Next Article in Journal
Drug-Induced Anaphylaxis in Children
Next Article in Special Issue
Putting Functional Gastrointestinal Disorders within the Spectrum of Inflammatory Disorders Can Improve Classification and Diagnostics of These Disorders
Previous Article in Journal
Urinary Viral Spectrum in Patients with Interstitial Cystitis/Bladder Pain Syndrome and the Clinical Efficacy of Valacyclovir Treatment
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Interferon Lambda Signaling Restrains Experimental Autoimmune Encephalomyelitis

1
Department of Dermatology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
2
Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
*
Authors to whom correspondence should be addressed.
Biomedicines 2024, 12(3), 526; https://doi.org/10.3390/biomedicines12030526
Submission received: 10 January 2024 / Revised: 19 February 2024 / Accepted: 21 February 2024 / Published: 26 February 2024

Abstract

:
IFN-λ is a type III interferon (IFN) with pleiotropic functions in modulating immune responses. To address its function in autoimmune neuroinflammation, we evaluated the development and progression of experimental autoimmune encephalitis (EAE) in IFNLR1 KO (Ifnlr1−/−) and C57Bl/6 (WT) mice following immunization with MOG35–55 peptide. The results show that Ifnlr1−/− mice developed significantly more severe EAE than WT littermates with a similar day of onset, suggesting the potential of IFN-λ in reducing disease severity. We next interrogated whether IFN-λ differentially modulates EAE induced by encephalitogenic Th1 cells or Th17 cells. Encephalitogenic Th1 or Th17 generated from WT donors were transferred into WT or Ifnlr1−/− recipient mice. Whereas encephalitogenic Th1 cells induced more severe EAE in Ifnlr1−/− than WT recipients, the disease severity induced by encephalitogenic Th17 cells was similar. Additionally, in vitro experiments showed that Ifnlr1−/− macrophages promoted the expansion of myelin peptide-reactive Th17 cells but not Th1 cells. Early in the disease, the spinal cords of EAE mice displayed a significantly greater proportion of Ly6CLy6G+ cells with CXCR2+CD62Llo phenotype, indicating activated neutrophils. These findings suggest that IFN-λ signaling restrains activation and migration of neutrophils to the CNS, potentially attenuating neutrophil-mediated disease progression in autoimmune neuroinflammation. Recombinant IFN-λ can be used as a potential therapeutic target for treatment of patients with multiple sclerosis as it has fewer side effects due to the restricted expression of its receptor.

1. Introduction

Type III interferons (IFNs), including IFN-λ1 through λ4, signal via a heterodimeric receptor complex consisting of interferon lambda receptor 1 (IFNLR1 or IL-28Rα) and interleukin-10 receptor 2 (IL-10R2) [1]. IFNLR1 is expressed on epithelial cells, neutrophils, dendritic cells, macrophages, and B cells [2,3]. IFNLR1 selectively binds type III IFNs, whereas IL10R2 is shared with other members of the IL-10 family. From a canonical signaling standpoint, signaling through the type I IFNR (IFNAR) and type III IFNR (IFNLR) are very similar in that both activate the Janus kinases (JAKs), tyrosine kinase 2 (TYK2) and JAK1, and the signal transducers and activators of transcription (STAT1 and STAT2) [3]. However, there are distinct differences in the outcome, dependent on cell type and tissue and the level of expression of IFNLR [3,4,5,6]. IFN-λ signaling in neutrophils is predominantly regulatory through mechanisms that include the inhibition of reactive oxygen species (ROS), NETosis, IL-1β expression, cell migration, expression of the transcription factor IRF1, and others [3,4,5,6,7,8,9,10]. The regulatory role of IFN-λ signaling in neutrophils has been reported in several autoimmune disorders, including multiple sclerosis (MS), systemic lupus erythematosus (SLE), and rheumatoid arthritis (RA) [3,7,11,12,13]. Recent research demonstrated the role of IFN-λ in the pathogenesis of central nervous system (CNS) autoimmune disease [14]. In the experimental autoimmune encephalomyelitis (EAE) model of MS, Ifnlr1−/− animals exhibited less severe clinical disease and decreased spinal cord axonal injury compared with wild-type (WT) animals. The underlying biological mechanism was attributed to myeloid cells [14].
To investigate the role of IFN-λ in EAE, we induced EAE in Ifnlr1−/− mice. Loss of IFNLR resulted in severe EAE in mice in comparison to their WT littermates. The effect was dependent on IFNLR1 expression on non-T cells. We found that specific neutrophil populations contributed to the development of autoimmune inflammation in the EAE model. Overall, the presented data indicate a role for IFN-λ in restraining neutrophils in the EAE model.

2. Materials and Methods

Mice. C57BL/6 mice were purchased from Jackson Laboratory (Bar Harbor, ME, USA) and bred at UAB. IFNLR1 knockout (KO) mice (Ifnlr1−/−) were provided by Dr. Herbert Virgin (Washington University School of Medicine, St. Louis, MO, USA). All animals were housed and treated in accordance with the National Institutes of Health guidelines. The approval number from the UAB Institutional Animal Use and Care Committee (IACUC) is 20631.
EAE induction. Age- and sex-matched wild-type (WT) C57BL/6 and Ifnlr1−/− mice were induced for EAE as described previously [15]. The emulsion, composed of 150 μg of MOG35–55 peptide (sourced from GL Biochem, Shanghai Ltd., Shanghai, China) and complete Freund’s adjuvant (CFA) containing 500 μg of Mycobacterium tuberculosis (BD), was prepared using a homogenizer (Thermofisher Scientific, Waltham, MA, USA) operating at a maximum speed of 30,000 RPM for 1 min and 30 s.
Briefly, mice were immunized with 150 μg of MOG35–55 peptide (MOGp) (GL Biochem, Shanghai Ltd.) emulsified in complete Freund’s adjuvant (CFA), followed by an intraperitoneal injection of 200 ng of Bordetella pertussis toxin (Difco Laboratories, Franklin Lakes, NJ, USA) in phosphate-buffered saline (PBS) on the day of immunization and on day 2. Mice were scored daily to assess clinical symptoms of EAE for 30 days. The EAE scoring system ranges from a score of 0 to 6 as described previously [15,16].

