Next Article in Journal
The Beneficial Effects of Ethanol Extract of the Microalga Phaeodactylum tricornutum in Alcoholic Liver Disease
Previous Article in Journal
Osmoregulation and Physiological Response of Largemouth Bass (Micropterus salmoides) Juvenile to Different Salinity Stresses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies

by
Costanza Stacchiotti
1,2,
Simona Mazzella di Regnella
1,
Miriam Cinotti
1,3,
Alida Spalloni
4,*,† and
Elisabetta Volpe
1,†
1
Molecular Neuroimmunology Unit, Santa Lucia Foundation, 00143 Rome, Italy
2
Department of Experimental Medicine, University of Rome Tor Vergata, 00133 Rome, Italy
3
Department of Biology and Biotechnology Charles Darwin, Sapienza University, 00185 Rome, Italy
4
Molecular Neurobiology Unit, Santa Lucia Foundation, 00143 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this study.
Int. J. Mol. Sci. 2025, 26(8), 3854; https://doi.org/10.3390/ijms26083854
Submission received: 18 March 2025 / Revised: 14 April 2025 / Accepted: 16 April 2025 / Published: 18 April 2025
(This article belongs to the Special Issue Amyotrophic Lateral Sclerosis Behind Motor Neurons)

Abstract

:
Neuroinflammation is an inflammatory response occurring within the central nervous system (CNS). The process is marked by the production of pro-inflammatory cytokines, chemokines, small-molecule messengers, and reactive oxygen species. Microglia and astrocytes are primarily involved in this process, while endothelial cells and infiltrating blood cells contribute to neuroinflammation when the blood–brain barrier (BBB) is damaged. Neuroinflammation is increasingly recognized as a pathological hallmark of several neurological diseases, including amyotrophic lateral sclerosis (ALS), and is closely linked to neurodegeneration, another key feature of ALS. In fact, neurodegeneration is a pathological trigger for inflammation, and neuroinflammation, in turn, contributes to motor neuron (MN) degeneration through the induction of synaptic dysfunction, neuronal death, and inhibition of neurogenesis. Importantly, resolution of acute inflammation is crucial for avoiding chronic inflammation and tissue destruction. Inflammatory processes are mediated by soluble factors known as cytokines, which are involved in both promoting and inhibiting inflammation. Cytokines with anti-inflammatory properties may exert protective roles in neuroinflammatory diseases, including ALS. In particular, interleukin (IL)-10, transforming growth factor (TGF)-β, IL-4, IL-13, and IL-9 have been shown to exert an anti-inflammatory role in the CNS. Other recently emerging immune regulatory cytokines in the CNS include IL-35, IL-25, IL-37, and IL-27. This review describes the current understanding of neuroinflammation in ALS and highlights recent advances in the role of anti-inflammatory cytokines within CNS with a particular focus on their potential therapeutic applications in ALS. Furthermore, we discuss current therapeutic strategies aimed at enhancing the anti-inflammatory response to modulate neuroinflammation in this disease.

1. Introduction

Neuroinflammation is a complex process that arises within the central nervous system (CNS) in response to injury, infections, autoimmunity or neurodegenerative diseases. The aim of this response is to protect the CNS by limiting the damage and facilitating the removal of pathogens, debris, and misfolded proteins. However, in some neurological diseases, the chronic inflammatory environment becomes detrimental and even contributes to neurodegeneration [1]. In this context, increasing evidence indicates that neuroinflammation contributes to motor neuron (MN) death, and influences the rate of disease progression in ALS [2,3]. The contributors to neuroinflammation can be broadly categorized into two main groups: resident CNS cells and infiltrating leukocytes, with their relative contributions varying among different kinds of CNS diseases [4]. In the CNS, astrocytes and microglia are the glial cells most responsive to changes in the microenvironment, capable of transitioning into a pro-inflammatory state. As the CNS damage progresses, glial cells release pro-inflammatory molecules to recruit peripheral blood cells, which can cross the damaged blood brain barrier (BBB), enter the CNS, and enhance neuroinflammation. During chronic neuroinflammation, glial cells may also produce neurotoxic factors which contribute to neurodegeneration. This mechanism has been largely investigated in multiple sclerosis (MS), characterized by a severely-compromised BBB, where infiltrating inflammatory monocytes, T lymphocytes, and B lymphocytes play a crucial role. In recent years, the view has emerged that brain barriers are not impermeable gates, but rather regulatory ones [5], and the disruption of this barrier control, which occurs in many neurological diseases [6], can lead to pathological leukocyte invasion from the periphery, thereby contributing to neuroinflammation in neurodegenerative diseases, including ALS.
However, a broad range of biological mechanisms are involved in the resolution of inflammation to restore tissue homeostasis and prevent the development of chronic inflammatory diseases.
In the CNS, glial cells and immune cells infiltrating the CNS possess the ability to release factors with anti-inflammatory properties, which can control the neuroinflammatory process by promoting tissue healing and homeostasis [7,8,9].
In this review, we discuss the contribution of anti-inflammatory cytokines in the resolution of neuroinflammation, highlighting the discoveries in preclinical and clinical studies and examining the potential therapeutic implications of engaging these molecules in ALS.

Neuroinflammation in ALS

ALS is a fatal CNS neurodegenerative disease, characterized by degeneration of MN. Several mechanisms contribute to MN degeneration in ALS, such as oxidative damage, formation of intracellular protein aggregates, damage to axonal transport systems, mitochondrial dysfunction, impairment of RNA metabolism and of DNA damage repair, and excessive activation of glutamate receptors leading to neurotoxicity [10]. In addition, increasing evidence indicates that also neuroinflammation contributes to MN injury and progression of the disease [2,3,11]. Inflammation in the CNS is mediated by microglia, astroglia, and infiltrating immune cells. The involvement of non-neuronal cell types in ALS pathology introduced the concept of non-cell autonomous components contributing to ALS disease [12] (Figure 1). In particular, most studies indicate that inflammatory cytokines released by astrocytes and microglia trigger an irreversible pathological process that leads to the non-cell autonomous death of MN in patients with ALS [13]. Moreover, a compromised BBB has been described in ALS mouse models and patients with ALS [14], suggesting that entry of activated peripheral immune cells into the CNS parenchyma of patients with ALS might contribute to the neuroinflammatory process (Figure 1).
Interestingly, immune dysregulation characterized by microglial cell activation in the spinal cord of ALS patients [15], elevated biomarkers of neuro-inflammation in the cerebrospinal fluid (CSF) of ALS patients correlating with disease severity [16], and pro-inflammatory gene profiles of blood monocytes from ALS patients [17] were associated with decreased blood levels of a subset of T lymphocytes [2,18], named T regulatory (Treg) cells, known to act as negative regulators of inflammation [19]. Importantly, reduced levels of Treg cells in the blood have been shown to correlate with disease progression at the time of collection and to serve as predictors of future rapid progression [20,21].
Central and peripheral inflammatory mechanisms are important contributors to ALS both in the context of familiar [22,23,24] and sporadic disease [25,26]. In the context of familiar ALS, it has been reported that mutations in genes such as superoxide dismutase 1 (SOD1) and TAR DNA-binding protein 43 (TARDBP) promote microglia activation [27] through nuclear factor-kappa B (NF-κB) and NLRP3 pathways [28], while mutations in chromosome 9 open reading frame 72 (C9orf72) [23], TANK binding kinase 1 (TBK1) [29], and optineurin (OPTN) [30] affect the inflammatory type I interferon response.
Overall, although it is still unclear what initiates the inflammatory processes, especially in patients with sporadic ALS, neuroinflammation has an important role in ALS pathogenesis, and its neutralization is crucial to curb excessive tissue damage and promote repair processes during neurodegeneration.

2. Anti-Inflammatory Cytokines in the CNS

Among the endogenous mechanisms for countering inflammation, immune molecules known as anti-inflammatory cytokines play a major role. Cytokines, including interferons, interleukins, and chemokines, are molecules canonically associated with the peripheral immune system and regulating the peripheral inflammatory response. However, numerous studies have now demonstrated that many of these cytokines and their receptors are expressed by resident CNS cells or by CNS-infiltrating immune cells. Increasing evidence indicates that some cytokines within the CNS may have an anti-inflammatory role by limiting the neuroinflammatory responses (Figure 2). These anti-inflammatory cytokines might effectively inhibit neuroinflammation, either by affecting CNS infiltrating immune cells (for example, by skewing macrophage polarization, by enhancing Treg cells, or by interfering with the activation of infiltrating B or T lymphocytes), and by reducing activation of CNS resident microglia and astrocytes.
Among anti-inflammatory cytokines, IL-10 and TGF-β were the first to be described. They are both produced by Treg cells, known to suppress pathological immune responses in the settings of transplantation, allergy and autoimmune diseases [31]. Other cytokines known to govern the regulation and resolution of inflammation are those produced by T helper 2 (Th2) cells, such as IL-4 and IL-13 [32,33]. Similarly, IL-9, originally associated with Th2 cells and then identified as a specific cytokine of a new Th subtype named Th9 [34], is emerging as potential regulator of inflammatory processes. Finally, other cytokines are emerging as potentially relevant in inhibiting neuroinflammation, such as IL-35, IL-25, IL-37, and IL-27 [35,36,37,38].

2.1. Interleukin-10 in the CNS and in ALS

IL-10 was first discovered for its ability to inhibit activation and effector functions of pro-inflammatory cells. IL-10 acts through a functional receptor complex composed of two subunits, IL-10R1 (IL-10Rα) and IL-10R2 (IL-10Rβ), and mainly activates Janus kinase (Jak) 1/Tyrosine (Tyk) kinase 2, and is the signal transducer and activator of the transcription 3 (STAT3) system [39]. IL-10 is expressed by many immune cell types and plays a critical role in balancing immune responses to limit chronic inflammatory diseases [40]. In particular, IL-10 inhibits the production of several inflammatory cytokines, such as tumor necrosis factor (TNF)-α, IL-1β, IL-6, and interferon (IFN)-γ secretion from monocytes/macrophages [41,42]. Moreover, IL-10 is a powerful inhibitor of the generation of reactive oxygen species (ROS), and increases the release of TNF receptors, which may antagonize the effects of TNF-α [40,42]. In the course of CNS pathology, IL-10 limits neuroinflammatory processes, similarly to what happens in peripheral sites, influencing resident macrophages in order to contain their inflammatory responses and promote the mechanisms that maintain tissue integrity.
The neuroprotective role of IL-10 was also described in MS, where the production of IL-10 by a rare subset of B lymphocytes called regulatory B (Breg) cells was associated with immunosuppressive functions [43,44]. In line with this, in experimental autoimmune encephalomyelitis (EAE), a murine demyelinating and paralyzing model of MS in IL-10-deficient animals develop a more severe disease, while IL-10-overexpressing mice are highly resistant to EAE [45]. Consistently, in a mouse model of Parkinson’s disease (PD), IL-10 gene therapy has been shown to elicit immunomodulation with enhanced suppression of neuroinflammation associated with dopaminergic neuron survival [46].
The role of IL-10 in ALS is emerging as protective. First, clinical studies report an increase of IL-10 levels in the blood of ALS patients compared to control subjects [47,48], and then higher levels of IL-10 in the blood predict longer disease duration in ALS patients [49]. Moreover, the beneficial role of IL-10 is confirmed by Furukawa et al., which showed a positive correlation between IL-10 levels in the CSF of ALS patients and Revised ALS Functional Rating Scale (ALSFRS-R) scores, indicating that high IL-10 is associated with low disease progression [50]. In transgenic mice mutants for SOD1G93A, which model the clinicopathology of ALS, overexpression of IL-10 in the spinal cord through recombinant adeno-associated virus (rAAV) vectors has been associated with extended survival of mice [51,52]. Similarly, gene-therapy-induced IL-10 overexpression in microglia of SOD1G93A mice significantly delays disease onset and increases survival [53]. In addition, a recent study demonstrated that intramuscular administration of IL-10 ameliorates disease progression and extends survival in SOD1G93A [54]. Consistently, overexpression of IL-10 in the same ALS mouse model reduces inflammatory genes involved in innate immunity [51,52], as well as astrocytosis and microgliosis in the lumbar spinal cord, while the blocking of IL-10 exacerbates inflammation [53] (Figure 3).

