Next Article in Journal
Peroxisome Proliferator FpPEX11 Is Involved in the Development and Pathogenicity in Fusarium pseudograminearum
Next Article in Special Issue
Is There Reduced Hemodynamic Brain Activation in Multiple Sclerosis Even with Undisturbed Cognition?
Previous Article in Journal
Sensitive Determination of Trace 4-Nitrophenol in Ambient Environment Using a Glassy Carbon Electrode Modified with Formamide-Converted Nitrogen-Doped Carbon Materials
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Interferon Beta-1a versus Combined Interferon Beta-1a and Oligodendrocyte-Specific FGFR1 Deletion in Experimental Autoimmune Encephalomyelitis

by
Ranjithkumar Rajendran
1,†,
Vinothkumar Rajendran
1,†,
Liza Gupta
1,
Kian Shirvanchi
1,
Darja Schunin
1,
Srikanth Karnati
2,
Mario Giraldo-Velásquez
3 and
Martin Berghoff
1,*
1
Experimental Neurology, Department of Neurology, University of Giessen, Klinikstrasse 33, 35385 Giessen, Germany
2
Institute of Anatomy and Cell Biology, University of Würzburg, Koellikerstrasse 6, 97080 Würzburg, Germany
3
Department of Neurology, Sozialstiftung Bamberg, Buger Strasse 80, 96049 Bamberg, Germany
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2022, 23(20), 12183; https://doi.org/10.3390/ijms232012183
Submission received: 5 September 2022 / Revised: 30 September 2022 / Accepted: 9 October 2022 / Published: 12 October 2022
(This article belongs to the Special Issue Molecular Signaling Pathways in Brain Pathology 2.0)

Abstract

:
Recombinant beta interferons-1 (IFNβ-1) are used as first line therapies in patients with relapsing multiple sclerosis (MS), a chronic inflammatory and neurodegenerative disease of the CNS. IFNβ-1a/b has moderate effects on the prevention of relapses and slowing of disease progression. Fibroblast growth factors (FGFs) and FGF receptors (FGFRs) are known to play a key role in the pathology of MS and its model EAE. To investigate the effects of short-term treatment with s.c. IFNβ-1a versus the combined application of s.c. IFNβ-1a and oligodendrocyte-specific deletion of FGFR1 (Fgfr1ind−/− mice) in MOG35-55-induced EAE. IFNβ-1a (30 mg/kg) was applied s.c. from days 0–7 p.i. of EAE in controls and Fgfr1ind−/− mice. FGFR signaling proteins associated with inflammation/degeneration in MS/EAE were analyzed by western blot in the spinal cord. Further, FGFR1 in Oli-neu oligodendrocytes were inhibited by PD166866 and treated with IFNβ-1a (400 ng/mL). Application of IFNβ-1a over 8 days resulted in less symptoms only at the peak of disease (days 9–11) compared to controls. Application of IFNβ-1a in Fgfr1ind−/− mice resulted in less symptoms primarily in the chronic phase of EAE. Fgfr1ind−/− mice treated with IFNβ-1a showed increased expression of pERK and BDNF. In Oli-neu oligodendrocytes, treatment with PD166866 and IFNβ-1a also showed an increased expression of pERK and BDNF/TrkB. These data suggest that the beneficial effects in the chronic phase of EAE and on signaling molecules associated with ERK and BDNF expression are caused by the modulation of FGFR1 and not by interferon beta-1a. FGFR may be a potential target for therapy in MS.

1. Introduction

Multiple sclerosis (MS) is a chronic inflammatory and neurodegenerative disease of the central nervous system. Immigration of immune cells into the CNS causes damage of myelin sheaths and oligodendrocytes, and the failure of remyelination plays a key role in this disease [1,2]. Currently approved disease-modifying treatments (DMT) primarily have anti-inflammatory effects in the periphery but do not pass the blood–brain barrier [3]. This also applies to recombinant beta interferons-1 (IFNβ-1a/b), which were the first DMTs available for relapsing MS (RMS) and are still widely applied [3]. Used as a first-line therapy, IFNβ-1a/b has moderate effects on the frequency of relapses and disease progression [3,4,5]. IFNβ-1 acts through a broad spectrum of immunomodulatory effects such as the alteration of antigen presentation, induction of regulatory T cells, reduction of matrix metalloproteinases, and inhibition of autoreactive lymphocytes and Th17 cell differentiation [3,6,7]. Overall, new insights into the underlying mechanisms and cross talk of IFN-β in other signaling pathways may reveal new treatment strategies to exploit the beneficial effects of IFN-β in patients with MS.
Recent research suggests that fibroblast growth factor (FGF)/FGF-receptor (FGFR) signaling pathways modulate the pathogenesis of MS through inflammatory and myelinating effects [8,9,10,11,12]. In post-mortem brain tissue, FGF2+ oligodendrocytes are present in demyelinating lesions, and FGF2 is considered to be an inhibitor of the differentiation of oligodendrocyte progenitor cells (OPC), thus comprising (re-) myelination [10,13], but also exerts neuroprotective effects such as improved neurogenesis [10,13,14]. FGF9 inhibits myelination, induces expression of pro-inflammatory chemokines, and may have an impact on the differentiation of oligodendrocytes [10]. In contrast, FGF1 is assumed to promote remyelination [11]. Oligodendrocyte-specific deletion of FGFR1 in MOG35-55-induces experimental autoimmune encephalomyelitis (EAE), an established mouse model for MS, ameliorates the disease’s course, reduces inflammation in demyelinating lesions, and results in less myelin degeneration and higher axonal density [15,16]. These effects were accompanied by the increased phosphorylation of ERK1/2 and Akt, which is reported to increase myelination [17]. Furthermore, the enhanced expression of BDNF and its receptor TrkB, and a decrease in LINGO-1, which inhibits the differentiation of oligodendrocytes [18], was found. The relevance of FGF/FGFR signaling in EAE was further supported by similar findings after the deletion of oligodendrocyte-specific FGFR2 [9]. Collectively, these data indicate a significant regulation of FGF/FGFR signaling pathways in autoimmune diseases such as MS and EAE.
Recent studies suggest that there exists some crosstalk between FGF and IFNβ pathways. Mice lacking FGFR1 and FGFR2 in keratinocytes overexpress type-I IFN-stimulated genes (ISGs) [19]. In addition, FGF signaling suppresses the basal expression of interferon-stimulated genes (ISGs) in keratinocytes and intestinal epithelial cells [19]. In vitro experiments with keratinocytes show that FGF7 or FGF10 suppress IFN-induced ISG expression via FGFR mediated ERK1/2 and AKT signaling [19]. Further, FGF7 prevents the trafficking of IFN-stimulated STAT1 in human airway epithelia, thus preventing the transcription of several IFN-induced genes [20]. In contrast, in human fetal astrocytes infected with the Zika virus, the blocking of FGF2 signaling leads to the increased expression of IFNβ gene and ISGs [21]. In mouse fibroblast and human cancer cells, Ras/MEK/ERK activation suppresses the transcription of IFN-inducible genes by downregulating IRF1, a transcriptional activator of the IFNβ gene [22], whereas in mouse embryo fibroblasts infected with the myxoma virus, ERK1/2 signaling promotes type-I IFN expression by the activation of IRF3 [23]. In human renal carcinoma cells, type-I IFN downregulates the expression of FGF2 [24]. Taken together, FGF/FGFR signaling by ERK/Akt is involved in Type-I IFN responses, though the exact mechanism of the crosslink remains elusive.
The present study aimed to investigate the effects of short-term treatment with s.c. IFNβ-1a (30 mg/kg over 8 days) versus the combined application of s.c. IFNβ-1a and the oligodendrocyte-specific deletion of FGFR1 (Fgfr1ind−/− mice) in MOG35-55-induced EAE. Compared with the treatment of IFNβ-1a in controls, the application of IFNβ-1a in Fgfr1ind−/− mice significantly reduced symptoms in the chronic phase of EAE, which is characterized by neurodegeneration. Effects on the disease’s course were accompanied by increased ERK phosphorylation and the increased expression of BDNF in the spinal cord, associated with neuroregeneration. In vitro, the combined treatment of Oli-neu oligodendrocytes with the FGFR1-inhibitor PD166866 and IFNβ-1a also resulted in the increased phosphorylation of ERK and increased BDNF/TrkB expression. These data suggest that the effects seen in chronic EAE are mediated by the inhibition of FGFR and not treatment with the beta interferon.

