1
|
Castellan M, Zamperin G, Foiani G, Zorzan M, Priore MF, Drzewnioková P, Melchiotti E, Vascellari M, Monne I, Crovella S, Leopardi S, De Benedictis P. Immunological findings of West Caucasian bat virus in an accidental host. J Virol 2025:e0191424. [PMID: 39846740 DOI: 10.1128/jvi.01914-24] [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: 11/15/2024] [Accepted: 12/09/2024] [Indexed: 01/24/2025] Open
Abstract
The Lyssavirus genus includes seventeen viral species able to cause rabies, an acute and almost invariably fatal encephalomyelitis of mammals. Rabies virus (RABV), which represents the type species of the genus, is a multi-host pathogen that over the years has undergone multiple events of host-switching, thus occupying several geographical and ecological niches. In contrast, non-RABV lyssaviruses are mainly confined within a single natural host with rare spillover events. In this scenario, unveiling the mechanisms underlying the host immune response against a virus is crucial to understand the dynamics of infection and to predict the probability of colonization/adaptation to a new target species. Presently, the host response to lyssaviruses has only been partially explored, with the majority of data extrapolated from RABV infection. West Caucasian bat virus (WCBV), a divergent lyssavirus, has recently been associated with a spillover event to a domestic cat, raising concern about the risks to public health due to the circulation of the virus in its natural host. Through this study we have investigated the immune response determined by the WCBV versus two widely known lyssaviruses. We selected the Syrian hamster as representative of an accidental host, and chose the intramuscular route in order to mimic the natural infection. In hamsters, WCBV was highly pathogenic, determining 100% lethality and mild encephalitis. In comparison with Duvenhage virus (DUVV) and RABV, we found that WCBV displayed an intermediate ability to promote cellular antiviral response, produce pro-inflammatory cytokines, and recruit and activate lymphocytes in the hamsters' central nervous system. IMPORTANCE Although all lyssaviruses cause fatal encephalomyelitis in mammals, they display a different host tropism and pathogenicity, with the ecology of Rabies virus (RABV) continually evolving and adapting to new host species. In 2020, West Caucasian bat virus (WCBV) was identified as the causative agent of rabies in a domestic cat in Italy. This event raised concerns about its public health consequences, due to the absence of biologicals against the infection. Our study investigates the host immune response triggered by WCBV in comparison with a pathogenic strain of RABV and the low pathogenic Duvenhage lyssavirus (DUVV), as a proxy to understand the mechanisms leading to lyssavirus spillover and pathogenicity. We overall confirm that previous evidence indicating an inverse relationship between lyssavirus pathogenicity and immune response is applicable for WCBV as well. Importantly, this work represents the first transcriptomic analysis of the WCBV interaction in the central nervous system with an accidental host.
Collapse
Affiliation(s)
- Martina Castellan
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Gianpiero Zamperin
- Viral Genomics and Transcriptomics Laboratory, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Greta Foiani
- Laboratory of Histopathology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Maira Zorzan
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Maria Francesca Priore
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Petra Drzewnioková
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Erica Melchiotti
- Laboratory of Histopathology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Marta Vascellari
- Laboratory of Histopathology, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Isabella Monne
- Viral Genomics and Transcriptomics Laboratory, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Sergio Crovella
- Laboratory of Animal Research Center (LARC), Qatar University, Doha, Qatar
| | - Stefania Leopardi
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| | - Paola De Benedictis
- Laboratory for Emerging Viral Zoonoses, WOAH Reference Laboratory for Rabies, FAO and National Reference Centre for Rabies, Department for Research and Innovation, Istituto Zooprofilattico Sperimentale delle Venezie, Legnaro, Italy
| |
Collapse
|
2
|
Guo X, Zhang M, Feng Y, Liu X, Wang C, Zhang Y, Wang Z, Zhang D, Guo Y. Transcriptome analysis of salivary glands of rabies-virus-infected mice. Front Microbiol 2024; 15:1354936. [PMID: 38380102 PMCID: PMC10877373 DOI: 10.3389/fmicb.2024.1354936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Rabies is a fatal zoonotic disease that poses a threat to public health. Rabies virus (RABV) is excreted in the saliva of infected animals, and is primarily transmitted by bite. The role of the salivary glands in virus propagation is significant, but has been less studied in the pathogenic mechanisms of RABV. To identify functionally important genes in the salivary glands, we used RNA sequencing (RNA-seq) to establish and analyze mRNA expression profiles in parotid tissue infected with two RABV strains, CVS-11 and PB4. The biological functions of differentially expressed genes (DEGs) were determined by Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis, which revealed 3,764 DEGs (678 up-regulated and 3,086 down-regulated) in the CVS-11 infected group and 4,557 DEGs (874 up-regulated and 3,683 down-regulated) in the PB4 infected group. Various biological processes are involved, including the salivary secretion pathway and the phosphatidylinositol 3-kinase-Akt (PI3K-Akt) signaling pathway. This study provides the first mapping of the transcriptome changes in response to RABV infection in parotid tissue, offering new insights into the study of RABV-affected salivary gland function and RABV pathogenic mechanisms in parotid tissue. The salivary gland-enriched transcripts may be potential targets of interest for rabies disease control.