2.1. Passive Transfer of Encephalitogenic Th1 and Th17 Cells

For passive induction of EAE, MOG35–55-reactive Th1 or Th17 cells were generated from C57Bl/6 mice as recently described [15]. Briefly, donor C57BL/6 WT mice were immunized with 150 µg MOG35–55 in CFA containing 500 μg of Mycobacterium tuberculosis (BD). No pertussis toxin was administered. Ten days following immunization, cells from peripheral lymph nodes and spleen were restimulated with 10 μg/mL MOG35–55 under either Th1 or Th17 polarizing conditions for 3 days. Th1 polarizing conditions consisted of 10 ng/mL IL-12 (BioLegend, San Diego, CA, USA) and 0.5 μg/mL anti-IL-4 (BioLegend) in complete Iscove’s modified Dulbecco’s medium (IMDM). Th17 polarizing conditions consisted of 20 ng/mL IL-6 (Tonbo Biosciences, San Diego, CA, USA), 20 ng/mL IL-23 (Bio-Legend), 1 ng/mL TGF-β1 (Tonbo Biosciences), 10 μg/mL anti-IFN-γ (BioLegend), and 0.5 μg/mL anti-IL-4 (BioLegend) in IMDM complete culture media. The differentiated cells were harvested and pooled and dead cells were removed using a Ficoll gradient. CD4 T cells were enriched using magnetic dynabeads (Thermo Fisher, Waltham, MA, USA). These cells were used as “donor cells”. WT and Ifnlr1−/− mice were injected with 2.5 × 106 cells (Th1 or Th17) resuspended in 500 μL of PBS via intravenous tail injections. Recipient mice were also intraperitoneally administered 200 ng of the pertussis toxin, the same as for active EAE, and they were scored daily to assess clinical symptoms of EAE for 30 days as described above [15].

2.2. Isolation of Peritoneal Macrophages and Co-Culture with CD4+T Cells

Thioglycolate (3%) injection (1.5 mL) was given intraperitoneally and, after 72 h, the mice were sacrificed and injected with chilled PBS in the peritoneum. The peritoneal exudate was collected and seeded onto non-tissue culture 100mm dishes in RPMI media with 10% fetal bovine serum (FBS) for 30–45 min to allow macrophages to adhere to the dishes. Subsequently, non-adherent cells were removed by washing twice with Hank’s balanced salt solution (HBSS). Adherent cells were lifted from culture dishes using cold (4 °C) HBSS with 10mM EDTA for 3–4 min. Purified CD4+ T cells from WT and Ifnlr1−/− MOGp-immunized mice were cultured at 1:3 ratio (macrophage/CD4 T cells ratio) with 20 ng/mL MOGp under Th1 or Th17 polarizing conditions for 72 h and analyzed for IL-17 and IFN-γ expressing cells by flow cytometry [15].

2.3. Flow Cytometric Analysis of Th1- and Th17-Polarized MOG-Specific T Cells

IFN-γ and IL-17-expressing cells were enumerated by flow cytometry as described previously [15,16]. Briefly, the cells were stimulated with PMA (50 ng/mL, Sigma Aldrich, St. Louis, MO, USA) and ionomycin (500 ng/mL, Sigma Aldrich) in the presence of brefeldin A (BioLegend) for 4 h at 37 °C. The cells were stained with Zombie NIR live/dead stain, followed by anti-CD4, anti-IFN-γ APC BV650, and anti-IL-17A AF488 (BioLegend, San Diego, CA, USA) antibodies. Flow cytometry data were acquired using an Attune NxT flow cytometer (Thermo Fisher) and analyzed using FlowJo-v10.9.0 software (BD).

2.4. Flow Cytometric Analysis of Neutrophil Populations

Mice immunized for active EAE, as described above, were deeply anesthetized on day 10 [15,17]. The mice were perfused and the spines were removed. The spinal cords were dissected and the cells were dispersed without enzymatic digestion and stained with antibodies to Ly6C, Ly6G, CXCR2, CD11b, and CD62L (BioLegend).

2.5. Statistical Analysis

The data are presented as means ± SEM. Statistical significance was determined using non-parametric (EAE) or parametric (in vitro experiments and cell populations) two-tailed Mann–Whitney t-test.

3. Results

3.1. IFN-λ Signaling Is Necessary to Restrain the Development of Severe EAE

As a first step to interrogate the role of IFN-λ in autoimmune neuroinflammation, we assessed the development and progression of EAE in WT and Ifnlr1−/− mice following immunization with MOG35–55 peptide (MOGp). EAE symptoms were monitored daily using a standard clinical score as described in the Methods section. The Ifnlr1−/− mice developed significantly more severe EAE than WT littermates (Figure 1).
Previously, we demonstrated that type 1 IFN receptor (IFNAR) signaling was necessary to restrain EAE induced by encephalitogenic Th1 cells but not Th17 cells [16]. We, therefore, interrogated whether IFN-λ signaling differentially modulates disease induced by encephalitogenic Th1 and Th17 cells. Ifnlr1−/− recipient mice of encephalitogenic Th1 cells developed significantly more severe EAE compared to WT recipient mice (p < 0.001) (Figure 2A). The day of onset of EAE as well as peak of disease was similar for WT and Ifnlr1−/− recipients; Ifnlr1−/− recipients reached peak of disease by day 12, about three days earlier than WT recipients. In contrast to Th1 cells, encephalitogenic Th17 cells induced EAE of equal severity in WT and Ifnlr1−/− mice (Figure 2B).

