Skip to main content

A detrimental role of RelB in mature oligodendrocytes during experimental acute encephalomyelitis

Abstract

Background

Multiple sclerosis (MS) is an autoimmune demyelinating disease of the central nervous system (CNS). It is firmly established that overactivation of the p65 (RelA) nuclear factor kappa B (NF-κB) transcription factor upregulates expression of inflammatory mediators in both immune and non-immune resident CNS cells and promotes inflammation during MS. In contrast to p65, NF-κB family member RelB regulates immune cell development and can limit inflammation. Although RelB expression is induced during inflammation in the CNS, its role in MS remains unknown.

Methods

To examine the role of RelB in non-immune CNS cells, we generated mice with RelB specifically deleted in astrocytes (RelBΔAST), oligodendrocytes (RelBΔOLIGO), or neural progenitor-derived cells (RelBΔNP). We used experimental autoimmune encephalomyelitis (EAE), an accepted mouse model of MS, to assess the effect of RelB deletion on disease outcomes and performed analysis on the histological, cellular, and molecular level.

Results

Despite being a negative regulator of inflammation, conditional knockout of RelB in non-immune resident CNS cells surprisingly decreased the severity of EAE. This protective effect was recapitulated by conditional deletion of RelB in oligodendrocytes but not astrocytes. Deletion of RelB in oligodendrocytes reduced disease severity, promoted survival of mature oligodendrocytes, and correlated with increased activation of p65 NF-κB.

Conclusions

These findings suggest that RelB fine tunes inflammation and cell death/survival during EAE. Importantly, our data points out the detrimental role RelB plays in controlling survival of mature oligodendrocytes, which could be explored as a viable option to treat MS in the future.

Background

Multiple sclerosis (MS) is a chronic inflammatory autoimmune demyelinating disease of the central nervous system (CNS) that manifests with symptoms such as muscle weakness, impaired motor skills and coordination, and sensory loss. Although the primary cause of the disease is not known, demyelination, primarily mediated by T cells that are reactive to myelin antigens, and ongoing inflammation are believed to cause damage to the CNS. MS lesions are commonly characterized by apoptotic loss of oligodendrocytes, loss of myelin-associated glycoproteins, immune cell infiltration, activation of resident glial cells, and axon degeneration [1, 2].

Ubiquitously expressed transcription factors of the nuclear factor kappa B (NF-κB) family regulate many cellular processes, including cell proliferation and survival, production of inflammatory mediators, differentiation of T cells, and maturation of dendritic cells [3, 4]. NF-κB family consists of five members: p65 (RelA), RelB, c-Rel, p50/p105, and p52/p100 that can form a variety of homo- and heterodimers. Under resting conditions, NF-κB proteins are sequestered in the cytoplasm to prevent aberrant activation. A typical inflammatory stimulus activates the canonical NF-κB pathway triggering degradation of inhibitory proteins, such as inhibitor of NF-κB alpha (IκBα), translocation of p65/p50 heterodimers to the nucleus, and induction of p65/p50-dependent genes [5]. It is now firmly established that overactivation of p65 in immune cells contributes to inflammation in MS. There is an increased nuclear localization of p65 in microglia and macrophages in active lesions of human MS patients [6]. Accordingly, an increased p65 DNA binding has been reported in rat spinal cords during experimental autoimmune encephalomyelitis (EAE), which is a rodent model of MS [6, 7]. Furthermore, microarray studies have demonstrated increased expression of NF-κB proteins in MS patients [8]. Conversely, preventing degradation of IκBα decreases the incidence and severity of EAE by decreasing antigen-specific T cell responses [9]. c-Rel, which is a target of the canonical NF-κB pathway, is also crucial for the development of EAE, and c-Rel knockout mice are completely protected from the disease. c-Rel knockout mice are characterized by decreased numbers of Th17 and Th1 autoreactive T cells [10].

In contrast to p65, RelB is mostly known as a target of the non-canonical NF-κB pathway, which is triggered by specific ligands that control development of lymphoid tissues [11]. These ligands induce processing of p100 to p52 and subsequent translocation of RelB/p52 heterodimers to the nucleus [12]. Interestingly, in the presence of high RelB expression [13], RelB/p50/IκBα complexes form and are activated by the canonical pathway [13, 14], but this is limited since RelB has higher affinity for p100/p52 than for p50 [15]. The crucial function of RelB in regulating immunity is best exemplified through knockout animal models. RelB knockout mice have a shorter lifespan and severe autoimmunity [11, 16, 17]. Because RelB plays a crucial role in immune cell development, particularly dendritic cells, RelB knockout mice lack Peyer’s patches, germinal centers, and dendritic cell networks [12, 18]. Furthermore, its loss results in a lack of negative T cell selection, and thus severe multiorgan inflammation with both T cell and monocytic infiltrates [19]. Nevertheless, RelB can also repress p65 activity through multiple mechanisms [20,21,22,23,24], which provides a negative feedback loop suppressing cytokine and chemokine expression. Indeed, RelB has been implicated in establishing tolerance in macrophages during septic shock [22]. We have recently shown that RelB also limits cytokine expression in astrocytes, which limits neuroinflammation [24]. Although RelB plays a crucial role in inflammation and immune cell development, its role has not been studied in animal models of MS.

While peripheral and local immune cells are central to the pathogenesis of MS, it is now evident that resident non-immune CNS cells are also critical. Indeed, activation of astrocytes during MS is an important process that directly contributes to the pathology of the disease [25,26,27]. NF-κB signaling is highly activated in reactive astrocytes and modulates expression of inflammatory mediators, cell death, and cell survival. Accordingly, overexpression of dominant-negative IκBα in astrocytes, which inhibits canonical NF-κB signaling, decreases EAE severity [28]. Multiple intrinsic signaling cascades also modulate susceptibility of oligodendrocytes to injury and thus disease severity [29,30,31]. Blocking p65 activation in oligodendrocytes has recently been found to increase oligodendrocyte death and reduce remyelination [32]. Although it is evident that p65 plays an important role in non-immune cells of the CNS during MS, the role of RelB has not been investigated. RelB is expressed at low levels in astrocytes [14, 24], but its expression is increased in astrocytes during EAE [21]. Since RelB suppresses cytokine expression in astrocytes and thus regulates neuroinflammation [24], we asked whether RelB in non-immune CNS cells modulates severity of EAE. Understanding the role of RelB in the pathogenesis of MS could provide clues for future therapeutic approaches.

