Skip to main content
Top
Published in: Journal of Neuroinflammation 1/2016

Open Access 01-12-2016 | Research

Glial cell activation, recruitment, and survival of B-lineage cells following MCMV brain infection

Authors: James R. Lokensgard, Manohar B. Mutnal, Sujata Prasad, Wen Sheng, Shuxian Hu

Published in: Journal of Neuroinflammation | Issue 1/2016

Login to get access

Abstract

Background

Chemokines produced by reactive glia drive migration of immune cells and previous studies from our laboratory have demonstrated that CD19+ B cells infiltrate the brain. In this study, in vivo and in vitro experiments investigated the role of reactive glial cells in recruitment and survival of B-lineage cells in response to (murine cytomegalovirus) MCMV infection.

Methods

Flow cytometric analysis was used to assess chemokine receptor expression on brain-infiltrating B cells. Real-time RT-PCR and ELISA were used to measure chemokine levels. Dual-immunohistochemical staining was used to co-localize chemokine production by reactive glia. Primary glial cell cultures and migration assays were used to examine chemokine-mediated recruitment. Astrocyte: B cell co-cultures were used to investigate survival and proliferation.

Results

The chemokine receptors CXCR3, CXCR5, CCR5, and CCR7 were detected on CD19+ cells isolated from the brain during MCMV infection. In particular, CXCR3 was found to be elevated on an increasing number of cells over the time course of infection, and it was the primary chemokine receptor expressed at 60 days post infection Quite different expression kinetics were observed for CXCR5, CCR5, and CCR7, which were elevated on the highest number of cells early during infection and decreased by 14, 30, and 60 days post infection Correspondingly, elevated levels of CXCL9, CXCL10, and CXCL13, as well as CCL5, were found within the brains of infected animals, and only low levels of CCL3 and CCL19 were detected. Differential expression of CXCL9/CXCL10 and CXCL13 between microglia and astrocytes was apparent, and B cells moved towards supernatants from MCMV-infected microglia, but not astrocytes. Pretreatment with neutralizing Abs to CXCL9 and CXCL10 inhibited this migration. In contrast, neutralizing Abs to the ligand of CXCR5 (i.e., CXCL13) did not significantly block chemotaxis. Proliferation of brain-infiltrating B cells was detected at 7 days post infection and persisted through the latest time tested (60 days post infection). Finally, astrocytes produce BAFF (B cell activating factor of the TNF family) and promote proliferation of B cells via cell-to-cell contact.

