Immune responses to RNA-virus infections of the CNS

Diane E Griffin, Diane E Griffin

Abstract

A successful outcome for the host of virus infection of the central nervous system (CNS) requires the elimination of the virus without damage to essential non-renewable cells, such as neurons. As a result, inflammatory responses must be tightly controlled, and many unique mechanisms seem to contribute to this control. In addition to being important causes of human disease, RNA viruses that infect the CNS provide useful models in which to study immune responses in the CNS. Recent work has shown the importance of innate immune responses in the CNS in controlling virus infection. And advances have been made in assessing the relative roles of cytotoxic T cells, antibodies and cytokines in the clearance of viruses from neurons, glial cells and meningeal cells.

Figures

Figure 1. Changes that occur in the…
Figure 1. Changes that occur in the CNS during the immune response to a neuronal virus infection.
a | In the normal uninfected central nervous system (CNS), endothelial cells express few adhesion molecules. Neurons maintain microglia in a quiescent state through CD200–CD200 receptor (CD200R) interactions and astrocytes contribute by the production of neurotrophins, such as brain-derived neurotrophic factor (BDNF), nerve-growth factor (NGF) and neurotrophin 3 (NT3), that interact with p75, a low-affinity neurotrophin receptor. Gangliosides are abundant and transforming growth factor-β (TGF-β) is constitutively produced by neurons and astrocytes. Activated T cells enter the CNS routinely through interaction with low levels of P-selectin expressed by endothelial cells, but leave or die soon thereafter. b | Virus infection of neurons initiates the early production of interferon-β (IFN-β), chemokines and pro-inflammatory cytokines, which results in further activation of microglia and increased expression of adhesion molecules, such as vascular-cell adhesion molecule 1 (VCAM1) and intercellular adhesion molecule 1 (ICAM1), by endothelial cells. c | By three to four days after infection, inflammatory cells — such as natural killer (NK) cells, macrophages and lymphocytes — that are activated in secondary lymphoid tissue begin to enter the CNS at regions where virus replication is occurring and chemokines, such as CCL19 and CCL21, are expressed by endothelial cells. T cells produce additional cytokines, such as interleukin-4 (IL-4) and IFN-γ, and the B cells produce antibody, which initiates clearance of virus from infected cells. TNF, tumour-necrosis factor.
Figure 2. Mechanisms of virus clearance from…
Figure 2. Mechanisms of virus clearance from neurons and microglia.
a | In neurons that survive infection with RNA viruses, infection is controlled initially by interferon-β (IFN-β) and subsequently by virus-specific antibody and IFN-γ produced by T cells and possibly neurons. This effectively inhibits the production of new virus, but does not necessarily eliminate virus RNA from the cell. CD4+ T cells that secrete cytokines, such as IFN-γ, and antibody-secreting B cells are resident for prolonged periods of time in the nervous system and probably have a role in long-term control of virus replication. b | Initial control of virus replication in infected glial cells involves IFN-β. Clearance involves cytolytic CD8+ T cells. CD4+ T cells supply help for CD8+ T cells and both subsets of T cells produce IFN-γ, which has an independent role in virus clearance from cells that survive. Long-term control of virus replication requires the local production of virus-specific antibody and possibly T cells.

References

    1. Ehrengruber MU, Ehler E, Billeter MA, Naim HY. Measles virus spreads in rat hippocampal neurons by cell-to-cell contact and in a polarized fashion. J. Virol. 2002;76:5720–5728.
    1. Stevenson PG, Freeman S, Bangham CR, Hawke S. Virus dissemination through the brain parenchyma without immunologic control. J. Immunol. 1997;159:1876–1884.
    1. Hoek RM, et al. Down-regulation of the macrophage lineage through interaction with OX2 (CD200) Science. 2000;290:1768–1771.
    1. Neumann H, Misgeld T, Matsumuro K, Wekerle H. Neurotrophins inhibit major histocompatibility class II inducibility of microglia: involvement of the p75 neurotrophin receptor. Proc. Natl Acad. Sci. USA. 1998;95:5779–5784.
    1. Johnson MD, Gold LI, Moses HL. Evidence for transforming growth factor-β expression in human leptomeningeal cells and transforming growth factor-β-like activity in human cerebrospinal fluid. Lab. Invest. 1992;67:360–368.
