Journal of Neuroinflammation

Published by Springer Nature

Online ISSN: 1742-2094

Articles


Table 1 List of the synthetic peptides spanning hPLP used in this study
Preparation of recombinant hΔPLP. (A) Scheme for the preparation and amplification of the synthetic hΔPLP DNA template, which is devoid of the hydrophobic putative transmembrane domains of hPLP, represented by the upper triangles, using the PCR overlap extension technique. (B) Alignment of deduced amino acid sequence of hΔPLP with full length native human PLP. The hΔPLP amino acid sequence derived from the DNA sequence of the pRSET/hΔPLP. The dot line in the hΔPLP sequence indicates the positions of the deleted hydrophobic domains. Underlined C and S represent the cysteine to serine substitutions introduced to minimizing incorrect refolding and to increase solubility of the expressed protein. (C) Coomassie Brilliant Blue-stained gel of the expressed and purified hΔPLP from bacteria; lane 1, bacterial host proteins before IPTG induction (20 μg of cellular proteins); lane 2, bacterial proteins after IPTG induction (20 μg cellular proteins; lane 3, hΔPLP (~1.5 μg) purified by metal chelate affinity chromatography on Ni2+-nitrilotriacetic acid agarose.
Mapping immunogenic DR15- and DQ6-associated PLP T-cell epitopes by immunization with individual overlapping peptides spanning hΔPLP. HLA-DRB1*1501-Tg, HLA-DQB1*0602-Tg and HLA-DRB1*1502-Tg (as control HLA-Tg line) mice were immunized (s.c.) each with an individual peptide (derived from native hPLP) of the overlapping peptides spanning hΔPLP. Ten days later, the primed LNC of immunized mice were analyzed ex-vivo for recall proliferative response to the immunizing peptide (0.5 - 5 μg/ml). Results expressed as stimulation index (S.I., mean cpm of antigen containing cultures/mean c.p.m of medium-containing cultures) are from one experiment with pooled draining LNC from two mice immunized with each individual peptide. Results are representative of three independent experiments.
Epitope-specificity of hΔPLP -primed T-cells. (A) Immunogenicity of hΔPLP in DRB1*1501- and DQB1*0602-Tg mice. HLA-Tg mice (two mice per Tg line) were immunized (s.c.) with 200 μg of hΔPLP in CFA. LNCs from draining LNs obtained 10 later were analyzed ex-vivo for their recall proliferative reponse to 5 μg/ml of hΔPLP or to hMOG (as control recombinant protein). Results (S.I.) are from pooled lymphocytes of each immunized HLA-Tg line. (B) HLA-class II restriction of hΔPLP primed LNCs. DRB1*1501- and DQB1*0602-hΔPLP-primed lymphocytes from (A) were analyzed in-vitro for their recall prolifrative response to hΔPLP in the absence or presence of blocking mAb specific for mouse CD4 (Leaf ™Purified anti-mouse CD4), CD8 (Leaf™ purified anti-mouse CD8a), or specific for human HLA-DR (L243) or HLA-DQ (L2) class-II molecules, or respective isotype control Abs.% inhibition - the S.I. calculated for the recall proliferative response in the presence of neutralizing Abs was divided by the S.I. of the response in the absence of neutralizing Abs (×100). (C, D) Antigenicity of mutated PLP175-194 peptide (PLP175-183S-194). DRB1*1501 (C) - and DQB1*0602 (D)- Tg mice (two mice/group) were immunized with 200 μg phPLP175-194 in CFA. Cells from draining LNs obtained 10 days later were analyzed ex-vivo for their recall proliferative response to 1 or 2.5 μg/ml of native phPLP175-194, PLP175-183S-194 mutant peptide, or hΔPLP. The proliferative response was measured as described in Methods. Results (S.I.) are from pooled lymphocytes of immunized HLA- Tg mice. (E, F) Ex-vivo analysis of immunodominant epitopes. Tg mice (two mice per HLA-Tg line) were immunized with 200 μg of in CFA at the flank, as described in Methods for the induction of EAE (protocol 2). Spleen cells were obtained from each of the immunized mice on day 14 after immunization and cultured in vitro in triplicates in the absence or presence of 1 or 2.5 μg/ml of each of the overlapping peptides (derived from native hPLP) spanning the hΔPLP, and in the presence of PLP175-183S-194 mutant peptide. Results are the mean S.I. of two individual spleens from mice immunized with same peptide in each HLA-Tg line. Results obtained from another independent experiment that was carried out in the same manner showed a similar pattern of reactivity to the overlapping peptides. *, and **, represent native PLP peptides, the cysteines of which were replaced by serine in the hΔPLP, where *, depicts peptides containing Cys within the nonameric core epitope for DRB1*1501 and/or DQB1*0602 molecule, but is not a major TCR or MHC binding residue, as predicted in silico: and **, depicts peptides containing Cys in the nonameric core epitope for DRB1*1501 and/or DQB1*0602 molecule that was predicted in silico to be a major TCR-contact or MHC-contact residue.
Clinical course of actively induced in HLA-DQB1*0602 Tg mice. EAE was induced by the indicated PLP peptide in CFA as described in methods (protocol 2): Pertussis toxin was administered immediately and 48 h after immunization. The mice received an identical booster immunization with PLP peptide in CFA on the flank one week later. I, incidence.

+2

DQB1*0602 rather than DRB1*1501 confers susceptibility to multiple sclerosis-like disease induced by proteolipid protein (PLP)
  • Article
  • Full-text available

February 2012

·

253 Reads

·

·

Chella S David

·

[...]

·

Avraham Ben-Nun
Multiple sclerosis (MS) is associated with pathogenic autoimmunity primarily focused on major CNS-myelin target antigens including myelin basic protein (MBP), proteolipidprotein (PLP), myelin oligodendrocyte protein (MOG). MS is a complex trait whereby the HLA genes, particularly class-II genes of HLA-DR15 haplotype, dominate the genetic contribution to disease-risk. Due to strong linkage disequilibrium in HLA-II region, it has been hard to establish precisely whether the functionally relevant effect derives from the DRB1*1501, DQA1*0102-DQB1*0602, or DRB5*0101 loci of HLA-DR15 haplotype, their combinations, or their epistatic interactions. Nevertheless, most genetic studies have indicated DRB1*1501 as a primary risk factor in MS. Here, we used 'HLA-humanized' mice to discern the potential relative contribution of DRB1*1501 and DQB1*0602 alleles to susceptibility to "humanized" MS-like disease induced by PLP, one of the most prominent and encephalitogenic target-antigens implicated in human MS. The HLA-DRB1*1501- and HLA-DQB1*0602-Tg mice (MHC-II(-/-)), and control non-HLA-DR15-relevant-Tg mice were immunized with a set of overlapping PLP peptides or with recombinant soluble PLP for induction of "humanized" MS-like disease, as well as for ex-vivo analysis of immunogenic/immunodominant HLA-restricted T-cell epitopes and associated cytokine secretion profile. PLP autoimmunity in both HLA-DR15-Tg mice was focused on 139-151 and 175-194 epitopes. Strikingly, however, the HLA-DRB1*1501-transgenics were refractory to disease induction by any of the overlapping PLP peptides, while HLA-DQB1*0602 transgenics were susceptible to disease induction by PLP139-151 and PLP175-194 peptides. Although both transgenics responded to both peptides, the PLP139-151- and PLP175-194-reactive T-cells were directed to Th1/Th17 phenotype in DQB1*0602-Tg mice and towards Th2 in DRB1*1501-Tg mice. While genome studies map a strong MS susceptibility effect to the region of DRB1*1501, our findings offer a rationale for potential involvement of pathogenic DQ6-associated autoimmunity in MS. Moreover, that DQB1*0602, but not DRB1*1501, determines disease-susceptibility to PLP in HLA-transgenics, suggests a potential differential, functional role for DQB1*0602 as a predisposing allele in MS. This, together with previously demonstrated disease-susceptibility to MBP and MOG in DRB1*1501-transgenics, also suggests a differential role for DRB1*1501 and DQB1*0602 depending on target antigen and imply a potential complex 'genotype/target antigen/phenotype' relationship in MS heterogeneity.
Download
Share

Combining nitric oxide release with anti-inflammatory activity preserves nigrostriatal dopaminergic innervation and prevents motor impairment in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease

November 2010

·

401 Reads

Current evidence suggests a role of neuroinflammation in the pathogenesis of Parkinson's disease (PD) and in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) model of basal ganglia injury. Reportedly, nonsteroidal anti-inflammatory drugs (NSAIDs) mitigate DAergic neurotoxicity in rodent models of PD. Consistent with these findings, epidemiological analysis indicated that certain NSAIDs may prevent or delay the progression of PD. However, a serious impediment of chronic NSAID therapy, particularly in the elderly, is gastric, renal and cardiac toxicity. Nitric oxide (NO)-donating NSAIDs, have a safer profile while maintaining anti-inflammatory activity of parent compounds. We have investigated the oral activity of the NO-donating derivative of flurbiprofen, [2-fluoro-α-methyl (1,1'-biphenyl)-4-acetic-4-(nitrooxy)butyl ester], HCT1026 (30 mg kg(-1) daily in rodent chow) in mice exposed to the parkinsonian neurotoxin MPTP. Ageing mice were fed with a control, flurbiprofen, or HCT1026 diet starting ten days before MPTP administration and continuing for all the experimental period. Striatal high affinity synaptosomal dopamine up-take, motor coordination assessed with the rotarod, tyrosine hydroxylase (TH)- and dopamine transporter (DAT) fiber staining, stereological cell counts, immunoblotting and gene expression analyses were used to assess MPTP-induced nigrostriatal DAergic toxicity and glial activation 1-40 days post-MPTP. HCT1026 was well tolerated and did not cause any measurable toxic effect, whereas flurbiprofen fed mice showed severe gastrointestinal side-effects. HCT1026 efficiently counteracted motor impairment and reversed MPTP-induced decreased synaptosomal [3H]dopamine uptake, TH- and DAT-stained fibers in striatum and TH+ neuron loss in substantia nigra pars compacta (SNpc), as opposed to age-matched mice fed with a control diet. These effects were associated to a significant decrease in reactive macrophage antigen-1 (Mac-1)-positive microglial cells within the striatum and ventral midbrain, decreased expression of iNOS, Mac-1 and NADPH oxidase (PHOX), and downregulation of 3-Nitrotyrosine, a peroxynitrite finger print, in SNpc DAergic neurons. Oral treatment with HCT1026 has a safe profile and a significant efficacy in counteracting MPTP-induced dopaminergic (DAergic) neurotoxicity, motor impairment and microglia activation in ageing mice. HCT1026 provides a novel promising approach towards the development of effective pharmacological neuroprotective strategies against PD.

