Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Front Immunol ; 12: 676686, 2021.
Article in English | MEDLINE | ID: mdl-34168647

ABSTRACT

Multiple Sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Once thought to be primarily driven by T cells, B cells are emerging as central players in MS immunopathogenesis. Interest in multiple B cell phenotypes in MS expanded following the efficacy of B cell-depleting agents targeting CD20 in relapsing-remitting MS and inflammatory primary progressive MS patients. Interestingly, these therapies primarily target non-antibody secreting cells. Emerging studies seek to explore B cell functions beyond antibody-mediated roles, including cytokine production, antigen presentation, and ectopic follicle-like aggregate formation. Importantly, memory B cells (Bmem) are rising as a key B cell phenotype to investigate in MS due to their antigen-experience, increased lifespan, and rapid response to stimulation. Bmem display diverse effector functions including cytokine production, antigen presentation, and serving as antigen-experienced precursors to antibody-secreting cells. In this review, we explore the cellular and molecular processes involved in Bmem development, Bmem phenotypes, and effector functions. We then examine how these concepts may be applied to the potential role(s) of Bmem in MS pathogenesis. We investigate Bmem both within the periphery and inside the CNS compartment, focusing on Bmem phenotypes and proposed functions in MS and its animal models. Finally, we review how current immunomodulatory therapies, including B cell-directed therapies and other immunomodulatory therapies, modify Bmem and how this knowledge may be harnessed to direct therapeutic strategies in MS.


Subject(s)
Antibodies, Monoclonal, Humanized/biosynthesis , Antigen Presentation , B-Lymphocytes/immunology , Cytokines/biosynthesis , Immunologic Memory , Multiple Sclerosis, Relapsing-Remitting/immunology , Animals , Antibodies, Monoclonal, Humanized/therapeutic use , Antigens, CD20/immunology , Central Nervous System/immunology , Disease Models, Animal , Humans , Immunologic Factors/therapeutic use , Immunomodulation , Inflammation/immunology , Multiple Sclerosis, Relapsing-Remitting/therapy , Phenotype
2.
Fluids Barriers CNS ; 18(1): 9, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33632258

ABSTRACT

Pilz et al. (Fluids Barriers CNS 17:7; 2020) investigated how CSF CXCL13 concentrations are influenced by CXCL13 serum concentrations and blood-CSF barrier (BCSFB) function, comparing the impact of serum CXCL13 levels and Qalbumin (CSF albumin/serum albumin) on CSF CXCL13 among patients with CNS inflammation categorized as CXCL13 negative, low, medium, or high. Among all CXCL13 groups, their results showed no correlation between CSF CXCL13 concentrations and serum CXCL13 or Qalbumin. The authors argue that, in contrast to other proteins, CXCL13 passage across the BCSFB does not occur, regardless of BCSFB function, and is instead solely influenced by intrathecal production. In contrast to the authors' findings, in our studies including both non-inflammatory neurological disorders (NIND; n = 62) and multiple sclerosis (MS) patients we observed a significant correlation between serum CXCL13 concentrations and CSF CXCL13 concentrations. We review several observations which may underlie these contrasting results, including (1) the impact of serum CXCL13 concentrations on CSF CXCL13 in patients with lower intrathecal CXCL13 production and thus lower CXCL13 concentrations (i.e. NIND and MS), (2) the proposed diffusion dynamics of the small molecule CXCL13 across the BCSFB, and (3) differing definitions of negative versus elevated CSF CXCL13 concentrations determined by an assay's relative sensitivity. In conclusion, we argue that for patients with moderately elevated CSF CXCL13 concentrations, serum CXCL13 concentrations influence CSF CXCL13 levels, and thus the appropriate corrections including incorporation of CSF/serum ratios and Qalbumin values should be utilized.