Collapse
Affiliation(s)
- Xin Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Maolin Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ye Feng
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, China
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, Beijing, China
| | - Xiaomin Liu
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Chongyang Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yannan Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zichen Wang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Danwei Zhang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yidi Guo
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
3
|
Wang S, Ding X, Li Z, Rao F, Xu H, Lu J, Ma X, Zhang M, Xie Z. Comprehensive analyses identify potential biomarkers for encephalitis in HIV infection. Sci Rep 2023; 13:18418. [PMID: 37891420 PMCID: PMC10611703 DOI: 10.1038/s41598-023-45922-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Accepted: 10/25/2023] [Indexed: 10/29/2023] Open
Abstract
Human immunodeficiency virus encephalitis (HIVE) is a severe neurological complication after HIV infection. Evidence shows that genetic factors play an important role in HIVE. The aim of the present study was to identify new potential therapeutic targets for HIVE. Differentially expressed gene (DEG), functional annotation and pathway, and protein-protein interaction analyses were performed to identify the hub genes associated with HIVE. Gene co-expression analysis was carried out to confirm the association between the hub genes and HIVE. Finally, the role of the hub genes in HIVE therapy was evaluated by conducting drug-gene interaction analysis. A total of 20 overlapping DEGs closely related to HIVE were identified. Functional annotation and pathway enrichment analysis indicated that the markedly enriched DEG terms included ion transport, type II interferon signaling, and synaptic signaling. Moreover, protein-protein interaction analysis revealed that 10 key HIVE-related genes were hub genes, including SCN8A, CDK5R2, GRM5, SCN2B, IFI44L, STAT1, SLC17A7, ISG15, FGF12, and FGF13. Furthermore, six hub genes were co-expressed with HIVE-associated host genes in human brain tissue. Finally, three hub genes (STAT1, ISG15, and SCN2B) interacted with several inflammation-associated drugs. These findings suggested that SCN8A, CDK5R2, GRM5, SCN2B, IFI44L, STAT1, SLC17A7, ISG15, FGF12, and FGF13 may be new targets for diagnosis and therapy of HIVE.
Collapse
Affiliation(s)
- Shitao Wang
- Department of Neurology, Affiliated Fuyang People's Hospital of Anhui Medical University, Fuyang, 236000, China.
