ming Zhou
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Yingying Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhaochen Luo
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Huanchun Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Min Cui
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhen F. Fu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
- Department of Pathology, University of Georgia, Athens, GA 30602, USA
| | - Ling Zhao
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China
| |
Collapse
|
45
|
Papa MP, Meuren LM, Coelho SVA, Lucas CGDO, Mustafá YM, Lemos Matassoli F, Silveira PP, Frost PS, Pezzuto P, Ribeiro MR, Tanuri A, Nogueira ML, Campanati L, Bozza MT, Paula Neto HA, Pimentel-Coelho PM, Figueiredo CP, de Aguiar RS, de Arruda LB. Zika Virus Infects, Activates, and Crosses Brain Microvascular Endothelial Cells, without Barrier Disruption. Front Microbiol 2017; 8:2557. [PMID: 29312238 PMCID: PMC5743735 DOI: 10.3389/fmicb.2017.02557] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 12/08/2017] [Indexed: 12/19/2022] Open
Abstract
Zika virus (ZIKV) has been associated to central nervous system (CNS) harm, and virus was detected in the brain and cerebrospinal fluids of microcephaly and meningoencephalitis cases. However, the mechanism by which the virus reaches the CNS is unclear. Here, we addressed the effects of ZIKV replication in human brain microvascular endothelial cells (HBMECs), as an in vitro model of blood brain barrier (BBB), and evaluated virus extravasation and BBB integrity in an in vivo mouse experimental model. HBMECs were productively infected by African and Brazilian ZIKV strains (ZIKVMR766 and ZIKVPE243), which induce increased production of type I and type III IFN, inflammatory cytokines and chemokines. Infection with ZIKVMR766 promoted earlier cellular death, in comparison to ZIKVPE243, but infection with either strain did not result in enhanced endothelial permeability. Despite the maintenance of endothelial integrity, infectious virus particles crossed the monolayer by endocytosis/exocytosis-dependent replication pathway or by transcytosis. Remarkably, both viruses' strains infected IFNAR deficient mice, with high viral load being detected in the brains, without BBB disruption, which was only detected at later time points after infection. These data suggest that ZIKV infects and activates endothelial cells, and might reach the CNS through basolateral release, transcytosis or transinfection processes. These findings further improve the current knowledge regarding ZIKV dissemination pathways.
Collapse
Affiliation(s)
- Michelle P. Papa
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Lana M. Meuren
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sharton V. A. Coelho
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carolina G. de Oliveira Lucas
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Yasmin M. Mustafá
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Flavio Lemos Matassoli
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paola P. Silveira
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula S. Frost
- Núcleo de Neurociências da Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Paula Pezzuto
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Milene R. Ribeiro
- Laboratório de Pesquisas em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
| | - Amilcar Tanuri
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mauricio L. Nogueira
- Laboratório de Pesquisas em Virologia, Faculdade de Medicina de São José do Rio Preto, São José do Rio Preto, Brazil
| | - Loraine Campanati
- Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcelo T. Bozza
- Departamento de Imunologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Heitor A. Paula Neto
- Laboratório de Alvos Moleculares, Departamento de Biotecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Pedro M. Pimentel-Coelho
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudia P. Figueiredo
- Núcleo de Neurociências da Faculdade de Farmácia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Renato S. de Aguiar
- Departamento de Genética, Instituto de Biologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Luciana B. de Arruda
- Departamento de Virologia, Instituto de Microbiologia Paulo de Góes, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
46
|
Nicotinic acetylcholine receptor alpha 1(nAChRα1) subunit peptides as potential antiviral agents against rabies virus. Int J Biol Macromol 2017; 104:180-188. [DOI: 10.1016/j.ijbiomac.2017.05.179] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Revised: 05/29/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022]
|
47
|
Menegas W, Uchida N, Watabe-Uchida M. A Self-Killing Rabies Virus That Leaves a Trace on the DNA. Trends Neurosci 2017; 40:589-591. [PMID: 28890212 DOI: 10.1016/j.tins.2017.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 08/24/2017] [Indexed: 11/26/2022]
Abstract
Although modified rabies viruses have emerged as a powerful tool for tracing the inputs to genetically defined populations of neurons, the toxicity of the virus has limited its utility. A recent study employed a self-inactivating rabies (SiR) virus that enables recording or manipulation of targeted neurons for months.
