Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Nat Immunol ; 21(7): 802-815, 2020 07.
Article in English | MEDLINE | ID: mdl-32541832

ABSTRACT

Microglia and central nervous system (CNS)-associated macrophages (CAMs), such as perivascular and meningeal macrophages, are implicated in virtually all diseases of the CNS. However, little is known about their cell-type-specific roles in the absence of suitable tools that would allow for functional discrimination between the ontogenetically closely related microglia and CAMs. To develop a new microglia gene targeting model, we first applied massively parallel single-cell analyses to compare microglia and CAM signatures during homeostasis and disease and identified hexosaminidase subunit beta (Hexb) as a stably expressed microglia core gene, whereas other microglia core genes were substantially downregulated during pathologies. Next, we generated HexbtdTomato mice to stably monitor microglia behavior in vivo. Finally, the Hexb locus was employed for tamoxifen-inducible Cre-mediated gene manipulation in microglia and for fate mapping of microglia but not CAMs. In sum, we provide valuable new genetic tools to specifically study microglia functions in the CNS.


Subject(s)
Brain/pathology , Encephalomyelitis, Autoimmune, Experimental/pathology , Facial Nerve Injuries/pathology , Microglia/metabolism , beta-Hexosaminidase beta Chain/metabolism , Animals , Brain/cytology , Brain/immunology , CRISPR-Cas Systems/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Facial Nerve Injuries/immunology , Gene Knock-In Techniques , Genes, Reporter/genetics , Genetic Loci/genetics , Humans , Intravital Microscopy , Luminescent Agents/chemistry , Luminescent Proteins/chemistry , Luminescent Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , Microglia/immunology , NIH 3T3 Cells , RNA-Seq , Single-Cell Analysis , Transfection , beta-Hexosaminidase beta Chain/genetics , Red Fluorescent Protein
2.
Stem Cells Dev ; 28(7): 489-496, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30704338

ABSTRACT

Normal facial nerve (FN) function is very important for human being. However, if injured, FN function is difficult to restore completely. Recently, many studies reported the immune regulation function of stem cells (SCs). However, the immunomodulation function of SCs on FN injury is still unclear. Our study aims to explore the mechanism of immunomodulation effect of Sprague-Dawley rat bone marrow-derived SCs (BMSCs) on FN injury and specially focus on the regulation of Th17 and the protection effects of BMSCs on central facial motor neurons (FMNs). First, rat FNs were harvested. FN and BMSCs were cultured together or separately and levels of transforming growth factor (TGF)-ß1, interleukin (IL)-6, hepatocyte growth factor (HGF), inducible nitric oxide synthase (iNOS), and prostaglandin E2 (PGE2) in supernatant were detected by enzyme-linked immunosorbent assay (ELISA). Then, after treating with or without local BMSCs injection, the proportion of Th17 in neck lymph nodes (LNs) was investigated in rat FN injury models. Furthermore, the apoptotic index of FMNs was studied in rat FN injury models that were treated with or without BMSCs. We found that BMSCs could secrete high levels of IL-6, HGF, PGE2, iNOS, and TGF-ß1 in culture. The percentage of Th17 of neck LNs in BMSCs-treated group was significantly lower than that in the control group. The apoptotic index of FMNs in BMSCs-treated group was significantly lower than that in the control group. In conclusion, our research indicates BMSCs could independently secrete cytokines IL-6, HGF, PGE2, iNOS, and TGF-ß1, and these cytokines could regulate the balance among subsets of CD4+ T cells and could protect FMNs by inhibiting neuron apoptosis.


Subject(s)
Bone Marrow Cells/immunology , Facial Nerve Injuries/therapy , Facial Nerve/immunology , Immunomodulation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Animals , Bone Marrow Cells/pathology , Facial Nerve/pathology , Facial Nerve Injuries/immunology , Facial Nerve Injuries/pathology , Mesenchymal Stem Cells/pathology , Rats , Rats, Sprague-Dawley , Th17 Cells/immunology , Th17 Cells/pathology
3.
Neuromolecular Med ; 19(2-3): 357-374, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28689354

ABSTRACT

Traumatic injuries to human peripheral nerves are frequently associated with damage to nerve surrounding tissues including muscles and blood vessels. Currently, most rodent models of peripheral nerve injuries (e.g., facial or sciatic nerve) employ surgical nerve transection with scissors or scalpels. However, such an isolated surgical nerve injury only mildly damages neighboring tissues and weakly activates an immune response. In order to provide a rodent nerve injury model accounting for such nerve-associated tissue damage and immune cell activation, we developed a drop tower-based facial nerve trauma model in mice. We compare nerve regeneration in this novel peripheral nerve trauma model with the established surgical nerve injury along several parameters. These include gene expression, histological and functional facial motoneuron (FMN) regeneration, facial nerve degeneration, immune cell activation and muscle damage. Regeneration-associated genes (RAGs; e.g., Atf3) were strongly induced in FMNs subjected to traumatic and surgical injury. Regeneration of FMNs and functional recovery of whisker movement were faster in traumatic versus complete surgical injury, thus cutting down experimentation time. Wallerian degeneration of distal nerve stumps was readily observed in this novel trauma injury model. Importantly, drop tower-inflicted facial nerve injury resulted in muscle damage, activation of muscle satellite cell markers (PAX7) and pronounced infiltration of immune cells to the injury site only in this model but not upon surgical nerve transection. Thus, we provide a novel rodent PNS trauma model that can be easily adopted to other PNS nerves such as the sciatic nerve. Since this nerve trauma model replicates multiple tissue damage frequently encountered in clinical routine, it will be well suited to identify molecular and cellular mechanisms of PNS nerve repair in wild-type and genetically modified rodents.