Subject(s)
Multiple Sclerosis , Nervous System Diseases , Chemokine CXCL13 , Humans , Inflammation
3.
Front Neurol ; 11: 616, 2020.
Article in English | MEDLINE | ID: mdl-32719651

ABSTRACT

Multiple sclerosis (MS) is the most common chronic inflammatory and neurodegenerative disease of the central nervous system (CNS). An interesting feature that this debilitating disease shares with many other inflammatory disorders is that susceptibility is higher in females than in males, with the risk of MS being three times higher in women compared to men. Nonetheless, while men have a decreased risk of developing MS, many studies suggest that males have a worse clinical outcome. MS exhibits an apparent sexual dimorphism in both the immune response and the pathophysiology of the CNS damage, ultimately affecting disease susceptibility and progression differently. Overall, women are predisposed to higher rates of inflammatory relapses than men, but men are more likely to manifest signs of disease progression and worse CNS damage. The observed sexual dimorphism in MS may be due to sex hormones and sex chromosomes, acting in parallel or combination. In this review, we outline current knowledge on the sexual dimorphism in MS and discuss the interplay of sex chromosomes, sex hormones, and the immune system in driving MS disease susceptibility and progression.

4.
J Vis Exp ; (159)2020 05 19.
Article in English | MEDLINE | ID: mdl-32510499

ABSTRACT

The central nervous system (CNS) is comprised of the brain and spinal cord and is enveloped by the meninges, membranous layers serving as a barrier between the periphery and the CNS. The CNS is an immunologically specialized site, and in steady state conditions, immune privilege is most evident in the CNS parenchyma. In contrast, the meninges harbor a diverse array of resident cells, including innate and adaptive immune cells. During inflammatory conditions triggered by CNS injury, autoimmunity, infection, or even neurodegeneration, peripherally derived immune cells may enter the parenchyma and take up residence within the meninges. These cells are thought to perform both beneficial and detrimental actions during CNS disease pathogenesis. Despite this knowledge, the meninges are often overlooked when analyzing the CNS compartment, because conventional CNS tissue extraction methods omit the meningeal layers. This protocol presents two distinct methods for the rapid isolation of murine CNS tissues (i.e., brain, spinal cord, and meninges) that are suitable for downstream analysis via single-cell techniques, immunohistochemistry, and in situ hybridization methods. The described methods provide a comprehensive analysis of CNS tissues, ideal for assessing the phenotype, function, and localization of cells occupying the CNS compartment under homeostatic conditions and during disease pathogenesis.


Subject(s)
Central Nervous System/cytology , Central Nervous System/immunology , Meninges/cytology , Meninges/immunology , Animals , Brain/cytology , Brain/immunology , Cell Aggregation , Cryopreservation , Demyelinating Diseases/pathology , Demyelinating Diseases/virology , Female , Leukocyte Common Antigens/metabolism , Mice , Paraffin Embedding , Spinal Cord/cytology , Spinal Cord/immunology , Theilovirus/physiology , Tissue Fixation
5.
Mult Scler J Exp Transl Clin ; 6(4): 2055217320981396, 2020.
Article in English | MEDLINE | ID: mdl-33403120

ABSTRACT

BACKGROUND: Clinicians caring for patients with Multiple Sclerosis (MS) need improved biomarkers to aid them in disease management. OBJECTIVE: We assessed the predictive value of the candidate biomarker CXCL13 index in comparison to oligoclonal bands (OCBs) and CSF neurofilament light (NfL) concentration, examining the ability of each biomarker to predict future disease activity in clinically and radiologically isolated syndromes, relapsing-remitting MS, and progressive MS. METHODS: Matched serum and CSF samples were obtained from 67 non-inflammatory neurologic disease patients and 67 MS patients. CSF and serum CXCL13 and CSF NfL were analyzed by Luminex and ELISA, respectively. CXCL13 data were also analyzed as CSF/serum ratios and indices. Electronic medical records were accessed to determine diagnosis, CSF profiles, and disease activity after the lumbar puncture. RESULTS: Among CXCL13 measures, CXCL13 index was the best predictor of future disease activity in MS patients (AUC = 0.82; CI = 0.69-0.95; p = 0.0002). CXCL13 index values were significantly elevated in activity-positive MS patients compared to activity-negative patients (p < 0.0001). As a single predictor, CXCL13 index outperformed both OCBs and CSF NfL in sensitivity, specificity, and positive and negative predictive value, for future disease activity in MS patients. Moreover, combining CXCL13 index and CSF NfL status improved sensitivity and predictive values for disease activity in MS patients. CONCLUSIONS: The CXCL13 index is an excellent candidate prognostic biomarker for disease activity in patients with MS.