Materials and methods

Mice

Mice with the RelB allele flanked by loxP sites generated by Dr. Ulrich Sibienlist (NIH) were bred with GFAP-Cre (Jackson Laboratory) to generate RelB astrocyte-specific conditional knockout mice (RelBΔAST), nestin-Cre (Jackson Laboratory) to generate RelB neural progenitor-specific conditional knockout mice (RelBΔNP), and CNPase-Cre (from Dr. X. Li, Cleveland Clinic, Cleveland, OH) to generate RelB oligodendrocyte-specific knockout mice (RelBΔOLIGO). Mice were housed at Virginia Commonwealth University according to guidelines of the Institutional Animal Care and Use Committee. The mouse protocols were approved by the Institutional Animal Care and Use Committee. Animals were housed in the animal facility, with a 12-h light/dark cycle, and provided water and standard laboratory chow ad libitum. Randomly chosen littermates (males and females) were used for all experiments. All animals were included for data analysis unless they reached a set humane endpoint (20% weight loss) before the end of experiment. The group sizes for each experiment are provided in figure legends. The disease progress was recorded for all experimental animals, while molecular analysis was performed in smaller animal groups that were analyzed using statistics. To establish statistical significance, data were analyzed by ANOVA (multiple comparisons) and both F and p values are indicated in the figure legends. Furthermore, post-hoc Sidak’s test was used for these multiple comparisons (p values from Sidak’s tests are also indicated). For comparisons of two groups, data were analyzed by T test and both F and p values are indicated in the figure legends. Asterisk designates statistical significance with both F and p values indicated.

Experimental autoimmune encephalomyelitis

Each mouse received subcutaneous 200 μg MOG35–55 peptide (AnaSpec) emulsified in CFA containing 500 μg Mycobacterium tuberculosis H37Ra (Difco, Detroit, MI) and intraperitoneal 200 ng pertussis toxin (Enzo Life Sciences, Farmingdale, NY). A booster dose of 200 ng pertussis toxin was administered 2 days after immunization. Mice were clinically scored and weighed daily, and the severity of the disease was quantified using a five-point scale: 0, no symptoms; 1, limp tail; 2, limp tail with loss of righting; 3, paralysis of single hind limb; 4, paralysis of both hind limbs; and 5, death. Two-three independent experiments were performed (as indicated in figure legends), and cumulative data are presented. Tissues were collected at day 15 from PBS- and MOG-injected animals for molecular/cellular analysis.

Cell culture

To prepare mouse cortical astrocytes, cerebral cortices were aseptically dissected and meninges were removed. Tissue was mechanically dissociated, incubated with trypsin and DNaseI at 37 °C for 30 min, and centrifuged. Tissue was filtered through a 70 μm filter and re-centrifuged. Cells were resuspended and plated in dishes pre-coated with poly-d-lysine. Cells were cultured in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal bovine serum, penicillin/streptomycin, sodium pyruvate, and non-essential amino acids.

Western blotting

Cells or ground flash frozen tissue were lysed in 10 mM Tris (pH 7.4), 150 mM sodium chloride, 1 mM EDTA, 1% Nonidet P-40, 1% Triton X-100, 1 mM sodium orthovanadate, 0.2 mM PMSF, and Pierce protease inhibitor mixture. Samples were separated on a 10% gel and transferred onto nitrocellulose membranes (GE Healthcare). Anti-β-tubulin (sc-9104), anti-RelB (sc-226), anti-p65 (sc-372) antibodies (Santa Cruz Biotechnology); anti-GAPDH (5174); and anti-phospho-p65(S536) (3031) antibodies (Cell Signaling) were used. Antigen-antibody complexes were visualized by enhanced chemiluminescence using Immobilon Western blotting kit (Millipore).

Quantitative PCR

Total RNA was prepared from flash frozen tissue with Trizol (Life Technologies), reverse transcribed with the high-capacity cDNA kit (Applied Biosystems), and amplified on the BioRad CFXConnect Real-time System. SYBR Green intron-spanning pre-design qPCR primers (BioRad) were used. Gene expression levels were normalized to GAPDH and represented as fold expression over control.

Immunofluorescence

Animals were perfused with 4% paraformaldehyde, tissue was embedded in optimal cutting medium, and 40 μm frozen sections were prepared. For fluoromyelin staining, slides were rehydrated in PBS for 20 min and then flooded with 1:300 diluted stain for 20 min. Counterstaining was done using Hoechst stain for 5 min, and slides were mounted using vectashield mounting medium (Vector Laboratories). Slides were imaged using the Zeiss LSM 700. For antibody staining, sections were incubated with primary anti-RelB, anti-GFAP, anti-p65, anti-phospho-p65 (all 1:300, Cell Signaling), anti-CC1 (1:200, Millipore), or anti-Iba1 (1:500, Wako) antibodies overnight at 4 °C. Subsequently, sections were incubated with Alexa Fluor 488 or Alexa Fluor 594 secondary antibodies (1:500, Invitrogen) for 1 h at room temperature. Slides were mounted and imaged as described above. No fluorescence crossover was found between the channels, and images were collected separately using the appropriate laser excitation. Images were analyzed using ImageJ.

Immunohistochemistry

Animals were perfused with 4% paraformaldehyde and tissue was paraffin-embedded, sectioned, and H&E-stained at the Cancer Mouse Models Core Facility (VCU, Richmond, VA). Slides were imaged using the Zeiss AxioImager A1 as indicated. Infiltration of immune cells was quantified by counting immune cells in the peripheral regions of the spinal cords.