Subject(s)
Facial Nerve Injuries/physiopathology , Facial Nerve/physiology , Models, Animal , Nerve Regeneration , Animals , Facial Nerve/surgery , Facial Nerve Injuries/genetics , Facial Nerve Injuries/immunology , Facial Nerve Injuries/pathology , Female , Gene Expression Regulation , Male , Masseter Muscle/pathology , Mice , Mice, Inbred C57BL , Motor Neurons/physiology , Nerve Regeneration/genetics , Nerve Regeneration/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Vibrissae/physiology , Wallerian Degeneration , Wounds, Nonpenetrating/physiopathology
4.
Mediators Inflamm ; 2016: 6131234, 2016.
Article in English | MEDLINE | ID: mdl-27194826

ABSTRACT

An increased risk of ALS has been reported for veterans, varsity athletes, and professional football players. The mechanism underlying the increased risk in these populations has not been identified; however, it has been proposed that motor nerve injury may trigger immune responses which, in turn, can accelerate the progression of ALS. Accumulating evidence indicates that abnormal immune reactions and inflammation are involved in the pathogenesis of ALS, but the specific immune cells involved have not been clearly defined. To understand how nerve injury and immune responses may contribute to ALS development, we investigated responses of CD4(+) T cell after facial motor nerve axotomy (FNA) at a presymptomatic stage in a transgenic mouse model of ALS (B6SJL SOD1(G93A)). SOD1(G93A) mice, compared with WT mice, displayed an increase in the basal activation state of CD4(+) T cells and higher frequency of Th17 cells, which were further enhanced by FNA. In conclusion, SOD1(G93A) mice exhibit abnormal CD4(+) T cell activation with increased levels of Th17 cells prior to the onset of neurological symptoms. Motor nerve injury exacerbates Th17 cell responses and may contribute to the development of ALS, especially in those who carry genetic susceptibility to this disease.


Subject(s)
Facial Nerve Injuries/metabolism , Facial Nerve Injuries/pathology , Motor Neurons/pathology , Superoxide Dismutase-1/metabolism , Th17 Cells/metabolism , Animals , Disease Models, Animal , Facial Nerve Injuries/immunology , Female , Mice , Mice, Transgenic , Motor Neurons/immunology , Motor Neurons/metabolism , Superoxide Dismutase-1/genetics , T-Lymphocytopenia, Idiopathic CD4-Positive/metabolism , Th17 Cells/immunology
5.
Neurosci Lett ; 526(2): 138-43, 2012 Sep 27.
Article in English | MEDLINE | ID: mdl-22922129

ABSTRACT

IL-2 is essential for T-helper regulatory (Treg) cell function and self-tolerance, and dysregulation of both endogenous brain and peripheral IL-2 gene expression may have important implications for neuronal injury and repair. We used an experimental approach combining mouse congenic breeding and immune reconstitution to test the hypothesis that the response of motoneurons to injury is modulated by the combined effects of IL2-mediated processes in the brain that modulate its endogenous neuroimmunological milieu, and IL2-mediated processes in the peripheral immune system that regulate T cell function (i.e., normal versus autoreactive Treg-deficient T cells). This experimental strategy enabled us to test our hypothesis by disentangling the effect of normal versus autoreactive T lymphocytes from the effect of endogenous brain IL-2 on microglial responsiveness (microglial phagocytic clusters normally associated with dead motoneurons and MHC2(+) activated microglia) and T cell trafficking, using the facial nerve axotomy model of injury. The results demonstrate that the loss of both brain and peripheral IL-2 had an additive effect on numbers of microglial phagocytic clusters at day 14 following injury, whereas the autoreactive status of peripheral T cells was the primary factor that determined the degree to which T cells entered the injured brain and contributed to increased microglial phagocytic clusters. Changes in activated MHC2(+) microglial in the injured FMN were associated with loss of endogenous brain IL-2 and/or peripheral IL-2. This model may provide greater understanding of the mechanisms involved in determining if T cells entering the injured central nervous system (CNS) have damaging or proregenerative effects.


Subject(s)
Axons/physiology , Brain/metabolism , Interleukin-2/metabolism , Microglia/physiology , Neuroimmunomodulation , T-Lymphocytes/physiology , Adoptive Transfer , Animals , Autoimmunity , Axons/immunology , Axotomy , Brain/immunology , Brain/pathology , Cell Movement , DNA-Binding Proteins/genetics , Facial Nerve Injuries/immunology , Facial Nerve Injuries/pathology , Immunocompromised Host/genetics , Interleukin-2/genetics , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/pathology , Motor Neurons/immunology , Motor Neurons/physiology , Motor Neurons/ultrastructure , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/immunology
6.
Brain Behav Immun ; 26(6): 886-90, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22426430

