Skip to main content
Immunology logoLink to Immunology
. 2018 May 10;155(1):112–122. doi: 10.1111/imm.12934

MicroRNA signature of central nervous system‐infiltrating dendritic cells in an animal model of multiple sclerosis

Mariah L Hoye 1, Angela S Archambault 1, Taylor M Gordon 1, Landon K Oetjen 2, Matthew D Cain 2, Robyn S Klein 2,3, Seth D Crosby 4, Brian S Kim 2,5,6, Timothy M Miller 1,3,, Gregory F Wu 1,3,5,
PMCID: PMC6099169  PMID: 29749614

Summary

Innate immune cells are integral to the pathogenesis of several diseases of the central nervous system (CNS), including multiple sclerosis (MS). Dendritic cells (DCs) are potent CD11c+ antigen‐presenting cells that are critical regulators of adaptive immune responses, particularly in autoimmune diseases such as MS. The regulation of DC function in both the periphery and CNS compartment has not been fully elucidated. One limitation to studying the role of CD11c+ DCs in the CNS is that microglia can upregulate CD11c during inflammation, making it challenging to distinguish bone marrow‐derived DCs (BMDCs) from microglia. Selective expression of microRNAs (miRNAs) has been shown to distinguish populations of innate cells and regulate their function within the CNS during neuro‐inflammation. Using the experimental autoimmune encephalomyelitis (EAE) murine model of MS, we characterized the expression of miRNAs in CD11c+ cells using a non‐biased murine array. Several miRNAs, including miR‐31, were enriched in CD11c+ cells within the CNS during EAE, but not LysM+ microglia. Moreover, to distinguish CD11c+ DCs from microglia that upregulate CD11c, we generated bone marrow chimeras and found that miR‐31 expression was specific to BMDCs. Interestingly, miR‐31‐binding sites were enriched in mRNAs downregulated in BMDCs that migrated into the CNS, and a subset was confirmed to be regulated by miR‐31. Finally, miR‐31 was elevated in DCs migrating through an in vitro blood–brain barrier. Our findings suggest miRNAs, including miR‐31, may regulate entry of DCs into the CNS during EAE, and could potentially represent therapeutic targets for CNS autoimmune diseases such as MS.

Keywords: dendritic cells, microRNAs, multiple sclerosis, neuroinflammation


Abbreviations

AD

Atopic dermatitis

Ago2

argonaute 2

APCs

antigen‐presenting cells

BBB

blood–brain barrier

BMDCs

bone marrow‐derived dendritic cells

CNS

central nervous system

DCs

dendritic cells

EAE

experimental autoimmune encephalomyelitis

LPS

lipopolysaccharide

miRAP

miRNA affinity and tagging purification

miRNAs

microRNAs

MS

multiple sclerosis

Tregs

T regulatory cells

Introduction

Multiple sclerosis (MS) – the prototypical inflammatory disease of the central nervous system (CNS) – is characterized by widely distributed inflammatory demyelinating plaques within the brain and spinal cord that can be associated with severe neurological disability.1 A hallmark feature of MS plaques is engagement of leucocytes with the blood–brain barrier (BBB) coupled with an influx of immune cells into the CNS from the periphery.2, 3 Using the common animal model for MS, experimental autoimmune encephalomyelitis (EAE), a variety of innate immune cells, including dendritic cells (DCs) and microglia, have been implicated in the promotion of neuro‐inflammation.4, 5 The mechanisms by which innate cells regulate adaptive immune responses in the CNS, coordinate damage to myelin and neurons, and respond to injury remain unclear.

MicroRNAs (miRNAs) are small, non‐coding RNAs that regulate the stability and translation of mRNAs.6 While numerous physiological processes are regulated by the ever‐expanding number of recognized miRNAs, the cell‐specific expression of miRNAs is elemental to their ultimate function.7 Furthermore, several miRNAs expressed by innate immune cells have been implicated in dysregulation of immune function that is associated with, and may be causal to, autoimmunity.8, 9 Thus, identification of miRNA signatures for various immune cells is likely to provide insight into the mechanisms by which innate immunity is involved in the pathogenesis of neuro‐inflammatory diseases such as MS. For example, a small number of miRNAs has been described as a signature for microglia in comparison to other innate immune cells.10 What remains to be determined is the role for miRNA in the specialization of different innate immune cell subsets, particularly during inflammatory diseases of the CNS.

We have previously found that DCs are critical regulators of adaptive immune responses in the CNS, and that the bone marrow‐derived fraction of DCs is necessary for T‐cell‐dependent neuro‐inflammation during EAE.11 Several miRNAs, including Let‐7c,12 miR‐142‐3p13 and miR‐34a,14 have been shown to regulate DC function. In what manner miRNA expression directs DC autoimmune behaviour during EAE has yet to be established. Therefore, we aimed to determine the miRNA signature of DCs that accumulate within the CNS during EAE.

To characterize the molecular regulation of DCs in EAE, we utilized an in vivo Cre‐LoxP murine system in which a tagged miRNA‐processing protein, argonaute 2 (Ago2), is expressed only in CD11c‐expressing cells. Immunoprecipitation of tagged Ago2 (tAgo2) from these cells facilitates isolation of CD11c‐specific miRNAs.15, 16 Using this system in combination with miRNA arrays, we found that miR‐31 and miR‐301 are significantly enriched in CD11c‐expressing mononuclear cells isolated from the CNS during EAE. Furthermore, the enrichment of miR‐31 is driven by entry of bone marrow‐derived DCs (BMDCs) and miR‐31 regulates their mRNA expression profiles. Moreover, miR‐31 serves as a signature of BMDCs migrating through endothelial cells in an in vitro BBB system.

Materials and methods

Mice

Animal protocols were in accordance with the ARRIVE guidelines, and were approved by the Institutional Animal Care and Use Committee of Washington University at St Louis. Wild‐type (WT) C57BL/6 (B6) mice were purchased from The Jackson Laboratory (Bar Harbor, ME). MiRNA tagging and affinity purification (miRAP) mice16 expressing a loxP‐flanked myc‐tagged Ago2 construct under the control of CMV beta‐actin enhancer promoter inserted in the ROSA locus (JAX ID: 017626) were crossed to CD11cCre mice (ItgaxCre)17 obtained from The Jackson Laboratory (JAX ID: 008068) to generate miRAPCD11c mice. Likewise, miRAPLysM mice were generated by breeding miRAP mice to LysMCre mice18 obtained from The Jackson Laboratory (JAX ID: 004781). Radiation bone marrow chimeras were generated by intravenous injection of 1 × 106 donor bone marrow cells into hosts irradiated with 1000 Gy.11 Bone marrow from mice with ubiquitous GFP expression (JAX ID: 003291)19 or eYFP driven by CD11c (JAX ID: 008829)20 was transplanted into irradiated mice. Mice were provided 0·1% Enrofloxacin (Baytril®; Bayer Healthcare, Shawnee Mission, KS) antibiotic PO ad libitum for 7 days post‐irradiation. Irradiated mice were used for experimentation after full immune reconstitution at 8 weeks following bone marrow transplantation.