6.
J Vis Exp ; (153)2019 11 29.
Article in English | MEDLINE | ID: mdl-31840664

ABSTRACT

Cerebrospinal fluid (CSF), a fluid found in the brain and the spinal cord, is of great importance to both basic and clinical science. The analysis of the CSF protein composition delivers crucial information in basic neuroscience research as well as neurological diseases. One caveat is that proteins measured in CSF may derive from both intrathecal synthesis and transudation from serum, and protein analysis of CSF can only determine the sum of these two components. To discriminate between protein transudation from the blood and intrathecally produced proteins in animal models as well as in humans, CSF protein profiling measurements using conventional protein analysis tools must include the calculation of the albumin CSF/serum quotient (Qalbumin), a marker of the integrity of the blood-brain interface (BBI), and the protein index (Qprotein/Qalbumin), an estimate of intrathecal protein synthesis. This protocol illustrates the entire procedure, from CSF and blood collection to quotients and indices calculations, for the quantitative measurement of intrathecal protein synthesis and BBI impairment in mouse models of neurological disorders.


Subject(s)
Cerebrospinal Fluid Proteins/chemistry , Cerebrospinal Fluid Proteins/metabolism , Albumins/cerebrospinal fluid , Albumins/chemistry , Albumins/metabolism , Animals , Biomarkers/cerebrospinal fluid , Humans , Mice , Serum Albumin , Specimen Handling
7.
Front Immunol ; 10: 1821, 2019.
Article in English | MEDLINE | ID: mdl-31428102

ABSTRACT

Persistent central nervous system (CNS) inflammation, as seen in chronic infections or inflammatory demyelinating diseases such as Multiple Sclerosis (MS), results in the accumulation of various B cell subsets in the CNS, including naïve, activated, memory B cells (Bmem), and antibody secreting cells (ASC). However, factors driving heterogeneous B cell subset accumulation and antibody (Ab) production in the CNS compartment, including the contribution of ectopic lymphoid follicles (ELF), during chronic CNS inflammation remain unclear and is a major gap in our understanding of neuroinflammation. We sought to address this gap using the Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) model of progressive MS. In this model, injection of the virus into susceptible mouse strains results in a persistent infection associated with demyelination and progressive disability. During chronic infection, the predominant B cell phenotypes accumulating in the CNS were isotype-switched B cells, including Bmem and ASC with naïve/early activated and transitional B cells present at low frequencies. B cell accumulation in the CNS during chronic TMEV-IDD coincided with intrathecal Ab synthesis in the cerebrospinal fluid (CSF). Mature and isotype-switched B cells predominately localized to the meninges and perivascular space, with IgG isotype-switched B cells frequently accumulating in the parenchymal space. Both mature and isotype-switched B cells and T cells occupied meningeal and perivascular spaces, with minimal evidence for spatial organization typical of ELF mimicking secondary lymphoid organs (SLO). Moreover, immunohistological analysis of immune cell aggregates revealed a lack of SLO-like ELF features, such as cell proliferation, cell death, and germinal center B cell markers. Nonetheless, flow cytometric assessment of B cells within the CNS showed enhanced expression of activation markers, including moderate upregulation of GL7 and expression of the costimulatory molecule CD80. B cell-related chemokines and trophic factors, including APRIL, BAFF, CXCL9, CXCL10, CCL19, and CXCL13, were elevated in the CNS. These results indicate that localization of heterogeneous B cell populations, including activated and isotype-switched B cell phenotypes, to the CNS and intrathecal Ab (ItAb) synthesis can occur independently of SLO-like follicles during chronic inflammatory demyelinating disease.