ABSTRACT

BACKGROUND: The production of neurotrophic factors, such as BDNF, has generally been considered an important mechanism of immune-mediated neuroprotection. However, the ability of T cells to produce BDNF remains controversial. METHODS: In the present study, we examined mRNA and protein of BDNF using RT-PCR and western blot, respectively, in purified and reactivated CD4(+) T cells. In addition, to determine the role of BDNF derived from CD4(+) T cells, the BDNF gene was specifically deleted in T cells using the Cre-lox mouse model system. RESULTS: Our results indicate that while both mRNA expression and protein secretion of BDNF in reactivated T cells were detected at 24 h, only protein could be detected at 72 h after reactivation. The results suggest a transient up-regulation of BDNF mRNA in reactivated T cells. Furthermore, in contrast to our hypothesis that the BDNF expression is necessary for CD4(+) T cells to mediate neuroprotection, mice with CD4(+) T cells lacking BDNF expression demonstrated a similar level of facial motoneuron survival compared to their littermates that expressed BDNF, and both levels were comparable to wild-type. The results suggest that the deletion of BDNF did not impair CD4(+) T cell-mediated neuroprotection. CONCLUSION: Collectively, while CD4(+) T cells are a potential source of BDNF after nerve injury, production of BDNF is not necessary for CD4(+) T cells to mediate their neuroprotective effects.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , CD4-Positive T-Lymphocytes/immunology , Facial Nerve Injuries/immunology , Animals , Axotomy , Blotting, Western , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/genetics , CD4-Positive T-Lymphocytes/metabolism , Cell Count , Cell Survival/physiology , Facial Nerve Injuries/metabolism , Female , Flow Cytometry , Mice , Mice, Knockout , Motor Neurons/physiology , RNA/biosynthesis , RNA/isolation & purification , Real-Time Polymerase Chain Reaction
7.
Metab Brain Dis ; 27(1): 23-8, 2012 Mar.
Article in English | MEDLINE | ID: mdl-21975535

ABSTRACT

It has been well documented that both estrogen and immune cells (CD4+ T cells) mediate neuroprotection in the mouse facial nerve axotomy model. Estrogen has been shown to play an important role in regulating the immune response. However, it is unclear whether immune cell-mediated neuroprotection is dependent on estrogen signaling. In this study, using FACS staining, we confirmed that the majority of CD4+ T cells express high levels of estrogen receptor-alpha (ERα), suggesting that CD4+ T cell-mediated neuroprotection may be modulated by estrogen signaling. We previously found that immunodeficient Rag-2KO mice showed a significant increase in axotomy-induced facial motoneuron death compared to immunocompetent wild-type mice. Therefore, we investigated axotomy-induced facial motoneuron loss in immunodeficient Rag-2KO mice that received 17ß-estradiol treatment or adoptive transfer of immune cells from mice lacking functional ERα. Our results indicate that while estradiol treatment failed to rescue facial motoneurons from axotomy-induced cell death in Rag-2KO mice, immune cells lacking ERα successfully restored facial motoneuron survival in Rag-2 KO mice to a wild-type level. Collectively, we concluded that CD4+ T cell-mediated neuroprotection is independent of estrogen action through ERα.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Estrogen Receptor alpha/metabolism , Estrogens/metabolism , Facial Nerve Injuries/pathology , Motor Neurons/physiology , Adoptive Transfer , Animals , Axotomy/methods , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Communication/immunology , Cell Survival/immunology , Cell Survival/physiology , DNA-Binding Proteins/immunology , DNA-Binding Proteins/metabolism , Disease Models, Animal , Estradiol/pharmacology , Estrogen Receptor alpha/immunology , Facial Nerve/immunology , Facial Nerve/pathology , Facial Nerve/surgery , Facial Nerve Injuries/immunology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/cytology , Motor Neurons/immunology , Motor Neurons/metabolism , Signal Transduction
8.
Brain Behav Immun ; 25(5): 820-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20723599

ABSTRACT

We have previously shown that immunodeficient mice exhibit significant facial motoneuron (FMN) loss compared to wild-type (WT) mice after a facial nerve axotomy. Interleukin-10 (IL-10) is known as a regulatory cytokine that plays an important role in maintaining the anti-inflammatory environment within the central nervous system (CNS). IL-10 is produced by a number of different cells, including Th2 cells, and may exert an anti-apoptotic action on neurons directly. In the present study, the role of IL-10 in mediating neuroprotection following facial nerve axotomy in Rag-2- and IL-10-deficient mice was investigated. Results indicate that IL-10 is neuroprotective, but CD4+ T cells are not the requisite source of IL-10. In addition, using real-time PCR analysis of laser microdissected brainstem sections, results show that IL-10 mRNA is constitutively expressed in the facial nucleus and that a transient, significant reduction of IL-10 mRNA occurs following axotomy under immunodeficient conditions. Dual labeling immunofluorescence data show, unexpectedly, that the IL-10 receptor (IL-10R) is constitutively expressed by facial motoneurons, but is selectively induced in astrocytes within the facial nucleus after axotomy. Thus, a non-CD4+ T cell source of IL-10 is necessary for modulating both glial and neuronal events that mediate neuroprotection of injured motoneurons, but only with the cooperation of CD4+ T cells, providing an avenue of novel investigation into therapeutic approaches to prevent or reverse motoneuron diseases, such as amyotrophic lateral sclerosis (ALS).


