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

Open Access 01-12-2018 | Research

Varicella zoster virus differentially alters morphology and suppresses proinflammatory cytokines in primary human spinal cord and hippocampal astrocytes

Authors: Andrew N. Bubak, Christina N. Como, Anna M. Blackmon, Dallas Jones, Maria A. Nagel

Published in: Journal of Neuroinflammation | Issue 1/2018

Login to get access

Abstract

Background

Varicella zoster virus (VZV) is a ubiquitous alphaherpesvirus that produces varicella and zoster. VZV can infect multiple cell types in the spinal cord and brain, including astrocytes, producing myelopathy and encephalopathy. While studies of VZV-astrocyte interactions are sparse, a recent report showed that quiescent primary human spinal cord astrocytes (qHA-sps) did not appear activated morphologically during VZV infection. Since astrocytes play a critical role in host defenses during viral infections of the central nervous system, we examined the cytokine responses of qHA-sps and quiescent primary human hippocampal astrocytes (qHA-hps) to VZV infection in vitro, as well as the ability of conditioned supernatant to recruit immune cells.

Methods

At 3 days post-infection, mock- and VZV-infected qHA-sps and qHA-hps were examined for morphological changes by immunofluorescence antibody assay using antibodies directed against glial fibrillary acidic protein and VZV. Conditioned supernatants were analyzed for proinflammatory cytokines [interleukin (IL)-1β, IL-2, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-13, interferon-gamma, and tumor necrosis factor-α] using the Meso Scale Discovery multiplex ELISA platform. Finally, the ability of conditioned supernatants to attract peripheral blood mononuclear cells (PBMCs) was determined using a chemotaxis assay. Quiescent primary human perineurial cells (qHPNCs) served as a control for VZV-induced cytokine production and PBMC migration. To confirm that the astrocytes have the ability to increase cytokine secretion, qHA-sps and qHA-hps were treated with IL-1β and examined for morphological changes and IL-6 secretion.

Results

VZV-infected qHA-sps displayed extensive cellular processes, whereas VZV-infected qHA-hps became swollen and clustered together. Astrocytes had the capacity to secrete IL-6 in response to IL-1β. Compared to mock-infected cells, VZV-infected qHA-sps showed significantly reduced secretion of IL-2, IL-4, IL-6, IL-12p70, and IL-13, while VZV-infected qHA-hps showed significantly reduced IL-8 secretion. In contrast, levels of all 10 cytokines examined were significantly increased in VZV-infected qHPNCs. Consistent with these results, conditioned supernatant from VZV-infected qHPNCs, but not that from VZV-infected qHA-sps and qHA-hps, recruited PBMCs.