| | - Xiangqian Ding
- Department of Neurosurgery, Qilu Hospital of Shandong University, Jinan, 250012, China
| | - Zongyou Li
- Department of Neurology, Affiliated Fuyang People's Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Feng Rao
- Department of Neurology, Affiliated Fuyang People's Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Hui Xu
- Department of Neurology, Affiliated Fuyang People's Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Jinghong Lu
- Department of Neurology, Affiliated Fuyang People's Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Xuelu Ma
- Department of Neurology, Affiliated Fuyang People's Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Mengen Zhang
- Department of Neurology, Affiliated Fuyang People's Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Zhenrong Xie
- The Medical Biobank, First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
| |
Collapse
|
4
|
Muangsanit P, Chailangkarn T, Tanwattana N, Wongwanakul R, Lekcharoensuk P, Kaewborisuth C. Hydrogel-based 3D human iPSC-derived neuronal culture for the study of rabies virus infection. Front Cell Infect Microbiol 2023; 13:1215205. [PMID: 37692167 PMCID: PMC10485840 DOI: 10.3389/fcimb.2023.1215205] [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: 05/01/2023] [Accepted: 08/08/2023] [Indexed: 09/12/2023] Open
Abstract
Background Rabies is a highly fatal infectious disease that poses a significant threat to human health in developing countries. In vitro study-based understanding of pathogenesis and tropism of different strains of rabies virus (RABV) in the central nervous system (CNS) is limited due to the lack of suitable culture models that recapitulate the complex communication pathways among host cells, extracellular matrices, and viruses. Therefore, a three-dimensional (3D) cell culture that mimics cell-matrix interactions, resembling in vivo microenvironment, is necessary to discover relevant underlying mechanisms of RABV infection and host responses. Methods The 3D collagen-Matrigel hydrogel encapsulating hiPSC-derived neurons for RABV infection was developed and characterized based on cell viability, morphology, and gene expression analysis of neuronal markers. The replication kinetics of two different strains of RABV [wild-type Thai (TH) and Challenge Virus Standard (CVS)-11 strains] in both 2D and 3D neuronal cultures were examined. Differential gene expression analysis (DEG) of the neuropathological pathway of RABV-infected 2D and 3D models was also investigated via NanoString analysis. Results The 3D hiPSC-derived neurons revealed a more physiologically interconnected neuronal network as well as more robust and prolonged maturation and differentiation than the conventional 2D monolayer model. TH and CVS-11 exhibited distinct growth kinetics in 3D neuronal model. Additionally, gene expression analysis of the neuropathological pathway observed during RABV infection demonstrated a vast number of differentially expressed genes (DEGs) in 3D model. Unlike 2D neuronal model, 3D model displayed more pronounced cellular responses upon infection with CVS-11 when compared to the TH-infected group, highlighting the influence of the cell environment on RABV-host interactions. Gene ontology (GO) enrichment of DEGs in the infected 3D neuronal culture showed alterations of genes associated with the inflammatory response, apoptotic signaling pathway, glutamatergic synapse, and trans-synaptic signaling which did not significantly change in 2D culture. Conclusion We demonstrated the use of a hydrogel-based 3D hiPSC-derived neuronal model, a highly promising technology, to study RABV infection in a more physiological environment, which will broaden our understanding of RABV-host interactions in the CNS.
Collapse
Affiliation(s)
- Papon Muangsanit
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Thanathom Chailangkarn
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Nathiphat Tanwattana
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
| | - Ratjika Wongwanakul
- National Nanotechnology Center (NANOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
| | - Porntippa Lekcharoensuk
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
- Department of Microbiology and Immunology, Faculty of Veterinary Medicine, Kasetsart University, Bangkok, Thailand
- Center for Advance Studies in Agriculture and Food, KU Institute Studies, Kasetsart University, Bangkok, Thailand
| | - Challika Kaewborisuth
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), Pathumthani, Thailand
- Interdisciplinary Program in Genetic Engineering and Bioinformatics, Graduate School, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
5
|
Embregts CW, Wentzel AS, den Dekker AT, van IJcken WF, Stadhouders R, GeurtsvanKessel CH. Rabies virus uniquely reprograms the transcriptome of human monocyte-derived macrophages. Front Cell Infect Microbiol 2023; 13:1013842. [PMID: 36798087 PMCID: PMC9927221 DOI: 10.3389/fcimb.2023.1013842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 01/18/2023] [Indexed: 02/03/2023] Open
Abstract
Macrophages are amongst the first immune cells that encounter rabies virus (RABV) at virus entry sites. Activation of macrophages is essential for the onset of a potent immune response, but insights into the effects of RABV on macrophage activation are scarce. In this study we performed high-throughput sequencing on RNA extracted from macrophages that were exposed to RABV for 48 hours, and compared their transcriptional profiles to that of non-polarized macrophages (M0), and macrophages polarized towards the canonical M1, M2a and M2c phenotypes. Our analysis revealed that RABV-stimulated macrophages show high expression of several M1, M2a and M2c signature genes. Apart from their partial resemblance to these phenotypes, unbiased clustering analysis revealed that RABV induces a unique and distinct polarization program. Closer examination revealed that RABV induced multiple pathways related to the interferon- and antiviral response, which were not induced under other classical polarization strategies. Surprisingly, our data show that RABV induces an activated rather than a fully suppressed macrophage phenotype, triggering virus-induced activation and polarization. This includes multiple genes with known antiviral (e.g. APOBEC3A, IFIT/OAS/TRIM genes), which may play a role in anti-RABV immunity.