Collapse
Affiliation(s)
- William Menegas
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Naoshige Uchida
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA
| | - Mitsuko Watabe-Uchida
- Center for Brain Science, Department of Molecular and Cellular Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
48
|
Prokaryotic Expression and Monoclonal Antibody Preparation of Rabies Virus Phosphoprotein. Jundishapur J Microbiol 2017. [DOI: 10.5812/jjm.13022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
49
|
Ohara S, Sota Y, Sato S, Tsutsui KI, Iijima T. Increased transgene expression level of rabies virus vector for transsynaptic tracing. PLoS One 2017; 12:e0180960. [PMID: 28700657 PMCID: PMC5507306 DOI: 10.1371/journal.pone.0180960] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Accepted: 06/23/2017] [Indexed: 12/27/2022] Open
Abstract
Viral vectors that can infect neurons transsynaptically and can strongly express foreign genes are useful for investigating the organization of neural circuits. We previously developed a propagation-competent rabies virus (RV) vector based on a highly attenuated HEP-Flury strain (rHEP5.0-CVSG), which selectively infects neurons and propagates between synaptically connected neurons in a retrograde direction. Its relatively low level of transgene expression, however, makes immunostaining necessary to visualize the morphological features of infected neurons. To increase the transgene expression level of this RV vector, in this study we focused on two viral proteins: the large protein (L) and matrix protein (M). We first attempted to enhance the expression of L, which is a viral RNA polymerase, by deleting the extra transcription unit and shortening the intergenic region between the G and L genes. This viral vector (rHEP5.0-GctL) showed increased transgene expression level with efficient transsynaptic transport. We next constructed an RV vector with a rearranged gene order (rHEP5.0-GML) with the aim to suppress the expression of M, which plays a regulatory role in virus RNA synthesis. Although this vector showed high transgene expression level, the efficiency of transsynaptic transport was low. To further evaluate the usability of rHEP5.0-GctL as a transsynaptic tracer, we inserted a fluorescent timer as a transgene, which changes the color of its fluorescence from blue to red over time. This viral vector enabled us the differentiation of primary infected neurons from secondary infected neurons in terms of the fluorescence wavelength. We expect this propagation-competent RV vector to be useful for elucidating the complex organization of the central nervous system.
Collapse
Affiliation(s)
- Shinya Ohara
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Yasuhiro Sota
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Sho Sato
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Ken-Ichiro Tsutsui
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
| | - Toshio Iijima
- Division of Systems Neuroscience, Tohoku University Graduate School of Life Sciences, Sendai, Japan
- * E-mail:
| |
Collapse
|
50
|
Life-Long Genetic and Functional Access to Neural Circuits Using Self-Inactivating Rabies Virus. Cell 2017; 170:382-392.e14. [PMID: 28689641 PMCID: PMC5509544 DOI: 10.1016/j.cell.2017.06.014] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Revised: 04/26/2017] [Accepted: 06/08/2017] [Indexed: 12/05/2022]
Abstract
Neural networks are emerging as the fundamental computational unit of the brain and it is becoming progressively clearer that network dysfunction is at the core of a number of psychiatric and neurodegenerative disorders. Yet, our ability to target specific networks for functional or genetic manipulations remains limited. Monosynaptically restricted rabies virus facilitates the anatomical investigation of neural circuits. However, the inherent cytotoxicity of the rabies largely prevents its implementation in long-term functional studies and the genetic manipulation of neural networks. To overcome this limitation, we developed a self-inactivating ΔG-rabies virus (SiR) that transcriptionally disappears from the infected neurons while leaving permanent genetic access to the traced network. SiR provides a virtually unlimited temporal window for the study of network dynamics and for the genetic and functional manipulation of neural circuits in vivo without adverse effects on neuronal physiology and circuit function. Self-inactivating rabies virus (SiR) provides life-long access to neural networks SiR has no adverse effects on neuronal physiology and network computations SiR allows open-ended Ca2+ imaging of network activity and functional intervention SiR allows generation of circuit-specific knockouts
Collapse
|