Conclusions

VZV-infected qHA-sps and qHA-hps have distinct morphological alterations and patterns of proinflammatory cytokine suppression that could contribute to ineffective viral clearance in VZV myelopathy and encephalopathy, respectively.
Literature
1.
go back to reference Kennedy PG, Grinfeld E, Gow JW. Latent varicella-zoster virus is located predominantly in neurons in human trigeminal ganglia. Proc Natl Acad Sci U S A. 1998;95:4658–62.CrossRefPubMedPubMedCentral Kennedy PG, Grinfeld E, Gow JW. Latent varicella-zoster virus is located predominantly in neurons in human trigeminal ganglia. Proc Natl Acad Sci U S A. 1998;95:4658–62.CrossRefPubMedPubMedCentral
2.
go back to reference Kennedy PG, Grinfeld E, Gow JW. Latent varicella-zoster virus in human dorsal root ganglia. Virology. 1999;258:451–4.CrossRefPubMed Kennedy PG, Grinfeld E, Gow JW. Latent varicella-zoster virus in human dorsal root ganglia. Virology. 1999;258:451–4.CrossRefPubMed
3.
go back to reference Nagel MA, Rempel A, Huntington J, Kim F, Choe A, Gilden D. Frequency and abundance of alphaherpesvirus DNA in human thoracic sympathetic ganglia. J Virol. 2014;88:8189–92.CrossRefPubMedPubMedCentral Nagel MA, Rempel A, Huntington J, Kim F, Choe A, Gilden D. Frequency and abundance of alphaherpesvirus DNA in human thoracic sympathetic ganglia. J Virol. 2014;88:8189–92.CrossRefPubMedPubMedCentral
4.
go back to reference Badani H, White T, Schulick N, Raeburn CD, Topkaya I, Gilden D, Nagel MA. Frequency of varicella zoster virus DNA in human adrenal glands. J Neuro-Oncol. 2016;22:400–2. Badani H, White T, Schulick N, Raeburn CD, Topkaya I, Gilden D, Nagel MA. Frequency of varicella zoster virus DNA in human adrenal glands. J Neuro-Oncol. 2016;22:400–2.
5.
go back to reference Gilden DH, Beinlich BR, Rubinstien EM, Stommel E, Swenson R, Rubinstein D, Mahalingam R. Varicella-zoster virus myelitis: an expanding spectrum. Neurology. 1994;44:1818–23.CrossRefPubMed Gilden DH, Beinlich BR, Rubinstien EM, Stommel E, Swenson R, Rubinstein D, Mahalingam R. Varicella-zoster virus myelitis: an expanding spectrum. Neurology. 1994;44:1818–23.CrossRefPubMed
9.
10.
go back to reference Horten B, Price RW, Jimenez D. Multifocal varicella-zoster virus leukoencephalitis temporally remote from herpes zoster. Ann Neurol. 1981;9:251–66.CrossRefPubMed Horten B, Price RW, Jimenez D. Multifocal varicella-zoster virus leukoencephalitis temporally remote from herpes zoster. Ann Neurol. 1981;9:251–66.CrossRefPubMed
11.
go back to reference Moulignier A, Pialoux G, Dega H, Dupont B, Huerre M, Baudrimont M. Brain stem encephalitis due to varicella-zoster virus in a patient with AIDS. Clin Infect Dis. 1995;20:1378–80.CrossRefPubMed Moulignier A, Pialoux G, Dega H, Dupont B, Huerre M, Baudrimont M. Brain stem encephalitis due to varicella-zoster virus in a patient with AIDS. Clin Infect Dis. 1995;20:1378–80.CrossRefPubMed
12.
go back to reference Carpenter JE, Clayton AC, Halling KC, Bonthius DJ, Buckingham EM, Jackson W, Dotzler SM, Card JP, Enquist LW, Grose C. Defensive perimeter in the central nervous system: predominance of astrocytes and astrogliosis during recovery from varicella-zoster virus encephalitis. J Virol. 2015;90:379–91.CrossRefPubMedPubMedCentral Carpenter JE, Clayton AC, Halling KC, Bonthius DJ, Buckingham EM, Jackson W, Dotzler SM, Card JP, Enquist LW, Grose C. Defensive perimeter in the central nervous system: predominance of astrocytes and astrogliosis during recovery from varicella-zoster virus encephalitis. J Virol. 2015;90:379–91.CrossRefPubMedPubMedCentral