Collapse
Affiliation(s)
- Carmen W.E. Embregts
- Department of Viroscience, Erasmus Medical Center, Rotterdam, Netherlands,*Correspondence: Carmen W.E. Embregts,
| | - Annelieke S. Wentzel
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, Netherlands
| | | | | | - Ralph Stadhouders
- Department of Pulmonary Medicine, Erasmus Medical Center, Rotterdam, Netherlands,Department of Cell Biology, Erasmus Medical Center, Rotterdam, Netherlands
| | | |
Collapse
|
6
|
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
|
7
|
Zhang J, Li Z, Lu H, Shi J, Gao R, Ma Y, Lan Y, Guan J, Zhao K, Gao F, He W. Evidence of Microglial Immune Response Following Coronavirus PHEV Infection of CNS. Front Immunol 2022; 12:804625. [PMID: 35082791 PMCID: PMC8784595 DOI: 10.3389/fimmu.2021.804625] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/20/2021] [Indexed: 12/14/2022] Open
Abstract
Porcine hemagglutinating encephalomyelitis virus (PHEV) is a highly neurotropic coronavirus that invades the host central nervous system (CNS) and causes neurological dysfunction. Microglia are key immune cells in the CNS, however, whether and how they response to PHEV infection remains unclear. Herein, microglial activation and proliferation were detected in the CNS of PHEV-infected mice, as along with the proinflammatory response. Moreover, the production of proinflammatory cytokines induced by moderately activated microglia limited viral replication in the early stage of infection. Microglial depletion assays showed that during late infection, excess activation of microglia aggravated neurological symptoms, BBB destruction, and peripheral monocyte/macrophage infiltration into the CNS. Using an in vitro brain slice model, PHEV was identified to specifically and moderately induce microglial activation in the absence of peripheral immune cells infiltration. Consistently, macrophage clearance from circulating blood indicated that peripheral monocytes/macrophages crossing the BBB of mice were responsible for excess activation of microglia and CNS damage in late PHEV infection. Overall, our findings provide evidence supporting a dual role for microglia in the host CNS in response to coronavirus PHEV invasion.
Collapse
Affiliation(s)
- Jing Zhang
- 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
| | - Huijun Lu
- Key Laboratory of Zoonosis Research, Ministry of Education, Institute of Zoonosis, Jilin University, Changchun, China
| | - Junchao Shi
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Rui Gao
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Ying Ma
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Yungang Lan
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jiyu Guan
- 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
| | - 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
|
8
|
Kim S, Larrous F, Varet H, Legendre R, Feige L, Dumas G, Matsas R, Kouroupi G, Grailhe R, Bourhy H. Early Transcriptional Changes in Rabies Virus-Infected Neurons and Their Impact on Neuronal Functions. Front Microbiol 2021; 12:730892. [PMID: 34970230 PMCID: PMC8713068 DOI: 10.3389/fmicb.2021.730892] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/25/2021] [Indexed: 12/24/2022] Open
Abstract
Rabies is a zoonotic disease caused by rabies virus (RABV). As rabies advances, patients develop a variety of severe neurological symptoms that inevitably lead to coma and death. Unlike other neurotropic viruses that can induce symptoms of a similar range, RABV-infected post-mortem brains do not show significant signs of inflammation nor the structural damages on neurons. This suggests that the observed neurological symptoms possibly originate from dysfunctions of neurons. However, many aspects of neuronal dysfunctions in the context of RABV infection are only partially understood, and therefore require further investigation. In this study, we used differentiated neurons to characterize the RABV-induced transcriptomic changes at the early time-points of infection. We found that the genes modulated in response to the infection are particularly involved in cell cycle, gene expression, immune response, and neuronal function-associated processes. Comparing a wild-type RABV to a mutant virus harboring altered matrix proteins, we found that the RABV matrix protein plays an important role in the early down-regulation of host genes, of which a significant number is involved in neuronal functions. The kinetics of differentially expressed genes (DEGs) are also different between the wild type and mutant virus datasets. The number of modulated genes remained constant upon wild-type RABV infection up to 24 h post-infection, but dramatically increased in the mutant condition. This result suggests that the intact viral matrix protein is important to control the size of host gene modulation. We then examined the signaling pathways previously studied in relation to the innate immune responses against RABV, and found that these pathways contribute to the changes in neuronal function-associated processes. We further examined a set of regulated genes that could impact neuronal functions collectively, and demonstrated in calcium imaging that indeed the spontaneous activity of neurons is influenced by RABV infection. Overall, our findings suggest that neuronal function-associated genes are modulated by RABV early on, potentially through the viral matrix protein-interacting signaling molecules and their downstream pathways.
