1
|
Porras AO, Morales MP, Santamaría G, Torres-Fernández O. Dendritic pathology and overexpression of MAP2 in Purkinje cells from mice inoculated with rabies virus. J Mol Histol 2024; 56:62. [PMID: 39739067 DOI: 10.1007/s10735-024-10348-5] [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: 09/05/2024] [Accepted: 12/22/2024] [Indexed: 01/02/2025]
Abstract
The effect of rabies virus infection on dendritic morphology and on the expression of the MAP2 protein in Purkinje cells in the cerebellum of mice was studied. ICR mice were inoculated with rabies virus, and six days later, the mice were sacrificed, the cerebellum was removed and processed for Golgi-Cox staining or MAP2 immunohistochemistry. Infection with rabies virus altered the dendritic pattern of Purkinje cells ranged from moderate changes to accentuated retraction in the dendritic tree of some Purkinje cells. The loss of dendritic branches in the samples of mice infected with RABV was also reflected in a decrease in intersections quantified using the Sholl technique, thus suggesting dendritic pathology. Immunoreactivity to MAP2 protein in the molecular layer of the cerebellum of control mice was mainly distributed in dendrites of Purkinje cells. Some somas were faintly stained. In infected mice immunoreactivity to MAP2 was intense in somas and dendrites of Purkinje cells and in some interneurons. These results are consistent with similar findings we previously reported for the cerebral cortex and spinal cord of rabies-infected mice. But they differ from studies in other pathologies where an association between dendritic pathology and loss of MAP2 immunoreactivity has been found. Our studies in rabies contribute to suggestion that MAP2 overexpression may also be associated with alterations in dendritic morphology. MAP2 protein contributes to maintaining cytoskeleton stability. However, in rabies, increased MAP2 expression here only determined by immunohistochemistry could destabilize the cytoskeleton of dendrites. Golgi staining is considered the gold standard for the study of dendritic morphology. Its association with changes in MAP2 expression appears to provide molecular support for the concept of dendritic pathology. These results contribute to the understanding of the effect of rabies virus infection on dendritic morphology. They therefore reinforce the idea that rabies not only has a dysfunctional effect on neurons, as some authors claim, but also affects their structure.
Collapse
Affiliation(s)
- Andrés Obdulio Porras
- Grupo de Morfología Celular, Instituto Nacional de Salud (INS), Av. Calle 26 No. 51-20, Bogotá, DC, 111321, Colombia
| | - María Paula Morales
- Grupo de Morfología Celular, Instituto Nacional de Salud (INS), Av. Calle 26 No. 51-20, Bogotá, DC, 111321, Colombia
| | - Gerardo Santamaría
- Grupo de Morfología Celular, Instituto Nacional de Salud (INS), Av. Calle 26 No. 51-20, Bogotá, DC, 111321, Colombia
| | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Instituto Nacional de Salud (INS), Av. Calle 26 No. 51-20, Bogotá, DC, 111321, Colombia.
| |
Collapse
|
2
|
Kiflu AB. The Immune Escape Strategy of Rabies Virus and Its Pathogenicity Mechanisms. Viruses 2024; 16:1774. [PMID: 39599888 PMCID: PMC11598914 DOI: 10.3390/v16111774] [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: 09/15/2024] [Revised: 11/01/2024] [Accepted: 11/06/2024] [Indexed: 11/29/2024] Open
Abstract
In contrast to most other rhabdoviruses, which spread by insect vectors, the rabies virus (RABV) is a very unusual member of the Rhabdoviridae family, since it has evolved to be fully adapted to warm-blooded hosts and spread directly between them. There are differences in the immune responses to laboratory-attenuated RABV and wild-type rabies virus infections. Various investigations showed that whilst laboratory-attenuated RABV elicits an innate immune response, wild-type RABV evades detection. Pathogenic RABV infection bypasses immune response by antagonizing interferon induction, which prevents downstream signal activation and impairs antiviral proteins and inflammatory cytokines production that could eliminate the virus. On the contrary, non-pathogenic RABV infection leads to immune activation and suppresses the disease. Apart from that, through recruiting leukocytes into the central nervous system (CNS) and enhancing the blood-brain barrier (BBB) permeability, which are vital factors for viral clearance and protection, cytokines/chemokines released during RABV infection play a critical role in suppressing the disease. Furthermore, early apoptosis of neural cells limit replication and spread of avirulent RABV infection, but street RABV strains infection cause delayed apoptosis that help them spread further to healthy cells and circumvent early immune exposure. Similarly, a cellular regulation mechanism called autophagy eliminates unused or damaged cytoplasmic materials and destroy microbes by delivering them to the lysosomes as part of a nonspecific immune defense mechanism. Infection with laboratory fixed RABV strains lead to complete autophagy and the viruses are eliminated. But incomplete autophagy during pathogenic RABV infection failed to destroy the viruses and might aid the virus in dodging detection by antigen-presenting cells, which could otherwise elicit adaptive immune activation. Pathogenic RABV P and M proteins, as well as high concentration of nitric oxide, which is produced during rabies virus infection, inhibits activities of mitochondrial proteins, which triggers the generation of reactive oxygen species, resulting in oxidative stress, contributing to mitochondrial malfunction and, finally, neuron process degeneration.
Collapse
Affiliation(s)
- 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
| |
Collapse
|
3
|
Liu X, Xu J, Zhang M, Wang H, Guo X, Zhao M, Duan M, Guan Z, Guo Y. RABV induces biphasic actin cytoskeletal rearrangement through Rac1 activity modulation. J Virol 2024; 98:e0060624. [PMID: 38809020 PMCID: PMC11264595 DOI: 10.1128/jvi.00606-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Accepted: 05/03/2024] [Indexed: 05/30/2024] Open
Abstract
Rabies virus (RABV) is highly lethal and triggers severe neurological symptoms. The neuropathogenic mechanism remains poorly understood. Ras-related C3 botulinum toxin substrate 1 (Rac1) is a Rho-GTPase that is involved in actin remodeling and has been reported to be closely associated with neuronal dysfunction. In this study, by means of a combination of pharmacological inhibitors, small interfering RNA, and specific dominant-negatives, we characterize the crucial roles of dynamic actin and the regulatory function of Rac1 in RABV infection, dominantly in the viral entry phase. The data show that the RABV phosphoprotein interacts with Rac1. RABV phosphoprotein suppress Rac1 activity and impedes downstream Pak1-Limk1-Cofilin1 signaling, leading to the disruption of F-actin-based structure formation. In early viral infection, the EGFR-Rac1-signaling pathway undergoes a biphasic change, which is first upregulated and subsequently downregulated, corresponding to the RABV entry-induced remodeling pattern of F-actin. Taken together, our findings demonstrate for the first time the role played by the Rac1 signaling pathway in RABV infection and may provide a clue for an explanation for the etiology of rabies neurological pathogenesis.IMPORTANCEThough neuronal dysfunction is predominant in fatal rabies, the detailed mechanism by which rabies virus (RABV) infection causes neurological symptoms remains in question. The actin cytoskeleton is involved in numerous viruses infection and plays a crucial role in maintaining neurological function. The cytoskeletal disruption is closely associated with abnormal nervous symptoms and induces neurogenic diseases. In this study, we show that RABV infection led to the rearrangement of the cytoskeleton as well as the biphasic kinetics of the Rac1 signal transduction. These results help elucidate the mechanism that causes the aberrant neuronal processes by RABV infection and may shed light on therapeutic development aimed at ameliorating neurological disorders.
Collapse
Affiliation(s)
- Xiaomin Liu
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Jing Xu
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Maolin Zhang
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Hualei Wang
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Xin Guo
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Mingxin Zhao
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Ming Duan
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Zhenhong Guan
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| | - Yidi Guo
- Institute of Zoonosis, College of Veterinary Medicine, Jilin University, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Changchun, China
| |
Collapse
|
4
|
Aliakbari S, Hasanzadeh L, Sayyah M, Amini N, Pourbadie HG. Induced expression of rabies glycoprotein in the dorsal hippocampus enhances hippocampal dependent memory in a rat model of Alzheimer's disease. J Neurovirol 2024; 30:274-285. [PMID: 38943023 DOI: 10.1007/s13365-024-01221-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 06/12/2024] [Accepted: 06/12/2024] [Indexed: 06/30/2024]
Abstract
The Rabies virus is a neurotropic virus that manipulates the natural cell death processes of its host to ensure its own survival and replication. Studies have shown that the anti-apoptotic effect of the virus is mediated by one of its protein named, rabies glycoprotein (RVG). Alzheimer's disease (AD) is characterized by the loss of neural cells and memory impairment. We aim to examine whether expression of RVG in the hippocampal cells can shield the detrimental effects induced by Aβ. Oligomeric form of Aβ (oAβ) or vehicle was bilaterally microinjected into the dorsal hippocampus of male Wistar rats. One week later, two μl (108 T.U. /ml) of the lentiviral vector carrying RVG gene was injected into their dorsal hippocampus (post-treatment). In another experiment, the lentiviral vector was microinjected one week before Aβ injection (pre-treatment). One week later, the rat's brain was sliced into cross-sections, and the presence of RVG-expressing neuronal cells was confirmed using fluorescent microscopy. Rats were subjected to assessments of spatial learning and memory as well as passive avoidance using the Morris water maze (MWM) and the Shuttle box apparatuses, respectively. Protein expression of AMPA receptor subunit (GluA1) was determined using western blotting technique. In MWM, Aβ treated rats showed decelerated acquisition of the task and impairment of reference memory. RVG expression in the hippocampus prevented and restored the deficits in both pre- and post- treatment conditions, respectively. It also improved inhibitory memory in the oAβ treated rats. RVG increased the expression level of GluA1 level in the hippocampus. Based on our findings, the expression of RVG in the hippocampus has the potential to enhance both inhibitory and spatial learning abilities, ultimately improving memory performance in an AD rat model. This beneficial effect is likely attributed, at least in part, to the increased expression of GluA1-containing AMPA receptors.
Collapse
Affiliation(s)
- Shayan Aliakbari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Leila Hasanzadeh
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Niloufar Amini
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | |
Collapse
|
5
|
Rengifo AC, Rivera J, Álvarez-Díaz DA, Naizaque J, Santamaria G, Corchuelo S, Gómez CY, Torres-Fernández O. Morphological and Molecular Changes in the Cortex and Cerebellum of Immunocompetent Mice Infected with Zika Virus. Viruses 2023; 15:1632. [PMID: 37631975 PMCID: PMC10458311 DOI: 10.3390/v15081632] [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: 05/31/2023] [Revised: 07/19/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Zika virus (ZIKV) disease continues to be a threat to public health, and it is estimated that millions of people have been infected and that there have been more cases of serious complications than those already reported. Despite many studies on the pathogenesis of ZIKV, several of the genes involved in the malformations associated with viral infection are still unknown. In this work, the morphological and molecular changes in the cortex and cerebellum of mice infected with ZIKV were evaluated. Neonatal BALB/c mice were inoculated with ZIKV intraperitoneally, and the respective controls were inoculated with a solution devoid of the virus. At day 10 postinoculation, the mice were euthanized to measure the expression of the markers involved in cortical and cerebellar neurodevelopment. The infected mice presented morphological changes accompanied by calcifications, as well as a decrease in most of the markers evaluated in the cortex and cerebellum. The modifications found could be predictive of astrocytosis, dendritic pathology, alterations in the regulation systems of neuronal excitation and inhibition, and premature maturation, conditions previously described in other models of ZIKV infection and microcephaly.
Collapse
Affiliation(s)
- Aura Caterine Rengifo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Jorge Rivera
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Diego Alejandro Álvarez-Díaz
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
- Genómica de Microorganismos Emergentes, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia
| | - Julián Naizaque
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Gerardo Santamaria
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Sheryll Corchuelo
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Claudia Yadira Gómez
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| | - Orlando Torres-Fernández
- Grupo de Morfología Celular, Dirección de Investigación en Salud Pública, Instituto Nacional de Salud (INS), Avenue 26 No. 51-20–Zone 6 CAN, Bogotá 111321, Colombia; (J.R.); (D.A.Á.-D.); (J.N.); (G.S.); (S.C.); (C.Y.G.); (O.T.-F.)
| |
Collapse
|
6
|
Toribio RE. Nonarboviral Equine Encephalitides. Vet Clin North Am Equine Pract 2022; 38:323-338. [PMID: 35811198 DOI: 10.1016/j.cveq.2022.04.007] [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: 10/17/2022] Open
Abstract
Several viruses transmitted by biological vectors or through direct contact, air, or ingestion cause neurologic disease in equids. Of interest are viruses of the Togaviridae, Flaviviridae, Rhabdoviridae, Herpesviridae, Bornaviridae, and Bunyaviridae families. Variable degree of inflammation is present with these viruses but lack of an inflammatory response does not rule out their presence. The goal of this article is to provide an overview on pathophysiologic and clinical aspects of nonarboviral equine encephalitides, specifically on lyssaviruses (rabies) and bornaviruses (Borna disease).