Antibodies to the inositol 1,4,5-trisphosphate receptor type 1 (ITPR1) in cerebellar ataxia

December 2014

·

787 Reads

We report on a serum autoantibody associated with cerebellar ataxia. Immunohistochemical studies of sera from four patients referred for autoantibody testing revealed binding of high-titer (up to 1:5,000) IgG antibodies, mainly IgG1, to the molecular layer, Purkinje cell layer, and white matter on mouse, rat, porcine, and monkey cerebellum sections. The antibody bound to PC somata, dendrites, and axons, resulting in a binding pattern similar to that reported for anti-Ca/anti-ARHGAP26, but did not react with recombinant ARHGAP26. Extensive control studies were performed to rule out a broad panel of previously described paraneoplastic and non-paraneoplastic anti-neural autoantibodies. The characteristic binding pattern as well as double staining experiments suggested inositol 1,4,5-trisphosphate receptor type 1 (ITPR1) as the target antigen. Verification of the antigen included specific neutralization of the tissue reaction following preadsorption with ITPR1 (but not ARHGAP26) and a dot-blot assay with purified ITPR1 protein. By contrast, anti-ARHGAP26-positive sera did not bind to ITPR1. In a parallel approach, a combination of histoimmunoprecipitation and mass spectrometry also identified ITPR1 as the target antigen. Finally, a recombinant cell-based immunofluorescence assay using HEK293 cells expressing ITPR1 and ARHGAP26, respectively, confirmed the identification of ITPR1. Mutations of ITPR1 have previously been implicated in spinocerebellar ataxia with and without cognitive decline. Our findings suggest a role of autoimmunity against ITPR1 in the pathogenesis of autoimmune cerebellitis and extend the panel of diagnostic markers for this disease.

Figure 1: The mRNA expression profiles of differentially expressed genes. Both the upregulated (A) and downregulated (B) genes were selected randomly. Gel images on top show the polymerase chain reaction (PCR) product bands stained with ethidium bromide; the band intensities are also presented graphically below for clarity. Lane numbers 1 to 8 indicate sham control (lanes 1, 2, 5, and 6) and permanent middle cerebral artery occlusion (PMCAO) treatment (lanes 3, 4, 7, and 8), respectively. P indicates pituitary adenylate cyclase-activating polypeptide (PACAP) treatment; C is the control (minus PACAP). GAPDH and beta-actin genes were used a positive control (C). Semi-quantitative RT-PCR was performed as described in Methods, and the specific 3’-UTR primers are detailed in Additional file 2: Table S2.
Correction to Transcriptomics and proteomics analyses of the PACAP38 influenced ischemic brain in permanent middle cerebral artery occlusion model mice [Journal of Neuroinflammation (2013) 10, 18] 10.1186/1742-2094-10-18

January 2013

·

38 Reads

The Figure 2, X-axis description of each sample was inverted in the original publication [1]. Figure 2 The mRNA expression profiles of differentially expressed genes. Both the upregulated (A) and downregulated (B) genes were selected randomly. Gel images on top show the polymerase chain reaction (PCR) product bands stained with ethidium bromide; the band intensities are also presented graphically below for clarity. Lane numbers 1 to 8 indicate sham control (lanes 1, 2, 5, and 6) and permanent middle cerebral artery occlusion (PMCAO) treatment (lanes 3, 4, 7, and 8), respectively. P indicates pituitary adenylate cyclase-activating polypeptide (PACAP) treatment; C is the control (minus PACAP). GAPDH and beta-actin genes were used a positive control (C). Semi-quantitative RT-PCR was performed as described in Methods, and the specific 3'-UTR primers are detailed in Additional file 2: Table S2 With reference to corrected Figure 2, we have the following revised text. On Page 9, left column: lines 19-24 should read as - "Similarly, Il6, S100a5, Il22, Il1b, Igf1, and Ccl2 were highly expressed at 6 h in the PACAP-treated ischemic brain, whereas their expression level decreased at 24 h compared to the PMCAO effect alone (Figure 2). Fgf21, Pitpnc1, and Epha3 genes showed an increase in expression at 24 h over PMCAO alone (Figure 2)." On Page 11, right column: lines 16-19 should read as - "In the ischemic hemisphere at 24 h, the PACAP plus PMCAO expression level of Il6 was also reduced compared to the PMCAO plus saline control." We regret any inconvenience that this inaccuracy in Figure 2 and therein the figure legend, which could not be properly corrected at the proof stage, in the originally published manuscript might have caused.

Microdialysis catheter inserted into the brain parenchyma of an anaesthetized rat.
Representative examples of summed PET images showing uptake of (R)-[11C]PK11195 in rat brain at baseline and at one and ten days after TBI or sham.
Mean SUV of the image-derived injection dose (IDID) with error bars over time for baseline (--), sham (О) and TBI (▴) animals at 10 days.
Dialysate glutamate concentrations as a function of time following CCI (♦ n = 3) and sham (о n = 3) (* p < 0.05). Vertical lines represent SD. The arrow indicates baseline ECF glutamate release (sham).
Representative images of (immuno)histochemical staining of brain tissue (TBI and control) with ED-1, OX-6, GFAP, Perl's, and Fluoro-Jade B ten days after the microdialysis experiment.
Increased cerebral (R)-[(11)C]PK11195 uptake and glutamate release in a rat model of traumatic brain injury: A longitudinal pilot study

June 2011

·

609 Reads

The aim of the present study was to investigate microglia activation over time following traumatic brain injury (TBI) and to relate these findings to glutamate release. Sequential dynamic (R)-[(11)C]PK11195 PET scans were performed in rats 24 hours before (baseline), and one and ten days after TBI using controlled cortical impact, or a sham procedure. Extracellular fluid (ECF) glutamate concentrations were measured using cerebral microdialysis. Brains were processed for histopathology and (immuno)-histochemistry. Ten days after TBI, (R)-[(11)C]PK11195 binding was significantly increased in TBI rats compared with both baseline values and sham controls (p < 0.05). ECF glutamate values were increased immediately after TBI (27.6 ± 14.0 μmol·L(-1)) as compared with the sham procedure (6.4 ± 3.6 μmol·L(-1)). Significant differences were found between TBI and sham for ED-1, OX-6, GFAP, Perl's, and Fluoro-Jade B. Increased cerebral uptake of (R)-[(11)C]PK11195 ten days after TBI points to prolonged and ongoing activation of microglia. This activation followed a significant acute posttraumatic increase in ECF glutamate levels.

Neuronal RING finger protein 11 (RNF11) regulates canonical NF-κB signaling

April 2012

·

182 Reads

The RING domain-containing protein RING finger protein 11 (RNF11) is a member of the A20 ubiquitin-editing protein complex and modulates peripheral NF-κB signaling. RNF11 is robustly expressed in neurons and colocalizes with a population of α-synuclein-positive Lewy bodies and neurites in Parkinson disease patients. The NF-κB pathway has an important role in the vertebrate nervous system, where the absence of NF-κB activity during development can result in learning and memory deficits, whereas chronic NF-κB activation is associated with persistent neuroinflammation. We examined the functional role of RNF11 with respect to canonical NF-κB signaling in neurons to gain understanding of the tight association of inflammatory pathways, including NF-κB, with the pathogenesis of neurodegenerative diseases. Luciferase assays were employed to assess NF-κB activity under targeted short hairpin RNA (shRNA) knockdown of RNF11 in human neuroblastoma cells and murine primary neurons, which suggested that RNF11 acts as a negative regulator of canonical neuronal NF-κB signaling. These results were further supported by analyses of p65 translocation to the nucleus following depletion of RNF11. Coimmunoprecipitation experiments indicated that RNF11 associates with members of the A20 ubiquitin-editing protein complex in neurons. Site-directed mutagenesis of the myristoylation domain, which is necessary for endosomal targeting of RNF11, altered the impact of RNF11 on NF-κB signaling and abrogated RNF11's association with the A20 ubiquitin-editing protein complex. A partial effect on canonical NF-κB signaling and an association with the A20 ubiquitin-editing protein complex was observed with mutagenesis of the PPxY motif, a proline-rich region involved in Nedd4-like protein interactions. Last, shRNA-mediated reduction of RNF11 in neurons and neuronal cell lines elevated levels of monocyte chemoattractant protein 1 and TNF-α mRNA and proteins, suggesting that NF-κB signaling and associated inflammatory responses are aberrantly regulated in the absence of RNF11. Our findings support the hypothesis that, in the nervous system, RNF11 negatively regulates canonical NF-κB signaling. Reduced or functionally compromised RNF11 could influence NF-κB-associated neuronal functions, including exaggerated inflammatory responses that may have implications for neurodegenerative disease pathogenesis and progression.