Subject(s)
Central Nervous System/immunology , Inflammation/immunology , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Theilovirus/immunology , Animals , Antibody Formation/immunology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biomarkers/metabolism , Central Nervous System/metabolism , Central Nervous System/virology , Demyelinating Diseases/immunology , Demyelinating Diseases/metabolism , Demyelinating Diseases/virology , Disease Models, Animal , Female , Germinal Center/immunology , Germinal Center/metabolism , Germinal Center/virology , Immunoglobulin G/immunology , Inflammation/metabolism , Inflammation/virology , Mice , Multiple Sclerosis/metabolism
8.
J Neuroinflammation ; 16(1): 109, 2019 May 22.
Article in English | MEDLINE | ID: mdl-31118079

ABSTRACT

BACKGROUND: The mechanisms driving multiple sclerosis (MS), the most common cause of non-traumatic disability in young adults, remain unknown despite extensive research. Especially puzzling are the underlying molecular processes behind the two major disease patterns of MS: relapsing-remitting and progressive. The relapsing-remitting course is exemplified by acute inflammatory attacks, whereas progressive MS is characterized by neurodegeneration on a background of mild-moderate inflammation. The molecular and cellular features differentiating the two patterns are still unclear, and the role of inflammation during progressive disease is a subject of active debate. METHODS: We performed a comprehensive analysis of the intrathecal inflammation in two clinically distinct mouse models of MS: the PLP139-151-induced relapsing experimental autoimmune encephalomyelitis (R-EAE) and the chronic progressive, Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). Microarray technology was first used to examine global gene expression changes in the spinal cord. Inflammation in the spinal cord was further assessed by immunohistochemical image analysis and flow cytometry. Levels of serum and cerebrospinal fluid (CSF) immunoglobulin (Ig) isotypes and chemokines were quantitated using Luminex Multiplex technology, whereas a capture ELISA was used to measure serum and CSF albumin levels. Finally, an intrathecal Ig synthesis index was established with the ratio of CSF and serum test results corrected as a ratio of their albumin concentrations. RESULTS: Microarray analysis identified an enrichment of B cell- and Ig-related genes upregulated in TMEV-IDD mice. We also demonstrated an increased level of intrathecal Ig synthesis as well as a marked infiltration of late differentiated B cells, including antibody secreting cells (ASC), in the spinal cord of TMEV-IDD, but not R-EAE mice. An intact blood-brain barrier in TMEV-IDD mice along with higher CSF levels of CXCL13, CXCL12, and CCL19 provides evidence for an intrathecal synthesis of chemokines mediating B cell localization to the central nervous system (CNS). CONCLUSIONS: Overall, these findings, showing increased concentrations of intrathecally produced Igs, substantial infiltration of ASC, and the presence of B cell supporting chemokines in the CNS of TMEV-IDD mice, but not R-EAE mice, suggest a potentially important role for Igs and ASC in the chronic progressive phase of demyelinating diseases.


Subject(s)
Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Spinal Cord/immunology , Theilovirus/immunology , Animals , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Mice , Multiple Sclerosis/pathology , Spinal Cord/pathology
9.
J Neuroimmunol ; 313: 34-40, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29153606

ABSTRACT

We evaluated the effects of pegylated-interferonß-1a (pegIFNß) therapy on intrathecal antibody responses, disability progression, and viral load in the CNS in mice infected with the Theiler's virus (TMEV), an animal model of progressive disability in Multiple Sclerosis (MS). The lack of a direct antiviral activity in the CNS, the absence of any effect upon the intrathecal immune response, and the failure to treat disease progression, indicate that the immunomodulatory effects of pegIFNß-1a likely occur in the systemic circulation rather than within the CNS. These results may be relevant to the relative lack of effect of IFNß in progressive MS relative to relapsing MS.


