Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2018

Open Access 01-12-2018 | Research

Human neural stem cell-derived neuron/astrocyte co-cultures respond to La Crosse virus infection with proinflammatory cytokines and chemokines

Authors: Brian E. Dawes, Junling Gao, Colm Atkins, Jacob T. Nelson, Kendra Johnson, Ping Wu, Alexander N. Freiberg

Published in: Journal of Neuroinflammation | Issue 1/2018

Login to get access

Abstract

Background

La Crosse virus (LACV) causes pediatric encephalitis in the USA. LACV induces severe inflammation in the central nervous system, but the recruitment of inflammatory cells is poorly understood. A deeper understanding of LACV-induced neural pathology is needed in order to develop treatment options. However, there is a severe limitation of relevant human neuronal cell models of LACV infection.

Methods

We utilized human neural stem cell (hNSC)-derived neuron/astrocyte co-cultures to study LACV infection in disease-relevant primary cells. hNSCs were differentiated into neurons and astrocytes and infected with LACV. To characterize susceptibility and responses to infection, we measured viral titers and levels of viral RNA, performed immunofluorescence analysis to determine the cell types infected, performed apoptosis and cytotoxicity assays, and evaluated cellular responses to infection using qRT-PCR and Bioplex assays.

Results

hNSC-derived neuron/astrocyte co-cultures were susceptible to LACV infection and displayed apoptotic responses as reported in previous in vitro and in vivo studies. Neurons and astrocytes are both targets of LACV infection, with neurons becoming the predominant target later in infection possibly due to astrocytic responses to IFN. Additionally, neuron/astrocyte co-cultures responded to LACV infection with strong proinflammatory cytokine, chemokine, as well as MMP-2, MMP-7, and TIMP-1 responses.