Evaluating the role of IL-11, a novel cytokine in the IL-6 family, in a mouse model of spinal cord injury

June 2012

·

141 Reads

Spinal cord injury (SCI) is a devastating condition with substantial functional and social morbidity. Previous research has established that the neuroinflammatory response plays a significant role in cord damage post-SCI. However, global immunosuppressive therapies have demonstrated mixed results. As a result, more specific therapies modulating inflammation after injury are needed. In this regard, research into cytokine signaling has demonstrated that cytokines of the gp130 family including IL-6 and leukemia inhibitory factor (LIF) play key roles in mediating damage to the spinal cord. Since members of the gp130 family all share a common signal transduction pathway via the JAK/STAT system, we performed the first study of a relatively new member of the gp130 family, IL-11, in SCI. A validated clip-compression mouse model of SCI was used to assess for temporal changes in expression of IL-11 and its receptor, IL-11Rα, post-SCI. To elucidate the role of IL-II in the pathophysiology of SCI, we compared differences in locomotor recovery (Basso Mouse Score; CatWalk), electrophysiological spinal cord signaling, histopathology, and the acute inflammatory neutrophil response in IL-11Rα knockouts with littermate wild-type C57BL/6 mice. We found an increase in gene expression of IL-11 in the spinal cord to a peak at twenty-four hours post-SCI with increases in IL-11Rα gene expression, peaking at seven days post-SCI. In spite of clear changes in the temporal expression of both IL-11 and its receptor, we found that there were no significant differences in motor function, electrophysiological signaling, histopathology, or neutrophil infiltration into the spinal cord between wild-type and knockout mice. This is the first study to address IL-11 in SCI. This study provides evidence that IL-11 signaling may not play as significant a role in SCI as other gp130 cytokines, which will ideally guide future therapy design and the signaling pathways those therapies target.

5, 8, 11, 14-eicosatetraynoic acid suppresses CCL2/MCP-1 expression in IFN-γ-stimulated astrocytes by increasing MAPK phosphatase-1 mRNA stability

February 2012

·

148 Reads

The peroxisome proliferator-activated receptor (PPAR)-α activator, 5,8,11,14-eicosatetraynoic acid (ETYA), is an arachidonic acid analog. It is reported to inhibit up-regulation of pro-inflammatory genes; however, its underlying mechanism of action is largely unknown. In the present study, we focused on the inhibitory action of ETYA on the expression of the chemokine, CCL2/MCP-1, which plays a key role in the initiation and progression of inflammation. To determine the effect of ETYA, primary cultured rat astrocytes and microglia were stimulated with IFN-γ in the presence of ETYA and then, expression of CCL2/MCP-1 and MAPK phosphatase (MKP-1) were determined using RT-PCR and ELISA. MKP-1 mRNA stability was evaluated by treating actinomycin D. The effect of MKP-1 and human antigen R (HuR) was analyzed by using specific siRNA transfection system. The localization of HuR was analyzed by immunocytochemistry and subcellular fractionation experiment. We found that ETYA suppressed CCL2/MCP-1 transcription and secretion of CCL2/MCP-1 protein through up-regulation of MKP-1mRNA levels, resulting in suppression of c-Jun N-terminal kinase (JNK) phosphorylation and activator protein 1 (AP1) activity in IFN-γ-stimulated brain glial cells. Moreover, these effects of ETYA were independent of PPAR-α. Experiments using actinomycin D revealed that the ETYA-induced increase in MKP-1 mRNA levels reflected an increase in transcript stability. Knockdown experiments using small interfering RNA demonstrated that this increase in MKP-1 mRNA stability depended on HuR, an RNA-binding protein known to promote enhanced mRNA stability. Furthermore, ETYA-induced, HuR-mediated mRNA stabilization resulted from HuR-MKP-1 nucleocytoplasmic translocation, which served to protect MKP-1 mRNA from the mRNA degradation machinery. ETYA induces MKP-1 through HuR at the post-transcriptional level in a receptor-independent manner. The mechanism revealed here suggests eicosanoids as potential therapeutic modulators of inflammation that act through a novel target.

M. corti infection elevates mRNA expression of TLRs 11–13 in brain. Total RNA was isolated from infected and mock infected control mouse brains at 1 wk, 3 wk, and 6 wk p.i. using Trizol reagent. Isolated RNA was reversed transcribed to cDNA by using random primers. Levels of TLR specific RNA and housekeeping gene 18S in these samples were measured by Real Time PCR analysis as described below using SYBR green as the detection dye. TLR specific mRNA levels were normalized to the mRNA level of the housekeeping gene 18S in the same sample and expressed in arbitrary units. The normalized values corresponding to the mRNA expression for each TLR gene are shown in the y-axis and represent the mean of three independent experiments. The data obtained were compared using a Student's t-test.
Expression of TLR11 in brains of mock infected control and NCC mice analyzed by immunofluorescence microscopy. Balb/c mice were infected intracranially with M. corti and sacrificed at various times p.i. Brain crysections were analyzed for expression of TLR11 and cell specific markers using fluorochrome conjugated antibodies. Nuclear staining DAPI is blue. (A) Mock infected control stained with TLR11 specific antibody (400×). (B) M. corti infection at 1 wk showing TLR11 positive staining in neurons (400×). (C) M. corti infection at 3 wk when maximum TLR11 expression was observed (400×). (D) M. corti infection at 6 wk showing TLR11 positive staining (400×). (E) Mock infected control mice stained with TLR11 and neuron specific antibody (Neun) (400×). The insert e' represents a 2× magnification of a selected area depicting TLR11 expression (Red) on neurons. (F) M. corti infection at 3 wk showing TLR11 positive neurons, merged image (yellow/orange; 400×). (G) Mock control mice stained with anti-TLR11 and anti-CD11b antibody (400×). (H) M. corti infection at 1 wk showing TLR11 and CD11b double positive cells, merged image (yellow/orange; 400×). Insert h' is a 2× magnification of a selected area to better illustrate TLR11 and CD11b double positive staining (I) The relative levels of TLR11 expression in mock and M. corti infected animals was calculated as described in experimental procedures.
Expression of TLR12 in brains of mock infected control and NCC mice analyzed by immunofluorescence microscopy. Balb/c mice were infected intracranially with M. corti and sacrificed at various times p.i. Brain cryosections were analyzed for expression of TLR12 and cell specific markers using fluorochrome conjugated antibodies. Nuclear staining DAPI is blue. (A) Mock infected control stained with TLR12 specific antibody (400×). (B) M. corti infection at 1 wk showing TLR12 positive staining (400×). (C) M. corti infection at 3 wk when maximum TLR12 expression was observed (400×). (D) M. corti infection at 6 wk showing TLR12 positive staining (400×). (E) Mock control mice stained with TLR12 and neuron specific antibody (Neun) (400×). The insert e' represents a 2× magnification of a selected area depicting TLR12 expression (Red) on neurons. (F) M. corti infection at 3 wk showing TLR12 positive neurons, merged image (yellow/orange; 400×). (G) The relative levels of TLR12 expression in mock and M. corti infected animals was calculated as described in experimental procedures.
Expression of TLR13 in brains of mock infected control and NCC mice analyzed by immunofluorescence microscopy. Balb/c mice were infected intracranially with M. corti and sacrificed at various times p.i. Brain cryosections were analyzed for expression of TLR13 and cell specific markers using fluorochrome conjugated antibodies. Nuclear staining DAPI is blue. (A) Mock infected control stained with TLR13 specific antibody (400×). (B) M. corti infection at 1 wk showing TLR13 positive staining in neurons (400×). (C) M. corti infection at 3 wk when maximum TLR13 expression was observed (400×). (D) M. corti infection at 6 wk showing TLR13 positive staining (400×). (E) Mock control mice stained with TLR13 and neuron specific antibody (Neun) (400×). The insert e' represents a 2× magnification of a selected area depicting TLR13 expression (Red) on neurons. (F) M. corti infection at 3 wk showing TLR13 positive neurons, merged image (yellow/orange; 400×). (G) M. corti infection at 3 wk showing TLR13 positive astrocytes and its processes, merged image (yellow/orange; 400×). The arrow points to the pial vessel expressing TLR13. (H) The relative levels of TLR13 expression in mock and M. corti infected animals was calculated as described in experimental procedures.
Expression and distribution of Toll-like receptors 11–13 in the brain during murine neurocysticercosis

January 2009

·

106 Reads

The functions of Toll-like receptors (TLRs) 11-13 in central nervous system (CNS) infections are currently unknown. Using a murine model of neurocysticercosis, we investigated the expression and distribution of TLRs 11-13 by using both gene specific real-time PCR analysis and in situ immunofluorescence microscopy in both control and neurocysticercosis brains. In the mock infected brain, mRNAs of TLRs 11-13 were constitutively expressed. Parasite infection caused an increase of both mRNAs and protein levels of all three TLRs by several fold. All three TLR proteins were present in both CNS and immune cell types. Among them TLR13 was expressed the most in terms of number of positive cells and brain areas expressing it, followed by TLR11 and TLR12 respectively. Among the nervous tissue cells, TLRs 11-13 protein levels appeared highest in neurons. However, TLR13 expression was also present in ependymal cells, endothelial cells of pial blood vessels, and astrocytes. In contrast, infiltrating CD11b and CD11c positive myeloid cells predominantly produced TLR11 protein, particularly early during infection at 1 wk post infection (approximately 50% cells). TLRs 12 and 13 proteins were present on approximately 5% of infiltrating immune cells. The infiltrating cells positive for TLRs 11-13 were mostly of myeloid origin, CD11b+ cells. This report provides a comprehensive analysis of the expression of TLRs 11-13 in normal and parasite infected mouse brains and suggests a role for them in CNS infections.