Subject(s)
Immunologic Factors/therapeutic use , Interferon-beta/therapeutic use , Multiple Sclerosis/drug therapy , Multiple Sclerosis/virology , Theilovirus/pathogenicity , Animals , Antibodies, Viral/blood , Disability Evaluation , Disease Models, Animal , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Mice , RNA, Messenger/metabolism , Rotarod Performance Test , Statistics, Nonparametric , Theilovirus/immunology , Viral Load
10.
J Neurovirol ; 23(6): 825-838, 2017 12.
Article in English | MEDLINE | ID: mdl-28913765

ABSTRACT

Teriflunomide is an oral therapy approved for the treatment of relapsing remitting multiple sclerosis (MS), showing both anti-inflammatory and antiviral properties. Currently, it is uncertain whether one or both of these properties may explain teriflunomide's beneficial effect in MS. Thus, to learn more about its mechanisms of action, we evaluated the effect of teriflunomide in the Theiler's encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) model, which is both a viral infection and an excellent model of the progressive disability of MS. We assessed the effects of the treatment on central nervous system (CNS) viral load, intrathecal immune response, and progressive neurological disability in mice intracranially infected with TMEV. In the TMEV-IDD model, we showed that teriflunomide has both anti-inflammatory and antiviral properties, but there seemed to be no impact on disability progression and intrathecal antibody production. Notably, benefits in TMEV-IDD were mostly mediated by effects on various cytokines produced in the CNS. Perhaps the most interesting result of the study has been teriflunomide's antiviral activity in the CNS, indicating it may have a role as an antiviral prophylactic and therapeutic compound for CNS viral infections.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , Cardiovirus Infections/drug therapy , Crotonates/pharmacology , Multiple Sclerosis/drug therapy , Toluidines/pharmacology , Animals , Antibodies, Viral/biosynthesis , Cardiovirus Infections/immunology , Cardiovirus Infections/virology , Cytokines/antagonists & inhibitors , Cytokines/biosynthesis , Disease Models, Animal , Disease Progression , Female , Hydroxybutyrates , Injections, Intraperitoneal , Mice , Multiple Sclerosis/immunology , Multiple Sclerosis/virology , Nitriles , Theilovirus/drug effects , Theilovirus/growth & development , Theilovirus/immunology , Viral Load/drug effects
11.
J Neurosci Methods ; 285: 58-68, 2017 Jun 15.
Article in English | MEDLINE | ID: mdl-28495370

ABSTRACT

BACKGROUND: CNS inflammation resulting from infection, injury, or neurodegeneration leads to accumulation of diverse B cell subsets. Although antibody secreting cells (ASC) within the inflamed CNS have been extensively examined, memory B cell (Bmem) characterization has been limited as they do not secrete antibody without stimulation. Moreover, unlike human Bmem, reliable surface markers for murine Bmem remain elusive. NEW METHOD: Using a viral encephalomyelitis model we developed a modified limiting dilution in vitro stimulation assay to convert CNS-derived virus specific Bmem into ASC. COMPARISON WITH EXISTING METHODS: Stimulation methods established for lymphoid tissue cells using prolonged stimulation with viral lysate resulted in substantial ASC loss and minimal Bmem to ASC conversion of CNS-derived cells. By varying stimulation duration, TLR activators, and culture supplements, we achieved optimal conversion by culturing cells with TLR7/8 agonist R848 in the presence of feeder cells for 2days. RESULTS: Flow cytometry markers CD38 and CD73 characterizing murine Bmem from lymphoid tissue showed more diverse expression patterns on corresponding CNS-derived B cell subsets. Using the optimized TLR7/8 stimulation protocol, we compared virus-specific IgG Bmem versus pre-existing ASC within the brain and spinal cord. Increasing Bmem frequencies during chronic infection mirrored kinetics of ASC. However, despite initially similar Bmem and ASC accumulation, Bmem prevailed in the brain, but were lower than ASC in the spinal cord during persistence. CONCLUSION: Simultaneous enumeration of antigen-specific Bmem and ASC using the Bmem assay optimized for CNS-derived cells enables characterization of temporal changes during microbial or auto-antigen induced neuroinflammation.