Conclusions

hNSC-derived neuron/astrocyte co-cultures reproduce key aspects of LACV infection in humans and mice and are useful models to study encephalitic viruses. Specifically, we show astrocytes to be susceptible to LACV infection and that neurons and astrocytes are important drivers of the inflammatory responses seen in LACV infection through the production of proinflammatory cytokines and chemokines.
Appendix
Available only for authorised users
Literature
1.
go back to reference Soldan SS, González-Scarano F. The Bunyaviridae. Handb Clin Neurol. 2014;123:449–63.CrossRef Soldan SS, González-Scarano F. The Bunyaviridae. Handb Clin Neurol. 2014;123:449–63.CrossRef
3.
go back to reference Haddow AD, Odoi A. The incidence risk, clustering, and clinical presentation of La Crosse virus infections in the Eastern United States, 2003-2007. PLoS One. 2009;4:2003–7. Haddow AD, Odoi A. The incidence risk, clustering, and clinical presentation of La Crosse virus infections in the Eastern United States, 2003-2007. PLoS One. 2009;4:2003–7.
6.
go back to reference Balfour HH, Siem A, Quie G, Ph D. CALIFORNIA I . Clinical ARBOVIRUS ( LA CROSSE ) findings with meningoencephalitis. 1973;52. Balfour HH, Siem A, Quie G, Ph D. CALIFORNIA I . Clinical ARBOVIRUS ( LA CROSSE ) findings with meningoencephalitis. 1973;52.
7.
go back to reference Winkler CW, Race B, Phillips K, Peterson KE. Capillaries in the olfactory bulb but not the cortex are highly susceptible to virus-induced vascular leak and promote viral neuroinvasion. Acta Neuropathol. Springer Berlin Heidelberg. 2015;130:233–45.CrossRef Winkler CW, Race B, Phillips K, Peterson KE. Capillaries in the olfactory bulb but not the cortex are highly susceptible to virus-induced vascular leak and promote viral neuroinvasion. Acta Neuropathol. Springer Berlin Heidelberg. 2015;130:233–45.CrossRef
8.
go back to reference Kalfayan B. Pathology of La Crosse virus infection in humans. Prog Clin Biol Res. United States. 1983;123:179–86.PubMed Kalfayan B. Pathology of La Crosse virus infection in humans. Prog Clin Biol Res. United States. 1983;123:179–86.PubMed
9.
go back to reference Bennett RS, Cress CM, Ward JM, Firestone CY, Murphy BR, Whitehead SS. La Crosse virus infectivity, pathogenesis, and immunogenicity in mice and monkeys. Virol J. 2008;5:1–15.CrossRef Bennett RS, Cress CM, Ward JM, Firestone CY, Murphy BR, Whitehead SS. La Crosse virus infectivity, pathogenesis, and immunogenicity in mice and monkeys. Virol J. 2008;5:1–15.CrossRef
10.
go back to reference Johnson RT. Pathogenesis of La Crosse virus in mice. Prog Clin Biol Res United States. 1983;123:139–44.PubMed Johnson RT. Pathogenesis of La Crosse virus in mice. Prog Clin Biol Res United States. 1983;123:139–44.PubMed
13.
go back to reference Winkler CW, Myers LM, Woods TA, Carmody AB, Taylor KG, Peterson KE. Lymphocytes have a role in protection, but not in pathogenesis, during La Crosse virus infection in mice. J Neuroinflammation. 2017;14:1–14.CrossRef Winkler CW, Myers LM, Woods TA, Carmody AB, Taylor KG, Peterson KE. Lymphocytes have a role in protection, but not in pathogenesis, during La Crosse virus infection in mice. J Neuroinflammation. 2017;14:1–14.CrossRef
18.
go back to reference Daniels BP, Holman DW, Cruz-Orengo L, Jujjavarapu H, Durrant DM, Klein RS. Viral pathogen-associated molecular patterns regulate blood-brain barrier integrity via competing innate cytokine signals. MBio. 2014;5:1–13.CrossRef Daniels BP, Holman DW, Cruz-Orengo L, Jujjavarapu H, Durrant DM, Klein RS. Viral pathogen-associated molecular patterns regulate blood-brain barrier integrity via competing innate cytokine signals. MBio. 2014;5:1–13.CrossRef
20.
go back to reference Jordan PM, Cain LD, Wu P. Astrocytes enhance long-term survival of cholinergic neurons differentiated from human fetal neural stem cells. J Neurosci Res. 2008;86:35–47 United States.CrossRef Jordan PM, Cain LD, Wu P. Astrocytes enhance long-term survival of cholinergic neurons differentiated from human fetal neural stem cells. J Neurosci Res. 2008;86:35–47 United States.CrossRef
22.