13.
go back to reference Assouline JG, Levin MJ, Major EO, Forghani B, Straus SE, Ostrove JM. Varicella-zoster virus infection of human astrocytes, Schwann cells, and neurons. Virology. 1990;179:834.CrossRefPubMed Assouline JG, Levin MJ, Major EO, Forghani B, Straus SE, Ostrove JM. Varicella-zoster virus infection of human astrocytes, Schwann cells, and neurons. Virology. 1990;179:834.CrossRefPubMed
14.
go back to reference Kennedy PG, Major EO, Williams RK, Straus SE. Down-regulation of glial fibrillary acidic protein expression during acute lytic varicella-zoster virus infection of cultured human astrocytes. Virology. 1994;205:558–62.CrossRefPubMed Kennedy PG, Major EO, Williams RK, Straus SE. Down-regulation of glial fibrillary acidic protein expression during acute lytic varicella-zoster virus infection of cultured human astrocytes. Virology. 1994;205:558–62.CrossRefPubMed
15.
go back to reference Bubak AN, Como CN, Blackmon AM, Frietze S, Mescher T, Jones D, Cohrs RJ, Paucek P, Baird NL, Nagel MA. Varicella zoster virus induces nuclear translocation of the neurokinin-1 receptor, promoting lamellipodia formation and viral spread in spinal astrocytes. J Infect Dis. 2018. https://doi.org/10.1093/infdis/jiy297 [Epub ahead of print].CrossRefPubMed Bubak AN, Como CN, Blackmon AM, Frietze S, Mescher T, Jones D, Cohrs RJ, Paucek P, Baird NL, Nagel MA. Varicella zoster virus induces nuclear translocation of the neurokinin-1 receptor, promoting lamellipodia formation and viral spread in spinal astrocytes. J Infect Dis. 2018. https://​doi.​org/​10.​1093/​infdis/​jiy297 [Epub ahead of print].CrossRefPubMed
17.
go back to reference Aloisi F, Carè A, Borsellino G, Gallo P, Rosa S, Bassani A, Cabibbo A, Testa U, Levi G, Peschle C. Production of hemolyphopoletic cytokines (IL-6, IL-8, colony-stimulating factors) by normal human astrocytes in response to IL-1 beta and tumor necrosis factor-alpha. J Immunol. 1992;149:2358–66.PubMed Aloisi F, Carè A, Borsellino G, Gallo P, Rosa S, Bassani A, Cabibbo A, Testa U, Levi G, Peschle C. Production of hemolyphopoletic cytokines (IL-6, IL-8, colony-stimulating factors) by normal human astrocytes in response to IL-1 beta and tumor necrosis factor-alpha. J Immunol. 1992;149:2358–66.PubMed
18.
go back to reference Benjamini Y, Krieger AM, Yekutieli D. Adaptive linear step-up procedures that control the false discovery rate. Biometrika. 2006;93:491–507.CrossRef Benjamini Y, Krieger AM, Yekutieli D. Adaptive linear step-up procedures that control the false discovery rate. Biometrika. 2006;93:491–507.CrossRef
19.
go back to reference Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA, Blumenthal PD, Vogel H, Steinberg GK, Edwards SM, Li G, Duncan JA III, Cheshier SH, Shuer LM, Chang EF, Grant GA, Gephart MG, Barres BA. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron. 2016;89:37–53.CrossRefPubMed Zhang Y, Sloan SA, Clarke LE, Caneda C, Plaza CA, Blumenthal PD, Vogel H, Steinberg GK, Edwards SM, Li G, Duncan JA III, Cheshier SH, Shuer LM, Chang EF, Grant GA, Gephart MG, Barres BA. Purification and characterization of progenitor and mature human astrocytes reveals transcriptional and functional differences with mouse. Neuron. 2016;89:37–53.CrossRefPubMed
20.
go back to reference Rehli M. Of mice and men: species variations of Toll-like receptor expression. Trends Immunol. 2002;23:375–8.CrossRefPubMed Rehli M. Of mice and men: species variations of Toll-like receptor expression. Trends Immunol. 2002;23:375–8.CrossRefPubMed
22.