Conclusions

CXCR3 is the primary chemokine receptor on CD19+ B cells persisting within the brain, and migration to microglial cell supernatants is mediated through this receptor. Correspondingly, microglial cells produce CXCL9 and CXCL10, but not CXCL13. Reactive astrocytes promote B cell proliferation.
Literature
1.
go back to reference Fragkoudis R, Ballany CM, Boyd A, Fazakerley JK. In Semliki Forest virus encephalitis, antibody rapidly clears infectious virus and is required to eliminate viral material from the brain, but is not required to generate lesions of demyelination. J Gen Virol. 2008;89:2565–8.CrossRefPubMed Fragkoudis R, Ballany CM, Boyd A, Fazakerley JK. In Semliki Forest virus encephalitis, antibody rapidly clears infectious virus and is required to eliminate viral material from the brain, but is not required to generate lesions of demyelination. J Gen Virol. 2008;89:2565–8.CrossRefPubMed
2.
go back to reference Griffin D, Levine B, Tyor W, Ubol S, Despres P. The role of antibody in recovery from alphavirus encephalitis. Immunol Rev. 1997;159:155–61.CrossRefPubMed Griffin D, Levine B, Tyor W, Ubol S, Despres P. The role of antibody in recovery from alphavirus encephalitis. Immunol Rev. 1997;159:155–61.CrossRefPubMed
3.
go back to reference Hooper DC, Phares TW, Fabis MJ, Roy A. The production of antibody by invading B cells is required for the clearance of rabies virus from the central nervous system. PLoS Negl Trop Dis. 2009;3:e535.CrossRefPubMedPubMedCentral Hooper DC, Phares TW, Fabis MJ, Roy A. The production of antibody by invading B cells is required for the clearance of rabies virus from the central nervous system. PLoS Negl Trop Dis. 2009;3:e535.CrossRefPubMedPubMedCentral
4.
go back to reference Levine B, Hardwick JM, Trapp BD, Crawford TO, Bollinger RC, Griffin DE. Antibody-mediated clearance of alphavirus infection from neurons. Science. 1991;254:856–60.CrossRefPubMed Levine B, Hardwick JM, Trapp BD, Crawford TO, Bollinger RC, Griffin DE. Antibody-mediated clearance of alphavirus infection from neurons. Science. 1991;254:856–60.CrossRefPubMed
5.
go back to reference Stewart BS, Demarest VL, Wong SJ, Green S, Bernard KA. Persistence of virus-specific immune responses in the central nervous system of mice after West Nile virus infection. BMC Immunol. 2011;12:6.CrossRefPubMedPubMedCentral Stewart BS, Demarest VL, Wong SJ, Green S, Bernard KA. Persistence of virus-specific immune responses in the central nervous system of mice after West Nile virus infection. BMC Immunol. 2011;12:6.CrossRefPubMedPubMedCentral
6.
go back to reference Phares TW, Marques CP, Stohlman SA, Hinton DR, Bergmann CC. Factors supporting intrathecal humoral responses following viral encephalomyelitis. J Virol. 2011;85:2589–98.CrossRefPubMed Phares TW, Marques CP, Stohlman SA, Hinton DR, Bergmann CC. Factors supporting intrathecal humoral responses following viral encephalomyelitis. J Virol. 2011;85:2589–98.CrossRefPubMed
7.
go back to reference Mutnal MB, Hu S, Lokensgard JR. Persistent humoral immune responses in the CNS limit recovery of reactivated murine cytomegalovirus. PLoS One. 2012;7:e33143.CrossRefPubMedPubMedCentral Mutnal MB, Hu S, Lokensgard JR. Persistent humoral immune responses in the CNS limit recovery of reactivated murine cytomegalovirus. PLoS One. 2012;7:e33143.CrossRefPubMedPubMedCentral
8.
go back to reference Cheeran MC, Lokensgard JR, Schleiss MR. Neuropathogenesis of congenital cytomegalovirus infection: disease mechanisms and prospects for intervention. Clin Microbiol Rev. 2009;22:99–126.CrossRefPubMedPubMedCentral Cheeran MC, Lokensgard JR, Schleiss MR. Neuropathogenesis of congenital cytomegalovirus infection: disease mechanisms and prospects for intervention. Clin Microbiol Rev. 2009;22:99–126.CrossRefPubMedPubMedCentral
9.