Table 2 Binding potential values of 11 C-[R]-PK11195 in various brain regions Binding potential (mean ± SD (95% CI))
Normalized time activity curves. Normalized time activity curves from various brain regions showing rapid 11C-[R]-PK11195 uptake followed by steady washout. Triangles: adults (≥18 years of age); squares: children (<18 years of age); SUV: standard uptake value.
Images of 11C-[R]-PK11195 brain positron emissiontomography scan. PK11195 brain PET scan showing PK11195 brain distribution in a child (upper row) and healthy normal adult (lower row). The overall brain PK11195 uptake is relatively low, suggested by much lower cortical PK11195 uptake compared to subcortical structures, and the pattern of PK11195 distribution is largely similar in both children and adults, although PK11195 uptake appears to be visually slightly higher in adults, particularly in the thalamus and midbrain. Highest PK11195 uptake is noted in the pituitary gland and midbrain, followed by thalamus, basal ganglia, occipital cortex, hippocampus and cerebellum. The rest of the cerebral cortex is showing low PK11195 uptake with minimal uptake in white matter. BG: basal ganglia; MB: midbrain; Cer: cerebellum; Cg: cingulum; F: frontal cortex; Hip: hippocampus; P: parietal cortex; Pit: pituitary gland; T: temporal cortex; Th: thalamus; R: right side; L: left side.
Effect of age on brain 11C-[R]-PK11195 uptake. Correlation analysis between age and PK11195 uptake (standardized uptake value; SUV) showed a significant age-related increase in brain PK11195 uptake.
Result of voxel-based analysis of 11C-[R]-PK11195 positron emission tomography scans. Voxel-wise statistical parametric mapping analysis of PK11195 standard uptake value images, carried out between adults and children, showed significantly higher PK11195 uptake in adults in the thalamus and midbrain region only.
Evaluation of age-related changes in translocator protein (TSPO) in human brain using 11C-[R]-PK11195 PET

October 2012

·

200 Reads

Background We studied the distribution and expression of translocator protein in the human brain using 11C-[R]-PK-11195 positron emission tomography (PK11195 PET) and evaluated age-related changes. Methods A dynamic PK11195 PET scan was performed in 15 normal healthy adults (mean age: 29 ±8.5 years (range: 20 to 49); 7 males) and 10 children (mean age: 8.8 ±5.2 years (range: 1.2 to 17); 5 males), who were studied for potential neuroinflammation but showed no focally increased PK11195 binding. The PET images were evaluated by calculating standard uptake values and regional binding potential, based on a simplified reference region model, as well as with a voxel-wise analysis using statistical parametric mapping. Results PK11195 uptake in the brain is relatively low, compared with the subcortical structures, and symmetrical. The overall pattern of PK11195 distribution in the brain does not change with age. PK11195 uptake was lowest in the frontal-parietal-temporal cortex and highest in the pituitary gland, midbrain, thalamus, basal ganglia, occipital cortex, hippocampus and cerebellum, in descending order. White matter showed negligible PK11195 uptake. Overall, brain PK11195 uptake increased with age, with midbrain and thalamus showing relatively higher increases with age compared with other brain regions. Conclusions The brain shows low PK11195 uptake, which is lower in the cortex and cerebellum compared with subcortical structures, suggesting a low level of translocator protein expression. There is no hemispheric asymmetry in PK11195 uptake and the overall pattern of PK11195 distribution in the brain does not change with age. However, brain PK11195 uptake increases with age, with the thalamus and midbrain showing relatively higher increases compared with other brain regions. This increase in uptake suggests an age-related increase in translocator protein expression or the number of cells expressing these receptors or both.

MicroRNA-124 as a novel treatment for persistent hyperalgesia

June 2012

·

280 Reads

Chronic pain is often associated with microglia activation in the spinal cord. We recently showed that microglial levels of the kinase G protein-coupled receptor kinase (GRK)2 are reduced in models of chronic pain. We also found that mice with a cell-specific reduction of around 50% in GRK2 level in microglia/macrophages (LysM-GRK2+/- mice) develop prolonged inflammatory hyperalgesia concomitantly with ongoing spinal microglia/macrophage activation. The microRNA miR-124 is thought to keep microglia/macrophages in brain and spinal cord in a quiescent state. In the present study, we investigated the contribution of miR-124 to regulation of hyperalgesia and microglia/macrophage activation in GRK2-deficient mice. In addition, we investigated the effect of miR-124 on chronic inflammatory and neuropathic pain in wild-type (WT) mice. Hyperalgesia was induced by intraplantar IL-1β in WT and LysM-GRK2+/- mice. We determined spinal cord microglia/macrophage miR-124 expression and levels of pro-inflammatory M1 and anti-inflammatory M2 activation markers. The effect of intrathecal miR-124 treatment on IL-1β-induced hyperalgesia and spinal M1/M2 phenotype, and on carrageenan-induced and spared nerve injury-induced chronic hyperalgesia in WT mice was analyzed. Transition from acute to persistent hyperalgesia in LysM-GRK2+/- mice is associated with reduced spinal cord microglia miR-124 levels. In our LysM-GRK2+/- mice, there was a switch towards a pro-inflammatory M1 phenotype together with increased pro-inflammatory cytokine production. Intrathecal administration of miR-124 completely prevented the transition to persistent pain in response to IL-1β in LysM-GRK2+/- mice. The miR-124 treatment also normalized expression of spinal M1/M2 markers of LysM-GRK2+/- mice. Moreover, intrathecal miR-124 treatment reversed the persistent hyperalgesia induced by carrageenan in WT mice and prevented development of mechanical allodynia in the spared nerve injury model of chronic neuropathic pain in WT mice. We present the first evidence that intrathecal miR-124 treatment can be used to prevent and treat persistent inflammatory and neuropathic pain. In addition, we show for the first time that persistent hyperalgesia in GRK2-deficient mice is associated with an increased ratio of M1/M2 type markers in spinal cord microglia/macrophages, which is restored by miR-124 treatment. We propose that intrathecal miR-124 treatment might be a powerful novel treatment for pathological chronic pain with persistent microglia activation.

Maternal immune activation by poly(I:C) induces expression of cytokines IL-1β and IL-13, chemokine MCP-1 and colony stimulating factor VEGF in fetal mouse brain

April 2012

·

311 Reads

Maternal viral infection during pregnancy is associated with an increase in the incidence of psychiatric disorders with presumed neurodevelopmental origin, including autism spectrum disorders and schizophrenia. The enhanced risk for developing mental illness appears to be caused by deleterious effects of innate immune response-associated factors on the development of the central nervous system, which predispose the offspring to pathological behaviors in adolescence and adulthood. To identify the immune response-associated soluble factors that may affect central nervous system development, we examined the effect of innate immune response activation by polyriboinosinic-polyribocytidylic acid (poly(I:C)), a synthetic analogue of viral double-stranded RNA, on the expression levels of pro- and anti-inflammatory cytokines, chemokines and colony stimulating factors in fetal and postnatal mouse brain 6 h and 24 h after treatment. C57BL/6J pregnant mice (gestational day 16) or newborn mice (postnatal day 4) received a single intraperitoneal injection of the synthetic analogue of viral double-stranded RNA poly(I:C) (20 mg/kg). Thirty-two immune response-associated soluble factors, including pro- and anti-inflammatory cytokines, chemokines and colony stimulating factors, were assayed 6 h and 24 h after poly(I:C) injection using multiplexed bead-based immunoassay (Milliplex Map) and processed in a Luminex 100 IS instrument. Maternal exposure to poly(I:C) at gestational day 16 induced a significant increase in cytokines interleukin (IL)-1β, IL-7 and IL-13; chemokines monocyte chemoattractant protein 1 (MCP-1), macrophage inflammatory protein (MIP)-1α, interferon gamma-induced protein (IP)-10 and monokine induced by IFN-gamma (MIG); and in the colony stimulating factor vascular endothelial growth factor (VEGF) in the fetal brain. IL-1β showed the highest concentration levels in fetal brains and was the only cytokine significantly up-regulated 24 h after maternal poly(I:C) injection, suggesting that IL-1β may have a deleterious impact on central nervous system development. In contrast, poly(I:C) treatment of postnatal day 4 pups induced a pronounced rise in chemokines and colony stimulating factors in their brains instead of the pro-inflammatory cytokine IL-1β. This study identified a significant increase in the concentration levels of the cytokines IL-1β and IL-13, the chemokine MCP-1 and the colony stimulating factor VEGF in the developing central nervous system during activation of an innate immune response, suggesting that these factors are mediators of the noxious effects of maternal immune activation on central nervous system development, with potential long-lasting effects on animal behavior.