3.2. IFN-λ Signaling in Macrophages Restrains the Expansion of Encephalitogenic Th17 Cells but Not Th1 Cells under Restimulation Conditions In Vitro

Considering that Th1-driven EAE but not Th17-driven EAE was aggravated in the absence of IFN-λ signaling, we tested the possibility that this represented differences in the function of IFN-λ signaling in antigen-presenting cells (APC) to regulate encephalitogenic Th1 vs. Th17 expansion. To address this possibility, we isolated CD4 T cells from spleens and lymph nodes of MOGp-immunized WT mice and restimulated them with MOGp using peritoneal macrophages from WT or Ifnlr1−/− mice as APC under Th17 or Th1 polarizing conditions. Under Th17 polarizing conditions, cultures containing Ifnlr−/− macrophages displayed a greater proportion of Th17 cells compared to those with WT macrophages (Figure 3A). In contrast, under Th1 polarizing conditions, WT and Ifnlr1−/− macrophages cultures contained similar proportion of Th1 cells (Figure 3B). From this data, it can be inferred that, in vitro, IFN-λ signaling in antigen presenting cells restrains the expansion of Th17 cells but not Th1 cells.

3.3. Neutrophil Populations (CD62Llo) in Spinal Cords Represent the Effector Population in EAE

Neutrophils express IFNLR and it is known that its engagement by IFN-λ inhibits their migration and production of ROS [7,10]. Therefore, we postulated that the less severe EAE observed in Ifnlr1 KO mice reflects the effect of IFN-λ on neutrophils. Consistent with this possibility, early in EAE disease (day 10 after immunization), the proportion of Ly6CLy6G+ cells representing neutrophils was significantly elevated in Ifnlr1−/− mice compared to WT mice (Figure 4A). The majority of neutrophils in Ifnlr1−/− mice displayed CXCR2CD62Llo phenotype indicating effector neutrophil population (Figure 4B) [18]. The frequency of CXCR2+CD62Lhi was similar in WT and Ifnlr1−/− mice. Though present in smaller numbers, the frequency of CXCR2 + CD62L- neutrophils was significantly lower in Ifnlr1−/− mice with EAE (Figure 4B).
A recent study showed that the loss of IFN-λ signaling increased the frequency of inflammatory macrophages in CNS of EAE mice [14]. As depicted in Figure 4C, no difference in the frequency of macrophages was observed in early EAE in the presented disease model (CD11b+LyC+LyG) (Figure 4C).

4. Discussion

In this report, we demonstrate that IFN-λ signaling is necessary to restrain autoimmune neuroinflammation severity in the EAE model of MS. We further showed that a transfer of encephalitogenic Th1 cells, but not Th17 cells, induced EAE with higher severity in naïve Ifnlr1−/− compared to WT mice.
In contrast to our findings, a recent study demonstrated that the Ifnlr1−/− mice developed less severe EAE than the WT mice [14]. The investigators conclude that IFN-λ signaling in myeloid cells, specifically in CD11c+ cells, was important in restraining EAE severity. The Ifnlr1−/− mice in this study are the same as those used by Manivasagam et al. [14]. Although we are unable to provide a definitive explanation for the underlying reasons for the disparate results between our two studies, a plausible explanation includes the differences in immunization approaches. Our immunization approach in C57Bl/6 leads to the development of chronic EAE with no recovery in disease severity (Figure 1). In the study by Manivasagam et al. [14], the Ifnlr1−/− mice developed chronic EAE; however, there was some degree of recovery. Observed differences in outcomes may reflect differences in experimental approaches including sites of immunization (upper back used here vs. flanks), immunization doses, and others, as previously reported for EAE outcome in Ifnlr1−/− mice [19,20,21,22].
Both adaptive and innate cells of the immune system contribute to the pathology of MS and EAE. Most studies in this field focused on the role of CD4+ T lymphocytes, which form part of the adaptive immune system as both mediators and regulators in disease pathogenesis [23]. Recent studies have suggested that the innate immune system also plays a vital role in the initiation and progression of MS by influencing the effector function of T and B cells [24]. The cells of the innate immune system can prevent autoimmunity by differentiation of regulatory T cells and by secretion of neurotrophic growth factors. The innate immune system can also play a pathogenic role by promoting the differentiation of Th1 and Th17 cells that drive acute inflammatory events associated with relapses in MS [24]. The effector cells express cytokines and activation markers that further activate innate immune cells [25]. Furthermore, innate immune cells have been implicated in the progressive phase of MS, as reflected by their activated phenotype in the periphery, which might be responsible for neurodegenerative changes in secondary progressive MS.