2.2. Transforming Growth Factor-β in the CNS and in ALS

Another molecule known to exert anti-inflammatory functions is TGF-β. TGF-β binds the TGF-β receptor, which signals through a serine/threonine kinase domain that phosphorylates the signaling mediator Smad. TGF-β is produced by several immune and non-immune cell types, but the primary source of TGF-β1, which is the most common isoform, are Treg cells [55]. A crucial role of TGF-β was shown by its ability to block nuclear factor-B activation and pro-inflammatory cytokines [56].
TGF-β has been associated with the amelioration of neurotoxicity by reducing glial activation, particularly the microglial pro-inflammatory phenotype. It also decreases the expression of pro-inflammatory factors produced by glial and immune cells, such as TNF-α, inducible nitric oxide synthase (iNOS), IFN-γ, and IL-1β, while promoting the differentiation of Treg cells and the production of the anti-inflammatory cytokine IL-10 [57]. However, in addition to the regulatory effects of TGF-β, it should be noted that TGF-β can also promote Th17 differentiation and IL-17 production [58,59] in inflamed tissues, such as brain tissue in MS patients [60]. These opposing actions may generate contradictory results regarding the therapeutic potential of TGF-β in MS [61].
In ALS, the expression of the genes encoding TGF-β and components of the TGF-β signaling pathways are altered in the spinal cord and skeletal muscle of ALS mice, as well as in muscle biopsies from sporadic ALS patients [62,63], suggesting an involvement in ALS pathogenesis. Notably, the expression of TGFβ-RII expression correlates with the progression of reactive astrogliosis [62], and astrocyte-derived TGF-β1 has been shown to accelerate disease progression in ALS mice [64]. Furthermore, a selective inhibitor of TGFBRI kinase activity extends animal survival in the ALS mouse model, leading to the hypothesis that TGF-β1 may act as a negative prognostic factor, rather than a protective one [64]. In ALS patients, elevated TGF-β1 levels in serum, along with a trend towards increased TGF-β1 in spinal cord tissues, have been associated with a potential anti-inflammatory protective effect in the early stages of the disease, and with a neurotoxic effect in later disease [65]. Consistently, a dual role of TGF-β1 in ALS disease progression is further supported in a transient zebrafish model, where knockdown of tgfb1a partially prevented motor axon abnormalities and locomotor deficits [66] (Figure 3).

2.3. Interleukin-4 in the CNS and in ALS

IL-4 is a cytokine whose anti-inflammatory properties were discovered through its ability to suppress TNF-α and IL-1β production in lipopolysaccharide (LPS)-activated human monocytes [67]. Since then, the list of anti-inflammatory properties of IL-4 has grown, and IL-4 has also been shown to induce the production of the IL-1 receptor antagonist (IL-1ra) [68] and a decoy type II receptor [69], as well as to reduce IFN-γ production by promoting GATA-3 expression [70]. IL-4 receptor complexes (type 1 and type 2) mediate IL-4 signaling through the activation of Jak pathways and STAT6 phosphorylation [69]. The type I IL-4 receptor consists of IL-4 receptor alpha (IL-4Rα) and common γ chain (γc), while the type II IL-4 receptor comprises IL-4Rα and IL-13Rα1.
IL-4 is produced by eosinophils, basophils, mast cells, innate lymphoid T cells (ILC)2, natural killer (NK) T cells, and Th2 cells [71]. It plays a key role in type 2 immune responses, contributing to resistance against helminth parasites, inactivating toxins by stimulating B cell antibody production, and promoting the expansion of eosinophils, basophils, and mast cells [72]. Beyond its immune functions, IL-4 plays a pivotal anti-inflammatory role in the CNS by modulating the microglial polarization. IL-4 facilitates the transition from M1 microglia, the pro-inflammatory phenotype, to M2 microglia, the anti-inflammatory phenotype. This shift is characterized by a reduction in M1 markers (IL-1β, IL-6, TNF-α) and an increase in M2 markers (Arginase-1, TGF-β, IL-10, and CD206) [73]. Yang et al. demonstrated that early intracerebral injection of IL-4 in a rat model of intracerebral hemorrhage (ICH) promotes neurofunctional recovery by enhancing M2 activation and improving neurobehavioral outcomes [74]. The beneficial effects of IL-4 have also been highlighted in MS. In the EAE mouse model of MS, CNS-specific IL-4 deficiency resulted in disease exacerbation, accompanied by increased infiltration of inflammatory cells [75]. Moreover, several studies have shown that IL-4 treatment can reverse disease progression in EAE models [76,77,78]. These findings underscore the critical role of IL-4 as a key regulator of neuroinflammation within the CNS.
In SOD1G93A mice, IL-4 gene therapy resulted in a general improvement in clinical outcomes during the early, slow progressive phase of the disease [79]. Additionally, Zhao et al. demonstrated that in the same model, IL-4 mediates the suppressive functions of Treg cells, including the inhibition of microglial cytotoxicity and the proliferation of T effector cells [80]. Interestingly, this suppressive effect on T cell effector proliferation is also mediated by other known anti-inflammatory cytokines, such as IL-10 and TGF-β [80]. Consistent with preclinical studies, Furukawa et al. reported increased IL-4 levels in the CSF of ALS patients compared to controls, as well as a negative correlation between IL-4 levels and disease progression in ALS patients [50]. In contrast, IL-4 levels in the CSF of ALS patients were negatively correlated with both the ALSFRS-R total and the bulbar subscales, indicating that the role IL-4 in ALS still remains unclear [81] (Figure 3).

2.4. Interleukin-13 in the CNS and in ALS

IL-13 shares many properties with IL-4, and it was initially identified as a T cell-derived cytokine that inhibits the production of inflammatory cytokines [82]. For this reason, it is typically classified as an anti-inflammatory interleukin. Like IL-4, IL-13 is secreted by Th2 cells, ILC2, NK T cells, as well as by mast cells, basophils, and eosinophils [83].
IL-13 has two receptors: the IL-4 type II receptor, composed of IL-4Rα and IL-13Rα1 chains, and IL-13Rα2 [84]; it shares a common signaling pathway with IL-4 [85]. IL-13 exerts similar functions to IL-4, including the regulation of M2 marker expression, such as the mannose receptor, Arginase-1, and IL-4Rα2, all of which are characteristic of anti-inflammatory macrophages [86,87]. In the context of CNS trauma IL-13 administration, either directly or through cell therapy, promotes an anti-inflammatory microglia/macrophage phenotype, and enhances the phagocytosis of damaged neurons in the peri-lesion areas, thereby providing neuroprotection [88,89,90,91,92]. The neuroprotective role of IL-13 has been further demonstrated by Li et al., who showed that IL-13 is upregulated in the brain and CSF of traumatic brain injury patients, with implications for synaptic plasticity and protection of neurons from excitotoxic death [93].
In addition, IL-13 has been reported to modulate brain inflammation by inducing the death of activated microglia, both in vitro and in vivo [94,95]. This effect is mediated by the enhancement of cyclooxygenase-2 (COX-2) expression, and by the production of PGE (2) and 15-deoxy-Delta(12,14)-PGJ(2) (15d-PGJ(2)), key factors involved in the resolution of brain inflammation [96]. In MS, the anti-inflammatory role of IL-13 has been widely reported. In an MS rat model, IL-13 reduced IL-1β and TNF expression as well as oxidative stress in macrophages, thereby mitigating disease severity [97]. In the cuprizone MS mouse model [98], IL-13 gene therapy conferred protection by enhancing the M2 phenotype of microglia and macrophages, thus limiting disease progression [99]. In EAE, treatment with human adipose tissue-derived stem cells (hADSCs) overexpressing IL-11 and IL-13 led to a reduction in the infiltration of inflammatory cells into the spinal cord and improved clinical outcomes [100]. In line with these findings, levels of IL-13 in the CSF of MS patients are positively correlated with axonal and neuronal integrity as well as with better performance in Multiple Sclerosis Functional Composite (MSFC) scoring [101].
Although the anti-inflammatory role of IL-13 in CNS has been largely demonstrated, its role in ALS has not been investigated. However, clinical data indicate an increase of IL-13 levels in the CSF of ALS patients compared to healthy subjects [81] and an increase of IL-13-producing T cells in the peripheral blood of ALS patients compared to healthy donors [102]. Importantly, the levels of IL-13 in CSF of ALS patients are inversely correlated with disease progression [103], while the percentage of IL-13-producing T cells in blood of ALS patients negatively correlates with the revised ALS functional rating scale scores, and positively correlates with the rate of disease progression [102]. These findings suggest that the precise role of IL-13 in ALS disease remains unclear (Figure 3).
Figure 3. The role of anti-inflammatory cytokines in amyotrophic lateral sclerosis (ALS). Schematic representation of the anti-inflammatory cytokines’ effects in ALS, based on preclinical data in ALS animal models and correlative studies based on ALS patient-derived samples and their clinical information (specific references are reported). Arrows indicate the impact of cytokine on neuroinflammation and/or disease progression: sharp arrows represent exacerbation (pathogenic role), and blunt arrows represent inhibition (protective role). The protective effect of IL-10 is supported by different studies, and the protective role of IL-9 is predicted by a single study. In contrast, controversial results are reported about the roles of TGF-β, IL-4, and IL-13. Created in https://BioRender.com. Refs. [45,46,47,48,49,50,58,60,61,62,75,76,77,98,99].
Figure 3. The role of anti-inflammatory cytokines in amyotrophic lateral sclerosis (ALS). Schematic representation of the anti-inflammatory cytokines’ effects in ALS, based on preclinical data in ALS animal models and correlative studies based on ALS patient-derived samples and their clinical information (specific references are reported). Arrows indicate the impact of cytokine on neuroinflammation and/or disease progression: sharp arrows represent exacerbation (pathogenic role), and blunt arrows represent inhibition (protective role). The protective effect of IL-10 is supported by different studies, and the protective role of IL-9 is predicted by a single study. In contrast, controversial results are reported about the roles of TGF-β, IL-4, and IL-13. Created in https://BioRender.com. Refs. [45,46,47,48,49,50,58,60,61,62,75,76,77,98,99].
Ijms 26 03854 g003

2.5. Interleukin-9 in the CNS and in ALS

In recent years, the anti-inflammatory role of IL-9 has emerged. Initially associated with Th2 cells [104,105], IL-9 is now recognized as a defining feature of a specific subset of IL-9-producing CD4+ T cells, known as Th9 cells [34,106]. Interestingly, the anti-inflammatory cytokines TGF-β and IL-4 drive naive CD4+ T cells towards the Th9 profile [34,107], suggesting a regulatory function for IL-9.
IL-9 exerts its action through binding with its specific receptor (IL-9R), composed of the common γ chain, shared by several cytokine receptors, and the specific α-chain (IL-9Rα). IL-9/IL-9R binding triggers Jak kinases activation and phosphorylation of STAT-1, STAT-3, and STAT-5 [108], or activation of the PI3K/Akt and MAPK/ERK pathways [109,110]. IL-9 is involved in the expansion of mast cell populations during allergic responses [111] and in asthma [112,113]. Moreover, IL-9 is involved in immune responses against intestinal nematodes, promoting parasite expulsion from the gut [114,115]. The anti-inflammatory properties of IL-9 were first demonstrated by its ability to reduce respiratory burst and inflammatory cytokine release in human monocytes and alveolar macrophages [116,117]. In line with this, human macrophages stimulated with IL-9 exhibit reduced expression of inflammatory markers and increased TGF-β production, suggesting that IL-9 decreases their activation state and enhances their anti-inflammatory properties [118]. This anti-inflammatory role was also observed in the brain of MS patients, where IL-9 expression was inversely correlated with macrophage/microglia activation [118]. Consistently, in the CSF of MS patients, IL-9 levels inversely correlate with neurofilament levels, a marker of neurodegeneration, as well as with levels of IL-17, which are indicative of neuroinflammation [119]. In line with these results, mice lacking IL-9R exhibit an increase in IL-17-producing cells during EAE, which is associated with enhanced disease progression [120]. Moreover, systemic and local administration of IL-9 in EAE reduces clinical disability by decreasing TNF release by microglia [121]. In contrast, studies using IL-9 neutralization and IL-9 receptor deficiency in the EAE model showed contrasting results [122,123], likely due to experimental differences that warrant further investigation. However, there are no studies on the role of IL-9 in ALS. To date, clinical data have reported an increase of IL-9 levels in the CSF of ALS patients compared to control subjects [81], and that IL-9 higher levels in the CSF predict longer survival in ALS patients [49], suggesting that IL-9 may play protective role in ALS (Figure 3).