EAE

We induced active EAE in standard fashion.11 Briefly, mice were immunized with residues 35–55 of myelin oligodendrocyte glycoprotein (MOG35–55) emulsified in complete Freund's adjuvant. Two‐hundred nanograms of pertussis toxin was administered at the time of immunization and 2 days later. Mice were graded on a scale of 0 (unaffected) to 5 (moribund or dead), with 1 representing a limp tail, 2 indicating hind limb paresis, 3 reflecting more severe hind limb paresis along with righting difficulty, and 4 indicative of hind limb paralysis.11

Atopic dermatitis (AD)

Six mice were treated with 50 μm MC903 (calcipotriol) or vehicle (ethanol) on each ear for 8 days to stimulate an inflammatory response in the skin.21 Lymph nodes and ears were harvested on the 8th day. Ears were separated with forceps and digested in 0·25 mg/ml Liberase TL (Roche/Sigma, St. Louis, MO) in Dulbecco's modified Eagle's minimum essential medium (DMEM) for 90 min at 37°. Single cell suspensions from lymph nodes and digested ear skin were filtered through 100‐μm filters and washed in media before staining for FACS as described above. Cells from individual mice or pooled mice were then used to isolate RNA.

miRAP and miRNA arrays

At day 21 post‐immunization, mice with a clinical score of 2 or greater were anaesthetized with an overdose of inhaled isoflurane and subsequently perfused with ice‐cold 1 × phosphate‐buffered saline (PBS). Spleen (unperfused) and spinal cord tissue were flash‐frozen in liquid nitrogen and frozen at −80°. MiRAP immunoprecipitation using an anti‐myc antibody (9E10 sc‐40; Santa Cruz, Dallas, TX) was performed as previously described.15, 16 A BCA assay was performed on all tissue homogenates to normalize protein input; 700 μl of Qiazol (Qiagen, Germantown, MD) was added directly to the beads and stored at −20° before proceeding with RNA isolation (miRNeasy 217004; Qiagen). Taqman low‐density rodent miRNA microarrays (version 3.0; Life Technologies, Waltham, MA) were performed with pre‐amplification on spinal cord and spleen tissue from three male mice. Microarrays were run on a 7900HT quantitative polymerase chain reaction (qPCR) machine (Thermo Fisher; Waltham, MA) for 40 cycles.

mRNA microarrays

Radiation bone marrow chimeras with BMDCs expressing eYFP were immunized with MOG35–55 as described above to induce active EAE. CD11c+eYFP+ cells were sorted by FACS at day 18 post‐immunization from the spleen and perfused CNS. RNA was isolated and amplified with the Nugen Ovation WTA kit (Nugen, San Carlos, CA) according to the NuGEN Ovation Pico WTA system v2 user guide and the Affymetrix GeneChip Expression Analysis Technical Manual, then hybridized to Affymetrix Mouse Gene 1·0ST arrays (Affymetrix, Santa Clara, CA). A GeneChip 3000 7G scanner was used to collect fluorescence signal. In order to determine the fold‐change of the genes of interest, Affymetrix normalized, background‐subtracted.cel files were imported into Partek Genomic Suite (v 5). In order to determine genes whose expression is altered between radiation‐resistant and radiation‐sensitive DCs, as well as radiation‐sensitive DCs within the CNS versus spleen, two‐way anova analysis was employed on log2‐transformed data. P‐values were corrected using step‐up multiple testing correction.22

miRNA expression validation

MiRNA array expression data were normalized to a geometric mean of all miRNAs expressed at CT < 30. DC miRNAs enriched in the CNS (spinal cord) as compared with periphery (spleen) were confirmed at a higher power (n = 6) with individual reverse transcription (RT)‐qPCR assays on a 7500 fast Real‐Time PCR system.

Flow cytometry and FACS sorts

Mice were perfused with 25 ml of ice‐cold PBS. Subsequently, brains and spinal cords were collected from perfused mice and homogenized to obtain single cell suspensions. CNS cells were purified by centrifugation for 30 min in a 95% Percoll (GE Healthcare, Chicago, IL) solution with EBSS (Sigma, St Louis, MO).11 Cells were incubated with the anti‐Fc receptor antibody 2·4G2 prior to the addition of antibodies. The following antibodies were purchased from BD Biosciences (Franklin Lakes, NJ): CD45‐FITC, CD8a‐APC‐H7, CD19‐APC‐H7, CD19‐BV510, B220–PE‐CF594, CD11b‐AlexaFluor‐700, MHCII‐v450. The following antibodies were purchased from eBioscience (San Diego, CA): MHCII‐Pacific Blue, CD11c‐PECy7. The following antibodies were purchased from BioLegend (San Diego, CA): MHCII (I‐A/I‐E)‐Pacific Blue, Thy1·1‐PerCP, CD4‐APC. Cells were acquired on a Gallios flow cytometer (Beckman Coulter, Brea, CA) and analysed with flowjo software (TreeStar, Ashland, OR) with doublets being excluded. Cells were sorted on a SY3200 cell sorter (Sony Biotechnology, San Jose, CA), and 700 μl of Qiazol was added directly to the cells before proceeding with RNA isolation (miRNeasy 217004; Qiagen).

To identify BMDCs that entered into the CNS during EAE, the bone marrow of mice expressing enhanced GFP under the actin promoter were used to generate chimeras. During EAE, we isolated cells using FACS by first gating on singlets from the lymphocyte population of cells isolated, as shown in Fig. S2. To isolate microglia, we excluded GFP+ cells and sorted the CD45intCD11bint population of cells. To isolate BMDCs, we gated the GFP+ cells and sorted the CD11c+ population of cells. CD11c−/CD11b− cells from the GFP+ population were also gated to sort CD3+ T‐cells. CD45+ GFPCD11bCD11c+ DCs and CD45GFPCD3+ T‐cells were sorted similarly from the single cell suspensions of the spleens from the same mice with EAE.