Several studies in EAE models indicate that neutrophils play an essential role in inflammation by producing cytokines and damaging the blood–brain barrier (BBB). Neutrophils can influence the manifestation of EAE by facilitating parenchymal brain inflammation [26]. Neutrophils are necessary and sufficient to induce demyelination in mouse models of MS [27,28]. Neutrophil expansion in the periphery and CNS is an early event in EAE and their depletion leads to delayed onset and attenuated disease [29,30,31,32]. Demyelination and axonal damage are directly orchestrated by autoreactive T cells, B cells, neutrophils, macrophages, and microglia, and indirectly through neutrophil-mediated release of pro-inflammatory factors including ROS, nitric oxide, matrix metalloproteinases (MMPs), and IL-1β [33,34,35]. While expansion of neutrophils early in EAE is linked to the expression of granulocyte-colony stimulating factor (G-CSF) [36], their recruitment into the CNS (brain and spinal cord) is dependent on Th1 and Th17 chemokines, CXCL1, CXCL2, and CXCL6, that activate the CXCR2 receptor [32,36,37]. Neutrophils are localized in active MS lesions at the sites of BBB leakage and actively contribute to BBB permeability [30,32,38,39]. Neutrophils mediate BBB breakdown via contact-dependent mechanisms and through the secretion of enzymes, myeloperoxidase (MPO), matrix metalloproteinases (MMPs), and ROS [40]. Mmp2−/− and Mmp9−/− double knockout mice are resistant to EAE development [41]. In MS patients, an increase in MMP-9 levels in CSF and blood correlates with disease activity [42,43]. MPO increases BBB permeability and neutrophil migration into the CNS [44] and induces the production of highly reactive ROS resulting in neuronal damage [45]. A neutrophil subset, polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs), plays a critical role in immune regulation [46,47] and was shown to ameliorate EAE through suppression of B cells, Th1, or Th17 cells via programmed death ligand 1 (PD-L1)/PD-1 interaction [48,49,50,51,52,53].
We found that Ifnlr1−/− mice exhibited an increased proportion of early-infiltrating neutrophils but not macrophages compared to WT mice. These neutrophils were predominantly CXCR2CD62lo, a population of activated neutrophils [18]. In EAE, neutrophils comprise a significant percentage of CNS-infiltrating cells prior to disease onset and relapse [26]. Disease was ameliorated when neutrophils were depleted prior to, but not after, the onset of disease or relapse, suggesting their essential function during the early steps of autoimmune neuroinflammation [26,36]. IFN-λ signaling exerts a regulatory function on neutrophils in MS, SLE, and RA [3,7,11,12,13]. The signal from IFN-λ regulates neutrophil activity via the inhibition of migration, ROS, NETosis, and expression of IL-1β and transcription factor IRF1 [3,4,5,6,7,8,9,10]. We also noted that IFNLR1-deficient macrophages are more efficient in promoting expansion of Th17 effector cells. Il-17A functions as a chemokine to promote recruitment of neutrophils and this could represent a mechanism for the greater numbers of neutrophils early during EAE in Ifnr1−/− mice [54]. Currently, the precise mechanism underlying the increased EAE severity in Ifnlr1−/− mice is unclear. Based on the findings presented here, we predict that a key property of IFN-λ is its ability to inhibit neutrophil migration into the CNS. This possibility is supported by a preliminary experiment using positron emission tomography (PET) and translocator protein (TSPO) as a tracer, which revealed an increase in early neutrophil migration into the CNS (spinal cord and brain).
In our previous study, we demonstrated that type 1 IFN (IFNAR) signaling was crucial in restraining EAE induced by encephalitogenic Th1 cells but not Th17 cells [16]. This current study further supports these findings, as we observed that IFNLR1 signaling is also necessary to restrain EAE induced by encephalitogenic Th1 cells but not Th17 cells (Figure 2). This was somewhat unexpected since, in vitro, Ifnlr1−/− macrophages promoted the expansion of in vivo-primed MOGp-reactive Th17 cells but not Th1 cells. The difference between in vivo and in vitro data may reflect the combined effect of other factors that are active in vivo but are not fully replicated in vitro. It is feasible that IFNLR signaling restrains Th17 plasticity to Th1 cells or other aspects of Th17 pathogenicity [17,55,56].
IFN-β is an important therapeutic option for MS and remains an important comparator for newer treatments for this disease. IFN-λ has a distinct advantage over type I IFNs (IFNα/β) as its receptor has restricted expression on cells and tissues. Additional research is required to fully understand the context-dependent effects of IFN-λs for optimization of treatment in MS and other autoimmune diseases.
Overall, our studies highlight the crucial role of type III interferon, IFN-λ signaling in restraining autoimmune neuroinflammation, potentially through the inhibition of neutrophil migration and/or activation. Importantly, our findings also suggest that IFN-λ exerts pleiotropic effects in EAE and MS which are context-dependent and may be anti-inflammatory or inflammatory, as previously demonstrated for IFN-γ [19].