2.6. Emerging Anti-Inflammatory Cytokines and Their Role in the CNS

In recent years, the anti-inflammatory roles of newly discovered cytokines such as IL-35, IL-25, IL-37, and IL-27 have emerged.
IL-35 was discovered in 2007 and is highly expressed by cells with immunosuppressive properties, including Treg cells [124], Breg cells [125,126], tolerogenic dendritic cells (tolDCs) [127], and M2 macrophages [128]. IL-35 is composed of the IL-12α chain (p35) and the IL-27β chain Epstein-Barr virus-induced gene 3 (Ebi3), and binds to its receptor IL-35R, which consists of the IL-12Rβ2 and gp130 chains. The binding of IL-35/IL-35R activates the Jak signaling cascade, leading to the phosphorylation of transcriptional activators STAT1, STAT4, and STAT3 [129]. Importantly, IL-35 has been shown to play a significant role in the CNS. For instance, IL-35 alleviates inflammatory responses induced by oxidative stress in a brain stroke model [128], and in hypoxic-ischemic encephalopathy [130]. In addition, in EAE, IL-35 attenuates neuroinflammation by inducing IL-10 production, ultimately reducing disease progression [36,125].
IL-25, also known as IL-17E, is a member of the IL-17 cytokine family with recognized anti-inflammatory properties [131]. To elicit its functional responses, IL-25 requires the presence of both IL-17RA and IL-17RB [132]. It is produced by activated Th2 cells, mast cells, eosinophils, and alveolar macrophages [133,134,135,136,137], and is known for its role in enhancing allergic inflammation by amplifying Th2 immune responses [133,138]. However, in recent years, several anti-inflammatory properties of IL-25 have been identified, including its ability to suppress IL-17-producing T cells [139], inhibit the release of inflammatory cytokines by activated monocytes [140], and induce the polarization of M2 macrophages [141]. These findings have led to the investigation into the effects of IL-25 in EAE, where Th17 cells and inflammatory macrophages play a key pathogenic role. Recent studies have revealed that IL-25 treatment reduces neuroinflammation and EAE disease severity [37,139], suggesting that IL-25 and IL-17, despite being members of the same cytokine family, play opposing roles in the pathogenesis of MS. Similar findings have been observed in mouse models of neuroinflammation, such as those involving the entorhinal cortex lesions [37].
IL-37, discovered in 2000, is a member of the IL-1 family, and is expressed in both extracellular and intracellular pro-inactive protein forms that are activated through proteolytic cleavage. The extracellular form of IL-37 binds the receptor composed of IL-18Rα and IL-1R8 chains, while the intracellular form binds the protein Smad3. Both forms of IL-37 transduce anti-inflammatory signals by suppressing the NF-κB and MAPK pathways while activating the Mer-PTEN-DOK signaling pathways [142]. IL-37 is secreted by innate immune cells and Treg cells [143,144], and plays anti-inflammatory functions in both innate and acquired immune responses by suppressing the activation and production of pro-inflammatory cytokines by monocytes and dendritic cells [145], while also enhancing the immunosuppressive properties of Treg cells [143]. Recent studies have highlighted the pivotal anti-inflammatory role of IL-37 also in the CNS. For instance, the presence of IL-37 in the brain after stroke, observed in both human post-mortem tissues and mouse models, is associated with reduced microglial activation [146], suggesting that IL-37 modulates post-stroke inflammation in the brain. Similarly, in the mouse model of Alzheimer’s disease (AD), IL-37 attenuates microglial activation [35]. Moreover, in EAE, IL-37 decreases the percentage of inflammatory infiltrates such as IFN-γ-producing T lymphocytes [147], and promotes the expansion of Treg cells [148].
IL-27, discovered in 2002, is a member of the IL-12 cytokine superfamily. IL-27 is composed of the Epstein–Barr virus-induced gene 3 (EBI3) and p28 chains, and it binds to a heterodimeric receptor formed by IL-27Rα and the signal transducer gp130 chains [149,150]. The engagement of IL-27R recruits Jak kinases, which predominantly activate the STAT1 and STAT3 pathways [151,152]. IL-27-mediated STAT1 activation in CD4 T cells was initially linked to IFN-γ production and Th1 polarization [149,153]. However, in the last few decades, anti-inflammatory functions of IL-27 have also been described, such as the suppression of IL-17 production [154,155] and the induction of IL-10 production by effector T lymphocytes [156].
IL-27 is produced by innate immune cells [157], B lymphocytes [158], Treg cells [159], and a subtype of Breg cells, known as innate-like IL-27-producing natural regulatory B-1a cell (i27-Breg) [160]. Importantly, the expression of IL-27 and its receptor has been detected in the CSF and post-mortem brain tissues of MS patients [161,162]; additionally, several studies have shown that IL-27 dampens inflammation and disease severity in EAE [38,154,156,163].

3. Anti-Inflammatory Therapeutic Approaches in ALS

Although evidence supports the anti-inflammatory role of cytokines into CNS, most therapeutic efforts aimed at modifying the neuro-inflammatory response in ALS patients have focused on drugs such as Anakinra (a recombinant analog of the endogenous antagonist IL-1Ra) [164,165], Masitinib (a pluripotent tyrosine kinase inhibitor) [166,167], Tocilizumab (the IL-6 receptor neutralizing antibody) [168], and Ibudilast (an inhibitor of toll like receptor 4 and phosphodiesterase 3 and 4) [169] (Table 1). Preliminary data from trials using these drugs are promising. However, these approaches primarily target inflammatory molecules, and such approaches have a high risk of harm, where toxicity may outweigh a beneficial drug effect. In this context, reinforcing the physiological anti-inflammatory response may provide a more effective approach to control neuroinflammatory states compared to drug-mediated immune suppression. Preclinical studies in this direction have shown exciting potential, and some clinical studies have started in recent years. Notably, infusions of autologous Treg cells in three ALS patients have shown a positive correlation with slowed disease progression according to the Appel ALS scale for each patient [170] (Table 1).
It is well known that interleukin 2 (IL-2) is crucial for the generation, activation and survival of Treg cells [171]. Since low dose IL-2 (ld-IL-2) administration selectively expands Treg cells in both mice and humans [172,173], the effects of ld-IL-2 in ALS was evaluated through a phase-2a, randomized, double-blind, placebo-controlled trial. Results from the trial showed that all individuals in both active treatment groups showed an increase in Treg cells. However, differences in neurofilament light chain protein (Nfl) concentrations, used as surrogate markers of efficacy, were not significantly modulated [174] (Table 1). The main limitation of this study was the small sample size, comprising a highly selected population of slowly progressing patients and a short treatment duration. Interestingly, this trial identified CD27 and Toll like receptor (TLR)9 [175], as well as metabolomic profiles [176], as baseline biomarkers that could predict Treg cell expansion after ld-IL-2 treatment.
Rapamycin, another molecule known to promote Treg cell expansion [177], was tested in a randomized, double-blind, placebo-controlled clinical trial in ALS patients. The results showed that treatment with a low dose of rapamycin is safe in patients with ALS, but failed to demonstrate any significant effect on Treg cells [178,179] (Table 1). Further trials focusing on different outcome measures are necessary to better understand the biological and clinical effects of rapamycin in ALS.
Table 1. Therapeutic approaches targeting neuroinflammation in amyotrophic lateral sclerosis (ALS).
Table 1. Therapeutic approaches targeting neuroinflammation in amyotrophic lateral sclerosis (ALS).
NameFunctional RoleClinical TrialPhase TrialClinical ResultsReference
AnakinraAntagonist of IL1RaNCT01277315Phase 2No significant reduction of ALSFRS-R[165]
MasitinibTyrosine kinase inhibitorNCT02588677Phase 2/3Significant decline of ALSFRS-R[167]
TocilizumabNeutralizes IL-6R-Pilot studyDecrease inflammation cytokines in PBMC[168]
IbudilastInhibitor of toll like receptor 4 and phosphodiesterase 3 and 4NCT02714036Phase 1No significant reductions in motor cortical glial activation measured by PBR28-PET SUVR or CNS neuroaxonal loss, measured by serum NfL[169]
Autologous Treg cells Induction of suppressive immune response-Phase 1Reduction of Appel ALS scale for each patient[170]
Low dose IL-2 Expansion of Treg cellsNCT02059759Phase 2Significant increase in Treg cells[174]
RapamycinExpansion of Treg cells-Pilot studyTreatment is safe[178]
RapamycinExpansion of Treg cellsNCT03359538Phase 2Significant increase in Treg cells [179]

4. Conclusions

In recent years, significant advances have been made in understanding neuroinflammation in ALS. It is increasingly recognized that anti-inflammatory cytokines may play a crucial role in regulating neuroinflammation in ALS. This review highlights cytokines with established anti-inflammatory roles, such as IL-4, IL-10, and TGF-β; cytokines with a newly recognized anti-inflammatory role, such as IL-13 and IL-9; and emerging anti-inflammatory cytokines, including IL-25, IL-25, IL-37, and IL-27. We also highlighted the identification of Treg, Breg, M2, Th2, and Th9 cells as both key sources and targets of these cytokines (Figure 2).
In conclusion, enhancing anti-inflammatory cytokines to amplify the regulatory immune response mediated by Treg, Breg, M2, and Th2 cells could represent an innovative therapeutic approach for ALS. While recent studies in this field have yielded encouraging results, several challenges remain, particularly concerning the optimal strategies for amplifying the anti-inflammatory immune response, as well as identifying potential biomarkers to fine-tune personalized therapeutic approaches. Progress in these areas is critical for the future development of new therapies aimed at modulating the immune response for the treatment of ALS and of diseases where neuroinflammation plays a key pathogenic role.

Author Contributions

E.V. and A.S. organized the different sections of the manuscript; E.V. and C.S. wrote the manuscript; A.S., M.C. and S.M.d.R. contributed to the writing of the manuscript. All authors approved the final version. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by FISM-Fondazione Italiana Sclerosi Multipla grant number FISM 2020/R-Single/007 to E.V.

Acknowledgments

We thank Giovanna Borsellino for helpful suggestions and critical reading of the manuscript.

Conflicts of Interest

The authors declare that they have no financial conflicts of interest.