Isolation and analysis of immune cells in the CNS

Brains from the mice were dissociated using Wheaton Dounce glass tissue grinders, strained through 70 μm filter, and subjected for centrifugation at 1500 rpm for 5 min at 4 °C. Pelleted cells were resuspended in 10 ml of 30% Percoll (Amersham Bioscience) and centrifuged onto a 70% Percoll for 30 min at 2600 rpm. Cells were collected at the 30–70% interface and stained with fluorescence-conjugated monoclonal antibodies against CD45 (clone 30-F11), CD11b (clone M1/70), CD4 (clone GK 1.5), CD8 (clone 53–6.7), and isotype control antibodies (Biolegend) were used to analyze cells.

Statistical analysis

Statistical analysis was performed using GraphPad Prism 7. Values are displayed as mean ± standard error. T tests and ANOVAs were performed as indicated. Post-hoc Sidak’s was used for multiple comparisons.

Results

Deletion of RelB in non-immune CNS cells reduces severity of EAE

Since RelB is upregulated in astrocytes during both EAE [33] and experimental LPS-induced neuroinflammation [24] and limits astrocytic cytokine production [24], we predicted that it may play important functions in non-immune CNS cells during EAE, and its deletion may exaggerate severity of the disease. To test this hypothesis, we crossed nestin-Cre mice with RelBloxp/loxp mice to generate nervous system-restricted RelB conditional knockout mice deficient of RelB in astrocytes, oligodendrocytes, neurons, and adult neural progenitors (Fig. 1a). These mice were born with expected Mendelian ratios (50% WT (RelBloxp/loxp) and 50% RelBΔNP (nestin-Cre; RelBloxp/loxp), were phenotypically normal, and displayed no obvious inflammation or neurological deficits. Surprisingly, RelBΔNP mice had reduced severity of EAE both at initiation and throughout the entire course of the disease (Fig. 1b, Additional file 1: Figure S1). These mice manifested EAE at a later day of onset, demonstrated a later day of disease peak, and had a lower peak score compared to WT littermates (Fig. 1c). Although nestin-Cre driver mice display metabolic phenotype [34], the unexpected reduced severity of the EAE in RelBΔNP mice was a result of RelB deletion since EAE severity was comparable in RelBloxp/loxp mice and nestin-Cre; RelBWT/WT mice (Additional file 1: Figure S2). In agreement with lower clinical scores, the RelBΔNP mice also had decreased immune cell infiltration in the white matter of the lumbar spinal cord, particularly along the ventral tracts (Fig. 2a). Decreased meningeal inflammation was also evident. Furthermore, flow cytometry analysis indicated decreased numbers of brain-infiltrating immune cells, including CD11b+ myeloid cells (both CD11b+ CD45low microglia and CD11b+ CD45high monocytes), and also CD4+ and CD8+ T cells (Fig. 2b, Additional file 1: Table S1). Since RelB negatively regulates cytokine expression in many cell types [35], including astrocytes [24], we examined cytokine mRNA levels in the spinal cords during EAE. RelBΔNP mice expressed anti-inflammatory IL-10 at lower level (Fig. 2c); however, expression of many proinflammatory cytokine and chemokine mRNAs were only incrementally increased in RelBΔNP mice in comparison to RelBloxp/loxp littermates. Thus, although deletion of RelB in the non-immune CNS cells slightly enhances cytokine expression during EAE, paradoxically RelB in these cells is protective.

Fig. 1
figure 1

Ablation of RelB in non-immune CNS cells reduces severity of experimental autoimmune encephalomyelitis. a Deletion of RelB was verified by western blotting in the brains, cultured mouse astrocytes, and spleens from WT (RelBloxP/loxP) and RelBΔNP mice. b, c EAE was induced and clinical scores were recorded for 23 days. n = 5–6 mice per group. b Multiple T tests (*p < 0.05). c T tests: day of onset (F = 4.54, p = 0.038); day of peak (F = 11.50, p = 0.036); peak score (F = 13.20, p = 0.047)

Fig. 2
figure 2

Decreased immune cell infiltration during EAE in RelBΔNP mice. EAE was induced in WT (RelBloxP/loxP) and RelBΔNP mice. Tissues were collected at the peak of disease. a Lumbar spinal cords (L2–L4) (left panels) were stained with hematoxylin and eosin. Quantification of immune cells infiltration into peripheral regions of spinal cords. n = 6, T test: F = 2.024, p = 0.0226). b Flow cytometry was conducted to quantify the indicated cells in the brains. n = 4 mice per group. T tests: CD45 (F = 6.486, p = 0.041); Cd11b+ (F = 1.956, p = 0.014); CD11b+CD45low (F = 3.497, p = 0.026). c Expression of the cytokines was examined by qPCR in lumbar spinal cords. n = 3–5 mice per group. IL-10, two-way ANOVA (F = 12.84, p = 0.004), Sidak’s test (p = 0.005)

Astrocytic RelB has a limited effect on the severity of EAE

Since knockout of RelB in non-immune CNS cells reduced severity of EAE, we sought to determine which cell type is contributing to this phenotype. Astrocytes are critical regulators of immune and inflammatory responses in the brain [36], and they upregulate expression of RelB during EAE [33]. To determine if astrocytic RelB regulates the severity of the disease, we used recently generated RelBΔAST mice [24] and their RelBloxp/loxp littermates in EAE experiments. The RelBΔAST mice are phenotypically normal and are generated with the expected Mendelian distribution. Although the onset of EAE in RelBΔAST mice was delayed in comparison to WT littermates (Fig. 3a), the severity of EAE (peak score and day of disease peak) was similar (Fig. 3b). In concordance with the similar disease severity, both RelBΔAST mice and WT littermates demonstrated a similar level of diffuse inflammatory cell infiltrate in the lumbar spinal cord, as well as some focal areas of immune cell infiltration in the white matter (Fig. 3c). We concluded that astrocytic RelB cannot account for the protective phenotype of RelBΔNP mice, suggesting that RelB plays critical protective role in another cell type.