Subject(s)
CD4-Positive T-Lymphocytes/physiology , Central Nervous System/immunology , Immunity, Cellular/physiology , Interleukin-10/physiology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/immunology , Central Nervous System/physiology , Enzyme-Linked Immunosorbent Assay , Facial Nerve Injuries/immunology , Facial Nerve Injuries/physiopathology , Female , Immunity, Cellular/immunology , Inflammation/immunology , Inflammation/physiopathology , Interleukin-10/immunology , Interleukin-10 Receptor alpha Subunit/immunology , Interleukin-10 Receptor alpha Subunit/physiology , Interleukin-10 Receptor beta Subunit/immunology , Interleukin-10 Receptor beta Subunit/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Neurons/immunology , Motor Neurons/physiology , Neurons/immunology , Neurons/physiology , Reverse Transcriptase Polymerase Chain Reaction
9.
J Neuroimmunol ; 216(1-2): 66-75, 2009 Nov 30.
Article in English | MEDLINE | ID: mdl-19818514

ABSTRACT

We have previously demonstrated a neuroprotective mechanism of facial motoneuron (FMN) survival after facial nerve axotomy that is dependent on CD4(+) Th2 cell interaction with peripheral antigen-presenting cells, as well as CNS resident microglia. To investigate this mechanism, we chose to study the Th2-associated chemokine, CCL11, and Th1-associated chemokine, CXCL11, in wild-type and presymptomatic mSOD1 mice after facial nerve axotomy. In this report, the results indicate that CCL11 is constitutively expressed in the uninjured facial motor nucleus, but CXCL11 is not expressed at all. Facial nerve axotomy induced a shift in CCL11 expression from FMN to astrocytes, whereas CXCL11 was induced in FMN. Differences in the number of CCL11- and CXCL11-expressing cells were observed between WT and mSOD1 mice after facial nerve axotomy.


Subject(s)
Chemokines/metabolism , Facial Nerve Injuries/immunology , Facial Nerve/immunology , Motor Neurons/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/immunology , Amyotrophic Lateral Sclerosis/metabolism , Animals , Astrocytes/cytology , Astrocytes/immunology , Astrocytes/metabolism , Axotomy , Cell Survival/immunology , Chemokine CCL11/metabolism , Chemokine CXCL11/metabolism , Disease Models, Animal , Facial Nerve/cytology , Facial Nerve/metabolism , Facial Nerve Injuries/metabolism , Facial Nerve Injuries/physiopathology , Female , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Neurons/cytology , Motor Neurons/metabolism , Nerve Regeneration/genetics , Nerve Regeneration/immunology , Superoxide Dismutase/genetics , Superoxide Dismutase-1
10.
J Neuroimmunol ; 212(1-2): 44-52, 2009 Jul 25.
Article in English | MEDLINE | ID: mdl-19481818

ABSTRACT

Major histocompatibility complex (MHC) class II is of critical importance for the induction of immune responses. Levels of MHC class II in the nervous system are normally low, but expression is up-regulated in many disease conditions. In rat and human, variation in the MHC class II transactivator gene (C2ta) is associated with differential expression of MHC class II and susceptibility to autoimmune disease. Here we have characterized the response to facial nerve transection in 7 inbred mouse strains (C57BL/6J, DBA/2J, 129X1/SvJ, BALB/cJ, SJL/J, CBA/J, and NOD). The results demonstrate differences in expression of C2ta and markers for MHC class I and II expression, glial activation, and T cell infiltration. Expression levels of C2ta and Cd74 followed similar patterns, in contrast to MHC class I and markers of glial activation. The regulatory region of the C2ta gene was subsequently sequenced in the four strains (C57BL/6/J, DBA/2J, SJL/J and 129X1/SvJ) that represented the phenotypical extremes with regard to C2ta/Cd74 expression. We found 3 single nucleotide polymorphisms in the type I (pI) and type III (pIII) promoters of C2ta, respectively. Higher expression of pI in 129X1/SvJ correlated with the pI haplotype specific for this strain. Furthermore, congenic strains carrying the 129X1/SvJ C2ta allele on B6 background displayed significantly higher C2ta and Cd74 expression compared to parental controls. We conclude that genetic polymorphisms in the type I promoter of C2ta regulates differential expression of MHC class II, but not MHC class I, Cd3 and other markers of glial activation.


Subject(s)
Facial Nerve Injuries/immunology , Genes, MHC Class II , Nuclear Proteins/genetics , Promoter Regions, Genetic , Trans-Activators/genetics , Animals , Antigens, Differentiation, B-Lymphocyte/analysis , Antigens, Differentiation, B-Lymphocyte/genetics , CD3 Complex/analysis , Female , Gene Expression Regulation , Genetic Variation , Histocompatibility Antigens Class II/analysis , Histocompatibility Antigens Class II/genetics , Immunohistochemistry , Mice , Mice, Inbred Strains , Nuclear Proteins/analysis , Polymorphism, Genetic , Protein Isoforms , Species Specificity , Trans-Activators/analysis
11.
J Neuroimmunol ; 199(1-2): 18-23, 2008 Aug 13.
Article in English | MEDLINE | ID: mdl-18538865