2. Results

2.1. Beneficial Effects in the Chronic Phase of EAE Are Due to the Conditional Deletion of FGFR1

To compare the effects of IFNβ-1a in controls and in Fgfr1ind−/− mice on the clinical course of MOG35-55-induced EAE, both groups received IFNβ-1a (30 mg/kg) from day 0 until day 7 p.i. (Figure 1B). Compared with the untreated controls, s.c. application of IFNβ-1a resulted in less symptoms at the peak of disease (days 11–13 p.i.; p < 0.05). Fgfr1ind−/− mice, which had received IFNβ-1a, showed a less severe disease course after the peak of disease (days 17, 19 p.i.; p < 0.05) and its chronic phase (day 27–36, 43–53 p.i.; p < 0.05) compared to IFNβ-1a treated controls (Figure 1B). Although not statistically significant, controls on IFNβ-1a had a mean EAE score of 2.1 ± 0.46, and Fgfr1ind−/− mice on IFNβ-1a had a mean score of 0.66 ± 0.42 at the end of the experiment (p = 0.056). These data suggest that IFNβ-1a applied to prevent relapses only shows short-term effects at the peak of disease in MOG35-55-induced EAE. The effects seen after the peak of disease and in the chronic phase of EAE are due to conditional FGFR1 gene deletion.

2.2. Increased Expression of pERK and BDNF in Fgfr1ind−/− Mice

To compare the effects of IFNβ-1a on FGF-dependent signaling in controls and Fgfr1ind−/− mice, the proteins ERK/Akt/p38 and TrkB/BDNF were examined by western blot in the spinal cord. On day 62 post-EAE induction, Fgfr1ind−/− mice on IFNβ-1a showed no regulation of FGFR1 protein expression (Figure 2A,B). There were no differences in the expression of the FGFR downstream molecules Akt and P38 or the TrkB receptor (Figure 2A,B). Compared with controls treated with IFNβ-1a, the increased phosphorylation of ERK (p < 0.01) and increased expression of BDNF (p < 0.05) were found (Figure 2A,B). The phosphorylation of STAT1 and STAT3 were not changed in Fgfr1ind−/− (Figure 2A,B). These findings suggest that the effect on the signaling protein pERK and the neurotrophin BDNF are mediated by conditional FGFR1 gene deletion.

2.3. Inhibition of FGFR1 and Application of IFNβ-1a Reduced Proliferation of Oli-Neu Oligodendrocytes

To study whether treatment of Oli-neu oligodendrocytes with the FGFR1 inhibitor PD166866 and/or IFNβ-1a alters the proliferation and cytotoxicity of oligodendrocytes, Oli-neu oligodendrocytes were treated with PD166866 (10 µM), IFNβ-1a (400 ng/mL, Rebif®) or combined IFNβ-1a and PD166866. Less proliferation of oligodendrocytes was found after FGFR1 inhibition (p < 0.001; Figure 3A,B), IFNβ-1a treatment (p < 0.001; Figure 3A,B) or a combined treatment of IFNβ-1a and FGFR1 inhibition (p < 0.001; Figure 3A,B) compared to controls. Increased cytotoxicity was observed after the application of PD166866 (p < 0.005; Figure 3D).

2.4. IFNβ-1a Treatment along with FGFR1 Inhibition Results in Altered FGFR Downstream Signalling

To study whether FGFR1 inhibition and/or treatment with IFNβ-1a regulates FGFR downstream signaling in oligodendrocytes, we analyzed the phosphorylation of ERK/Akt/p38 and the expression of TrkB/BDNF. FGFR1 protein expression was lower after the application of PD166866 (p < 0.01) (Figure 4A,B). ERK phosphorylation was increased by FGFR inhibition (p < 0.05) and the combined treatment of PD166866 and IFNβ-1a (p < 0.01) (Figure 4A,B). Phosphorylation of Akt and p38 was not altered by any of the treatments (Figure 4A,B). STAT1 and STAT3 phosphorylation was increased by IFNβ-1a (pSTAT1: p < 0.05; pSTAT3: p < 0.001), and the combined treatment of IFNβ-1a and PD166866 (pSTAT1: p < 0.001; pSTAT3: p < 0.01 (Figure 4A,B). BDNF and TrkB expression was higher after the combined treatment of IFNβ-1a and PD166866 (BDNF: p < 0.05; TrkB: p < 0.05) (Figure 4A,C). Further, FGFR1 inhibition enhanced TrkB protein expression (p < 0.05). These in vitro results indicate that the combined treatment of IFNβ-1a and PD166866 alters FGFR1 signaling and increases the expression of pERK/TrkB/BDNF in Oli-neu oligodendrocytes.