Fig. 3
figure 3

Astrocyte-restricted ablation of RelB delays the onset of EAE. EAE was induced in WT (RelBloxP/loxP) and RelBΔAST mice. a, b Clinical scores were recorded for 24 days. n = 15–16 mice per group. a Multiple T tests (*p < 0.05). b T tests: day of onset (F = 4.274, p = 0.013); day of peak (F = 2.129, p = 0.184); peak score (F = 1.390, p = 0.554). c At the peak of disease, lumbar spinal cords (L2–L4) (left panels) were stained with hematoxylin and eosin. Quantification of immune cells infiltration into peripheral regions of spinal cords. n = 6, T test: F = 4.879, t test p = 0.824

Deletion of RelB in oligodendrocytes attenuates EAE disease severity

Because death of oligodendrocytes is a hallmark of both MS and EAE [37,38,39], we asked whether deletion of RelB in oligodendrocytes could explain the reduced EAE severity in RelBΔNP mice. We generated oligodendrocyte-specific RelB conditional knockout mice (RelBΔOLIGO) by crossing RelBloxp/loxp mice with CNPase-Cre driver mice. The RelBΔOLIGO mice were phenotypically normal and generated progeny as expected. RelB was efficiently deleted from oligodendrocytes since it no longer was detected in CC1-positive cells of RelBΔOLIGO mice, but it was present in CC1-positive cells of WT littermates (Fig. 4a). Interestingly, induction of EAE in RelBΔOLIGO mice resulted in reduced severity of the disease in comparison to WT littermates (Fig. 4b). Similar to RelBΔNP mice, RelBΔOLIGO mice had a lower peak score and later day of disease onset (Fig. 4c); however, the day of disease peak was not affected. The decreased severity of the EAE in RelBΔOLIGO mice correlated with a decrease in diffuse inflammatory cell infiltrate throughout the lumbar spinal cord with small scattered focal areas of inflammation (Fig. 4d). There was also a trending decrease in immune cell infiltration in the brain as detected by flow cytometry, which was particularly evident for CD4+ T cells (Fig. 4e).

Fig. 4
figure 4

Deletion of RelB in mature oligodendrocytes reduces the severity of EAE. a RelB was visualized in CC1-positive oligodendrocytes in cortical brain sections from WT (RelBloxP/loxP) and RelBΔOLIGO mice by IF. Anti-CC1 and anti-RelB antibodies were used, and sections were counterstained with Hoechst. be EAE was induced in WT and RelBΔOLIGO mice. b, c Clinical scores were recorded for 23 days. n = 5–9 mice per group. b Multiple T tests (*p < 0.05). c T tests: day of onset (F = 1.11, p = 0.01); day of peak (F = 1.619, p = 0.65); peak score (F = 3.659, p = 0.02). d At the peak of disease, lumbar spinal cords were stained with hematoxylin and eosin (L2–L4) (left panels). Quantification of immune cells infiltration into peripheral regions of spinal cords. n = 6, T test: F = 9.212, p = 0.0321. e Flow cytometry was conducted to quantify the indicated cells in the brains. n = 3–4 mice per group, T tests

Oligodendrocyte-restricted ablation of RelB prevents demyelination and decreases loss of mature oligodendrocytes

To determine why RelBΔOLIGO mice develop EAE with reduced severity, we first investigated whether the disease correlates with decreased inflammation characterized by lower expression of cytokines. Activated microglia and reactive astrocytes, but not oligodendrocytes, are the major producers of inflammatory mediators during EAE [40]. Not surprisingly, there was no significant difference between expression of many cytokines and chemokines between RelBΔOLIGO mice and RelBloxp/loxp littermates (Fig. 5a). Although expression of major proinflammatory cytokine mRNAs, such as IL-1β and TNFα, was comparable, IFNγ and IL-10 mRNA levels were interestingly significantly lower in the spinal cords of RelBΔOLIGO mice. In concordance with these findings, numbers of both Iba1+ myeloid cells (Fig. 5b) and GFAP+ reactive astrocytes (Fig. 5c) were significantly diminished in RelBΔOLIGO mice. Furthermore, RelBΔOLIGO mice had a higher intensity of myelin staining in the lumbar spinal cords compared to WT littermates (Fig. 5d). Oligodendrocyte death followed by regeneration from oligodendrocyte progenitor cells (OPCs) is characteristic of EAE and is a determinant of the clinical phenotype of paralysis [30, 41]. Since IFNγ is known to induce apoptosis of oligodendrocyte progenitors [42, 43], we examined the expression of markers for OPCs (NG2, OLIG2) and mature oligodendrocytes (MBP, PLP). Although levels of IFNγ were significantly lower in RelBΔOLIGO mice (Fig. 5a), expression of NG2 and OLIG2 in the lumbar spinal cords were not changed (Fig. 5e), suggesting similar survival of OPCs. In contrast, RelBΔOLIGO mice were characterized by significantly higher expression of MBP and PLP mRNA in the lumbar spinal cords, indicating that deletion of RelB prevents mature oligodendrocyte loss during EAE (Fig. 5e). Since RelB can inhibit p65 [23, 44, 45] and p65 promotes survival of oligodendrocytes in vitro [46, 47] and during EAE [32], we hypothesized that RelBΔOLIGO mice were protected due to exaggerated activation of p65. To evaluate this, we first analyzed expression and activation of p65 in CC1-positive mature oligodendrocytes by immunofluorescence. As expected, there was a greater increase in CC1-positive oligodendrocytes in the spinal cords of RelBΔOLIGO mice than in RelBloxp/loxp littermates (Fig. 5f), which was consistent with the elevated expression of MBP and PLP (Fig. 5e). These CC1-positive oligodendrocytes were also p65-positive (Fig. 6a). Next, we evaluated phosphorylation of p65 on Serine 536 that correlates with p65 activation [48]. We found that activated, phosphorylated p65 was present in the nuclei of CC1-positive oligodendrocytes in both RelBΔOLIGO mice and their WT littermates (Fig. 6b). Importantly, quantification of these data revealed that the percent of CC1-positive cells that co-stain with p-p65(Ser536) was significantly higher during EAE in RelBΔOLIGO mice than in WT littermates (Fig. 6c). These data suggest that activation of p65 in CC1-positive oligodendrocytes is exaggerated in the absence of RelB, promoting survival of mature oligodendrocytes and subsequently limiting severity of EAE in RelBΔOLIGO mice.