go back to reference Cheeran MC, Jiang Z, Hu S, Ni HT, Palmquist JM, Lokensgard JR. Cytomegalovirus infection and interferon-gamma modulate major histocompatibility complex class I expression on neural stem cells. J Neurovirol. 2008;14:437–47.CrossRefPubMedPubMedCentral Cheeran MC, Jiang Z, Hu S, Ni HT, Palmquist JM, Lokensgard JR. Cytomegalovirus infection and interferon-gamma modulate major histocompatibility complex class I expression on neural stem cells. J Neurovirol. 2008;14:437–47.CrossRefPubMedPubMedCentral
10.
go back to reference Cheeran MC, Gekker G, Hu S, Palmquist JM, Lokensgard JR. T cell-mediated restriction of intracerebral murine cytomegalovirus infection displays dependence upon perforin but not interferon-gamma. J Neurovirol. 2005;11:274–80.CrossRefPubMed Cheeran MC, Gekker G, Hu S, Palmquist JM, Lokensgard JR. T cell-mediated restriction of intracerebral murine cytomegalovirus infection displays dependence upon perforin but not interferon-gamma. J Neurovirol. 2005;11:274–80.CrossRefPubMed
11.
go back to reference Reddehase MJ, Mutter W, Munch K, Buhring HJ, Koszinowski UH. CD8-positive T lymphocytes specific for murine cytomegalovirus immediate-early antigens mediate protective immunity. J Virol. 1987;61:3102–8.PubMedPubMedCentral Reddehase MJ, Mutter W, Munch K, Buhring HJ, Koszinowski UH. CD8-positive T lymphocytes specific for murine cytomegalovirus immediate-early antigens mediate protective immunity. J Virol. 1987;61:3102–8.PubMedPubMedCentral
12.
go back to reference Reddehase MJ, Balthesen M, Rapp M, Jonjic S, Pavic I, Koszinowski UH. The conditions of primary infection define the load of latent viral genome in organs and the risk of recurrent cytomegalovirus disease. J Exp Med. 1994;179:185–93.CrossRefPubMed Reddehase MJ, Balthesen M, Rapp M, Jonjic S, Pavic I, Koszinowski UH. The conditions of primary infection define the load of latent viral genome in organs and the risk of recurrent cytomegalovirus disease. J Exp Med. 1994;179:185–93.CrossRefPubMed
13.
go back to reference Mutnal MB, Hu S, Little MR, Lokensgard JR. Memory T cells persisting in the brain following MCMV infection induce long-term microglial activation via interferon-gamma. J Neurovirol. 2011;17:424–37.CrossRefPubMedPubMedCentral Mutnal MB, Hu S, Little MR, Lokensgard JR. Memory T cells persisting in the brain following MCMV infection induce long-term microglial activation via interferon-gamma. J Neurovirol. 2011;17:424–37.CrossRefPubMedPubMedCentral
14.
go back to reference Reddehase MJ, Podlech J, Grzimek NK. Mouse models of cytomegalovirus latency: overview. J Clin Virol. 2002;25 Suppl 2:S23–36.CrossRefPubMed Reddehase MJ, Podlech J, Grzimek NK. Mouse models of cytomegalovirus latency: overview. J Clin Virol. 2002;25 Suppl 2:S23–36.CrossRefPubMed
15.
go back to reference Shapiro-Shelef M, Calame K. Regulation of plasma-cell development. Nat Rev Immunol. 2005;5:230–42.CrossRefPubMed Shapiro-Shelef M, Calame K. Regulation of plasma-cell development. Nat Rev Immunol. 2005;5:230–42.CrossRefPubMed
16.
go back to reference Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immunity due to long-lived plasma cells. Immunity. 1998;8:363–72.CrossRefPubMed Slifka MK, Antia R, Whitmire JK, Ahmed R. Humoral immunity due to long-lived plasma cells. Immunity. 1998;8:363–72.CrossRefPubMed
17.
go back to reference Brandes M, Legler DF, Spoerri B, Schaerli P, Moser B. Activation-dependent modulation of B lymphocyte migration to chemokines. Int Immunol. 2000;12:1285–92.CrossRefPubMed Brandes M, Legler DF, Spoerri B, Schaerli P, Moser B. Activation-dependent modulation of B lymphocyte migration to chemokines. Int Immunol. 2000;12:1285–92.CrossRefPubMed
18.