References

  1. DiSabato, D.J.; Quan, N.; Godbout, J.P. Neuroinflammation: The devil is in the details. J. Neurochem. 2016, 139 (Suppl. S2), 136–153. [Google Scholar] [CrossRef]
  2. Beers, D.R.; Appel, S.H. Immune dysregulation in amyotrophic lateral sclerosis: Mechanisms and emerging therapies. Lancet Neurol. 2019, 18, 211–220. [Google Scholar] [CrossRef]
  3. Chakraborty, A.; Diwan, A. Biomarkers and molecular mechanisms of Amyotrophic Lateral Sclerosis. AIMS Neurosci. 2022, 9, 423–443. [Google Scholar] [CrossRef]
  4. Waisman, A.; Liblau, R.S.; Becher, B. Innate and adaptive immune responses in the CNS. Lancet Neurol. 2015, 14, 945–955. [Google Scholar] [CrossRef] [PubMed]
  5. Shechter, R.; London, A.; Schwartz, M. Orchestrated leukocyte recruitment to immune-privileged sites: Absolute barriers versus educational gates. Nat. Rev. Immunol. 2013, 13, 206–218. [Google Scholar] [CrossRef]
  6. Sweeney, M.D.; Sagare, A.P.; Zlokovic, B.V. Blood-brain barrier breakdown in Alzheimer disease and other neurodegenerative disorders. Nat. Rev. Neurol. 2018, 14, 133–150. [Google Scholar] [CrossRef]
  7. Kwon, H.S.; Koh, S.H. Neuroinflammation in neurodegenerative disorders: The roles of microglia and astrocytes. Transl. Neurodegener. 2020, 9, 42. [Google Scholar] [CrossRef]
  8. Tang, Y.; Le, W. Differential Roles of M1 and M2 Microglia in Neurodegenerative Diseases. Mol. Neurobiol. 2016, 53, 1181–1194. [Google Scholar] [CrossRef]
  9. Yang, Q.Q.; Zhou, J.W. Neuroinflammation in the central nervous system: Symphony of glial cells. Glia 2019, 67, 1017–1035. [Google Scholar] [CrossRef]
  10. Boillee, S.; Vande Velde, C.; Cleveland, D.W. ALS: A disease of motor neurons and their nonneuronal neighbors. Neuron 2006, 52, 39–59. [Google Scholar] [CrossRef]
  11. Mead, R.J.; Shan, N.; Reiser, H.J.; Marshall, F.; Shaw, P.J. Amyotrophic lateral sclerosis: A neurodegenerative disorder poised for successful therapeutic translation. Nat. Rev. Drug Discov. 2023, 22, 185–212. [Google Scholar] [CrossRef] [PubMed]
  12. Chen, H.; Kankel, M.W.; Su, S.C.; Han, S.W.S.; Ofengeim, D. Exploring the genetics and non-cell autonomous mechanisms underlying ALS/FTLD. Cell Death Differ. 2018, 25, 648–662. [Google Scholar] [CrossRef] [PubMed]
  13. Lee, J.; Hyeon, S.J.; Im, H.; Ryu, H.; Kim, Y.; Ryu, H. Astrocytes and Microglia as Non-cell Autonomous Players in the Pathogenesis of ALS. Exp. Neurobiol. 2016, 25, 233–240. [Google Scholar] [CrossRef] [PubMed]
  14. Garbuzova-Davis, S.; Sanberg, P.R. Blood-CNS Barrier Impairment in ALS patients versus an animal model. Front. Cell Neurosci. 2014, 8, 21. [Google Scholar] [CrossRef]
  15. Troost, D.; Van den Oord, J.J.; Vianney de Jong, J.M. Immunohistochemical characterization of the inflammatory infiltrate in amyotrophic lateral sclerosis. Neuropathol. Appl. Neurobiol. 1990, 16, 401–410. [Google Scholar] [CrossRef]
  16. Gille, B.; De Schaepdryver, M.; Dedeene, L.; Goossens, J.; Claeys, K.G.; Van Den Bosch, L.; Tournoy, J.; Van Damme, P.; Poesen, K. Inflammatory markers in cerebrospinal fluid: Independent prognostic biomarkers in amyotrophic lateral sclerosis? J. Neurol. Neurosurg. Psychiatry 2019, 90, 1338–1346. [Google Scholar] [CrossRef]
  17. Zhao, W.; Beers, D.R.; Hooten, K.G.; Sieglaff, D.H.; Zhang, A.; Kalyana-Sundaram, S.; Traini, C.M.; Halsey, W.S.; Hughes, A.M.; Sathe, G.M.; et al. Characterization of Gene Expression Phenotype in Amyotrophic Lateral Sclerosis Monocytes. JAMA Neurol. 2017, 74, 677–685. [Google Scholar] [CrossRef] [PubMed]
  18. Mantovani, S.; Garbelli, S.; Pasini, A.; Alimonti, D.; Perotti, C.; Melazzini, M.; Bendotti, C.; Mora, G. Immune system alterations in sporadic amyotrophic lateral sclerosis patients suggest an ongoing neuroinflammatory process. J. Neuroimmunol. 2009, 210, 73–79. [Google Scholar] [CrossRef]
  19. Wing, K.; Sakaguchi, S. Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nat. Immunol. 2010, 11, 7–13. [Google Scholar] [CrossRef]
  20. Henkel, J.S.; Beers, D.R.; Wen, S.; Rivera, A.L.; Toennis, K.M.; Appel, J.E.; Zhao, W.; Moore, D.H.; Powell, S.Z.; Appel, S.H. Regulatory T-lymphocytes mediate amyotrophic lateral sclerosis progression and survival. EMBO Mol. Med. 2013, 5, 64–79. [Google Scholar] [CrossRef]
  21. Sheean, R.K.; McKay, F.C.; Cretney, E.; Bye, C.R.; Perera, N.D.; Tomas, D.; Weston, R.A.; Scheller, K.J.; Djouma, E.; Menon, P.; et al. Association of Regulatory T-Cell Expansion with Progression of Amyotrophic Lateral Sclerosis: A Study of Humans and a Transgenic Mouse Model. JAMA Neurol. 2018, 75, 681–689. [Google Scholar] [CrossRef] [PubMed]
  22. Coque, E.; Salsac, C.; Espinosa-Carrasco, G.; Varga, B.; Degauque, N.; Cadoux, M.; Crabe, R.; Virenque, A.; Soulard, C.; Fierle, J.K.; et al. Cytotoxic CD8(+) T lymphocytes expressing ALS-causing SOD1 mutant selectively trigger death of spinal motoneurons. Proc. Natl. Acad. Sci. USA 2019, 116, 2312–2317. [Google Scholar] [CrossRef]
  23. McCauley, M.E.; O’Rourke, J.G.; Yanez, A.; Markman, J.L.; Ho, R.; Wang, X.; Chen, S.; Lall, D.; Jin, M.; Muhammad, A.; et al. C9orf72 in myeloid cells suppresses STING-induced inflammation. Nature 2020, 585, 96–101. [Google Scholar] [CrossRef]
  24. Yu, C.H.; Davidson, S.; Harapas, C.R.; Hilton, J.B.; Mlodzianoski, M.J.; Laohamonthonkul, P.; Louis, C.; Low, R.R.J.; Moecking, J.; De Nardo, D.; et al. TDP-43 Triggers Mitochondrial DNA Release via mPTP to Activate cGAS/STING in ALS. Cell 2020, 183, 636–649. [Google Scholar] [CrossRef] [PubMed]
  25. Murdock, B.J.; Famie, J.P.; Piecuch, C.E.; Pawlowski, K.D.; Mendelson, F.E.; Pieroni, C.H.; Iniguez, S.D.; Zhao, L.; Goutman, S.A.; Feldman, E.L. NK cells associate with ALS in a sex- and age-dependent manner. JCI Insight 2021, 6, e147129. [Google Scholar] [CrossRef]
  26. Murdock, B.J.; Zhou, T.; Kashlan, S.R.; Little, R.J.; Goutman, S.A.; Feldman, E.L. Correlation of Peripheral Immunity with Rapid Amyotrophic Lateral Sclerosis Progression. JAMA Neurol. 2017, 74, 1446–1454. [Google Scholar] [CrossRef]
  27. Xiao, Q.; Zhao, W.; Beers, D.R.; Yen, A.A.; Xie, W.; Henkel, J.S.; Appel, S.H. Mutant SOD1(G93A) microglia are more neurotoxic relative to wild-type microglia. J. Neurochem. 2007, 102, 2008–2019. [Google Scholar] [CrossRef]
  28. Zhao, W.; Beers, D.R.; Bell, S.; Wang, J.; Wen, S.; Baloh, R.H.; Appel, S.H. TDP-43 activates microglia through NF-kappaB and NLRP3 inflammasome. Exp. Neurol. 2015, 273, 24–35. [Google Scholar] [CrossRef]
  29. Gerbino, V.; Kaunga, E.; Ye, J.; Canzio, D.; O’Keeffe, S.; Rudnick, N.D.; Guarnieri, P.; Lutz, C.M.; Maniatis, T. The Loss of TBK1 Kinase Activity in Motor Neurons or in All Cell Types Differentially Impacts ALS Disease Progression in SOD1 Mice. Neuron 2020, 106, 789–805.e5. [Google Scholar] [CrossRef]
  30. Markovinovic, A.; Ljutic, T.; Beland, L.C.; Munitic, I. Optineurin Insufficiency Disbalances Proinflammatory and Anti-inflammatory Factors by Reducing Microglial IFN-beta Responses. Neuroscience 2018, 388, 139–151. [Google Scholar] [CrossRef]
  31. Levings, M.K.; Bacchetta, R.; Schulz, U.; Roncarolo, M.G. The role of IL-10 and TGF-beta in the differentiation and effector function of T regulatory cells. Int. Arch. Allergy Immunol. 2002, 129, 263–276. [Google Scholar] [CrossRef] [PubMed]
  32. Raphael, I.; Nalawade, S.; Eagar, T.N.; Forsthuber, T.G. T cell subsets and their signature cytokines in autoimmune and inflammatory diseases. Cytokine 2015, 74, 5–17. [Google Scholar] [CrossRef] [PubMed]
  33. Kokubo, K.; Onodera, A.; Kiuchi, M.; Tsuji, K.; Hirahara, K.; Nakayama, T. Conventional and pathogenic Th2 cells in inflammation, tissue repair, and fibrosis. Front. Immunol. 2022, 13, 945063. [Google Scholar] [CrossRef]
  34. Veldhoen, M.; Uyttenhove, C.; van Snick, J.; Helmby, H.; Westendorf, A.; Buer, J.; Martin, B.; Wilhelm, C.; Stockinger, B. Transforming growth factor-beta ‘reprograms’ the differentiation of T helper 2 cells and promotes an interleukin 9-producing subset. Nat. Immunol. 2008, 9, 1341–1346. [Google Scholar] [CrossRef]
  35. Lonnemann, N.; Hosseini, S.; Ohm, M.; Geffers, R.; Hiller, K.; Dinarello, C.A.; Korte, M. IL-37 expression reduces acute and chronic neuroinflammation and rescues cognitive impairment in an Alzheimer’s disease mouse model. Elife 2022, 11, e75889. [Google Scholar] [CrossRef] [PubMed]
  36. Duffy, S.S.; Keating, B.A.; Perera, C.J.; Lees, J.G.; Tonkin, R.S.; Makker, P.G.S.; Carrive, P.; Butovsky, O.; Moalem-Taylor, G. Regulatory T Cells and Their Derived Cytokine, Interleukin-35, Reduce Pain in Experimental Autoimmune Encephalomyelitis. J. Neurosci. 2019, 39, 2326–2346. [Google Scholar] [CrossRef]
  37. Maiorino, C.; Khorooshi, R.; Ruffini, F.; Lobner, M.; Bergami, A.; Garzetti, L.; Martino, G.; Owens, T.; Furlan, R. Lentiviral-mediated administration of IL-25 in the CNS induces alternative activation of microglia. Gene Ther. 2013, 20, 487–496. [Google Scholar] [CrossRef]
  38. Casella, G.; Finardi, A.; Descamps, H.; Colombo, F.; Maiorino, C.; Ruffini, F.; Patrone, M.; Degano, M.; Martino, G.; Muzio, L.; et al. IL-27, but not IL-35, inhibits neuroinflammation through modulating GM-CSF expression. Sci. Rep. 2017, 7, 16547. [Google Scholar] [CrossRef]
  39. Saraiva, M.; O’Garra, A. The regulation of IL-10 production by immune cells. Nat. Rev. Immunol. 2010, 10, 170–181. [Google Scholar] [CrossRef]
  40. Mosser, D.M.; Zhang, X. Interleukin-10: New perspectives on an old cytokine. Immunol. Rev. 2008, 226, 205–218. [Google Scholar] [CrossRef]
  41. Moore, K.W.; de Waal Malefyt, R.; Coffman, R.L.; O’Garra, A. Interleukin-10 and the interleukin-10 receptor. Annu. Rev. Immunol. 2001, 19, 683–765. [Google Scholar] [CrossRef] [PubMed]
  42. de Waal Malefyt, R.; Abrams, J.; Bennett, B.; Figdor, C.G.; de Vries, J.E. Interleukin 10(IL-10) inhibits cytokine synthesis by human monocytes: An autoregulatory role of IL-10 produced by monocytes. J. Exp. Med. 1991, 174, 1209–1220. [Google Scholar] [CrossRef] [PubMed]
  43. Kim, Y.; Kim, G.; Shin, H.J.; Hyun, J.W.; Kim, S.H.; Lee, E.; Kim, H.J. Restoration of regulatory B cell deficiency following alemtuzumab therapy in patients with relapsing multiple sclerosis. J. NeuroInflamm. 2018, 15, 300. [Google Scholar] [CrossRef]
  44. Piancone, F.; Saresella, M.; Marventano, I.; La Rosa, F.; Zoppis, M.; Agostini, S.; Longhi, R.; Caputo, D.; Mendozzi, L.; Rovaris, M.; et al. B Lymphocytes in Multiple Sclerosis: Bregs and BTLA/CD272 Expressing-CD19+ Lymphocytes Modulate Disease Severity. Sci. Rep. 2016, 6, 29699. [Google Scholar] [CrossRef] [PubMed]