Author Contributions

M.A.S.: methodology, software, validation, and writing—original draft preparation; S.J.D.: methodology, software, and data curation; Z.H.: writing—reviewing and editing; C.R.: conceptualization, methodology, software, validation, investigation, writing—reviewing and editing, and supervision; N.Y.: conceptualization, investigation, writing—original draft preparation, writing—reviewing and editing, supervision, and funding. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported in part by 1R01AR071157-01A1 (NY), R21 AI149267 (CR), and R21 MD015319 (CR) from the National Institute of Health.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available on request from the corresponding author.

Conflicts of Interest

None of the authors has a potential conflict of interest with this submission.

References

  1. Lazear, H.M.; Schoggins, J.W.; Diamond, M.S. Shared and Distinct Functions of Type I and Type III Interferons. Immunity 2019, 50, 907–923. [Google Scholar] [CrossRef] [PubMed]
  2. Lazear, H.M.; Nice, T.J.; Diamond, M.S. Interferon-λ: Immune Functions at Barrier Surfaces and Beyond. Immunity 2015, 43, 15–28. [Google Scholar] [CrossRef]
  3. Manivasagam, S.; Klein, R.S. Type III Interferons: Emerging Roles in Autoimmunity. Front. Immunol. 2021, 12, 764062. [Google Scholar] [CrossRef] [PubMed]
  4. Kotenko, S.V.; Rivera, A.; Parker, D.; Durbin, J.E. Type III IFNs: Beyond Antiviral Protection. Semin. Immunol. 2019, 43, 101303. [Google Scholar] [CrossRef] [PubMed]
  5. Forero, A.; Ozarkar, S.; Li, H.; Lee, C.H.; Hemann, E.A.; Nadjsombati, M.S.; Hendricks, M.R.; So, L.; Green, R.; Roy, C.N.; et al. Differential Activation of the Transcription Factor IRF1 Underlies the Distinct Immune Responses Elicited by Type I and Type III Interferons. Immunity 2019, 51, 451–464.e6. [Google Scholar] [CrossRef]
  6. Casazza, R.L.; Lazear, H.M. Why Is IFN-λ Less Inflammatory? One IRF Decides. Immunity 2019, 51, 415–417. [Google Scholar] [CrossRef] [PubMed]
  7. Blazek, K.; Eames, H.L.; Weiss, M.; Byrne, A.J.; Perocheau, D.; Pease, J.E.; Doyle, S.; McCann, F.; Williams, R.O.; Udalova, I.A. IFN-λ Resolves Inflammation via Suppression of Neutrophil Infiltration and IL-1β Production. J. Exp. Med. 2015, 212, 845–853. [Google Scholar] [CrossRef]
  8. Galani, I.E.; Triantafyllia, V.; Eleminiadou, E.-E.; Koltsida, O.; Stavropoulos, A.; Manioudaki, M.; Thanos, D.; Doyle, S.E.; Kotenko, S.V.; Thanopoulou, K.; et al. Interferon-λ Mediates Non-Redundant Front-Line Antiviral Protection against Influenza Virus Infection without Compromising Host Fitness. Immunity 2017, 46, 875–890.e6. [Google Scholar] [CrossRef]
  9. Rivera, A. Interferon Lambda’s New Role as Regulator of Neutrophil Function. J. Interf. Cytokine Res. 2019, 39, 609–617. [Google Scholar] [CrossRef]
  10. Broggi, A.; Tan, Y.; Granucci, F.; Zanoni, I. IFN-λ Suppresses Intestinal Inflammation by Non-Translational Regulation of Neutrophil Function. Nat. Immunol. 2017, 18, 1084–1093. [Google Scholar] [CrossRef]
  11. Goel, R.R.; Wang, X.; O’neil, L.J.; Nakabo, S.; Hasneen, K.; Gupta, S.; Wigerblad, G.; Blanco, L.P.; Kopp, J.B.; Morasso, M.I.; et al. Interferon Lambda Promotes Immune Dysregulation and Tissue Inflammation in TLR7-Induced Lupus. Proc. Natl. Acad. Sci. USA 2020, 117, 5409–5419. [Google Scholar] [CrossRef] [PubMed]
  12. Goel, R.R.; Kotenko, S.V.; Kaplan, M.J. Interferon Lambda in Inflammation and Autoimmune Rheumatic Diseases. Nat. Rev. Rheumatol. 2021, 17, 349–362. [Google Scholar] [CrossRef] [PubMed]
  13. Goel, R.R.; Nakabo, S.; Dizon, B.L.; Urban, A.; Waldman, M.; Howard, L.; Darnell, D.; Buhaya, M.; Carmona-Rivera, C.; Hasni, S.; et al. Lupus-like Autoimmunity and Increased Interferon Response in Patients with STAT3-Deficient Hyper-IgE Syndrome. J. Allergy Clin. Immunol. 2021, 147, 746–749.e9. [Google Scholar] [CrossRef] [PubMed]
  14. Manivasagam, S.; Williams, J.L.; Vollmer, L.L.; Bollman, B.; Bartleson, J.M.; Ai, S.; Wu, G.F.; Klein, R.S. Targeting IFN-λ Signaling Promotes Recovery from Central Nervous System Autoimmunity. J. Immunol. 2022, 208, 1341–1351. [Google Scholar] [CrossRef] [PubMed]
  15. Duesman, S.J.; Ortega-Francisco, S.; Olguin-Alor, R.; Acevedo-Dominguez, N.A.; Sestero, C.M.; Chellappan, R.; De Sarno, P.; Yusuf, N.; Salgado-Lopez, A.; Segundo-Liberato, M.; et al. Transforming Growth Factor Receptor III (Betaglycan) Regulates the Generation of Pathogenic Th17 Cells in EAE. Front. Immunol. 2023, 14, 1088039. [Google Scholar] [CrossRef] [PubMed]
  16. Axtell, R.C.; De Jong, B.A.; Boniface, K.; Van Der Voort, L.F.; Bhat, R.; De Sarno, P.; Naves, R.; Han, M.; Zhong, F.; Castellanos, J.G.; et al. T Helper Type 1 and 17 Cells Determine Efficacy of Interferon-β in Multiple Sclerosis and Experimental Encephalomyelitis. Nat. Med. 2010, 16, 406–412. [Google Scholar] [CrossRef]
  17. Axtell, R.C.; Xu, L.; Barnum, S.R.; Raman, C. CD5-CK2 Binding/Activation-Deficient Mice Are Resistant to Experimental Autoimmune Encephalomyelitis: Protection Is Associated with Diminished Populations of IL-17-Expressing T Cells in the Central Nervous System. J. Immunol. 2006, 177, 8542–8549. [Google Scholar] [CrossRef]
  18. Capucetti, A.; Albano, F.; Bonecchi, R. Multiple Roles for Chemokines in Neutrophil Biology. Front. Immunol. 2020, 11, 1259. [Google Scholar] [CrossRef]