go back to reference Lokensgard JR, Hu S, Sheng W, vanOijen M, Cox D, Cheeran MC, Peterson PK. Robust expression of TNF-alpha, IL-1beta, RANTES, and IP-10 by human microglial cells during nonproductive infection with herpes simplex virus. J Neuro-Oncol. 2001;7:208–19. Lokensgard JR, Hu S, Sheng W, vanOijen M, Cox D, Cheeran MC, Peterson PK. Robust expression of TNF-alpha, IL-1beta, RANTES, and IP-10 by human microglial cells during nonproductive infection with herpes simplex virus. J Neuro-Oncol. 2001;7:208–19.
23.
go back to reference Li J, Hu S, Zhou L, Ye L, Wang X, Ho J, Ho W. Interferon lambda inhibits herpes simplex virus type I infection of human astrocytes and neurons. Glia. 2011;59:58–67.CrossRefPubMed Li J, Hu S, Zhou L, Ye L, Wang X, Ho J, Ho W. Interferon lambda inhibits herpes simplex virus type I infection of human astrocytes and neurons. Glia. 2011;59:58–67.CrossRefPubMed
24.
go back to reference Cheeran MC, Hu S, Yager SL, Gekker G, Peterson PK, Lokensgard JR. Cytomegalovirus induces cytokine and chemokine production differentially in microglia and astrocytes: antiviral implications. J Neuro-Oncol. 2001;7:135–47. Cheeran MC, Hu S, Yager SL, Gekker G, Peterson PK, Lokensgard JR. Cytomegalovirus induces cytokine and chemokine production differentially in microglia and astrocytes: antiviral implications. J Neuro-Oncol. 2001;7:135–47.
25.
go back to reference Nitkiewicz J, Borjabad A, Morgello S, Murray J, Chao W, Emdad L, Fisher PF, Potash MJ, Volsky DJ. HIV induces expression of complement component C3 in astrocytes by NF-kappaB-dependent activation of interleukin-6 synthesis. J Neuroinflammation. 2017;14:23.CrossRefPubMedPubMedCentral Nitkiewicz J, Borjabad A, Morgello S, Murray J, Chao W, Emdad L, Fisher PF, Potash MJ, Volsky DJ. HIV induces expression of complement component C3 in astrocytes by NF-kappaB-dependent activation of interleukin-6 synthesis. J Neuroinflammation. 2017;14:23.CrossRefPubMedPubMedCentral
26.
go back to reference Stefanik M, Formanova P, Bily T, Vancova M, Eyer L, Palus M, Salat J, Braconi CT, Zanotto PMA, Gould EA, Ruzek D. Characterisation of Zika virus infection in primary human astrocytes. BMC Neurosci. 2018;19:5.CrossRefPubMedPubMedCentral Stefanik M, Formanova P, Bily T, Vancova M, Eyer L, Palus M, Salat J, Braconi CT, Zanotto PMA, Gould EA, Ruzek D. Characterisation of Zika virus infection in primary human astrocytes. BMC Neurosci. 2018;19:5.CrossRefPubMedPubMedCentral
27.
go back to reference Palus M, Bily T, Elsterova J, Langhansova H, Salat J, Vancova M, Ruzek D. Infection and injury of human astrocytes by tick-borne encephalitis virus. J Gen Virol. 2014;95:2411–26.CrossRefPubMed Palus M, Bily T, Elsterova J, Langhansova H, Salat J, Vancova M, Ruzek D. Infection and injury of human astrocytes by tick-borne encephalitis virus. J Gen Virol. 2014;95:2411–26.CrossRefPubMed
29.
go back to reference Feng M, Guo S, Fan S, Zeng X, Zhang Y, Liao Y, Wang J, Zhao T, Wang L, Che Y, Wang K, Ma N, Liu L, Yue L, Li Q. The preferential infection of astrocytes by enterovirus 71 plays a key role in the viral neurogenic pathogenesis. Front Cell Infect Microbiol. 2016;6:192.PubMedPubMedCentral Feng M, Guo S, Fan S, Zeng X, Zhang Y, Liao Y, Wang J, Zhao T, Wang L, Che Y, Wang K, Ma N, Liu L, Yue L, Li Q. The preferential infection of astrocytes by enterovirus 71 plays a key role in the viral neurogenic pathogenesis. Front Cell Infect Microbiol. 2016;6:192.PubMedPubMedCentral
30.