  45. Bettelli, E.; Das, M.P.; Howard, E.D.; Weiner, H.L.; Sobel, R.A.; Kuchroo, V.K. IL-10 is critical in the regulation of autoimmune encephalomyelitis as demonstrated by studies of IL-10- and IL-4-deficient and transgenic mice. J. Immunol. 1998, 161, 3299–3306. [Google Scholar] [CrossRef]
  46. Bido, S.; Nannoni, M.; Muggeo, S.; Gambare, D.; Ruffini, G.; Bellini, E.; Passeri, L.; Iaia, S.; Luoni, M.; Provinciali, M.; et al. Microglia-specific IL-10 gene delivery inhibits neuroinflammation and neurodegeneration in a mouse model of Parkinson’s disease. Sci. Transl. Med. 2024, 16, eadm8563. [Google Scholar] [CrossRef]
  47. Brodovitch, A.; Boucraut, J.; Delmont, E.; Parlanti, A.; Grapperon, A.M.; Attarian, S.; Verschueren, A. Combination of serum and CSF neurofilament-light and neuroinflammatory biomarkers to evaluate ALS. Sci. Rep. 2021, 11, 703. [Google Scholar] [CrossRef]
  48. Guo, J.; Yang, X.; Gao, L.; Zang, D. Evaluating the levels of CSF and serum factors in ALS. Brain Behav. 2017, 7, e00637. [Google Scholar] [CrossRef]
  49. Su, X.W.; Simmons, Z.; Mitchell, R.M.; Kong, L.; Stephens, H.E.; Connor, J.R. Biomarker-based predictive models for prognosis in amyotrophic lateral sclerosis. JAMA Neurol. 2013, 70, 1505–1511. [Google Scholar] [CrossRef]
  50. Furukawa, T.; Matsui, N.; Fujita, K.; Nodera, H.; Shimizu, F.; Miyamoto, K.; Takahashi, Y.; Kanda, T.; Kusunoki, S.; Izumi, Y.; et al. CSF cytokine profile distinguishes multifocal motor neuropathy from progressive muscular atrophy. Neurol. Neuroimmunol. Neuroinflamm. 2015, 2, e138. [Google Scholar] [CrossRef]
  51. Ayers, J.I.; Fromholt, S.; Sinyavskaya, O.; Siemienski, Z.; Rosario, A.M.; Li, A.; Crosby, K.W.; Cruz, P.E.; DiNunno, N.M.; Janus, C.; et al. Widespread and efficient transduction of spinal cord and brain following neonatal AAV injection and potential disease modifying effect in ALS mice. Mol. Ther. 2015, 23, 53–62. [Google Scholar] [CrossRef] [PubMed]
  52. Strickland, M.R.; Ibanez, K.R.; Yaroshenko, M.; Diaz, C.C.; Borchelt, D.R.; Chakrabarty, P. IL-10 based immunomodulation initiated at birth extends lifespan in a familial mouse model of amyotrophic lateral sclerosis. Sci. Rep. 2020, 10, 20862. [Google Scholar] [CrossRef]
  53. Gravel, M.; Beland, L.C.; Soucy, G.; Abdelhamid, E.; Rahimian, R.; Gravel, C.; Kriz, J. IL-10 Controls Early Microglial Phenotypes and Disease Onset in ALS Caused by Misfolded Superoxide Dismutase 1. J. Neurosci. 2016, 36, 1031–1048. [Google Scholar] [CrossRef]
  54. Fabbrizio, P.; Margotta, C.; D’Agostino, J.; Suanno, G.; Quetti, L.; Bendotti, C.; Nardo, G. Intramuscular IL-10 Administration Enhances the Activity of Myogenic Precursor Cells and Improves Motor Function in ALS Mouse Model. Cells 2023, 12, 1016. [Google Scholar] [CrossRef]
  55. Vignali, D.A.; Collison, L.W.; Workman, C.J. How regulatory T cells work. Nat. Rev. Immunol. 2008, 8, 523–532. [Google Scholar] [CrossRef]
  56. Mirshafiey, A.; Mohsenzadegan, M. TGF-beta as a promising option in the treatment of multiple sclerosis. Neuropharmacology 2009, 56, 929–936. [Google Scholar] [CrossRef] [PubMed]
  57. Chen, J.H.; Ke, K.F.; Lu, J.H.; Qiu, Y.H.; Peng, Y.P. Protection of TGF-beta1 against neuroinflammation and neurodegeneration in Abeta1-42-induced Alzheimer’s disease model rats. PLoS ONE 2015, 10, e0116549. [Google Scholar] [CrossRef]
  58. Veldhoen, M.; Hocking, R.J.; Atkins, C.J.; Locksley, R.M.; Stockinger, B. TGFbeta in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity 2006, 24, 179–189. [Google Scholar] [CrossRef]
  59. Volpe, E.; Servant, N.; Zollinger, R.; Bogiatzi, S.I.; Hupe, P.; Barillot, E.; Soumelis, V. A critical function for transforming growth factor-beta, interleukin 23 and proinflammatory cytokines in driving and modulating human T(H)-17 responses. Nat. Immunol. 2008, 9, 650–657. [Google Scholar] [CrossRef]
  60. Park, H.; Li, Z.; Yang, X.O.; Chang, S.H.; Nurieva, R.; Wang, Y.H.; Wang, Y.; Hood, L.; Zhu, Z.; Tian, Q.; et al. A distinct lineage of CD4 T cells regulates tissue inflammation by producing interleukin 17. Nat. Immunol. 2005, 6, 1133–1141. [Google Scholar] [CrossRef]
  61. Esmaeilzadeh, A.; Mohammadi, V.; Elahi, R. Transforming growth factor beta (TGF-beta) pathway in the immunopathogenesis of multiple sclerosis (MS); molecular approaches. Mol. Biol. Rep. 2023, 50, 6121–6131. [Google Scholar] [CrossRef]
  62. Meroni, M.; Crippa, V.; Cristofani, R.; Rusmini, P.; Cicardi, M.E.; Messi, E.; Piccolella, M.; Tedesco, B.; Ferrari, V.; Soraru, G.; et al. Transforming growth factor beta 1 signaling is altered in the spinal cord and muscle of amyotrophic lateral sclerosis mice and patients. Neurobiol. Aging 2019, 82, 48–59. [Google Scholar] [CrossRef]
  63. Phatnani, H.P.; Guarnieri, P.; Friedman, B.A.; Carrasco, M.A.; Muratet, M.; O’Keeffe, S.; Nwakeze, C.; Pauli-Behn, F.; Newberry, K.M.; Meadows, S.K.; et al. Intricate interplay between astrocytes and motor neurons in ALS. Proc. Natl. Acad. Sci. USA 2013, 110, E756–E765. [Google Scholar] [CrossRef]
  64. Endo, F.; Komine, O.; Fujimori-Tonou, N.; Katsuno, M.; Jin, S.; Watanabe, S.; Sobue, G.; Dezawa, M.; Wyss-Coray, T.; Yamanaka, K. Astrocyte-derived TGF-beta1 accelerates disease progression in ALS mice by interfering with the neuroprotective functions of microglia and T cells. Cell Rep. 2015, 11, 592–604. [Google Scholar] [CrossRef]
  65. Peters, S.; Zitzelsperger, E.; Kuespert, S.; Iberl, S.; Heydn, R.; Johannesen, S.; Petri, S.; Aigner, L.; Thal, D.R.; Hermann, A.; et al. The TGF-beta System as a Potential Pathogenic Player in Disease Modulation of Amyotrophic Lateral Sclerosis. Front. Neurol. 2017, 8, 669. [Google Scholar] [CrossRef]
  66. Gonzalez, D.; Cuenca, X.; Allende, M.L. Knockdown of tgfb1a partially improves ALS phenotype in a transient zebrafish model. Front. Cell Neurosci. 2024, 18, 1384085. [Google Scholar] [CrossRef] [PubMed]
  67. Hart, P.H.; Vitti, G.F.; Burgess, D.R.; Whitty, G.A.; Piccoli, D.S.; Hamilton, J.A. Potential antiinflammatory effects of interleukin 4: Suppression of human monocyte tumor necrosis factor alpha, interleukin 1, and prostaglandin E2. Proc. Natl. Acad. Sci. USA 1989, 86, 3803–3807. [Google Scholar] [CrossRef]
  68. Fenton, M.J.; Buras, J.A.; Donnelly, R.P. IL-4 reciprocally regulates IL-1 and IL-1 receptor antagonist expression in human monocytes. J. Immunol. 1992, 149, 1283–1288. [Google Scholar] [CrossRef]
  69. Colotta, F.; Re, F.; Muzio, M.; Bertini, R.; Polentarutti, N.; Sironi, M.; Giri, J.G.; Dower, S.K.; Sims, J.E.; Mantovani, A. Interleukin-1 type II receptor: A decoy target for IL-1 that is regulated by IL-4. Science 1993, 261, 472–475. [Google Scholar] [CrossRef]
  70. Ferber, I.A.; Lee, H.J.; Zonin, F.; Heath, V.; Mui, A.; Arai, N.; O’Garra, A. GATA-3 significantly downregulates IFN-gamma production from developing Th1 cells in addition to inducing IL-4 and IL-5 levels. Clin. Immunol. 1999, 91, 134–144. [Google Scholar] [CrossRef]
  71. Luzina, I.G.; Keegan, A.D.; Heller, N.M.; Rook, G.A.; Shea-Donohue, T.; Atamas, S.P. Regulation of inflammation by interleukin-4: A review of “alternatives”. J. Leukoc. Biol. 2012, 92, 753–764. [Google Scholar] [CrossRef]
  72. Heeb, L.E.M.; Egholm, C.; Boyman, O. Evolution and function of interleukin-4 receptor signaling in adaptive immunity and neutrophils. Genes. Immun. 2020, 21, 143–149. [Google Scholar] [CrossRef] [PubMed]
  73. Guo, J.; Tang, X.; Deng, P.; Hui, H.; Chen, B.; An, J.; Zhang, G.; Shi, K.; Wang, J.; He, Y.; et al. Interleukin-4 from curcumin-activated OECs emerges as a central modulator for increasing M2 polarization of microglia/macrophage in OEC anti-inflammatory activity for functional repair of spinal cord injury. Cell Commun. Signal 2024, 22, 162. [Google Scholar] [CrossRef] [PubMed]
  74. Yang, J.; Ding, S.; Huang, W.; Hu, J.; Huang, S.; Zhang, Y.; Zhuge, Q. Interleukin-4 Ameliorates the Functional Recovery of Intracerebral Hemorrhage Through the Alternative Activation of Microglia/Macrophage. Front. Neurosci. 2016, 10, 61. [Google Scholar] [CrossRef]
  75. Ponomarev, E.D.; Maresz, K.; Tan, Y.; Dittel, B.N. CNS-derived interleukin-4 is essential for the regulation of autoimmune inflammation and induces a state of alternative activation in microglial cells. J. Neurosci. 2007, 27, 10714–10721. [Google Scholar] [CrossRef]
  76. Shaw, M.K.; Lorens, J.B.; Dhawan, A.; DalCanto, R.; Tse, H.Y.; Tran, A.B.; Bonpane, C.; Eswaran, S.L.; Brocke, S.; Sarvetnick, N.; et al. Local delivery of interleukin 4 by retrovirus-transduced T lymphocytes ameliorates experimental autoimmune encephalomyelitis. J. Exp. Med. 1997, 185, 1711–1714. [Google Scholar] [CrossRef]
  77. Furlan, R.; Poliani, P.L.; Galbiati, F.; Bergami, A.; Grimaldi, L.M.; Comi, G.; Adorini, L.; Martino, G. Central nervous system delivery of interleukin 4 by a nonreplicative herpes simplex type 1 viral vector ameliorates autoimmune demyelination. Hum. Gene Ther. 1998, 9, 2605–2617. [Google Scholar] [CrossRef]
  78. Vogelaar, C.F.; Mandal, S.; Lerch, S.; Birkner, K.; Birkenstock, J.; Buhler, U.; Schnatz, A.; Raine, C.S.; Bittner, S.; Vogt, J.; et al. Fast direct neuronal signaling via the IL-4 receptor as therapeutic target in neuroinflammation. Sci. Transl. Med. 2018, 10, eaao2304. [Google Scholar] [CrossRef]
  79. Rossi, C.; Cusimano, M.; Zambito, M.; Finardi, A.; Capotondo, A.; Garcia-Manteiga, J.M.; Comi, G.; Furlan, R.; Martino, G.; Muzio, L. Interleukin 4 modulates microglia homeostasis and attenuates the early slowly progressive phase of amyotrophic lateral sclerosis. Cell Death Dis. 2018, 9, 250. [Google Scholar] [CrossRef]
  80. Zhao, W.; Beers, D.R.; Liao, B.; Henkel, J.S.; Appel, S.H. Regulatory T lymphocytes from ALS mice suppress microglia and effector T lymphocytes through different cytokine-mediated mechanisms. Neurobiol. Dis. 2012, 48, 418–428. [Google Scholar] [CrossRef]
  81. Femiano, C.; Bruno, A.; Gilio, L.; Buttari, F.; Dolcetti, E.; Galifi, G.; Azzolini, F.; Borrelli, A.; Furlan, R.; Finardi, A.; et al. Inflammatory signature in amyotrophic lateral sclerosis predicting disease progression. Sci. Rep. 2024, 14, 19796. [Google Scholar] [CrossRef] [PubMed]
  82. Minty, A.; Chalon, P.; Derocq, J.M.; Dumont, X.; Guillemot, J.C.; Kaghad, M.; Labit, C.; Leplatois, P.; Liauzun, P.; Miloux, B.; et al. Interleukin-13 is a new human lymphokine regulating inflammatory and immune responses. Nature 1993, 362, 248–250. [Google Scholar] [CrossRef] [PubMed]
  83. Junttila, I.S. Tuning the Cytokine Responses: An Update on Interleukin (IL)-4 and IL-13 Receptor Complexes. Front. Immunol. 2018, 9, 888. [Google Scholar] [CrossRef] [PubMed]