Collapse
Affiliation(s)
- Seonhee Kim
- Technology Development Platform, Institut Pasteur Korea, Seongnam, South Korea
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
- Université de Paris, Doctoral School Bio Sorbonne Paris Cité, Paris, France
| | - Florence Larrous
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| | - Hugo Varet
- Institut Pasteur, Université de Paris, Hub de Bioinformatique et Biostatistique, Département Biologie Computationnelle, Paris, France
- Institut Pasteur, Université de Paris, Plate-Forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | - Rachel Legendre
- Institut Pasteur, Université de Paris, Hub de Bioinformatique et Biostatistique, Département Biologie Computationnelle, Paris, France
- Institut Pasteur, Université de Paris, Plate-Forme Technologique Biomics, Centre de Ressources et Recherches Technologiques (C2RT), Paris, France
| | - Lena Feige
- Technology Development Platform, Institut Pasteur Korea, Seongnam, South Korea
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
- Université de Paris, Doctoral School Bio Sorbonne Paris Cité, Paris, France
| | - Guillaume Dumas
- Department of Psychiatry, CHU Sainte-Justine Research Center, University of Montreal, Montreal, QC, Canada
- Mila, Quebec Artificial Intelligence Institute, University of Montreal, Montreal, QC, Canada
| | - Rebecca Matsas
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Georgia Kouroupi
- Laboratory of Cellular and Molecular Neurobiology-Stem Cells, Department of Neurobiology, Hellenic Pasteur Institute, Athens, Greece
| | - Regis Grailhe
- Technology Development Platform, Institut Pasteur Korea, Seongnam, South Korea
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, Paris, France
| |
Collapse
|
9
|
Feige L, Sáenz-de-Santa-María I, Regnault B, Lavenir R, Lepelletier A, Halacu A, Rajerison R, Diop S, Nareth C, Reynes JM, Buchy P, Bourhy H, Dacheux L. Transcriptome Profile During Rabies Virus Infection: Identification of Human CXCL16 as a Potential New Viral Target. Front Cell Infect Microbiol 2021; 11:761074. [PMID: 34804996 PMCID: PMC8602097 DOI: 10.3389/fcimb.2021.761074] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Accepted: 09/23/2021] [Indexed: 12/24/2022] Open
Abstract
Rabies virus (RABV), the causative agent for rabies disease is still presenting a major public health concern causing approximately 60,000 deaths annually. This neurotropic virus (genus Lyssavirus, family Rhabdoviridae) induces an acute and almost always fatal form of encephalomyelitis in humans. Despite the lethal consequences associated with clinical symptoms of rabies, RABV limits neuro-inflammation without causing major histopathological lesions in humans. Nevertheless, information about the mechanisms of infection and cellular response in the central nervous system (CNS) remain scarce. Here, we investigated the expression of inflammatory genes involved in immune response to RABV (dog-adapted strain Tha) in mice, the most common animal model used to study rabies. To better elucidate the pathophysiological mechanisms during natural RABV infection, we compared the inflammatory transcriptome profile observed at the late stage of infection in the mouse brain (cortex and brain stem/cerebellum) with the ortholog gene expression in post-mortem brain biopsies of rabid patients. Our data indicate that the inflammatory response associated with rabies is more pronounced in the murine brain compared to the human brain. In contrast to murine transcription profiles, we identified CXC motif chemokine ligand 16 (CXCL16) as the only significant differentially expressed gene in post-mortem brains of rabid patients. This result was confirmed in vitro, in which Tha suppressed interferon alpha (IFN-α)-induced CXCL16 expression in human CNS cell lines but induced CXCL16 expression in IFN-α-stimulated murine astrocytes. We hypothesize that RABV-induced modulation of the CXCL16 pathway in the brain possibly affects neurotransmission, natural killer (NK) and T cell recruitment and activation. Overall, we show species-specific differences in the inflammatory response of the brain, highlighted the importance of understanding the potential limitations of extrapolating data from animal models to humans.