Collapse
Affiliation(s)
- Ramiro E Toribio
- College of Veterinary Medicine, The Ohio State University, 601 Vernon Tharp Street, Columbus, OH 43210, USA.
| |
Collapse
|
7
|
Liu X, Nawaz Z, Guo C, Ali S, Naeem MA, Jamil T, Ahmad W, Siddiq MU, Ahmed S, Asif Idrees M, Ahmad A. Rabies Virus Exploits Cytoskeleton Network to Cause Early Disease Progression and Cellular Dysfunction. Front Vet Sci 2022; 9:889873. [PMID: 35685339 PMCID: PMC9172992 DOI: 10.3389/fvets.2022.889873] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Accepted: 04/14/2022] [Indexed: 11/17/2023] Open
Abstract
Rabies virus (RABV) is a cunning neurotropic pathogen and causes top priority neglected tropical diseases in the developing world. The genome of RABV consists of nucleoprotein (N), phosphoprotein (P), matrix protein (M), glycoprotein (G), and RNA polymerase L protein (L), respectively. The virus causes neuronal dysfunction instead of neuronal cell death by deregulating the polymerization of the actin and microtubule cytoskeleton and subverts the associated binding and motor proteins for efficient viral progression. These binding proteins mainly maintain neuronal structure, morphology, synaptic integrity, and complex neurophysiological pathways. However, much of the exact mechanism of the viral-cytoskeleton interaction is yet unclear because several binding proteins of the actin-microtubule cytoskeleton are involved in multifaceted pathways to influence the retrograde and anterograde axonal transport of RABV. In this review, all the available scientific results regarding cytoskeleton elements and their possible interactions with RABV have been collected through systematic methodology, and thereby interpreted to explain sneaky features of RABV. The aim is to envisage the pathogenesis of RABV to understand further steps of RABV progression inside the cells. RABV interacts in a number of ways with the cell cytoskeleton to produce degenerative changes in the biochemical and neuropathological trails of neurons and other cell types. Briefly, RABV changes the gene expression of essential cytoskeleton related proteins, depolymerizes actin and microtubules, coordinates the synthesis of inclusion bodies, manipulates microtubules and associated motors proteins, and uses actin for clathrin-mediated entry in different cells. Most importantly, the P is the most intricate protein of RABV that performs complex functions. It artfully operates the dynein motor protein along the tracks of microtubules to assist the replication, transcription, and transport of RABV until its egress from the cell. New remedial insights at subcellular levels are needed to counteract the destabilization of the cytoskeleton under RABV infection to stop its life cycle.
Collapse
Affiliation(s)
- Xilin Liu
- Department of Hand Surgery, Presidents' Office of China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zeeshan Nawaz
- Department of Microbiology, Government College University Faisalabad, Faisalabad, Pakistan
| | - Caixia Guo
- Department of Hand Surgery, Presidents' Office of China-Japan Union Hospital of Jilin University, Changchun, China
| | - Sultan Ali
- Faculty of Veterinary Science, Institute of Microbiology, University of Agriculture, Faisalabad, Pakistan
| | - Muhammad Ahsan Naeem
- Department of Basic Sciences, University College of Veterinary and Animal Sciences, Narowal, Pakistan
| | - Tariq Jamil
- Department of Clinical Sciences, Section of Epidemiology and Public Health, College of Veterinary and Animal Sciences, Jhang, Pakistan
| | - Waqas Ahmad
- Department of Clinical Sciences, University College of Veterinary and Animal Sciences, Narowal, Pakistan
| | - Muhammad Usman Siddiq
- Institute of Microbiology, University of Veterinary and Animal Sciences, Lahore, Pakistan
| | - Sarfraz Ahmed
- Department of Basic Sciences, University College of Veterinary and Animal Sciences, Narowal, Pakistan
| | - Muhammad Asif Idrees
- Department of Pathobiology, University College of Veterinary and Animal Sciences, Narowal, Pakistan
| | - Ali Ahmad
- Department of Pathobiology, University College of Veterinary and Animal Sciences, Narowal, Pakistan
| |
Collapse
|
8
|
Appolinário CM, Daly JM, Emes RD, Marchi FA, Ribeiro BLD, Megid J. Gene Expression Profile Induced by Two Different Variants of Street Rabies Virus in Mice. Viruses 2022; 14:v14040692. [PMID: 35458422 PMCID: PMC9031335 DOI: 10.3390/v14040692] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/20/2022] [Accepted: 03/24/2022] [Indexed: 12/10/2022] Open
Abstract
Pathogenicity and pathology of rabies virus (RABV) varies according to the variant, but the mechanisms are not completely known. In this study, gene expression profile in brains of mice experimentally infected with RABV isolated from a human case of dog rabies (V2) or vampire bat-acquired rabies (V3) were analyzed. In total, 138 array probes associated with 120 genes were expressed differentially between mice inoculated with V2 and sham-inoculated control mice at day 10 post-inoculation. A single probe corresponding to an unannotated gene was identified in V3 versus control mice. Gene ontology (GO) analysis revealed that all of the genes upregulated in mice inoculated with V2 RABV were involved in the biological process of immune defense against pathogens. Although both variants are considered pathogenic, inoculation by the same conditions generated different gene expression results, which is likely due to differences in pathogenesis between the dog and bat RABV variants. This study demonstrated the global gene expression in experimental infection due to V3 wild-type RABV, from the vampire bat Desmodus rotundus, an important source of infection for humans, domestic animals and wildlife in Latin America.
Collapse
Affiliation(s)
- Camila M. Appolinário
- Faculdade de Medicina Veterinária e Zootecnia, Universidade Estadual Paulista, Julio de Mesquita Filho, Distrito de Rubião Júnior, s/n, CEP, Botucatu 18618-970, SP, Brazil;
- Correspondence: (C.M.A.); (J.M.)
| | - Janet M. Daly
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK; (J.M.D.); (R.D.E.)
| | - Richard D. Emes
- School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough LE12 5RD, UK; (J.M.D.); (R.D.E.)
| | | | - Bruna Leticia Devidé Ribeiro
- Faculdade de Medicina Veterinária e Zootecnia, Universidade Estadual Paulista, Julio de Mesquita Filho, Distrito de Rubião Júnior, s/n, CEP, Botucatu 18618-970, SP, Brazil;
| | - Jane Megid
- Faculdade de Medicina Veterinária e Zootecnia, Universidade Estadual Paulista, Julio de Mesquita Filho, Distrito de Rubião Júnior, s/n, CEP, Botucatu 18618-970, SP, Brazil;
- Correspondence: (C.M.A.); (J.M.)
| |
Collapse
|
9
|
Harsha PK, Ranganayaki S, Yale G, Dey G, Mangalaparthi KK, Yarlagadda A, Chandrasekhar Sagar BK, Mahadevan A, Srinivas Bharath MM, Mani RS. Mitochondrial Dysfunction in Rabies Virus-Infected Human and Canine Brains. Neurochem Res 2022; 47:1610-1636. [PMID: 35229271 DOI: 10.1007/s11064-022-03556-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 11/26/2022]
Abstract
Rabies is a fatal encephalitis caused by the Rabies lyssavirus (RABV). The presence of minimal neuropathological changes observed in rabies indicates that neuronal dysfunction, rather than neuronal death contributes to the fatal outcome. The role of mitochondrial changes has been suggested as a possible mechanism for neuronal dysfunction in rabies. However, these findings are mostly based on studies that have employed experimental models and laboratory-adapted virus. Studies on brain tissues from naturally infected human and animal hosts are lacking. The current study investigated the role of mitochondrial changes in rabies by morphological, biochemical and proteomic analysis of RABV-infected human and canine brains. Morphological analysis showed minimal inflammation with preserved neuronal and disrupted mitochondrial structure in both human and canine brains. Proteomic analysis revealed involvement of mitochondrial processes (oxidative phosphorylation, cristae formation, homeostasis and transport), synaptic proteins and autophagic pathways, with over-expression of subunits of mitochondrial respiratory complexes. Consistent with these findings, human and canine brains displayed elevated activities of complexes I (p < 0.05), IV (p < 0.05) and V (p < 0.05). However, this did not result in elevated ATP production (p < 0.0001), probably due to lowered mitochondrial membrane potential as noted in RABV-infected cells in culture. These could lead to mitochondrial dysfunction and mitophagy as indicated by expression of FKBP8 (p < 0.05) and PINK1 (p < 0.001)/PARKIN (p > 0.05) and ensuing autophagy, as shown by the status of LCIII (p < 0.05), LAMP1 (p < 0.001) and pertinent ultrastructural markers. We propose that altered mitochondrial bioenergetics and cristae architecture probably induce mitophagy, leading to autophagy and consequent neuronal dysfunction in rabies.
Collapse
Affiliation(s)
- Pulleri Kandi Harsha
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Sathyanarayanan Ranganayaki
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | | | - Gourav Dey
- Manipal Academy of Higher Education, Manipal, India
- Institute of Bioinformatics, Bangalore, India
| | | | - Anusha Yarlagadda
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - B K Chandrasekhar Sagar
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India
| | - M M Srinivas Bharath
- Department of Clinical Psychopharmacology and Neurotoxicology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.
| | - Reeta S Mani
- Department of Neurovirology, National Institute of Mental Health and Neurosciences (NIMHANS), Bangalore, India.
| |
Collapse
|
10
|
Comprehensive analysis of protein acetylation and glucose metabolism inmouse brains infected with rabies virus. J Virol 2021; 96:e0194221. [PMID: 34878915 DOI: 10.1128/jvi.01942-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Rabies, caused by rabies virus (RABV), is a widespread zoonosis that is nearly 100% fatal. Alteration of the metabolic environment affects viral replication and the immune response during viral infection. In this study, glucose uptake was increased in mouse brains at the late stage of infection with different RABV strains (lab-attenuated CVS strain and wild-type DRV strain). To illustrate the mechanism underlying glucose metabolism alteration, comprehensive analysis of lysine acetylation and target analysis of energy metabolites in mouse brains infected with CVS and DRV strains were performed. A total of 156 acetylated sites and 115 acetylated proteins were identified as significantly different during RABV infection. Compared to CVS- and mock-infected mice, the lysine acetylation levels of glycolysis and tricarboxylic acid (TCA) cycle enzymes were decreased, and enzyme activity was upregulated in DRV-infected mouse brains. Metabolomic analysis revealed that high levels of oxaloacetate (OAA) in RABV-infected mouse brains. Specifically, the OAA level in CVS-infected mouse brains was higher than that in DRV-infected mouse brains, which contributed to the enhancement of the metabolic rate at the substrate level. Finally, we confirmed that OAA could reduce excessive neuroinflammation in CVS-infected mouse brains by inhibiting JNK and P38 phosphorylation. Taken together, this study provides fresh insight into the different strategies the host adapts to regulate glucose metabolism for energy requirements after different RABV strain infection and suggest that OAA treatment could be a potential strategy to prevent neural damage during RABV infection. IMPORTANCE Both viral replication and the host immune response are highly energy-dependent. It is important to understand how the rabies virus affects energy metabolism in the brain. Glucose is the direct energy source for cell metabolism. Previous studies have revealed that there is some association between acetylation and metabolic processes. In this study, comprehensive protein acetylation and glucose metabolism analysis were conducted to compare glucose metabolism in mouse brains infected with different RABV strains. Our study demonstrates that the regulation of enzyme activity by acetylation and OAA accumulation at the substrate level are two strategies for the host to respond to the energy requirements after RABV infection. Our study also indicates the potential role OAA could play in neuronal protection by suppressing excessive neuroinflammation.
Collapse
|
11
|
Scott TP, Nel LH. Lyssaviruses and the Fatal Encephalitic Disease Rabies. Front Immunol 2021; 12:786953. [PMID: 34925368 PMCID: PMC8678592 DOI: 10.3389/fimmu.2021.786953] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/17/2021] [Indexed: 12/12/2022] Open
Abstract
Lyssaviruses cause the disease rabies, which is a fatal encephalitic disease resulting in approximately 59,000 human deaths annually. The prototype species, rabies lyssavirus, is the most prevalent of all lyssaviruses and poses the greatest public health threat. In Africa, six confirmed and one putative species of lyssavirus have been identified. Rabies lyssavirus remains endemic throughout mainland Africa, where the domestic dog is the primary reservoir - resulting in the highest per capita death rate from rabies globally. Rabies is typically transmitted through the injection of virus-laden saliva through a bite or scratch from an infected animal. Due to the inhibition of specific immune responses by multifunctional viral proteins, the virus usually replicates at low levels in the muscle tissue and subsequently enters the peripheral nervous system at the neuromuscular junction. Pathogenic rabies lyssavirus strains inhibit innate immune signaling and induce cellular apoptosis as the virus progresses to the central nervous system and brain using viral protein facilitated retrograde axonal transport. Rabies manifests in two different forms - the encephalitic and the paralytic form - with differing clinical manifestations and survival times. Disease symptoms are thought to be due mitochondrial dysfunction, rather than neuronal apoptosis. While much is known about rabies, there remain many gaps in knowledge about the neuropathology of the disease. It should be emphasized however, that rabies is vaccine preventable and dog-mediated human rabies has been eliminated in various countries. The global elimination of dog-mediated human rabies in the foreseeable future is therefore an entirely feasible goal.