3. Discussion

Fgfr1ind−/− mice treated with IFNβ-1a showed less symptoms after the peak of disease and the chronic phase of EAE. Application of IFNβ-1a in controls had effects on symptoms at the peak of disease. The beneficial effects on the disease course in Fgfr1ind−/− mice were associated with the activation of ERK phosphorylation and increased expression of the neurotrophin BDNF. In agreement with these EAE data, the in vitro findings showed that treatment of oligodendrocytes with IFNβ-1a or the FGFR1 inhibitor PD166866 enhances the phosphorylation of ERKand increases the expression of BDNF and its receptor TrkB.
IFNβ-1a is a first-line treatment of relapsing MS (RMS) with anti-inflammatory effects in the periphery but not within the CNS [7]. In agreement with these facts, IFNβ-1a was effective at the peak of EAE, when immune cells cross over the blood–brain barrier and cause destruction of oligodendrocytes and myelin in the CNS [1,2]. IFNβ-1a and other substances given for multiple sclerosis have anti-inflammatory effects, reduce the number of relapses, and slow disease progression [3]. Neurodegeneration, which is mainly seen in the chronic phase of EAE, is more difficult to target. In progressive MS, neurodegeneration is the leading pathology, and to date, no substance significantly slows disease progression in these patients.
In MS [11,25,26] and EAE models [9,15,16], FGF signaling pathways have been shown to be involved in the pathology. In MOG35-55-induced EAE, the conditional deletion of FGFR1 or FGFR2 in oligodendrocytes has a number of beneficial effects primarily in the chronic, neurodegenerative phase of EAE. In demyelinating spinal cord lesions, there are fewer lymphocyte and macrophage/microglia infiltrates, and myelin and axons are better preserved [9,15]. The beneficial effects seen in Fgfr1ind−/− mice or Fgfr2ind−/− mice were associated with changes in signaling molecules and the BDNF/TrkB pathway [9,14,15,27]. In the present study, the clinical findings in the chronic phase of EAE could be reproduced.
To extend the knowledge on FGF signaling in EAE, controls and Fgfr1ind−/− mice were both treated with a standard dose of IFNβ-1a from the time of EAE induction. The clinical findings as well as effects on the signaling cascade are similar to those of recent EAE experiments in Fgfr1ind−/− mice or Fgfr2ind−/− mice, suggesting that the findings are due to the conditional deletion of FGFR1 and are not a result of IFNβ-1a application. In this study, the reduction in disease severity in Fgfr1ind−/− mice was accompanied by increased ERK phosphorylation and the increased expression of BDNF. In recent EAE experiments, downstream mediators of mitogen-activated protein kinases (MAPKs) such as ERK1/2 have been shown to regulate myelin formation. Mice without ERK1/2 activity show microglial activation, disturbed axonal integrity, and a partial loss of oligodendrocytes [28]. It is possible that the effects of phosphorylated ERK are mediated by BDNF signaling. Upregulated ERK and IP3/Akt signaling pathways enhance BDNF secretion, followed by the improved viability of neurons [29]. Investigations on the effects of recombinant BDNF on EAE support its neuroregenerative effects [30], whereas dysregulated BDNF secretion in MS patients is related with reduced neuroprotection [31]. Moreover, the increased expression of BDNF in T cells can be detected near demyelinating white matter lesions in EAE, implying that BDNF inhibits parenchymal injury and participates in neuroinflammatory responses during repair processes [32]. These data indicate that enhanced BDNF expression may be a reason for the ameliorated EAE disease course of Fgfr1ind−/− mice.
Like in the EAE model, in vitro findings support the effects of FGFR modulation. The application of an FGFR inhibitor in oligodendrocytes resulted in enhanced BDNF/TrkB expression. It is important to note that BDNF/TrkB signaling is not only mediated by FGFR, but also belongs to the broad spectrum of IFNβ-1a-STAT signaling pathways. Under IFNβ-1a treatment, patients with RR-MS show elevated BDNF serum levels [33,34,35,36,37]. Further, in this study, yhe phosphorylation of STAT1/3 was increased by IFNβ-1a. IFNβ exerts its immunomodulatory, beneficial effects by the induction of T-reg cells and IL-10 secretion by binding to interferon receptors [38,39]. IFN-receptors, in turn, activate the Janus kinase (JAK) to phosphorylate STAT1 and STAT2 [39]. In contrast, in vivo, IFNβ-1a treatment does not regulate STAT1/3 and TrkB expression in the spinal cord at the chronic phase of EAE. The discrepancy/differences between the findings of protein expression from in vitro and in vivo experiments can be explained by (a) the duration of treatment: the time of IFNβ-1a administration in the EAE study and IFNβ-1a exposure in cell cultures, (b) time point: proteins were analyzed 62 days after IFNβ-1a administration and 24 h after IFNβ-1a exposure in cell cultures, (c) concentration: the presence of IFNβ-1a was different in the spinal cord compared to cells exposed in vitro. Besides the activation of the JAK/STAT pathway, recruitment of kinases such as PI3K, Akt, p38, JNK, and ERK corresponds to the increased ERK and BDNF expression in this study [40]. Taken together, EAE and in vitro findings result in increased pERK and BDNF signaling consequently creating a promyelinating and neuroprotective milieu in the CNS tissue, which manifests in an ameliorated EAE disease course.
In summary, this study does not provide evidence for a crosstalk mechanism between IFNβ-1a and FGFR1 signaling. FGFRs may be a potential target for therapy of EAE and patients with MS and may be more promising than treatment with interferon beta-1a.

4. Materials and Methods

4.1. Ethics Statement and Housing Conditions

All scientific procedures on animals were approved by the regional council of Hesse, Giessen, Germany (20/23-Nr. 31/2008) in accordance with the German Animal Welfare Act and the European legislation for the protection of animals used for scientific purposes (2010/63/EU). AVMA guidelines for euthanasia of animals were followed. All efforts were made to minimize pain and suffering. All mice were housed in a controlled environment and kept to a 12 h light/dark cycle. Mice had free access to a standard pellet diet (Altromin 1324 TPF, Altromin Spezialfutter GmbH, Lage, Germany) and autoclaved water ad libitum.