ABSTRACT

The temporal relationship between severity of peripheral axonal injury and T lymphocyte trafficking to the neuronal cell bodies of origin in the brain has been unclear. We sought to test the hypothesis that greater neuronal death induced by disparate forms of peripheral nerve injury would result in differential patterns of T cell infiltration and duration at the cell bodies of origin in the brain and that these measures would correlate with the magnitude of neuronal death over time and cumulative neuronal loss. To test this hypothesis, we compared the time course of CD3(+) T cell infiltration and neuronal death (assessed by CD11b(+) perineuronal microglial phagocytic clusters) following axonal crush versus axonal resection injuries, two extreme variations of facial nerve axotomy that result in mild versus severe neuronal loss, respectively, in the facial motor nucleus. We also quantified the number of facial motor neurons present at 49 days post-injury to determine whether differences in the levels of neuronal death between nerve crush and resection correlated with differences in cumulative neuronal loss. Between 1 and 7 days post-injury when levels of neuronal death were minimal, we found that the rate of accumulation and magnitude of the T cell response was similar following nerve crush and resection. Differences in the T cell response were apparent by 14 days post-injury when the level of neuronal death following resection was substantially greater than that seen in crush injury. For nerve resection, the peak of neuronal death at 14 days post-resection was followed by a maximal T cell response one week later at 21 days. Differences in the level of neuronal death between the two injuries across the time course tested reflected differences in cumulative neuronal loss at 49 days post-injury. Altogether, these data suggest that the trafficking of T cells to the injured FMN is dependent upon the severity of peripheral nerve injury and associated neuronal death.


Subject(s)
Brain/immunology , Facial Nerve Injuries/immunology , Facial Nerve Injuries/pathology , Neurons/immunology , T-Lymphocytes/immunology , Animals , Axotomy , Brain/pathology , CD3 Complex/metabolism , Cell Death/immunology , Chemotaxis, Leukocyte/immunology , Facial Nerve/pathology , Immunohistochemistry , Male , Mice , Mice, Inbred C57BL , Nerve Crush , Neurons/pathology , Retrograde Degeneration/immunology
12.
Brain Behav Immun ; 22(4): 528-37, 2008 May.
Article in English | MEDLINE | ID: mdl-18024079

ABSTRACT

We have previously shown that CD4(+) T helper (Th) 2 cells, but not Th1 cells, participate in the rescue of mouse facial motoneurons (FMN) from axotomy-induced cell death. Recently, a number of other CD4(+) T cell subsets have been identified in addition to the Th1 and Th2 effector subsets, including Th17, inducible T regulatory type 1 (Tr1), and naturally thymus-born Foxp3(+) regulatory (Foxp3(+) Treg) cells. These subsets regulate the nature of a T cell-mediated immune response. Th1 and Th17 cells are pro-inflammatory subsets, while Th2, Tr1, and Foxp3(+) Treg cells are anti-inflammatory subsets. Pro-inflammatory responses in the central nervous system are thought to be neurodestructive, while anti-inflammatory responses are considered neuroprotective. However, it remains to be determined if another CD4(+) T cell subset, other than the Th2 cell, develops after peripheral nerve injury and participates in FMN survival. In the present study, we used FACS analysis to determine the temporal frequency of Th1, Th17, Th2, Tr1 and Foxp3(+) Treg CD4(+) T cell subset development in C57BL/6 wild type mice after facial nerve transection at the stylomastoid foramen in the mouse. The results indicate that all of the known CD4(+) T cell subsets develop and expand in number within the draining lymph node, with a peak in number primarily at 7 days postoperative (dpo), followed by a decline at 9 dpo. In addition to the increase in subset frequency over time, FACS analysis of individual cells showed that the level of cytokine expressed per cell also increased for interferon-gamma (IFN-gamma), interleukin (IL)-10 and IL-17, but not IL-4. Additional control double-cytokine labeling experiments were done which indicate that, at 7dpo, the majority of cells indeed have committed to a specific phenotype and express only 1 cytokine. Collectively, our findings indicate for the first time that there is no preferential activation and expansion of any single CD4(+) T cell subset after peripheral nerve injury but, rather, that both pro-inflammatory and anti-inflammatory CD4(+) T cells develop.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Facial Nerve Diseases/immunology , Facial Nerve Injuries/immunology , Facial Nerve/immunology , T-Lymphocyte Subsets/immunology , Animals , Axotomy , CD4-Positive T-Lymphocytes/cytology , Cell Survival , Facial Nerve/cytology , Facial Nerve Diseases/pathology , Facial Nerve Injuries/pathology , Female , Immunophenotyping , Mice , Mice, Inbred C57BL , Motor Neurons/immunology , T-Lymphocyte Subsets/cytology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology
13.
Exp Neurol ; 208(1): 92-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17761165