  19. Naves, R.; Singh, S.P.; Cashman, K.S.; Rowse, A.L.; Axtell, R.C.; Steinman, L.; Mountz, J.D.; Steele, C.; De Sarno, P.; Raman, C. The Interdependent, Overlapping, and Differential Roles of Type I and II IFNs in the Pathogenesis of Experimental Autoimmune Encephalomyelitis. J. Immunol. 2013, 191, 2967–2977. [Google Scholar] [CrossRef]
  20. Kavrochorianou, N.; Evangelidou, M.; Markogiannaki, M.; Tovey, M.; Thyphronitis, G.; Haralambous, S. IFNAR Signaling Directly Modulates T Lymphocyte Activity, Resulting in Milder Experimental Autoimmune Encephalomyelitis Development. J. Leukoc. Biol. 2016, 99, 175–188. [Google Scholar] [CrossRef]
  21. Guo, B.; Chang, E.Y.; Cheng, G. The Type I IFN Induction Pathway Constrains Th17-Mediated Autoimmune Inflammation in Mice. J. Clin. Investig. 2008, 118, 1680–1690. [Google Scholar] [CrossRef] [PubMed]
  22. Prinz, M.; Schmidt, H.; Mildner, A.; Knobeloch, K.-P.; Hanisch, U.-K.; Raasch, J.; Merkler, D.; Detje, C.; Gutcher, I.; Mages, J.; et al. Distinct and Nonredundant In Vivo Functions of IFNAR on Myeloid Cells Limit Autoimmunity in the Central Nervous System. Immunity 2008, 28, 675–686. [Google Scholar] [CrossRef] [PubMed]
  23. Van Kaer, L.; Postoak, J.L.; Wang, C.; Yang, G.; Wu, L. Innate, Innate-like and Adaptive Lymphocytes in the Pathogenesis of MS and EAE. Cell. Mol. Immunol. 2019, 16, 531–539. [Google Scholar] [CrossRef] [PubMed]
  24. Gandhi, R.; Laroni, A.; Weiner, H.L. Role of the Innate Immune System in the Pathogenesis of Multiple Sclerosis. J. Neuroimmunol. 2010, 221, 7–14. [Google Scholar] [CrossRef] [PubMed]
  25. Monney, L.; Sabatos, C.A.; Gaglia, J.L.; Ryu, A.; Waldner, H.; Chernova, T.; Manning, S.; Greenfield, E.A.; Coyle, A.J.; Sobel, R.A.; et al. Th1-Specific Cell Surface Protein Tim-3 Regulates Macrophage Activation and Severity of an Autoimmune Disease. Nature 2002, 415, 536–541. [Google Scholar] [CrossRef]
  26. Pierson, E.R.; Wagner, C.A.; Goverman, J.M. The Contribution of Neutrophils to CNS Autoimmunity. Clin. Immunol. 2018, 189, 23–28. [Google Scholar] [CrossRef]
  27. Liu, L.; Belkadi, A.; Darnall, L.; Hu, T.; Drescher, C.; Cotleur, A.C.; Padovani-Claudio, D.; He, T.; Choi, K.; E Lane, T.; et al. CXCR2-Positive Neutrophils Are Essential for Cuprizone-Induced Demyelination: Relevance to Multiple Sclerosis. Nat. Neurosci. 2010, 13, 319–326. [Google Scholar] [CrossRef]
  28. Grist, J.J.; Marro, B.S.; Skinner, D.D.; Syage, A.R.; Worne, C.; Doty, D.J.; Fujinami, R.S.; Lane, T.E. Induced CNS Expression of CXCL1 Augments Neurologic Disease in a Murine Model of Multiple Sclerosis via Enhanced Neutrophil Recruitment. Eur. J. Immunol. 2018, 48, 1199–1210. [Google Scholar] [CrossRef]
  29. Nygårdas, P.T.; Määttä, J.A.; Hinkkanen, A.E. Chemokine Expression by Central Nervous System Resident Cells and Infiltrating Neutrophils during Experimental Autoimmune Encephalomyelitis in the BALB/c Mouse. Eur. J. Immunol. 2000, 30, 1911–1918. [Google Scholar] [CrossRef]
  30. Christy, A.L.; Walker, M.E.; Hessner, M.J.; Brown, M.A. Mast Cell Activation and Neutrophil Recruitment Promotes Early and Robust Inflammation in the Meninges in EAE. J. Autoimmun. 2013, 42, 50–61. [Google Scholar] [CrossRef]
  31. McColl, S.R.; Staykova, M.A.; Wozniak, A.; Fordham, S.; Bruce, J.; Willenborg, D.O. Treatment with Anti-Granulocyte Antibodies Inhibits the Effector Phase of Experimental Autoimmune Encephalomyelitis. J. Immunol. 1998, 161, 6421–6426. [Google Scholar] [CrossRef]
  32. Carlson, T.; Kroenke, M.; Rao, P.; Lane, T.E.; Segal, B. The Th17–ELR+ CXC Chemokine Pathway Is Essential for the Development of Central Nervous System Autoimmune Disease. J. Exp. Med. 2008, 205, 811–823. [Google Scholar] [CrossRef]
  33. Opdenakker, G.; Proost, P.; Van Damme, J. Microbiomic and Posttranslational Modifications as Preludes to Autoimmune Diseases. Trends Mol. Med. 2016, 22, 746–757. [Google Scholar] [CrossRef]
  34. Lan, M.; Tang, X.; Zhang, J.; Yao, Z. Insights in Pathogenesis of Multiple Sclerosis: Nitric Oxide May Induce Mitochondrial Dysfunction of Oligodendrocytes. Prog. Neurobiol. 2017, 29, 39–53. [Google Scholar] [CrossRef] [PubMed]
  35. Muri, L.; Leppert, D.; Grandgirard, D.; Leib, S.L. MMPs and ADAMs in Neurological Infectious Diseases and Multiple Sclerosis. Cell. Mol. Life Sci. 2019, 76, 3097–3116. [Google Scholar] [CrossRef] [PubMed]
  36. Rumble, J.M.; Huber, A.K.; Krishnamoorthy, G.; Srinivasan, A.; Giles, D.A.; Zhang, X.; Wang, L.; Segal, B.M. Neutrophil-Related Factors as Biomarkers in EAE and MS. J. Exp. Med. 2015, 212, 23–35. [Google Scholar] [CrossRef] [PubMed]
  37. Stoolman, J.S.; Duncker, P.C.; Huber, A.K.; Giles, D.A.; Washnock-Schmid, J.M.; Soulika, A.M.; Segal, B.M. An IFNγ/CXCL2 Regulatory Pathway Determines Lesion Localization during EAE. J. Neuroinflammation 2018, 15, 208. [Google Scholar] [CrossRef] [PubMed]
  38. Aubé, B.; Lévesque, S.A.; Paré, A.; Chamma, E.; Kébir, H.; Gorina, R.; Lécuyer, M.-A.; Alvarez, J.I.; De Koninck, Y.; Engelhardt, B.; et al. Neutrophils Mediate Blood–Spinal Cord Barrier Disruption in Demyelinating Neuroinflammatory Diseases. J. Immunol. 2014, 193, 2438–2454. [Google Scholar] [CrossRef] [PubMed]
