IL-37 expression reduces acute and chronic neuroinflammation and rescues cognitive impairment in an Alzheimer’s disease mouse model

  1. Niklas Lonnemann
  2. Shirin Hosseini
  3. Melanie Ohm
  4. Robert Geffers
  5. Karsten Hiller
  6. Charles A Dinarello  Is a corresponding author
  7. Martin Korte  Is a corresponding author
  1. Department of Cellular Neurobiology, Zoological Institute, Germany
  2. Neuroinflammation and Neurodegeneration Group, Helmholtz Centre for Infection Research, Germany
  3. BRICS - Braunschweig Integrated Centre of Systems Biology, Germany
  4. Genome Analytics Group, Helmholtz Center for Infection Research, Germany
  5. Department of Computational Biology of Infection Research, Helmholtz Centre for Infection Research, Germany
  6. Department of Medicine, University of Colorado Denver, United States
  7. Department of Medicine, Radboud University, Medical Center, Netherlands
8 figures, 2 tables and 1 additional file

Figures

Figure 1 with 1 supplement
Primary microglial cells from IL-37tg mice showed decreased pro-inflammatory cytokine release after stimulation by LPS.

(A) WT and IL-37tg primary microglial cells (P3-5) were plated and stimulated without LPS or with LPS. Cells were treated for 6 hr (B) and 24 hr (B–E). Cells from homozygous transgenic animals released less pro-inflammatory cytokines IL-6 (C), TNF- (D) and IL-1β (E) under different LPS concentration (B-E n=4–9). (F) IL37 mRNA was analyzed in IL-37tg microglial cells at specific intervals after addition of LPS. (G) Levels of IL-6 were measured from the same cells as in (F) and were detectable only after 24 hr (n=3). (H) Levels of IL-6 were measured in WT after addition of increasing concentrations of recombinant IL-37b and 100 ng LPS (H n=6–18). Data are presented as mean ± SEM. * p<0.05, ** p<0.01, *** p<0.001 compared to WT. (B+F + H: 1-way ANOVA with multiple comparison; C-E+G: t-test).

Figure 1—source data 1

Release of pro-inflammatory cytokines by microglia after LPS stimulation.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig1-data1-v2.xlsx
Figure 1—figure supplement 1
Pro-inflammatory cytokine release by primary astrocytes after LPS stimulation.

(A–B) Primary astrocytes respond to an LPS stimulus (100 ng/ml for 24 hr) with increased levels of the pro-inflammatory cytokines IL-6 (A) and TNF-α (B). In contrast to microglial cells, astrocytes from hIL-37tg mice showed no reduction in these two pro-inflammatory cytokines (n=5-10).

Figure 1—figure supplement 1—source data 1

Release of pro-inflammatory cytokines by astrocytes after LPS stimulation.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig1-figsupp1-data1-v2.xlsx
Primary microglial cells isolated from IL-37tg mice exhibited reduced levels of inflammation-associated intracellular metabolites after stimulation with LPS.

WT and IL-37tg microglia were stimulated with 10 ng/ml LPS (based on highly sensitive metabolomics assessments). Metabolomic analysis was performed on these cells. (A) Heatmap of identified significantly altered metabolites after or without LPS stimulation (described as Z-score). (B–C) Significant effects in WT, treated with LPS, were seen with respect to itaconate (B) and succinate (C), whereas these changes were significantly reduced in IL-37tg microglial cells treated with LPS compared with WT cells (n=11). Data are presented as mean ± SEM. *** p<0.001 (two-way ANOVA with multiple comparison).

Figure 2—source data 1

Inflammation-associated intracellular metabolites in primary microglia after LPS stimulation.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig2-data1-v2.xlsx
Figure 3 with 2 supplements
IL-37tg mice showed decreased pro-inflammatory cytokine release and less activated microglia after the stimulus of LPS.

(A) WT and IL-37tg mice were stimulated with saline or LPS. (B) IL-37tg animals exhibited significantly less weight loss compared to WT mice. However, IL-37tg mice also had a significant weight change compared to saline treated mice (n=10–22). Microglial cell activation was analyzed by FACS method. (C–D) Microglial cells were identified as CD11b+ and CD45low cells and analyzed for CD68 expression. IL-37tg mice had a lower percentage of cells with CD68 expression compared with WT mice after LPS stimulation (n=4) (C–D). (E) In addition, WT mice exhibited a significant increase in IL-1β levels after LPS treatment, whereas IL-37tg mice did not (n=6–8). (F–I) Morphological analysis of microglial cells showed an increased number of IBA-1-positive cells in WT animals treated with LPS compared with saline-treated animals. In contrast, there is no increased IBA-1-positive cell number in IL-37tg animals after LPS stimulation (n=9–18; n=18–30 cells for processes). (J) Representative images of IBA-1-positive cells (red) and DAPI (blue); scale bar 40 µm. Data are shown as mean ± SEM. * p<0.05, ** p<0.01, *** p<0.001, (B-I: 2-way ANOVA with multiple comparison).

Figure 3—source data 1

Neuroinflammatory status of mouse brain after systemic LPS challenge.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig3-data1-v2.xlsx
Figure 3—figure supplement 1
Pro-inflammatory cytokines in brain lysates after peripheral LPS challenge.

(A–B) Pro-inflammatory cytokines IL-6 and TNF-α were measured in brain lysates from mice treated with LPS or saline (control). WT Mice showed slightly increased levels of IL-6 after LPS compared with mice treated with saline, whereas hIL-37tg mice did not show increased IL-6 levels (A) (n=7-10); TNF-α levels were not altered in both WT and hIL-37tg mice after LPS challenge (B) (n=3-5).

Figure 3—figure supplement 1—source data 1

Pro-inflammatory cytokines in mouse brain after systemic LPS challenge.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig3-figsupp1-data1-v2.xlsx
Figure 3—figure supplement 2
FACS analysis of brain cells after peripheral LPS administration.

(A–B) Fluorescent activated cell sorting (FACS) method was used to analyze microglial activation with respect to CD68 expression (n=4). In forward/side scatter (FSC/SSC), the region of interest (ROI) was selected (A) and analyzed for two monocyte markers (CD11b and CD45) (B). (C–J) CD68 expressing cells were analyzed in microglia (CD11b+/CD45low) (C–E), macrophages (CD11b+/CD45high) (F–H), and leukocytes (CD11b-/CD45high) (I–J). (K) The number of cells in the different populations is shown in the experimental groups.

IL-37tg mice showed rescued synaptic plasticity and restored loss of spine density after stimulation by LPS compared with WT animals.

(A) WT animals stimulated with LPS showed significant impairment of theta burst stimulation-induced LTP (TBS) compared with WT, which were treated with saline. (B) In contrast, IL-37tg mice showed no significant impairment of LTP after LPS treatment. (C) Mean LTP magnitude (average of 55–60 min after TBS) was significantly lower in WT mice treated with LPS, while IL-37tg mice showed no significant differences (n of mice 3–4; n of acute slices 11–17). (D–F) Spine density in apical dendrites of the CA1 hippocampal neurons and in the superior DG neurons was significantly decreased in WT mice treated with LPS, whereas spine density of IL-37tg animals treated with LPS was not affected (n of mice 3–4; n of dendrites 13–25) (D and E). (F) Representative images of dendritic spines of hippocampal CA1 neurons in the tested groups were shown; scale bar 5 µm. (G) WT acute slices stimulated with LPS showed significant impairment of TBS-induced LTP compared with WT acute slices treated with ACSF. (H) In contrast, acute slices from IL-37tg mice showed no significant impairment of LTP after LPS treatment. (I) Mean LTP magnitude (mean of 55–60 min after TBS) was significantly lower in acute slices from WT mice treated with LPS, whereas slices from IL-37tg mice showed no significant differences (n of mice 3–4; n of acute slices 14–17). Data are presented as mean ± SEM. * p<0.05, *** p<0.001, (A-I: two-way ANOVA with multiple comparison).

Figure 4—source data 1

Assessment of synaptic plasticity after LPS challenge.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig4-data1-v2.xlsx
Figure 5 with 1 supplement
Injection of recombinant IL-37 into WT mice showed restoration of cognitive deficits and reduced release of pro-inflammatory cytokines after IL-1β-mediated immunostimulation.

(A) WT mice were pretreated with either saline or rIL-37 for three consecutive days and then injected with saline or IL-1β. (B) WT mice pretreated with saline and then stimulated with IL-1β failed to perform the Y-maze test, whereas WT mice pretreated with rIL-37 and then stimulated with IL-1β performed the test without deficits (n=4). (C and D) Although the pro-inflammatory cytokine levels of IL-6 and IL-1α were significantly increased in stimulated WT mice pretreated with rIL-37 compared with the control group, the mice treated with rIL-37 showed a significant decrease in cytokine levels after immunostimulation with IL-1β compared with saline treated group (n=4–6). Data are presented as mean ± SEM. ** p<0.01, *** p<0.001, (B-D: two-way ANOVA with multiple comparison).