  84. Wills-Karp, M.; Finkelman, F.D. Untangling the complex web of IL-4- and IL-13-mediated signaling pathways. Sci. Signal 2008, 1, pe55. [Google Scholar] [CrossRef]
  85. Bhattacharjee, A.; Shukla, M.; Yakubenko, V.P.; Mulya, A.; Kundu, S.; Cathcart, M.K. IL-4 and IL-13 employ discrete signaling pathways for target gene expression in alternatively activated monocytes/macrophages. Free Radic. Biol. Med. 2013, 54, 1–16. [Google Scholar] [CrossRef]
  86. Luo, M.; Zhao, F.; Cheng, H.; Su, M.; Wang, Y. Macrophage polarization: An important role in inflammatory diseases. Front. Immunol. 2024, 15, 1352946. [Google Scholar] [CrossRef]
  87. Sica, A.; Mantovani, A. Macrophage plasticity and polarization: In vivo veritas. J. Clin. Investig. 2012, 122, 787–795. [Google Scholar] [CrossRef]
  88. Hamzei Taj, S.; Le Blon, D.; Hoornaert, C.; Daans, J.; Quarta, A.; Praet, J.; Van der Linden, A.; Ponsaerts, P.; Hoehn, M. Targeted intracerebral delivery of the anti-inflammatory cytokine IL13 promotes alternative activation of both microglia and macrophages after stroke. J. Neuroinflamm. 2018, 15, 174. [Google Scholar] [CrossRef]
  89. Van Broeckhoven, J.; Erens, C.; Sommer, D.; Scheijen, E.; Sanchez, S.; Vidal, P.M.; Dooley, D.; Van Breedam, E.; Quarta, A.; Ponsaerts, P.; et al. Macrophage-based delivery of interleukin-13 improves functional and histopathological outcomes following spinal cord injury. J. Neuroinflamm. 2022, 19, 102. [Google Scholar] [CrossRef]
  90. Kolosowska, N.; Keuters, M.H.; Wojciechowski, S.; Keksa-Goldsteine, V.; Laine, M.; Malm, T.; Goldsteins, G.; Koistinaho, J.; Dhungana, H. Peripheral Administration of IL-13 Induces Anti-inflammatory Microglial/Macrophage Responses and Provides Neuroprotection in Ischemic Stroke. Neurotherapeutics 2019, 16, 1304–1319. [Google Scholar] [CrossRef]
  91. Miao, W.; Zhao, Y.; Huang, Y.; Chen, D.; Luo, C.; Su, W.; Gao, Y. IL-13 Ameliorates Neuroinflammation and Promotes Functional Recovery after Traumatic Brain Injury. J. Immunol. 2020, 204, 1486–1498. [Google Scholar] [CrossRef] [PubMed]
  92. Dooley, D.; Lemmens, E.; Vangansewinkel, T.; Le Blon, D.; Hoornaert, C.; Ponsaerts, P.; Hendrix, S. Cell-Based Delivery of Interleukin-13 Directs Alternative Activation of Macrophages Resulting in Improved Functional Outcome after Spinal Cord Injury. Stem. Cell Rep. 2016, 7, 1099–1115. [Google Scholar] [CrossRef] [PubMed]
  93. Li, S.; Olde Heuvel, F.; Rehman, R.; Aousji, O.; Froehlich, A.; Li, Z.; Jark, R.; Zhang, W.; Conquest, A.; Woelfle, S.; et al. Interleukin-13 and its receptor are synaptic proteins involved in plasticity and neuroprotection. Nat. Commun. 2023, 14, 200. [Google Scholar] [CrossRef] [PubMed]
  94. Shin, W.H.; Lee, D.Y.; Park, K.W.; Kim, S.U.; Yang, M.S.; Joe, E.H.; Jin, B.K. Microglia expressing interleukin-13 undergo cell death and contribute to neuronal survival in vivo. Glia 2004, 46, 142–152. [Google Scholar] [CrossRef]
  95. Yang, M.S.; Park, E.J.; Sohn, S.; Kwon, H.J.; Shin, W.H.; Pyo, H.K.; Jin, B.; Choi, K.S.; Jou, I.; Joe, E.H. Interleukin-13 and -4 induce death of activated microglia. Glia 2002, 38, 273–280. [Google Scholar] [CrossRef]
  96. Yang, M.S.; Ji, K.A.; Jeon, S.B.; Jin, B.K.; Kim, S.U.; Jou, I.; Joe, E. Interleukin-13 enhances cyclooxygenase-2 expression in activated rat brain microglia: Implications for death of activated microglia. J. Immunol. 2006, 177, 1323–1329. [Google Scholar] [CrossRef]
  97. Cash, E.; Minty, A.; Ferrara, P.; Caput, D.; Fradelizi, D.; Rott, O. Macrophage-inactivating IL-13 suppresses experimental autoimmune encephalomyelitis in rats. J. Immunol. 1994, 153, 4258–4267. [Google Scholar] [CrossRef]
  98. Torkildsen, O.; Brunborg, L.A.; Myhr, K.M.; Bo, L. The cuprizone model for demyelination. Acta Neurol. Scand. Suppl. 2008, 188, 72–76. [Google Scholar] [CrossRef]
  99. Guglielmetti, C.; Le Blon, D.; Santermans, E.; Salas-Perdomo, A.; Daans, J.; De Vocht, N.; Shah, D.; Hoornaert, C.; Praet, J.; Peerlings, J.; et al. Interleukin-13 immune gene therapy prevents CNS inflammation and demyelination via alternative activation of microglia and macrophages. Glia 2016, 64, 2181–2200. [Google Scholar] [CrossRef]
  100. Azimzadeh, M.; Mahmoodi, M.; Kazemi, M.; Hakemi, M.G.; Jafarinia, M.; Eslami, A.; Salehi, H.; Amirpour, N. The immunoregulatory and neuroprotective effects of human adipose derived stem cells overexpressing IL-11 and IL-13 in the experimental autoimmune encephalomyelitis mice. Int. Immunopharmacol. 2020, 87, 106808. [Google Scholar] [CrossRef]
  101. Rossi, S.; Mancino, R.; Bergami, A.; Mori, F.; Castelli, M.; De Chiara, V.; Studer, V.; Mataluni, G.; Sancesario, G.; Parisi, V.; et al. Potential role of IL-13 in neuroprotection and cortical excitability regulation in multiple sclerosis. Mult. Scler. 2011, 17, 1301–1312. [Google Scholar] [CrossRef] [PubMed]
  102. Shi, N.; Kawano, Y.; Tateishi, T.; Kikuchi, H.; Osoegawa, M.; Ohyagi, Y.; Kira, J. Increased IL-13-producing T cells in ALS: Positive correlations with disease severity and progression rate. J. Neuroimmunol. 2007, 182, 232–235. [Google Scholar] [CrossRef]
  103. Xu, C.Z.; Huan, X.; Luo, S.S.; Zhong, H.H.; Zhao, C.B.; Chen, Y.; Zou, Z.Y.; Chen, S. Serum cytokines profile changes in amyotrophic lateral sclerosis. Heliyon 2024, 10, e28553. [Google Scholar] [CrossRef] [PubMed]
  104. Schmitt, E.; Van Brandwijk, R.; Fischer, H.G.; Rude, E. Establishment of different T cell sublines using either interleukin 2 or interleukin 4 as growth factors. Eur. J. Immunol. 1990, 20, 1709–1715. [Google Scholar] [CrossRef]
  105. Goswami, R.; Kaplan, M.H. A brief history of IL-9. J. Immunol. 2011, 186, 3283–3288. [Google Scholar] [CrossRef]
  106. Dardalhon, V.; Awasthi, A.; Kwon, H.; Galileos, G.; Gao, W.; Sobel, R.A.; Mitsdoerffer, M.; Strom, T.B.; Elyaman, W.; Ho, I.C.; et al. IL-4 inhibits TGF-beta-induced Foxp3+ T cells and, together with TGF-beta, generates IL-9+ IL-10+ Foxp3(-) effector T cells. Nat. Immunol. 2008, 9, 1347–1355. [Google Scholar] [CrossRef]
  107. Stassen, M.; Schmitt, E.; Bopp, T. From interleukin-9 to T helper 9 cells. Ann. N. Y. Acad. Sci. 2012, 1247, 56–68. [Google Scholar] [CrossRef] [PubMed]
  108. Demoulin, J.B.; Renauld, J.C. Interleukin 9 and its receptor: An overview of structure and function. Int. Rev. Immunol. 1998, 16, 345–364. [Google Scholar] [CrossRef]
  109. Yin, T.; Keller, S.R.; Quelle, F.W.; Witthuhn, B.A.; Tsang, M.L.; Lienhard, G.E.; Ihle, J.N.; Yang, Y.C. Interleukin-9 induces tyrosine phosphorylation of insulin receptor substrate-1 via JAK tyrosine kinases. J. Biol. Chem. 1995, 270, 20497–20502. [Google Scholar] [CrossRef]
  110. Chakraborty, S.; Kubatzky, K.F.; Mitra, D.K. An Update on Interleukin-9: From Its Cellular Source and Signal Transduction to Its Role in Immunopathogenesis. Int. J. Mol. Sci. 2019, 20, 2113. [Google Scholar] [CrossRef]
  111. Noelle, R.J.; Nowak, E.C. Cellular sources and immune functions of interleukin-9. Nat. Rev. Immunol. 2010, 10, 683–687. [Google Scholar] [CrossRef] [PubMed]
  112. Levitt, R.C.; McLane, M.P.; MacDonald, D.; Ferrante, V.; Weiss, C.; Zhou, T.; Holroyd, K.J.; Nicolaides, N.C. IL-9 pathway in asthma: New therapeutic targets for allergic inflammatory disorders. J. Allergy Clin. Immunol. 1999, 103, S485–S491. [Google Scholar] [CrossRef]
  113. Doherty, T.A.; Broide, D.H. Insights into the biology of IL-9 in asthma. J. Allergy Clin. Immunol. 2022, 150, 585–586. [Google Scholar] [CrossRef] [PubMed]
  114. Faulkner, H.; Humphreys, N.; Renauld, J.C.; Van Snick, J.; Grencis, R. Interleukin-9 is involved in host protective immunity to intestinal nematode infection. Eur. J. Immunol. 1997, 27, 2536–2540. [Google Scholar] [CrossRef]
  115. Faulkner, H.; Renauld, J.C.; Van Snick, J.; Grencis, R.K. Interleukin-9 enhances resistance to the intestinal nematode Trichuris muris. Infect. Immun. 1998, 66, 3832–3840. [Google Scholar] [CrossRef]
  116. Pilette, C.; Ouadrhiri, Y.; Van Snick, J.; Renauld, J.C.; Staquet, P.; Vaerman, J.P.; Sibille, Y. Oxidative burst in lipopolysaccharide-activated human alveolar macrophages is inhibited by interleukin-9. Eur. Respir. J. 2002, 20, 1198–1205. [Google Scholar] [CrossRef] [PubMed]
  117. Pilette, C.; Ouadrhiri, Y.; Van Snick, J.; Renauld, J.C.; Staquet, P.; Vaerman, J.P.; Sibille, Y. IL-9 inhibits oxidative burst and TNF-alpha release in lipopolysaccharide-stimulated human monocytes through TGF-beta. J. Immunol. 2002, 168, 4103–4111. [Google Scholar] [CrossRef]
  118. Donninelli, G.; Saraf-Sinik, I.; Mazziotti, V.; Capone, A.; Grasso, M.G.; Battistini, L.; Reynolds, R.; Magliozzi, R.; Volpe, E. Interleukin-9 regulates macrophage activation in the progressive multiple sclerosis brain. J. Neuroinflamm. 2020, 17, 149. [Google Scholar] [CrossRef]
  119. Ruocco, G.; Rossi, S.; Motta, C.; Macchiarulo, G.; Barbieri, F.; De Bardi, M.; Borsellino, G.; Finardi, A.; Grasso, M.G.; Ruggieri, S.; et al. T helper 9 cells induced by plasmacytoid dendritic cells regulate interleukin-17 in multiple sclerosis. Clin. Sci. 2015, 129, 291–303. [Google Scholar] [CrossRef]
  120. Elyaman, W.; Bradshaw, E.M.; Uyttenhove, C.; Dardalhon, V.; Awasthi, A.; Imitola, J.; Bettelli, E.; Oukka, M.; van Snick, J.; Renauld, J.C.; et al. IL-9 induces differentiation of TH17 cells and enhances function of FoxP3+ natural regulatory T cells. Proc. Natl. Acad. Sci. USA 2009, 106, 12885–12890. [Google Scholar] [CrossRef]
  121. Guadalupi, L.; Vanni, V.; Balletta, S.; Caioli, S.; De Vito, F.; Fresegna, D.; Sanna, K.; Nencini, M.; Donninelli, G.; Volpe, E.; et al. Interleukin-9 protects from microglia- and TNF-mediated synaptotoxicity in experimental multiple sclerosis. J. Neuroinflamm. 2024, 21, 128. [Google Scholar] [CrossRef] [PubMed]
  122. Li, H.; Nourbakhsh, B.; Ciric, B.; Zhang, G.X.; Rostami, A. Neutralization of IL-9 ameliorates experimental autoimmune encephalomyelitis by decreasing the effector T cell population. J. Immunol. 2010, 185, 4095–4100. [Google Scholar] [CrossRef] [PubMed]
  123. Nowak, E.C.; Weaver, C.T.; Turner, H.; Begum-Haque, S.; Becher, B.; Schreiner, B.; Coyle, A.J.; Kasper, L.H.; Noelle, R.J. IL-9 as a mediator of Th17-driven inflammatory disease. J. Exp. Med. 2009, 206, 1653–1660. [Google Scholar] [CrossRef]
  124. Collison, L.W.; Workman, C.J.; Kuo, T.T.; Boyd, K.; Wang, Y.; Vignali, K.M.; Cross, R.; Sehy, D.; Blumberg, R.S.; Vignali, D.A. The inhibitory cytokine IL-35 contributes to regulatory T-cell function. Nature 2007, 450, 566–569. [Google Scholar] [CrossRef]
