scispace - formally typeset
Search or ask a question

Showing papers by "Igor Smirnov published in 2014"


Journal ArticleDOI
TL;DR: Transfer of a monoclonal T cell population reactive to the central nervous system (CNS) antigen, myelin oligodendrocyte glycoprotein (MOG), was sufficient to improve cognitive task performance in otherwise impaired OTII mice, raising the possibility that the antigen-specificity requirement of pro-cognitive T cells may be directed against CNS-derived self-antigens.
Abstract: Numerous methods of T cell depletion lead to impairment of learning and memory function in mice. While adoptive transfer of whole splenocytes rescues learning behavior impairments, the precise sub-population and antigenic specificity of the T cells mediating the rescue remains unknown. Using several transgenic mouse models in combination with adoptive transfers, we demonstrate the necessity of an antigen-specific CD4+ T cell compartment in normal spatial learning and memory, as measured by the Morris water maze (MWM). Moreover, transfer of a monoclonal T cell population reactive to the central nervous system (CNS) antigen, myelin oligodendrocyte glycoprotein (MOG), was sufficient to improve cognitive task performance in otherwise impaired OTII mice, raising the possibility that the antigen-specificity requirement of pro-cognitive T cells may be directed against CNS-derived self-antigens.

102 citations


Journal ArticleDOI
TL;DR: It is shown that meningeal T-cell composition is coupled to the CNS-draining deep cervical lymph nodes (dCLNs), whose surgical removal interrupted the normal flow of meningealing T-cells and resulted in cognitive impairment.
Abstract: We recently identified the meninges, which envelopes the outer and ventricular surfaces of the brain and spinal cord, as a candidate site for beneficial T-cell interaction with the CNS 3. The meninges are permissive to circulating lymphocytes, harbor MHCII-expressing cell types, and are well situated to broadly affect CNS function 3, 7, 8. To study the dynamics of CD4+ T-cells at this site, we employed a parabiotic mouse model, enabling us to characterize the relative intra-tissue cell turnover rate in the CD4+ compartment. By day 15 after parabiotic surgery, CD4+ T-cells equilibrate across the blood and lymphoid tissues, however, their turnover in the meninges (and CNS draining dCLN) was severely delayed (Fig. 1a), suggesting that the entry of CD4+ T-cells was gated, passively or actively, by the meninges. We broadly define “passive gating” as an intrinsically CD4-restrictive meningeal environment, and “active gating” as permissive but regulated entry. Figure 1 (a) Ly5.1 (Black) and Ly5.2 (Blue) mice were parabiotically joined. After 15 days, each mouse was perfused with PBS and the indicated tissues were analyzed for self and donor CD4+ T-cells by FACS, showing slow relative T-cell turnover in the meninges ... To differentiate between active and passive gating, we transferred wild-type Ly5.1 splenocytes into OTII mice. We predicted that given the low CD4+ T-cell turnover in the meninges, seeding by transferred wild-type T-cells (i.e., their “passive gating”) would occur more slowly here than in other tissues. Surprisingly however, the meninges were the most receptive of all the analyzed tissues, with the largest percentage of their total CD4+ T-cells originating from transferred Ly5.1 splenocytes (Fig. 1b). We interpreted this result to mean the meninges is not globally restrictive to CD4+ T-cell entry and residence. Polyclonal wild-type T-cells are known to expand homeostatically under conditions of lymphopenia or low clonal diversity9. To determine whether homeostatic expansion was driving T-cells from the blood into the meninges, we connected OTII transgenic mice parabiotically with wild-type partners (Fig. 1c). Here, as in the adoptive transfer model, the OTII mouse meninges accepted impressive numbers of donor CD4+ T-cells from the blood. Surprisingly, whereas “naive” CD44− T-cells equilibrated evenly between parabiotic partners, CD44hi “activated/memory” T-cells did not (Fig. 1d). Not only can this explain the lack of T-cell accumulation in the meninges of the wild-type partner, but it also serves as a cautionary tale concerning a major assumption of the parabiotic model, namely that the unified circulatory system guarantees homogenous leukocyte circulation. In fact, mixing of CD11b+ monocyte populations was also severely diminished (Fig. 1e), suggesting either that some cell types cannot pass through the unified capillary junctions or that they extravasate directly into the site of surgical injury. Both the uniquely slow T-cell turnover rate in the wild type meninges and the increased seeding of the meninges of OTII mice by wild type T-cells was also apparent in the dCLNs. To explore possible coupling of the meninges and dCLNs, and to find out how interruption of this coupling might affect the T-cell composition of the meninges, we surgically resected dCLNs of wild-type mice. Two weeks after surgery the numbers of CD4+ T-cells in the meninges of operated mice were substantially increased (Fig. 1f). To find out what effect, if any, dCLN resection might have on behavioral outcomes, we subjected resected and sham operated mice to the Morris Water Maze (MWM) behavioral task, and observed significant spatial learning and memory impairment in the dCLN resected group–particularly in the reversal phase of the assay (Fig. 1g). While the notion that T-cells are required for normal learning and memory is not an intuitive one, every mode of T-cell depletion attempted so far substantiates this necessity (reviewed in 10). This phenomenon is particularly interesting given that the cognitive decline seen in patients undergoing chemotherapy, and in those suffering from age-related senility or HIV-associated dementia, is correlated with a collapse in adaptive immunity that occurs most saliently within the CD4+ T-cell compartment. As we begin to ask what role(s) T-cells are playing in higher brain functions, honing in on the specific localization and migratory behavior of T-cells that operate in the meningeal spaces of the CNS is a likely prerequisite to understanding their ultimate function. Additionally, while the phenotypic readout in this study is learning and memory, a general understanding of T-cell makeup and behavior in the meninges may also be applicable to CNS pathologies such as MS, in which lymphocyte accumulation in the meninges is thought to precede parenchymal infiltration 7.

76 citations


Journal ArticleDOI
TL;DR: It is demonstrated that neuronal survival after CNS injury is impaired when Treg cells are either removed or added, and the phenotype of macrophages at the site, however, is affected: both addition and removal of Treg negatively impact the numbers of Macrophages with alternatively activated (tissue-building) phenotype.
Abstract: Previous research investigating the roles of T effector (T(eff)) and T regulatory (T(reg)) cells after injury to the CNS has yielded contradictory conclusions, with both protective and destructive functions being ascribed to each of these T cell subpopulations In this work, we study this dichotomy by examining how regulation of the immune system affects the response to CNS trauma We show that, in response to CNS injury, T(eff) and T(reg) subsets in the CNS-draining deep cervical lymph nodes are activated, and surgical resection of these lymph nodes results in impaired neuronal survival Depletion of T(reg), not surprisingly, induces a robust T(eff) response in the draining lymph nodes and is associated with impaired neuronal survival Interestingly, however, injection of exogenous T(reg) cells, which limits the spontaneous beneficial immune response after CNS injury, also impairs neuronal survival We found that no T(reg) accumulate at the site of CNS injury, and that changes in T(reg) numbers do not alter the amount of infiltration by other immune cells into the site of injury The phenotype of macrophages at the site, however, is affected: both addition and removal of T(reg) negatively impact the numbers of macrophages with alternatively activated (tissue-building) phenotype Our data demonstrate that neuronal survival after CNS injury is impaired when T(reg) cells are either removed or added With this exacerbation of neurodegeneration seen with both addition and depletion of T(reg), we recommend exercising extreme caution when considering the therapeutic targeting of T(reg) cells after CNS injury, and possibly in chronic neurodegenerative conditions

70 citations