Collapse
Affiliation(s)
| | - Louis Hendrik Nel
- Global Alliance for Rabies Control, Manhattan, KS, United States
- Department of Biochemistry, Genetics and Microbiology, Faculty of Natural and Agricultural Sciences, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
12
|
Feige L, Zaeck LM, Sehl-Ewert J, Finke S, Bourhy H. Innate Immune Signaling and Role of Glial Cells in Herpes Simplex Virus- and Rabies Virus-Induced Encephalitis. Viruses 2021; 13:2364. [PMID: 34960633 PMCID: PMC8708193 DOI: 10.3390/v13122364] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 12/19/2022] Open
Abstract
The environment of the central nervous system (CNS) represents a double-edged sword in the context of viral infections. On the one hand, the infectious route for viral pathogens is restricted via neuroprotective barriers; on the other hand, viruses benefit from the immunologically quiescent neural environment after CNS entry. Both the herpes simplex virus (HSV) and the rabies virus (RABV) bypass the neuroprotective blood-brain barrier (BBB) and successfully enter the CNS parenchyma via nerve endings. Despite the differences in the molecular nature of both viruses, each virus uses retrograde transport along peripheral nerves to reach the human CNS. Once inside the CNS parenchyma, HSV infection results in severe acute inflammation, necrosis, and hemorrhaging, while RABV preserves the intact neuronal network by inhibiting apoptosis and limiting inflammation. During RABV neuroinvasion, surveilling glial cells fail to generate a sufficient type I interferon (IFN) response, enabling RABV to replicate undetected, ultimately leading to its fatal outcome. To date, we do not fully understand the molecular mechanisms underlying the activation or suppression of the host inflammatory responses of surveilling glial cells, which present important pathways shaping viral pathogenesis and clinical outcome in viral encephalitis. Here, we compare the innate immune responses of glial cells in RABV- and HSV-infected CNS, highlighting different viral strategies of neuroprotection or Neuroinflamm. in the context of viral encephalitis.
Collapse
Affiliation(s)
- Lena Feige
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 28 Rue Du Docteur Roux, 75015 Paris, France;
| | - Luca M. Zaeck
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (L.M.Z.); (S.F.)
| | - Julia Sehl-Ewert
- Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany;
| | - Stefan Finke
- Institute of Molecular Virology and Cell Biology, Friedrich-Loeffler-Institut (FLI), Federal Institute of Animal Health, Südufer 10, 17493 Greifswald-Insel Riems, Germany; (L.M.Z.); (S.F.)
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology, 28 Rue Du Docteur Roux, 75015 Paris, France;
| |
Collapse
|
13
|
Farahtaj F, Alizadeh L, Gholami A, Khosravy MS, Bashar R, Gharibzadeh S, Mahmoodzadeh Niknam H, Ghaemi A. Differential pathogenesis of intracerebral and intramuscular inoculation of street rabies virus and CVS-11 strains in a mouse model. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2021; 24:943-950. [PMID: 34712425 PMCID: PMC8528248 DOI: 10.22038/ijbms.2021.54264.12188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 05/25/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVES The mechanisms of rabies evasion and immunological interactions with the host defense have not been completely elucidated. Here, we evaluated the dynamic changes in the number of astrocytes, microglial and neuronal cells in the brain following intramuscular (IM) and intracerebral (IC) inoculations of street rabies virus (SRV). MATERIALS AND METHODS The SRV isolated from a jackal and CVS-11 were used to establish infection in NMRI-female mice. The number of astrocytes (by expression of GFAP), microglial (by Iba1), and neuronal cells (by MAP-2) in the brain following IM and IC inoculations of SRV were evaluated by immunohistochemistry and H & E staining 7 to 30 days post-infection. RESULTS Increased numbers of astrocytes and microglial cells in dead mice infected by SRV via both IC and IM routes were recorded. The number of neuronal cells in surviving mice was decreased only in IC-infected mice, while in the dead group, this number was decreased by both routes.The risk of death in SRV-infected mice was approximately 3 times higher than in the CVS-11 group. In IC-inoculated mice, viral dilution was the only influential factor in mortality, while the type of strain demonstrated a significant impact on the mortality rate in IM inoculations. CONCLUSION Our results suggested that microglial cells and their inflammatory cytokines may not contribute to the neuroprotection and recovery in surviving mice following intracerebral inoculation of SRV. An unexpected decrease in MAP2 expression via intramuscular inoculation indicates the imbalance in the integrity and stability of neuronal cytoskeleton which aggravates rabies infection.
Collapse
Affiliation(s)
- Firozeh Farahtaj
- National Center for Reference & Research on Rabies, Institut Pasteur of Iran, Tehran, Iran
| | - Leila Alizadeh
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Gholami
- Viral vaccine Production, Pasteur Institute of Iran, Karaj, Iran
| | | | - Rouzbeh Bashar
- National Center for Reference & Research on Rabies, Institut Pasteur of Iran, Tehran, Iran
| | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics, Research Center for Emerging and Reemerging of Infectious Diseases, Institut Pasteur of Iran, Tehran, Iran
| | | | - Amir Ghaemi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran
| |
Collapse
|
14
|
Farahtaj F, Gholami A, Khosravy MS, Gharibzadeh S, Niknam HM, Ghaemi A. Enhancement of immune responses by co-stimulation of TLR3 - TLR7 agonists as a potential therapeutics against rabies in mouse model. Microb Pathog 2021; 157:104971. [PMID: 34029660 DOI: 10.1016/j.micpath.2021.104971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 04/18/2021] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
Rabies is always fatal, when post-exposure prophylaxis is administered after the onset of clinical symptoms. To date, there is no effective treatment of rabies once clinical symptoms has initiated. Therefore, we aimed to provide evidences which indicate the promising effects of combination treatment with TLR agonists following rabies infection. Four groups of rabies infected-mice (10-mice/group) were treated with PolyI:C 50 μg (a TLR3 agonist), Imiquimod50 μg (a TLR7 agonist), (Poly + Imi)25 μg and (Poly + Imi)50 μg respectively. The immune responses in each experimental groups were investigated in the brain through evaluation of GFAP, MAP2, CD4, HSP70, TLR3, TLR7 and apoptotic cell expression as well as determination of IFN-γ, TNF-α and IL-4, levels. The treatment with combination of agonists (Poly + Imi)50 μg/mouse resulted a 75% decrease of mortality rate and better extended survival time following street rabies virus infection. Higher number of CD4+T cells, TLR3 and TLR7 expression in the brain parenchyma observed in the groups receiving both combined agonist therapies at the levels of 25 μg and 50 μg. In spite of decreased number of neuronal cell, significant higher number of astrocytes was shown in the group given (Poly + Imi)25 μg. The obtained results also pointed to the dramatic decrease of HSP70 expression in all groups of infected mice whereas higher number of apoptotic cells and Caspase 8 expression were recorded in (Poly + Imi)25 μg treated group. Furthermore, the cytokine profile consisting the increased levels of TNF-α, IFN-γ and IL-4 revealed that both humoral and cellular responses were highly modulated in combination therapy of 50 μg of Imiquimod and Poly I:C. Reduced viral load as quantified by real-time PCR of rabies N gene expression in the brain also correlated with the better survival of agonist-treated groups of mice. Based on obtained results, we have presented evidences of beneficial utilization of combined agonist therapy composed of TLR3/TLR7 ligands. This treatment regimen extended survival of infected mice and decreased significantly their mortality rate. We believe that the results of synergy-inducing protection of both TLR3/TLR7 agonists lead to the enhancement of innate immune responses cells residing in the CNS which warrant the studies to further understanding of crosstalk mechanisms in cellular immunity against rabies in the future.
Collapse
Affiliation(s)
- Firouzeh Farahtaj
- Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | - Alireza Gholami
- Viral Vaccine Production, Pasteur Institute of Iran, Karaj, Iran
| | | | - Safoora Gharibzadeh
- Department of Epidemiology and Biostatistics, Research Center for Emerging and Reemerging of Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | | | - Amir Ghaemi
- Department of Virology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
15
|
Ghassemi S, Asgari T, Mirzapour-Delavar H, Aliakbari S, Pourbadie HG, Prehaud C, Lafon M, Gholami A, Azadmanesh K, Naderi N, Sayyah M. Lentiviral Expression of Rabies Virus Glycoprotein in the Rat Hippocampus Strengthens Synaptic Plasticity. Cell Mol Neurobiol 2021; 42:1429-1440. [PMID: 33462779 DOI: 10.1007/s10571-020-01032-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 12/16/2020] [Indexed: 11/30/2022]
Abstract
Rabies virus (RABV) is a neurotropic virus exclusively infecting neurons in the central nervous system. RABV encodes five proteins. Among them, the viral glycoprotein (RVG) plays a key role in viral entry into neurons and rabies pathogenesis. It was shown that the nature of the C-terminus of the RABV G protein, which possesses a PDZ-binding motif (PBM), modulates the virulence of the RABV strain. The neuronal protein partners recruited by this PBM may alter host cell function. This study was conducted to investigate the effect of RVG on synaptic function in the hippocampal dentate gyrus (DG) of rat. Two μl (108 T.U./ml) of the lentiviral vector containing RVG gene was injected into the DG of rat hippocampus. After 2 weeks, the rat's brain was cross-sectioned and RVG-expressing cells were detected by fluorescent microscopy. Hippocampal synaptic activity of the infected rats was then examined by recording the local field potentials from DG after stimulation of the perforant pathway. Short-term synaptic plasticity was also assessed by double pulse stimulation. Expression of RVG in DG increased long-term potentiation population spikes (LTP-PS), whereas no facilitation of LTP-PS was found in neurons expressing δRVG (deleted PBM). Furthermore, RVG and δRVG strengthened paired-pulse facilitation. Heterosynaptic long-term depression (LTD) in the DG was significantly blocked in RVG-expressing group compared to the control group. This blockade was dependent to PBM motif as rats expressing δRVG in the DG-expressed LTD comparable to the RVG group. Our data demonstrate that RVG expression facilitates both short- and long-term synaptic plasticity in the DG indicating that it may involve both pre- and postsynaptic mechanisms to alter synaptic function. Further studies are needed to elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Soheil Ghassemi
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | - Tara Asgari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Shayan Aliakbari
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran
| | | | - Christophe Prehaud
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris, France
| | - Monique Lafon
- Institut Pasteur, Unité de Neuroimmunologie Virale, UMR 3569, CNRS, Paris, France
| | - Alireza Gholami
- WHO Collaborating Center for Reference and Research on Rabies, Pasteur Institute of Iran, Tehran, Iran
| | | | - Nima Naderi
- Department of Pharmacology and Toxicology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Sayyah
- Department of Physiology and Pharmacology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
16
|
Lan Y, Li Z, Wang Z, Wang X, Wang G, Zhang J, Hu S, Zhao K, Xu B, Gao F, He W. An Experimental Model of Neurodegenerative Disease Based on Porcine Hemagglutinating Encephalomyelitis Virus-Related Lysosomal Abnormalities. Mol Neurobiol 2020; 57:5299-5306. [PMID: 32876841 PMCID: PMC7463228 DOI: 10.1007/s12035-020-02105-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/27/2020] [Indexed: 01/01/2023]
Abstract
Lysosomes are involved in pathogenesis of a variety of neurodegenerative diseases and play a large role in neurodegenerative disorders caused by virus infection. However, whether virus-infected cells or animals can be used as experimental models of neurodegeneration in humans based on virus-related lysosomal dysfunction remain unclear. Porcine hemagglutinating encephalomyelitis virus displays neurotropism in mice, and neural cells are its targets for viral progression. PHEV infection was confirmed to be a risk factor for neurodegenerative diseases in the present. The findings demonstrated for the first time that PHEV infection can lead to lysosome disorders and showed that the specific mechanism of lysosome dysfunction is related to PGRN expression deficiency and indicated similar pathogenesis compared with human neurodegenerative diseases upon PHEV infection. Trehalose can also increase progranulin expression and rescue abnormalities in lysosomal structure in PHEV-infected cells. In conclusion, these results suggest that PHEV probably serve as a disease model for studying the pathogenic mechanisms and prevention of other degenerative diseases.