go back to reference Grigoryan S, Kinchington PR, Yang IH, Selariu A, Zhu H, Yee M, et al. Retrograde axonal transport of VZV: kinetic studies in hESC-derived neurons. J Neuro-Oncol. 2012;18:462–70. Grigoryan S, Kinchington PR, Yang IH, Selariu A, Zhu H, Yee M, et al. Retrograde axonal transport of VZV: kinetic studies in hESC-derived neurons. J Neuro-Oncol. 2012;18:462–70.
25.
go back to reference Markus A, Lebenthal-Loinger I, Yang IH, Kinchington PR, Goldstein RS. An in vitro model of latency and reactivation of Varicella zoster virus in human stem cell-derived neurons. PLoS Pathog. 2015;11:1–22.CrossRef Markus A, Lebenthal-Loinger I, Yang IH, Kinchington PR, Goldstein RS. An in vitro model of latency and reactivation of Varicella zoster virus in human stem cell-derived neurons. PLoS Pathog. 2015;11:1–22.CrossRef
26.
go back to reference Lee KS, Zhou W, Scott-McKean JJ, Emmerling KL, Cai GY, Krah DL, et al. Human sensory neurons derived from induced pluripotent stem cells support Varicella-zoster virus infection. PLoS One. 2012;7:e53010.CrossRef Lee KS, Zhou W, Scott-McKean JJ, Emmerling KL, Cai GY, Krah DL, et al. Human sensory neurons derived from induced pluripotent stem cells support Varicella-zoster virus infection. PLoS One. 2012;7:e53010.CrossRef
28.
go back to reference Cai Y, Wu P, Ozen M, Yu Y, Wang J, Ittmann M, et al. Gene expression profiling and analysis of signaling pathways involved in priming and differentiation of human neural stem cells. Neuroscience. 2006;138:133–48.CrossRef Cai Y, Wu P, Ozen M, Yu Y, Wang J, Ittmann M, et al. Gene expression profiling and analysis of signaling pathways involved in priming and differentiation of human neural stem cells. Neuroscience. 2006;138:133–48.CrossRef
29.
go back to reference Tarasenko YI, Yu Y, Jordan PM, Bottenstein J, Wu P. Effect of growth factors on proliferation and phenotypic differentiation of human fetal neural stem cells. J Neurosci Res. 2004;78:625–36.CrossRef Tarasenko YI, Yu Y, Jordan PM, Bottenstein J, Wu P. Effect of growth factors on proliferation and phenotypic differentiation of human fetal neural stem cells. J Neurosci Res. 2004;78:625–36.CrossRef
30.
go back to reference McGrath EL, Rossi SL, Gao J, Widen SG, Grant AC, Dunn TJ, et al. Differential responses of human fetal brain neural stem cells to Zika virus infection. Stem Cell Reports. 2017;8:715–27.CrossRef McGrath EL, Rossi SL, Gao J, Widen SG, Grant AC, Dunn TJ, et al. Differential responses of human fetal brain neural stem cells to Zika virus infection. Stem Cell Reports. 2017;8:715–27.CrossRef
31.
go back to reference Yun T, Park A, Hill TE, Pernet O, Beaty SM, Juelich TL, et al. Efficient reverse genetics reveals genetic determinants of budding and fusogenic differences between Nipah and Hendra viruses and enables real-time monitoring of viral spread in small animal models of Henipavirus infection. J Virol. 2015;89(2):1242-1253. Available from: http://jvi.asm.org/lookup/doi/10.1128/JVI.02583-14.CrossRef Yun T, Park A, Hill TE, Pernet O, Beaty SM, Juelich TL, et al. Efficient reverse genetics reveals genetic determinants of budding and fusogenic differences between Nipah and Hendra viruses and enables real-time monitoring of viral spread in small animal models of Henipavirus infection. J Virol. 2015;89(2):1242-1253. Available from: http://​jvi.​asm.​org/​lookup/​doi/​10.​1128/​JVI.​02583-14.CrossRef
32.
go back to reference Herculano-Houzel S. The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia. 2014;62:1377–91 United States.CrossRef Herculano-Houzel S. The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution. Glia. 2014;62:1377–91 United States.CrossRef
35.
go back to reference Costello DA, Lynch MA. Toll-like receptor 3 activation modulates hippocampal network excitability, via glial production of interferon-?? Hippocampus. 2013;23:696–707.CrossRef Costello DA, Lynch MA. Toll-like receptor 3 activation modulates hippocampal network excitability, via glial production of interferon-?? Hippocampus. 2013;23:696–707.CrossRef
36.