go back to reference Corcione A, Casazza S, Ferretti E, Giunti D, Zappia E, Pistorio A, Gambini C, Mancardi GL, Uccelli A, Pistoia V. Recapitulation of B cell differentiation in the central nervous system of patients with multiple sclerosis. Proc Natl Acad Sci U S A. 2004;101:11064–9.CrossRefPubMedPubMedCentral Corcione A, Casazza S, Ferretti E, Giunti D, Zappia E, Pistorio A, Gambini C, Mancardi GL, Uccelli A, Pistoia V. Recapitulation of B cell differentiation in the central nervous system of patients with multiple sclerosis. Proc Natl Acad Sci U S A. 2004;101:11064–9.CrossRefPubMedPubMedCentral
19.
go back to reference Serafini B, Rosicarelli B, Magliozzi R, Stigliano E, Aloisi F. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol. 2004;14:164–74.CrossRefPubMed Serafini B, Rosicarelli B, Magliozzi R, Stigliano E, Aloisi F. Detection of ectopic B-cell follicles with germinal centers in the meninges of patients with secondary progressive multiple sclerosis. Brain Pathol. 2004;14:164–74.CrossRefPubMed
20.
go back to reference Kowarik MC, Cepok S, Sellner J, Grummel V, Weber MS, Korn T, Berthele A, Hemmer B. CXCL13 is the major determinant for B cell recruitment to the CSF during neuroinflammation. J Neuroinflammation. 2012;9:93.CrossRefPubMedPubMedCentral Kowarik MC, Cepok S, Sellner J, Grummel V, Weber MS, Korn T, Berthele A, Hemmer B. CXCL13 is the major determinant for B cell recruitment to the CSF during neuroinflammation. J Neuroinflammation. 2012;9:93.CrossRefPubMedPubMedCentral
21.
go back to reference Muehlinghaus G, Cigliano L, Huehn S, Peddinghaus A, Leyendeckers H, Hauser AE, Hiepe F, Radbruch A, Arce S, Manz RA. Regulation of CXCR3 and CXCR4 expression during terminal differentiation of memory B cells into plasma cells. Blood. 2005;105:3965–71.CrossRefPubMed Muehlinghaus G, Cigliano L, Huehn S, Peddinghaus A, Leyendeckers H, Hauser AE, Hiepe F, Radbruch A, Arce S, Manz RA. Regulation of CXCR3 and CXCR4 expression during terminal differentiation of memory B cells into plasma cells. Blood. 2005;105:3965–71.CrossRefPubMed
22.
go back to reference Marques CP, Kapil P, Hinton DR, Hindinger C, Nutt SL, Ransohoff RM, Phares TW, Stohlman SA, Bergmann CC. CXCR3-dependent plasma blast migration to the central nervous system during viral encephalomyelitis. J Virol. 2011;85:6136–47.CrossRefPubMedPubMedCentral Marques CP, Kapil P, Hinton DR, Hindinger C, Nutt SL, Ransohoff RM, Phares TW, Stohlman SA, Bergmann CC. CXCR3-dependent plasma blast migration to the central nervous system during viral encephalomyelitis. J Virol. 2011;85:6136–47.CrossRefPubMedPubMedCentral
23.
go back to reference Metcalf TU, Baxter VK, Nilaratanakul V, Griffin DE. Recruitment and retention of B cells in the central nervous system in response to alphavirus encephalomyelitis. J Virol. 2013;87:2420–9.CrossRefPubMedPubMedCentral Metcalf TU, Baxter VK, Nilaratanakul V, Griffin DE. Recruitment and retention of B cells in the central nervous system in response to alphavirus encephalomyelitis. J Virol. 2013;87:2420–9.CrossRefPubMedPubMedCentral
24.
go back to reference Stoddart CA, Cardin RD, Boname JM, Manning WC, Abenes GB, Mocarski ES. Peripheral blood mononuclear phagocytes mediate dissemination of murine cytomegalovirus. J Virol. 1994;68:6243–53.PubMedPubMedCentral Stoddart CA, Cardin RD, Boname JM, Manning WC, Abenes GB, Mocarski ES. Peripheral blood mononuclear phagocytes mediate dissemination of murine cytomegalovirus. J Virol. 1994;68:6243–53.PubMedPubMedCentral
25.
go back to reference Cheeran MC, Gekker G, Hu S, Min X, Cox D, Lokensgard JR. Intracerebral infection with murine cytomegalovirus induces CXCL10 and is restricted by adoptive transfer of splenocytes. J Neurovirol. 2004;10:152–62.CrossRefPubMed Cheeran MC, Gekker G, Hu S, Min X, Cox D, Lokensgard JR. Intracerebral infection with murine cytomegalovirus induces CXCL10 and is restricted by adoptive transfer of splenocytes. J Neurovirol. 2004;10:152–62.CrossRefPubMed