Collapse
Affiliation(s)
- Lena Feige
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | | | | | - Rachel Lavenir
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | - Anthony Lepelletier
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | - Ala Halacu
- National Agency for Public Health, Chișinău, Moldova
| | | | - Sylvie Diop
- Infectious Diseases Department, National and University Hospital Center of Fann-Dakar, Dakar, Senegal
| | | | - Jean-Marc Reynes
- Virology Unit, Institut Pasteur de Madagascar, Tananarive, Madagascar
| | - Philippe Buchy
- Virology Unit, Institut Pasteur in Cambodia, Phnom Penh, Cambodia
| | - Hervé Bourhy
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| | - Laurent Dacheux
- Institut Pasteur, Université de Paris, Lyssavirus Epidemiology and Neuropathology Unit, National Reference Center for Rabies, WHO Collaborating Center for Reference and Research on Rabies, Department of Global Health, Paris, France
| |
Collapse
|
10
|
Kanu B, Kia GSN, Aimola IA, Korie GC, Tekki IS. Rabies virus infection is associated with alterations in the expression of parvalbumin and secretagogin in mice brain. Metab Brain Dis 2021; 36:1267-1275. [PMID: 33783673 PMCID: PMC8008021 DOI: 10.1007/s11011-021-00717-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 03/14/2021] [Indexed: 12/21/2022]
Abstract
Infection with the deadly rabies virus (RABV) leads to alteration of cellular gene expression. The RABV, similar to other neurodegenerative diseases may be implicated in neuronal death due to an imbalance in Ca2+ homeostasis. Parvalbumin (PV) and Secretagogin (Scgn), two members of the Calcium-Binding Proteins (CBPs) are useful neuronal markers responsible for calcium regulation and buffering with possible protective roles against infections. This study investigated whether infection with rabies virus causes variance in expression levels of PV and Scgn using the Challenge virus standard (CVS) and Nigerian Street Rabies virus (SRV) strains. Forty-eight, 4-week-old BALB/c mice strains were divided into two test groups and challenged with Rabies virus (RABV) infection and one control group. The presence of RABV antigen was verified by direct fluorescent antibody test (DFAT) and real-time quantitative PCR (qRT-PCR) was used to assess PV and Scgn gene expression. Infection with both virus strains resulted in significant (p < 0.05) increases in expression during early infection. Mid-infection phase caused reduced expression for both genes. However, as infection progressed to the terminal phase, a lower increase in expression was measured. Gene expression and viral load correlation indicated no positive relationship. Neurons with these CBPs may have a greater capacity to buffer calcium and be more resistant to degenerative changes caused by RABV. This implies that, when PV and Scgn expression levels are kept adequately high, the integrity of neurons may be maintained and degeneration caused by RABV infection may be prevented or stopped, hence, these are possible constituents of effective rabies therapy.
Collapse
Affiliation(s)
- Brenda Kanu
- Department of Biochemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria.
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria.