Representative disease course of SJL mice. 5 mice were immunized with PLPp/CFA with addition of pertussis toxin. 5 mice were immunized with PLPp and CpG in IFA. The mean clinical EAE score is shown with standard deviation. The differences in disease scores between the two groups on days 10, 11 and 12 were highly significant (p < 0.001).
Frequencies of IFN-γ (A), IL-2 (B) and IL-17 (C) producing cells in spleen cells of animals immunized with PLPp in CpG/IFA after stimulation with PLPp, CpG or combined activation. Spleens were isolated on day 14 after immunization, and single cell suspensions were made as described in Materials and Methods. Cells were plated at a density of 1.000.000 cells per well. The cumulative data of 7 animals tested individually in two independent experiments are shown. Measurements were conducted in triplicate wells each. Each data point indicates one individual animal, lines represent the mean. p values are given in Section 3.
Frequencies of IFN-γ (A), IL-2 (B) and IL-17 (C) producing cells in spleens of animals immunized with PLPp in CFA and pertussis toxin after stimulation with PLPp, CpG or combined activation. Spleens were isolated on day 12 after immunization, and single cell suspensions were made as described in Materials and Methods. Cells were plated at a density of 1.000.000 cells per well. The cumulative data of 7 animals tested individually in two independent experiments are shown. Measurements were conducted in triplicate wells each. Each data point indicates one individual animal, lines represent the mean. p values are given in Section 3.
Frequencies of IFN-γ (A), IL-2 (B) and IL-17 (C) producing PLPp specific cells in spinal cords of animals immunized with PLPp in CFA and pertussis toxin after stimulation with PLPp, CpG or combined activation. Spinal cords were isolated on day 12 after immunization, and single cell suspensions were made as described in Materials and Methods. Cells were plated at a density of 100.000 cells per well. Raw data is adjusted to 1.000.000 cells per well for a direct comparison with frequencies of cytokine-producing spleen cells. Each bar represents two independent experiments with pooled spinal cord cultures of totally 7 mice (4 mice in the first experiment, 3 mice in the second experiment) due to the small number of mononuclear cells which can be recovered the CNS. Measurements were conducted in duplicate wells each. p values are given in Section 3.
Adoptive transfer of PLPp-stimulated and PLPp/CpG-stimulated PLPp-specific T cells into SJL mice with actively induced EAE. Female SJL mice injected with PLPp/CFA and PTX as described above were (either at day 9 or day 10) injected with cells re-stimulated in vitro either with PLPp (A) or PLPp/CpG (B). A control group received PBS at day 9 or day 10, respectively (C). The disease course of all animals is depicted individually. Further details are described in the results section.
Alteration of T cell cytokine production in PLPp-139-151-induced EAE in SJL mice by an immunostimulatory CpG Oligonucleotide

May 2011

·

108 Reads

Experimental autoimmune encephalomyelitis (EAE) is--in certain aspects--regarded as an animal model of the human CNS autoimmune disease multiple sclerosis (MS). While in EAE CNS-autoantigen-specific immunity is induced in a defined way, the initial processes leading to CNS autoimmunity in humans are so far unknown. Despite essential restrictions, which exist regarding the interpretation of EAE data towards MS, EAE might be a useful model to study certain basic aspects of CNS autoimmunity. Studies in MS have demonstrated that established autoimmune pathology can be critically influenced by environmental factors, in particular viral and bacterial infections. To investigate this interaction, EAE as an instrument to study CNS autoimmunity under defined conditions appears to be a suitable experimental tool. For this reason, we here investigated the influence of the Toll-like-receptor (TLR) ligand CpG oligonucleotide (CpG) on already established CNS autoimmunity in murine proteolipid protein (PLP)-induced EAE in SJL mice. CpG were found to co-stimulate PLPp-specific IFN-γ production in the peripheral immune system and in the CNS. However, CpG induced Interleukin (IL)-17 production in the inflamed CNS both alone and in combination with additional PLPp stimulation. These findings might indicate a mechanism by which systemic infections and the microbial stimuli associated with them may influence already existing CNS autoimmune pathology.

The cytokine tumor necrosis factor-like weak inducer of apoptosis and its receptor fibroblast growth factor-inducible 14 have a neuroprotective effect in the central nervous system

March 2012

·

92 Reads

Cerebral cortical neurons have a high vulnerability to the harmful effects of hypoxia. However, the brain has the ability to detect and accommodate to hypoxic conditions. This phenomenon, known as preconditioning, is a natural adaptive process highly preserved among species whereby exposure to sub-lethal hypoxia promotes the acquisition of tolerance to a subsequent lethal hypoxic injury. The cytokine tumor necrosis factor-like weak inducer of apoptosis (TWEAK) and its receptor fibroblast growth factor-inducible 14 (Fn14) are found in neurons and their expression is induced by exposure to sub-lethal hypoxia. Accordingly, in this work we tested the hypothesis that the interaction between TWEAK and Fn14 induces tolerance to lethal hypoxic and ischemic conditions. Here we used in vitro and in vivo models of hypoxic and ischemic preconditioning, an animal model of transient middle cerebral artery occlusion and mice and neurons genetically deficient in TWEAK, Fn14, or tumor necrosis factor alpha (TNF-α) to investigate whether treatment with recombinant TWEAK or an increase in the expression of endogenous TWEAK renders neurons tolerant to lethal hypoxia. We used enzyme-linked immunosorbent assay to study the effect of TWEAK on the expression of neuronal TNF-α, Western blot analysis to investigate whether the effect of TWEAK was mediated by activation of mitogen-activated protein kinases and immunohistochemical techniques and quantitative real-time polymerase chain reaction analysis to study the effect of TWEAK on apoptotic cell death. We found that either treatment with recombinant TWEAK or an increase in the expression of TWEAK and Fn14 induce hypoxic and ischemic tolerance in vivo and in vitro. This protective effect is mediated by neuronal TNF-α and activation of the extracellular signal-regulated kinases 1 and 2 pathway via phosphorylation and inactivation of the B-cell lymphoma 2-associated death promoter protein. Our work indicate that the interaction between TWEAK and Fn14 triggers the activation of a cell signaling pathway that results in the induction of tolerance to lethal hypoxia and ischemia. These data indicate that TWEAK may be a potential therapeutic strategy to protect the brain from the devastating effects of an ischemic injury.

Microglia and neuroinflammation: a pathological perspective <http://PM:15285801

August 2004

·

326 Reads

Microglia make up the innate immune system of the central nervous system and are key cellular mediators of neuroinflammatory processes. Their role in central nervous system diseases, including infections, is discussed in terms of a participation in both acute and chronic neuroinflammatory responses. Specific reference is made also to their involvement in Alzheimer's disease where microglial cell activation is thought to be critically important in the neurodegenerative process.

Figure 1: Decrease of miR-15b and miR-16 expression in hyperglycemia and transfection-induced fold changes. (A) Fold changes of microRNA (miR)-15b and miR-16 expression are shown. After 3 days of retinal endothelial cell (REC) culture in a high-glucose (25 mM) medium, the expression of both miR-15b and miR-16 was reduced (0.6- and 0.2-fold change, respectively) compared to that of the normal-glucose (NG; 5 mM) group. (B, C) Transfection-induced fold changes of miR-15b and miR-16 expression in REC. REC were transfected with mimics (30 nM of final concentration) of miR-15b and/or miR-16 to increase the level of expression in a hyperglycemic condition. Approximately 167- and 54-fold increases (miR-15b and miR-16, respectively) were detected following transfection with the mimic, compared to the high glucose (HG) control. The y-axis is a logarithmic scale. *P < 0.05 versus HG, N = 3; data are mean ± S.E.M.
Table 1 Fold changes of miR-15b and miR-16 expression after transfection with miR-mimics
Figure 4 Changes of insulin receptor (Tyr1150/1151) phosphorylation and IGFBP-3 levels. Western blot results for phosphorylated insulin receptor (IR) (Tyr1150/1151) to total IR (A) and insulin-like growth factor binding protein-3 (IGFBP-3) (B) on retinal endothelial cells (REC) cultured in normal glucose (NG, 5 mM) or high glucose (HG, 25 mM) and transfected groups. (A) The levels of IR Tyr1150/1151 phosphorylation were increased by the overexpression of miR-15b and miR-16 in REC under hyperglycemic conditions. (B) miR-15b significantly increased the levels of IGFBP-3 in REC in hyperglycemia. A representative blot is shown. # P < 0.05 versus NG, *P < 0.05 versus HG, ## P < 0.01 versus NG, **P < 0.01 versus HG, N = 6 ~ 7; data are mean ± S.E.M.
Figure 5 Effects of miR-15b and miR-16 on Akt phosphorylation and cleaved caspase 3 in hyperglycemia. REC were cultured in normal glucose (NG, 5 mM) or high glucose (HG, 25 mM). (A) Western blot results for phosphorylated Akt to total Akt on retinal endothelial cells (REC). Overexpression of microRNA (miR)-15b and miR-16 increased the levels of Akt phosphorylation, which was reduced in the control HG condition. A representative blot is shown. (B) Enzyme-linked immunosorbent assay (ELISA) results for cleaved caspase 3. miR-16 and miR-15b/16 significantly reduced the level of cleaved caspase 3 in hyperglycemia. # P < 0.05 versus NG, *P < 0.05 versus HG, N = 4; data are mean ± S.E.M.
miR-15b/16 protects primary human retinal microvascular endothelial cells against hyperglycemia-induced increases in tumor necrosis factor alpha and suppressor of cytokine signaling 3

December 2015

·

217 Reads

Mechanisms underlying the pathology of diabetic retinopathy are still not completely understood. Increased understanding of potential cellular pathways responsive to hyperglycemia is essential to develop novel therapeutic strategies for diabetic retinopathy. Emerging evidence shows the impact of microRNA (miR) as a potential novel therapeutic target. The purpose of our study was to test the hypothesis that miR-15b and miR-16 are altered by hyperglycemia in retinal endothelial cells (REC), and that miR-15b/16 play key roles in regulating insulin signaling through a reduction in TNFα- and suppressor of cytokine signaling 3 (SOCS3)-mediated insulin resistance pathways. Human REC were maintained in normal (5 mM) glucose or transferred to high-glucose medium (25 mM) for 3 days. REC were transfected with miRNA mimics (hsa-miR-15b-5p and hsa-miR-16-5p) 48 h before cell harvest. A final concentration of 30 nM was used when transfected separately (miR-15b and miR-16) and 15 nM was used in combination (miR-15b + miR-16). A negative control group was treated with an equal concentration of a mimic negative control. The levels of miRNA overexpression were verified using quantitative reverse transcription-polymerase chain reaction and real-time PCR. Western blot analyses were performed to study the levels of phosphorylated Akt (Serine 473), Akt, SOCS3, insulin receptor, phosphorylated insulin receptor (tyrosine 1150/1151), and insulin receptor phosphorylated on Tyr960. In addition, ELISA was used to examine cleaved caspase 3 and TNFα. Analyses were done using unpaired Student t test. Data are presented as mean ± S.E.M. We demonstrated that the expression of miR-15b and miR-16 was reduced in human REC cultured in hyperglycemia. Overexpression of miR-15b and/or miR-16 reduced TNFα and SOCS3 levels, while increasing insulin-like growth factor binding protein-3 (IGFBP-3) levels and the phosphorylation of insulin receptor (IR)(Tyr1150/1151) in REC cultured in hyperglycemia. These, in turn, led to an increase of Akt phosphorylation and decreased cleavage of caspase 3. miR-15b and miR-16 play a role in the inhibition of insulin resistance via reduced TNFα and SOCS3 signaling and increased IGFBP-3 levels, resulting in REC protection from hyperglycemia-induced apoptosis. This outcome suggests that both miR-15b and miR-16 are potential therapeutic targets for therapeutics for the diabetic retina.