4.2. Generation of FGFR1 Conditional Knockout Mice

Fgfr1ind−/−geflox flox (B6.129S4-Fgfr1tm5.1Sor/J), Plp-creERT (B6.Cg-Tg(Plp1-cre/ERT)3Pop/J) and C57BL/6 mice (Mus musculus) were purchased from The Jackson Laboratories (Bar Harbor, ME, USA). Fgfr1ind−/−geflox flox (B6.129S4-Fgfr1tm5.1Sor/J) mice were crossbred with Plp-creERT (B6.Cg-Tg(Plp1-cre/ERT)3Pop/J) mice and backcrossed to C57BL/6J (Final mice line: B6.Cg-Tg(Plp1-cre/ERT)3Pop Fgfr1tm5.1Sor). The generation of conditional FGFR1 knockout mice was achieved as described earlier [15,16]. Briefly, the genomic DNA was isolated (DirectPCR-Tail, Peqlab, Erlangen, Germany) to identify the deletion of the FGFR1 floxed allele and PLP transgene, and mice were genotyped by PCR. For the induction of Cre recombinase, 4-week-old B6.Cg-Tg(PLP1-cre/ERT)3Pop Fgfr1tm5.1Sor female mice were administered with 1 mg of tamoxifen (Sigma-Aldrich, Steinheim, Germany) in 100 µL sunflower oil/ethanol i.p. over 5 consecutive days (referred to as Fgfr1ind−/−). B6.Cg-Tg(PLP1-cre/ERT)3Pop Fgfr1tm5.1Sor littermate control mice received a sunflower oil/ethanol mixture (referred to as controls).

4.3. EAE Induction and Evaluation of Symptoms

EAE induction and evaluation of symptoms were performed as described earlier [15]. Briefly, EAE was induced in 8-to-12-week-old female Fgfr1ind−/− and control mice by subcutaneous injection of 300 µg myelin oligodendrocyte glycoprotein peptides (MOG35-55; Charité Hospital, Berlin, Germany) emulsified in complete Freund’s adjuvant (Sigma, Steinheim, Germany) containing 10 mg Mycobacterium tuberculosis (Difco, Detroit, MI, USA). Intraperitoneal injections of 300 ng pertussis toxin (Calbiochem, Darmstadt, Germany) were given at the time of immunization and 48 h later. The EAE disease course was monitored until day 62 in a blinded manner. Mice were euthanized at day 62 p.i. for the chronic phase. Mice were evaluated daily in a blinded fashion using the following 5-scale score criteria: 0 to 5 where 0 = normal, 0.5 = distal tail weakness, 1 = complete tail weakness, 1.5 = mild hind limb weakness, 2 = ascending hind limb weakness, 2.5 = severe hind limb weakness, 3 = hind limb paralysis, 3.5 = hind limb paralysis and moderate forelimb weakness, 4 = hind limb paralysis and severe forelimb weakness, 4.5 = tetraplegia and incontinence, and 5 = moribund/death.

4.4. Administration of IFNβ-1a

IFNβ-1a (Rebif®) was kindly provided by Merck Serono GmbH, Darmstadt, Germany. In this study, we used three groups of mice: control, IFNβ-1a alone (referred as IFNβ-1a), and IFNβ-1a treated Fgfr1ind−/− mice (referred as Fgfr1ind−/− IFNβ-1a). IFNβ-1a (30 mg/kg) were subcutaneously injected daily from day 0 to day 7 after induction of EAE (Figure 1A).

4.5. In Vitro Oli-Neu Oligodendrocyte Cultures

Oli-neu oligodendrocyte adherent cell lines were used in this study. Cells were cultured at 37 °C in 5% CO2. The Oli-neu cell line was maintained in DMEM medium supplemented with B27 supplement (Invitrogen), 0.011% sodium pyruvate, 500 nM Tri-lodo-L-Thyronine, 520 nM L-Thyroxine. Poly-L-Lysine was coated in a 75 cm2 culture flask for at least 5 h or overnight, and washed with 1X PBS for 3 times before use. Oli-neu cells were seeded in the Poly-L-Lysine coated flask, and above 80% confluent flasks were used for experiments.

4.6. FGFR1 Inhibitor PD166866 and Application of Interferon Beta 1a in Oli-Neu Cells In Vitro

PD166866, a potent FGFR1 tyrosine kinase inhibitor, was used in this study. PD166866 was purchased from Sigma-Aldrich (Steinheim, Germany) and dissolved in DMSO, and above 80% confluent cells were treated with a 10 µM concentration of PD166866 for 3 h. Effects of IFNβ-1a and FGFR1 inhibition on proliferation, cytotoxicity, FGFR signaling, and TrkB/BDNF protein expression in Oli-neu oligodendrocytes were analyzed. Oli-neu culture was performed as described previously [41]. Briefly, experiments were performed at passages 8–12. To assess the effects of IFNβ-1a and FGFR1 inhibition on cell proliferation, FGFR signaling, and myelin proteins, Oli-neu cells were treated with FGF2 (20 ng/mL), PD166866 (10 μM), IFNβ-1a (400 ng/mL, Rebif®), IFNβ-1a + PD166866, and DMSO. For FGFR signaling activation, stimulation with FGF2 was performed. After 24 h of incubation, protein isolation and western blot were carried out [41].

4.7. Protein Extraction and Western Blot Analysis

Spinal cord tissue was homogenized in lysis buffer with Tissue ruptor (Qiagen Instruments, Hombrechtikon, Switzerland). In vitro experiments were performed upon respective treatments, cells were scraped from the flask, cell pellets were lysed, cell lysates were centrifuged at 14,000 rpm in 4 °C for 30 min, and proteins were collected and stored at −20 °C. Protein concentrations were quantified (Pierce® BCA Protein Assay Kit, Thermo Scientific, Rockford, IL, USA) and normalized. Thirty micrograms of proteins were fractionated by denaturing gel electrophoresis (10% SDS-PAGE), transferred (Trans Blot, Semi dry Transfer cell, BioRad, Hercules, CA, USA) to a nitrocellulose membrane (GE Healthcare, AmershamTM Hybond ECL, Buckinghamshire, UK), and blocked with 5% BSA for 1 h. The membranes were incubated overnight at 4 °C with target-protein-specific primary antibodies, followed by 1 h incubation with respective secondary antibodies (Table 1). The immunoreactive bands of target proteins were detected with SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific, Rockford, IL, USA) using an ECL ChemoCam Imager (INTAS Science Imaging Instruments GmbH, Göttingen, Germany). GAPDH was used as a loading control. Protein band densities were analyzed by ImageJ 1.53b software (National Institute of Health, Bethesda, AR, USA).

4.8. Proliferation Assays

To assess the effects of IFNβ-1a and FGFR1 inhibition on the proliferation of Oli-neu oligodendrocytes, a WST proliferation assay and tryphan blue staining were utilized. Oli-neu oligodendrocytes were seeded and maintained in culture until the cells tightly adhered to the bottom of the plates. Cells were incubated for 24 h with fresh medium containing FGF2 (20 ng/mL), PD166866 (10 µM), IFNβ-1a (400 ng/mL, Rebif®), and IFNβ-1a + PD166866. After 24, 48, and 72 h of incubation, manual counting was performed using a Neubauer improved chamber (Karl Hecht “Assistant”, Altnau TG, Switzerland) and trypan blue dye (Carl Roth, Karlsruhe, Germany) as described by others [42]. For photometric evaluation of proliferation, cells were seeded in a final volume of 100 µL/well (1 × 105 cells/mL) before treatment. After 24 h of treatment, a WST-1 assay (Roche Applied Science, Mannheim, Germany) was carried with 96-well plates (Greiner Bio-One, Frickenhausen, Germany). Absorption was measured by an ELISA-Reader (Multiscan EX, Thermo Fisher Scientific, Langenselbold, Germany) at a wavelength of 405 nm using a reference wavelength of 492 nm.