| | - Grace S N Kia
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria
- Department of Veterinary Public Health, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
| | - Idowu A Aimola
- Department of Biochemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria
| | - George C Korie
- Department of Biochemistry, Ahmadu Bello University, Zaria, Kaduna State, Nigeria
- Africa Centre of Excellence for Neglected Tropical Diseases and Forensic Biotechnology, Ahmadu Bello University Centre, Zaria, Kaduna State, Nigeria
| | - Ishaya S Tekki
- Central Diagnostics Laboratory, National Veterinary Research Institute, Vom, Plateau State, Nigeria
| |
Collapse
|
11
|
Assessing Rabies Vaccine Protection against a Novel Lyssavirus, Kotalahti Bat Lyssavirus. Viruses 2021; 13:v13050947. [PMID: 34065574 PMCID: PMC8161192 DOI: 10.3390/v13050947] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Revised: 05/07/2021] [Accepted: 05/12/2021] [Indexed: 12/20/2022] Open
Abstract
Rabies is a fatal encephalitis caused by an important group of viruses within the Lyssavirus genus. The prototype virus, rabies virus, is still the most commonly reported lyssavirus and causes approximately 59,000 human fatalities annually. The human and animal burden of the other lyssavirus species is undefined. The original reports for the novel lyssavirus, Kotalahti bat lyssavirus (KBLV), were based on the detection of viral RNA alone. In this report we describe the successful generation of a live recombinant virus, cSN-KBLV; where the full-length genome clone of RABV vaccine strain, SAD-B19, was constructed with the glycoprotein of KBLV. Subsequent in vitro characterisation of cSN-KBLV is described here. In addition, the ability of a human rabies vaccine to confer protective immunity in vivo following challenge with this recombinant virus was assessed. Naïve or vaccinated mice were infected intracerebrally with a dose of 100 focus-forming units/30 µL of cSN-KBLV; all naïve mice and 8% (n = 1/12) of the vaccinated mice succumbed to the challenge, whilst 92% (n = 11/12) of the vaccinated mice survived to the end of the experiment. This report provides strong evidence for cross-neutralisation and cross-protection of cSN-KBLV using purified Vero cell rabies vaccine.
Collapse
|
12
|
Luo J, Zhang B, Wu Y, Guo X. Amino Acid Mutation in Position 349 of Glycoprotein Affect the Pathogenicity of Rabies Virus. Front Microbiol 2020; 11:481. [PMID: 32308648 PMCID: PMC7145897 DOI: 10.3389/fmicb.2020.00481] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 03/05/2020] [Indexed: 12/25/2022] Open
Abstract
Rabies, caused by rabies virus (RABV), is a zoonotic disease infecting mammals including humans. Studies have confirmed that glycoprotein (G) is most related to RABV pathogenicity. In the present study, to discover more amino acid sites related to viral pathogenicity, artificial mutants have been constructed in G of virulent strain GD-SH-01 backbone. Results showed that pathogenicity of GD-SH-01 significantly decreased when Gly349 was replaced by Glu349 through in vivo assays. Gly349→Glu349 of G did not significantly influence viral growth and spread in NA cells. Gly349→Glu349 of G increased the immunogenicity of GD-SH-01 in periphery and induced more expression of interferon alpha (IFN-α) in the brain in mice. It was observed that Gly349→Glu349 of G led to enhanced blood–brain barrier (BBB) permeability at day 5 postinfection. All together, these data revealed that Gly349→Glu349 of G mutation decreased RABV pathogenicity through enhanced immune response and increased BBB permeability. This study provides a new referenced site G349 that could attenuate pathogenicity of RABV.
Collapse
Affiliation(s)
- Jun Luo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Boyue Zhang
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yuting Wu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Xiaofeng Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| |
Collapse
|
13
|
Asian Elephant T Cell Responses to Elephant Endotheliotropic Herpesvirus. J Virol 2018; 92:JVI.01951-17. [PMID: 29263271 PMCID: PMC5827410 DOI: 10.1128/jvi.01951-17] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/14/2017] [Indexed: 12/20/2022] Open
Abstract
Elephant endotheliotropic herpesvirus (EEHV) can cause lethal hemorrhagic disease in juvenile Asian elephants, an endangered species. One hypothesis to explain this vulnerability of some juvenile elephants is that they fail to mount an effective T cell response to the virus. To our knowledge, there have been no studies of Asian elephant T cell responses to EEHV. To address this deficiency, we validated the gamma interferon (IFN-γ) enzyme-linked immunospot assay for tracking antigen-directed T cell activity by monitoring rabies-specific responses in vaccinated elephants. In addition, we generated monoclonal antibodies to Asian elephant CD4 and CD8 to facilitate phenotypic T cell profiling. Using these tools, we screened healthy elephants with a history of EEHV infection for reactivity against nine EEHV proteins whose counterparts in other herpesviruses