Collapse
Affiliation(s)
- Yungang Lan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zi Li
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhenzhen Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xinran Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Gaili Wang
- Jilin Academy of Animal Husbandry and Veterinary Medicine, Changchun, Jilin, China
| | - Jing Zhang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shiyu Hu
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Kui Zhao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Baofeng Xu
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, 130021, China
| | - Feng Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Wenqi He
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
17
|
Fares M, Cochet-Bernoin M, Gonzalez G, Montero-Menei CN, Blanchet O, Benchoua A, Boissart C, Lecollinet S, Richardson J, Haddad N, Coulpier M. Pathological modeling of TBEV infection reveals differential innate immune responses in human neurons and astrocytes that correlate with their susceptibility to infection. J Neuroinflammation 2020; 17:76. [PMID: 32127025 PMCID: PMC7053149 DOI: 10.1186/s12974-020-01756-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Accepted: 02/21/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Tick-borne encephalitis virus (TBEV) is a member of the Flaviviridae family, Flavivirus genus, which includes several important human pathogens. It is responsible for neurological symptoms that may cause permanent disability or death, and, from a medical point of view, is the major arbovirus in Central/Northern Europe and North-Eastern Asia. TBEV tropism is critical for neuropathogenesis, yet little is known about the molecular mechanisms that govern the susceptibility of human brain cells to the virus. In this study, we sought to establish and characterize a new in vitro model of TBEV infection in the human brain and to decipher cell type-specific innate immunity and its relation to TBEV tropism and neuropathogenesis. METHOD Human neuronal/glial cells were differentiated from neural progenitor cells and infected with the TBEV-Hypr strain. Kinetics of infection, cellular tropism, and cellular responses, including innate immune responses, were characterized by measuring viral genome and viral titer, performing immunofluorescence, enumerating the different cellular types, and determining their rate of infection and by performing PCR array and qRT-PCR. The specific response of neurons and astrocytes was analyzed using the same approaches after enrichment of the neuronal/glial cultures for each cellular subtype. RESULTS We showed that infection of human neuronal/glial cells mimicked three major hallmarks of TBEV infection in the human brain, namely, preferential neuronal tropism, neuronal death, and astrogliosis. We further showed that these cells conserved their capacity to mount an antiviral response against TBEV. TBEV-infected neuronal/glial cells, therefore, represented a highly relevant pathological model. By enriching the cultures for either neurons or astrocytes, we further demonstrated qualitative and quantitative differential innate immune responses in the two cell types that correlated with their particular susceptibility to TBEV. CONCLUSION Our results thus reveal that cell type-specific innate immunity is likely to contribute to shaping TBEV tropism for human brain cells. They describe a new in vitro model for in-depth study of TBEV-induced neuropathogenesis and improve our understanding of the mechanisms by which neurotropic viruses target and damage human brain cells.
Collapse
Affiliation(s)
- Mazigh Fares
- UMR1161 Virologie, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
- MRC-University of Glasgow Centre for Virus Research, Glasgow, Scotland, UK
| | - Marielle Cochet-Bernoin
- UMR1161 Virologie, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Gaëlle Gonzalez
- UMR1161 Virologie, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Claudia N Montero-Menei
- CRCINA, UMR 1232, INSERM, Université de Nantes, Université d'Angers, F-49933, Angers, France
| | - Odile Blanchet
- Centre de Ressources Biologiques, CHU Angers, BB-0033-00038, Angers, France
| | | | | | - Sylvie Lecollinet
- UMR1161 Virologie, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Jennifer Richardson
- UMR1161 Virologie, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Nadia Haddad
- UMR BIPAR 956, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France
| | - Muriel Coulpier
- UMR1161 Virologie, Anses, INRAE, Ecole Nationale Vétérinaire d'Alfort, Université Paris-Est, Maisons-Alfort, France.
| |
Collapse
|
18
|
Ahmed MS, Body MH, El-Neweshy MS, ALrawahi AH, Al-Abdawani M, Eltahir HA, ALmaewaly MG. Molecular characterization and diagnostic investigations of rabies encephalitis in camels (Camelus dromedaries) in Oman: a retrospective study. Trop Anim Health Prod 2020; 52:2163-2168. [PMID: 32124182 DOI: 10.1007/s11250-020-02239-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/13/2020] [Indexed: 10/24/2022]
Abstract
The accurate and early diagnosis of rabies is critical for undertaking public health measures in animals. The aim of the current study was to identify the molecular characterization of the circulating rabies virus (RABV) among camels (Camelus dromedaries) in Oman and to evaluate the efficacy of the histopathology and reverse transcription polymerase chain reaction (RT-PCR) as diagnostic tools of acute rabies encephalitis in camels in comparison with direct fluorescent antibody test (dFAT). Of the forty-five brain samples from suspected camels submitted to the Animal Health Research Center in Oman (2009-2013), 22 cases were positive by dFAT and RT-PCR. Two positive samples were subjected for N gene nucleotide sequencing and phylogenetic analysis (accession numbers GU353156 and KC883998 for brain samples collected in 2009 and 2011, respectively). The specificity and sensitivity of histopathology were 100% and 81%, respectively, while in RT-PCR were 100% and 100%, respectively. The neuropathological changes were presence of intracytoplasmic inclusions (Negri bodies) in the pyramidal neurons of the hippocampus beside prominent cerebral and cerebellar congestion and hemorrhage. Neuronal necrosis with satellitosis and neuronophagia were also noticed in the cerebrum of affected brains. Conclusively, there was one genetic group of RABV with 99% homology circulating in Omani camels. Also, it is concluded that histopathological examination is a safe and reliable diagnostic tool when only formalin-fixed and paraffin-embedded material is available, but the negative results should be reaffirmed by dFAT or RT-PCR.
Collapse
Affiliation(s)
- Mohamed S Ahmed
- Animal Health Research Center, Directorate General of Agriculture and Livestock Research, Ministry of Agriculture and Fisheries, Muscat, Sultanate of Oman.,Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El Sheikh, 33516, Egypt
| | - Mohammad H Body
- Animal Health Research Center, Directorate General of Agriculture and Livestock Research, Ministry of Agriculture and Fisheries, Muscat, Sultanate of Oman
| | - Mahmoud S El-Neweshy
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, Behera, Alexandria, 22785, Egypt. .,Central Laboratory for Animal Health, Ministry of Agriculture and Fisheries, Muscat, Sultanate of Oman.
| | - Abdulmajeed H ALrawahi
- Animal Health Research Center, Directorate General of Agriculture and Livestock Research, Ministry of Agriculture and Fisheries, Muscat, Sultanate of Oman
| | - Mohsin Al-Abdawani
- Animal Health Research Center, Directorate General of Agriculture and Livestock Research, Ministry of Agriculture and Fisheries, Muscat, Sultanate of Oman
| | - Hatim A Eltahir
- Animal Health Research Center, Directorate General of Agriculture and Livestock Research, Ministry of Agriculture and Fisheries, Muscat, Sultanate of Oman.,Central Veterinary Research Laboratory, Khartoum, Sudan
| | - Mahir G ALmaewaly
- Animal Health Research Center, Directorate General of Agriculture and Livestock Research, Ministry of Agriculture and Fisheries, Muscat, Sultanate of Oman
| |
Collapse
|
19
|
Pei J, Huang F, Wu Q, Luo Z, Zhang Y, Ruan J, Li Y, Zhou M, Fu Z, Zhao L. Codon optimization of G protein enhances rabies virus-induced humoral immunity. J Gen Virol 2019; 100:1222-1233. [PMID: 31259681 DOI: 10.1099/jgv.0.001299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Rabies, caused by rabies virus (RABV), is a fatal zoonosis, which still poses a threat to public health in most parts of the world. Glycoprotein of RABV is the only viral surface protein, which is critical for the induction of virus-neutralizing antibodies (VNA). In order to improve the production of VNA, recombinant RABVs containing two copies of G gene and codon-optimized G gene were constructed by using reverse genetics, named LBNSE-dG and LBNSE-dOG, respectively. After being inoculated into the mouse brains, LBNSE-dOG induced more apoptosis and recruited more inflammatory cells than LBNSE-dG and LBNSE, resulting in reduced virulence in vivo. After intramuscular (im) immunization in mice, LBNSE-dOG promoted the formation of germinal centres (GCs), the recruitment of GC B cells and the generation of antibody-secreting cells (ASCs) in the draining lymph nodes (LNs). Consistently, LBNSE-dOG boosted the production of VNA and provided better protection against lethal RABV challenge than LBNSE-dG and LBNSE when it was used as both live and inactivated vaccines. Our results demonstrate that the codon-optimized RABV LBNSE-dOG displays attenuated pathogenicity and enhanced immunogenicity, therefore it could be a potential candidate for the next generation of rabies vaccines.
Collapse
Affiliation(s)
- Jie Pei
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China
| | - Fei Huang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China
| | - Qiong Wu
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China
| | - Zhaochen Luo
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - YaChun Zhang
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China
| | - Juncheng Ruan
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China
| | - Yingying Li
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China
| | - Ming Zhou
- Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
| | - ZhenFang Fu
- Department of Pathology, University of Georgia, Athens, GA, USA.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China.,College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China
| | - Ling Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, PR China.,Key Laboratory of Preventive Veterinary Medicine of Hubei Province, Huazhong Agricultural University, Wuhan, PR China.,State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, PR China
| |
Collapse
|
20
|
Overexpression of MAP2 and NF-H Associated with Dendritic Pathology in the Spinal Cord of Mice Infected with Rabies Virus. Viruses 2018; 10:v10030112. [PMID: 29509660 PMCID: PMC5869505 DOI: 10.3390/v10030112] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Revised: 01/29/2018] [Accepted: 02/08/2018] [Indexed: 12/13/2022] Open
Abstract
Rabies is a viral infection that targets the nervous system, specifically neurons. The clinical manifestations of the disease are dramatic and their outcome fatal; paradoxically, conventional histopathological descriptions reveal only subtle changes in the affected nervous tissue. Some researchers have considered that the pathophysiology of rabies is based more on biochemical changes than on structural alterations, as is the case with some psychiatric diseases. However, we believe that it has been necessary to resort to other methods that allow us to analyze the effect of the infection on neurons. The Golgi technique is the gold standard for studying the morphology of all the components of a neuron and the cytoskeletal proteins are the structural support of dendrites and axons. We have previously shown, in the mouse cerebral cortex and now with this work in spinal cord, that rabies virus generates remarkable alterations in the morphological pattern of the neurons and that this effect is associated with the increase in the expression of two cytoskeletal proteins (MAP2 and NF-H). It is necessary to deepen the investigation of the pathogenesis of rabies in order to find therapeutic alternatives to a disease to which the World Health Organization classifies as a neglected disease.
Collapse
|
21
|
Immunological aspects of rabies: a literature review. Arch Virol 2017; 162:3251-3268. [PMID: 28726129 DOI: 10.1007/s00705-017-3484-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 06/27/2017] [Indexed: 02/08/2023]
Abstract
Rabies is a lethal disease caused by the neurotropic virus rabies virus (RABV), and it remains an important public health problem globally. It is known that the host immune response is important for control of viral infection and promoting viral clearance. In this context, it is well documented that, in addition to RABV neutralizing antibody, interferons and cell-mediated immunity also have an important role in preventing the establishment of disease. On the other hand, RABV suppresses host immunity through different mechanisms, for example, direct inhibition of host gene expression, sequestration of pathogen-associated molecular patterns, or modification of cytokine signalling pathways, which hinder the protective host immune responses to RABV infection. Here, we review the immunological aspects of rabies, highlighting innate and adaptive immunity, as well as the host evasion immune mechanisms used by the virus. Finally, we briefly discuss how this knowledge can direct new research and be harnessed for future therapeutic strategies.
Collapse
|
22
|
Shim SM, Cheon HS, Jo C, Koh YH, Song J, Jeon JP. Elevated Epstein-Barr Virus Antibody Level is Associated with Cognitive Decline in the Korean Elderly. J Alzheimers Dis 2016; 55:293-301. [DOI: 10.3233/jad-160563] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
23
|
Scott TP, Nel LH. Subversion of the Immune Response by Rabies Virus. Viruses 2016; 8:v8080231. [PMID: 27548204 PMCID: PMC4997593 DOI: 10.3390/v8080231] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 08/11/2016] [Accepted: 08/12/2016] [Indexed: 12/24/2022] Open
Abstract
Rabies has affected mankind for several centuries and is one of the oldest known zoonoses. It is peculiar how little is known regarding the means by which rabies virus (RABV) evades the immune response and kills its host. This review investigates the complex interplay between RABV and the immune system, including the various means by which RABV evades, or advantageously utilizes, the host immune response in order to ensure successful replication and spread to another host. Different factors that influence immune responses—including age, sex, cerebral lateralization and temperature—are discussed, with specific reference to RABV and the effects on host morbidity and mortality. We also investigate the role of apoptosis and discuss whether it is a detrimental or beneficial mechanism of the host’s response to infection. The various RABV proteins and their roles in immune evasion are examined in depth with reference to important domains and the downstream effects of these interactions. Lastly, an overview of the means by which RABV evades important immune responses is provided. The research discussed in this review will be important in determining the roles of the immune response during RABV infections as well as to highlight important therapeutic target regions and potential strategies for rabies treatment.