  39. Steinbach, K.; Piedavent, M.; Bauer, S.; Neumann, J.T.; Friese, M.A. Neutrophils Amplify Autoimmune Central Nervous System Infiltrates by Maturing Local APCs. J. Immunol. 2013, 191, 4531–4539. [Google Scholar] [CrossRef]
  40. Garg, N.; Smith, T.W. An Update on Immunopathogenesis, Diagnosis, and Treatment of Multiple Sclerosis. Brain Behav. 2015, 5, e00362. [Google Scholar] [CrossRef]
  41. Song, J.; Wu, C.; Korpos, E.; Zhang, X.; Agrawal, S.M.; Wang, Y.; Faber, C.; Schäfers, M.; Körner, H.; Opdenakker, G.; et al. Focal MMP-2 and MMP-9 Activity at the Blood-Brain Barrier Promotes Chemokine-Induced Leukocyte Migration. Cell Rep. 2015, 10, 1040–1054. [Google Scholar] [CrossRef]
  42. Leppert, D.; Ford, J.; Stabler, G.; Grygar, C.; Lienert, C.; Huber, S.; Miller, K.M.; Hauser, S.L.; Kappos, L. Matrix Metalloproteinase-9 (Gelatinase B) Is Selectively Elevated in CSF during Relapses and Stable Phases of Multiple Sclerosis. Brain 1998, 121, 2327–2334. [Google Scholar] [CrossRef]
  43. Waubant, E.; Goodkin, D.E.; Gee, L.; Bacchetti, P.; Sloan, R.; Stewart, T.; Andersson, P.-B.; Stabler, G.; Miller, K. Serum MMP-9 and TIMP-1 Levels Are Related to MRI Activity in Relapsing Multiple Sclerosis. Neurology 1999, 53, 1397. [Google Scholar] [CrossRef]
  44. Lau, D.; Mollnau, H.; Eiserich, J.P.; Freeman, B.A.; Daiber, A.; Gehling, U.M.; Brümmer, J.; Rudolph, V.; Münzel, T.; Heitzer, T.; et al. Myeloperoxidase Mediates Neutrophil Activation by Association with CD11b/CD18 Integrins. Proc. Natl. Acad. Sci. USA 2005, 102, 431–436. [Google Scholar] [CrossRef]
  45. Kubota, K.; Saiwai, H.; Kumamaru, H.; Maeda, T.; Ohkawa, Y.; Aratani, Y.; Nagano, T.; Iwamoto, Y.; Okada, S. Myeloperoxidase Exacerbates Secondary Injury by Generating Highly Reactive Oxygen Species and Mediating Neutrophil Recruitment in Experimental Spinal Cord Injury. Spine 2012, 37, 1363–1369. [Google Scholar] [CrossRef]
  46. Veglia, F.; Perego, M.; Gabrilovich, D. Myeloid-Derived Suppressor Cells Coming of Age. Nat. Immunol. 2018, 19, 108–119. [Google Scholar] [CrossRef] [PubMed]
  47. Gabrilovich, D.I.; Nagaraj, S. Myeloid-Derived Suppressor Cells as Regulators of the Immune System. Nat. Rev. Immunol. 2009, 9, 162–174. [Google Scholar] [CrossRef] [PubMed]
  48. Knier, B.; Hiltensperger, M.; Sie, C.; Aly, L.; Lepennetier, G.; Engleitner, T.; Garg, G.; Muschaweckh, A.; Mitsdörffer, M.; Koedel, U.; et al. Myeloid-Derived Suppressor Cells Control B Cell Accumulation in the Central Nervous System during Autoimmunity. Nat. Immunol. 2018, 19, 1341–1351. [Google Scholar] [CrossRef] [PubMed]
  49. White, M.P.J.; Webster, G.; Leonard, F.; La Flamme, A.C. Innate IFN-γ Ameliorates Experimental Autoimmune Encephalomyelitis and Promotes Myeloid Expansion and PDL-1 Expression. Sci. Rep. 2018, 8, 259. [Google Scholar] [CrossRef] [PubMed]
  50. Khorooshi, R.; Marczynska, J.; Dieu, R.S.; Wais, V.; Hansen, C.R.; Kavan, S.; Thomassen, M.; Burton, M.; Kruse, T.; Webster, G.A.; et al. Innate Signaling within the Central Nervous System Recruits Protective Neutrophils. Acta Neuropathol. Commun. 2020, 8, 2. [Google Scholar] [CrossRef] [PubMed]
  51. Ioannou, M.; Alissafi, T.; Lazaridis, I.; Deraos, G.; Matsoukas, J.; Gravanis, A.; Mastorodemos, V.; Plaitakis, A.; Sharpe, A.; Boumpas, D.; et al. Crucial Role of Granulocytic Myeloid-Derived Suppressor Cells in the Regulation of Central Nervous System Autoimmune Disease. J. Immunol. 2012, 188, 1136–1146. [Google Scholar] [CrossRef] [PubMed]
  52. Zehntner, S.P.; Brickman, C.; Bourbonnière, L.; Remington, L.; Caruso, M.; Owens, T. Neutrophils That Infiltrate the Central Nervous System Regulate T Cell Responses. J. Immunol. 2005, 174, 5124–5131. [Google Scholar] [CrossRef] [PubMed]
  53. Melero-Jerez, C.; Suardíaz, M.; Lebrón-Galán, R.; Marín-Bañasco, C.; Oliver-Martos, B.; Machín-Díaz, I.; Fernández, Ó.; de Castro, F.; Clemente, D. The Presence and Suppressive Activity of Myeloid-Derived Suppressor Cells Are Potentiated after Interferon-β Treatment in a Murine Model of Multiple Sclerosis. Neurobiol. Dis. 2019, 127, 13–31. [Google Scholar] [CrossRef] [PubMed]
  54. McGinley, A.M.; Sutton, C.E.; Edwards, S.C.; Leane, C.M.; Decourcey, J.; Teijeiro, A.; Hamilton, J.A.; Boon, L.; Djouder, N.; Mills, K.H. Interleukin-17A Serves a Priming Role in Autoimmunity by Recruiting IL-1β-Producing Myeloid Cells that Promote Pathogenic T Cells. Immunity 2020, 52, 342–356.e6. [Google Scholar] [CrossRef]
  55. Weaver, C.T.; Harrington, L.E.; Mangan, P.R.; Gavrieli, M.; Murphy, K.M. Th17: An Effector CD4 T Cell Lineage with Regulatory T Cell Ties. Immunity 2006, 24, 677–688. [Google Scholar] [CrossRef]
  56. Lee, Y.; Kuchroo, V. Defining the Functional States of Th17 cells. F1000Research 2015, 4, 132. [Google Scholar] [CrossRef]
Figure 1. IFNLR1 knockout (Ifnlr1−/−) mice develop significantly more severe EAE compared to wild-type (WT) mice. EAE clinical score of Ifnlr1−/− (red, n = 10) and WT (green n = 10) mice immunized with MOG35–55 peptide in CFA. Mice were immunized with 150 µg MOG35–55 peptide in CFA and the clinical course of disease was determined. Mice with an EAE clinical score above 4 or a drop in weight of over 20% were humanely euthanized and removed from future analysis. Data represent mean ± SEM; ** p < 0.01.
