The central nervous system (CNS) is a major target for acute encephalitic viral infections, as well as a reservoir of latent/persisting viruses. Whileeffective immune control of persisting viruses in immunocompetent individuals is reflected by the absence of overt neurological deficits or pathology, this balance is highly tenuous. Studies supported by this award were the first to demonstrate that intrathecal antibody secreting cells (ASC) were critical to control CNS viral persistence even despite early T cell mediated control and presence of anti-viral serum antibody (Ab). We have also defined the regulation of ACS recruitment into the CNS by identifying the crucial chemokine receptor and chemokine. The reliance on local ASC for prolonged Ab output during neurotropic coronavirus infection is a critical observation, as it provides a potent non lytic mechanism of sustained immune control applicable to numerous neurotropic infections. Indeed, a vital local protective role of ASC is supported by other experimental CNS infections, particularly by RNA viruses such as Sindbis, Semliki Forest, and Rabies viruses. Intrathecal Ab synthesis is also well documented in humans during infections associated with neurological complications and the demyelinating disease multiple sclerosis (MS). However, virtually nothing is known about the origin, maintenance, and relevance of ASC in the CNS or other specialized microenvironments. The overall goal is to define the regulation of protective, yet minimally destructive ASC within the CNS, to combat infections where virus replicates in the CNS in the absence of overt blood brain barrier damage or inflammatory signals.
The Specific Aims are to 1) determine the role of CNS resident cells in mediating ASC entry and localization within the CNS; and 2) determine the relative contribution of migratory ASC versus circulating, non Ab secreting memory B cells in maintaining ASC within the CNS. In situ hybridization combined with confocal microscopy in Aim 1 will determine the spatial relationship between chemokines, viral antigen, ASC localization and the microvasculature. The results will further reveal a central role of astrocytes in regulating ASC recruitment.
Aim 2 uses transgenic mice in which germinal center derived ASC and memory B cells are phenotypically marked, to specifically test the role of virus specific B cells in contributing to ASC maintenance during chronic CNS infection. Adoptive transfers will define whether the initial burst of ASC in the CNS is sufficient to sustain local protective capacity. Non of these parameters have been studied during viral or autoimmune encephalomyelitis. The ability to trace ASC and Bmem using transgenic markers provides a versatile innovative approach to dissect humoral immunity in a non-lymphoid organ prone to persisting infection. The contribution of peripherally activated B cells rather than lymphoid tissue neogenesis, in sustaining intrathecal humoral responses during persistent infections associated with limited ongoing inflammation will be beneficial for understanding protective responses during human CNS infections caused by measles virus, rubella virus, JC virus, and HIV.

Public Health Relevance

The central nervous system (CNS) is a major target for numerous acute encephalitic viral infections, as well as a reservoir of latent/persisting viruses. Antibody secreting cells (ASC) in the CNS provide an effective protective component in controlling persistent CNS infections. The overall goals of this proposal are to determine how resident CNS cells contribute to ASC entry into the CNS and to determine the relative contribution of distinct B cell subsets to long term ASC survival and antiviral function. Insights into the precursor B cells giving rise to ASC in the CNS are essential for vaccination and therapeutic treatment of viral encephalitis and chronic inflammation.

Agency
National Institute of Health (NIH)
Institute
National Institute of Allergy and Infectious Diseases (NIAID)
Type
High Priority, Short Term Project Award (R56)
Project #
2R56AI047249-11
Application #
8507825
Study Section
Special Emphasis Panel (ZRG1-IMM-G (03))
Program Officer
Park, Eun-Chung
Project Start
2012-08-01
Project End
2013-08-31
Budget Start
2012-08-01
Budget End
2013-08-31
Support Year
11
Fiscal Year
2012
Total Cost
$392,500
Indirect Cost
$142,500
Name
Cleveland Clinic Lerner
Department
Other Basic Sciences
Type
Schools of Medicine
DUNS #
135781701
City
Cleveland
State
OH
Country
United States
Zip Code
44195
Phares, Timothy W; DiSano, Krista D; Stohlman, Stephen A et al. (2014) Progression from IgD+ IgM+ to isotype-switched B cells is site specific during coronavirus-induced encephalomyelitis. J Virol 88:8853-67
Phares, Timothy W; DiSano, Krista D; Hinton, David R et al. (2013) IL-21 optimizes T cell and humoral responses in the central nervous system during viral encephalitis. J Neuroimmunol 263:43-54
Phares, Timothy W; Stohlman, Stephen A; Hinton, David R et al. (2013) Astrocyte-derived CXCL10 drives accumulation of antibody-secreting cells in the central nervous system during viral encephalomyelitis. J Virol 87:3382-92
Phares, Timothy W; Stohlman, Stephen A; Bergmann, Cornelia C (2013) Intrathecal humoral immunity to encephalitic RNA viruses. Viruses 5:732-52
Phares, Timothy W; Stohlman, Stephen A; Hwang, Mihyun et al. (2012) CD4 T cells promote CD8 T cell immunity at the priming and effector site during viral encephalitis. J Virol 86:2416-27
Phares, Timothy W; Stohlman, Stephen A; Hinton, David R et al. (2012) Enhanced CD8 T-cell anti-viral function and clinical disease in B7-H1-deficient mice requires CD4 T cells during encephalomyelitis. J Neuroinflammation 9:269
Marques, Cristina P; Kapil, Parul; Hinton, David R et al. (2011) CXCR3-dependent plasma blast migration to the central nervous system during viral encephalomyelitis. J Virol 85:6136-47
Parra, Gabriel I; Bergmann, Cornelia C; Phares, Timothy W et al. (2010) Gamma interferon signaling in oligodendrocytes is critical for protection from neurotropic coronavirus infection. J Virol 84:3111-5
Phares, Timothy W; Stohlman, Stephen A; Hinton, David R et al. (2010) Enhanced antiviral T cell function in the absence of B7-H1 is insufficient to prevent persistence but exacerbates axonal bystander damage during viral encephalomyelitis. J Immunol 185:5607-18
Phares, Timothy W; Ramakrishna, Chandran; Parra, Gabriel I et al. (2009) Target-dependent B7-H1 regulation contributes to clearance of central nervous system infection and dampens morbidity. J Immunol 182:5430-8

Showing the most recent 10 out of 11 publications