To identify CD11c+ resident and migratory DCs in the draining lymph from mice with AD‐induced skin inflammation, we isolated cells using FACS by first gating on singlets and then excluding CD3+, CD9+ and NK1·1+ cells from the CD45+ population. We then sorted CD11c+ MHCIIlow (resident DCs) and CD11c+ MHCIIhigh (migratory DCs) populations.23 To isolate CD11c+ cells that infiltrated the ear, we again excluded the CD3CD9NK1·1+ cells from the CD45+ population, and isolated CD11chi, CD207low cells.

Migration assays

Bone marrow‐derived dendritic cells were generated in vitro as previously reported.24 Briefly, bone marrow from tibia and fibulae were cultured in RPMI medium containing 10% fetal bovine serum and supplemented with 10 ng/ml granulocyte‐macrophage colony‐stimulating factor (R&D Systems, Minneapolis, MN) for 7–10 days. BBB cultures were utilized as described previously;25 0·5–1·0 × 106 BMDCs were added to the top of the BBB cultures, and 16 hr later media was removed from the top and bottom chambers of the transwell apparatus. Cells were spun down and 700 μl of Qiazol was added directly to the cells before proceeding with RNA isolation (miRNeasy 217004; Qiagen).

Luciferase assays

The 3′‐untranslated regions (UTRs) of putative miR‐31 targets were cloned into the Dual Glo luciferase vector (Promega, Madison, WI) using the primers listed in Table S1. Mouse miR‐31 was cloned into the pSilencer vector (Thermo Fisher) using the primers: GGACACTCTAAGGACCACTGC (forward) and CGAGACAGACCAAGTCACAGG (reverse). Site‐directed mutagenesis was used to mutate the first four nucleotides of the miR‐31‐binding site in the IL‐34 3′‐UTR; 30 000 HEK 293 cells were plated in a 96‐well dish (Thermo Fisher), and transfected with 100 ng of dual glo vector and 100 ng of miR‐31 using FuGENE 6 (Promega). Firefly luciferase (Fluc) luminescence was monitored 24 hr post‐transfection using the Dual Glo assay kit (Promega, E2920), and was normalized to a control renilla luciferase (Rluc). Three–six technical replicates for each condition were performed for each biological replicate (n = 3).

Results

Migratory CD11c+ cells have a miRNA expression profile distinct from microglia in EAE spinal cord tissue

Several distinct antigen‐presenting cells (APCs) are present within the CNS during inflammatory demyelination.5, 26 DCs, generally identified by expression of the integrin CD11c, are potent APCs involved in the initiation and propagation of neuro‐inflammation in MS and EAE.11, 27, 28 We sought to examine miRNA expression by DCs to define the potential regulatory mechanism of this important APC subset during EAE. We utilized the murine miRAP system15, 16 to selectively isolate miRNA from CD11c+ DCs during EAE. MiRNA from the spleens and spinal cords of miRAPCD11c mice with EAE was isolated at day 21 post‐immunization (Fig. 1a). MiRNA microarray analysis of miRAPCD11c spinal cord tissue identified several miRNAs with elevated expression compared with the spleen (Data S1; Fig. S1). As confirmed by individual RT‐qPCR assays, miR‐31, miR‐301a, miR‐301b and miR‐339‐5p expressions were elevated in DCs within the CNS of miRAPCD11c mice during EAE (Fig. 1b). To explore the selective nature of miRNA expression by CD11c+ cells, we compared the expression of these miRNAs with innate cells expressing the myeloid marker LysM, which, in a perfused mouse, labels resident microglia.29, 30 In comparison to miRAPLysM mice at the peak of EAE, significant elevations of miR‐31, miR‐301a and miR‐339‐5p were observed in CNS of miRAPCD11c mice, suggesting a selective enrichment in CD11c+ DCs compared with microglial cells expressing LysM31 (Fig. 1c). Consistent with isolation of LysM+ microglia, the expression of miR‐155, a miRNA important in myeloid cells during EAE,32, 33, 34 was enriched in miRAPLysM as compared with miRAPCD11c EAE spinal cord tissue (Fig. 1d). These data suggest that migratory CD11c+ cells in EAE spinal cord tissue have a miRNA expression profile distinct from LysM+ microglia.

Figure 1.

Figure 1

The miRNA signature of migratory CD11c+ cells in experimental autoimmune encephalomyelitis (EAE) spinal cord tissue is distinct from LysM+ microglia. (a) Schematic of miRNA affinity and tagging purification (miRAP) approach where CD11c‐Cre mice are crossed with mice expressing LSL‐tAgo2. miRAP from spleen and spinal cord of CD11c Cre, LSL‐tAgo2 mice isolates miRNAs only from CD11c+ DCs. (b) miR‐31, 339‐5p, 301a and 301b levels are enriched in CD11c+ cells of EAE spinal cord tissue as compared to spleen; n = 6/tissue. (c, d) miRAP of CD11c+ cells as compared with LysM+ microglia in the spinal cord indicates elevated miR‐339‐5p, miR‐301a and miR‐31 is specific to CD11c+ cells, whereas miR‐155 is more enriched in LysM+ microglia; n = 6/line. Values represented as mean ± SEM. Relative expression normalized to a geometric mean of miR‐24 and miR‐191. Student's two‐tailed, unpaired t‐tests with Bonferroni correction for multiple (4) comparisons (b, c–d). Adjusted P‐values: *P ≤ 0·05, **P ≤ 0·01, ***P ≤ 0·001, ****P ≤ 0·0001.

MiR‐31 expression in migratory CD11c+ cells in EAE spinal cord is driven selectively by BMDCs and not resident microglia

Expression of CD11c by both infiltrating DCs and CNS resident innate immune cells can confound the identification of DCs within the CNS during EAE.35 As BMDCs are considered essential and sufficient for antigen presentation during EAE,11, 27, 36 we generated radiation bone marrow chimeric mice in which miRAPCD11c bone marrow was transplanted into lethally irradiated B6 or miRAPCD11c host mice. We performed miRAP and isolated RNA at the peak of disease from the spinal cords and spleens of chimeric mice, and ran miRNA arrays (Data S1). In agreement with miRAPCD11c mice with EAE, elevated expression of miR‐31 was observed in the spinal cords of miRAPCD11c → miRAPCD11c chimeric mice (Fig. 2a). While this did not reach statistical significance (P = 0·1), the magnitude of the elevation was nearly identical to the upregulation that was found in Fig. 1a. Furthermore, DC expression of miR‐31 in miRAPCD11c → WT mice was significantly elevated within the spinal cord in comparison to the spleen, suggesting the expression of miR‐31 by CD11c‐expressing cells in the spinal cord during EAE is driven by BMDCs, not CNS resident innate immune cells (Fig. 2a). To confirm this finding, we isolated bone marrow‐derived CD11bhiCD11cint/hi DCs and CD45intCD11bint microglia from the spinal cord of EAE mice by FACS. In these sorted cells, we found that miR‐31 was highly and selectively enriched in BMDCs as compared with resident microglia (Fig. 2d). These data, however, do not distinguish BMDCs from CNS‐infiltrating monocytes that differentiate into DCs.