go back to reference Zhang X, Zheng Z, Shu B, Liu X, Zhang Z, Liu Y, Bai B, Hu Q, Mao P, Wang H. Human astrocytic cells support persistent coxsackievirus B3 infection. J Virol. 2013;87:12407–21.CrossRefPubMedPubMedCentral Zhang X, Zheng Z, Shu B, Liu X, Zhang Z, Liu Y, Bai B, Hu Q, Mao P, Wang H. Human astrocytic cells support persistent coxsackievirus B3 infection. J Virol. 2013;87:12407–21.CrossRefPubMedPubMedCentral
31.
go back to reference Lin X, Wang R, Zhang J, Sun X, Zou Z, Wang S, Jin M. Insights into human astrocyte response to H5N1 infection by microarray analysis. Viruses. 2015;7:2618–40.CrossRefPubMedPubMedCentral Lin X, Wang R, Zhang J, Sun X, Zou Z, Wang S, Jin M. Insights into human astrocyte response to H5N1 infection by microarray analysis. Viruses. 2015;7:2618–40.CrossRefPubMedPubMedCentral
32.
go back to reference He W, Zhao A, Anees A, Li Y, Ashraf U, Chen Z, Song Y, Chen H, Cao S, Ye J. p21-activated kinase 4 signaling promotes Japanese encephalitis virus-mediated inflammation in astrocytes. Front Cell Infect Microbiol. 2017;7:271.CrossRefPubMedPubMedCentral He W, Zhao A, Anees A, Li Y, Ashraf U, Chen Z, Song Y, Chen H, Cao S, Ye J. p21-activated kinase 4 signaling promotes Japanese encephalitis virus-mediated inflammation in astrocytes. Front Cell Infect Microbiol. 2017;7:271.CrossRefPubMedPubMedCentral
33.
go back to reference Molofsky AV, Kelley KW, Tsai HH, Redmond SA, Chang SM, Madireddy L, Chan JR, Baranzini SE, Ullian EM, Rowitch DH. Astrocyte-encoded positional cues maintain sensorimotor circuit integrity. Nature. 2014;509:189–94.CrossRefPubMedPubMedCentral Molofsky AV, Kelley KW, Tsai HH, Redmond SA, Chang SM, Madireddy L, Chan JR, Baranzini SE, Ullian EM, Rowitch DH. Astrocyte-encoded positional cues maintain sensorimotor circuit integrity. Nature. 2014;509:189–94.CrossRefPubMedPubMedCentral
34.
go back to reference Panatier A, Vallée J, Haber M, Murai KK, Lacaille JC, Robitaille R. Astrocytes are endogenous regulators of basal transmission at central synapses. Cell. 2011;146:785–98.CrossRefPubMed Panatier A, Vallée J, Haber M, Murai KK, Lacaille JC, Robitaille R. Astrocytes are endogenous regulators of basal transmission at central synapses. Cell. 2011;146:785–98.CrossRefPubMed
35.
go back to reference Haustein MD, Kracun S, Lu XH, Shih T, Jackson-Weaver O, Tong X, Xu J, Yang XW, O'Dell TJ, Marvin JS, Ellisman MH, Bushong EA, Looger LL, Khakh BS. Conditions and constraints for astrocyte calcium signaling in the hippocampal mossy fiber pathway. Neuron. 2014;82:413–29.CrossRefPubMedPubMedCentral Haustein MD, Kracun S, Lu XH, Shih T, Jackson-Weaver O, Tong X, Xu J, Yang XW, O'Dell TJ, Marvin JS, Ellisman MH, Bushong EA, Looger LL, Khakh BS. Conditions and constraints for astrocyte calcium signaling in the hippocampal mossy fiber pathway. Neuron. 2014;82:413–29.CrossRefPubMedPubMedCentral
36.
go back to reference Wang X, Zhang X, Yu Z, Zhang Q, Huang D, Yu S. Long-term outcomes of varicella zoster virus infection-related myelitis in 10 immunocompetent patients. J Neuroimmunol. 2018;321:36–40.CrossRefPubMed Wang X, Zhang X, Yu Z, Zhang Q, Huang D, Yu S. Long-term outcomes of varicella zoster virus infection-related myelitis in 10 immunocompetent patients. J Neuroimmunol. 2018;321:36–40.CrossRefPubMed
Metadata
Title
Varicella zoster virus differentially alters morphology and suppresses proinflammatory cytokines in primary human spinal cord and hippocampal astrocytes
Authors
Andrew N. Bubak
Christina N. Como
Anna M. Blackmon
Dallas Jones
Maria A. Nagel
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/s12974-018-1360-9

Other articles of this Issue 1/2018

Journal of Neuroinflammation 1/2018 Go to the issue