4.9. Cytotoxicity Assay

Cytotoxic effects of IFNβ-1a and FGFR1 inhibition in Oli-neu oligodendrocytes were studied by the measuring of lactate dehydrogenase (LDH) in the supernatant of incubated cells (as described in proliferation assay) (96-well plate, 0.5 × 104 cells/well) using a prefabricated kit (Cytotoxicity Detection Kit (LDH), Roche Diagnostics, Mannheim, Germany). Cell plates were centrifuged for 10 min at 250× g, and the supernatant was used to measure absorbance at 492 nm (reference wavelength: 620 nm) to calculate cytotoxicity.

4.10. Quantification and Statistical Analysis

All analyses were performed in a blinded fashion. EAE scores were analyzed using a two-way ANOVA with Tukey’s post hoc test. Western blot data were analyzed using a t-test. Statistical analysis and graph preparation was performed using GraphPad Prism 9 (GraphPad Software, San Diego, CA, USA). Graphical displays were prepared using BioRender web application. Data are expressed as the mean ± standard error of mean (SEM). * indicates p ≤ 0.05, ** indicates p ≤ 0.01, *** indicates p ≤ 0.001.

Author Contributions

Conceptualization, M.B., M.G.-V. and R.R.; methodology, M.B., M.G.-V. and R.R.; validation, formal analysis, investigation, R.R., V.R., L.G., S.K. and M.B.; writing—original draft preparation, writing—review and editing, R.R., V.R., K.S., D.S., M.G.-V., S.K. and M.B.; supervision, M.B.; project administration, M.B.; funding acquisition, M.B. All authors have read and agreed to the published version of the manuscript.

Funding

The study was supported by Merck Serono GmbH, Germany.

Institutional Review Board Statement

The study was conducted according to the guidelines of Justus Liebig University, Giessen, and approved by the regional council of Hesse, Giessen, Germany (GI 20/23-Nr. 31/2008) in accordance with the German Animal Welfare Act and the European legislation for the protection of animals.

Data Availability Statement

The data presented in this study are available upon request to corresponding author.

Conflicts of Interest

The authors declare no competing financial interest.