ABSTRACT

Following facial nerve resection in the mouse, a substantial number of neurons reside in an atrophied state (characterized by cell shrinkage and decreased ability to uptake Nissl stain), which can be reversed by re-injury. The mechanisms mediating the reversal of neuronal atrophy remain unclear. Although T cells have been shown to prevent neuronal loss following peripheral nerve injury, it was unknown whether T cells play a role in mediating the reversal of axotomy-induced neuronal atrophy. Thus, we used a facial nerve re-injury model to test the hypothesis that the reversal of neuronal atrophy would be impaired in recombinase activating gene-2 knockout (RAG-2 KO) mice, which lack functional T and B cells. Measures of neuronal survival were compared in the injured facial motor nucleus (FMN) of RAG-2 KO and wild-type (WT) mice that received a resection of the right facial nerve followed by re-injury of the same nerve 10 weeks later ("chronic resection+re-injury") or a resection of the right facial nerve followed by sham re-injury of the same nerve 10 weeks later ("chronic resection+sham"). We recently demonstrated that prior exposure to neuronal injury elicited a marked increase in T cell trafficking indicative of a T cell memory response when the contralateral FMN was injured later in adulthood. We examined if such a T cell memory response would also occur in the current re-injury model. RAG-2 KO mice showed no reversal of neuronal atrophy whereas WT mice showed a robust response. The reversal of atrophy in WT mice was not accompanied by a T cell memory response. Although the number of CD4(+) and CD8(+) T cells in the injured FMN did not differ from each other, double-negative T cells appear to be recruited in response to neuronal injury. Re-injury did not result in increased expression of MHC2 by microglia. Our findings suggest that T cells may be involved in reversing the axotomy-induced atrophy of injured neurons.


Subject(s)
Facial Nerve Injuries/complications , Facial Nerve Injuries/pathology , Immunologic Deficiency Syndromes/complications , Microglia/pathology , Pons/pathology , T-Lymphocyte Subsets/pathology , Wounds, Penetrating/pathology , Animals , Atrophy , Axotomy , DNA-Binding Proteins/deficiency , Facial Nerve/surgery , Facial Nerve Injuries/immunology , Histocompatibility Antigens Class II/analysis , Immunologic Memory , Mice , Mice, Knockout , Microglia/immunology , Neurons/pathology , Postoperative Period , Reoperation , T-Lymphocytes/immunology , Wounds, Penetrating/complications , Wounds, Penetrating/immunology
14.
Neurosci Lett ; 417(2): 160-4, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17418948

ABSTRACT

IL-15 is a potent T cell chemoattractant, and this cytokine and its unique alpha subunits, IL-15R alpha, can modify immune cell expression of several T cell chemokines and their receptors. Facial nerve axotomy in mice leads to T cell migration across an intact blood-brain-barrier (BBB), and under certain conditions T cells can provide neuroprotection to injured neurons in the facial motor nucleus (FMN). Although chemokines and chemoattractant cytokines are thought to be responsible for T cell migration to the injured cell bodies, data addressing this question are lacking. This study tested the hypothesis that T cell homing to the axotomized FMN would be impaired in knockout (KO) mice with the IL-15 and IL-15R alpha genes deleted, and sought to determine if microglial responsiveness and motoneuron death are affected. Both IL-15KO and IL-15R alpha KO mice exhibited a marked reduction in CD3(+) T cells and had fewer MHC2(+) activated microglia in the injured FMN than their respective WT controls at day 14 post-axotomy. Although there was a relative absence of T cell recruitment into the axotomized FMN in both knockout strains, IL-15R alpha KO mice had five times more motoneuron death (characterized by perineuronal microglial clusters engulfing dead motoneurons) than their WT controls, whereas dead neurons in IL-15KO did not differ from their WT controls. Further studies are needed to dissect the mechanisms that underlie these observations (e.g., central vs. peripheral immune contributions).


Subject(s)
Chemotaxis, Leukocyte/immunology , Facial Nerve Injuries/immunology , Gliosis/immunology , Interleukin-15/immunology , Motor Neurons/immunology , Nerve Degeneration/immunology , Animals , Axotomy , Chemotaxis, Leukocyte/genetics , Facial Nerve/immunology , Facial Nerve/metabolism , Facial Nerve Injuries/genetics , Facial Nerve Injuries/metabolism , Female , Gliosis/genetics , Interleukin-15/genetics , Interleukin-15 Receptor alpha Subunit/genetics , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Microglia/metabolism , Motor Neurons/metabolism , Motor Neurons/pathology , Nerve Degeneration/genetics , Nerve Degeneration/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
J Neuroimmunol ; 181(1-2): 93-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17045343

ABSTRACT

We have previously demonstrated that CD4(+) T cells transiently rescue facial motoneurons (FMN) from axotomy-induced death in immunodeficient mice. Three subpopulations of motoneurons have been observed within the facial motor nucleus following axotomy: one that always survives axotomy (50%), one that is amenable to rescue from axotomy-induced death through the addition of neurotrophic factors or CD4(+) T cells (30-40%), and one that always dies after axotomy (10-15%). The objective of this study was to anatomically map the extent of axotomy-induced cell death and immune cell rescue in the facial nucleus to study the differential survival capabilities of each subpopulation. Wild-type (WT) mice, recombinase activating gene 2 knockout (RAG-2 KO) mice, and RAG-2 KO mice reconstituted with CD4(+) T cells were subjected to right facial nerve axotomy. At 4 weeks post-axotomy, topographical mapping of axotomy-induced cell death throughout the rostro-caudal extent of the facial nucleus was accomplished in accordance with previously published maps of the subnuclear arrangement of the facial neurons. The results indicate that all 3 subpopulations of FMN can be found in each of the subnuclear groups throughout the entire rostro-caudal extent of the facial nucleus. These data are discussed in context of recent work in amyotrophic lateral sclerosis, a fatal motoneuron disease.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Facial Nerve Injuries/immunology , Facial Nerve Injuries/pathology , Motor Neurons/cytology , Motor Neurons/immunology , Animals , Axotomy , CD4-Positive T-Lymphocytes/cytology , Cell Communication/immunology , Cell Death/immunology , Cell Survival/immunology , DNA-Binding Proteins/genetics , Facial Nerve/immunology , Facial Nerve/pathology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout
16.
Exp Neurol ; 201(1): 212-24, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16806176