Fig. 5
figure 5

Deletion of RelB in oligodendrocytes prevents loss of mature oligodendrocytes and demyelination. EAE was induced in WT (RelBloxP/loxP) and RelBΔOLIGO mice. Tissues were collected at the peak of disease. a, e Expression was analyzed by qPCR in lumbar spine tissue. n = 8–14 mice per group. T tests: IL-10 (F = 4.243, p = 0.025); IFNg (F = 1.384, p = 0.032); MBP (F = 22.57, p = 0.03); PLP (F = 5.116, p = 0.04). b, c Immunofluorescence of lumbar spinal cord sections stained with anti-IBA1 (b) and anti-GFAP (c) antibodies. Sections were counterstained with Hoechst. d Lumbar spinal cord sections stained with fluoromyelin and counterstained with Hoechst. f Immunofluorescence of lumbar spinal cord sections stained with anti-CC1. Quantification of CC1 positive cells (IF). Images were taken at least two regions from three different animals per genotype. Percentages of CC1-positive cells to nuclei labeled with Hoechst. Measurements were calculated manually using the cell counter tool in ImageJ (n = 6–7 per group, T test; (F = 1.846, p = 0.04)

Fig. 6
figure 6

Increased activation of p65 NF-κB in oligodendrocytes of RelBΔOLIGO mice. EAE was induced in WT (RelBloxP/loxP) and RelBΔOLIGO mice. Tissues were collected at the peak of disease. a, b Sections of the lumbar spinal cords were stained with anti-p65, anti-pp65, and anti-CC1 antibodies, and counterstained with Hoechst. Higher magnifications are shown (right panels). c Quantification of pp65/CC1 double-positive cells (IF) in spinal cords of WT (RelBloxP/loxP) and RelBΔOLIGO mice. Percentages were calculated as the ratio of pp65/CC1 double labeled cells per total CC1 cells measured using the cell counter tool in ImageJ. (n = 6–7 per group, T test: (F = 1.359, p = 0.04)

Discussion

RelB has been mostly recognized as a regulator of immune cell development and differentiation that is critical for B cell homeostasis and establishment of lymph nodes and germinal centers [12]. RelB also regulates osteoclast differentiation [49]. These functions of RelB are controlled by the non-canonical NF-κB pathway and RelB/p52 complexes. In contrast, RelB/p50 complexes have recently been identified as critical targets of the necroptosis-independent RIPK3-dependent pathway, promoting inflammation in dendritic cells [50]. Interestingly, oligodendrocyte death involves both necroptosis and RIP3K activation [51, 52]. Over the past decade, RelB also emerged as a critical negative regulator of inflammatory responses in non-lymphoid cells. Multiple mechanisms by which RelB negatively regulates inflammation have been proposed to date, including inhibition of p65 translocation and binding [23], epigenetic silencing of chromatin [22], and dimer switching [44]. RelB also limits production of inflammatory cytokines and curbs inflammatory memory in astrocytes [24]. Indeed, our data confirm that RelB limits cytokine production in non-immune CNS cells during EAE since RelBΔNP mice have increased proinflammatory and decreased anti-inflammatory cytokine expression. Unexpectedly however, RelBΔNP mice displayed reduced severity of EAE indicating that RelB plays other important functions that are in fact not related to the control of cytokine production or inflammation. These conclusions are additionally supported by our findings that deletion of RelB in astrocytes, which are known to produce substantial quantities of inflammatory cytokines [53], has limited effect on the severity of EAE. Furthermore, although oligodendrocytes are not known to produce significant amounts of cytokines, deletion of RelB in these cells significantly reduced severity of EAE in RelBΔOLIGO mice. Thus, the protective effect of RelB deletion in non-immune CNS cells during EAE does not seem to be related to its anti-inflammatory functions but rather non-inflammatory effects in oligodendrocytes. Of note, although we did not find a direct correlation between the severity of EAE and expression of proinflammatory mediators, this can be explained by relatively aggressive disease with high cytokine and chemokine expression in both control and RelB-deficient animals.

We found that RelB exhibits deleterious effects in oligodendrocytes, and these cells are the main targets of autoreactive T cell-mediated response during EAE, which leads to mature oligodendrocyte death, axon degeneration, and increased disease severity [29, 30]. Although deletion of RelB in CNP-expressing oligodendrocytes decreased the amount of IFNγ, known to induce apoptosis of OPCs during EAE [42, 43], it had no effect on the abundance of oligodendrocyte progenitor markers, suggesting similar survival of OPCs. Nevertheless, enhanced differentiation to mature oligodendrocytes in the absence of RelB cannot be fully excluded. Expression of mature oligodendrocyte markers was significantly higher during EAE in RelBΔOLIGO mice. These results suggest that RelB may limit survival of mature oligodendrocytes, promotes their death during EAE, or suppresses differentiation of OPCs.

Previous studies have demonstrated that the canonical activation of p65/p50 complexes promote oligodendrocyte survival during inflammation [32, 46, 47]. Although the exact mechanism remains elusive, p65 is known to control expression of several anti-apoptotic genes including cIAPs, cFLIP, Bcl-2, Bcl-xL, TRAF1, and TRAF2 [4]. Indeed, we found increased activation of p65 in oligodendrocytes during EAE in RelBΔOLIGO mice. The increased p65 activation in oligodendrocytes can be explained by the lack of RelB-mediated inhibition that is accomplished by previously described mechanisms [22,23,24, 44]. However, it is also possible that a proposed p100/IκBδ-mediated crosstalk between canonical and non-canonical NF-κB pathways [54] regulates p65 activation and thus mature oligodendrocyte survival. Since RelB is known to stabilize p100/IκBδ [55], its deletion may limit the amount of p100/IκBδ in oligodendrocytes and thus indirectly remove p100/IκBδ-mediated inhibition of the p65/p50 activation. It is highly probable that all these RelB-dependent mechanisms function in vivo to fine tune inflammation and cell death/survival. Our data suggest that specific targeting of RelB in oligodendrocytes in the future could be explored as a viable option to limit oligodendrocyte loss during MS.