Figure 5—source data 1

Suppressive effect of rIL-37 on the inflammatory response induced by IL-1β.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig5-data1-v2.xlsx
Figure 5—figure supplement 1
Pro-inflammatory cytokines in brain lysates after peripheral pretreatment with recombinant IL-37 followed by LPS challenge.

(A–C) IL-1β (A), IL-6 (B), and TNF-α (C) measured in control and LPS-treated WT or rIL-37-pretreated WT mice (n=3-4).

Figure 5—figure supplement 1—source data 1

Pro-inflammatory cytokine levels induced by LPS in the brain of rIL-37 treated mice.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig5-figsupp1-data1-v2.xlsx
Figure 6 with 2 supplements
APP/PS1-IL37tg double transgenic mice showed lower pro-inflammatory cytokine expression and reduced activation of microglia, as well as lower numbers of amyloid plaques compared with APP/PS1 mice.

(A) WT, APP/PS1, and APP /PS1-IL37tg mice were analyzed at 6, 9–12, and 20–23 months of age. (B–C) Pro-inflammatory cytokine levels of IL-6 and IL-1β (although for IL-1β was not statistically significant) were increased in 9–12 months old APP/PS1 mice compared with WT mice, whereas APP/PS1-IL37tg mice showed no increase in pro-inflammatory cytokines (n=6–10). (D–G) Microglial cells were identified as CD11b+ and CD45low cells and analyzed for CD68 expression. Nine to 12-month-old APP/PS1 mice showed a significantly increased percentage of cells with CD68 expression compared with WT mice, whereas APP/PS1-IL37tg mice did not show a significant increase (D and F). Although 6- and 20–23 month-old APP/PS1-IL37tg mice had a significantly higher amount of CD68-expressing cells compared with WT mice, the percentage of CD68-expressing cells was reduced compared with APP/PS1 animals (6 months: n=4–6; 9–12 months: n=3–6; 20–23 months: n=3–7). (H–J) Plaque analysis showed significantly lower plaque burden (I) and reduced plaque size (J) in the hippocampal (Hp) and cortex (Cx) regions compared between APP/PS1-IL37tg mice and APP/PS1 (n=27–42). Representative image of Congo red staining in 30 µm sections; scale bar 500 µm (H). (K–M) Aβ uptake by microglial cells by measuring single cells in FACS system. Gating strategy for positive Cx3CR1-GFP cells and positive staining for MXO4 (K–L). Quantified analysis of Aβ uptake showing significantly higher uptake in APP/PS1-IL37tg cells compared with APP/PS1 cells (n=5–11) (M). Data are presented as mean ± SEM. * p<0.05, ** p<0.01, *** p<0.001 compared to WT, # p<0.05, ## p<0.01, ### p<0.001 compared to APP/PS1. (B-G and M: one-way ANOVA with multicolumn comparison; I-J: t-test).

Figure 6—source data 1

Neuroinflammatory status in the brain of APP/PS1 mice.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig6-data1-v2.xlsx
Figure 6—figure supplement 1
Congo red staining of brain sections from APP/PS1 and APP /PS1-IL37 mice.

(A–B) Plaque staining in the cortex area of APP/PS1 mice (A) and APP/PS1-IL37tg mice (B). A single plaque is shown in the lower right inset. (C–D) Plaque staining in the hippocampal region of APP/PS1 mice (C) and APP/PS1-IL37tg mice (D). Individual plaques are shown in the lower left inset (scale bar 200 µm).

Figure 6—figure supplement 2
FACS analysis of microglial cells regarding their Methoxy-X04 uptake.

(A–C) Gating strategy of CX3CR1-GFP+cell. (D–E) Gating strategy for MX04-(neg) and MX04+(pos) cells depending on the Pacific-Blue-Methoxy-X04 signal. (G–I) Cell counts of MX04- and MX04 + cells in WT, APP/PS1 and APP/PS1-IL37tg (n=5-11).

Figure 7 with 1 supplement
APP/PS1-IL37tg double transgenic mice showed improvements in behavioral tests and synaptic plasticity compared with APP/PS1 mice.