ABSTRACT

The CD4(+) T lymphocyte has recently been found to promote facial motoneuron (FMN) survival after nerve injury. Signal Transducer and Activator of Transcription (STAT)4 and STAT6 are key proteins involved in the CD4(+) T cell differentiation pathways leading to T helper type (Th)1 and Th2 cell development, respectively. To determine which CD4(+) T cell subset mediates FMN survival, the facial nerve axotomy paradigm was applied to STAT4-deficient (-/-) and STAT6-/- mice. A significant decrease in FMN survival 4 weeks after axotomy was observed in STAT6-/- mice compared to wild-type (WT) or STAT4-/- mice. Reconstituting STAT6-/- mice with CD4(+) T cells obtained from WT mice promoted WT levels of FMN survival after injury. Furthermore, rescue of FMN from axotomy-induced cell death in recombination activating gene (RAG)-2-/- mice (lacking T and B cells) could be achieved only by reconstitution with CD4(+) T cells expressing functional STAT6 protein. To determine if either the Th1 cytokine, interferon-gamma (IFN-gamma) or the Th2 cytokine IL-4 is involved in mediating FMN survival, facial nerve axotomy was applied to IFN-gamma-/- and IL-4-/- mice. A significant decrease in FMN survival after axotomy occurred in IL-4-/- but not in IFN-gamma-/- mice compared to WT mice, indicating that IL-4 but not IFN-gamma is important for FMN survival after nerve injury. In WT mice, intracellular IFN-gamma vs. IL-4 expression was examined in CD4(+) T cells from draining cervical lymph nodes 14 days after axotomy, and substantial increase in the production of both CD4(+) effector T cell subsets was found. Collectively, these data suggest that STAT6-mediated CD4(+) T cell differentiation into the Th2 subset is necessary for FMN survival. A hypothesis relevant to motoneuron disease progression is presented.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Facial Nerve/physiopathology , Interleukin-4/physiology , Motor Neurons/physiology , STAT6 Transcription Factor/physiology , Animals , Axotomy , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , Cell Survival/immunology , Cell Survival/physiology , Facial Nerve/immunology , Facial Nerve/surgery , Facial Nerve Injuries/genetics , Facial Nerve Injuries/immunology , Facial Nerve Injuries/physiopathology , Genotype , Interferon-gamma/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/physiology , Interleukin-4/biosynthesis , Interleukin-4/genetics , Lymph Nodes/cytology , Lymph Nodes/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Motor Neurons/cytology , Motor Neurons/immunology , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/physiology , STAT6 Transcription Factor/genetics , Signal Transduction/physiology
17.
J Neuroimmune Pharmacol ; 1(1): 11-9, 2006 Mar.
Article in English | MEDLINE | ID: mdl-18040787

ABSTRACT

Motoneurons are large multipolar neurons with cell bodies located in the brainstem and spinal cord, and peripheral axons ending in neuromuscular junctions. Peripheral nerve damage, outside the blood-brain barrier (BBB), results in both retrograde changes centrally and anterograde changes along the length of the axon distal to the lesion site. Often, peripheral nerve damage is accompanied by motoneuron cell death, unless axon regrowth and target reconnection occur so that the target muscle can provide essential neurotrophic factors. It is essential that the motoneuron cell body survive during the process of reconnection so that the source for essential axon-rebuilding proteins is assure(of a fact)/ensured (results). A commonly used peripheral injury paradigm is that of facial nerve transection at its exit from the skull through the stylomastoid foramen so that nerve reconnection to the facial muscle tissue is permanently prevented. This model system allows for the study of the mechanisms responsible for maintaining facial motoneuron (FMN) cell body survival, without the complicating factor of axon regrowth. Injury to the nervous system results in an immune response that is either neuroprotective or neurodestructive. Findings suggest that FMN survival after facial nerve axotomy depends on the action of a CD4(+) T cell that is initially activated peripherally and subsequently reactivated centrally. This review will summarize what is known about the neural-immune players involved in FMN survival and repair, so that the pharmacological manipulation of this interaction will one day become evident for the clinical management of neurological situations.


Subject(s)
Facial Nerve Injuries/immunology , Motor Neurons/immunology , Nerve Regeneration/immunology , Wallerian Degeneration/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , Chemotaxis, Leukocyte/immunology , Facial Nerve Injuries/pathology , Humans , Lymphocyte Activation/immunology , Motor Neurons/pathology , Wallerian Degeneration/pathology
18.
Brain Behav Immun ; 19(2): 173-80, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15664790