Role of IL-16 in CD4 T cell-mediated regulation of relapsing multiple sclerosis

April 2015

·

55 Reads

In an important article published in Nature Medicine, Liu and colleagues described a novel CD4+ FoxA1+ regulatory T (Treg) cell population as distinct regulators of relapsing-remitting multiple sclerosis (RRMS) and experimental autoimmune encephalomyelitis (EAE). CD4+ FoxA1+ Treg cells appear as key regulators of responsiveness to therapy with interferon beta (IFN-β) in RRMS patients. Data indicate that CD4+FoxA1+ FOXP3− Treg cells develop within the central nervous system (CNS), and a potential of cerebellar granule neurons (CGN) in generation of CD4+FoxA1+PD-L1hiFOXP3− Treg cells from encephalitogenic CD4+ T cells. A CD4 co-receptor specific ligand, IL-16, governs trafficking and biological properties of CD4+ T cells irrespective of their activation state. Functions of IL-16, relevant to Treg cells, include expansion of CD4+CD25+ T cells in long-term cultures with IL-2, de novo induction of FOXP-3 and migration of FOXP-3+ T cells. IL-16 is highly conserved across species including human and mouse. CGN and neurons in hippocampus contain neuronal-IL-16 (NIL-16), splice variant of immune IL-16, and express CD4 molecule. In a CD4-dependent manner, IL-16 supports cultured CGN survival. Concomitant studies of RRMS lesions and corresponding MOG35–55-induced relapsing EAE in (B6 × SJL)F1 (H-2b/s) mice discovered similar roles of IL-16 in regulation of relapsing disease. In RRMS and EAE relapse, peak levels of IL-16 and active caspase-3 correlated with CD4+ T cell infiltration and levels of T-bet, Stat-1(Tyr701), and phosphorylated neurofilaments of axonal cytoskeleton [NF (M + H) P], suggesting a role of locally produced IL-16 in regulation of CD4+ Th1 inflammation and axonal damage, respectively. IL-16 was abundantly present in CD4+ T cells, followed by CD20+ B, CD8+ T, CD83+ dendritic cells, and Mac-1+ microglia. Apart from lesions, bioactive IL-16 was located in normal-appearing white matter (NAWM) and normal-appearing grey matter (NAGM) in RRMS brain and spinal cord. A cytokine IL-16 emerges as an important regulator of relapsing MS and EAE. Better understanding of immune cell-neuron interactions mediated by IL-16 will foster development of more specific CD4+ T cell subset-targeted therapies to prevent or ameliorate progression of neuroinflammation and axonal and neuronal damage. Translational studies necessitate corresponding EAE models.

Production of IL-16 correlates with CD4+ Th1 inflammation and phosphorylation of axonal cytoskeleton in multiple sclerosis lesions

February 2006

·

172 Reads

Multiple sclerosis (MS) is a central nervous system-specific autoimmune, demyelinating and neurodegenerative disease. Infiltration of lesions by autoaggressive, myelin-specific CD4+Th1 cells correlates with clinical manifestations of disease. The cytokine IL-16 is a CD4+ T cell-specific chemoattractant that is biased towards CD4+ Th1 cells. IL-16 precursor is constitutively expressed in lymphocytes and during CD4+ T cell activation; active caspase-3 cleaves and releases C-terminal bioactive IL-16. Previously, we used an animal model of MS to demonstrate an important role for IL-16 in regulation of autoimmune inflammation and subsequent axonal damage. This role of IL-16 in MS is largely unexplored. Here we examine the regulation of IL-16 in relation to CD4+ Th1 infiltration and inflammation-related changes of axonal cytoskeleton in MS lesions. We measured relative levels of IL-16, active caspase-3, T-bet, Stat-1 (Tyr 701), and phosphorylated NF(M+H), in brain and spinal cord lesions from MS autopsies, using western blot analysis. We examined samples from 39 MS cases, which included acute, subacute and chronic lesions, as well as adjacent, normal-appearing white and grey matter. All samples were taken from patients with relapsing remitting clinical disease. We employed two-color immunostaining and confocal microscopy to identify phenotypes of IL-16-containing cells in frozen tissue sections from MS lesions. We found markedly increased levels of pro- and secreted IL-16 (80 kD and 22 kD, respectively) in MS lesions compared to controls. Levels of IL-16 peaked in acute, diminished in subacute, and were elevated again in chronic active lesions. Compared to lesions, lower but still appreciable IL-6 levels were measured in normal-appearing white matter adjacent to active lesions. Levels of IL-16 corresponded to increases in active-caspase-3, T-bet and phosphorylated Stat-1. In MS lesions, we readily observed IL-16 immunoreactivity confined to infiltrating CD3+, T-bet+ and active caspase-3+ mononuclear cells. We present evidence suggesting that IL-16 production occurs in MS lesions. We show correlations between increased levels of secreted IL-16, CD4+ Th1 cell inflammation, and phosphorylation of axonal cytoskeleton in MS lesions. Overall, the data suggest a possible role for IL-16 in regulation of inflammation and of subsequent changes in the axonal cytoskeleton in MS.

IFN-γ protects from lethal IL-17 mediated viral encephalomyelitis independent of neutrophils

May 2012

·

93 Reads

The interplay between IFN-γ, IL-17 and neutrophils during CNS inflammatory disease is complex due to cross-regulatory factors affecting both positive and negative feedback loops. These interactions have hindered the ability to distinguish the relative contributions of neutrophils, Th1 and Th17 cell-derived effector molecules from secondary mediators to tissue damage and morbidity. Encephalitis induced by a gliatropic murine coronavirus was used as a model to assess the direct contributions of neutrophils, IFN-γ and IL-17 to virus-induced mortality. CNS inflammatory conditions were selectively manipulated by adoptive transfer of virus-primed wild-type (WT) or IFN-γ deficient (GKO) memory CD4+ T cells into infected SCID mice, coupled with antibody-mediated neutrophil depletion and cytokine blockade. Transfer of GKO memory CD4+ T cells into infected SCID mice induced rapid mortality compared to recipients of WT memory CD4+ T cells, despite similar virus control and demyelination. In contrast to recipients of WT CD4+ T cells, extensive neutrophil infiltration and IL-17 expression within the CNS in recipients of GKO CD4+ T cells provided a model to directly assess their contribution(s) to disease. Recipients of WT CD4+ T cells depleted of IFN-γ did not express IL-17 and were spared from mortality despite abundant CNS neutrophil infiltration, indicating that mortality was not mediated by excessive CNS neutrophil accumulation. By contrast, IL-17 depletion rescued recipients of GKO CD4+ T cells from rapid mortality without diminishing neutrophils or reducing GM-CSF, associated with pathogenic Th17 cells in CNS autoimmune models. Furthermore, co-transfer of WT and GKO CD4+ T cells prolonged survival in an IFN-γ dependent manner, although IL-17 transcription was not reduced. These data demonstrate that IL-17 mediates detrimental clinical consequences in an IFN-γ-deprived environment, independent of extensive neutrophil accumulation or GM-CSF upregulation. The results also suggest that IFN-γ overrides the detrimental IL-17 effector responses via a mechanism downstream of transcriptional regulation.