Subject(s)
Antibody-Producing Cells/physiology , B-Lymphocytes/cytology , Central Nervous System/pathology , Hepatitis, Viral, Animal/complications , Inflammation/etiology , Inflammation/pathology , Animals , Antibody-Producing Cells/drug effects , B-Lymphocytes/drug effects , Cell Differentiation , Cell Movement , Central Nervous System/drug effects , Central Nervous System/virology , Cyclopropanes/pharmacology , Cytokines/metabolism , Disease Models, Animal , Flow Cytometry , Guanosine/analogs & derivatives , Guanosine/pharmacology , Imidazoles/pharmacology , In Vitro Techniques , Mice , Mice, Inbred C57BL , Murine hepatitis virus/pathogenicity , Spinal Cord/pathology , Spinal Cord/virology , Spleen/cytology , Time Factors , Toll-Like Receptor 1/antagonists & inhibitors , Toll-Like Receptor 1/metabolism
12.
Brain Behav Immun ; 60: 71-83, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27658544

ABSTRACT

Central nervous system (CNS) inflammation associated with viral infection and autoimmune disease results in the accumulation of B cells in various differentiation stages. However, the contribution between peripheral and CNS activation remains unclear. During gliatropic coronavirus induced encephalomyelitis, accumulation of protective antibody secreting cells is preceded by infiltration of B cells with a naïve and early differentiation phenotype (Phares et al., 2014). Investigation of the temporal dynamics of B cell activation in draining cervical lymph nodes (CLN) and the CNS revealed that peak CNS infiltration of early activated, unswitched IgD+ and IgM+ B cells coincided with polyclonal activation in CLN. By contrast, isotype-switched IgG+ B cells did not accumulate until peripheral germinal center formation. In the CNS, unswitched B cells were confined to the perivascular space and meninges, with only rare B cell clusters, while isotype-switched B cells localized to parenchymal areas. Although ectopic follicle formation was not observed, more differentiated B cell subsets within the CNS expressed the germinal center marker GL7, albeit at lower levels than CLN counterparts. During chronic infection, CNS IgDint and IgD- B cell subsets further displayed sustained markers of proliferation and CD4 T cell help, which were only transiently expressed in the CLN. A contribution of local CD4 T cell help to sustain B cell activation was supported by occasional B cells adjacent to T cells. The results suggest that accumulation of differentiated B cell subsets within the CNS is largely dictated by peripheral activation, but that local events contribute to their sustained activation independent of ectopic follicle formation.


Subject(s)
B-Lymphocytes/virology , Central Nervous System/virology , Coronavirus Infections/immunology , Encephalomyelitis/virology , Lymphocyte Activation/immunology , Animals , B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Movement/immunology , Central Nervous System/immunology , Coronavirus Infections/virology , Encephalomyelitis/immunology , Mice, Inbred C57BL
13.
Brain Behav Immun ; 54: 128-139, 2016 May.
Article in English | MEDLINE | ID: mdl-26795429

ABSTRACT

Elevated CXCL13 within the central nervous system (CNS) correlates with humoral responses in several neuroinflammatory diseases, yet its role is controversial. During coronavirus encephalomyelitis CXCL13 deficiency impaired CNS accumulation of memory B cells and antibody-secreting cells (ASC) but not naïve/early-activated B cells. However, despite diminished germinal center B cells and follicular helper T cells in draining lymph nodes, ASC in bone marrow and antiviral serum antibody were intact in the absence of CXCL13. The data demonstrate that CXCL13 is not essential in mounting effective peripheral humoral responses, but specifically promotes CNS accumulation of differentiated B cells.