Figure 2.

Figure 2

miR‐31 enrichment in central nervous system (CNS) localized CD11c+ cells is driven by bone marrow‐derived dendritic cells (BMDCs), not resident CD11c+ cells during experimental autoimmune encephalomyelitis (EAE). (a) miRNA affinity and tagging purification (miRAP) of all CD11c+ cells or specifically CD11c+ BMDCs from EAE tissue confirms enrichment of miR‐31 in the spinal cord as compared with spleen, particularly in CD11c+ BMDCs; n = 6/condition/tissue. CD45intCD11bint microglia (b) and CD11bhiCD11cint/hi BMDCs (c) were isolated by FACS from the spinal cord (n = 4–8) of mice with EAE. (d) Expression of miR‐31 by CD11bhiCD11cint/hi BMDCs and CD45intCD11bint microglia isolated by FACS from the spinal cord of mice with EAE; n = 4–8. (e) miR‐34b‐3p is selectively depleted from CNS‐infiltrating BMDCs as compared with resident CNS CD11c+ cells. (f) Expression of miR‐34b‐3p by CD11cint/hi BMDCs and CD45intCD11bint microglia isolated by FACS from the spinal cord of mice with EAE; n = 4–8. Values represented as mean ± SEM. Relative expression normalized to a geometric mean of miR‐24 and miR‐191 (a, e) or U6 snRNA (d, f). One‐way anova with multiple comparisons (Dunnett's) (a, e) or Student's two‐tailed, unpaired t‐tests (d, f). P‐values: *P ≤ 0·05, **P ≤ 0·01, ***P ≤ 0·001.

Concomitant with the selective expression of miR‐31 in BMDCs, we also found that miR‐34b‐3p was selectively depleted in CD11c+ cells arising from the bone marrow (Fig. 2e). Thus, miR‐34b‐3p appears to be restricted to resident CNS immune cells that upregulate CD11c during EAE. Consistent with this hypothesis, we found that miR‐34b‐3p expression was higher in microglia than in bone marrow‐derived CD11bhiCD11cint/hi cells isolated by FACS from the CNS during EAE (Fig. 2f).

Because miR‐31 has been reported to be elevated in T‐cells,37 we compared miR‐31 expression in various mononuclear cells sorted by FACS from the spleens and CNS of mice with EAE. Intriguingly, compared with miR‐31 expression by CD11bhiCD11cint/hi DCs, CD3+ T‐cells within the periphery of mice with EAE expressed a low level of miR‐31 (Fig. S2f). However, upon entry into the CNS, expression of miR‐31 was higher in CD3+ T‐cells than CD11bhiCD11cint/hi DCs (Fig. S2g).

miR‐31‐binding sites are enriched in mRNAs downregulated in BMDCs that migrate into the CNS during EAE

To determine the functional relevance of elevated miR‐31 in BMDCs that migrate into the CNS during EAE, we generated chimeric mice expressing eYFP exclusively by bone marrow‐derived CD11c+ cells. At the peak of EAE induced by MOG35–55 immunization, microarrays were performed on mRNA isolated from BMDCs in the spleen and CNS. Because miRNAs typically downregulate their target mRNAs, we queried mRNAs that were downregulated by ≥ 1·5‐fold in brain BMDCs versus spleen BMDCs, and found a significant number were putative miR‐31 targets (Fig. 3a; P = 0·013, χ2 test with Yates’ correction). These data suggest that miR‐31 elevation in BMDCs in CNS tissue during EAE functionally modulates BMDCs cellular expression profiles. To confirm miR‐31 regulation of mRNAs downregulated in BMDCs that migrate into the CNS, we performed luciferase assays on a subset of targets. Of the four candidates tested, three were confirmed to be regulated by miR‐31 (Fig. 3b–e). The full list of mRNA changes is included in Data S1.

Figure 3.

Figure 3

miR‐31 sites are enriched in mRNAs downregulated in central nervous system (CNS) bone marrow‐derived dendritic cells (BMDCs) compared with spleen BMDCs. (a) We found that of the 4480 mRNAs downregulated by ≥ 1·5‐fold in CNS BMDCs compared with spleen BMDCs, 101 transcripts were putative miR‐31 targets (Targetscan.org). This is a significant enrichment when compared with the mouse genome of ~23 000 genes. (b–e) Putative miR‐31 targets, Hiat1 (b), Srp54b (d) and Tspan31 (e) were confirmed using luciferase assays; the luminescence activity of the reporter bearing the corresponding 3′‐untranslated region (UTR) was downregulated in the presence of miR‐31 as compared with scrambled miRNA control. Putative miR‐31 target, Ndrg3, (c) was not regulated by miR‐31. n = 10/condition (three biological replicates with three–four technical replicates each). Values represented as mean ± SEM. Student's two‐tailed, unpaired t‐tests (b–e). P‐values: *P ≤ 0·05, **P ≤ 0·01, ***P ≤ 0·001.