Collapse
Affiliation(s)
- Terence P Scott
- Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0002, South Africa.
| | - Louis H Nel
- Department of Microbiology and Plant Pathology, University of Pretoria, Pretoria 0002, South Africa.
| |
Collapse
|
24
|
Shuangshoti S, Thorner PS, Teerapakpinyo C, Thepa N, Phukpattaranont P, Intarut N, Lumlertdacha B, Tepsumethanon V, Hemachudha T. Intracellular Spread of Rabies Virus Is Reduced in the Paralytic Form of Canine Rabies Compared to the Furious Form. PLoS Negl Trop Dis 2016; 10:e0004748. [PMID: 27253394 PMCID: PMC4890772 DOI: 10.1371/journal.pntd.0004748] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Accepted: 05/10/2016] [Indexed: 11/18/2022] Open
Abstract
Studies of the furious and paralytic forms of canine rabies at the early stage of disease have shown a more rapid viral colonization of the cerebral hemispheres in the furious form, as measured by viral antigen within neuronal cell bodies and viral RNA levels. Measurement of cellular processes separate from neuronal cell body provides a visual record of the spread of rabies virus which occurs across synapses. In this study, the amount of rabies viral antigen within cell processes was quantitatively assessed by image analysis in a cohort of naturally rabies infected non-vaccinated dogs (5 furious and 5 paralytic) that were sacrificed shortly after developing illness. Measurements were taken at different levels of the spinal cord, brain stem, and cerebrum. Results were compared to the amount of rabies viral antigen in neuronal cell bodies. Generally, the amount of rabies viral antigen in cell processes decreased in a rostral direction, following the pattern for the amount of rabies viral antigen in neuronal cell bodies and the percentage of involved cell bodies. However, there was a delay in cell process involvement following cell body involvement, consistent with replication occurring in the cell body region and subsequent transport out to cell processes. Greater amounts of antigen were seen in cell processes in dogs with the furious compared to paralytic form, at all anatomic levels examined. This difference was even evident when comparing (1) neurons with similar amounts of antigen, (2) similar percentages of involved neurons, and (3) anatomic levels that showed 100% positive neurons. These findings suggest that intracellular transport of the virus may be slower in the paralytic form, resulting in slower viral propagation. Possible mechanisms might involve host-specific differences in intracellular virus transport. The latter could be cytokine-mediated, since previous studies have documented greater inflammation in the paralytic form. Dogs with rabies can show the furious or paralytic form. The virus spreads from nerve cell to nerve cell via connections in cell processes. There are greater amounts of virus in the nerve cell bodies in the furious form. Studying cell processes separate from cell body provides a visual record of the spread of rabies virus. The amount of rabies viral protein within cell processes was measured in dogs with rabies (5 furious and 5 paralytic) sacrificed shortly after developing illness. The amount of viral protein in cell processes decreased from spinal cord to brain, as did the amount of viral protein in cell bodies and the percentage of involved cell bodies. However, there was a delay in cell process involvement following cell body involvement, consistent with the virus replicating in the cell body region and later moving out to cell processes. Greater amounts of viral protein were seen in cell processes in dogs with the furious compared to paralytic form, by comparing nerve cells with similar amounts of antigen, or similar percentages of involved nerve cells. These findings suggest that intracellular transport of the virus may be slower in paralytic rabies, resulting in slower viral spread in the brain.
Collapse
Affiliation(s)
- Shanop Shuangshoti
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Chulalongkorn GenePRO Center, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- WHO Collaborating Center for Research and Training on Viral Zoonoses, Bangkok, Thailand
- * E-mail:
| | - Paul Scott Thorner
- Department of Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Department of Laboratory Medicine, Hospital for Sick Children and University of Toronto, Toronto, Canada
| | - Chinachote Teerapakpinyo
- Chulalongkorn GenePRO Center, Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Nisachol Thepa
- Department of Clinical Pathology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Pornchai Phukpattaranont
- Department of Electrical Engineering, Faculty of Engineering, Prince of Songkla University, Songkhla, Thailand
| | - Nirun Intarut
- Epidemiology Unit, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | | | | | - Thiravat Hemachudha
- WHO Collaborating Center for Research and Training on Viral Zoonoses, Bangkok, Thailand
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
25
|
Li L, Wang H, Jin H, Cao Z, Feng N, Zhao Y, Zheng X, Wang J, Li Q, Zhao G, Yan F, Wang L, Wang T, Gao Y, Tu C, Yang S, Xia X. Interferon-inducible GTPase: a novel viral response protein involved in rabies virus infection. Arch Virol 2016; 161:1285-93. [PMID: 26906695 DOI: 10.1007/s00705-016-2795-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 02/14/2016] [Indexed: 11/25/2022]
Abstract
Rabies virus infection is a major public health concern because of its wide host-interference spectrum and nearly 100 % lethality. However, the interactions between host and virus remain unclear. To decipher the authentic response in the central nervous system after rabies virus infection, a dynamic analysis of brain proteome alteration was performed. In this study, 104 significantly differentially expressed proteins were identified, and intermediate filament, interferon-inducible GTPases, and leucine-rich repeat-containing protein 16C were the three outstanding groups among these proteins. Interferon-inducible GTPases were prominent because of their strong upregulation. Moreover, quantitative real-time PCR showed distinct upregulation of interferon-inducible GTPases at the level of transcription. Several studies have shown that interferon-inducible GTPases are involved in many biological processes, such as viral infection, endoplasmic reticulum stress response, and autophagy. These findings indicate that interferon-inducible GTPases are likely to be a potential target involved in rabies pathogenesis or the antiviral process.
Collapse
Affiliation(s)
- Ling Li
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Hualei Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225000, China.
| | - Hongli Jin
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China.,Changchun SR Biological Technology Co., Ltd., Changchun, 130012, China
| | - Zengguo Cao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Na Feng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225000, China
| | - Yongkun Zhao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225000, China
| | - Xuexing Zheng
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225000, China
| | - Jianzhong Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China.,Department of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, 453003, China
| | - Qian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Guoxing Zhao
- College of Veterinary Medicine, Jilin University, Changchun, 130062, China.,Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Lina Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Tiecheng Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Yuwei Gao
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China.,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225000, China
| | - Changchun Tu
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China
| | - Songtao Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225000, China.
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Institute of Military Veterinary, Academy of Military Medical Sciences, Changchun, 130122, China. .,Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Disease and Zoonoses, Yangzhou, 225000, China.
| |
Collapse
|
26
|
Davis BM, Rall GF, Schnell MJ. Everything You Always Wanted to Know About Rabies Virus (But Were Afraid to Ask). Annu Rev Virol 2015; 2:451-71. [PMID: 26958924 DOI: 10.1146/annurev-virology-100114-055157] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The cultural impact of rabies, the fatal neurological disease caused by infection with rabies virus, registers throughout recorded history. Although rabies has been the subject of large-scale public health interventions, chiefly through vaccination efforts, the disease continues to take the lives of about 40,000-70,000 people per year, roughly 40% of whom are children. Most of these deaths occur in resource-poor countries, where lack of infrastructure prevents timely reporting and postexposure prophylaxis and the ubiquity of domestic and wild animal hosts makes eradication unlikely. Moreover, although the disease is rarer than other human infections such as influenza, the prognosis following a bite from a rabid animal is poor: There is currently no effective treatment that will save the life of a symptomatic rabies patient. This review focuses on the major unanswered research questions related to rabies virus pathogenesis, especially those connecting the disease progression of rabies with the complex dysfunction caused by the virus in infected cells. The recent applications of cutting-edge research strategies to this question are described in detail.
Collapse
Affiliation(s)
| | - Glenn F Rall
- Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| | - Matthias J Schnell
- Department of Microbiology and Immunology and.,Jefferson Vaccine Center, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, 19107; .,Fox Chase Cancer Center, Philadelphia, Pennsylvania 19111
| |
Collapse
|
27
|
Diabolical effects of rabies encephalitis. J Neurovirol 2015; 22:8-13. [PMID: 25994917 DOI: 10.1007/s13365-015-0351-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 04/24/2015] [Accepted: 04/30/2015] [Indexed: 12/19/2022]
Abstract
Rabies is an acute encephalomyelitis in humans and animals caused by rabies virus (RABV) infection. Because the neuropathological changes are very mild in rabies, it has been assumed that neuronal dysfunction likely explains the severe clinical disease. Recently, degenerative changes have been observed in neuronal processes (dendrites and axons) in experimental rabies. In vitro studies have shown evidence of oxidative stress that is caused by mitochondrial dysfunction. Recent work has shown that the RABV phosphoprotein (P) interacts with mitochondrial Complex I leading to overproduction of reactive oxygen species, which results in injury to axons. Amino acids at positions 139 to 172 of the P are critical in this process. Rabies vectors frequently show behavioral changes. Aggressive behavior with biting is important for transmission of the virus to new hosts at a time when virus is secreted in the saliva. Aggression is associated with low serotonergic activity in the brain. Charlton and coworkers performed studies in experimentally infected striped skunks with skunk rabies virus and observed aggressive behavioral responses. Heavy accumulation of RABV antigen was found in the midbrain raphe nuclei, indicating that impaired serotonin neurotransmission from the brainstem may account for the aggressive behavior. We now have an improved understanding of how RABV causes neuronal injury and how the infection results in behavioral changes that promote viral transmission to new hosts.
Collapse
|
28
|
Abstract
Rabies is a highly lethal disease caused by the neurotropic rabies virus (RABV), and it remains an important public health problem globally. Effective vaccines have been developed for pre- and post-exposure prophylaxis (PEP). PEP is only effective if it is initiated promptly after recognizing exposure. Once neurological symptoms develop, however, it is widely accepted that there is no effective treatment available. Recent studies indicate that the presence of RABV-specific immunity (i.e. Virus neutralizing antibodies, VNA) and the transient enhancement of the BBB permeability are absolutely required for effective virus clearance from the CNS. In principle, it has been shown in mice using various live-attenuated RABVs or recombinant RABVs expressing three copies of the G or expressing chemokine/cytokines, which can induce high levels of VNA in the serum and also capable of transiently enhancing the BBB permeability that it is possible to clear the virus from CNS. Also, it has been demonstrated that, intravenous administration of VNA together with MCP-1 (shown to transiently open up BBB) can clear RABV from the CNS in both immunocompetent and immunocompromised mice, as late as 5 days after lethal challenge. Novel therapeutic approaches aimed at allowing the peripheral VNA to cross the BBB by administration of the VNA in combination with biological or chemical agents that can transiently open up the BBB would be useful to establish an effective therapy for rabies in humans. In this review, we focus on the some of the approaches that can be used to meet the challenges in the field of rabies treatment.
Collapse
Affiliation(s)
- C W Gnanadurai
- Department of Pathology, College of Veterinary Medicine, University of Georgia Athens, USA
| | - C T Huang
- Department of Pathology, College of Veterinary Medicine, University of Georgia Athens, USA
| | - D Kumar
- Department of Pathology, College of Veterinary Medicine, University of Georgia Athens, USA
| | - Zhen F Fu
- Department of Pathology, College of Veterinary Medicine, University of Georgia Athens, USA; State-key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, China
| |
Collapse
|
29
|
Mehta SM, Banerjee SM, Chowdhary AS. Postgenomics biomarkers for rabies—the next decade of proteomics. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2015; 19:67-79. [PMID: 25611201 DOI: 10.1089/omi.2014.0127] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rabies is one of the oldest diseases known to mankind. The pathogenic mechanisms by which rabies virus infection leads to development of neurological disease and death are still poorly understood. Analysis of rabies-infected proteomes may help identify novel biomarkers for antemortem diagnosis of the disease and target molecules for therapeutic intervention. This article offers a literature synthesis and critique of the differentially expressed proteins that have been previously reported from various in vitro/in vivo model systems and naturally infected clinical specimens. The emerging data collectively indicate that, in addition to the obvious alterations in proteins involved in synapse and neurotransmission, a majority of cytoskeletal proteins are relevant as well, providing evidence of neuronal degeneration. An interesting observation is that certain molecules, such as KPNA4, could be potential diagnostic markers for rabies. Importantly, proteomic studies with body fluids such as cerebrospinal fluid provide newer insights into antemortem diagnosis. In order to develop a complete integrative biology picture, it is essential to analyze the entire CNS (region-wise) and in particular, the brain. We suggest the use of laboratory animal models over cell culture systems using a combinatorial proteomics approach, as the former is a closer match to the actual host response. While most studies have focused on the terminal stages of the disease in mice, a time-series analysis could provide deeper insights for therapy. Postgenomics technologies such as proteomics warrant more extensive applications in rabies and similar diseases impacting public health around the world.