26.
go back to reference Cheeran MC, Hu S, Palmquist JM, Bakken T, Gekker G, Lokensgard JR. Dysregulated interferon-gamma responses during lethal cytomegalovirus brain infection of IL-10-deficient mice. Virus Res. 2007;130:96–102.CrossRefPubMedPubMedCentral Cheeran MC, Hu S, Palmquist JM, Bakken T, Gekker G, Lokensgard JR. Dysregulated interferon-gamma responses during lethal cytomegalovirus brain infection of IL-10-deficient mice. Virus Res. 2007;130:96–102.CrossRefPubMedPubMedCentral
27.
go back to reference Ford AL, Goodsall AL, Hickey WF, Sedgwick JD. Normal adult ramified microglia separated from other central nervous system macrophages by flow cytometric sorting. Phenotypic differences defined and direct ex vivo antigen presentation to myelin basic protein-reactive CD4+ T cells compared. J Immunol. 1995;154:4309–21.PubMed Ford AL, Goodsall AL, Hickey WF, Sedgwick JD. Normal adult ramified microglia separated from other central nervous system macrophages by flow cytometric sorting. Phenotypic differences defined and direct ex vivo antigen presentation to myelin basic protein-reactive CD4+ T cells compared. J Immunol. 1995;154:4309–21.PubMed
28.
go back to reference Marten NW, Stohlman SA, Zhou J, Bergmann CC. Kinetics of virus-specific CD8+-T-cell expansion and trafficking following central nervous system infection. J Virol. 2003;77:2775–8.CrossRefPubMedPubMedCentral Marten NW, Stohlman SA, Zhou J, Bergmann CC. Kinetics of virus-specific CD8+-T-cell expansion and trafficking following central nervous system infection. J Virol. 2003;77:2775–8.CrossRefPubMedPubMedCentral
29.
go back to reference Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001;25:402–8.CrossRefPubMed Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) Method. Methods. 2001;25:402–8.CrossRefPubMed
30.
go back to reference Mutnal MB, Hu S, Schachtele SJ, Lokensgard JR. Infiltrating regulatory B cells control neuroinflammation following viral brain infection. J Immunol. 2014;193:6070–80.CrossRefPubMedPubMedCentral Mutnal MB, Hu S, Schachtele SJ, Lokensgard JR. Infiltrating regulatory B cells control neuroinflammation following viral brain infection. J Immunol. 2014;193:6070–80.CrossRefPubMedPubMedCentral
31.
go back to reference Steinert EM, Schenkel JM, Fraser KA, Beura LK, Manlove LS, Igyarto BZ, Southern PJ, Masopust D. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell. 2015;161:737–49.CrossRefPubMedPubMedCentral Steinert EM, Schenkel JM, Fraser KA, Beura LK, Manlove LS, Igyarto BZ, Southern PJ, Masopust D. Quantifying memory CD8 T cells reveals regionalization of immunosurveillance. Cell. 2015;161:737–49.CrossRefPubMedPubMedCentral
32.
go back to reference Liu RX, Wei Y, Zeng QH, Chan KW, Xiao X, Zhao XY, Chen MM, Ouyang FZ Chen DP, Zheng L, et al. Chemokine (C-X-C motif) receptor 3-positive B cells link interleukin-17 inflammation to protumorigenic macrophage polarization in human hepatocellular carcinoma. Hepatology. 2015;62:1779–90.CrossRefPubMed Liu RX, Wei Y, Zeng QH, Chan KW, Xiao X, Zhao XY, Chen MM, Ouyang FZ Chen DP, Zheng L, et al. Chemokine (C-X-C motif) receptor 3-positive B cells link interleukin-17 inflammation to protumorigenic macrophage polarization in human hepatocellular carcinoma. Hepatology. 2015;62:1779–90.CrossRefPubMed
33.
go back to reference Vincent FB, Saulep-Easton D, Figgett WA, Fairfax KA, Mackay F. The BAFF/APRIL system: emerging functions beyond B cell biology and autoimmunity. Cytokine Growth Factor Rev. 2013;24:203–15.CrossRefPubMed Vincent FB, Saulep-Easton D, Figgett WA, Fairfax KA, Mackay F. The BAFF/APRIL system: emerging functions beyond B cell biology and autoimmunity. Cytokine Growth Factor Rev. 2013;24:203–15.CrossRefPubMed
34.
go back to reference Krumbholz M, Theil D, Derfuss T, Rosenwald A, Schrader F, Monoranu CM, Kalled SL, Hess DM, Serafini B, Aloisi F, et al. BAFF is produced by astrocytes and up-regulated in multiple sclerosis lesions and primary central nervous system lymphoma. J Exp Med. 2005;201:195–200.CrossRefPubMedPubMedCentral Krumbholz M, Theil D, Derfuss T, Rosenwald A, Schrader F, Monoranu CM, Kalled SL, Hess DM, Serafini B, Aloisi F, et al. BAFF is produced by astrocytes and up-regulated in multiple sclerosis lesions and primary central nervous system lymphoma. J Exp Med. 2005;201:195–200.CrossRefPubMedPubMedCentral
35.
go back to reference Hauser AE, Debes GF, Arce S, Cassese G, Hamann A, Radbruch A, Manz RA. Chemotactic responsiveness toward ligands for CXCR3 and CXCR4 is regulated on plasma blasts during the time course of a memory immune response. J Immunol. 2002;169:1277–82.CrossRefPubMed Hauser AE, Debes GF, Arce S, Cassese G, Hamann A, Radbruch A, Manz RA. Chemotactic responsiveness toward ligands for CXCR3 and CXCR4 is regulated on plasma blasts during the time course of a memory immune response. J Immunol. 2002;169:1277–82.CrossRefPubMed
36.
go back to reference Ellis SL, Gysbers V, Manders PM, Li W, Hofer MJ, Muller M, Campbell IL. The cell-specific induction of CXC chemokine ligand 9 mediated by IFN-gamma in microglia of the central nervous system is determined by the myeloid transcription factor PU.1. J Immunol. 2010;185:1864–77.CrossRefPubMedPubMedCentral Ellis SL, Gysbers V, Manders PM, Li W, Hofer MJ, Muller M, Campbell IL. The cell-specific induction of CXC chemokine ligand 9 mediated by IFN-gamma in microglia of the central nervous system is determined by the myeloid transcription factor PU.1. J Immunol. 2010;185:1864–77.CrossRefPubMedPubMedCentral
37.
go back to reference Guillemin GJ, Croitoru-Lamoury J, Dormont D, Armati PJ, Brew BJ. Quinolinic acid upregulates chemokine production and chemokine receptor expression in astrocytes. Glia. 2003;41:371–81.CrossRefPubMed Guillemin GJ, Croitoru-Lamoury J, Dormont D, Armati PJ, Brew BJ. Quinolinic acid upregulates chemokine production and chemokine receptor expression in astrocytes. Glia. 2003;41:371–81.CrossRefPubMed
38.
go back to reference Rainey-Barger EK, Rumble JM, Lalor SJ, Esen N, Segal BM, Irani DN. The lymphoid chemokine, CXCL13, is dispensable for the initial recruitment of B cells to the acutely inflamed central nervous system. Brain Behav Immun. 2011;25:922–31.CrossRefPubMed Rainey-Barger EK, Rumble JM, Lalor SJ, Esen N, Segal BM, Irani DN. The lymphoid chemokine, CXCL13, is dispensable for the initial recruitment of B cells to the acutely inflamed central nervous system. Brain Behav Immun. 2011;25:922–31.CrossRefPubMed
39.
go back to reference Rolink AG, Melchers F. BAFFled B cells survive and thrive: roles of BAFF in B-cell development. Curr Opin Immunol. 2002;14:266–75.CrossRefPubMed Rolink AG, Melchers F. BAFFled B cells survive and thrive: roles of BAFF in B-cell development. Curr Opin Immunol. 2002;14:266–75.CrossRefPubMed
40.
go back to reference Rolink AG, Tschopp J, Schneider P, Melchers F. BAFF is a survival and maturation factor for mouse B cells. Eur J Immunol. 2002;32:2004–10.CrossRefPubMed Rolink AG, Tschopp J, Schneider P, Melchers F. BAFF is a survival and maturation factor for mouse B cells. Eur J Immunol. 2002;32:2004–10.CrossRefPubMed
41.
go back to reference Belnoue E, Pihlgren M, McGaha TL, Tougne C, Rochat AF, Bossen C, Schneider P, Huard B, Lambert PH, Siegrist CA. APRIL is critical for plasmablast survival in the bone marrow and poorly expressed by early-life bone marrow stromal cells. Blood. 2008;111:2755–64.CrossRefPubMed Belnoue E, Pihlgren M, McGaha TL, Tougne C, Rochat AF, Bossen C, Schneider P, Huard B, Lambert PH, Siegrist CA. APRIL is critical for plasmablast survival in the bone marrow and poorly expressed by early-life bone marrow stromal cells. Blood. 2008;111:2755–64.CrossRefPubMed
42.
go back to reference Benson MJ, Dillon SR, Castigli E, Geha RS, Xu S, Lam KP, Noelle RJ. Cutting edge: the dependence of plasma cells and independence of memory B cells on BAFF and APRIL. J Immunol. 2008;180:3655–9.CrossRefPubMed Benson MJ, Dillon SR, Castigli E, Geha RS, Xu S, Lam KP, Noelle RJ. Cutting edge: the dependence of plasma cells and independence of memory B cells on BAFF and APRIL. J Immunol. 2008;180:3655–9.CrossRefPubMed
43.
go back to reference Magliozzi R, Columba-Cabezas S, Serafini B, Aloisi F. Intracerebral expression of CXCL13 and BAFF is accompanied by formation of lymphoid follicle-like structures in the meninges of mice with relapsing experimental autoimmune encephalomyelitis. J Neuroimmunol. 2004;148:11–23.CrossRefPubMed Magliozzi R, Columba-Cabezas S, Serafini B, Aloisi F. Intracerebral expression of CXCL13 and BAFF is accompanied by formation of lymphoid follicle-like structures in the meninges of mice with relapsing experimental autoimmune encephalomyelitis. J Neuroimmunol. 2004;148:11–23.CrossRefPubMed
44.
go back to reference Serafini B, Severa M, Columba-Cabezas S, Rosicarelli B, Veroni C, Chiappetta G, Magliozzi R, Reynolds R, Coccia EM, Aloisi F. Epstein-Barr virus latent infection and BAFF expression in B cells in the multiple sclerosis brain: implications for viral persistence and intrathecal B-cell activation. J Neuropathol Exp Neurol. 2010;69:677–93.CrossRefPubMed Serafini B, Severa M, Columba-Cabezas S, Rosicarelli B, Veroni C, Chiappetta G, Magliozzi R, Reynolds R, Coccia EM, Aloisi F. Epstein-Barr virus latent infection and BAFF expression in B cells in the multiple sclerosis brain: implications for viral persistence and intrathecal B-cell activation. J Neuropathol Exp Neurol. 2010;69:677–93.CrossRefPubMed
45.
go back to reference Marsters SA, Yan M, Pitti RM, Haas PE, Dixit VM, Ashkenazi A. Interaction of the TNF homologues BLyS and APRIL with the TNF receptor homologues BCMA and TACI. Curr Biol. 2000;10:785–8.CrossRefPubMed Marsters SA, Yan M, Pitti RM, Haas PE, Dixit VM, Ashkenazi A. Interaction of the TNF homologues BLyS and APRIL with the TNF receptor homologues BCMA and TACI. Curr Biol. 2000;10:785–8.CrossRefPubMed
46.
go back to reference Yan M, Marsters SA, Grewal IS, Wang H, Ashkenazi A, Dixit VM. Identification of a receptor for BLyS demonstrates a crucial role in humoral immunity. Nat Immunol. 2000;1:37–41.CrossRefPubMed Yan M, Marsters SA, Grewal IS, Wang H, Ashkenazi A, Dixit VM. Identification of a receptor for BLyS demonstrates a crucial role in humoral immunity. Nat Immunol. 2000;1:37–41.CrossRefPubMed
47.
go back to reference Thompson JS, Schneider P, Kalled SL, Wang L, Lefevre EA, Cachero TG, MacKay F, Bixler SA, Zafari M, Liu ZY, et al. BAFF binds to the tumor necrosis factor receptor-like molecule B cell maturation antigen and is important for maintaining the peripheral B cell population. J Exp Med. 2000;192:129–35.CrossRefPubMedPubMedCentral Thompson JS, Schneider P, Kalled SL, Wang L, Lefevre EA, Cachero TG, MacKay F, Bixler SA, Zafari M, Liu ZY, et al. BAFF binds to the tumor necrosis factor receptor-like molecule B cell maturation antigen and is important for maintaining the peripheral B cell population. J Exp Med. 2000;192:129–35.CrossRefPubMedPubMedCentral
Metadata
Title
Glial cell activation, recruitment, and survival of B-lineage cells following MCMV brain infection
Authors
James R. Lokensgard
Manohar B. Mutnal
Sujata Prasad
Wen Sheng
Shuxian Hu
Publication date
01-12-2016
Publisher
BioMed Central
Published in
Journal of Neuroinflammation / Issue 1/2016
Electronic ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-016-0582-y

Other articles of this Issue 1/2016

Journal of Neuroinflammation 1/2016 Go to the issue