go back to reference Gesuete R, Packard AEB, Vartanian KB, Conrad VK, Stevens SL, Bahjat FR, et al. Poly-ICLC preconditioning protects the blood-brain barrier against ischemic injury in vitro through type i interferon signaling. J Neurochem. 2012;123:75–85.CrossRef Gesuete R, Packard AEB, Vartanian KB, Conrad VK, Stevens SL, Bahjat FR, et al. Poly-ICLC preconditioning protects the blood-brain barrier against ischemic injury in vitro through type i interferon signaling. J Neurochem. 2012;123:75–85.CrossRef
37.
go back to reference Savarin C, Bergmann CC, Hinton DR, Stohlman SA. MMP-independent role of TIMP-1 at the blood brain barrier during viral encephalomyelitis. ASN Neuro. United States. 2013;5:e00127.CrossRef Savarin C, Bergmann CC, Hinton DR, Stohlman SA. MMP-independent role of TIMP-1 at the blood brain barrier during viral encephalomyelitis. ASN Neuro. United States. 2013;5:e00127.CrossRef
38.
go back to reference Groters S, Alldinger S, Baumgartner W. Up-regulation of mRNA for matrix metalloproteinases-9 and -14 in advanced lesions of demyelinating canine distemper leukoencephalitis. Acta Neuropathol. Germany. 2005;110:369–82.CrossRef Groters S, Alldinger S, Baumgartner W. Up-regulation of mRNA for matrix metalloproteinases-9 and -14 in advanced lesions of demyelinating canine distemper leukoencephalitis. Acta Neuropathol. Germany. 2005;110:369–82.CrossRef
39.
go back to reference Toft-Hansen H, Buist R, Sun X-J, Schellenberg A, Peeling J, Owens T. Metalloproteinases control brain inflammation induced by pertussis toxin in mice overexpressing the chemokine CCL2 in the central nervous system. J Immunol. United States. 2006;177:7242–9.CrossRef Toft-Hansen H, Buist R, Sun X-J, Schellenberg A, Peeling J, Owens T. Metalloproteinases control brain inflammation induced by pertussis toxin in mice overexpressing the chemokine CCL2 in the central nervous system. J Immunol. United States. 2006;177:7242–9.CrossRef
40.
go back to reference Buhler LA, Samara R, Guzman E, Wilson CL, Krizanac-Bengez L, Janigro D, et al. Matrix metalloproteinase-7 facilitates immune access to the CNS in experimental autoimmune encephalomyelitis. BMC Neurosci. England. 2009;10:17.CrossRef Buhler LA, Samara R, Guzman E, Wilson CL, Krizanac-Bengez L, Janigro D, et al. Matrix metalloproteinase-7 facilitates immune access to the CNS in experimental autoimmune encephalomyelitis. BMC Neurosci. England. 2009;10:17.CrossRef
41.
go back to reference Conant K, McArthur JC, Griffin DE, Sjulson L, Wahl LM, Irani DN. Cerebrospinal fluid levels of MMP-2, 7, and 9 are elevated in association with human immunodeficiency virus dementia. Ann Neurol. United States. 1999;46:391–8.CrossRef Conant K, McArthur JC, Griffin DE, Sjulson L, Wahl LM, Irani DN. Cerebrospinal fluid levels of MMP-2, 7, and 9 are elevated in association with human immunodeficiency virus dementia. Ann Neurol. United States. 1999;46:391–8.CrossRef
42.
go back to reference Gardner J, Ghorpade A. Tissue inhibitor of metalloproteinase (TIMP)-1: the TIMPed balance of matrix metalloproteinases in the central nervous system. J Neurosci Res. United States. 2003;74:801–6.CrossRef Gardner J, Ghorpade A. Tissue inhibitor of metalloproteinase (TIMP)-1: the TIMPed balance of matrix metalloproteinases in the central nervous system. J Neurosci Res. United States. 2003;74:801–6.CrossRef
48.
go back to reference Hill E, Nagel D, Parri R, Coleman M. Stem cell-derived astrocytes: are they physiologically credible? J Physiol. 2016;594:6595–606.CrossRef Hill E, Nagel D, Parri R, Coleman M. Stem cell-derived astrocytes: are they physiologically credible? J Physiol. 2016;594:6595–606.CrossRef
49.
go back to reference Banisadr G, Gosselin R-D, Mechighel P, Kitabgi P, Rostene W, Parsadaniantz SM. Highly regionalized neuronal expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) in rat brain: evidence for its colocalization with neurotransmitters and neuropeptides. J Comp Neurol. United States. 2005;489:275–92.CrossRef Banisadr G, Gosselin R-D, Mechighel P, Kitabgi P, Rostene W, Parsadaniantz SM. Highly regionalized neuronal expression of monocyte chemoattractant protein-1 (MCP-1/CCL2) in rat brain: evidence for its colocalization with neurotransmitters and neuropeptides. J Comp Neurol. United States. 2005;489:275–92.CrossRef