References

  1. Dendrou, C.A.; Fugger, L.; Friese, M.A. Immunopathology of multiple sclerosis. Nat. Rev. Immunol. 2015, 15, 545–558. [Google Scholar] [CrossRef] [PubMed]
  2. Franklin, R.J.M.; Ffrench-Constant, C. Regenerating CNS myelin—From mechanisms to experimental medicines. Nat. Rev. Neurosci. 2017, 18, 753–769. [Google Scholar] [CrossRef] [PubMed]
  3. Haghikia, A.; Hohlfeld, R.; Gold, R.; Fugger, L. Therapies for multiple sclerosis: Translational achievements and outstanding needs. Trends Mol. Med. 2013, 19, 309–319. [Google Scholar] [CrossRef] [PubMed]
  4. Compston, A.; Coles, A. Multiple sclerosis. Lancet 2008, 372, 1502–1517. [Google Scholar] [CrossRef]
  5. Hauser, S.L.; Cree, B.A. Treatment of Multiple Sclerosis: A Review. Am. J. Med. 2020, 133, 1380–1390.e2. [Google Scholar] [CrossRef]
  6. Kasper, L.H.; Reder, A.T. Immunomodulatory activity of interferon-beta. Ann. Clin. Transl. Neurol. 2014, 1, 622–631. [Google Scholar] [CrossRef] [PubMed]
  7. Kieseier, B.C. The mechanism of action of interferon-beta in relapsing multiple sclerosis. CNS Drugs 2011, 25, 491–502. [Google Scholar] [CrossRef]
  8. Furusho, M.; Roulois, A.J.; Franklin, R.J.M.; Bansal, R. Fibroblast growth factor signaling in oligodendrocyte-lineage cells facilitates recovery of chronically demyelinated lesions but is redundant in acute lesions. Glia 2015, 63, 1714–1728. [Google Scholar] [CrossRef] [Green Version]
  9. Kamali, S.; Rajendran, R.; Stadelmann, C.; Karnati, S.; Rajendran, V.; Giraldo-Velasquez, M.; Berghoff, M. Oligodendrocyte-specific deletion of FGFR2 ameliorates MOG35-55-induced EAE through ERK and Akt signalling. Brain Pathol. 2021, 31, 297–311. [Google Scholar] [CrossRef] [PubMed]
  10. Lindner, M.; Thümmler, K.; Arthur, A.; Brunner, S.; Elliott, C.; McElroy, D.; Mohan, H.; Williams, A.; Edgar, J.; Schuh, C.; et al. Fibroblast growth factor signalling in multiple sclerosis: Inhibition of myelination and induction of pro-inflammatory environment by FGF9. Brain 2015, 138, 1875–1893. [Google Scholar] [CrossRef] [Green Version]
  11. Mohan, H.; Friese, A.; Albrecht, S.; Krumbholz, M.; Elliott, C.L.; Arthur, A.; Menon, R.; Farina, C.; Junker, A.; Stadelmann, C.; et al. Transcript profiling of different types of multiple sclerosis lesions yields FGF1 as a promoter of remyelination. Acta Neuropathol.Commun. 2014, 2, 168. [Google Scholar] [CrossRef] [Green Version]
  12. Rajendran, R.; Böttiger, G.; Stadelmann, C.; Karnati, S.; Berghoff, M. FGF/FGFR Pathways in Multiple Sclerosis and in Its Disease Models. Cells 2021, 10, 884. [Google Scholar] [CrossRef] [PubMed]
  13. Thümmler, K.; Rom, E.; Zeis, T.; Lindner, M.; Brunner, S.; Cole, J.J.; Arseni, D.; Mücklisch, S.; Edgar, J.M.; Schaeren-Wiemers, N.; et al. Polarizing receptor activation dissociates fibroblast growth factor 2 mediated inhibition of myelination from its neuroprotective potential. Acta Neuropathol. Commun. 2019, 7, 212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Rottlaender, A.; Villwock, H.; Addicks, K.; Kuerten, S. Neuroprotective role of fibroblast growth factor-2 in experimental autoimmune encephalomyelitis. Immunology 2011, 133, 370–378. [Google Scholar] [CrossRef] [PubMed]
  15. Rajendran, R.; Velásquez, M.G.; Stadelmann, C.; Berghoff, M. Oligodendroglial fibroblast growth factor receptor 1 gene targeting protects mice from experimental autoimmune encephalomyelitis through ERK/AKT phosphorylation. Brain Pathol. 2017, 28, 212–224. [Google Scholar] [CrossRef]
  16. Rajendran, R.; Rajendran, V.; Giraldo-Velasquez, M.; Megalofonou, F.F.; Gurski, F.; Stadelmann, C.; Karnati, S.; Berghoff, M. Oligodendrocyte-Specific Deletion of FGFR1 Reduces Cerebellar Inflammation and Neurodegeneration in MOG35-55-Induced EAE. Int. J. Mol. Sci. 2021, 22, 9495. [Google Scholar] [CrossRef] [PubMed]
  17. Flores, A.I.; Narayanan, S.P.; Morse, E.N.; Shick, H.E.; Yin, X.; Kidd, G.; Avila, R.L.; Kirschner, D.A.; Macklin, W.B. Constitutively Active Akt Induces Enhanced Myelination in the CNS. J. Neurosci. 2008, 28, 7174–7183. [Google Scholar] [CrossRef] [Green Version]
  18. Mi, S.; Hu, B.; Hahm, K.; Luo, Y.; Hui, E.S.K.; Yuan, Q.; Wong, W.M.; Wang, L.; Su, H.; Chu, T.-H.; et al. LINGO-1 antagonist promotes spinal cord remyelination and axonal integrity in MOG-induced experimental autoimmune encephalomyelitis. Nat. Med. 2007, 13, 1228–1233. [Google Scholar] [CrossRef] [PubMed]
  19. Maddaluno, L.; Urwyler, C.; Rauschendorfer, T.; Meyer, M.; Stefanova, D.; Spörri, R.; Wietecha, M.; Ferrarese, L.; Stoycheva, D.; Bender, D.; et al. Antagonism of interferon signaling by fibroblast growth factors promotes viral replication. EMBO Mol. Med. 2020, 12, e11793. [Google Scholar] [CrossRef]
  20. Prince, L.S.; Karp, P.H.; Moninger, T.O.; Welsh, M.J. KGF alters gene expression in human airway epithelia: Potential regulation of the inflammatory response. Physiol. Genom. 2001, 6, 81–89. [Google Scholar] [CrossRef]
  21. Limonta, D.; Jovel, J.; Kumar, A.; Lu, J.; Hou, S.; Airo, A.M.; Lopez-Orozco, J.; Wong, C.P.; Saito, L.; Branton, W.; et al. Fibroblast Growth Factor 2 Enhances Zika Virus Infection in Human Fetal Brain. J. Infect. Dis. 2019, 220, 1377–1387. [Google Scholar] [CrossRef]
  22. Komatsu, Y.; Christian, S.L.; Ho, N.; Pongnopparat, T.; Licursi, M.; Hirasawa, K. Oncogenic Ras inhibits IRF1 to promote viral oncolysis. Oncogene 2014, 34, 3985–3993. [Google Scholar] [CrossRef] [Green Version]
  23. Wang, F.; Ma, Y.; Barrett, J.W.; Gao, X.; Loh, J.; Barton, E.; Virgin, H.W.; McFadden, G. Disruption of Erk-dependent type I interferon induction breaks the myxoma virus species barrier. Nat. Immunol. 2004, 5, 1266–1274. [Google Scholar] [CrossRef]
  24. Singh, R.K.; Gutman, M.; Bucana, C.D.; Sanchez, R.; Llansa, N.; Fidler, I.J. Interferons alpha and beta down-regulate the expression of basic fibroblast growth factor in human carcinomas. Proc. Natl. Acad. Sci. USA 1995, 92, 4562–4566. [Google Scholar] [CrossRef] [Green Version]
  25. Clemente, D.; Ortega, M.C.; Arenzana, F.J.; De Castro, F. FGF-2 and Anosmin-1 Are Selectively Expressed in Different Types of Multiple Sclerosis Lesions. J. Neurosci. 2011, 31, 14899–14909. [Google Scholar] [CrossRef] [Green Version]