Figure 1. IFNLR1 knockout (Ifnlr1−/−) mice develop significantly more severe EAE compared to wild-type (WT) mice. EAE clinical score of Ifnlr1−/− (red, n = 10) and WT (green n = 10) mice immunized with MOG35–55 peptide in CFA. Mice were immunized with 150 µg MOG35–55 peptide in CFA and the clinical course of disease was determined. Mice with an EAE clinical score above 4 or a drop in weight of over 20% were humanely euthanized and removed from future analysis. Data represent mean ± SEM; ** p < 0.01.
Biomedicines 12 00526 g001
Figure 2. IFN-λ signaling plays an important role in restraining EAE induced by encephalitogenic Th1 cells but not for Th17 cells. For passive induction of EAE, MOG35–55 reactive Th17 or Th1 cells were generated from immunized donor C57Bl/6 mice. Ten days following immunization, cells from peripheral lymph nodes and spleen were restimulated with 10 μg/mL MOG35–55 under either Th17 or Th1 polarizing conditions for 3 days. These cells were used as “donor cells”. WT (n = 10) and Ifnlr1−/− (n = 10) mice were injected with 2.5 × 106 cells (Th17 (A) or Th1 (B)) resuspended in 500 μL of PBS via intravenous tail injections. Recipient mice were also intraperitoneally administered 200 ng of the pertussis toxin, the same as for active EAE, and they were scored daily to assess clinical symptoms of EAE for 30 days. Data represent mean ± SEM; *** p < 0.0001.
Figure 2. IFN-λ signaling plays an important role in restraining EAE induced by encephalitogenic Th1 cells but not for Th17 cells. For passive induction of EAE, MOG35–55 reactive Th17 or Th1 cells were generated from immunized donor C57Bl/6 mice. Ten days following immunization, cells from peripheral lymph nodes and spleen were restimulated with 10 μg/mL MOG35–55 under either Th17 or Th1 polarizing conditions for 3 days. These cells were used as “donor cells”. WT (n = 10) and Ifnlr1−/− (n = 10) mice were injected with 2.5 × 106 cells (Th17 (A) or Th1 (B)) resuspended in 500 μL of PBS via intravenous tail injections. Recipient mice were also intraperitoneally administered 200 ng of the pertussis toxin, the same as for active EAE, and they were scored daily to assess clinical symptoms of EAE for 30 days. Data represent mean ± SEM; *** p < 0.0001.
Biomedicines 12 00526 g002
Figure 3. IFN-λ signaling in macrophages restrains the expansion of encephalitogenic Th17 cells but not Th1 cells. Encephalitogenic Th17 cell expansion by Ifnlr1−/− macrophages. The proportion of (A) IL-17+ or (B) IFN-γ+ CD4 T cells in cultures following 3-day restimulation with MOGp in the presence of peritoneal macrophages from WT or Ifnr1−/− mice. Each dot represents macrophages from an individual mouse. CD4 T cells were obtained from MOGp-immunized WT mice. Data represent mean ± SEM; ** p < 0.01; ns = not significant.
Figure 3. IFN-λ signaling in macrophages restrains the expansion of encephalitogenic Th17 cells but not Th1 cells. Encephalitogenic Th17 cell expansion by Ifnlr1−/− macrophages. The proportion of (A) IL-17+ or (B) IFN-γ+ CD4 T cells in cultures following 3-day restimulation with MOGp in the presence of peritoneal macrophages from WT or Ifnr1−/− mice. Each dot represents macrophages from an individual mouse. CD4 T cells were obtained from MOGp-immunized WT mice. Data represent mean ± SEM; ** p < 0.01; ns = not significant.
Biomedicines 12 00526 g003
Figure 4. Neutrophil subpopulations in spinal cords of WT and Ifnlr1 knockout mice with EAE. Single-cell suspension of perfused spinal cord from WT or Ifnlr1−/− mice with EAE were stained for expression of Ly6C, Ly6G, CD11b, CD62L, and CXCR2. Neutrophils were gated as Ly6G+Ly6Clo (A) and then examined for expression of CXCR2 and CD11b or CXCR2 and CD62L (B). The proportion of Ly6G + Ly6C-, CD11b + CXCR2-, CXCR2-CD62Llo, and CXCR2 + CD62lo in WT and Ifnlr1−/− mice (C). Macrophages were gated as CD11b+LyC+LyG+ cells (D). Total number of mice n = 3 of each genotype. * p < 0.05; ** p < 0.01; ns = not significant.
Figure 4. Neutrophil subpopulations in spinal cords of WT and Ifnlr1 knockout mice with EAE. Single-cell suspension of perfused spinal cord from WT or Ifnlr1−/− mice with EAE were stained for expression of Ly6C, Ly6G, CD11b, CD62L, and CXCR2. Neutrophils were gated as Ly6G+Ly6Clo (A) and then examined for expression of CXCR2 and CD11b or CXCR2 and CD62L (B). The proportion of Ly6G + Ly6C-, CD11b + CXCR2-, CXCR2-CD62Llo, and CXCR2 + CD62lo in WT and Ifnlr1−/− mice (C). Macrophages were gated as CD11b+LyC+LyG+ cells (D). Total number of mice n = 3 of each genotype. * p < 0.05; ** p < 0.01; ns = not significant.
Biomedicines 12 00526 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Sherwani, M.A.; Duesman, S.J.; Hel, Z.; Raman, C.; Yusuf, N. Interferon Lambda Signaling Restrains Experimental Autoimmune Encephalomyelitis. Biomedicines 2024, 12, 526. https://doi.org/10.3390/biomedicines12030526

AMA Style

Sherwani MA, Duesman SJ, Hel Z, Raman C, Yusuf N. Interferon Lambda Signaling Restrains Experimental Autoimmune Encephalomyelitis. Biomedicines. 2024; 12(3):526. https://doi.org/10.3390/biomedicines12030526

Chicago/Turabian Style

Sherwani, Mohammad Asif, Samuel J. Duesman, Zdenek Hel, Chander Raman, and Nabiha Yusuf. 2024. "Interferon Lambda Signaling Restrains Experimental Autoimmune Encephalomyelitis" Biomedicines 12, no. 3: 526. https://doi.org/10.3390/biomedicines12030526

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