ABSTRACT

Numerous studies have shown that motoneuron survival can be facilitated by neurotrophic factors (NTF) after injury. However, the ability of specific NTF to rescue facial motoneurons (FMN) from axotomy-induced death in immunodeficient mice has not been tested. Therefore, one goal of this study was to determine if brain-derived neurotrophic factor (BDNF), an NTF with a known ability to rescue FMN from axotomy-induced death, supports FMN from axotomy-induced death in recombinase activating gene-2 knockout (RAG-2 KO) mice that lack functional T and B lymphocytes. Nerve growth factor, which has been shown not to play a role in motoneuron survival, was used as a negative control. Brain derived neurotrophic factor treatment restored FMN survival to wild-type (WT) control levels 4 weeks post-operative (wpo) (80% +/- 1.9, 83% +/- 2.4, respectively). The second goal of this study was to begin to elucidate if CD4+ T cells produce NTF after facial nerve axotomy. Cervical lymph nodes were collected from WT mice 9 days post-operative, re-activated with anti-CD3 and supernatant collected 24 h later. Immediately after injury, the supernatant was administered to RAG-2 KO mice leading to an increase in FMN survival equivalent to WT controls (80% +/- 1.4, 84% +/- 2.1, respectively, 4 wpo). In addition, cervical lymph node supernatant treated with anti-BDNF attenuated FMN rescue in RAG-2 KO mice (62% +/- 3.3) 4 wpo. These data lend support to the hypothesis that CD4+ T cells produce NTF that support motoneuron survival before target reconnection occurs.


Subject(s)
Brain-Derived Neurotrophic Factor/physiology , Facial Nerve/physiology , Motor Neurons/cytology , Nerve Regeneration/immunology , Neuroimmunomodulation/physiology , Animals , Axotomy , B-Lymphocytes/immunology , Brain-Derived Neurotrophic Factor/genetics , Cell Survival/physiology , DNA-Binding Proteins/genetics , Facial Nerve/cytology , Facial Nerve Injuries/immunology , Facial Nerve Injuries/physiopathology , Female , Immunocompromised Host , Lymph Nodes/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes/immunology
19.
Brain Behav Immun ; 19(1): 12-9, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15581733

ABSTRACT

In the field of neuroimmunology, an emerging area of research involves the role that the immune system plays in neural injury and repair. Such immune:neural interactions may involve both neuroprotective and neurodestruction actions. To begin to address the compelling, and clinically relevant, issue of how the immune system impacts neural reparative processes, we combined the well described facial nerve injury paradigm, a simple neural injury model, with various immunodeficient mouse models, in order to delineate the contributing immune cells/factors involved in neural injury and repair. We have discovered a role for the CD4+ T cell in mediating facial motoneuron survival after facial nerve injury in the mouse. In this review, we present an overview of our work to date in this field and discuss future directions relevant to understanding key elements in the crosstalk between the immune:neural systems that develops subsequent to injury and/or trauma.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Survival/immunology , Facial Nerve Injuries/immunology , Facial Nerve/immunology , Motor Neurons/immunology , Animals , Axotomy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, SCID , Motor Neurons/pathology , Nerve Degeneration/immunology , Nerve Regeneration/immunology , Neuroimmunomodulation/physiology , Recovery of Function/immunology , Species Specificity
20.
J Neuroimmunol ; 156(1-2): 132-45, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15465604

ABSTRACT

Co-stimulatory factors are involved in different forms of brain pathology and play an important role in the activation of T-cells. In the current study, we explored the regulation of B7.2, a prominent member of the B7 family of costimulatory factors, in the facial motor nucleus (FMN) following facial axotomy and systemic application of lipopolysaccharide (LPS, endotoxin) using light and electron immunohistochemistry and cytokine-receptor-deficient mice. Facial axotomy led to a gradual increase of B7.2 immunoreactivity (IR) on microglial cell surface; similar effects were also observed following application of LPS, but both effects were not additive, suggesting overlapping or saturated signaling pathways. Some B7.2-IR was already present on activated microglia surrounding injured neurons at days 1-4 after injury, but became particularly intense during neuronal cell death, peaking at day 14. Previous studies revealed that these late microglial changes are accompanied by a strong increase in the expression of proinflammatory cytokines such as interleukin-1 beta (IL1beta) tumor necrosis factor-alpha (TNFalpha) and interferon gamma (IFNgamma) [J. Neurosci. 18 (1998a) 5804]. Here, deletion of the receptors for these cytokines-IL1R1, TNFR1 or TNFR2, but not IFNgammaR1-caused a strong and significant reduction in B7.2-IR in reactive microglial cells, compared with their wild type (WT) controls on the same genetic strain background, with a 31% decrease in IL1R1-/- , 39% in TNFR1-/- and 49% in TNFR2-/- mice. These data underscore the significance of IL1beta, TNFalpha and LPS, and their receptors, as potent inflammatory signals that regulate the cellular response in the injured brain as well as the interaction with the rapidly recruited immune system. The broad susceptibility of B7.2 regulation to a wide range of different inflammatory signals also points to its role as a sensor of molecular pathology, and a factor that plays an important accessory role in allowing and shaping the microglia/T-cell interaction in the injured central nervous system.


Subject(s)
Antigens, CD/physiology , Endotoxins/pharmacology , Facial Nerve Injuries/immunology , Facial Nerve Injuries/metabolism , Membrane Glycoproteins/physiology , Microglia/physiology , Receptors, Interleukin-1/physiology , Receptors, Tumor Necrosis Factor, Type II/physiology , Receptors, Tumor Necrosis Factor, Type I/physiology , Animals , Axotomy , B7-2 Antigen , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/metabolism , Phagocytes/drug effects , Phagocytes/metabolism , Phagocytes/physiology , Receptors, Interleukin-1 Type I
SELECTION OF CITATIONS
SEARCH DETAIL
...