are known to induce T cell responses in their natural hosts. We identified glycoprotein B (gB) and the putative regulatory protein E40 as the most immunogenic T cell targets (IFN-γ responses in five of seven elephants), followed by the major capsid protein (IFN-γ responses in three of seven elephants). We also observed that IFN-γ responses were largely from CD4+ T cells. We detected no activity against the predicted major immediate early (E44) and large tegument (E34) proteins, both immunodominant T cell targets in humans latently infected with cytomegalovirus. These studies identified EEHV-specific T cells in Asian elephants for the first time, lending insight into the T cell priming that might be required to protect against EEHV disease, and will guide the design of effective vaccine strategies. IMPORTANCE Endangered Asian elephants are facing many threats, including lethal hemorrhagic disease from elephant endotheliotropic herpesvirus (EEHV). EEHV usually establishes chronic, benign infections in mature Asian elephants but can be lethal to juvenile elephants in captivity and the wild. It is the leading cause of death in captive Asian elephants in North America and Europe. Despite the availability of sensitive tests and protocols for treating EEHV-associated illness, these measures are not always effective. The best line of defense would be a preventative vaccine. We interrogated normal healthy elephants previously infected with EEHV for T cell responses to nine EEHV proteins predicted to induce cellular immune responses. Three proteins elicited IFN-γ responses, suggesting their potential usefulness as vaccine candidates. Our work is the first to describe T cell responses to a member of the proposed fourth subfamily of mammalian herpesviruses, the Deltaherpesvirinae, within a host species in the clade Afrotheria. An EEHV vaccine would greatly contribute to the health care of Asian and African elephants that are also susceptible to this disease.
Collapse
|
14
|
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
|
15
|
Gene Expression Profiling with Cre-Conditional Pseudorabies Virus Reveals a Subset of Midbrain Neurons That Participate in Reward Circuitry. J Neurosci 2017; 37:4128-4144. [PMID: 28283558 DOI: 10.1523/jneurosci.3193-16.2017] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 02/06/2017] [Accepted: 02/27/2017] [Indexed: 01/29/2023] Open
Abstract
The mesolimbic dopamine pathway receives inputs from numerous regions of the brain as part of a neural system that detects rewarding stimuli and coordinates a behavioral response. The capacity to simultaneously map and molecularly define the components of this complex multisynaptic circuit would thus advance our understanding of the determinants of motivated behavior. To accomplish this, we have constructed pseudorabies virus (PRV) strains in which viral propagation and fluorophore expression are activated only after exposure to Cre recombinase. Once activated in Cre-expressing neurons, the virus serially labels chains of presynaptic neurons. Dual injection of GFP and mCherry tracing viruses simultaneously illuminates nigrostriatal and mesolimbic circuitry and shows no overlap, demonstrating that PRV transmission is confined to synaptically connected neurons. To molecularly profile mesolimbic dopamine neurons and their presynaptic inputs, we injected Cre-conditional GFP virus into the NAc of (anti-GFP) nanobody-L10 transgenic mice and immunoprecipitated translating ribosomes from neurons infected after retrograde tracing. Analysis of purified RNA revealed an enrichment of transcripts expressed in neurons of the dorsal raphe nuclei and lateral hypothalamus that project to the mesolimbic dopamine circuit. These studies identify important inputs to the mesolimbic dopamine pathway and further show that PRV circuit-directed translating ribosome affinity purification can be broadly applied to identify molecularly defined neurons comprising complex, multisynaptic circuits.SIGNIFICANCE STATEMENT The mesolimbic dopamine circuit integrates signals from key brain regions to detect and respond to rewarding stimuli. To further define this complex multisynaptic circuit, we constructed a panel of Cre recombinase-activated pseudorabies viruses (PRVs) that enabled retrograde tracing of neural inputs that terminate on Cre-expressing neurons. Using these viruses and Retro-TRAP (translating ribosome affinity purification), a previously reported molecular profiling method, we developed a novel technique that provides anatomic as well as molecular information about the neural components of polysynaptic circuits. We refer to this new method as PRV-Circuit-TRAP (PRV circuit-directed TRAP). Using it, we have identified major projections to the mesolimbic dopamine circuit from the lateral hypothalamus and dorsal raphe nucleus and defined a discrete subset of transcripts expressed in these projecting neurons, which will allow further characterization of this important pathway. Moreover, the method we report is general and can be applied to the study of other neural circuits.
Collapse
|