MP treatment ameliorates EAE in DA rats and decreases infiltration of cells into the CNS. DA rats were immunized with SCH-CFA. Commencing on the day when first neurological signs appeared (designated as day 1), DA rats were injected daily for 3 days with methylprednisolone (MP, 50 mg/kg body weight) or with phosphate-buffered saline (PBS). The rats were checked daily for clinical signs of EAE. For the determination of the clinical course of EAE, 19 animals per group (out of four independent experiments) were used. (A). Three hours after the last injection of MP some animals were sacrificed, and cells infiltrating spinal cord, splenocytes, peripheral blood cells, and draining lymph node cells were isolated and counted (B). *p < 0.05 represents statistically significant difference between MP and PBS group.
MP inhibits IFN-γ, but not IL-17 production, in cells infiltrating the CNS of DA rats with EAE. SCC and DLNC were isolated from DA rats immunized with SCH-CFA and injected with MP or PBS, as described in Methods. RNA was isolated from SCC and RT-PCR was performed (A), or SCC were grown in medium and ELISA was performed from cell-free supernatants obtained after 72 hours of cultivation (B). Alternatively, SCC from PBS-treated (C) and MP-treated (D) rats were stained for IFN-γ and IL-17. Results of intracellular staining (C, D) are representative of three independent experiments. The numbers in the plots are means ± SD of the proportions of cells present in the corresponding quadrant obtained in four independent experiments. DLNC were cultivated without (E) or with (F) 1 μg/ml of ConA for 72 hours and cell-culture supernatants were analyzed by ELISA. *p < 0.05 represents a statistically significant difference between the MP and PBS groups.
MP does not affect CD4+ cell proportion or apoptosis rate among CNS-infiltrating cells. SCC were isolated from DA rats immunized with SCH-CFA, and injected with MP or with PBS, as described in Methods. SCC were stained with anti-CD4 and anti-CD11b specific antibodies (A), with anti-CD4 and anti-CD62L or anti-OX40 or anti-CD25 specific antibodies (B), or with anti-CD4 specific antibody and Annexin V (C). Alternatively, SCC from PBS-treated (D) and MP-treated (E) rats were stained with anti-CD4 antibody and intracellular staining for IFN-γ and IL-17 was performed. The results presented in B-E are gated to CD4+ cells. Results of intracellular staining (D, E) are representative of three independent experiments. *p < 0.05 represents a statistically significant difference between the MP and PBS groups.
RU486 prevents inhibition of IFN-γ and IL-17 by MP. (A) DA rats were immunized with SCH-CFA. Commencing on the day when first neurological signs appeared (designated as day 1) DA rats were injected daily for 3 days with methylprednisolone (MP, 50 mg/kg body weight) and/or RU486 (25 mg/kg body weight) or with phosphate-buffered saline (PBS). The rats were checked daily for clinical signs of EAE. For the determination of the clinical course of EAE, 15 animals per group (out of three independent experiments) were used. (B, C) CLNC from healthy animals were stimulated with ConA (1 μg/ml) in the presence or absence of MP (10 ng/ml) and/or RU486 (5 ng/ml) for 24 hours. Cell-free supernatants of the cultures were analyzed with ELISA for IFN-γ (B) and IL-17 (C) concentrations. (D) SCC were isolated from DA rats immunized with SCH-CFA and injected with PBS, or with MP, or with MP+RU486, as described in Methods. Cell-free supernatants of cultures of SCC grown in medium for 72 hours were analyzed with ELISA for IFN-γ and IL-17 concentrations. *p < 0.05 represents a statistically significant difference between the MP and PBS treatment groups, and 'p < 0.05 represents a statistically significant difference between the MP and MP+RU486 treatment groups.
Methylprednisolone inhibits IFN-?? and IL-17 expression and production by cells infiltrating central nervous system in experimental autoimmune encephalomyelitis

December 2009

·

81 Reads

Glucocorticoids have been shown to be effective in the treatment of autoimmune diseases of the CNS such as multiple sclerosis and its animal model, experimental autoimmune encephalomyelitis (EAE). However, the mechanisms and the site of glucocorticoids' actions are still not completely defined. The aim of this study was to investigate the in vivo effect of the synthetic glucocorticoid methylprednisolone (MP) on the expression and production of proinflammatory cytokines interferon (IFN)-gamma and interleukin (IL)-17 by cells infiltrating CNS tissue. Experimental autoimmune encephalomyelitis was induced in Dark Agouti (DA) rats by immunization with rat spinal cord homogenate mixed with adjuvants. Commencing on the day when the first EAE signs appeared, DA rats were injected daily for 3 days with MP and/or RU486, an antagonist of glucocorticoid receptor. Cytokine production and gene expression in CNS-infiltrating cells and lymph node cells were measured using ELISA and real time PCR, respectively. Treatment of rats with MP ameliorated EAE, and the animals recovered without relapses. Further, MP inhibited IFN-gamma and IL-17 expression and production in cells isolated from the CNS of DA rats with EAE after the last injection of MP. The observed effect of MP in vivo treatment was not mediated through depletion of CD4+ T cells among CNS infiltrating cells, or through induction of their apoptosis within the CNS. Finally, the glucocorticoid receptor-antagonist RU486 prevented the inhibitory effect of MP on IFN-gamma and IL-17 production both in vitro and in vivo, thus indicating that the observed effects of MP were mediated through glucocorticoid receptor-dependent mechanisms. Taken together, these results demonstrate that amelioration of EAE by exogenous glucocorticoids might be, at least partly, ascribed to the limitation of effector cell functions in the target tissue.

Functional interleukin-17 receptor A is expressed in central nervous system glia and upregulated in experimental autoimmune encephalomyelitis

May 2009

·

679 Reads

Interleukin-17A (IL-17A) is the founding member of a novel family of inflammatory cytokines that plays a critical role in the pathogenesis of many autoimmune diseases, including multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE). IL-17A signals through its receptor, IL-17RA, which is expressed in many peripheral tissues; however, expression of IL-17RA in the central nervous system (CNS) and its role in CNS inflammation are not well understood. EAE was induced in C57Bl/6 mice by immunization with myelin oligodendroglial glycoprotein. IL-17RA expression in the CNS was compared between control and EAE mice using RT-PCR, in situ hybridization, and immunohistochemistry. Cell-type specific expression was examined in isolated astrocytic and microglial cell cultures. Cytokine and chemokine production was measured in IL-17A treated cultures to evaluate the functional status of IL-17RA. Here we report increased IL-17RA expression in the CNS of mice with EAE, and constitutive expression of functional IL-17RA in mouse CNS tissue. Specifically, astrocytes and microglia express IL-17RA in vitro, and IL-17A treatment induces biological responses in these cells, including significant upregulation of MCP-1, MCP-5, MIP-2 and KC chemokine secretion. Exogenous IL-17A does not significantly alter the expression of IL-17RA in glial cells, suggesting that upregulation of chemokines by glial cells is due to IL-17A signaling through constitutively expressed IL-17RA. IL-17RA expression is significantly increased in the CNS of mice with EAE compared to healthy mice, suggesting that IL-17RA signaling in glial cells can play an important role in autoimmune inflammation of the CNS and may be a potential pathway to target for therapeutic interventions.

Differential effects of interleukin-17 receptor signaling on innate and adaptive immunity during central nervous system bacterial infection

June 2012

·

55 Reads

Although IL-17A (commonly referred to as IL-17) has been implicated in the pathogenesis of central nervous system (CNS) autoimmune disease, its role during CNS bacterial infections remains unclear. To evaluate the broader impact of IL-17 family members in the context of CNS infection, we utilized IL-17 receptor (IL-17R) knockout (KO) mice that lack the ability to respond to IL-17, IL-17F and IL-17E (IL-25). In this article, we demonstrate that IL-17R signaling regulates bacterial clearance as well as natural killer T (NKT) cell and gamma-delta (γδ) T cell infiltrates during Staphylococcus aureus-induced brain abscess formation. Specifically, when compared with wild-type (WT) animals, IL-17R KO mice exhibited elevated bacterial burdens at days 7 and 14 following S. aureus infection. Additionally, IL-17R KO animals displayed elevated neutrophil chemokine production, revealing the ability to compensate for the lack of IL-17R activity. Despite these differences, innate immune cell recruitment into brain abscesses was similar in IL-17R KO and WT mice, whereas IL-17R signaling exerted a greater influence on adaptive immune cell recruitment. In particular, γδ T cell influx was increased in IL-17R KO mice at day 7 post-infection. In addition, NK1.1high infiltrates were absent in brain abscesses of IL-17R KO animals and, surprisingly, were rarely detected in the livers of uninfected IL-17R KO mice. Although IL-17 is a key regulator of neutrophils in other infection models, our data implicate an important role for IL-17R signaling in regulating adaptive immunity during CNS bacterial infection.