(A) WT, APP/PS1 and APP/PS1-IL37tg mice were analyzed at 9–12 months of age. (B–D) The cognitive deficits of APP/PS1 mice in the spatial learning test of the Morris Water Maze could be restored in APP/PS1-IL37tg animals. WT APP/PS1 and APP/PS1-IL37tg mice showed learning behavior during the training phase of the spatial learning test. APP/PS1 animals showed higher escape latency during acquisition on day 3–6 compared to WT mice (B). WT Mice and APP/PS1-IL37tg mice show a significant preference for the target quadrant (TQ), whereas APP/PS1 mice showed no preference (C). Representative heat maps of mice from each group demonstrated the results of the reference test (D) (n=10–12). (E–G) The LTP deficits in APP/PS1 mice could be rescued in APP/PS1-IL37tg mice in the induction phase (20–25 min). However, LTP deficits were not restored in APP/PS1-IL37tg animals in the maintenance phase (75–80 min) (n of animals 3–4; n of slices 12–18). (H–I) Dendritic spine density was significantly reduced in APP/PS1 animals compared to WT, whereas there was no significant reduction in spine density in APP/PS1-IL37tg (n=21–23), scale bar 5 µm. Data are presented as mean ± SEM. * p<0.05, ** p<0.01, *** p<0.001 compared to WT, ^^ p<0.01, ^^^ p<0.001 compared to NT (non-target quadrants); (B+E: two-way ANOVA with multiple comparison; C: t-test; C-I: one-way ANOVA with multiple comparison).

Figure 7—source data 1

Assessment of spatial learning and synaptic plasticity in APP/PS1 mice.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig7-data1-v2.xlsx
Figure 7—figure supplement 1
Age of the trained mice in the behavioral experiment.

Average age of each genotype trained in the Morris water maze is displayed (n=10-12).

Figure 7—figure supplement 1—source data 1

Age range of mice trained in the Morris water maze.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig7-figsupp1-data1-v2.xlsx
Figure 8 with 1 supplement
RNA sequencing of microglia isolated from WT and IL-37tg mice after LPS challenge in vivo.

(A) Heatmap shows z-normalized gene expression profiles of the 500 most highly regulated genes (out of 11014 genes with adjusted pValue <0.05). Two clusters of co-regulated genes were identified, representing up- and down-regulated genes after LPS treatment. Each column shows expression data of individual mouse transcriptomes. (B) Gene Set Enrichment Analysis (GSEA) was performed for the 500 most up-regulated genes, which are also shown in the heat map. Each cluster (1 and 2) was tested for enriched gene sets defined by the Reactome Pathway Database (https://reactome.org/). The number of genes that could be linked to gene sets in the Reactome Pathway Database is indicated under each cluster name. The terms for the major gene sets are shown. The size of the circles corresponds to the ratio of genes found in each gene set. Significance of enrichment is expressed by a blue-red color code. (C) Differential expression comparing WT and IL-37tg after LPS stimulation is shown by the Volcano plot. Log2FC and the corresponding adjusted pValue are shown for each gene analyzed (18,049 in total). The dashed lines show the limits for significant gene regulation: –1 1, FDR <0.05. (D–K) Changes in gene expression after LPS injection in WT and IL-37tg mice for candidate genes are shown (n=2-3).

Figure 8—source data 1

RNA sequencing data of microglia isolated from mice after systemic LPS challenge.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig8-data1-v2.xlsx
Figure 8—figure supplement 1
RNA sequencing of microglia isolated from WT and IL-37tg mice after LPS challenge in vivo.

(A–D) The changes in expression levels after LPS injection in WT and IL-37tg mice for the candidate genes are shown (n=2-3).

Figure 8—figure supplement 1—source data 1

Expression levels of candidate genes in microglia isolated from mice after systemic LPS challenge.

https://cdn.elifesciences.org/articles/75889/elife-75889-fig8-figsupp1-data1-v2.xlsx