Conclusions

Overall, our data demonstrate that although RelB suppresses cytokine production by non-immune cells in the CNS, it plays an additional deleterious role in controlling survival of mature oligodendrocytes during EAE. Thus, RelB fine tunes inflammation and cell death/survival during EAE, which potentially could be explored as an option to treat MS in the future.

Availability of data and materials

Information regarding the experimental methods used, and the data in this paper are available to scientific communities upon direct contact to the authors. Individual requests for shipment of mice to AAALAC accredited institutions will be honored. An appropriately signed MTA will be required, as well as permission from original sources of RelBLoxp/LoxP mice (NIH).

Abbreviations

CNS:

Central nervous system

EAE:

Experimental autoimmune encephalomyelitis

IκBα:

Inhibitor of NF-κB alpha

MS:

Multiple sclerosis

NF-κB:

Nuclear factor kappa B

OPC:

Oligodendrocyte precursor cells

p50:

Nuclear factor kappa B p50 subunit

p52:

Nuclear factor kappa B p52 subunit

p65:

Nuclear factor kappa B p65 subunit (RelA)

RelB:

Transcription factor RelB

References

  1. McQualter JL, Bernard CC. Multiple sclerosis: a battle between destruction and repair. J Neurochem. 2007;100(2):295–306.

    Article  CAS  PubMed  Google Scholar 

  2. Lucchinetti C, Bruck W, Noseworthy J. Multiple sclerosis: recent developments in neuropathology, pathogenesis, magnetic resonance imaging studies and treatment. Curr Opin Neurol. 2001;14(3):259–69.

    Article  CAS  PubMed  Google Scholar 

  3. Hoffmann A, Natoli G, Ghosh G. Transcriptional regulation via the NF-kappaB signaling module. Oncogene. 2006;25(51):6706–16.

    Article  CAS  PubMed  Google Scholar 

  4. Karin M, Lin A. NF-kappaB at the crossroads of life and death. Nat Immunol. 2002;3(3):221–7.

    Article  CAS  PubMed  Google Scholar 

  5. Shih VF, Tsui R, Caldwell A, Hoffmann A. A single NFkappaB system for both canonical and non-canonical signaling. Cell Res. 2011;21(1):86–102.

    Article  CAS  PubMed  Google Scholar 

  6. Gveric D, Kaltschmidt C, Cuzner ML, Newcombe J. Transcription factor NF-kappaB and inhibitor I kappaBalpha are localized in macrophages in active multiple sclerosis lesions. J Neuropathol Exp Neurol. 1998;57(2):168–78.

    Article  CAS  PubMed  Google Scholar 

  7. Pahan K, Schmid M. Activation of nuclear factor-kB in the spinal cord of experimental allergic encephalomyelitis. Neurosci Lett. 2000;287(1):17–20.

    Article  CAS  PubMed  Google Scholar 

  8. Lock C, Hermans G, Pedotti R, Brendolan A, Schadt E, Garren H, et al. Gene-microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med. 2002;8(5):500–8.

    Article  CAS  PubMed  Google Scholar 

  9. Hwang I, Ha D, Ahn G, Park E, Joo H, Jee Y. Experimental autoimmune encephalomyelitis: association with mutual regulation of RelA (p65)/NF-kappaB and phospho-IkappaB in the CNS. Biochem Biophys Res Commun. 2011;411(2):464–70.

    Article  CAS  PubMed  Google Scholar 

  10. Chen G, Hardy K, Pagler E, Ma L, Lee S, Gerondakis S, et al. The NF-kappaB transcription factor c-Rel is required for Th17 effector cell development in experimental autoimmune encephalomyelitis. J Immunol. 2011;187(9):4483–91.

    Article  CAS  PubMed  Google Scholar 

  11. Burkly L, Hession C, Ogata L, Reilly C, Marconi LA, Olson D, et al. Expression of relB is required for the development of thymic medulla and dendritic cells. Nature. 1995;373(6514):531–6.

    Article  CAS  PubMed  Google Scholar 

  12. Yilmaz ZB, Weih DS, Sivakumar V, Weih F. RelB is required for Peyer's patch development: differential regulation of p52-RelB by lymphotoxin and TNF. EMBO J. 2003;22(1):121–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Shih VF, Davis-Turak J, Macal M, Huang JQ, Ponomarenko J, Kearns JD, et al. Control of RelB during dendritic cell activation integrates canonical and noncanonical NF-kappaB pathways. Nat Immunol. 2012;13(12):1162–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Bhardwaj R, Yester JW, Singh SK, Biswas DD, Surace MJ, Waters MR, et al. RelB/p50 complexes regulate cytokine-induced YKL-40 expression. J Immunol. 2015;194(6):2862–70.

    Article  CAS  PubMed  Google Scholar 

  15. Solan NJ, Miyoshi H, Carmona EM, Bren GD, Paya CV. RelB cellular regulation and transcriptional activity are regulated by p100. J Biol Chem. 2002;277(2):1405–18.

    Article  CAS  PubMed  Google Scholar 

  16. Gerondakis S, Grumont R, Gugasyan R, Wong L, Isomura I, Ho W, et al. Unravelling the complexities of the NF-kappaB signalling pathway using mouse knockout and transgenic models. Oncogene. 2006;25(51):6781–99.

    Article  CAS  PubMed  Google Scholar 

  17. Barton D, HogenEsch H, Weih F. Mice lacking the transcription factor RelB develop T cell-dependent skin lesions similar to human atopic dermatitis. Eur J Immunol. 2000;30(8):2323–32.