    1. Fabry Z, et al. TGF-β2 decreases migration of lymphocytes in vitro and homing of cells into the central nervous system in vivo. J. Immunol. 1995;155:325–332.
    1. Muller U, et al. Functional role of type I and type II interferons in antiviral defense. Science. 1994;264:1918–1921.
    1. Fiette L, et al. Theilers virus infection of 129SV mice that lack the interferon-α/β or interferon-γ receptors. J. Exp. Med. 1995;181:2069–2076.
    1. Ryman K, Klimstra W, Nguyen K, Biron C, Johnston R. α/β interferon protects adult mice from fatal Sindbis virus infection and is an important determinant of cell and tissue tropism. J. Virol. 2000;74:3366–3378.
    1. Byrnes AP, Durbin JE, Griffin DE. Control of Sindbis virus infection by antibody in interferon-deficient mice. J. Virol. 2000;74:3905–3908.
    1. Sandberg K, Eloranta ML, Campbell IL. Expression of α/β interferons (IFN-α/β) and their relationship to IFN-α/β-induced genes in lymphocytic choriomeningitis. J. Virol. 1994;68:7358–7366.
    1. Erlandsson L, et al. Interferon-β is required for interferon-α production in mouse fibroblasts. Curr. Biol. 1998;8:223–226.
    1. Akwa Y, et al. Transgenic expression of IFN-α in the central nervous system of mice protects against lethal neurotropic viral infection but induces inflammation and neurodegeneration. J. Immunol. 1998;161:5016–5026.
    1. McLaurin J, Antel JP, Yong VW. Immune and non-immune actions of interferon-β-1b on primary human neural cells. Multiple Sclerosis. 1995;1:10–19.
    1. Boutros T, Croze E, Yong VW. Interferon-β is a potent promoter of nerve growth factor production by astrocytes. J. Neurochem. 1997;69:939–946.
    1. Chabot S, Yong VW. Interferon-β-1b increases interleukin-10 in a model of T cell–microglia interaction: relevance to MS. Neurology. 2000;55:1497–1505.
    1. van Pesch V, van Eyll O, Michiels T. The leader protein of Theiler's virus inhibits immediate-early α/β interferon production. J. Virol. 2001;75:7811–7817.
    1. Neumann H. Control of glial immune function by neurons. Glia. 2001;36:191–199.
    1. Neumann H, Schmidt H, Wilharm E, Behrens L, Wekerle H. Interferon γ gene expression in sensory neurons: evidence for autocrine gene regulation. J. Exp. Med. 1997;186:2023–2031.
    1. Harrison JK, et al. Role for neuronally derived fractalkine in mediating interactions between neurons and CX3CR1-expressing microglia. Proc. Natl Acad. Sci. USA. 1998;95:10896–10901.
    1. Rappert A, et al. Secondary lymphoid tissue chemokine (CCL21) activates CXCR3 to trigger a Cl(-) current and chemotaxis in murine microglia. J. Immunol. 2002;168:3221–3226.
    1. Chapman GA, et al. Fractalkine cleavage from neuronal membranes represents an acute event in the inflammatory response to excitotoxic brain damage. J. Neurosci. 2000;20:1–5.
    1. Jung S, et al. Analysis of fractalkine receptor CX3CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol. Cell Biol. 2000;20:4106–4114.
    1. Maciejewski-Lenoir D, Chen S, Feng L, Maki R, Bacon KB. Characterization of fractalkine in rat brain cells: migratory and activation signals for CX3CR-1-expressing microglia. J. Immunol. 1999;163:1628–1635.
    1. Asensio VC, Campbell IL. Chemokine gene expression in the brains of mice with lymphocytic choriomeningitis. J. Virol. 1997;71:7832–7840.
    1. Lane TE, et al. Dynamic regulation of α- and β-chemokine expression in the central nervous system during mouse hepatitis virus-induced demyelinating disease. J. Immunol. 1998;160:970–978.
    1. Wesselingh SL, Levine B, Fox RJ, Choi S, Griffin DE. Intracerebral cytokine mRNA expression during fatal and nonfatal alphavirus encephalitis suggests a predominant type 2 T cell response. J. Immunol. 1994;152:1289–1297.
    1. Parra B, Hinton DR, Lin MT, Cua DJ, Stohlman SA. Kinetics of cytokine mRNA expression in the central nervous system following lethal and nonlethal coronavirus-induced acute encephalomyelitis. Virology. 1997;233:260–270.
    1. Chang JR, Zaczynska E, Katsetos CD, Platsoucas CD, Oleszak EL. Differential expression of TGF-β, IL-2, and other cytokines in the CNS of Theiler's murine encephalomyelitis virus-infected susceptible and resistant strains of mice. Virology. 2000;278:346–360.
    1. Galelli A, Baloul L, Lafon M. Abortive rabies virus central nervous infection is controlled by T lymphocyte local recruitment and induction of apoptosis. J. Neurovirol. 2000;6:359–372.
    1. Middleton J, Patterson AM, Gardner L, Schmutz C, Ashton BA. Leukocyte extravasation: chemokine transport and presentation by the endothelium. Blood. 2002;100:3853–3860.
    1. Harling-Berg CJ, Park TJ, Knopf PM. Role of the cervical lymphatics in the TH2-type hierarchy of CNS immune regulation. J. Neuroimmunol. 1999;101:111–127.
    1. Fischer HG, Reichmann G. Brain dendritic cells and macrophages/microglia in central nervous system inflammation. J. Immunol. 2001;166:2717–2726.
    1. Pope JG, Vanderlugt CL, Rahbe SM, Lipton HL, Miller SD. Characterization of and functional antigen presentation by central nervous system mononuclear cells from mice infected with Theiler's murine encephalomyelitis virus. J. Virol. 1998;72:7762–7771.
    1. McMenamin PG. Distribution and phenotype of dendritic cells and resident tissue macrophages in the dura mater, leptomeninges, and choroid plexus of the rat brain as demonstrated in wholemount preparations. J. Comp. Neurol. 1999;405:553–562.
    1. Cserr HF, Knopf PM. Cervical lymphatics, the blood–brain barrier and the immunoreactivity of the brain: a new view. Immunol. Today. 1992;13:507–512.
    1. Stevenson PG, Hawke S, Sloan DJ, Bangham CR. The immunogenicity of intracerebral virus infection depends on anatomical site. J. Virol. 1997;71:145–151.
    1. Carrithers MD, Visintin I, Kang SJ, Janeway CA., Jr. Differential adhesion molecule requirements for immune surveillance and inflammatory recruitment. Brain. 2000;123:1092–1101.
    1. Irani DN, Griffin DE. Regulation of lymphocyte homing into the brain during viral encephalitis at various states of infection. J. Immunol. 1996;156:3850–3857.
    1. Wekerle H, Linington C, Lassmann H, Meyermann R. Cellular immune reactivity within the CNS. Trends Neurosci. 1986;9:271–277.
    1. Hickey WF, Hsu BL, Kimura H. T-lymphocyte entry into the central nervous system. J. Neurosci. Res. 1991;28:254–260.
    1. Piccio L, et al. Molecular mechanisms involved in lymphocyte recruitment in inflamed brain microvessels: critical roles for P-selectin glycoprotein ligand-1 and heterotrimeric G(i)-linked receptors. J. Immunol. 2002;168:1940–1949.
    1. Hickey WF. Basic principles of immunological surveillance of the normal central nervous system. Glia. 2001;36:118–124.
    1. Licinio J, Wong ML. Pathways and mechanisms for cytokine signaling of the central nervous system. J. Clin. Invest. 1997;100:2941–2947.
    1. Alt C, Laschinger M, Engelhardt B. Functional expression of the lymphoid chemokines CCL19 (ELC) and CCL21 (SLC) at the blood–brain barrier suggests their involvement in G-protein-dependent lymphocyte recruitment into the central nervous system during experimental autoimmune encephalomyelitis. Eur. J. Immunol. 2002;32:2133–2144.
    1. Kimura T, Griffin DE. The role of CD8+ T cells and major histocompatibility complex class I expression in the central nervous system of mice infected with neurovirulent Sindbis virus. J. Virol. 2000;74:6117–6125.
    1. Suzumura A, et al. Induction of glial cell MHC antigen expression in neurotropic coronavirus infections. Characterization of the H-2-inducing soluble factor elaborated by infected brain cells. J. Immunol. 1988;140:2068–2072.
    1. Massa PT, Whitney LW, Wu C, Ropka SL, Jarosinski KW. A mechanism for selective induction of 2′–5′ oligoadenylate synthetase, anti-viral state, but not MHC class I genes by interferon-β in neurons. J. Neurovirol. 1999;5:161–171.
    1. Jarosinski KW, Whitney LW, Massa PT. Specific deficiency in nuclear factor-κB activation in neurons of the central nervous system. Lab. Invest. 2001;81:1275–1288.
    1. Neumann H, Schmidt H, Cavalie A, Jenne D, Wekerle H. Major histocompatibility complex (MHC) class I gene expression in single neurons of the central nervous system: differential regulation by interferon (IFN)-γ and tumor necrosis factor (TNF)-α. J. Exp. Med. 1997;185:305–316.
    1. Pereira RA, Simmons A. Cell surface expression of H2 antigens on primary sensory neurons in response to acute but not latent herpes simplex virus infection in vivo. J. Virol. 1999;73:6484–6489.
    1. Sedgwick JD, Ford AL, Foulcher E, Airriess R. Central nervous system microglial cell activation and proliferation follows direct interaction with tissue-infiltrating T cell blasts. J. Immunol. 1998;160:5320–5330.
    1. Irani DN, Griffin DE. Isolation of brain parenchymal lymphocytes for flow cytometric analysis. Application to acute viral encephalitis. J. Immunol. Methods. 1991;139:223–231.
    1. Hatalski CG, Hickey WF, Lipkin WI. Evolution of the immune response in the central nervous system following infection with Borna disease virus. J. Neuroimmunol. 1998;90:137–142.
    1. Haring JS, Pewe LL, Perlman S. High-magnitude, virus-specific CD4 T-cell response in the central nervous system of coronavirus-infected mice. J. Virol. 2001;75:3043–3047.
    1. Stohlman SA, Bergmann CC, Lin MT, Cua DJ, Hinton DR. CTL effector function within the central nervous system requires CD4+ T cells. J. Immunol. 1998;160:2896–2904.
    1. Tschen SI, et al. Recruitment kinetics and composition of antibody-secreting cells within the central nervous system following viral encephalomyelitis. J. Immunol. 2002;168:2922–2929.
    1. Dorries R. The role of T-cell-mediated mechanisms in virus infections of the nervous system. Curr. Top. Microbiol. Immunol. 2001;253:219–245.
    1. Pashenkov M, Link H. Dendritic cells and immune responses in the central nervous system. Trends Immunol. 2002;23:69–70.
    1. Marten NW, Stohlman SA, Bergmann CC. Role of viral persistence in retaining CD8+ T cells within the central nervous system. J. Virol. 2000;74:7903–7910.
    1. Pearce BD, Hobbs MV, McGraw TS, Buchmeier MJ. Cytokine induction during T-cell-mediated clearance of mouse hepatitis virus from neurons in vivo. J. Virol. 1994;68:5483–5495.
    1. Rowell JF, Griffin DE. The inflammatory response to nonfatal Sindbis virus infection of the nervous system is more severe in SJL than in BALB/c mice and is associated with low levels of IL-4 mRNA and high levels of IL-10-producing CD4+ T cells. J. Immunol. 1999;162:1624–1632.
    1. Griffin DE. Immunoglobulins in the cerebrospinal fluid: changes during acute viral encephalitis in mice. J. Immunol. 1981;126:27–31.
    1. Irani DN, Lin K-I, Griffin DE. Brain-derived gangliosides regulate the cytokine production and proliferation of activated T cells. J. Immunol. 1996;157:4333–4340.
    1. Irani DN, Lin K-I, Griffin DE. Regulation of brain-derived T cells during acute central nervous system inflammation. J. Immunol. 1997;158:2318–2326.
    1. Massa PT. Specific suppression of major histocompatibility complex class I and class II genes in astrocytes by brain-enriched gangliosides. J. Exp. Med. 1993;178:1357–1363.
    1. Irani DN. The susceptibility of mice to immune-mediated neurologic disease correlates with the degree to which their lymphocytes resist the effects of brain-derived gangliosides. J. Immunol. 1998;161:2746–2752.
    1. Tontsch U, Rott O. Cortical neurons selectively inhibit MHC class II induction in astrocytes but not in microglial cells. Int. Immunol. 1993;5:249–254.
    1. Ford AL, Foulcher E, Lemckert FA, Sedgwick JD. Microglia induce CD4 T lymphocyte final effector function and death. J. Exp. Med. 1996;184:1737–1745.
    1. Gordon LB, Nolan SC, Ksander BR, Knopf PM, Harling-Berg CJ. Normal cerebrospinal fluid suppresses the in vitro development of cytotoxic T cells: role of the brain microenvironment in CNS immune regulation. J. Neuroimmunol. 1998;88:77–84.
    1. Hailer NP, Heppner FL, Haas D, Nitsch R. Astrocytic factors deactivate antigen presenting cells that invade the central nervous system. Brain Pathol. 1998;8:459–474.
    1. Bauer J, et al. T-cell apoptosis in inflammatory brain lesions: destruction of T cells does not depend on antigen recognition. Am. J. Pathol. 1998;153:715–724.
    1. Gold R, et al. Antigen presentation by astrocytes primes rat T lymphocytes for apoptotic cell death. A model for T-cell apoptosis in vivo. Brain. 1996;119:651–659.
    1. Medana I, et al. Fas ligand (CD95L) protects neurons against perforin-mediated T lymphocyte cytotoxicity. J. Immunol. 2001;167:674–681.
    1. Flugel A, et al. Neuronal FasL induces cell death of encephalitogenic T lymphocytes. Brain Pathol. 2000;10:353–364.
    1. Shin DH, et al. Fas ligand mRNA expression in the mouse central nervous system. J. Neuroimmunol. 2002;123:50–57.
    1. Hawke S, Stevenson PG, Freeman S, Bangham CR. Long-term persistence of activated cytotoxic T lymphocytes after viral infection of the central nervous system. J. Exp. Med. 1998;187:1575–1582.
    1. Fujinami RS, Rosenthal A, Lampert PW, Zurbriggen A, Yamada M. Survival of athymic (nu/nu) mice after Theiler's murine encephalomyelitis virus infection by passive administration of neutralizing monoclonal antibody. J. Virol. 1989;63:2081–2087.
    1. Fleming JO, Shubin RA, Sussman MA, Casteel N, Stohlman SA. Monoclonal antibodies to the matrix (E1) glycoprotein of mouse hepatitis virus protect mice from encephalitis. Virology. 1989;168:162–167.
    1. Levine B, et al. Antibody-mediated clearance of alphavirus infection from neurons. Science. 1991;254:856–860.
    1. Perry LL, Lodmell DL. Role of CD4+ and CD8+ T cells in murine resistance to street rabies virus. J. Virol. 1991;65:3429–3434.
    1. Tyor WR, Wesselingh S, Levine B, Griffin DE. Long term intraparenchymal Ig secretion after acute viral encephalitis in mice. J. Immunol. 1992;149:4016–4020.
    1. Hatalski CG, Hickey WF, Lipkin WI. Humoral immunity in the central nervous system of Lewis rats infected with Borna disease virus. J. Neuroimmunol. 1998;90:128–136.
    1. Ubol S, Levine B, Lee S-H, Greenspan NS, Griffin DE. Roles of immunoglobulin valency and the heavy-chain constant domain in antibody-mediated downregulation of Sindbis virus replication in persistently infected neurons. J. Virol. 1995;69:1990–1993.
    1. Despres P, Griffin JW, Griffin DE. Effects of anti-E2 monoclonal antibody on Sindbis virus replication in AT3 cells expressing Bcl-2. J. Virol. 1995;69:7006–7014.
    1. Despres P, Griffin JW, Griffin DE. Antiviral activity of α-interferon in Sindbis virus-infected cells is restored by anti-E2 monoclonal antibody treatment. J. Virol. 1995;69:7345–7348.
    1. Hooper DC, et al. Collaboration of antibody and inflammation in clearance of rabies virus from the central nervous system. J. Virol. 1998;72:3711–3719.
    1. Destombes J, et al. Persistent poliovirus infection in mouse motoneurons. J. Virol. 1997;71:1621–1628.
    1. Liu T, Khanna KM, Carriere BN, Hendricks RL. γ-interferon can prevent herpes simplex virus type 1 reactivation from latency in sensory neurons. J. Virol. 2001;75:11178–11184.
    1. Neumann H, Cavalie A, Jenne DE, Wekerle H. Induction of MHC class 1 genes in neurons. Science. 1995;269:549–552.
    1. Medana IM, et al. MHC class I-restricted killing of neurons by virus-specific CD8+ T lymphocytes is effected through the Fas–FasL, but not the perforin pathway. Eur. J. Immunol. 2000;30:3623–3633.