Collapse
Affiliation(s)
- Shraddha M Mehta
- Department of Virology and Immunology, Haffkine Institute for Training , Research and Testing, Parel, Mumbai, India
| | | | | |
Collapse
|
30
|
Chai Q, He WQ, Zhou M, Lu H, Fu ZF. Enhancement of blood-brain barrier permeability and reduction of tight junction protein expression are modulated by chemokines/cytokines induced by rabies virus infection. J Virol 2014; 88:4698-710. [PMID: 24522913 PMCID: PMC3993813 DOI: 10.1128/jvi.03149-13] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/05/2014] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Infection with laboratory-attenuated rabies virus (RABV) enhances blood-brain barrier (BBB) permeability, which has been demonstrated to be an important factor for host survival, since it allows immune effectors to enter the central nervous system (CNS) and clear RABV. To probe the mechanism by which RABV infection enhances BBB permeability, the expression of tight junction (TJ) proteins in the CNS was investigated following intracranial inoculation with laboratory-attenuated or wild-type (wt) RABV. BBB permeability was significantly enhanced in mice infected with laboratory-attenuated, but not wt, RABV. The expression levels of TJ proteins (claudin-5, occludin, and zonula occludens-1) were decreased in mice infected with laboratory-attenuated, but not wt, RABV, suggesting that enhancement of BBB permeability is associated with the reduction of TJ protein expression in RABV infection. RABV neither infects the brain microvascular endothelial cells (BMECs) nor modulates the expression of TJ proteins in BMECs. However, brain extracts prepared from mice infected with laboratory-attenuated, but not wt, RABV reduced TJ protein expression in BMECs. It was found that brain extracts from mice infected with laboratory-attenuated RABV contained significantly higher levels of inflammatory chemokines/cytokines than those from mice infected with wt RABV. Pathway analysis indicates that gamma interferon (IFN-γ) is located in the center of the cytokine network in the RABV-infected mouse brain, and neutralization of IFN-γ reduced both the disruption of BBB permeability in vivo and the downregulation of TJ protein expression in vitro. These findings indicate that the enhancement of BBB permeability and the reduction of TJ protein expression are due not to RABV infection per se but to virus-induced inflammatory chemokines/cytokines. IMPORTANCE Previous studies have shown that infection with only laboratory-attenuated, not wild-type, rabies virus (RABV) enhances blood-brain barrier (BBB) permeability, allowing immune effectors to enter the central nervous system (CNS) and clear RABV from the CNS. This study investigated the mechanism by which RABV infection enhances BBB permeability. It was found that RABV infection enhances BBB permeability by downregulation of tight junction (TJ) protein expression in the brain microvasculature. It was further found that it is not RABV infection per se but the chemokines/cytokines induced by RABV infection that downregulate the expression of TJ proteins and enhance BBB permeability. Blocking some of these cytokines, such as IFN-γ, ameliorated both the disruption of BBB permeability and the downregulation of TJ protein expression. These studies may provide a foundation for developing therapeutics for clinical rabies, such as medication that could be used to enhance BBB permeability.
Collapse
Affiliation(s)
- Qingqing Chai
- State-Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Wen Q. He
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Ming Zhou
- State-Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Huijun Lu
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| | - Zhen F. Fu
- State-Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, USA
| |
Collapse
|
31
|
Hirano M, Yoshii K, Sakai M, Hasebe R, Ichii O, Kariwa H. Tick-borne flaviviruses alter membrane structure and replicate in dendrites of primary mouse neuronal cultures. J Gen Virol 2014; 95:849-861. [PMID: 24394700 DOI: 10.1099/vir.0.061432-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Neurological diseases caused by encephalitic flaviviruses are severe and associated with high levels of mortality. However, detailed mechanisms of viral replication in the brain and features of viral pathogenesis remain poorly understood. We carried out a comparative analysis of replication of neurotropic flaviviruses: West Nile virus, Japanese encephalitis virus and tick-borne encephalitis virus (TBEV), in primary cultures of mouse brain neurons. All the flaviviruses multiplied well in primary neuronal cultures from the hippocampus, cerebral cortex and cerebellum. The distribution of viral-specific antigen in the neurons varied: TBEV infection induced accumulation of viral antigen in the neuronal dendrites to a greater extent than infection with other viruses. Viral structural proteins, non-structural proteins and dsRNA were detected in regions in which viral antigens accumulated in dendrites after TBEV replication. Replication of a TBEV replicon after infection with virus-like particles of TBEV also induced antigen accumulation, indicating that accumulated viral antigen was the result of viral RNA replication. Furthermore, electron microscopy confirmed that TBEV replication induced characteristic ultrastructural membrane alterations in the neurites: newly formed laminal membrane structures containing virion-like structures. This is the first report describing viral replication in and ultrastructural alterations of neuronal dendrites, which may cause neuronal dysfunction. These findings encourage further work aimed at understanding the molecular mechanisms of viral replication in the brain and the pathogenicity of neurotropic flaviviruses.
Collapse
Affiliation(s)
- Minato Hirano
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Kentaro Yoshii
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Mizuki Sakai
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Rie Hasebe
- Laboratory of Veterinary Hygiene, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Osamu Ichii
- Laboratory of Anatomy, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| | - Hiroaki Kariwa
- Laboratory of Public Health, Graduate School of Veterinary Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
32
|
|
33
|
Mitochondrial dysfunction in rabies virus infection of neurons. J Neurovirol 2013; 19:537-49. [PMID: 24277436 DOI: 10.1007/s13365-013-0214-6] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Revised: 09/20/2013] [Accepted: 10/01/2013] [Indexed: 12/21/2022]
Abstract
Infection with the challenge virus standard-11 (CVS) strain of fixed rabies virus induces neuronal process degeneration in adult mice after hindlimb footpad inoculation. CVS-induced axonal swellings of primary rodent dorsal root ganglion neurons are associated with 4-hydroxy-2-nonenal protein adduct staining, indicating a critical role of oxidative stress. Mitochondrial dysfunction is the major cause of oxidative stress. We hypothesized that CVS infection induces mitochondrial dysfunction leading to oxidative stress. We investigated the effects of CVS infection on several mitochondrial parameters in different cell types. CVS infection significantly increased maximal uncoupled respiration and complex IV respiration and complex I and complex IV activities, but did not affect complex II-III or citrate synthase activities. Increases in complex I activity, but not complex IV activity, correlated with susceptibility of the cells to CVS infection. CVS infection maintained coupled respiration and rate of proton leak, indicating a tight mitochondrial coupling. Possibly as a result of enhanced complex activity and efficient coupling, a high mitochondrial membrane potential was generated. CVS infection reduced the intracellular ATP level and altered the cellular redox state as indicated by a high NADH/NAD+ ratio. The basal production of reactive oxygen species (ROS) was not affected in CVS-infected neurons. However, a higher rate of ROS generation occurred in CVS-infected neurons in the presence of mitochondrial substrates and inhibitors. We conclude that CVS infection induces mitochondrial dysfunction leading to ROS overgeneration and oxidative stress.
Collapse
|
34
|
Gnanadurai CW, Zhou M, He W, Leyson CM, Huang CT, Salyards G, Harvey SB, Chen Z, He B, Yang Y, Hooper DC, Dietzchold B, Fu ZF. Presence of virus neutralizing antibodies in cerebral spinal fluid correlates with non-lethal rabies in dogs. PLoS Negl Trop Dis 2013; 7:e2375. [PMID: 24069466 PMCID: PMC3777866 DOI: 10.1371/journal.pntd.0002375] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Accepted: 07/01/2013] [Indexed: 12/03/2022] Open
Abstract
Background Rabies is traditionally considered a uniformly fatal disease after onset of clinical manifestations. However, increasing evidence indicates that non-lethal infection as well as recovery from flaccid paralysis and encephalitis occurs in laboratory animals as well as humans. Methodology/Principal Findings Non-lethal rabies infection in dogs experimentally infected with wild type dog rabies virus (RABV, wt DRV-Mexico) correlates with the presence of high level of virus neutralizing antibodies (VNA) in the cerebral spinal fluid (CSF) and mild immune cell accumulation in the central nervous system (CNS). By contrast, dogs that succumbed to rabies showed only little or no VNA in the serum or in the CSF and severe inflammation in the CNS. Dogs vaccinated with a rabies vaccine showed no clinical signs of rabies and survived challenge with a lethal dose of wild-type DRV. VNA was detected in the serum, but not in the CSF of immunized dogs. Thus the presence of VNA is critical for inhibiting virus spread within the CNS and eventually clearing the virus from the CNS. Conclusions/Significance Non-lethal infection with wt RABV correlates with the presence of VNA in the CNS. Therefore production of VNA within the CNS or invasion of VNA from the periphery into the CNS via compromised blood-brain barrier is important for clearing the virus infection from CNS, thereby preventing an otherwise lethal rabies virus infection. Inexorable lethality is still commonly attributed to rabies infection, although there is increasing evidence for non-lethal infection and even recovery from clinical rabies in various animal species and humans. This paper reports non-lethal infection in dogs. The striking difference between dogs that survived a wt RABV infection and dogs that succumbed to the infection is that the surviving dogs showed high level of VNA in the serum and in the CSF, as well as mild immune cell accumulation in the CNS, whereas dogs that succumbed to disease showed little or no VNA in the serum or in the CSF and developed severe CNS inflammation. Considering the role of VNA in clearing the virus from the CNS, production of VNA within the CNS or infiltration of VNA from the periphery into the CNS across the blood-brain barrier appears to be important for clearing the virus from CNS thereby preventing a lethal rabies infection.
Collapse
Affiliation(s)
- Clement W. Gnanadurai
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Ming Zhou
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Wenqi He
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Christina M. Leyson
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Chien-tsun Huang
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Gregory Salyards
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Stephen B. Harvey
- Department of Population Health, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Zhenhai Chen
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Biao He
- Department of Infectious Diseases, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
| | - Yang Yang
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
- State-key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
| | - D. C. Hooper
- Departments of Cancer Biology and Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Berhnard Dietzchold
- Departments of Cancer Biology and Neurological Surgery, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Zhen F. Fu
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia, United States of America
- State-key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- * E-mail:
| |
Collapse
|
35
|
Zhang G, Wang H, Mahmood F, Fu ZF. Rabies virus glycoprotein is an important determinant for the induction of innate immune responses and the pathogenic mechanisms. Vet Microbiol 2013; 162:601-613. [PMID: 23265241 PMCID: PMC3568536 DOI: 10.1016/j.vetmic.2012.11.031] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Revised: 11/20/2012] [Accepted: 11/22/2012] [Indexed: 12/24/2022]
Abstract
Our previous studies have suggested that street and fixed rabies viruses (RABVs) induce diseases in the mouse model via different mechanisms. In the present study, attempts were made to determine if it is the glycoprotein (G) that is responsible for the observed differences in the pathogenic mechanisms. To this end, an infectious clone from fixed virus B2c was established and used as a backbone for exchange of the G from street viruses. The rate of viral replication, expression of viral proteins, and the induction of innate immune responses were compared in cells or in mice infected with each of the viruses. Furthermore, the infiltration of inflammatory cells into the CNS and the enhancement of blood-brain barrier (BBB) permeability were also compared. It was found that fixed viruses induced stronger innate immune responses (expression of chemokines, infiltration of inflammatory cells, and enhancement of BBB permeability) than street RABV or recombinant viruses expressing the G from street RABVs. Fixed viruses induce disease via an immune-mediated pathogenic mechanism while street viruses or recombinant viruses expressing the G from street RABVs induce diseases via a mechanism other than immune-mediated pathogenesis. Therefore, RABV G is an important determinant for the induction of innate immune responses and consequently the pathogenic mechanisms.
Collapse
Affiliation(s)
- Guoqing Zhang
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Hualei Wang
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Fazal Mahmood
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | - Zhen F Fu
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA; State-Key Laboratory of Agricultural Microbiology, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, China.
| |
Collapse
|
36
|
Venugopal AK, Ghantasala SSK, Selvan LDN, Mahadevan A, Renuse S, Kumar P, Pawar H, Sahasrabhuddhe NA, Suja MS, Ramachandra YL, Prasad TSK, Madhusudhana SN, HC H, Chaerkady R, Satishchandra P, Pandey A, Shankar SK. Quantitative proteomics for identifying biomarkers for Rabies. Clin Proteomics 2013; 10:3. [PMID: 23521751 PMCID: PMC3660221 DOI: 10.1186/1559-0275-10-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 03/14/2013] [Indexed: 12/25/2022] Open
Abstract
INTRODUCTION Rabies is a fatal acute viral disease of the central nervous system, which is a serious public health problem in Asian and African countries. Based on the clinical presentation, rabies can be classified into encephalitic (furious) or paralytic (numb) rabies. Early diagnosis of this disease is particularly important as rabies is invariably fatal if adequate post exposure prophylaxis is not administered immediately following the bite. METHODS In this study, we carried out a quantitative proteomic analysis of the human brain tissue from cases of encephalitic and paralytic rabies along with normal human brain tissues using an 8-plex isobaric tags for relative and absolute quantification (iTRAQ) strategy. RESULTS AND CONCLUSION We identified 402 proteins, of which a number of proteins were differentially expressed between encephalitic and paralytic rabies, including several novel proteins. The differentially expressed molecules included karyopherin alpha 4 (KPNA4), which was overexpressed only in paralytic rabies, calcium calmodulin dependent kinase 2 alpha (CAMK2A), which was upregulated in paralytic rabies group and glutamate ammonia ligase (GLUL), which was overexpressed in paralytic as well as encephalitic rabies. We validated two of the upregulated molecules, GLUL and CAMK2A, by dot blot assays and further validated CAMK2A by immunohistochemistry. These molecules need to be further investigated in body fluids such as cerebrospinal fluid in a larger cohort of rabies cases to determine their potential use as antemortem diagnostic biomarkers in rabies. This is the first study to systematically profile clinical subtypes of human rabies using an iTRAQ quantitative proteomics approach.