    Article  CAS  PubMed  Google Scholar 

  18. Weih DS, Yilmaz ZB, Weih F. Essential role of RelB in germinal center and marginal zone formation and proper expression of homing chemokines. J Immunol. 2001;167(4):1909–19.

    Article  CAS  PubMed  Google Scholar 

  19. Naspetti M, Aurrand-Lions M, DeKoning J, Malissen M, Galland F, Lo D, et al. Thymocytes and RelB-dependent medullary epithelial cells provide growth-promoting and organization signals, respectively, to thymic medullary stromal cells. Eur J Immunol. 1997;27(6):1392–7.

    Article  CAS  PubMed  Google Scholar 

  20. Jacque E, Tchenio T, Piton G, Romeo PH, Baud V. RelA repression of RelB activity induces selective gene activation downstream of TNF receptors. Proc Natl Acad Sci U S A. 2005;102(41):14635–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Kiebala M, Polesskaya O, Yao Z, Perry SW, Maggirwar SB. Nuclear factor-kappa B family member RelB inhibits human immunodeficiency virus-1 Tat-induced tumor necrosis factor-alpha production. PLoS One. 2010;5(7):e11875.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Yoza BK, Hu JY, Cousart SL, Forrest LM, McCall CE. Induction of RelB participates in endotoxin tolerance. J Immunol. 2006;177(6):4080–5.

    Article  CAS  PubMed  Google Scholar 

  23. Marienfeld R, May MJ, Berberich I, Serfling E, Ghosh S, Neumann M. RelB forms transcriptionally inactive complexes with RelA/p65. J Biol Chem. 2003;278(22):19852–60.

    Article  CAS  PubMed  Google Scholar 

  24. Gupta AS, Waters MR, Biswas DD, Brown LN, Surace MJ, Floros C, et al. RelB controls adaptive responses of astrocytes during sterile inflammation. Glia. 2019;67(8):1449–61.

    PubMed  PubMed Central  Google Scholar 

  25. Wang Y, Zhou CF. Involvement of interferon-gamma and its receptor in the activation of astrocytes in the mouse hippocampus following entorhinal deafferentation. Glia. 2005;50(1):56–65.

    Article  PubMed  Google Scholar 

  26. Kang Z, Altuntas CZ, Gulen MF, Liu C, Giltiay N, Qin H, et al. Astrocyte-restricted ablation of interleukin-17-induced Act1-mediated signaling ameliorates autoimmune encephalomyelitis. Immunity. 2010;32(3):414–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Qian Y, Liu C, Hartupee J, Altuntas CZ, Gulen MF, Jane-Wit D, et al. The adaptor Act1 is required for interleukin 17-dependent signaling associated with autoimmune and inflammatory disease. Nat Immunol. 2007;8(3):247–56.

    Article  CAS  PubMed  Google Scholar 

  28. Brambilla R, Persaud T, Hu X, Karmally S, Shestopalov VI, Dvoriantchikova G, et al. Transgenic inhibition of astroglial NF-kappa B improves functional outcome in experimental autoimmune encephalomyelitis by suppressing chronic central nervous system inflammation. J Immunol. 2009;182(5):2628–40.

    Article  CAS  PubMed  Google Scholar 

  29. Prineas JW, Parratt JD. Oligodendrocytes and the early multiple sclerosis lesion. Ann Neurol. 2012;72(1):18–31.

    Article  PubMed  Google Scholar 

  30. Hisahara S, Okano H, Miura M. Caspase-mediated oligodendrocyte cell death in the pathogenesis of autoimmune demyelination. Neurosci Res. 2003;46(4):387–97.

    Article  CAS  PubMed  Google Scholar 

  31. Lin W, Lin Y, Li J, Fenstermaker AG, Way SW, Clayton B, et al. Oligodendrocyte-specific activation of PERK signaling protects mice against experimental autoimmune encephalomyelitis. J Neurosci. 2013;33(14):5980–91.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Stone S, Jamison S, Yue Y, Durose W, Schmidt-Ullrich R, Lin W. NF-kappaB activation protects oligodendrocytes against inflammation. J Neurosci. 2017;37(38):9332–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mayo L, Trauger SA, Blain M, Nadeau M, Patel B, Alvarez JI, et al. Regulation of astrocyte activation by glycolipids drives chronic CNS inflammation. Nat Med. 2014;20(10):1147–56.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Harno E, Cottrell EC, White A. Metabolic pitfalls of CNS Cre-based technology. Cell Metab. 2013;18(1):21–8.

    Article  CAS  PubMed  Google Scholar 

  35. Tully JE, Nolin JD, Guala AS, Hoffman SM, Roberson EC, Lahue KG, et al. Cooperation between classical and alternative NF-kappaB pathways regulates proinflammatory responses in epithelial cells. Am J Respir Cell Mol Biol. 2012;47(4):497–508.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Claycomb KI, Johnson KM, Winokur PN, Sacino AV, Crocker SJ. Astrocyte regulation of CNS inflammation and remyelination. Brain Sci. 2013;3(3):1109–27.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Dowling P, Husar W, Menonna J, Donnenfeld H, Cook S, Sidhu M. Cell death and birth in multiple sclerosis brain. J Neurol Sci. 1997;149(1):1–11.

    Article  CAS  PubMed  Google Scholar 

  38. Wolswijk G. Oligodendrocyte survival, loss and birth in lesions of chronic-stage multiple sclerosis. Brain. 2000;123(Pt 1):105–15.

    Article  PubMed  Google Scholar 

  39. Pender MP, Nguyen KB, McCombe PA, Kerr JF. Apoptosis in the nervous system in experimental allergic encephalomyelitis. J Neurol Sci. 1991;104(1):81–7.