Detection of interleukin 17 (IL-17) receptors (R) IL-17R A and IL-17R B on mouse Schwann cells (SCs), using immunocytochemistry and rtPCR analysis. (A-C) immunocytochemistry for IL-17R A, IL-17R B with mainly membrane-bound expression (D-F). (G-I) Real-time PCR results of IL-17R mRNA expression of mouse dorsal root ganglia (DRG) co-cultures. Cultures were treated with IL-17 for 21 days at the indicated concentrations. Stimulation gave rise to a strong decrease in IL-17R A expression at the mRNA level under stimulation with 0.5 and 50 mg/mL IL-17, with a remaining expression of 30% (***P ≤0.001). A dose-dependent decrease was detected for IL-17R B and IL-17R C. The remaining expression of IL-17R B was 58% with 0.5 ng/mL IL-17 (**P ≤0.01) and 14% with 50 ng/mL IL-17 (***P ≤0.001). Similarly, the expression of IL-17R C was significantly reduced at the mRNA level to 78% for 0.5 ng/mL IL-17 (*P ≤0.05) and 33% for 50 ng/mL IL-17 (***P ≤0.001). For all three receptors and the concentrations applied, an analysis of variance between the three independent experiments revealed no significant difference. AU, arbitrary unit; DAPI, diamidino-2-phenylindole.
Myelin morphology and quantification after interleukin 17 (IL-17) treatment. (A-E) For myelin quantification, mouse dorsal root ganglia (DRG) co-cultures were exposed to IL-17 for 21 days at the indicated concentrations and subsequently stained, using Sudan black dye. Images show myelin layer (α) and Schwann cell body (β) are separated from the next internode by Ranvier’s node (γ) and neuronal cell bodies (δ). In analysis of three independent experiments, quantification revealed a myelin ratio (internodes to neurons) reduction to 66% for 0.5 ng/mL IL-17 (**P = <0.01), to 55% for 5 ng/mL IL-17 (**P = <0.01), and to 42% for 50 ng/mL IL-17 (**P = <0.01), each compared with control stimulations. (F) Internodal distances after exposure to IL-17 revealed a significant decrease with 5 ng/mL IL-17 (*P = <0.05) and 50 ng/mL IL-17 (*P = <0.05). Analysis of variances between the independent experiments revealed no significant difference. (G) Myelin quantification of Sudan-stained DRG co-cultures after treatment with IL-17, an IL-17-neutralizing antibody (αIL-17ab), and co-stimulation of three independent experiments, respectively. The myelin-inhibitory effect of IL-17 was reduced by 81% after supplementation with αIL-17ab. Myelination was restored to the base level, similar to treatment with the antibody alone.
mRNA expression of myelin genes P0, and KROX-20 of rat Schwann cells (rSCs) and myelin morphology after stimulation with interleukin 17 (IL-17). (A) mRNA expression of myelin genes P0 and KROX-20 is normalized to glyceraldehyde 3-phosphate dehydrogenase (GAPDH) and to control cultures. A significant decrease in KROX-20 mRNA expression to 51% (**P ≤0.01) was seen on stimulation with 5 and 50 ng/mL IL-17. P0 mRNA showed a significant down-regulation to 84% expression with 5 ng/mL IL-17 (*P ≤0.05) and a non-significant down-regulation to 92% expression with 50 ng/mL IL-17. For P0 and KROX-20 and both concentrations applied; an analysis of variance between the three independent experiments revealed no significant difference. (B) Ultrastructure of the myelin layer of dorsal root ganglia (DRG) co-cultures after IL-17 stimulation (5 ng/mL IL-17, top row) and control cultures (bottom row). Cultures were stimulated from day 6 after preparation and fixed after 28 days in vitro. The left images depict a longitudinal section and the right images a traverse section referring to the axonal fiber. Myelin showed no morphological alterations after IL-17 stimulation. AU, arbitrary unit; Ax, axon; My, myelin layer.
Neuronal outgrowth, Schwann cell (SC) viability, and Matrix metalloproteinases (MMP) activity in Interleukin 17 (IL-17) treated cultures. (A) Neuronal outgrowth was assessed in mouse dorsal root ganglia (DRG) co-cultures after exposure for 21 days to IL-17 at the indicated concentrations. Representative neurofilament (NF)-stained images of the cultures are depicted. (B) Quantification of NF, using densitometry analysis, normalized to background fluorescence and control stimulation. Analysis did not reveal a significant alteration in neuronal outgrowth. (C) SC viability was analyzed after IL-17 stimulation for 7 days at the indicated concentrations. Concentrations between 0.5 and 50 ng/mL did not significantly alter SC viability. Graph for viability and neurofilament analysis shows results of three independent experiments (D, E). MMP activity in IL-17-treated DRG co-culture supernatants was analyzed using gelatine zymography and quantified densitometrically. Representative zymography with the densitometry analysis is depicted. Overall, three independent experiments were performed and analyzed, exhibiting comparable results. AU, arbitrary unit. IL-17 led to a dose-dependent increase in inflammatory MMP-9 activity (E; 92 kDa), whereas the active and the inactive form of MMP-2 (D; MMP-2 inactive: 72 kDa, active: 62 kDa) decreased in activity.
Major histocompatibility complex (MHC) I and II as well as Transporter associated with antigen presentation II (TAPII) were analyzed, using immunocytochemistry on rat Schwann cells (SCs). Corresponding merges are shown in the bottom rows. Treatment of SCs with IL-17 was performed at concentrations of 0.5 and 50 ng/mL. Graphs to the right show densitometry quantification. SCs showed expression of MHCI > TAPII > MHCII, which increased after IL-17 treatment. (A) MHCI was mainly detected in the cytoplasm and the expression increased in a dose-dependent manner after IL-17 treatment, significant for 0.5 ng/mL and 50 ng/mL (**P ≤0.01). (B) MHCII revealed a fainter basic expression emphasizing the nucleus and was found significantly increased after 0.5 ng/mL IL-17 stimulation (**P ≤ 0.01). (C) TAPII was detected in the nucleus and cytoplasm. We detected a dose-dependent tendency but no significantly increased expression after IL-17 stimulation. For MHCI, TAPII and MHCII and the concentrations applied, analysis of variance between the independent experiments revealed no significant difference. DAPI, 4′, 6-diamidino-2-phenylindole.
Interleukin-17 impedes Schwann cell-mediated myelination

March 2014

·

238 Reads

Pro-inflammatory cytokines are known to have deleterious effects on Schwann cells (SCs). Interleukin 17 (IL-17) is a potent pro-inflammatory cytokine that exhibits relevant effects during inflammation in the peripheral nervous system (PNS), and IL-17-secreting cells have been reported within the endoneurium in proximity to the SCs. Here, we analyzed the effects of IL-17 on myelination and the immunological properties of SCs. Dorsal root ganglia (DRG) co-cultures containing neurons and SCs from BL6 mice were used to define the impact of IL-17 on myelination and on SC differentiation; primary SCs were analyzed for RNA and protein expression to define the putative immunological alignment of the SCs. SCs were found to functionally express the IL-17 receptors A and B. In DRG cultures, stimulation with IL-17 resulted in reduced myelin synthesis, while pro-myelin gene expression was suppressed at the mRNA level. Neuronal outgrowth and SC viability, as well as structural myelin formation, remained unaffected. Co-cultures exhibited SC-relevant pro-inflammatory markers, such as matrix metalloproteinase 9 and SCs significantly increased the expression of the major histocompatibility complex (MHC) I and exhibited a slight, nonsignificant increase in expression of MHCII, and a transporter associated with antigen presentation (TAP) II molecules relevant for antigen processing and presentation. IL-17 may act as a myelin-suppressive mediator in the peripheral nerve, directly propagating SC-mediated demyelination, paralleled by an inflammatory alignment of the SCs. Further analyses are warranted to elucidate the role of IL-17 during inflammation in the PNS in vivo, which could be useful in the development of target therapies.

Figure 1: Cytokines titers of the multiplex assay. Cytokine titers of the control subjects (n = 14), the patients with sporadic Creutzfeldt-Jakob disease (CJD, n = 14) and those with autoimmune encephalitis (n = 14). *P < 0.05, **P < 0.01, ***P < 0.001.
Table 1 Clinical profiles of patients with sporadic Creutzfeldt-Jakob disease
Figure 2: Correlation of cytokine titers and clinical parameters. (A) Correlation between cerebrospinal fluid interleukin-17 (IL-17) titers and disease duration before sampling in the patients with sporadic Creutzfeldt-Jakob disease. (B) Correlation between cerebrospinal fluid IL-17 and total protein titers in the patients with sporadic Creutzfeldt-Jakob disease. Total protein titer was used as a general inflammatory marker in the central nervous system.
Table 2 Cytokine profiles in the cerebrospinal fluid
Increased interleukin-17 in the cerebrospinal fluid in sporadic Creutzfeldt-Jakob disease: A case-control study of rapidly progressive dementia

November 2013

·

81 Reads

Inflammatory responses in the cerebrospinal fluid (CSF) of patients with sporadic Creutzfeldt-Jakob disease (sCJD) remain elusive. We conducted a case-control study, in which 14 patients with sCJD, 14 with noninflammatory neurological disorders, and 14 with autoimmune encephalitis were enrolled. We used the suspension array system to measure the concentrations of 27 cytokines in CSF. The cytokine titers of the three groups were compared, and the correlation between the relevant cytokine titers and clinical parameters was investigated in the patients with sCJD. Levels of the two cytokines interleukin (IL)-1 receptor antagonist and IL-17 were significantly elevated in the patients with sCJD compared with those in the patients with noninflammatory neurological disorders: IL-17 levels in sCJD were approximately ten times higher than in the noninflammatory neurological disorders (mean, 35.46 vs. 3.45 pg/ml; P < 0.001) but comparable to that in encephalitis (mean, 32.16 pg/ml). In contrast, levels of classical proinflammatory cytokines such as IL-12(p70) and tumor necrosis factor-alpha were increased only in encephalitis. Although not significant, IL-17 titers tended to be higher in the patients with shorter disease duration before CSF sampling (r = -0.452; P = 0.104) and in those with lower CSF total protein concentrations (r = -0.473; P = 0.086). IL-17 is significantly increased in CSF in sCJD, which can be an early event in the pathogenesis of sCJD.

Table 1 Distribution of the TLR2 -196 to -174 del polymorphism in LOAD cases and controls. 
Table 2 Logistic regression analysis of the -196 to -174 del polymorphism within TLR2. 
Toll-like receptor 2 -196 to -174 del polymorphism influences the susceptibility of Han Chinese people to Alzheimer's disease

October 2011

·

66 Reads

Toll-like receptor 2 (TLR2) represents a reasonable functional and positional candidate gene for Alzheimer's disease (AD) as it is located under the linkage region of AD on chromosome 4q, and functionally is involved in the microglia-mediated inflammatory response and amyloid-β clearance. The -196 to -174 del polymorphism affects the TLR2 gene and alters its promoter activity. We recruited 800 unrelated Northern Han Chinese individuals comprising 400 late-onset AD (LOAD) patients and 400 healthy controls matched for gender and age. The -196 to -174 del polymorphism in the TLR2 gene was genotyped using the polymerase chain reaction (PCR) method. There were significant differences in genotype (P = 0.026) and allele (P = 0.009) frequencies of the -196 to -174 del polymorphism between LOAD patients and controls. The del allele was associated with an increased risk of LOAD (OR = 1.31, 95% CI = 1.07-1.60, Power = 84.9%). When these data were stratified by apolipoprotein E (ApoE) ε4 status, the observed association was confined to ApoE ε4 non-carriers. Logistic regression analysis suggested an association of LOAD with the polymorphism in a recessive model (OR = 1.64, 95% CI = 1.13-2.39, Bonferroni corrected P = 0.03). Our data suggest that the -196 to -174 del/del genotype of TLR2 may increase risk of LOAD in a Northern Han Chinese population.

Top-cited authors