Collapse
Affiliation(s)
- Abhilash K Venugopal
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Department of Biotechnology, Kuvempu University, Shimoga, 577451, India
| | - S Sameer Kumar Ghantasala
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Department of Biotechnology, Kuvempu University, Shimoga, 577451, India
| | - Lakshmi Dhevi N Selvan
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Anita Mahadevan
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore, 560029, India
| | - Santosh Renuse
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
| | - Praveen Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
| | - Harsh Pawar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Rajiv Gandhi University of Health Sciences, Bangalore, 560041, India
| | - Nandini A Sahasrabhuddhe
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Manipal University, Madhav Nagar, Manipal, Karnataka, 576104, India
| | - Mooriyath S Suja
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore, 560029, India
| | | | - Thottethodi S Keshava Prasad
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, 690525, India
- Manipal University, Madhav Nagar, Manipal, Karnataka, 576104, India
- Bioinformatics Centre, School of Life Sciences, Pondicherry University, Pondicherry, 605014, India
| | - Shampur N Madhusudhana
- Department of Neurovirology, National Institute of Mental Health and Neuro Sciences, Bangalore, 560029, India
| | - Harsha HC
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
| | - Raghothama Chaerkady
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India
| | | | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, 733 N. Broadway, BRB 527, Baltimore, MD, 21205, USA
- Department of Biological Chemistry, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Susarla K Shankar
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bangalore, 560029, India
| |
Collapse
|
37
|
|
38
|
Zandi F, Eslami N, Torkashvand F, Fayaz A, Khalaj V, Vaziri B. Expression changes of cytoskeletal associated proteins in proteomic profiling of neuroblastoma cells infected with different strains of rabies virus. J Med Virol 2012; 85:336-47. [PMID: 23168799 DOI: 10.1002/jmv.23458] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2012] [Indexed: 12/17/2022]
Abstract
Rabies virus invades the nervous system, induces neuronal dysfunction and causes death of the host. The disruption of the cytoskeletal integrity and synaptic structures of the neurons by rabies virus has been postulated as a possible basis for neuronal dysfunction. In the present study, a two-dimensional electrophoresis/mass spectrometry proteomics analysis of neuroblastoma cells revealed a significant effect of a virulent strain of rabies virus on the host cytoskeleton related proteins which was quite different from that of an attenuated strain. Vimentin, actin cytoplasmic 1 isoform, profilin I, and Rho-GDP dissociation inhibitor were host cell cytoskeletal related proteins changed by the virulent strain. The proteomics data indicated that the virulent strain of rabies virus induces significant expression changes in the vimentin and actin cytoskeleton networks of neurons which could be a strong clue for the relation of cytoskeletal integrity distraction and rabies virus pathogenesis. In addition, the expression alteration of other host proteins, particularly some structural and regulatory proteins may have potential roles in rabies virus pathogenesis.
Collapse
Affiliation(s)
- Fatemeh Zandi
- Protein Chemistry Unit, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | | | | | | | | | | |
Collapse
|
39
|
Song Y, Hou J, Qiao B, Li Y, Xu Y, Duan M, Guan Z, Zhang M, Sun L. Street rabies virus causes dendritic injury and F-actin depolymerization in the hippocampus. J Gen Virol 2012; 94:276-283. [PMID: 23114630 PMCID: PMC3709620 DOI: 10.1099/vir.0.047480-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Rabies is an acute viral infection of the central nervous system and is typically fatal in humans and animals; however, its pathogenesis remains poorly understood. In this study, the morphological changes of dendrites and dendritic spines in the CA1 region of the hippocampus were investigated in mice that were infected intracerebrally with an MRV strain of the street rabies virus. Haematoxylin and eosin and fluorescence staining analysis of brain sections from the infected mice showed very few morphological changes in the neuronal bodies and neuronal processes. However, we found a significant decrease in the number of dendritic spines. Primary neuronal cultures derived from the hippocampus of mice (embryonic day 16.5) that were infected with the virus also showed an obvious decrease in the number of dendritic spines. Furthermore, the decrease in the number of dendritic spines was related to the depolymerization of actin filaments (F-actin). We propose that the observed structural changes can partially explain the severe clinical disease that was found in experimental models of street rabies virus infections.
Collapse
Affiliation(s)
- Yan Song
- Nursing College, Beihua University, 3999 Huashan Road, Jilin 132013, PR China.,Key Laboratory of Zoonoses, Ministry of Education, Institute of Zoonoses, Jilin University, 5333 Xian Road, Changchun 130062, PR China.,Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun 130021, PR China
| | - Jinli Hou
- Key Laboratory of Zoonoses, Ministry of Education, Institute of Zoonoses, Jilin University, 5333 Xian Road, Changchun 130062, PR China.,Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun 130021, PR China
| | - Bin Qiao
- Key Laboratory of Zoonoses, Ministry of Education, Institute of Zoonoses, Jilin University, 5333 Xian Road, Changchun 130062, PR China.,Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun 130021, PR China
| | - Yanchao Li
- Department of Histology and Embryology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun, 130021, PR China
| | - Ye Xu
- Medical Research Laboratory, Jilin Medical College, Jilin Road, Jilin 132013, PR China
| | - Ming Duan
- Key Laboratory of Zoonoses, Ministry of Education, Institute of Zoonoses, Jilin University, 5333 Xian Road, Changchun 130062, PR China.,Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun 130021, PR China
| | - Zhenhong Guan
- Key Laboratory of Zoonoses, Ministry of Education, Institute of Zoonoses, Jilin University, 5333 Xian Road, Changchun 130062, PR China.,Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun 130021, PR China
| | - Maolin Zhang
- Key Laboratory of Zoonoses, Ministry of Education, Institute of Zoonoses, Jilin University, 5333 Xian Road, Changchun 130062, PR China.,Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun 130021, PR China
| | - Liankun Sun
- Key Laboratory of Zoonoses, Ministry of Education, Institute of Zoonoses, Jilin University, 5333 Xian Road, Changchun 130062, PR China.,Department of Pathophysiology, Norman Bethune College of Medicine, Jilin University, Xinming Road, Changchun 130021, PR China
| |
Collapse
|
40
|
Gomme EA, Wirblich C, Addya S, Rall GF, Schnell MJ. Immune clearance of attenuated rabies virus results in neuronal survival with altered gene expression. PLoS Pathog 2012; 8:e1002971. [PMID: 23071441 PMCID: PMC3469654 DOI: 10.1371/journal.ppat.1002971] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Accepted: 08/30/2012] [Indexed: 01/23/2023] Open
Abstract
Rabies virus (RABV) is a highly neurotropic pathogen that typically leads to mortality of infected animals and humans. The precise etiology of rabies neuropathogenesis is unknown, though it is hypothesized to be due either to neuronal death or dysfunction. Analysis of human brains post-mortem reveals surprisingly little tissue damage and neuropathology considering the dramatic clinical symptomology, supporting the neuronal dysfunction model. However, whether or not neurons survive infection and clearance and, provided they do, whether they are functionally restored to their pre-infection phenotype has not been determined in vivo for RABV, or any neurotropic virus. This is due, in part, to the absence of a permanent “mark” on once-infected cells that allow their identification long after viral clearance. Our approach to study the survival and integrity of RABV-infected neurons was to infect Cre reporter mice with recombinant RABV expressing Cre-recombinase (RABV-Cre) to switch neurons constitutively expressing tdTomato (red) to expression of a Cre-inducible EGFP (green), permanently marking neurons that had been infected in vivo. We used fluorescence microscopy and quantitative real-time PCR to measure the survival of neurons after viral clearance; we found that the vast majority of RABV-infected neurons survive both infection and immunological clearance. We were able to isolate these previously infected neurons by flow cytometry and assay their gene expression profiles compared to uninfected cells. We observed transcriptional changes in these “cured” neurons, predictive of decreased neurite growth and dysregulated microtubule dynamics. This suggests that viral clearance, though allowing for survival of neurons, may not restore them to their pre-infection functionality. Our data provide a proof-of-principle foundation to re-evaluate the etiology of human central nervous system diseases of unknown etiology: viruses may trigger permanent neuronal damage that can persist or progress in the absence of sustained viral antigen. Rabies is an ancient and fatal neurological disease of animals and humans, caused by infection of the central nervous system (CNS) with Rabies virus (RABV). It is estimated that nearly 55,000 human RABV fatalities occur each year, though this number is likely much higher due to unreported exposures or failure of diagnosis. No treatment has been identified to cure disease after onset of symptoms. Neurovirologists still do not know the cause of rabies' dramatic symptoms and fatality, though it is thought to be due to neuronal loss or dysfunction. Here, we use a novel approach to permanently and genetically tag infected cells so that they can be identified after the infection has been cleared. This allowed us to define neuronal survival time following infection, and to assess neuronal function through gene expression analysis. We found that RABV infection does not lead to loss of neurons, but rather induces a permanent change in gene expression that may be related to the ability of RABV to cause permanent CNS disease. Our study provides evidence that viral infection of the brain can initiate long-term changes that may have consequences for nervous system health, even after the virus has been cleared from the CNS.
Collapse
Affiliation(s)
- Emily A. Gomme
- Department of Microbiology and Immunology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Christoph Wirblich
- Department of Microbiology and Immunology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Sankar Addya
- Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
| | - Glenn F. Rall
- Fox Chase Cancer Center, Philadelphia, Pennsylvania, United States of America
| | - Matthias J. Schnell
- Department of Microbiology and Immunology, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Kimmel Cancer Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- Jefferson Vaccine Center, Jefferson Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
41
|
Koraka P, Martina BEE, Roose JM, van Thiel PPAM, van Amerongen G, Kuiken T, Osterhaus ADME. In vitro and in vivo isolation and characterization of Duvenhage virus. PLoS Pathog 2012; 8:e1002682. [PMID: 22654660 PMCID: PMC3359985 DOI: 10.1371/journal.ppat.1002682] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Accepted: 03/22/2012] [Indexed: 12/25/2022] Open
Abstract
A fatal human case of Duvenhage virus (DUVV) infection in a Dutch traveller who had returned from Kenya was reported in 2007. She exhibited classical symptoms of rabies encephalitis with distinct pathological findings. In the present study we describe the isolation and characterization of DUVV in vitro and its passage in BALB/c mice. The virus proved to be neuroinvasive in both juvenile and adult mice, resulting in about 50% lethality upon peripheral infection. Clinical signs in infected mice were those of classical rabies. However, the distribution of viral antigen expression in the brain differed from that of classical rabies virus infection and neither inclusion bodies nor neuronal necrosis were observed. This is the first study to describe the in vitro and in vivo isolation and characterization of DUVV. Lyssaviruses have been known for centuries to cause lethal encephalitis in animals and humans, representing a serious public health problem especially in developing countries. Little is known about the way that lyssaviruses in general, and Duvenhage virus in particular cause disease. Studies of pathogenesis have been hampered by the fact that the virus has not yet been propagated and characterized extensively. In this paper, we describe the characterization of Duvenhage virus in vitro. Further, we characterized the virus in BALB/c mice. We compared Duvenhage virus with a wild type rabies virus (silver-haired bat rabies virus) and we found that while in vitro the differences of these two viruses were not significant, the in vivo characteristics of these two viruses differed significantly. Histological analyses of infected mouse brains suggest that differences in virulence may be associated with difference in tropism. Elucidating the differences in pathogenesis between different lyssaviruses might help us in the design of novel treatment protocols.
Collapse
Affiliation(s)
- Penelope Koraka
- Department of Virology, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
42
|
Wang H, Zhang G, Wen Y, Yang S, Xia X, Fu ZF. Intracerebral administration of recombinant rabies virus expressing GM-CSF prevents the development of rabies after infection with street virus. PLoS One 2011; 6:e25414. [PMID: 21980450 PMCID: PMC3182207 DOI: 10.1371/journal.pone.0025414] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2011] [Accepted: 09/02/2011] [Indexed: 12/22/2022] Open
Abstract
Recently it was found that prior immunization with recombinant rabies virus (RABV) expressing granulocyte-macrophage colony-stimulating factor (GM-CSF) (LBNSE-GM-CSF) resulted in high innate/adaptive immune responses and protection against challenge with virulent RABV (Wen et al., JVI, 2011). In this study, the ability of LBNSE-GM-CSF to prevent animals from developing rabies was investigated in mice after infection with lethal doses of street RABV. It was found that intracerebral administration of LBNSE-GM-CSF protected more mice from developing rabies than sham-treated mice as late as day 5 after infection with street RABV. Intracerebral administration of LBNSE-GM-CSF resulted in significantly higher levels of chemokine/cytokine expression and more infiltration of inflammatory and immune cells into the central nervous system (CNS) than sham-administration or administration with UV-inactivated LBNSE-GM-CSF. Enhancement of blood-brain barrier (BBB) permeability and increases in virus neutralizing antibodies (VNA) were also observed in mice treated with LBNSE-GM-CSF. On the other hand, intracerebral administration with UV-inactivated LBNSE-GM-CSF did not increase protection despite the fact that VNA were induced in the periphery. However, intracerebral administration with chemoattractant protein-1 (MCP-1, also termed CCL2) increased significantly the protective efficacy of UV-inactivated LBNSE-GM-CSF. Together these studies confirm that direct administration of LBNSE-GM-CSF can enhance the innate and adaptive immunity as well as the BBB permeability, thus allowing infiltration of inflammatory cells and other immune effectors enter into the CNS to clear the virus and prevent the development of rabies.