    Article  CAS  PubMed  Google Scholar 

  40. Schmitz T, Chew LJ. Cytokines and myelination in the central nervous system. ScientificWorldJournal. 2008;8:1119–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mc Guire C, Volckaert T, Wolke U, Sze M, de Rycke R, Waisman A, et al. Oligodendrocyte-specific FADD deletion protects mice from autoimmune-mediated demyelination. J Immunol. 2010;185(12):7646–53.

    Article  CAS  PubMed  Google Scholar 

  42. Itoh T, Horiuchi M, Itoh A. Interferon-triggered transcriptional cascades in the oligodendroglial lineage: a comparison of induction of MHC class II antigen between oligodendroglial progenitor cells and mature oligodendrocytes. J Neuroimmunol. 2009;212(1–2):53–64.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Horiuchi M, Itoh A, Pleasure D, Ozato K, Itoh T. Cooperative contributions of interferon regulatory factor 1 (IRF1) and IRF8 to interferon-gamma-mediated cytotoxic effects on oligodendroglial progenitor cells. J Neuroinflammation. 2011;8:8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Saccani S, Pantano S, Natoli G. Modulation of NF-kappaB activity by exchange of dimers. Mol Cell. 2003;11(6):1563–74.

    Article  CAS  PubMed  Google Scholar 

  45. Chen X, El Gazzar M, Yoza BK, McCall CE. The NF-kappaB factor RelB and histone H3 lysine methyltransferase G9a directly interact to generate epigenetic silencing in endotoxin tolerance. J Biol Chem. 2009;284(41):27857–65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Nicholas RS, Wing MG, Compston A. Nonactivated microglia promote oligodendrocyte precursor survival and maturation through the transcription factor NF-kappa B. Eur J Neurosci. 2001;13(5):959–67.

    Article  CAS  PubMed  Google Scholar 

  47. Hamanoue M, Yoshioka A, Ohashi T, Eto Y, Takamatsu K. NF-kappaB prevents TNF-alpha-induced apoptosis in an oligodendrocyte cell line. Neurochem Res. 2004;29(8):1571–6.

    Article  CAS  PubMed  Google Scholar 

  48. Mattioli I, Sebald A, Bucher C, Charles RP, Nakano H, Doi T, et al. Transient and selective NF-kappa B p65 serine 536 phosphorylation induced by T cell costimulation is mediated by I kappa B kinase beta and controls the kinetics of p65 nuclear import. J Immunol. 2004;172(10):6336–44.

    Article  CAS  PubMed  Google Scholar 

  49. Vaira S, Johnson T, Hirbe AC, Alhawagri M, Anwisye I, Sammut B, et al. RelB is the NF-kappaB subunit downstream of NIK responsible for osteoclast differentiation. Proc Natl Acad Sci U S A. 2008;105(10):3897–902.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Moriwaki K, Balaji S, McQuade T, Malhotra N, Kang J, Chan FK. The necroptosis adaptor RIPK3 promotes injury-induced cytokine expression and tissue repair. Immunity. 2014;41(4):567–78.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ofengeim D, Ito Y, Najafov A, Zhang Y, Shan B, DeWitt JP, et al. Activation of necroptosis in multiple sclerosis. Cell Rep. 2015;10(11):1836–49.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Yuan J, Amin P, Ofengeim D. Necroptosis and RIPK1-mediated neuroinflammation in CNS diseases. Nat Rev Neurosci. 2019;20(1):19–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Liddelow SA, Barres BA. Reactive astrocytes: production, function, and therapeutic potential. Immunity. 2017;46(6):957–67.

    Article  CAS  PubMed  Google Scholar 

  54. Shih VF, Kearns JD, Basak S, Savinova OV, Ghosh G, Hoffmann A. Kinetic control of negative feedback regulators of NF-kappaB/RelA determines their pathogen- and cytokine-receptor signaling specificity. Proc Natl Acad Sci U S A. 2009;106(24):9619–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Maier HJ, Marienfeld R, Wirth T, Baumann B. Critical role of RelB serine 368 for dimerization and p100 stabilization. J Biol Chem. 2003;278(40):39242–50.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by NIH grants R21NS100698, R21NS102802, and R01AI093718 (to T.K.) and by the Intramural Research Program of the NIAID/NIH (to U.S). ASG was supported by the F30CA203447 fellowships from the NIH. Microscopy was performed at the VCU Microscopy Facility, supported, in part, by funding from NIH-NCI Cancer Center Support Grant P30 CA016059. Authors state no conflict of interest.

Author information

Authors and Affiliations

Authors

Contributions

ASG planned and performed most experiments, with assistance from DDB, LB, KM, AH, and MM. US created RelBloxP/loxP mice and edited the manuscript. TK conceived the study and contributed to planning of the experiments. TK and ASG drafted the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Tomasz Kordula.

Ethics declarations

Ethics approval

Mice were housed at Virginia Commonwealth University according to guidelines of the Institutional Animal Care and Use Committee (IACUC). The mouse protocols were approved by IACUC.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Additional file

Additional file 1:

Figure S1. Ablation of RelB in non-immune CNS cells reduces severity of EAE. EAE was induced and clinical scores were recorded for 15 days (n = 15–16 mice per group, Figure S2. Nestin-driven expression of CRE has no effect on severity of EAE. EAE was induced and clinical scores were recorded for 23 days (n = 5–8 mice per group, *p < 0.05, T-test). Figure S3. Deletion of RelB in mature oligodendrocytes reduces the severity of EAE. EAE was induced and clinical scores were recorded for 15 days (n = 12–17 mice per group, *p < 0.05, T-test). Table S1. Infiltration of lymphocytes, CD4+, and CD8+ cells into brains during EAE. EAE was induced, clinical scores were recorded, and flow cytometry was conducted to quantify the indicated cells in the brains. n = 4 mice per group. (PDF 240 kb)

Rights and permissions

Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gupta, A.S., Biswas, D.D., Brown, L.S.N. et al. A detrimental role of RelB in mature oligodendrocytes during experimental acute encephalomyelitis. J Neuroinflammation 16, 161 (2019). https://doi.org/10.1186/s12974-019-1548-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s12974-019-1548-7

Keywords