Tables

Key resources table
Reagent type (species) or resourceDesignationSource or referenceIdentifiersAdditional information
antibodyAnti-IBA1 (Rabbit polyclonal)Synaptic SystemsCat#234003, RRID:AB_106419621:1000
antibodyCy2 AffiniPure Goat Anti-Rabbit IgG (H+L) (Rabbit polyclonal)Jackson ImmunoResearch LaboratoriesCat# 111-225-144, RRID:AB_23380211:500
antibodyCy3 AffiniPure Goat Anti-Mouse IgG +IgM (H+L) (Mouse polyclonal)Jackson ImmunoResearch LaboratoriesCat#115-165-068, RRID:AB_23386861:500
antibodyCy3 AffiniPure Goat Anti-Rabbit IgG (H+L) (Rabbit polyclonal)Jackson ImmunoResearch LaboratoriesCat#111-165-144, RRID:AB_23380061:500
antibodymouse CD68-PE
Clone REA835 (mouse monoclonal)
Miltenyi BiotecCat# 130-112-8561:50
antibodymouse CD11b-PerCP-Vio700
Clone REA592 (mouse monoclonal)
Miltenyi BiotecCat# 130-113-8091:50
antibodymouse CD45-APC (mouse monoclonal)Miltenyi BiotecCat# 130-110-7981:50
chemical compound, drugBovine Serum AlbuminSigma-AldrichCat# A7906
chemical compound, drugCaCl2ApplichemLot: 4U010421
chemical compound, drugcOmplete Protease Inhibitor CocktailSigma-AldrichCat# 04693116001
chemical compound, drugDAPISigma-AldrichCat# D9542
chemical compound, drugD-glucoseRothArt.-Nr. HN06.3
chemical compound, drugEvans Blue tetrasodium saltTocrisCat# 0845
chemical compound, drugFluoro-Gel mounting mediumElectron Microscopy SciencesCat# 17985–10
chemical compound, drugGBSSSigma-AldrichG9779-500ML
chemical compound, drugGibco DMEMFisher ScientificCat# 31885023
chemical compound, drugGibco Fetal Bovine SerumFisher ScientificCat# 11573397
chemical compound, drugGibco HBSS 10 XFisher ScientificCat# 14065049
chemical compound, drugGibco L-GlutamineFisher ScientificCat# 15410314
chemical compound, drugGlycineApplichemCat# A1067
chemical compound, drugKClApplichemLot: 0000574737
chemical compound, drugKH2PO4ApplichemLot: 4Q016683
chemical compound, drugMethoxy-XO4Abcamab142818
chemical compound, drugMgSO4ApplichemLot: 3E000057
chemical compound, drugNaClApplichemLot: 8Q012497
chemical compound, drugNaHCO3RothArt.-Nr. HN01.1
chemical compound, drugPermount Mounting MediumFisher ScientificCat# SP15-100
chemical compound, drugPoly-L-lysine solutionSigma-AldrichCAS# 25988-63-0
peptide, recombinant proteinRecombinant IL-37Moretti et al., 2014N/A
peptide, recombinant proteinRecombinant IL-1βKim et al., 2013N/A
chemical compound, drugTriton X-100 Molecular Biology grade BCApplichemCat# A4975
chemical compound, drugTrypsin-EDTA Solution 10 XSigma-AldrichCAS Nr. 9002-07-7
chemical compound, drugTWEEN 20Sigma-AldrichCat# P9416
commercial assay or kitBiozym Blue Probe qPCR Kit Separate ROXBiozymCat# 331456 S
commercial assay or kitFD Congo-Red Solution KitFD NeuroTechnologies, Inc.Cat# PS108
commercial assay or kitFD Rapid GolgiStain KitFD NeuroTechnologies, Inc.Cat# PK401
commercial assay or kitHigh-Capacity cDNA Reverse Transcription KitThermo FisherCat# 4368814
commercial assay or kitMacherey-Nagel NucleoSpin RNAThermo FisherProduct Code 15373604