Subject(s)
B-Lymphocytes/immunology , Central Nervous System/immunology , Chemokine CXCL13/immunology , Coronavirus Infections/immunology , Encephalomyelitis/immunology , Animals , B-Lymphocytes/pathology , Cell Movement/immunology , Coronavirus Infections/pathology , Encephalomyelitis/pathology , Female , Immunoglobulin Class Switching/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Helper-Inducer/immunology
14.
J Virol ; 88(16): 8853-67, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24872583

ABSTRACT

UNLABELLED: Various infections in the central nervous system (CNS) trigger B cell accumulation; however, the relative dynamics between viral replication and alterations in distinct B cell subsets are largely unknown. Using a glia-tropic coronavirus infection, which is initiated in the brain but rapidly spreads to and predominantly persists in the spinal cord, this study characterizes longitudinal changes in B cell subsets at both infected anatomical sites. The phase of T cell-dependent, antibody-independent control of infectious virus was associated with a similar recruitment of naive/early-activated IgD(+) IgM(+) B cells into both the brain and spinal cord. This population was progressively replaced by CD138(-) IgD(-) IgM(+) B cells, isotype-switched CD138(-) IgD(-) IgM(-) memory B cells (B(mem)), and CD138(+) antibody-secreting cells (ASC). A more rapid transition to B(mem) and ASC in spinal cord than in brain was associated with higher levels of persisting viral RNA and transcripts encoding factors promoting B cell migration, differentiation, and survival. The results demonstrate that naive/early-activated B cells are recruited early during coronavirus CNS infection but are subsequently replaced by more differentiated B cells. Furthermore, viral persistence, even at low levels, is a driving force for accumulation of isotype-switched B(mem) and ASC. IMPORTANCE: Acute and chronic human CNS infections are associated with an accumulation of heterogeneous B cell subsets; however, their influence on viral load and disease is unclear. Using a glia-tropic coronavirus model, we demonstrate that the accumulation of B cells ranging from early-activated to isotype-switched differentiation stages is both temporally and spatially orchestrated. Acutely infected brains and spinal cords indiscriminately recruit a homogeneous population of early-activated B cells, which is progressively replaced by diverse, more differentiated subsets. The latter process is accelerated by elevated proinflammatory responses associated with viral persistence. The results imply that early-recruited B cells do not have antiviral function but may contribute to the inflammatory environment or act as antigen-presenting cells. Moreover, CNS viral persistence is a driving force promoting differentiated B cells with protective potential.


Subject(s)
B-Lymphocytes/immunology , Coronavirus Infections/immunology , Coronavirus/immunology , Encephalomyelitis/immunology , Immunoglobulin Class Switching/immunology , Immunoglobulin D/immunology , Immunoglobulin M/immunology , Animals , Antibody-Producing Cells/immunology , Antibody-Producing Cells/virology , Antigen-Presenting Cells/immunology , B-Lymphocytes/virology , Brain/immunology , Brain/virology , Cell Differentiation/immunology , Cell Movement/immunology , Coronavirus Infections/virology , Encephalomyelitis/virology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , RNA, Viral/immunology , Spinal Cord/immunology , Spinal Cord/virology
15.
J Neuroimmunol ; 263(1-2): 43-54, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23992866

ABSTRACT

Acute coronavirus encephalomyelitis is controlled by T cells while humoral responses suppress virus persistence. This study defines the contribution of interleukin (IL)-21, a regulator of T and B cell function, to central nervous system (CNS) immunity. IL-21 receptor deficiency did not affect peripheral T cell activation or trafficking, but dampened granzyme B, gamma interferon and IL-10 expression by CNS T cells and reduced serum and intrathecal humoral responses. Viral control was already lost prior to humoral CNS responses, but demyelination remained comparable. These data demonstrate a critical role of IL-21 in regulating CNS immunity, sustaining viral persistence and preventing mortality.


Subject(s)
B-Lymphocyte Subsets/immunology , Coronavirus Infections/immunology , Encephalitis, Viral/immunology , Immunity, Humoral , Interleukins/physiology , T-Lymphocyte Subsets/immunology , Animals , Central Nervous System/immunology , Central Nervous System/metabolism , Central Nervous System/virology , Coronavirus/immunology , Coronavirus Infections/metabolism , Coronavirus Infections/pathology , Encephalitis, Viral/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Virus Replication/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...