Collapse
Affiliation(s)
- Hualei Wang
- Department of Pathology, University of Georgia, Athens, Georgia, United States of America
- Institute of Veterinary Medicine, Chinese Academy of Military Medical Sciences, Changchun, Jilin, China
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Guoqing Zhang
- Department of Pathology, University of Georgia, Athens, Georgia, United States of America
| | - Yongjun Wen
- Department of Pathology, University of Georgia, Athens, Georgia, United States of America
| | - Songtao Yang
- Institute of Veterinary Medicine, Chinese Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Xianzhu Xia
- Institute of Veterinary Medicine, Chinese Academy of Military Medical Sciences, Changchun, Jilin, China
| | - Zhen F. Fu
- Department of Pathology, University of Georgia, Athens, Georgia, United States of America
- Department of Infectious Diseases, University of Georgia, Athens, Georgia, United States of America
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- * E-mail:
| |
Collapse
|
43
|
Abstract
Human rabies is almost invariably fatal, and globally it remains an important public health problem. Our knowledge of rabies pathogenesis has been learned mainly from studies performed in experimental animal models, and a number of unresolved issues remain. In contrast with the neural pathway of spread, there is still no credible evidence that hematogenous spread of rabies virus to the central nervous system plays a significant role in rabies pathogenesis. Although neuronal dysfunction has been thought to explain the neurological disease in rabies, recent evidence indicates that structural changes involving neuronal processes may explain the severe clinical disease and fatal outcome. Endemic dog rabies results in an ongoing risk to humans in many resource-limited and resource-poor countries, whereas rabies in wildlife is important in North America and Europe. In human cases in North America, transmission from bats is most common, but there is usually no history of a bat bite and there may be no history of contact with bats. Physicians may not recognize typical features of rabies in North America and Europe. Laboratory diagnostic evaluation for rabies includes rabies serology plus skin biopsy, cerebrospinal fluid, and saliva specimens for rabies virus antigen and/or RNA detection. Methods of postexposure rabies prophylaxis, including wound cleansing and administration of rabies vaccine and human rabies immune globulin, are highly effective after recognized exposure. Although there have been rare survivors of human rabies, no effective therapy is presently available. Therapeutic coma (midazolam and phenobarbital), ketamine, and antiviral therapies (known as the "Milwaukee protocol") were given to a rabies survivor, but this therapy was likely not directly responsible for the favorable outcome. New therapeutic approaches for human rabies need to be developed. A better understanding of basic mechanisms involved in rabies pathogenesis may be helpful in the development of potential new therapies for the future.
Collapse
Affiliation(s)
- Alan C Jackson
- Departments of Internal Medicine (Neurology) and Medical Microbiology, University of Manitoba, Winnipeg, MB, Canada,
| |
Collapse
|
44
|
Role of oxidative stress in rabies virus infection of adult mouse dorsal root ganglion neurons. J Virol 2010; 84:4697-705. [PMID: 20181692 DOI: 10.1128/jvi.02654-09] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Rabies virus infection of dorsal root ganglia (DRG) was studied in vitro with cultured adult mouse DRG neurons. Recent in vivo studies of transgenic mice that express the yellow fluorescent protein indicate that neuronal process degeneration, involving both dendrites and axons, occurs in mice infected with the challenge virus standard (CVS) strain of rabies virus by footpad inoculation. Because of the similarities of the morphological changes in experimental rabies and in diabetic neuropathy and other diseases, we hypothesize that neuronal process degeneration occurs as a result of oxidative stress. DRG neurons were cultured from adult ICR mice. Two days after plating, they were infected with CVS. Immunostaining was evaluated with CVS- and mock-infected cultures for neuron specific beta-tubulin, rabies virus antigen, and amino acid adducts of 4-hydroxy-2-nonenal (4-HNE) (marker of lipid peroxidation and hence oxidative stress). Neuronal viability (by trypan blue exclusion), terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining, and axonal growth were also assessed with the cultures. CVS infected 33 to 54% of cultured DRG neurons. Levels of neuronal viability and TUNEL staining were similar in CVS- and mock-infected DRG neurons. There were significantly more 4-HNE-labeled puncta at 2 and 3 days postinfection in CVS-infected cultures than in mock-infected cultures, and axonal outgrowth was reduced at these time points in CVS infection. Axonal swellings with 4-HNE-labeled puncta were also associated with aggregations of actively respiring mitochondria. We have found evidence that rabies virus infection in vitro causes axonal injury of DRG neurons through oxidative stress. Oxidative stress may be important in vivo in rabies and may explain previous observations of the degeneration of neuronal processes.
Collapse
|
45
|
Abstract
Rabies is a top-ten infectious killer and is newly re-emergent worldwide. Focusing on the unique aspects of the pathophysiology and immunology of rabies, we improvised a strategy that led to the first survivor of rabies without rabies prophylaxis. Our data support the long-standing speculation that rabies is a disorder of neurotransmission. Improvements in human survival have better delineated the immune and metabolic responses to rabies. We anticipate a new generation of rabies biologicals in the very near future, as well as the more remote possibility of rescue monoclonal antibodies engineered from several recent survivors of human rabies. The general approach to rabies treatment serves as a model for a more complex, physiological approach to treating infectious neurological disorders.
Collapse
Affiliation(s)
- Rodney E Willoughby
- Pediatric Infectious Diseases, Medical College of Wisconsin, PO Box 1997, Milwaukee, WI 53201-1997, USA
| |
Collapse
|
46
|
Kuang Y, Lackay SN, Zhao L, Fu ZF. Role of chemokines in the enhancement of BBB permeability and inflammatory infiltration after rabies virus infection. Virus Res 2009; 144:18-26. [PMID: 19720239 PMCID: PMC2760941 DOI: 10.1016/j.virusres.2009.03.014] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2009] [Revised: 03/23/2009] [Accepted: 03/25/2009] [Indexed: 02/08/2023]
Abstract
Induction of innate immunity, particularly through the induction of interferon and chemokines, by rabies virus (RABV) infection has been reported to be inversely correlated with pathogenicity. To further investigate the association between the expression of chemokines and RABV infection, laboratory-attenuated RABV (B2C) and wild-type (wt) RABV (DRV) were administered to Balb/c mice intramuscularly. Chemokine expression, inflammatory cell infiltration, and blood-brain barrier (BBB) permeability were evaluated at various time points after infection. At day 3 post-infection (p.i.) there was very little inflammation in the central nervous system (CNS) and BBB permeability did not change in mice infected with either virus when compared with mock-infected mice. At 6 day p.i., infection with B2C induced the expression of inflammatory chemokines and infiltration of inflammatory cells into the CNS, while these changes were minimal in DRV-infected mice. Furthermore, infection with B2C significantly enhanced BBB permeability comparing to infection with DRV. Among the upregulated chemokines, the expression of IP-10 was best correlated with infiltration of inflammatory cells into the CNS and enhancement of BBB permeability. These data indicate that laboratory-attenuated RABV induces expression of chemokines and infiltration of inflammatory cells into the CNS. Upregulation of chemokines by B2C may have triggered the change in BBB permeability, which helps infiltration of inflammatory cells into the CNS, and thus attenuation of RABV.
Collapse
Affiliation(s)
- Yi Kuang
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, GA 30602, USA
| | | | | | | |
Collapse
|
47
|
Gholami A, Kassis R, Real E, Delmas O, Guadagnini S, Larrous F, Obach D, Prevost MC, Jacob Y, Bourhy H. Mitochondrial dysfunction in lyssavirus-induced apoptosis. J Virol 2008; 82:4774-84. [PMID: 18321977 PMCID: PMC2346764 DOI: 10.1128/jvi.02651-07] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2007] [Accepted: 02/22/2008] [Indexed: 12/25/2022] Open
Abstract
Lyssaviruses are highly neurotropic viruses associated with neuronal apoptosis. Previous observations have indicated that the matrix proteins (M) of some lyssaviruses induce strong neuronal apoptosis. However, the molecular mechanism(s) involved in this phenomenon is still unknown. We show that for Mokola virus (MOK), a lyssavirus of low pathogenicity, the M (M-MOK) targets mitochondria, disrupts the mitochondrial morphology, and induces apoptosis. Our analysis of truncated M-MOK mutants suggests that the information required for efficient mitochondrial targeting and dysfunction, as well as caspase-9 activation and apoptosis, is held between residues 46 and 110 of M-MOK. We used a yeast two-hybrid approach, a coimmunoprecipitation assay, and confocal microscopy to demonstrate that M-MOK physically associates with the subunit I of the cytochrome c (cyt-c) oxidase (CcO) of the mitochondrial respiratory chain; this is in contrast to the M of the highly pathogenic Thailand lyssavirus (M-THA). M-MOK expression induces a significant decrease in CcO activity, which is not the case with M-THA. M-MOK mutations (K77R and N81E) resulting in a similar sequence to M-THA at positions 77 and 81 annul cyt-c release and apoptosis and restore CcO activity. As expected, the reverse mutations, R77K and E81N, introduced in M-THA induce a phenotype similar to that due to M-MOK. These features indicate a novel mechanism for energy depletion during lyssavirus-induced apoptosis.
Collapse
Affiliation(s)
- Alireza Gholami
- Unité Postulante de Recherche et d'Expertise Dynamique des Lyssavirus et Adaptation à l'Hôte, Institut Pasteur, 25 rue du Docteur Roux, 75724 Paris Cedex 15, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Rabies vaccines. Vaccines (Basel) 2008. [DOI: 10.1016/b978-1-4160-3611-1.50031-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023] Open
|
49
|
Abstract
Various technological developments have revitalized the approaches employed to study the disease of rabies. In particular, reverse genetics has facilitated the generation of novel viruses used to improve our understanding of the fundamental aspects of rabies virus (RABV) biology and pathogenicity and yielded novel constructs potentially useful as vaccines against rabies and other diseases. Other techniques such as high throughput methods to examine the impact of rabies virus infection on host cell gene expression and two hybrid systems to explore detailed protein-protein interactions also contribute substantially to our understanding of virus-host interactions. This review summarizes much of the increased knowledge about rabies that has resulted from such studies but acknowledges that this is still insufficient to allow rational attempts at curing those who present with clinical disease.
Collapse
Affiliation(s)
- Susan A Nadin-Davis
- Centre of Expertise for Rabies, Ottawa Laboratory (Fallowfield), Canadian Food Inspection Agency, Ottawa, ON, Canada
| | | |
Collapse
|
50
|
Structural abnormalities in neurons are sufficient to explain the clinical disease and fatal outcome of experimental rabies in yellow fluorescent protein-expressing transgenic mice. J Virol 2007; 82:513-21. [PMID: 17942540 DOI: 10.1128/jvi.01677-07] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Under natural conditions and in some experimental models, rabies virus infection of the central nervous system causes relatively mild histopathological changes, without prominent evidence of neuronal death despite its lethality. In this study, the effects of rabies virus infection on the structure of neurons were investigated with experimentally infected transgenic mice expressing yellow fluorescent protein (YFP) in neuronal subpopulations. Six-week-old mice were inoculated in the hind-limb footpad with the CVS strain of fixed virus or were mock infected with vehicle (phosphate-buffered saline). Brain regions were subsequently examined by light, epifluorescent, and electron microscopy. In moribund CVS-infected mice, histopathological changes were minimal in paraffin-embedded tissue sections, although mild inflammatory changes were present. Terminal deoxynucleotidyltransferase-mediated dUTP-biotin nick end labeling and caspase-3 immunostaining showed only a few apoptotic cells in the cerebral cortex and hippocampus. Silver staining demonstrated the preservation of cytoskeletal integrity in the cerebral cortex. However, fluorescence microscopy revealed marked beading and fragmentation of the dendrites and axons of layer V pyramidal neurons in the cerebral cortex, cerebellar mossy fibers, and axons in brainstem tracts. At an earlier time point, when mice displayed hind-limb paralysis, beading was observed in a few axons in the cerebellar commissure. Toluidine blue-stained resin-embedded sections from moribund YFP-expressing animals revealed vacuoles within the perikarya and proximal dendrites of pyramidal neurons in the cerebral cortex and hippocampus. These vacuoles corresponded with swollen mitochondria under electron microscopy. Vacuolation was also observed ultrastructurally in axons and in presynaptic nerve endings. We conclude that the observed structural changes are sufficient to explain the severe clinical disease with a fatal outcome in this experimental model of rabies.
Collapse
|