commercial assay or kitpegGOLD TriFastAvantorN/A
commercial assay or kitProtoScript II First Strand cDNA Synthesis KitNew England Biolabs Inc.Cat# E6560
strain, strain background (Mus musculus)B6;C3-Tg(APPswe,PSEN1dE9)85Dbo/Mmjax miceThe Jackson LaboratoryCat# 005864
strain, strain background (Mus musculus)C57BL/6 J OlaHsd miceHarlan-Winkelmann or JanvierCat# 057 (H-W)
strain, strain background (Mus musculus)Human Interleukin-37 transgenic miceNold et al., 2010N/A
software, algorithmANY-mazeStoeltingRRID:SCR_014289
https://www.stoeltingco.com/
software, algorithmFlowJoFlowJohttps://www.flowjo.com/solutions/flowjo
software, algorithmImageJWane Rasband NIH, USAN/A
software, algorithmIntraCell Version 1.5(C)2000 Institute for Neurobiology MagdeburgN/A
software, algorithmPrism 8GraphPadhttps://www.graphpad.com/scientific-software/prism/
software, algorithmVideo Mot 2TSE Systemshttps://www.tse-systems.com
software, algorithmG*Power Version 3.1.9.4Heinrich Heine University Düsseldorf, Germanyhttp://www.psychologie.hhu.de/arbeitsgruppen/allgemeine-psychologie-und-arbeitspsychologie/gpower.html
Table 1
Statistical table.
FigureNameTestMulti-comparison
1B100 ng/ml 6 hrOne-way ANOVAF(2,9)=2,428; p=0.1434Tukey’s0,8947WT vs. HET
0,1460WT vs. HOM
100 ng/ml 24 hrOne-way ANOVAF(2,11)=7,941; p=0.0073Tukey’s0,0539WT vs. HET
0,0067WT vs. HOM
1 µg/ml 6 hrOne-way ANOVAF(2,9)=12,74; p=0.0024Tukey’s0,0090WT vs. HET
0,0037WT vs. HOM
1 µg/ml 24 hrOne-way ANOVAF(2,11)=3,915; p=0.0520Tukey’s0,3586WT vs. HET
0,0426WT vs. HOM
1CIL-6 10 ng/mlttestt=1,742 df = 16; p=0.1007WT vs. IL37tg
IL-6 100 ng/mlttestt=3,597 df = 16; p=0.0024WT vs. IL37tg
IL-6 1 µg/mlttestt=5,127 df = 16; p=0.0001WT vs. IL37tg
1DTNF-α 10 ng/mlttestt=3,376 df = 16; p=0.0038WT vs. IL37tg
TNF-α 100 ng/mlttestt=6,774 df = 16; p<0.0001WT vs. IL37tg
TNF-α 1 µg/mlttestt=6,264 df = 16; p<0.0001WT vs. IL37tg
1EIL-1β 10 ng/mlttestt=2,390 df = 16; p=0.0295WT vs. IL37tg
IL-1β 100 ng/mlttestt=5,027 df = 16; p=0.0001WT vs. IL37tg
IL-1β 1 µg/mlttestt=5,852 df = 16; p<0.0001WT vs. IL37tg
1FIL-37 mRNAOne-way ANOVAF(7,15)=2,629; p=0.0550Fisher’s LSD0.01710 hr vs. 1 hr
0.00860 hr vs. 4 hr
0.02380 hr vs. 24 hr
1GIL-6ttestt=20,39 df = 4; p<0.0001WT vs. IL37tg
1HrIL-37 100 ng/mlOne-way ANOVAF(2,30)=5,135; p=0.0121Tukey’s0.1675WT vs. 100 ng
0.0133WT vs. 500 ng
rIL-37 1 µg/mlOne-way ANOVAF(2,30)=3,512; p=0.0426Tukey’s0.2457WT vs. 100 ng
0.0498WT vs. 500 ng
2BItaconate
Treatment
Two-way ANOVAF(1,20)=174,9; p<0.0001Bonferroni’s<0.0001WT NIL vs. LPS
<0.0001IL37 NIL vs. LPS
Itaconate
Genotype
Two-way ANOVAF(1,20)=27,55; p<0.0001Bonferroni’s>0.9999NIL WT vs. IL37
<0.0001LPS WT vs. IL37
2CSuccinate
Treatment
Two-way ANOVAF(1,20)=98,40; p<0.0001Bonferroni’s<0.0001WT NIL vs. LPS
0.0005IL37 NIL vs. LPS
Succinate
Genotype
Two-way ANOVAF(1,20)=17,01; p=0.0005Bonferroni’s>0.9999NIL WT vs. IL37
<0.0001LPS WT vs. IL37
3BBody weight
Treatment
Two-way ANOVAF(1,17)=220,5; p<0.0001Bonferroni’s<0.0001WT NIL vs. LPS