  125. Shen, P.; Roch, T.; Lampropoulou, V.; O’Connor, R.A.; Stervbo, U.; Hilgenberg, E.; Ries, S.; Dang, V.D.; Jaimes, Y.; Daridon, C.; et al. IL-35-producing B cells are critical regulators of immunity during autoimmune and infectious diseases. Nature 2014, 507, 366–370. [Google Scholar] [CrossRef]
  126. Wang, R.X.; Yu, C.R.; Dambuza, I.M.; Mahdi, R.M.; Dolinska, M.B.; Sergeev, Y.V.; Wingfield, P.T.; Kim, S.H.; Egwuagu, C.E. Interleukin-35 induces regulatory B cells that suppress autoimmune disease. Nat. Med. 2014, 20, 633–641. [Google Scholar] [CrossRef]
  127. Dixon, K.O.; van der Kooij, S.W.; Vignali, D.A.; van Kooten, C. Human tolerogenic dendritic cells produce IL-35 in the absence of other IL-12 family members. Eur. J. Immunol. 2015, 45, 1736–1747. [Google Scholar] [CrossRef] [PubMed]
  128. Xiao, T.; Qu, H.; Zeng, Z.; Li, C.; Wan, J. Interleukin-35 from Interleukin-4-Stimulated Macrophages Alleviates Oxygen Glucose Deprivation/Re-oxygenation-Induced Neuronal Cell Death via the Wnt/beta-Catenin Signaling Pathway. Neurotox. Res. 2022, 40, 420–431. [Google Scholar] [CrossRef]
  129. Huang, A.; Cheng, L.; He, M.; Nie, J.; Wang, J.; Jiang, K. Interleukin-35 on B cell and T cell induction and regulation. J. Inflamm. 2017, 14, 16. [Google Scholar] [CrossRef]
  130. Liu, G.; Li, M.; Qian, S.; Yu, L.; Qian, L.; Feng, X. Interleukin-35 exhibits protective effects in a rat model of hypoxic-ischemic encephalopathy through the inhibition of microglia-mediated inflammation. Transl. Pediatr. 2022, 11, 651–662. [Google Scholar] [CrossRef]
  131. Moseley, T.A.; Haudenschild, D.R.; Rose, L.; Reddi, A.H. Interleukin-17 family and IL-17 receptors. Cytokine Growth Factor Rev. 2003, 14, 155–174. [Google Scholar] [CrossRef] [PubMed]
  132. Rickel, E.A.; Siegel, L.A.; Yoon, B.R.; Rottman, J.B.; Kugler, D.G.; Swart, D.A.; Anders, P.M.; Tocker, J.E.; Comeau, M.R.; Budelsky, A.L. Identification of functional roles for both IL-17RB and IL-17RA in mediating IL-25-induced activities. J. Immunol. 2008, 181, 4299–4310. [Google Scholar] [CrossRef] [PubMed]
  133. Angkasekwinai, P.; Park, H.; Wang, Y.H.; Wang, Y.H.; Chang, S.H.; Corry, D.B.; Liu, Y.J.; Zhu, Z.; Dong, C. Interleukin 25 promotes the initiation of proallergic type 2 responses. J. Exp. Med. 2007, 204, 1509–1517. [Google Scholar] [CrossRef] [PubMed]
  134. Fort, M.M.; Cheung, J.; Yen, D.; Li, J.; Zurawski, S.M.; Lo, S.; Menon, S.; Clifford, T.; Hunte, B.; Lesley, R.; et al. IL-25 induces IL-4, IL-5, and IL-13 and Th2-associated pathologies in vivo. Immunity 2001, 15, 985–995. [Google Scholar] [CrossRef]
  135. Ikeda, K.; Nakajima, H.; Suzuki, K.; Kagami, S.; Hirose, K.; Suto, A.; Saito, Y.; Iwamoto, I. Mast cells produce interleukin-25 upon Fc epsilon RI-mediated activation. Blood 2003, 101, 3594–3596. [Google Scholar] [CrossRef]
  136. Kang, C.M.; Jang, A.S.; Ahn, M.H.; Shin, J.A.; Kim, J.H.; Choi, Y.S.; Rhim, T.Y.; Park, C.S. Interleukin-25 and interleukin-13 production by alveolar macrophages in response to particles. Am. J. Respir. Cell Mol. Biol. 2005, 33, 290–296. [Google Scholar] [CrossRef]
  137. Wang, Y.H.; Angkasekwinai, P.; Lu, N.; Voo, K.S.; Arima, K.; Hanabuchi, S.; Hippe, A.; Corrigan, C.J.; Dong, C.; Homey, B.; et al. IL-25 augments type 2 immune responses by enhancing the expansion and functions of TSLP-DC-activated Th2 memory cells. J. Exp. Med. 2007, 204, 1837–1847. [Google Scholar] [CrossRef]
  138. Tamachi, T.; Maezawa, Y.; Ikeda, K.; Kagami, S.; Hatano, M.; Seto, Y.; Suto, A.; Suzuki, K.; Watanabe, N.; Saito, Y.; et al. IL-25 enhances allergic airway inflammation by amplifying a TH2 cell-dependent pathway in mice. J. Allergy Clin. Immunol. 2006, 118, 606–614. [Google Scholar] [CrossRef]
  139. Kleinschek, M.A.; Owyang, A.M.; Joyce-Shaikh, B.; Langrish, C.L.; Chen, Y.; Gorman, D.M.; Blumenschein, W.M.; McClanahan, T.; Brombacher, F.; Hurst, S.D.; et al. IL-25 regulates Th17 function in autoimmune inflammation. J. Exp. Med. 2007, 204, 161–170. [Google Scholar] [CrossRef]
  140. Caruso, R.; Stolfi, C.; Sarra, M.; Rizzo, A.; Fantini, M.C.; Pallone, F.; MacDonald, T.T.; Monteleone, G. Inhibition of monocyte-derived inflammatory cytokines by IL-25 occurs via p38 Map kinase-dependent induction of Socs-3. Blood 2009, 113, 3512–3519. [Google Scholar] [CrossRef]
  141. Cao, Q.; Wang, C.; Zheng, D.; Wang, Y.; Lee, V.W.; Wang, Y.M.; Zheng, G.; Tan, T.K.; Yu, D.; Alexander, S.I.; et al. IL-25 induces M2 macrophages and reduces renal injury in proteinuric kidney disease. J. Am. Soc. Nephrol. 2011, 22, 1229–1239. [Google Scholar] [CrossRef] [PubMed]
  142. Su, Z.; Tao, X. Current Understanding of IL-37 in Human Health and Disease. Front. Immunol. 2021, 12, 696605. [Google Scholar] [CrossRef] [PubMed]
  143. Shuai, X.; Wei-min, L.; Tong, Y.L.; Dong, N.; Sheng, Z.Y.; Yao, Y.M. Expression of IL-37 contributes to the immunosuppressive property of human CD4+CD25+ regulatory T cells. Sci. Rep. 2015, 5, 14478. [Google Scholar] [CrossRef]
  144. Rudloff, I.; Cho, S.X.; Lao, J.C.; Ngo, D.; McKenzie, M.; Nold-Petry, C.A.; Nold, M.F. Monocytes and dendritic cells are the primary sources of interleukin 37 in human immune cells. J. Leukoc. Biol. 2017, 101, 901–911. [Google Scholar] [CrossRef]
  145. Nold, M.F.; Nold-Petry, C.A.; Zepp, J.A.; Palmer, B.E.; Bufler, P.; Dinarello, C.A. IL-37 is a fundamental inhibitor of innate immunity. Nat. Immunol. 2010, 11, 1014–1022. [Google Scholar] [CrossRef] [PubMed]
  146. Zhang, S.R.; Nold, M.F.; Tang, S.C.; Bui, C.B.; Nold, C.A.; Arumugam, T.V.; Drummond, G.R.; Sobey, C.G.; Kim, H.A. IL-37 increases in patients after ischemic stroke and protects from inflammatory brain injury, motor impairment and lung infection in mice. Sci. Rep. 2019, 9, 6922. [Google Scholar] [CrossRef]
  147. Sanchez-Fernandez, A.; Zandee, S.; Amo-Aparicio, J.; Charabati, M.; Prat, A.; Garlanda, C.; Eisenmesser, E.Z.; Dinarello, C.A.; Lopez-Vales, R. IL-37 exerts therapeutic effects in experimental autoimmune encephalomyelitis through the receptor complex IL-1R5/IL-1R8. Theranostics 2021, 11, 1–13. [Google Scholar] [CrossRef]
  148. Yazdani, R.; Naziri, H.; Azizi, G.; Ciric, B.; Askari, M.; Ahmadi, A.M.; Aseervatham, J.; Zhang, G.X.; Rostami, A. IL-37 suppresses CNS autoimmunity by increasing the frequency of Treg cells and reducing CD4 + T cell-derived IL-10 production. J. Neuroinflamm. 2024, 21, 301. [Google Scholar] [CrossRef]
  149. Pflanz, S.; Timans, J.C.; Cheung, J.; Rosales, R.; Kanzler, H.; Gilbert, J.; Hibbert, L.; Churakova, T.; Travis, M.; Vaisberg, E.; et al. IL-27, a heterodimeric cytokine composed of EBI3 and p28 protein, induces proliferation of naive CD4+ T cells. Immunity 2002, 16, 779–790. [Google Scholar] [CrossRef]
  150. Pflanz, S.; Hibbert, L.; Mattson, J.; Rosales, R.; Vaisberg, E.; Bazan, J.F.; Phillips, J.H.; McClanahan, T.K.; de Waal Malefyt, R.; Kastelein, R.A. WSX-1 and glycoprotein 130 constitute a signal-transducing receptor for IL-27. J. Immunol. 2004, 172, 2225–2231. [Google Scholar] [CrossRef]
  151. Hibbert, L.; Pflanz, S.; De Waal Malefyt, R.; Kastelein, R.A. IL-27 and IFN-alpha signal via Stat1 and Stat3 and induce T-Bet and IL-12Rbeta2 in naive T cells. J. Interferon Cytokine Res. 2003, 23, 513–522. [Google Scholar] [CrossRef]
  152. Carl, J.W.; Bai, X.F. IL27: Its roles in the induction and inhibition of inflammation. Int. J. Clin. Exp. Pathol. 2008, 1, 117–123. [Google Scholar]
  153. Yoshimura, T.; Takeda, A.; Hamano, S.; Miyazaki, Y.; Kinjyo, I.; Ishibashi, T.; Yoshimura, A.; Yoshida, H. Two-sided roles of IL-27: Induction of Th1 differentiation on naive CD4+ T cells versus suppression of proinflammatory cytokine production including IL-23-induced IL-17 on activated CD4+ T cells partially through STAT3-dependent mechanism. J. Immunol. 2006, 177, 5377–5385. [Google Scholar] [CrossRef]
  154. Batten, M.; Li, J.; Yi, S.; Kljavin, N.M.; Danilenko, D.M.; Lucas, S.; Lee, J.; de Sauvage, F.J.; Ghilardi, N. Interleukin 27 limits autoimmune encephalomyelitis by suppressing the development of interleukin 17-producing T cells. Nat. Immunol. 2006, 7, 929–936. [Google Scholar] [CrossRef]
  155. Stumhofer, J.S.; Laurence, A.; Wilson, E.H.; Huang, E.; Tato, C.M.; Johnson, L.M.; Villarino, A.V.; Huang, Q.; Yoshimura, A.; Sehy, D.; et al. Interleukin 27 negatively regulates the development of interleukin 17-producing T helper cells during chronic inflammation of the central nervous system. Nat. Immunol. 2006, 7, 937–945. [Google Scholar] [CrossRef]
  156. Fitzgerald, D.C.; Zhang, G.X.; El-Behi, M.; Fonseca-Kelly, Z.; Li, H.; Yu, S.; Saris, C.J.; Gran, B.; Ciric, B.; Rostami, A. Suppression of autoimmune inflammation of the central nervous system by interleukin 10 secreted by interleukin 27-stimulated T cells. Nat. Immunol. 2007, 8, 1372–1379. [Google Scholar] [CrossRef]
  157. Aldridge, D.L.; Moodley, D.; Park, J.; Phan, A.T.; Rausch, M.; White, K.F.; Ren, Y.; Golin, K.; Radaelli, E.; Kedl, R.; et al. Endogenous IL-27 during toxoplasmosis limits early monocyte responses and their inflammatory activation by pathological T cells. mBio 2024, 15, e0008324. [Google Scholar] [CrossRef]
  158. Wu, X.Z.; Shi, X.Y.; Zhai, K.; Yi, F.S.; Wang, Z.; Wang, W.; Pei, X.B.; Xu, L.L.; Wang, Z.; Shi, H.Z. Activated naive B cells promote development of malignant pleural effusion by differential regulation of T(H)1 and T(H)17 response. Am. J. Physiol. Lung Cell Mol. Physiol. 2018, 315, L443–L455. [Google Scholar] [CrossRef]
  159. Lin, C.H.; Wu, C.J.; Cho, S.; Patkar, R.; Huth, W.J.; Lin, L.L.; Chen, M.C.; Israelsson, E.; Betts, J.; Niedzielska, M.; et al. Selective IL-27 production by intestinal regulatory T cells permits gut-specific regulation of T(H)17 cell immunity. Nat. Immunol. 2023, 24, 2108–2120. [Google Scholar] [CrossRef]
  160. Choi, J.K.; Yu, C.R.; Bing, S.J.; Jittayasothorn, Y.; Mattapallil, M.J.; Kang, M.; Park, S.B.; Lee, H.S.; Dong, L.; Shi, G.; et al. IL-27-producing B-1a cells suppress neuroinflammation and CNS autoimmune diseases. Proc. Natl. Acad. Sci. USA 2021, 118, e2109548118. [Google Scholar] [CrossRef]
  161. Senecal, V.; Deblois, G.; Beauseigle, D.; Schneider, R.; Brandenburg, J.; Newcombe, J.; Moore, C.S.; Prat, A.; Antel, J.; Arbour, N. Production of IL-27 in multiple sclerosis lesions by astrocytes and myeloid cells: Modulation of local immune responses. Glia 2016, 64, 553–569. [Google Scholar] [CrossRef]