Elevated miR‐31 expression in Cd11c+ DCs may be broadly representative of DCs that migrate into inflamed tissue

To test whether elevated miR‐31 expression is specific to DCs that migrate into the CNS during EAE or whether elevated miR‐31 is generally representative of BMDCs infiltrating inflamed tissues, we used a murine model of skin inflammation resembling AD. In this paradigm, leucocytes, including DCs, migrate to the site of irritation in the ears induced by the vitamin D3 analogue, MC903. We isolated migratory DCs from the ear and lymph nodes, as well as resident DCs in the lymph nodes, by FACS (Fig. S3a–d). Interestingly, we found that migratory DCs in the lymph nodes did not have elevated miR‐31 expression, but DCs infiltrating into the skin did have elevated miR‐31 expression as compared with resident DCs in the lymph nodes (Fig. S3e). These data suggest that elevated miR‐31 may be representative of DCs that localize to inflamed tissue rather than those migrating in general.

miR‐31 is enriched in BMDCs that migrate through an in vitro BBB irrespective of lipopolysaccharide (LPS)‐mediated DC activation

Because the expression of miR‐31 by DCs is derived from DCs originating outside of the CNS compartment, we hypothesized that engagement of DCs with tissue resident cells may prompt an increase in miR‐31 expression. To test this hypothesis, we used cultured BMDCs in an in vitro BBB model.25 DCs were incubated in the top chamber of the BBB system, and were harvested from both top and bottom compartments after 16 hr. Expression of the innate cell‐associated miRNA, miR‐155, was similar in DCs from both the top and bottom wells (Fig. 4a). In contrast, DCs traversing the BBB into the bottom chamber had a nearly two‐fold increase in miR‐31 (Fig. 4b). To determine whether DC activation further influenced DC migration, we activated DCs with LPS prior to incubation with BBB cultures. To confirm that DCs were responding to LPS stimulation, we measured the levels of miR‐155 and miR‐31 in LPS‐activated and ‐non‐activated DCs. As expected, LPS‐activated DCs had a robust increase in miR‐155 expression, whereas miR‐31 was not changed (Fig. 4c). This suggests miR‐31 expression is not upregulated in response to DC activation by LPS. The application of these activated DCs resulted in a similar enriched expression of miR‐31 in DCs that migrate into the lower chamber, while the miR‐155 levels were still not influenced by passing through the in vitro BBB (Fig. 4d,e). These data suggest that miR‐31 expression by DCs is not substantially driven by LPS‐mediated activation, and its elevation in DCs that migrate into the CNS is irrespective of LPS‐mediated activation status. However, other stimulants may differentially affect miR‐31 expression in DCs or their migratory propensity.

Figure 4.

Figure 4

MiR‐31 is increased in dendritic cells (DCs) that migrate through an in vitro blood–brain barrier (BBB). (a, b) miR‐31, but not miR‐155, is elevated in cultured bone marrow‐derived dendritic cells (BMDCs) that migrate through (bottom DCs) an in vitro BBB assay, n = 6–9/condition. (c) Lipopolysaccharide (LPS) stimulation of BMDCs results in increased expression of miR‐155, but not miR‐31; n = 2/condition. (d, e) LPS‐activated DCs also have elevated miR‐31, but not miR‐155, upon migrating through (bottom DCs) an in vitro BBB, n = 6/condition. Values represented as mean ± SEM. Relative expression normalized to U6 snRNA. Student's unpaired, two‐tailed t‐test (a, b, d, e). P ‐values: *P ≤ 0·05, **P ≤ 0·01.

Discussion

Dysregulation of DC function is considered an underpinning of autoimmunity in the brain and spinal cord during MS. An important consideration regarding the role of DCs in MS autoimmunity is the specialized compartmental nature of the CNS. At steady‐state, DCs are found scattered sparsely within the CNS, primarily affiliated with the vasculature and meninges.27, 38 In EAE, entry and accumulation of DCs within the CNS is driven in large part by recruitment from bone marrow progenitors.11, 39 Because BMDCs are critical and sufficient for antigen presentation during EAE,11, 27, 36 understanding their entry into the CNS is of great therapeutic interest. However, one limitation to studying DC function in EAE is the possible contamination of microglia that upregulate the DC marker CD11c during neuroinflammation.

MicroRNAs regulate hundreds of targets and, accordingly, shape the expression profile of a cell. Thus, miRNAs often exhibit cell type‐enriched expression and have been shown to distinguish various innate immune cell populations.10 Furthermore, miRNAs can also influence cellular migration, most notably in cancer metastasis.40, 41 To this end, we sought to characterize the miRNA expression profile of migratory BMDCs, distinct from microglia, in the CNS compartment during EAE.

Our finding that miR‐31 expression is enriched in BMDCs that pass through the BBB both in vivo and in vitro suggests that miR‐31 may participate in CNS, or general inflamed tissue, localization of peripheral DCs. A precedent for this concept was reported in vascular development, whereby miR‐126‐3p mediates perivascular and endothelial interactions.42 More abundant evidence for miRNA regulation of cellular migration stems from the field of tumour biology. For example, miR‐223 facilitates gastric cancer cell migration and invasion,40 while miR‐515‐5p expression is associated with reduced migration and metastasis of multiple cancer lines.41 Thus, miR‐31 may act to regulate the trafficking of DCs through the BBB during inflammation. Consistent with a functional role for elevated miR‐31 in modulating a DC phenotype associated with infiltration of inflamed tissue, we found that miR‐31‐binding sites were enriched in mRNAs downregulated in DCs that migrated into the CNS during EAE (Fig. 3a). The confirmed miR‐31‐downregulated mRNAs (Hiat1, Srp54b and Tspan31) will be interesting candidates for further studies into the phenotypic changes that contribute to DC migration into inflamed tissue.

Alternatively, the milieu of the inflamed CNS microenvironment may drive DC expression of miR‐31 upon invasion of the CNS and other inflamed tissue. Again, similar observations have been made in tumour studies, including metastatic infiltration into the brain.43, 44 The potential for specific miRNA changes in DCs based on interactions beyond the BBB during EAE and MS raises the intriguing possibility that miR‐31 acts to perpetuate inflammation within the local tissue environment. Indeed, miR‐31 targets IL‐34, which is a critical component for microglial survival as well as maintaining BBB integrity.45, 46 Using a luciferase reporter assay, we found that miR‐31 robustly represses IL‐34 (Fig. S4), which predicts a loss of IL‐34 by infiltrating DCs within the CNS during EAE. While IL‐34 is expressed most abundantly in neurons within the naive CNS, it is possible that miR‐31‐mediated suppression of IL‐34 by DCs assists ongoing BBB damage during EAE. Further exploration of the effects from miR‐31‐dependent IL‐34 expression changes in vivo during EAE is warranted.