<0.0001IL37 NIL vs. LPS
Body weight
Genotype
Two-way ANOVAF(1,41)=1,486; p=0.2299Bonferroni’s>0.9999NIL WT vs. IL37
0.0139LPS WT vs. IL37
3DCD68
Treatment
Two-way ANOVAF(1,6)=19,52; p=0.0045Bonferroni’s0.0017WT NIL vs. LPS
>0.9999IL37 NIL vs. LPS
CD68
Genotype
Two-way ANOVAF(1,6)=48,42; p=0.0004Bonferroni’s0.0167NIL WT vs. IL37
<0.0001LPS WT vs. IL37
3EIL-1β TreatmentTwo-way ANOVAF(1,7)=9,399;p=0.0182Bonferroni’s0.0244WT NIL vs. LPS
0.7541IL37 NIL vs. LPS
IL-1β GenotypeTwo-way ANOVAF(1,7)=3,182;p=0.0935Bonferroni’s>0.9999NIL WT vs. IL37
0.0366LPS WT vs. IL37
3FIba1 CA1 TreatmentTwo-way ANOVAF(1,25)=5,222;p=0.0311Bonferroni’s0.0024WT NIL vs. LPS
>0.9999IL37 NIL vs. LPS
Iba1 CA1 GenotypeTwo-way ANOVAF(1,34)=4,951;p=0.0328Bonferroni’s>0.9999NIL WT vs. IL37
0.0027LPS WT vs. IL37
3GProcesses CA1 TreatmentTwo-way ANOVAF(1,46)=1,066; p=0.3073Bonferroni’s>0.9999WT NIL vs. LPS
0.6229IL37 NIL vs. LPS
Processes CA1 GenotypeTwo-way ANOVAF(1,58)=1,238; p=0.2704Bonferroni’s0.5670NIL WT vs. IL37
>0.9999LPS WT vs. IL37
3HIba1 Cortex TreatmentTwo-way ANOVAF(1,25)=15,97; p=0.0005Bonferroni’s0.0002WT NIL vs. LPS
0.3234IL37 NIL vs. LPS
Iba1 Cortex GenotypeTwo-way ANOVAF(1,34)=5,159; p=0.0296Bonferroni’s>0.9999NIL WT vs. IL37
0.0054LPS WT vs. IL37
3IProcesses Cortex TreatmentTwo-way ANOVAF(1,104)=9,865; p=0.0022Bonferroni’s0.1401WT NIL vs. LPS
0.0232IL37 NIL vs. LPS
Processes Cortex GenotypeTwo-way ANOVAF(1,104)=0,119; p=0.7308Bonferroni’s>0.9999NIL WT vs. IL37
0.8147LPS WT vs. IL37
4ALTP WTTwo-way ANOVAF(1,28)=4,459; p=0.0438Fisher’s LSD
Time point 43–80
<0.05WT NIL vs. LPS
4BLTP IL37Two-way ANOVAF(1,23)=0,085; p=0.7734IL37 NIL vs. LPS
4CLTP last 5 min TreatmentTwo-way ANOVAF(1,22)=7,887;p=0.0102Bonferroni’s0.0043WT NIL vs. LPS
>0.9999IL37 NIL vs. LPS
LTP last 5 min GenotypeTwo-way ANOVAF(1,29)=1,552; p=0.2228
4DSpines CA1 TreatmentTwo-way ANOVAF(1,32)=22,81; p<0.0001Bonferroni’s<0.0001WT NIL vs. LPS
0.1145IL37 NIL vs. LPS
Spines CA1 GenotypeTwo-way ANOVAF(1,44)=26,68; p<0.0001Bonferroni’s0.0657NIL WT vs. IL37
<0.0001LPS WT vs. IL37
4ESpines DG TreatmentTwo-way ANOVAF(1,26)=3,307; p=0.0805Bonferroni’s0.0062WT NIL vs. LPS
>0.9999IL37 NIL vs. LPS
Spines DG GenotypeTwo-way ANOVAF(1,36)=0,529; p=0.4718Bonferroni’s0.5222NIL WT vs. IL37
0.0295LPS WT vs. IL37
4GLTP WTTwo-way ANOVAF(1,30)=4,925; p=0.0342Fisher’s LSD
Time point 39–80
<0.05WT NIL vs. LPS
4HLTP IL37Two-way ANOVAF(1,26)=0,01395; p=0.9069IL37 NIL vs. LPS
4ILTP last 5 min TreatmentTwo-way ANOVAF(1,27)=4,613; p=0.0409Bonferroni’s0.0285WT NIL vs. LPS
>0.9999IL37 NIL vs. LPS
LTP last 5 min GenotypeTwo-way ANOVAF(1,29)=1,747; p=0.1966
5AY-Maze TreatmentTwo-way ANOVAF(1,6)=18,25; p=0.0052Bonferroni’s0.0046WT NIL vs. IL1
0.7313IL37 NIL vs. IL1
Y-Maze pre-treatmentTwo-way ANOVAF(1,6)=9,899; p=0.0199Bonferroni’s>0.9999NIL WT vs. IL37
0.0024IL1 WT vs. IL37
5BIL-6 TreatmentTwo-way ANOVAF(1,16)=264,6; p<0.0001Bonferroni’s<0.0001WT NIL vs. IL1
<0.0001IL37 NIL vs. IL1