  162. Lalive, P.H.; Kreutzfeldt, M.; Devergne, O.; Metz, I.; Bruck, W.; Merkler, D.; Pot, C. Increased interleukin-27 cytokine expression in the central nervous system of multiple sclerosis patients. J. Neuroinflamm. 2017, 14, 144. [Google Scholar] [CrossRef]
  163. Fitzgerald, D.C.; Ciric, B.; Touil, T.; Harle, H.; Grammatikopolou, J.; Das Sarma, J.; Gran, B.; Zhang, G.X.; Rostami, A. Suppressive effect of IL-27 on encephalitogenic Th17 cells and the effector phase of experimental autoimmune encephalomyelitis. J. Immunol. 2007, 179, 3268–3275. [Google Scholar] [CrossRef]
  164. Mertens, M.; Singh, J.A. Anakinra for rheumatoid arthritis: A systematic review. J. Rheumatol. 2009, 36, 1118–1125. [Google Scholar] [CrossRef]
  165. Maier, A.; Deigendesch, N.; Muller, K.; Weishaupt, J.H.; Krannich, A.; Rohle, R.; Meissner, F.; Molawi, K.; Munch, C.; Holm, T.; et al. Interleukin-1 Antagonist Anakinra in Amyotrophic Lateral Sclerosis—A Pilot Study. PLoS ONE 2015, 10, e0139684. [Google Scholar] [CrossRef]
  166. Trias, E.; Ibarburu, S.; Barreto-Nunez, R.; Babdor, J.; Maciel, T.T.; Guillo, M.; Gros, L.; Dubreuil, P.; Diaz-Amarilla, P.; Cassina, P.; et al. Post-paralysis tyrosine kinase inhibition with masitinib abrogates neuroinflammation and slows disease progression in inherited amyotrophic lateral sclerosis. J. Neuroinflamm. 2016, 13, 177. [Google Scholar] [CrossRef]
  167. Mora, J.S.; Genge, A.; Chio, A.; Estol, C.J.; Chaverri, D.; Hernandez, M.; Marin, S.; Mascias, J.; Rodriguez, G.E.; Povedano, M.; et al. Masitinib as an add-on therapy to riluzole in patients with amyotrophic lateral sclerosis: A randomized clinical trial. Amyotroph. Lateral Scler. Frontotemporal Degener. 2020, 21, 5–14. [Google Scholar] [CrossRef]
  168. Mizwicki, M.T.; Fiala, M.; Magpantay, L.; Aziz, N.; Sayre, J.; Liu, G.; Siani, A.; Chan, D.; Martinez-Maza, O.; Chattopadhyay, M.; et al. Tocilizumab attenuates inflammation in ALS patients through inhibition of IL6 receptor signaling. Am. J. Neurodegener. Dis. 2012, 1, 305–315. [Google Scholar]
  169. Babu, S.; Hightower, B.G.; Chan, J.; Zurcher, N.R.; Kivisakk, P.; Tseng, C.J.; Sanders, D.L.; Robichaud, A.; Banno, H.; Evora, A.; et al. Ibudilast (MN-166) in amyotrophic lateral sclerosis- an open label, safety and pharmacodynamic trial. Neuroimage Clin. 2021, 30, 102672. [Google Scholar] [CrossRef]
  170. Thonhoff, J.R.; Beers, D.R.; Zhao, W.; Pleitez, M.; Simpson, E.P.; Berry, J.D.; Cudkowicz, M.E.; Appel, S.H. Expanded autologous regulatory T-lymphocyte infusions in ALS: A phase I, first-in-human study. Neurol. Neuroimmunol. Neuroinflamm. 2018, 5, e465. [Google Scholar] [CrossRef]
  171. Malek, T.R.; Bayer, A.L. Tolerance, not immunity, crucially depends on IL-2. Nat. Rev. Immunol. 2004, 4, 665–674. [Google Scholar] [CrossRef]
  172. Zorn, E.; Nelson, E.A.; Mohseni, M.; Porcheray, F.; Kim, H.; Litsa, D.; Bellucci, R.; Raderschall, E.; Canning, C.; Soiffer, R.J.; et al. IL-2 regulates FOXP3 expression in human CD4+CD25+ regulatory T cells through a STAT-dependent mechanism and induces the expansion of these cells in vivo. Blood 2006, 108, 1571–1579. [Google Scholar] [CrossRef]
  173. Tang, Q.; Adams, J.Y.; Penaranda, C.; Melli, K.; Piaggio, E.; Sgouroudis, E.; Piccirillo, C.A.; Salomon, B.L.; Bluestone, J.A. Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity 2008, 28, 687–697. [Google Scholar] [CrossRef]
  174. Camu, W.; Mickunas, M.; Veyrune, J.L.; Payan, C.; Garlanda, C.; Locati, M.; Juntas-Morales, R.; Pageot, N.; Malaspina, A.; Andreasson, U.; et al. Repeated 5-day cycles of low dose aldesleukin in amyotrophic lateral sclerosis (IMODALS): A phase 2a randomised, double-blind, placebo-controlled trial. EBioMedicine 2020, 59, 102844. [Google Scholar] [CrossRef]
  175. Giovannelli, I.; Bayatti, N.; Brown, A.; Wang, D.; Mickunas, M.; Camu, W.; Veyrune, J.L.; Payan, C.; Garlanda, C.; Locati, M.; et al. Amyotrophic lateral sclerosis transcriptomics reveals immunological effects of low-dose interleukin-2. Brain Commun. 2021, 3, fcab141. [Google Scholar] [CrossRef]
  176. Alarcan, H.; Bruno, C.; Emond, P.; Raoul, C.; Vourc’h, P.; Corcia, P.; Camu, W.; Veyrune, J.L.; Garlanda, C.; Locati, M.; et al. Pharmacometabolomics applied to low-dose interleukin-2 treatment in amyotrophic lateral sclerosis. Ann. N. Y. Acad. Sci. 2024, 1536, 82–91. [Google Scholar] [CrossRef]
  177. Battaglia, M.; Stabilini, A.; Roncarolo, M.G. Rapamycin selectively expands CD4+CD25+FoxP3+ regulatory T cells. Blood 2005, 105, 4743–4748. [Google Scholar] [CrossRef]
  178. Mandrioli, J.; D’Amico, R.; Zucchi, E.; Gessani, A.; Fini, N.; Fasano, A.; Caponnetto, C.; Chio, A.; Dalla Bella, E.; Lunetta, C.; et al. Rapamycin treatment for amyotrophic lateral sclerosis: Protocol for a phase II randomized, double-blind, placebo-controlled, multicenter, clinical trial (RAP-ALS trial). Med. Baltim. 2018, 97, e11119. [Google Scholar] [CrossRef]
  179. Mandrioli, J.; D’Amico, R.; Zucchi, E.; De Biasi, S.; Banchelli, F.; Martinelli, I.; Simonini, C.; Lo Tartaro, D.; Vicini, R.; Fini, N.; et al. Randomized, double-blind, placebo-controlled trial of rapamycin in amyotrophic lateral sclerosis. Nat. Commun. 2023, 14, 4970. [Google Scholar] [CrossRef]
Figure 1. Cell-autonomous and non-cell-autonomous mechanisms of motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Microenvironment and genetic factors might trigger motor neuron intrinsic changes (cell-autonomous mechanism) and several other cell types across the central nervous system (CNS) (non-cell autonomous mechanism). Among non-cell autonomous mechanisms, the most important is the turning of astrocytes and microglia into a pro-inflammatory state. The activation of glia cells in ALS leads to production of neurotoxic factors, which contribute to neurodegeneration, and of pro-inflammatory cytokines involved in the drastic loss of blood-brain barrier (BBB) integrity, which causes the invasion of leukocytes from blood into the CNS parenchyma. The entry of inflammatory monocytes, T lymphocytes, and B lymphocytes into the CNS enhances neuroinflammation by releasing inflammatory cytokines and reactive oxygen species to the tissue. Long and chronic neuroinflammation is linked to impaired tissue function and neurodegeneration, which further amplify inflammatory mechanisms in a pathological feedback loop.
Figure 1. Cell-autonomous and non-cell-autonomous mechanisms of motor neuron degeneration in amyotrophic lateral sclerosis (ALS). Microenvironment and genetic factors might trigger motor neuron intrinsic changes (cell-autonomous mechanism) and several other cell types across the central nervous system (CNS) (non-cell autonomous mechanism). Among non-cell autonomous mechanisms, the most important is the turning of astrocytes and microglia into a pro-inflammatory state. The activation of glia cells in ALS leads to production of neurotoxic factors, which contribute to neurodegeneration, and of pro-inflammatory cytokines involved in the drastic loss of blood-brain barrier (BBB) integrity, which causes the invasion of leukocytes from blood into the CNS parenchyma. The entry of inflammatory monocytes, T lymphocytes, and B lymphocytes into the CNS enhances neuroinflammation by releasing inflammatory cytokines and reactive oxygen species to the tissue. Long and chronic neuroinflammation is linked to impaired tissue function and neurodegeneration, which further amplify inflammatory mechanisms in a pathological feedback loop.
Ijms 26 03854 g001
Figure 2. Anti-inflammatory response counteracts the neuroinflammation. In neurodegenerative diseases, the blood-brain barrier (BBB) integrity is impaired, and invasion of leukocytes from blood into CNS parenchyma leads to neuroinflammation. However, immune cells with anti-inflammatory properties might infiltrate the CNS and produce specific anti-inflammatory cytokines contrasting neuroinflammation, and consequently neurodegeneration. In particular, the cytokines involved in this process are IL-10, TGF-β, IL-4, IL-13, IL-9, IL-35, IL-25, IL-37, IL-27, and the immune cells involved are Treg, Breg, microglia M2, macrophage M2, Th2, and Th9 cells. Cytokines may exert either driving than effector functions: IL-4 induces Th2 cells; IL-4 and TGF-β drive Th9 polarization; IL-2, TGF-β, IL-37, and IL-10 are involved in Treg expansion; IL-10 drives Breg; IL-4, IL-10, IL-13, IL-9, IL-25, IL-37, IL-27, and TGF-β polarize microglia and macrophages towards the M2 anti-inflammatory profile.
Figure 2. Anti-inflammatory response counteracts the neuroinflammation. In neurodegenerative diseases, the blood-brain barrier (BBB) integrity is impaired, and invasion of leukocytes from blood into CNS parenchyma leads to neuroinflammation. However, immune cells with anti-inflammatory properties might infiltrate the CNS and produce specific anti-inflammatory cytokines contrasting neuroinflammation, and consequently neurodegeneration. In particular, the cytokines involved in this process are IL-10, TGF-β, IL-4, IL-13, IL-9, IL-35, IL-25, IL-37, IL-27, and the immune cells involved are Treg, Breg, microglia M2, macrophage M2, Th2, and Th9 cells. Cytokines may exert either driving than effector functions: IL-4 induces Th2 cells; IL-4 and TGF-β drive Th9 polarization; IL-2, TGF-β, IL-37, and IL-10 are involved in Treg expansion; IL-10 drives Breg; IL-4, IL-10, IL-13, IL-9, IL-25, IL-37, IL-27, and TGF-β polarize microglia and macrophages towards the M2 anti-inflammatory profile.
Ijms 26 03854 g002
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

Stacchiotti, C.; Mazzella di Regnella, S.; Cinotti, M.; Spalloni, A.; Volpe, E. Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies. Int. J. Mol. Sci. 2025, 26, 3854. https://doi.org/10.3390/ijms26083854

AMA Style

Stacchiotti C, Mazzella di Regnella S, Cinotti M, Spalloni A, Volpe E. Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies. International Journal of Molecular Sciences. 2025; 26(8):3854. https://doi.org/10.3390/ijms26083854

Chicago/Turabian Style

Stacchiotti, Costanza, Simona Mazzella di Regnella, Miriam Cinotti, Alida Spalloni, and Elisabetta Volpe. 2025. "Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies" International Journal of Molecular Sciences 26, no. 8: 3854. https://doi.org/10.3390/ijms26083854

APA Style

Stacchiotti, C., Mazzella di Regnella, S., Cinotti, M., Spalloni, A., & Volpe, E. (2025). Neuroinflammation and Amyotrophic Lateral Sclerosis: Recent Advances in Anti-Inflammatory Cytokines as Therapeutic Strategies. International Journal of Molecular Sciences, 26(8), 3854. https://doi.org/10.3390/ijms26083854

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