56.
go back to reference Smith PM, Wolcott RM, Chervenak R, Jennings SR. Control of acute cutaneous herpes simplex virus infection: T cell-mediated viral clearance is dependent upon interferon-gamma (IFN-gamma). Virology. United States. 1994;202:76–88.CrossRef Smith PM, Wolcott RM, Chervenak R, Jennings SR. Control of acute cutaneous herpes simplex virus infection: T cell-mediated viral clearance is dependent upon interferon-gamma (IFN-gamma). Virology. United States. 1994;202:76–88.CrossRef
57.
go back to reference Rodriguez M, Zoecklein LJ, Howe CL, Pavelko KD, Gamez JD, Nakane S, et al. Gamma interferon is critical for neuronal viral clearance and protection in a susceptible mouse strain following early intracranial Theiler’s murine encephalomyelitis virus infection. J Virol. United States. 2003;77:12252–65.CrossRef Rodriguez M, Zoecklein LJ, Howe CL, Pavelko KD, Gamez JD, Nakane S, et al. Gamma interferon is critical for neuronal viral clearance and protection in a susceptible mouse strain following early intracranial Theiler’s murine encephalomyelitis virus infection. J Virol. United States. 2003;77:12252–65.CrossRef
58.
go back to reference Burdeinick-Kerr R, Griffin DE. Gamma interferon-dependent, noncytolytic clearance of sindbis virus infection from neurons in vitro. J Virol United States. 2005;79:5374–85.CrossRef Burdeinick-Kerr R, Griffin DE. Gamma interferon-dependent, noncytolytic clearance of sindbis virus infection from neurons in vitro. J Virol United States. 2005;79:5374–85.CrossRef
61.
go back to reference Mathieu C, Guillaume V, Sabine A, Ong KC, Wong KT, Legras-Lachuer C, et al. Lethal Nipah virus infection induces rapid overexpression of cxcl10. PLoS One. 2012;7:e32157.CrossRef Mathieu C, Guillaume V, Sabine A, Ong KC, Wong KT, Legras-Lachuer C, et al. Lethal Nipah virus infection induces rapid overexpression of cxcl10. PLoS One. 2012;7:e32157.CrossRef
63.
go back to reference Lin CC, Wu YJ, Heimrich B, Schwemmle M. Absence of a robust innate immune response in rat neurons facilitates persistent infection of Borna disease virus in neuronal tissue. Cell Mol Life Sci. 2013;70:4399–410.CrossRef Lin CC, Wu YJ, Heimrich B, Schwemmle M. Absence of a robust innate immune response in rat neurons facilitates persistent infection of Borna disease virus in neuronal tissue. Cell Mol Life Sci. 2013;70:4399–410.CrossRef
64.
go back to reference Reed C, Lin K, Wilhelmsen C, Friedrich B, Nalca A, Keeney A, et al. Aerosol exposure to Rift Valley fever virus causes earlier and more severe neuropathology in the murine model, which has important implications for therapeutic development. PLoS Negl Trop Dis. 2013;7:e2156.CrossRef Reed C, Lin K, Wilhelmsen C, Friedrich B, Nalca A, Keeney A, et al. Aerosol exposure to Rift Valley fever virus causes earlier and more severe neuropathology in the murine model, which has important implications for therapeutic development. PLoS Negl Trop Dis. 2013;7:e2156.CrossRef
65.
go back to reference Dodd KA, McElroy AK, Jones TL, Zaki SR, Nichol ST, Spiropoulou CF. Rift Valley fever virus encephalitis is associated with an ineffective systemic immune response and activated T cell infiltration into the CNS in an immunocompetent mouse model. PLoS Negl Trop Dis. 2014;8:e2874.CrossRef Dodd KA, McElroy AK, Jones TL, Zaki SR, Nichol ST, Spiropoulou CF. Rift Valley fever virus encephalitis is associated with an ineffective systemic immune response and activated T cell infiltration into the CNS in an immunocompetent mouse model. PLoS Negl Trop Dis. 2014;8:e2874.CrossRef
Metadata
Title
Human neural stem cell-derived neuron/astrocyte co-cultures respond to La Crosse virus infection with proinflammatory cytokines and chemokines
Authors
Brian E. Dawes
Junling Gao
Colm Atkins
Jacob T. Nelson
Kendra Johnson
Ping Wu
Alexander N. Freiberg
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2018
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1356-5

Other articles of this Issue 1/2018

Journal of Neuroinflammation 1/2018 Go to the issue