IL-6 pre-treatmentTwo-way ANOVAF(1,16)=10,27; p=0.0055Bonferroni’s0.8327NIL WT vs. IL37
0.0004IL1 WT vs. IL37
5CIL-1α TreatmentTwo-way ANOVAF(1,16)=132,0; p<0.0001Bonferroni’s<0.0001WT NIL vs. IL1
<0.0001IL37 NIL vs. IL1
IL-1α pre-treatmentTwo-way ANOVAF(1,16)=17,47; p=0.0007Bonferroni’s>0.9999NIL WT vs. IL37
0.0003IL1 WT vs. IL37
6BIL-1βOne-way ANOVAF(2,19)=5,224; p=0.0156Fisher’s LSD0.0903WT vs. APP
0.2480WT vs. APP-IL37
0.0046APP vs. APP-IL37
6CIL-6One-way ANOVAF(2,21)=3,600; p=0.0452Fisher’s LSD0.0220WT vs. APP
0.5264WT vs. APP-IL37
0.0466APP vs. APP-IL37
6ECD68 9–12 mOne-way ANOVAF(2,11)=14,39; p=0.0008Bonferroni’s0.0008WT vs. APP
0.0529WT vs. APP-IL37
0.04494APP vs. APP-IL37
6FCD68 6 mOne-way ANOVAF(2,12)=13,45; p=0.0009Bonferroni’s0.0007WT vs. APP
0.0263WT vs. APP-IL37
0.0985APP vs. APP-IL37
6GCD68 20–23 mOne-way ANOVAF(2,12)=13,38; p=0.0009Bonferroni’s0.0007WT vs. APP
0.0359WT vs. APP-IL37
0.0383APP vs. APP-IL37
6IPlaque load
Hp
ttestt=2,403 df = 74; p=0.0187APP vs. APP-IL37
Plaque load
Cx
ttestt=4,953 df = 63; p<0.0001APP vs. APP-IL37
6JPlaque size
Hp
ttestt=2,296 df = 74; p=0.0245APP vs. APP-IL37
Plaque size
Cx
ttestt=5,949 df = 63; p<0.0001APP vs. APP-IL37
6GAbeta uptakeOne-way ANOVAF(2,22)=136; p<0.0001Bonferroni’s0.0001WT vs. APP
0.0001WT vs. APP-IL37
0.0026APP vs. APP-IL37
7BLatency WTOne-way ANOVAF(7,88)=5,842; p<0.0001WT
Latency APPOne-way ANOVAF(7,72)=3,477; p=0.0029APP
Latency APP-IL37One-way ANOVAF(7,80)=4,445; p=0.0003APP-IL37
Latency2-way ANOVAF(2,30)=10,50; p=0.0003Fisher’s LSD
Day 1
<0.05WT vs. APP
Day 3–6<0.01
7CPT
WT
ttestt=11,45 df = 22; p<0,0001NT vs. TQ
PT
APP
ttestt=0,9874 df = 18; p=0.3365NT vs. TQ
PT
APP-IL37
ttestt=3,705 df = 20; p=0.0014NT vs. TQ
PT TQsOne-way ANOVAF(2,30)=8,415; p=0.0013Turkey‘s0.0009WT vs. APP
0,0505WT vs. APP-IL37
7ELTPTwo-way ANOVAF(2,45)=9,286; p=0.0004Fisher’s LSD
Time point 21–80
<0.0005WT vs. APP
Fisher’s LSD
Time point 41–80
<0.05WT vs. APP-IL37
7FMean LTP 20–25 minOne-way ANOVAF(2,45)=7,090; p=0.0021Turkey‘s0.0016WT vs. APP
0.5444WT vs. APP-IL37
7GMean LTP 75–80 minOne-way ANOVAF(2,45)=9,354; p=0.0004Turkey‘s0.0014WT vs. APP
0.0023WT vs. APP-IL37
7ISpine densityOne-way ANOVAF(2,63)=6.318;p=0.0032Turkey‘s0.0026WT vs. APP
0.0536WT vs. APP-IL37

Additional files

Download links

A two-part list of links to download the article, or parts of the article, in various formats.

Downloads (link to download the article as PDF)

Open citations (links to open the citations from this article in various online reference manager services)

Cite this article (links to download the citations from this article in formats compatible with various reference manager tools)

  1. Niklas Lonnemann
  2. Shirin Hosseini
  3. Melanie Ohm
  4. Robert Geffers
  5. Karsten Hiller
  6. Charles A Dinarello
  7. Martin Korte
(2022)
IL-37 expression reduces acute and chronic neuroinflammation and rescues cognitive impairment in an Alzheimer’s disease mouse model
eLife 11:e75889.
https://doi.org/10.7554/eLife.75889