  26. Sarchielli, P.; Di Filippo, M.; Ercolani, M.V.; Chiasserini, D.; Mattioni, A.; Bonucci, M.; Tenaglia, S.; Eusebi, P.; Calabresi, P. Fibroblast growth factor-2 levels are elevated in the cerebrospinal fluid of multiple sclerosis patients. Neurosci. Lett. 2008, 435, 223–228. [Google Scholar] [CrossRef]
  27. Ruffini, F.; Furlan, R.; Poliani, P.; Brambilla, E.; Marconi, P.; Bergami, A.; Desina, G.; Glorioso, J.; Comi, G.; Martino, G. Fibroblast growth factor-II gene therapy reverts the clinical course and the pathological signs of chronic experimental autoimmune encephalomyelitis in C57BL/6 mice. Gene Ther. 2001, 8, 1207–1213. [Google Scholar] [CrossRef] [Green Version]
  28. Ishii, A.; Furusho, M.; Dupree, J.L.; Bansal, R. Role of ERK1/2 MAPK signaling in the maintenance of myelin and axonal integrity in the adult CNS. J. Neurosci. 2014, 34, 16031–16045. [Google Scholar] [CrossRef] [Green Version]
  29. Almeida, R.D.; Manadas, B.J.; Melo, C.V.; Gomes, J.R.; Mendes, C.S.; Graos, M.M.; Carvalho, R.F.; Carvalho, A.P.; Duarte, C.B. Neuroprotection by BDNF against glutamate-induced apoptotic cell death is mediated by ERK and PI3-kinase pathways. Cell Death Differ. 2005, 12, 1329–1343. [Google Scholar] [CrossRef] [Green Version]
  30. Kopec, B.M.; Kiptoo, P.; Zhao, L.; Rosa-Molinar, E.; Siahaan, T.J. Noninvasive Brain Delivery and Efficacy of BDNF to Stimulate Neuroregeneration and Suppression of Disease Relapse in EAE Mice. Mol. Pharm. 2020, 17, 404–416. [Google Scholar] [CrossRef]
  31. Azoulay, D.; Urshansky, N.; Karni, A. Low and dysregulated BDNF secretion from immune cells of MS patients is related to reduced neuroprotection. J. Neuroimmunol. 2008, 195, 186–193. [Google Scholar] [CrossRef] [PubMed]
  32. Wang, N.; Tian, B. Brain-derived neurotrophic factor in autoimmune inflammatory diseases (Review). Exp. Ther. Med. 2021, 22, 1292. [Google Scholar] [CrossRef] [PubMed]
  33. Azoulay, D.; Mausner-Fainberg, K.; Urshansky, N.; Fahoum, F.; Karni, A. Interferon-beta therapy up-regulates BDNF secretion from PBMCs of MS patients through a CD40-dependent mechanism. J. Neuroimmunol. 2009, 211, 114–119. [Google Scholar] [CrossRef] [PubMed]
  34. Caggiula, M.; Batocchi, A.P.; Frisullo, G.; Angelucci, F.; Patanella, A.K.; Sancricca, C.; Nociti, V.; Tonali, P.A.; Mirabella, M. Neurotrophic factors in relapsing remitting and secondary progressive multiple sclerosis patients during interferon beta therapy. Clin. Immunol. 2006, 118, 77–82. [Google Scholar] [CrossRef] [PubMed]
  35. Dhib-Jalbut, S.; Sumandeep, S.; Valenzuela, R.; Ito, K.; Patel, P.; Rametta, M. Immune response during interferon beta-1b treatment in patients with multiple sclerosis who experienced relapses and those who were relapse-free in the START study. J. Neuroimmunol. 2013, 254, 131–140. [Google Scholar] [CrossRef]
  36. Hamamcioglu, K.; Reder, A.T. Interferon-beta regulates cytokines and BDNF: Greater effect in relapsing than in progressive multiple sclerosis. Mult. Scler. 2007, 13, 459–470. [Google Scholar] [CrossRef]
  37. Mehrpour, M.; Akhoundi, F.H.; Delgosha, M.; Keyvani, H.; Motamed, M.R.; Sheibani, B.; Meysamie, A. Increased Serum Brain-derived Neurotrophic Factor in Multiple Sclerosis Patients on Interferon-beta and Its Impact on Functional Abilities. Neurologist 2015, 20, 57–60. [Google Scholar] [CrossRef] [PubMed]
  38. Cauwels, A.; Van Lint, S.; Rogge, E.; Verhee, A.; Eeckhout, B.V.D.; Pang, S.; Prinz, M.; Kley, N.; Uzé, G.; Tavernier, J. Targeting IFN activity to both B cells and plasmacytoid dendritic cells induces a robust tolerogenic response and protection against EAE. Sci. Rep. 2021, 11, 21575. [Google Scholar] [CrossRef] [PubMed]
  39. Platanias, L.C. Mechanisms of type-I- and type-II-interferon-mediated signalling. Nat. Rev. Immunol. 2005, 5, 375–386. [Google Scholar] [CrossRef] [PubMed]
  40. van Boxel-Dezaire, A.H.; Rani, M.S.; Stark, G.R. Complex Modulation of Cell Type-Specific Signaling in Response to Type I Interferons. Immunity 2006, 25, 361–372. [Google Scholar] [CrossRef] [PubMed]
  41. Rajendran, R.; Böttiger, G.; Dentzien, N.; Rajendran, V.; Sharifi, B.; Ergün, S.; Stadelmann, C.; Karnati, S.; Berghoff, M. Effects of FGFR Tyrosine Kinase Inhibition in OLN-93 Oligodendrocytes. Cells 2021, 10, 1318. [Google Scholar] [CrossRef] [PubMed]
  42. Strober, W. Trypan Blue Exclusion Test of Cell Viability. Curr. Protoc. Immunol. 2015, 111, A3.B.1–A3.B.3. [Google Scholar] [CrossRef]
Figure 1. Experimental design and the clinical outcome of EAE scores. (A) Tamoxifen-induced FGFR1 knockout was created in 4-week-old mice, and at 8 weeks of age, control and Fgfr1ind−/− mice were immunized subcutaneously with MOG35-55-peptide emulsified in CFA. Both mice group received 30 mg/kg IFNβ-1a from day 0–7 post-EAE-induction, and the development of neurological symptoms was monitored for 62 days. (B) A mean EAE clinical score of untreated control mice (n = 5), IFNβ-1a-treated control mice (n = 5) compared with IFNβ-1a-treated Fgfr1ind−/− mice (n = 6). IFNβ-1a-treated Fgfr1ind−/− mice showed a significant reduction of clinical symptoms. Data are expressed as the mean + SEM for each group. * p < 0.05.
Figure 1. Experimental design and the clinical outcome of EAE scores. (A) Tamoxifen-induced FGFR1 knockout was created in 4-week-old mice, and at 8 weeks of age, control and Fgfr1ind−/− mice were immunized subcutaneously with MOG35-55-peptide emulsified in CFA. Both mice group received 30 mg/kg IFNβ-1a from day 0–7 post-EAE-induction, and the development of neurological symptoms was monitored for 62 days. (B) A mean EAE clinical score of untreated control mice (n = 5), IFNβ-1a-treated control mice (n = 5) compared with IFNβ-1a-treated Fgfr1ind−/− mice (n = 6). IFNβ-1a-treated Fgfr1ind−/− mice showed a significant reduction of clinical symptoms. Data are expressed as the mean + SEM for each group. * p < 0.05.
Ijms 23 12183 g001
Figure 2. Protein expression of FGFR signaling and TrkB/BDNF in spinal cord lysates at day 62 p.i. (A,B). Protein expression of FGFR1, pAKT, pP38, pSTAT1/3, and TrkB were not different between IFNβ-1a-treated controls and IFNβ-1a-treated Fgfr1ind−/− mice. The increased phosphorylation of ERK and increased BDNF protein expression was found in IFNβ-1a-treated Fgfr1ind−/− mice’s spinal cords compared to the control mice. n = 4/group. Data are expressed as the mean ± SEM. ns = not significant, * p < 0.05, ** p < 0.01.