    1. Bilzer T, Stitz L. Immune-mediated brain atrophy. CD8+ T cells contribute to tissue destruction during borna disease. J. Immunol. 1994;153:818–823.
    1. Patterson CE, Lawrence DM, Echols LA, Rall GF. Immune-mediated protection from measles virus-induced central nervous system disease is noncytolytic and γ-interferon dependent. J. Virol. 2002;76:4497–4506.
    1. Binder G, Griffin D. Interferon-γ-mediated site specific clearance of alphavirus from CNS neurons. Science. 2001;293:303–306.
    1. Komatsu T, Ireland DD, Chen N, Reiss CS. Neuronal expression of NOS-1 is required for host recovery from viral encephalitis. Virology. 1999;258:389–395.
    1. Oldstone MB, Blount P, Southern PJ, Lampert PW. Cytoimmunotherapy for persistent virus infection reveals a unique clearance pattern from the central nervous system. Nature. 1986;321:239–243.
    1. Pereira RA, Simon MM, Simmons A. Granzyme A, a noncytolytic component of CD8+ cell granules, restricts the spread of herpes simplex virus in the peripheral nervous systems of experimentally infected mice. J. Virol. 2000;74:1029–1032.
    1. Lin MT, Hinton DR, Stohlman SA. Mechanisms of viral clearance in perforin-deficient mice. Adv. Exp. Med. Biol. 1998;440:431–436.
    1. Robertson B, Kong G, Peng Z, Bentivoglio M, Kristensson K. Interferon-γ-responsive neuronal sites in the normal rat brain: receptor protein distribution and cell activation revealed by Fos induction. Brain Res. Bull. 2000;52:61–74.
    1. Eneroth A, et al. Interferon-γ-like immunoreactivity in sensory neurons may influence the replication of Sendai and mumps viruses. J. Neurosci. Res. 1992;31:487–493.
    1. Lindsley MD, Rodriguez M. Characterization of the inflammatory response in the central nervous system of mice susceptible or resistant to demyelination by Theiler's virus. J. Immunol. 1989;142:2677–2682.
    1. Korner H, et al. Nucleocapsid or spike protein-specific CD4+ T lymphocytes protect against coronavirus-induced encephalomyelitis in the absence of CD8+ T cells. J. Immunol. 1991;147:2317–2323.
    1. Lin MT, Stohlman SA, Hinton DR. Mouse hepatitis virus is cleared from the central nervous systems of mice lacking perforin-mediated cytolysis. J. Virol. 1997;71:383–391.
    1. Yamaguchi K, Goto N, Kyuwa S, Hayami M, Toyoda Y. Protection of mice from a lethal coronavirus infection in the central nervous system by adoptive transfer of virus-specific T cell clones. J. Neuroimmunol. 1991;32:1–9.
    1. Stohlman SA, Bergmann CC, Van der Veen RC, Hinton DR. Mouse hepatits virus-specific cytotoxic T lymphocytes protect from lethal infection without eliminating virus from the central nervous system. J. Virol. 1995;69:684–694.
    1. Parra B, et al. IFN-γ is required for viral clearance from central nervous system oligodendroglia. J. Immunol. 1999;162:1641–1647.
    1. Parra B, et al. Contributions of Fas–Fas ligand interactions to the pathogenesis of mouse hepatitis virus in the central nervous system. J. Virol. 2000;74:2447–2450.
    1. Marten NW, et al. Contributions of CD8+ T cells and viral spread to demyelinating disease. J. Immunol. 2000;164:4080–4088.
    1. Marten NW, et al. Selection of CD8+ T cells with highly focused specificity during viral persistence in the central nervous system. J. Immunol. 1999;162:3905–3914.
    1. Bergmann CC, Altman JD, Hinton D, Stohlman SA. Inverted immunodominance and impaired cytolytic function of CD8+ T cells during viral persistence in the central nervous system. J. Immunol. 1999;163:3379–3387.
    1. Ramakrishna C, Stohlman SA, Atkinson RD, Shlomchik MJ, Bergmann CC. Mechanisms of central nervous system viral persistence: the critical role of antibody and B cells. J. Immunol. 2002;168:1204–1211.
    1. Morales S, Parra B, Ramakrishna C, Blau DM, Stohlman SA. B-cell-mediated lysis of cells infected with the neurotropic JHM strain of mouse hepatitis virus. Virology. 2001;286:160–167.

Source: PubMed

3
Prenumerera