Intriguingly, activation of DCs using LPS did not induce miR‐31 expression, nor influence the migration of DCs through an in vitro BBB (Fig. 4c,e). While several miRNAs have been implicated in DC activation, including miR‐34a, let‐7c and miR‐155,12, 47, 48 miRNA regulation of DC migration through the BBB has not been characterized extensively. Expression of both chemokine receptors49 and adhesion molecules50 is considered critical for DC migration into the CNS, and both have been shown to be targets of miRNA regulation.51 The maturation state of DCs influences the combination of chemokine and cytokine expression,52 thus adding to the complexity of DC trafficking into the CNS. In dissecting the steps involved in DC infiltration through the BBB, it is feasible that distinct molecular pathways for activation and trafficking are utilized, thus enabling additional levels of regulation, some of which may be miRNA‐mediated. Indeed, the separation of differentiation and activation of DCs from their migration was recently described by Rhee and colleagues, who found that PTPN12 is essential for DC migration from the periphery to secondary lymphoid organs without influencing differentiation, maturation or cytokine production.53 Our data suggest that miR‐31 induction in DCs may be regulated by factors other than those involved in DC maturation, thus identifying a distinct miRNA‐dependent regulatory mechanism for DC migration during neuro‐inflammation.

Recently, a report demonstrated the importance of miR‐31 in EAE based on its role in T‐cell lineage commitment.37 Using a murine conditional miR‐31 knockout, Zhang and colleagues demonstrated that miR‐31 serves to inhibit the generation of peripheral T regulatory cells (Tregs).37 The function of miR‐31 during EAE may thus serve dual effects on driving the pathogenesis of MS by coordinating a layered inflammatory response in both the innate as well as adaptive immune responses. Our data found that miR‐31 expression is higher in CD11c+ DCs in the peripherary as compared with CD3+ T‐cells (Fig. S2f), but that upon entry into the CNS, CD3+ T‐cells have increased expression of miR‐31 (Fig. S2g). Because DCs have higher miR‐31 expression than T‐cells in spleen and DCs are among the first cells to enter the CNS during EAE,11, 54 it is possible that DC entry, and expression of miR‐31, may be requisite for T‐cell engagement and subsequent induction of miR‐31 expression. Moreover, if DC expression of miR‐31 influences BBB permeability, then migration of miR‐31‐expressing DCs could enable entry of T‐cells. To elucidate the complex interplay between miR‐31 expression in DCs and T‐cells, the conditional knockout mice generated by Zhang and colleagues37 should be used to study the role of miR‐31 in CD11c+ DCs during EAE.

More fundamental is the question of what defines DCs versus microglia within the inflamed CNS. Compared with peripheral myeloid cells, microglia have a specific miRNA expression pattern.10 Importantly, use of radiation bone marrow chimeras segregates microglia that express CD11c from BMDCs.11, 35 Our data demonstrate a signature of infiltrating BMDCs within the CNS during EAE distinct from microglia (Fig. 2a). In contrast, miR‐34b‐3p was found to be significantly elevated in the spinal cord of miRAPCD11c → miRAPCD11c chimeric mice, but not in the spinal cord of miRAPCD11c → WT mice (Fig. 2e), suggesting miR‐34b‐3p may signify a subset of microglia adopting a DC phenotype. Differentiating unique miRNA expressed by DCs from microglia represents one essential step toward targeting individual contributions by each innate cell during EAE and MS. This work could be further expanded upon by differentiating BMDCs from DCs derived from CNS‐infiltrating monocytes.

Evidence continues to build for a critical role of miRNA in DC function in MS. Our results indicate miR‐31 may function in directing DC migration into the CNS or may be induced in DCs by the inflamed tissue environment, which could negatively influence DC mRNA expression profiles and/or BBB permeability. Interestingly, the induction of miR‐31 does not seem to be influenced by LPS‐mediated DC activation, indicating potentially separate levels of regulation for DC maturation and migration. Furthermore, while resident microglia can upregulate CD11c in EAE, miR‐31 is sufficient to distinguish BMDCs from these resident immune cells. Finally, our work clarifies the role of miR‐31 in EAE, demonstrating a functional role of miR‐31 in DCs, in addition to the previously described role of miR‐31 in Tregs. This study further highlights the importance of miR‐31 in EAE, and suggests miR‐31 inhibition may serve as a therapeutic for reducing inflammatory responses within the CNS by targeting DC expression.

Disclosure

The authors declare no competing financial interests.

Supporting information

Data S1. Raw microRNA array data and mRNA array data.

Figure S1. Volcano plot of miRNA microarray data generated from spleen and spinal cords of CD11c‐Cre, LSL‐tAgo2 mice following miRAP. Several miRNAs were significantly enriched (P < 0.1, log2FC > 1.5) in CD11c+ DCs in the spinal cord versus spleen. miR31, 339‐5p and miR‐301a/b were selected for validation because they were abundant in both tissues (CT < 30).

Figure S2. MiR‐31 expression in CD3+ T‐cells and CD11c+ cells in EAE spleen and spinal cord.

Figure S3. miR‐31 is elevated in DCs that migrate into inflamed tissue.

Figure S4. miR‐31 regulates IL‐34 expression through 3′‐UTR binding.

Table S1. Primers used to generate putative miR‐31 target 3′‐UTR constructs.

Acknowledgements

MLH, ASA, TMG, LKO and MDC performed experiments. MLH and GFW designed the study. RSK, SDC, BSK and TMM provided new techniques and reagents. MLH, ASA, TMG, MDC, LKO, SDC, BSK, RSK, TMM and GFW edited the manuscript. MLH and GFW wrote the manuscript. The authors thank The Molecular Profiling Facility at The University of Pennsylvania for quantitative mRNA profiling. The authors also thank Cassie A.‐L. Parks for assistance with these studies. The authors are grateful to Dr Anne Cross for critical review of the manuscript. Funding was provided by the Hope Center for Neurologic Diseases, and the National Institute of Neurological Disorders and Stroke [R01NS083678 (GFW), R01NS078398 (TMM), R25NS09098502 (TMG), and F31NS092340 (MLH)].

Contributor Information

Timothy M. Miller, Email: miller.t@wustl.edu

Gregory F. Wu, Email: gfwu@wustl.edu.

References

  • 1. Frohman EM, Racke MK, Raine CS. Multiple sclerosis–the plaque and its pathogenesis. N Engl J Med 2006; 354:942–55. [DOI] [PubMed] [Google Scholar]
  • 2. Rangachari M, Kuchroo VK. Using EAE to better understand principles of immune function and autoimmune pathology. J Autoimmun 2013; 45:31–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Wu GF, Alvarez E. The immunopathophysiology of multiple sclerosis. Neurol Clin 2011; 29:257–78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Goldmann T, Prinz M. Role of microglia in CNS autoimmunity. Clin Dev Immunol 2013; 2013:208 093. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Chastain EM, Duncan DS, Rodgers JM, Miller SD. The role of antigen presenting cells in multiple sclerosis. Biochim Biophys Acta 2011; 1812:265–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. He L, Hannon GJ. MicroRNAs: small RNAs with a big role in gene regulation. Nat Rev Genet 2004; 5:522–31. [DOI] [PubMed] [Google Scholar]
  • 7. Shukla GC, Singh J, Barik S. MicroRNAs: processing, maturation, target recognition and regulatory functions. Mol Cell Pharmacol 2011; 3:83–92. [PMC free article] [PubMed] [Google Scholar]
  • 8. Chan EK, Ceribelli A, Satoh M. MicroRNA‐146a in autoimmunity and innate immune responses. Ann Rheum Dis 2013; 72 (Suppl 2):ii90–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Salama A, Fichou N, Allard M, Dubreil L, De Beaurepaire L, Viel A et al MicroRNA‐29b modulates innate and antigen‐specific immune responses in mouse models of autoimmunity. PLoS ONE 2014; 9:e106153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Butovsky O, Jedrychowski MP, Moore CS, Cialic R, Lanser AJ, Gabriely G et al Identification of a unique TGF‐beta‐dependent molecular and functional signature in microglia. Nat Neurosci 2014; 17:131–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11. Wu GF, Shindler KS, Allenspach EJ, Stephen TL, Thomas HL, Mikesell RJ et al Limited sufficiency of antigen presentation by dendritic cells in models of central nervous system autoimmunity. J Autoimmun 2011; 36:56–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Kim SJ, Gregersen PK, Diamond B. Regulation of dendritic cell activation by microRNA let‐7c and BLIMP1. J Clin Invest 2013; 123:823–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Mildner A, Chapnik E, Manor O, Yona S, Kim KW, Aychek T et al Mononuclear phagocyte miRNome analysis identifies miR‐142 as critical regulator of murine dendritic cell homeostasis. Blood 2013; 121:1016–27. [DOI] [PubMed] [Google Scholar]
  • 14. Huang A, Yang Y, Chen S, Xia F, Sun D, Fang D et al MiR‐34a promotes DCs development and inhibits their function on T cell activation by targeting WNT1. Oncotarget 2017; 8:10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Hoye ML, Koval ED, Wegener AJ, Hyman TS, Yang C, O'Brien DR et al MicroRNA profiling reveals marker of motor neuron disease in ALS models. J Neurosci 2017; 37:5574–86. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. He M, Liu Y, Wang X, Zhang MQ, Hannon GJ, Huang ZJ. Cell‐type‐based analysis of microRNA profiles in the mouse brain. Neuron 2012; 73:35–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Caton ML, Smith‐Raska MR, Reizis B. Notch‐RBP‐J signaling controls the homeostasis of CD8‐ dendritic cells in the spleen. J Exp Med 2007; 204:1653–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Clausen BE, Burkhardt C, Reith W, Renkawitz R, Forster I. Conditional gene targeting in macrophages and granulocytes using LysMcre mice. Transgenic Res 1999; 8:265–77. [DOI] [PubMed] [Google Scholar]
  • 19. Okabe M, Ikawa M, Kominami K, Nakanishi T, Nishimune Y. ‘Green mice’ as a source of ubiquitous green cells. FEBS Lett 1997; 407:313–9. [DOI] [PubMed] [Google Scholar]
  • 20. Lindquist RL, Shakhar G, Dudziak D, Wardemann H, Eisenreich T, Dustin ML et al Visualizing dendritic cell networks in vivo . Nat Immunol 2004; 5:1243–50. [DOI] [PubMed] [Google Scholar]
  • 21. Oetjen LK, Mack MR, Feng J, Whelan TM, Niu H, Guo CJ et al Sensory neurons co‐opt classical immune signaling pathways to mediate chronic itch. Cell 2017; 171:217–28. e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22. Dunnett CW, Tamhane AC. A step‐up multiple test procedure. J Am Statistic Assoc 1992; 87:162–70. [Google Scholar]
  • 23. Idoyaga J, Fiorese C, Zbytnuik L, Lubkin A, Miller J, Malissen B et al Specialized role of migratory dendritic cells in peripheral tolerance induction. J Clin Investig 2013; 123:844–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Archambault AS, Carrero JA, Barnett LG, McGee NG, Sim J, Wright JO et al Cutting edge: conditional MHC class II expression reveals a limited role for B cell antigen presentation in primary and secondary CD4 T cell responses. J Immunol 2013; 191:545–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Daniels BP, Holman DW, Cruz‐Orengo L, Jujjavarapu H, Durrant DM, Klein RS. Viral pathogen‐associated molecular patterns regulate blood‐brain barrier integrity via competing innate cytokine signals. MBio 2014; 5:e01476–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26. Becher B, Bechmann I, Greter M. Antigen presentation in autoimmunity and CNS inflammation: how T lymphocytes recognize the brain. J Mol Med 2006; 84:532–43. [DOI] [PubMed] [Google Scholar]
  • 27. Greter M, Heppner FL, Lemos MP, Odermatt BM, Goebels N, Laufer T et al Dendritic cells permit immune invasion of the CNS in an animal model of multiple sclerosis. Nat Med 2005; 11:328–34. [DOI] [PubMed] [Google Scholar]
  • 28. Wu GF, Laufer TM. The role of dendritic cells in multiple sclerosis. Curr Neurol Neurosci Rep 2007; 7:245–52. [DOI] [PubMed] [Google Scholar]
  • 29. Kaindl AM, Degos V, Peineau S, Gouadon E, Chhor V, Loron G et al Activation of microglial N‐methyl‐D‐aspartate receptors triggers inflammation and neuronal cell death in the developing and mature brain. Ann Neurol 2012; 72:536–49. [DOI] [PubMed] [Google Scholar]
  • 30. Orthgiess J, Gericke M, Immig K, Schulz A, Hirrlinger J, Bechmann I et al Neurons exhibit Lyz2 promoter activity in vivo: implications for using LysM‐Cre mice in myeloid cell research. Eur J Immunol 2016; 46:1529–32. [DOI] [PubMed] [Google Scholar]
  • 31. Goldmann T, Wieghofer P, Muller PF, Wolf Y, Varol D, Yona S et al A new type of microglia gene targeting shows TAK1 to be pivotal in CNS autoimmune inflammation. Nat Neurosci 2013; 16:1618–26. [DOI] [PubMed] [Google Scholar]
  • 32. Mehta A, Baltimore D. MicroRNAs as regulatory elements in immune system logic. Nat Rev Immunol 2016; 16:279–94. [DOI] [PubMed] [Google Scholar]
  • 33. Moore CS, Rao VT, Durafourt BA, Bedell BJ, Ludwin SK, Bar‐Or A et al miR‐155 as a multiple sclerosis‐relevant regulator of myeloid cell polarization. Ann Neurol 2013; 74:709–20. [DOI] [PubMed] [Google Scholar]
  • 34. Murugaiyan G, Beynon V, Mittal A, Joller N, Weiner HL. Silencing microRNA‐155 ameliorates experimental autoimmune encephalomyelitis. J Immunol 2011; 187:2213–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Ponomarev ED, Shriver LP, Maresz K, Dittel BN. Microglial cell activation and proliferation precedes the onset of CNS autoimmunity. J Neurosci Res 2005; 81:374–89. [DOI] [PubMed] [Google Scholar]
  • 36. Bailey SL, Schreiner B, McMahon EJ, Miller SD. CNS myeloid DCs presenting endogenous myelin peptides ‘preferentially’ polarize CD4 + T(H)‐17 cells in relapsing EAE. Nat Immunol 2007; 8:172–80. [DOI] [PubMed] [Google Scholar]
  • 37. Zhang L, Ke F, Liu Z, Bai J, Liu J, Yan S et al MicroRNA‐31 negatively regulates peripherally derived regulatory T‐cell generation by repressing retinoic acid‐inducible protein 3. Nat Commun 2015; 6:7639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Anandasabapathy N, Victora GD, Meredith M, Feder R, Dong B, Kluger C et al Flt3L controls the development of radiosensitive dendritic cells in the meninges and choroid plexus of the steady‐state mouse brain. J Exp Med 2011; 208:1695–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. King IL, Dickendesher TL, Segal BM. Circulating Ly‐6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. Blood 2009; 113:3190–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Li X, Zhang Y, Zhang H, Liu X, Gong T, Li M et al miRNA‐223 promotes gastric cancer invasion and metastasis by targeting tumor suppressor EPB41L3. Mol Cancer Res 2011; 9:824–33. [DOI] [PubMed] [Google Scholar]
  • 41. Pardo OE, Castellano L, Munro CE, Hu Y, Mauri F, Krell J et al miR‐515‐5p controls cancer cell migration through MARK4 regulation. EMBO Rep 2016; 17:570–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Pitzler L, Auler M, Probst K, Frie C, Bergmeier V, Holzer T et al miR‐126‐3p promotes matrix‐dependent perivascular cell attachment, migration and intercellular interaction. Stem Cells 2016; 34:1297–309. [DOI] [PubMed] [Google Scholar]
  • 43. Mitra AK, Chiang CY, Tiwari P, Tomar S, Watters KM, Peter ME et al Microenvironment‐induced downregulation of miR‐193b drives ovarian cancer metastasis. Oncogene 2015; 34:5923–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Subramani A, Alsidawi S, Jagannathan S, Sumita K, Sasaki AT, Aronow B et al The brain microenvironment negatively regulates miRNA‐768‐3p to promote K‐ras expression and lung cancer metastasis. Sci Rep 2013; 3:2392. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Jin S, Sonobe Y, Kawanokuchi J, Horiuchi H, Cheng Y, Wang Y et al Interleukin‐34 restores blood‐brain barrier integrity by upregulating tight junction proteins in endothelial cells. PLoS One 2014; 9:e115981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Ulland TK, Wang Y, Colonna M. Regulation of microglial survival and proliferation in health and diseases. Semin Immunol 2015; 27:410–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Dunand‐Sauthier I, Santiago‐Raber M‐L, Capponi L, Vejnar CE, Schaad O, Irla M et al Silencing of c‐Fos expression by microRNA‐155 is critical for dendritic cell maturation and function. Blood 2011; 117:4490–500. [DOI] [PubMed] [Google Scholar]
  • 48. Kurowska‐Stolarska M, Alivernini S, Melchor EG, Elmesmari A, Tolusso B, Tange C et al MicroRNA‐34a dependent regulation of AXL controls the activation of dendritic cells in inflammatory arthritis. Nat Commun 2017; 8:15 877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Clarkson BD, Walker A, Harris MG, Rayasam A, Sandor M, Fabry Z. CCR2‐dependent dendritic cell accumulation in the central nervous system during early effector experimental autoimmune encephalomyelitis is essential for effector T cell restimulation in situ and disease progression. J Immunol 2015; 194:531–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Jain P, Coisne C, Enzmann G, Rottapel R, Engelhardt B. Alpha4beta1 integrin mediates the recruitment of immature dendritic cells across the blood‐brain barrier during experimental autoimmune encephalomyelitis. J Immunol 2010; 184:7196–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Smyth LA, Boardman DA, Tung SL, Lechler R, Lombardi G. MicroRNAs affect dendritic cell function and phenotype. Immunology 2015; 144:197–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52. Sagar D, Foss C, El Baz R, Pomper MG, Khan ZK, Jain P. Mechanisms of dendritic cell trafficking across the blood‐brain barrier. J Neuroimmune Pharmacol 2012; 7:74–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53. Rhee I, Zhong MC, Reizis B, Cheong C, Veillette A. Control of dendritic cell migration, T cell‐dependent immunity, and autoimmunity by protein tyrosine phosphatase PTPN12 expressed in dendritic cells. Mol Cell Biol 2014; 34:888–99. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54. McMahon EJ, Bailey SL, Castenada CV, Waldner H, Miller SD. Epitope spreading initiates in the CNS in two mouse models of multiple sclerosis. Nat Med 2005; 11:335–9. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Data S1. Raw microRNA array data and mRNA array data.

Figure S1. Volcano plot of miRNA microarray data generated from spleen and spinal cords of CD11c‐Cre, LSL‐tAgo2 mice following miRAP. Several miRNAs were significantly enriched (P < 0.1, log2FC > 1.5) in CD11c+ DCs in the spinal cord versus spleen. miR31, 339‐5p and miR‐301a/b were selected for validation because they were abundant in both tissues (CT < 30).

Figure S2. MiR‐31 expression in CD3+ T‐cells and CD11c+ cells in EAE spleen and spinal cord.

Figure S3. miR‐31 is elevated in DCs that migrate into inflamed tissue.

Figure S4. miR‐31 regulates IL‐34 expression through 3′‐UTR binding.

Table S1. Primers used to generate putative miR‐31 target 3′‐UTR constructs.


Articles from Immunology are provided here courtesy of British Society for Immunology

RESOURCES