Figure 2. Protein expression of FGFR signaling and TrkB/BDNF in spinal cord lysates at day 62 p.i. (A,B). Protein expression of FGFR1, pAKT, pP38, pSTAT1/3, and TrkB were not different between IFNβ-1a-treated controls and IFNβ-1a-treated Fgfr1ind−/− mice. The increased phosphorylation of ERK and increased BDNF protein expression was found in IFNβ-1a-treated Fgfr1ind−/− mice’s spinal cords compared to the control mice. n = 4/group. Data are expressed as the mean ± SEM. ns = not significant, * p < 0.05, ** p < 0.01.
Ijms 23 12183 g002
Figure 3. The proliferation and cytotoxicity effects of IFNβ-1a and FGFR1 inhibition by PD166866 in oligodendrocytes in vitro. Oli-neu oligodendrocyte cells were incubated for 24 h with culture medium containing FGF2 (20 ng/mL), PD166866 (10 µM), IFNβ-1a (400 ng/mL, Rabiff), IFNβ-1a + PD166866. (AC) Proliferation were analyzed by counting the number of cells using light microscopy on day 1, 2, and 3. Cell counting assays showed FGFR1 inhibition by PD166866, IFNβ-1a, and IFNβ-1a + PD166866 decreases the proliferation of Oli-neu cells. (D) LDH cytotoxicity assays showed cytotoxicity effects of FGFR1 inhibition by PD166866. Representative microscopic images of Oli-neu cells under different treatments are shown in 20× magnification (A). Data are presented as mean ± SEM from three independent experiments, * p < 0.05, *** p < 0.001.
Figure 3. The proliferation and cytotoxicity effects of IFNβ-1a and FGFR1 inhibition by PD166866 in oligodendrocytes in vitro. Oli-neu oligodendrocyte cells were incubated for 24 h with culture medium containing FGF2 (20 ng/mL), PD166866 (10 µM), IFNβ-1a (400 ng/mL, Rabiff), IFNβ-1a + PD166866. (AC) Proliferation were analyzed by counting the number of cells using light microscopy on day 1, 2, and 3. Cell counting assays showed FGFR1 inhibition by PD166866, IFNβ-1a, and IFNβ-1a + PD166866 decreases the proliferation of Oli-neu cells. (D) LDH cytotoxicity assays showed cytotoxicity effects of FGFR1 inhibition by PD166866. Representative microscopic images of Oli-neu cells under different treatments are shown in 20× magnification (A). Data are presented as mean ± SEM from three independent experiments, * p < 0.05, *** p < 0.001.
Ijms 23 12183 g003
Figure 4. Effect of IFNβ-1a and FGFR1 inhibition by PD166866 in FGFR downstream signaling proteins in oligodendrocytes in vitro. Oli-neu oligodendrocyte cells were incubated with culture medium containing FGF2 (20 ng/mL), PD166866 (10 µM), IFNβ-1a (400 ng/mL, Rabiff), IFNβ-1a + PD166866. (A,B) FGFR1 protein expression was significantly lower in cells treated with PD166866 compared to controls. Protein expression of pERK, BDNF (C; PD with IFN) and TrkB were increased after PD166866, IFNβ-1a, and PD166866 with IFNβ-1a. Increased phosphorylation of STAT1 and 3 were found in cells treated with PD166866 as well as PD166866 with IFNβ-1a. (D) Overview of the effect of IFNβ-1a and FGFR1 inhibition by PD166866 in FGFR downstream signaling proteins in oligodendrocytes. Data are presented as mean ± SEM from three independent experiments, * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001.
Figure 4. Effect of IFNβ-1a and FGFR1 inhibition by PD166866 in FGFR downstream signaling proteins in oligodendrocytes in vitro. Oli-neu oligodendrocyte cells were incubated with culture medium containing FGF2 (20 ng/mL), PD166866 (10 µM), IFNβ-1a (400 ng/mL, Rabiff), IFNβ-1a + PD166866. (A,B) FGFR1 protein expression was significantly lower in cells treated with PD166866 compared to controls. Protein expression of pERK, BDNF (C; PD with IFN) and TrkB were increased after PD166866, IFNβ-1a, and PD166866 with IFNβ-1a. Increased phosphorylation of STAT1 and 3 were found in cells treated with PD166866 as well as PD166866 with IFNβ-1a. (D) Overview of the effect of IFNβ-1a and FGFR1 inhibition by PD166866 in FGFR downstream signaling proteins in oligodendrocytes. Data are presented as mean ± SEM from three independent experiments, * p < 0.05, ** p < 0.01, *** p < 0.001, **** p < 0.0001.
Ijms 23 12183 g004
Table 1. List of antibodies used for Western blot.
Table 1. List of antibodies used for Western blot.
NameHostMol. WeightMethodArt. NoManufacturer
Primary Antibodies
Anti-pERKRabbit44, 42 kDaWB4370sCell Signaling Tech., Danvers, MA, USA
Anti-pAktRabbit60 kDaWB4060sCell Signaling Tech., Danvers, MA, USA
Anti-pP38Rabbit43 kDaWB9212sCell Signaling Tech., Danvers, MA, USA
Anti-pSTAT1Rabbit84, 91 kDaWB9171sCell Signaling Tech., Danvers, MA, USA
Anti-pSTAT3Rabbit79, 86WB9145sCell Signaling Tech., Danvers, MA, USA
Anti-FGFR1Rabbit110 kDaWBsc-57132Santa Cruz Biotech., Dallas, CA, USA
Anti-Trk BRabbit145 kDaWBsc-377218Santa Cruz Biotech., Dallas, CA, USA
Anti-BDNFRabbit14 kDaWBsc-65514Santa Cruz Biotech., Dallas, CA, USA
Anti-GAPDHMouse37 kDaWBsc-365062Santa Cruz Biotech., Dallas, CA, USA
Secondary Antibodies
Anti-rabbit-HRPGoat 7074Cell Signaling Tech., Danvers, MA, USA
Anti-mouse-HRPHorse 7076Cell Signaling Tech., Danvers, MA, USA
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Rajendran, R.; Rajendran, V.; Gupta, L.; Shirvanchi, K.; Schunin, D.; Karnati, S.; Giraldo-Velásquez, M.; Berghoff, M. Interferon Beta-1a versus Combined Interferon Beta-1a and Oligodendrocyte-Specific FGFR1 Deletion in Experimental Autoimmune Encephalomyelitis. Int. J. Mol. Sci. 2022, 23, 12183. https://doi.org/10.3390/ijms232012183

AMA Style

Rajendran R, Rajendran V, Gupta L, Shirvanchi K, Schunin D, Karnati S, Giraldo-Velásquez M, Berghoff M. Interferon Beta-1a versus Combined Interferon Beta-1a and Oligodendrocyte-Specific FGFR1 Deletion in Experimental Autoimmune Encephalomyelitis. International Journal of Molecular Sciences. 2022; 23(20):12183. https://doi.org/10.3390/ijms232012183

Chicago/Turabian Style

Rajendran, Ranjithkumar, Vinothkumar Rajendran, Liza Gupta, Kian Shirvanchi, Darja Schunin, Srikanth Karnati, Mario Giraldo-Velásquez, and Martin Berghoff. 2022. "Interferon Beta-1a versus Combined Interferon Beta-1a and Oligodendrocyte-Specific FGFR1 Deletion in Experimental Autoimmune Encephalomyelitis" International Journal of Molecular Sciences 23, no. 20: 12183. https://doi.org/10.3390/ijms232012183

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop