Open access peer-reviewed chapter

Cellular Cytotoxicity and Multiple Sclerosis

Written By

Annie M.L. Willson and Margaret A. Jordan

Submitted: 09 May 2022 Reviewed: 02 June 2022 Published: 29 June 2022

DOI: 10.5772/intechopen.105681

From the Edited Volume

Cytotoxicity - Understanding Cellular Damage and Response

Edited by Anil Sukumaran and Mahmoud Ahmed Mansour

Chapter metrics overview

106 Chapter Downloads

View Full Metrics

Abstract

Multiple sclerosis (MS) is an autoimmune disease in which discrete central nervous system lesions result from perivascular immune cell infiltration associated with damage to myelin (demyelination), oligodendrocytes and neurons. This culminates in debilitating neurological symptoms, primarily affecting women in their child-bearing years. Both the innate and adaptive branches of the immune system have been implicated in disease initiation and progression, and although the underlying cause remains elusive, there is compelling evidence for a complex interaction between genetic and environmental factors, leading to inflammation and neurodegeneration. Both direct cellular toxicity and antibody-dependent cellular cytotoxicity (ADCC) involving several cell types have been identified in playing major roles. These cells and their interactions in the pathogenesis of MS will be discussed.

Keywords

  • multiple sclerosis
  • cellular cytotoxicity
  • cell subsets
  • human
  • MS mouse models

1. Introduction

Multiple sclerosis (MS) is the most prevalent neurological disease among young adults in developed countries, with approximately 2.8 million people being affected worldwide [1]. It principally affects women in their prime, with diagnosis typically occurring between the ages of 20 and 40. The disease is debilitating due to central nervous system (CNS) damage resulting from activated lymphocytes migrating across the blood brain barrier (BBB) and engaging in a proinflammatory response. This causes cells to attack and destroy the myelin sheaths that coat the axons of neurons of the brain, spinal cord and optic nerve, as well as the myelinating cells or oligodendrocytes, and the axons themselves [2]. As neurons receive sensory input from external sources and send motor commands to the muscles by relaying interneuron electrical impulses, breakdown causes interruption to the signals being sent around the body, and dependent on where the damage occurs, results in different signs and symptoms. These can include vision impairment, muscle spasms and numbness, bladder and bowel issues, fatigue and difficulty walking [3]. Most people with MS have progressive neurological disability which, though not usual, can culminate in death [4]. The area of damage or scarring caused by the immune system attack is called a lesion or plaque, and can be visualised by magnetic resonance imaging (MRI). A definitive diagnosis of MS is made when these plaques are shown to be reoccurring and when there is the clear presence of clinical symptoms [3].

Two major types of MS have been recognised, primary progressive multiple sclerosis (PPMS), diagnosed in approximately 15% of patients and which results in steady progression of disease from onset, and relapsing remitting multiple sclerosis (RRMS), which affects approximately 80% of patients and is characterised by periods of relapse separated by periods of remit without worsening of symptoms [5, 6, 7]. Most patients with an initial diagnosis of RRMS will, within 20 years of diagnosis, progress to secondary progressive multiple sclerosis (SPMS) where the stages between relapse and remit shorten and there is a steady decline with an increase in symptoms and disease progression [8]. Up to approximately 5% of MS patients have progressive relapsing multiple sclerosis (PRMS) and this characterised by steady disease progression with occasional relapses [9].

The exact cause of MS is still unknown, however research has determined that it is an autoimmune disease, arising from complex interactions between environmental and genetic influences. There is a latitude incidence variance, with prevalence of MS increased the further one is from the equator; sunlight and vitamin D are therefore being investigated as disease triggers [1, 10, 11, 12]. Childhood exposure to bacteria and viruses have also been investigated, due to a person’s disease risk being set as the incidence of the region they moved to prior to puberty [13, 14]. Of note, every patient with MS have previously been exposed to Epstein–Barr virus (EBV) [15, 16]. Smoking also increases a person’s risk and worsens symptoms following diagnosis [17].

Although the disease is not inherited, it has a genetic component, with those having an affected first degree relative exhibiting an increased incidence of disease [18], and twin studies indicate that there is a 30% chance of developing disease in the second twin if the first has been diagnosed with MS [19]. Genome wide association studies (GWAS) have identified more than 230 genes associated with a person’s MS risk, several being immune genes, particularly those of T cells, B cells, natural killer (NK) cells, monocytes and microglia, implicating involvement of both major branches of the immune system, the innate and adaptive immune responses, in initiation and progression of disease [20, 21, 22, 23]. These studies are supported by several human and animal model functional studies [24, 25, 26].

Cellular toxicity, or the ability to kill other cells, is an important effector mechanisms of the immune system to protect us from infections, cancer or autoimmune diseases. There is a close association between inflammation and neurodegeneration, and cellular toxicity has been implicated as a having a major role in MS [27]. The main players are CD8, or cytotoxic, T cells and NK cells. Cellular toxicity can operate by many mechanisms including NK cell release of lytic granules containing perforin or granzymes to kill directly, or by inducing death receptor-mediated apoptosis via tumour necrosis factor (ligand) superfamily member 10 (TRAIL) or Fas Ligand (FasL) expression on CD8 T cells [28]. There are also antibody-dependent cell-mediated cytotoxic mechanisms (ADCC), where B cells produce antigen specific antibodies or immunoglobulins, that will coat a pathogen or foreign body, marking them for killing or destruction through cell to cell cytoloysis by effector immune cells expressing FcγRIIIA (CD16A), including classical NK cells, monocytes/macrophages, neutrophils, eosinophils, NKT cells, or γδT cells (reviewed in [29]).

Advertisement

2. Innate and adaptive immunity

The immune system of vertebrates is commonly divided into two main complementary parts, innate and adaptive immunity, the bridge between which is critical for an efficient and effective immune response.

The innate immune system is evolutionary the most primitive, where there is non-specific response to a broad class of antigens. The haematopoietic cells involved include macrophages, dendritic cells, mast cells, neutrophils, eosinophils, NK cells and NKT cells. Although 1908 Nobel Prize winner, Elie Metchnikoff, first described an important role for the innate immune system [30], it is only now being recognised as a critical regulator of human inflammatory disease. Innate immunity involves the recognition of infected cells through surface recognition receptors. These are termed pattern recognition receptors (PRRs) which recognise pathogen associated molecular patterns (PAMPs) unique to non-vertebrate cells, including bacteria and fungi. They are also on internal vesicle membranes for recognition of viral ssRNA and dsRNA and for distinguishing lysed bacterial components [31]. Cytotoxic innate lymphocytes can lyse abnormal or infected cells through the release of cytotoxic granules containing perforin or granzymes, and antigen presenting cells (APCs) can be activated by the innate immune system to present pathogen antigens on their surface. Once activated they will migrate to secondary lymph organs to present their antigen to T cells, and in so doing also activate the adaptive immune system response [32, 33]. The innate immune system therefore functions through a combination of cellular defences and humoral components to defend against nonspecific antigens before activating B and T cells, triggering an adaptive immune response. Speed is the main advantage of innate immunity, with a protective inflammatory response being generated within minutes of pathogen exposure.

Another part of innate immunity is the complement system, which is made up of several small proteins that have been synthesised in the liver and circulate in the blood as active precursors that when stimulated are proteolytically cleaved to release cytokines, leading to a cascade of reactions, ultimately resulting in complement activation or fixation [34]. As the name suggests, they complement or enhance the ability of antibodies and phagocytic cells to clear damaged or diseased cells by promoting inflammation and attack of the cell membrane of the pathogen. Antibodies, generated by the adaptive immune system, can activate the complement system.

Adaptive immunity, sometimes referred to as acquired immunity, is highly specialised and helps to protect the body by recognising antigens, whether they are foreign to the host’s immune system (exogenous), produced by intracellular bacteria or viruses (intracellular) or produced by the host (autoantigen). The adaptive immune system also remembers previously encountered antigens, leading to quicker response times [35]. T and B lymphocytes are the main cells mediating adaptive immunity, with T cells being further divided into the cytotoxic CD8 T cells and CD4 T cells that constitute several classes of what are commonly referred to as “helper T cells”. These cell have produced highly specific receptors for recognition of hundreds or even thousands of antigens through genetic recombination, and this facilitates pathogen specific immunologic effectors pathways, the generation of immunological memory and the regulation of host immune homoeostasis [36].

CD8 T cells recognise infected cells through interaction of T cell receptors with antigens presented by major histocompatibility complex (MHC) class I on the infected cell. The target cell is then killed by the release of cytotoxins, such as perforin and granzymes, from the CD8 T cell [28]. CD4 T cells, on the other hand, recognise antigens presented in the context of MHC II on an APC. Binding to MHC II molecules activates CD4 T cells to release cytokines, which can stimulate CD8 T cells, macrophages and B cells to form an immune response (reviewed in [37]). They can, for example, release cytokines as instructors to CD8 T cells to release cytotoxins, or to B cells to produce pathogen specific antibodies. They therefore instigate and shape adaptive immune responses dependent on the cytokines they release. These can be mainly Th1, or inflammatory, in nature, such as IFN-γ and IL-12, responsible for the control of intracellular pathogens, or polarised to a more anti-inflammatory Th2 response, where cytokines such as IL-4, IL-5 or IL-13 are produced [38, 39]. A disturbance in this Th1/Th2 response can have severe consequences, be they more Th2 in nature, driving asthma and allergy, or Th1 driven, resulting in autoimmune diseases, including MS (reviewed in [40]). A couple of the more recently identified CD4 T cells subsets include Th17 cells that are characterised by production of IL-17 and IL-23, and have been linked to inflammatory diseases, and T regulatory (Treg) cells, which are important in maintaining homeostasis and tolerance of the immune system [41, 42, 43]. Tregs express the transcription factor FoxP3 which is essential for their development and function [44, 45, 46]. In humans, mutations in FOXP3 have been found to result in immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, providing evidence that anomalies of Tregs can cause autoimmune disease and allergy [47].

During production of the T cell receptor (TCR) on T cells and B cell receptor (BCR) on B cells, random genetic recombination events can lead to receptors being produced that are specific to autoantigens [48, 49]. To prevent reaction to self, cells undergo central and peripheral tolerance events through which autoreactive cells are apoptotically removed, first in the primary lymphoid organs of the thymus (T cell) and bone marrow (B cell), and if this fails, in the secondary lymphoid organs after cells migrate to the periphery [49]. Self-reactive antibodies account for 55–75% of all antibodies expressed by early immature B cells, including polyreactive and anti-nuclear specificities [49]. However, it is estimated that the majority of newly produced B cells do not reach maturity, and during central and peripheral tolerance most of the self-reactive B cells are removed. If both selection processes fail in T or B cells, this will result in T and B cells able to react with the body’s own cells and tissues. These events lead to autoimmune disease.

Advertisement

3. Autoimmune disease

Inflammation as a response of the body to infection or cell injury is a well-known concept that dates back to the beginning of medicine. However, Metchnikoff pointed out that although normally a method of protection, inflammation that exceeds normal bounds can cause disease [27]. Even with this knowledge, it was not until the 1950s that inflammation was recognised as inducing an autoimmune reaction responsible for disease. Autoimmune disease is characterised by an excessive immune response against self, often resulting in inflammation and tissue destruction, in the absence of a threat to the organism [50, 51]. Aberrant immune responses have been associated with over 80 disorders, including multiple sclerosis, and affects 5–7% of the population [52]. Clinical observations over the past decade have suggested that the prevalence of all autoimmune disease, not just MS, is increasing, bringing the issue to the forefront of scientific interest [53, 54]. Successful treatment of autoimmune disease is also of great societal interest, as they are commonly characterised by chronic natures, ongoing health care costs, and debilitating issues resulting in loss of productivity.

Immunological self-tolerance is maintained in part by Tregs. Tregs are CD4 T cells that actively and dominantly supress lymphocytes, particularly self-reactive T cells in the normal periphery that exist despite the deletion mechanisms in the thymus [43]. Natural CD25+ CD4 Tregs utilise several modes of suppression, including cell contact dependent mechanisms, such as the killing of APCs or responder T cells by granzyme and perforin, and by mediation of soluble factors, such as the secretion of immunosuppressive cytokines like IL-10, TGF-β or IL-35, or deprivation of cytokines necessary for expansion and survival of responder T cells (reviewed in [55, 56]).

Optimal T cell function relies on a carefully maintained state of equilibrium. When one subpopulation of T helper cells is activated, others are modulated or inhibited to promote the most specific effector response to the threat [57]. The cellular development of Tregs shares a common cytokine with Th17 cells, TGF-β [41, 42]. Th17 cells are the opposing force to Tregs, serving as an effector lymphocyte population that plays a key role in autoimmunity [41, 42]. At homeostasis, Th17 cells promote gut barrier defence, granulopoiesis, granulocyte chemotaxis and immunity against extracellular pathogen [58]. IL-17 induces granulopoesis indirectly through the stimulation of fibroblasts, epithelial and endothelial cells to secrete GM-CSF, IL-6, IL-8 and MIP-2, with IL-8 and MIP-2 enhancing chemotaxis of neutrophils [59, 60]. While Th17 cell mediated immunity is crucial for maintaining mucosal and haematopoietic homeostasis, too strong a response can induce autoimmunity. The relationship between Tregs and effector Th17 must remain balanced to provide the optimal functional immunity and health of an organism.

Another theory of immune regulation is the hypothesis of homeostasis between Th1 and Th2 cells. The subpopulations can be distinguished by the cytokines they produce and the expression of difference cell surface molecules. Th1 cells are responsible for cell mediated immunity, phagocyte dependent protective responses, B cell activation and production of opsonising antibodies such as IgG1, whereas Th2 cells produce cytokines that are responsible for strong antibody production, eosinophil activation and inhibition of several macrophage functions, thus providing phagocyte independent protective responses [61]. Th2 cells are also responsible for the general activation of B cells. When the Th1/Th2 paradigm is thrown out of balance by failure of central or peripheral tolerance, immunological disorders can occur due to uncontrolled responses [61].

Advertisement

4. MS and the immune system

MS arises when there is an imbalance in the body’s immune response, shifting it from a beneficial immune process that fights infection and disease towards a self-aggressive immune attack on the cells within the CNS (Figure 1). Genetic and environmental factor interaction may facilitate movement of autoreactive T cells, macrophages and NK cells and demyelinating antibodies from the periphery to the CNS. In the periphery self-antigens can be presented on MHC II molecules by APCs to TCRs on T cells, thereby activating proinflammatory T cells [48]. The activated T cells can then migrate through the blood brain barrier to the brain and spinal cord [2]. Once in the CNS the T cells can be reactivated by CNS antigens presented on MHC II by other APCs, primarily microglial cells [62]. Secretion of proinflammatory Th1 cytokines by the reactivated T cells can induce CNS inflammation by activating macrophages, B cells and other T cells [63]. The antibodies can also initiate a complement cascade resulting in assembly of the membrane attack complex, forming pores in the myelin membranes.

Figure 1.

Multiple sclerosis pathogenesis. Autoreactive T cells, macrophages and NK cells, and demyelinating antibodies, may migrate across a compromised blood brain barrier. T cells are reactivated in the central nervous system by antigen presenting cells (APC). Anti-inflammatory cytokines released by Th2 cells can stimulate B cells to differentiate into plasma cells that secrete demyelinating antibodies. Alternatively, the release of proinflammatory cytokines by Th1 cells can enhance immune response, via activation of other immune cells such as CD8 T cells and macrophages to attack the myelin sheath and oligodendrocytes causing demyelination and the development of clinical symptoms of MS (created with BioRender.com by A Willson).

Advertisement

5. Cell types

While inflammation and neurodegeneration are correlated in active lesions, research suggests that neurodegeneration may become independent from inflammation in progressive disease [64]. There are many MS therapeutics that suppress proinflammatory cytokines or their effector functions, but not all treatments show equal efficacy and can cause unintended effects. Currently, there is no cure. It is thus becoming clear that there is a need to elucidate the different populations important in initiating and progressing disease, and by studying their interactive networks, identify possible areas for targeted intervention.

5.1 T cells

While there is overwhelming evidence of a role for T cells in the pathogenesis of MS, further studies in humans and in the mouse model of disease, experimental autoimmune encephalomyelitis (EAE), provides compelling evidence that other cell types play major roles. Linkage to the Human Leukocyte antigen (HLA) locus, including MHC I and II genes, was the first genetic locus identified, and still provides today the strongest linkage to MS. Further studies have identified an extended HLA haplotype, HLA-DRB1*15:01, DQA1*0102, DQB1*0602, within the MHC class II region [65]. As MHC II molecules specifically present peptide antigens to activate CD4 T cells, this suggests that CD4 T cells are important in initiation and progression of MS.

Th1 cells are a lineage of CD4 effector T cells that promote cell mediated immune responses and are necessary for defence against intracellular viral and bacterial pathogens. They were originally believed to be the main pathogenic T cells in MS, not only because susceptibility genes were linked to MHC II molecules, but also because immune surveillance of a healthy brain to scan for infection, showed favouring towards infiltration by Th1 cells, and therapeutic strategies designed to induce a shift from Th1 to Th2 immune response resulted in beneficial outcomes in MS patients [66, 67, 68].

The development of Th1 cells is coupled to the involvement of cell-extrinsic and cell-intrinsic factors, including signal transducer activator 1 (STAT1), the transcription factor Tbx21, IL-21 and STAT4 [69]. The CD4-Th1 model for MS was further supported by a trial performed in 1987, which found that administering IFN-γ to RRMS patients exacerbated disease. An accompanying increase in circulating monocytes bearing class II (HLA-DR) surface antigens suggested that the attacks induced by the treatment were immunologically mediated [70].

Th1 cells are also known to drive EAE. However, it was found that transgenic mice that lacked Th1 cells developed more severe EAE, thereby contradicting the Th1 cell theory for MS [71]. This conundrum was partially resolved following further investigation involving IL-23, a heterodimer cytokine composed of a unique p19 subunit and a common p40 subunit shared with IL-12. IL-23 promotes development of Th17 cells as opposed to Th1 cells [72]. Early studies on Th17 cells therefore dismissed a role for the previously favoured Th1 cells, but more recent research suggests that both cell types may play distinct roles in pathology [73]. It was suggested that Th1 cells accessed the CNS initially and subsequently facilitated the recruitment of Th17 cells [73].

Analysis of CNS tissue revealed distinct histopathological features and immune profiles depending on cytokine modulated T cells. IL-12p70 driven disease was characterised by macrophage-rich infiltrates, however in IL-23 driven lesions it was found that neutrophils and the growth factor, granulocyte colony stimulating factor (CSF), were the most prominent [74]. Research has shown that while IL-23 is commonly associated with the expansion of Th17 cells or the stabilisation of the Th17 phenotype, a similar course of EAE has been reported following the transfer of MOG-specific T cells into either wild type or IL-23 knockout mice [75]. This suggests that once encephalitogenic cells have been generated, EAE can develop in the absence of IL-23. IL-23 may therefore only be necessary for disease induction and not the effector phase of disease.

While MHC II molecules were found to be the strongest associated with MS in genetic studies, the MHC I HLA-A*0301 allele, independent of the HLA II haplotype DRB1*15,DQB1*06, was found to be increased in MS patients [65]. There was also a negative association with the MHC I HLA-A*0201 and disease [76]. As MHC I molecules are recognised by CD8 T cells, this suggests that CD8 T cells play a role in MS.

In one of the first studies that shifted from a CD4 T cell focus, CD8 T cells outnumbered the CD4 T cell subset in all parenchyma samples from MS patients, regardless of the MS type, duration or speed of disease progression [77]. Research has also shown that APCs, including dendritic cells (DCs), interact with T cells and proliferating lymphocytes, predominately CD8 T cells, at the margins of chronic active MS lesions [78]. CD8 T cells have also been found within active lesions of RRMS patients [77]. These T cells, and to a lesser extent, compartmentally differentially distributed B cells, have been shown to correlate with disease progression and damage.

CD8 T cells are an important subpopulation of MHC I restricted T cells, and are mediators of adaptive immunity. Cytotoxic T cells specialise in direct killing of cells that are infected, particularly with viruses, or are cancerous or damaged in other ways. Cytotoxic cells rely on two mechanisms for lytic activity: granule-dependent cytotoxicity (reviewed in [79]) and death receptor dependent cytotoxicity (reviewed in [80]). The principle mechanism used is granule-dependent cytotoxicity. In lesion prone areas of the CNS, T lymphocytes, including CD8 cytotoxic T lymphocytes (CTLs), are recruited to the affected tissue and brain cells are stimulated to present antigens to the T lymphocytes via de novo expression of MHC molecules. Although levels of MHC I and MHC II are very low in normal CNS parenchyma, neural injury leads to a massive increase in activated and phagocytotic microglial, which can serve as competent APCs [81]. To develop into functioning CD8 T cells, the TCR must recognise the MHC-peptide combination along with the costimulatory signal from APCs. While classical MHC I molecules necessary for CD8 T cell activation are not usually expressed on neural cells, they are induced in most inflammatory and degenerative CNS diseases [82].

Oligodendrocytes lack expression of costimulatory molecules and are thus unable to trigger the full effector of T cells, however they have been known to express MHC I in vitro [83]. Therefore, despite the lack of complete activation of the T cells, oligodendrocytes may still be targets of primed CTLs. MHC I expressing oligodendrocytes are susceptible to lysis by blood donor derived CD8 CTLs [83]. IFN-γ treated human oligodendrocytes also express Fas/CD95, and are therefore susceptible to death receptor dependent cytotoxicity [84]. Another component of the CNS, the neurons, were found to be capable of expressing MHC I when treated with IFN-γ [85, 86]. Medana and colleagues in 2000 discovered that hippocampal neurons were highly susceptible to direct application of cytotoxic granules, but showed no signs of perforin mediated lysis or membrane damage following attack by CTLs [87]. This effect was not observed in any other cell type.

Research to date indicates that all cellular elements of the CNS may act as targets to CTLs but that susceptibility and cytotoxic pathways involved vary dependent on the cell type and the immune activations during the course of the inflammatory process.

5.2 B cells

Historically, B cells have not been recognised as major players in regulatory function in the development of autoimmune diseases, although the identification of autoantibodies produced by autoreactive plasma cells and their pathogenic consequences are widely accepted [88]. B cells are considered effector cells as well as cells with immunoregulatory potential. B cells in MS patients express increased levels of costimulatory molecules, increasing the stimulation of antigen-reactive T cells [89]. It has been reported that MS patients have increased levels of IL-6 and GM-CSF, correlating with increased Th17 cells [90, 91]. B cell targeted therapies utilise B cell depleting monoclonal antibodies against the B cell marker CD20. These antibodies trigger B cell lysis through antibody dependent cellular cytotoxicity, complement dependent cytotoxicity or apoptosis induction [92].

5.3 NK cells

Administration of daclizumab, an alpha subunit of IL-2 receptor blocking monoclonal antibody, to MS patients was found to strongly reduce brain inflammation. This therapy, while being associated with a decline in circulating CD4 and CD8 T cells, also correlated with a significant expansion of CD56bright NK cells in vivo. This provided supporting evidence of NK cell-mediated negative immunoregulation of T cells during daclizumab treatment [93], and the identification of NK cells in association with MS, where positive outcome was possibly due to the treatment’s effect of increasing the NK cell numbers [94, 95].

For decades, NK cells have been classified as a component of the innate immune system. However, evidence suggests that, like B and T cells, NK cells are educated during development, possess antigen-specific receptors, undergo clonal expansion and generate memory cells (reviewed in [96]). Research originally suggested that NK cells developed and underwent differentiation within the bone marrow, however more recent extensive ex vivo characterisation of haematopoietic precursor cells (HPCs) and downstream NK cell development intermediates (NKDIs) reveals that they are enriched in secondary lymphoid tissues (STLs), including the tonsils, spleen and lymph nodes [97, 98, 99, 100]. This suggests that NK cells in humans can differentiate in the SLTs, and may do so preferentially.

Human NK cells are phenotypically defined by expression of CD56 and the lack of CD3 expression [101]. CD56 is the 140-kDa isoform of neural cell adhesion molecule (NCAM) found on NK cells and a minority of T cells [102]. NK cells are categorised into two distinct populations depending on the cell surface density of CD56. The majority of human NK cells, approximately 90%, express low levels of CD56 (CD56dim) and high levels of FCyRIII (CD16), while the minority express higher levels of CD56 (CD56bright) [103]. CD56bright NK cells have long being associated with an immunoregulatory role, due to increased production of NK-derived immunoregulatory cytokines, including IFN-γ, TNF-β, IL-10, IL-13 and GM-CSF, and reduced cytotoxicity compared to CD56dim NK cells [104]. CD56bright NK cells express receptors for cytokines such as IL-12, IL-15 and IL-18, produced by APCs, which can trigger proliferation of CD56bright NK cells and their production of molecules, including IFN-γ, IL-10 and IL-13 [104]. It has been demonstrated that DCs are a key source of cytokines for the activation of CD56bright NK cells [105]. Modulation and proliferation of CD56bright NK cells can also occur due to DC-derived IL-27 [105]. Activated NK cells can modulate the function of APCs by stimulating monocytes to produce TNF-α and kill immature DCs by a perforin-dependent process referred to as DC editing [106, 107].

However, more recent research has challenged this commonly accepted concept of CD56bright as the primary source of immunoregulatory cytokines. Studies have shown that CD56dim NK cells are also a major source of proinflammatory cytokines and chemokines that are induced rapidly after target cell recognition [108, 109].

The absence of MHC class I molecules, as indicated by virally infected cells or cancerous cells with MHC I downregulated, is not always sufficient to induce NK cell mediated death, suggesting that there must be activating receptors on NK cells whose affinity for target cell ligands dominates over the inhibitory signals of the NK cell. Some activating receptors identified include NKG2D, the NCR, and NKp80 [110, 111, 112]. NKG2D is the best characterised of these activating NK cell receptors. It is a c-type lectin-like receptor expressed on the surface of all human NK cells and recognises at least six ligands, each with a MHC class I homology [113]. Following receptor-ligand interaction, NKG2D phosphorylates an adaptor protein that recruits and activates phosphatidylinositol-3 (PI-3) kinase, which results in perforin-dependent cytotoxicity [114, 115]. Gunesh et al. found that the deletion of CD56 on the NK92 cell line lead to impaired cytotoxic function. The knockout CD56 cells failed to polarise during immunological synapse formation and had severely impaired exocytosis of lytic granules [116].

Treatment of MS patients with IFN-β caused an expansion of CD56bright NK cells, and resulted in the population of CD56dim cells being diminished [117]. The study also found that the proportion of CD56bright NK cells was significantly higher in the secondary lymphoid tissues compared to the peripheral blood for the control group [117]. This suggested that CD56bright NK cells may preferably locate within secondary lymphoid tissues, where they are able to interact with T cells and contribute to control of disease activity in MS [117].

There is an ongoing debate as to whether NK cells have a predominately beneficial or detrimental role in EAE, made even more complex by the lack of CD56 expression on murine NK cells. Studies have shown that enhancing the regulatory features of NK cells ameliorates the disease course of EAE. When the interaction between NKG2A and its ligand Qa-1 (the murine equivalent to the human HLA-E) expressed on target cells were blocked by antibodies specific for either antigen, it was found that NKG2A-expressing NK cells in particular decreased CNS inflammation by killing microglial and T cells [118, 119].

Enrichment of NK cells through treatment with IL-2 coupled with a monoclonal antibody specific for IL-2 (IL-2 mAb) was also found to ameliorate EAE [120]. The IL-2 mAb supplements the proliferation of NK and CD8 T cells in mice by increasing the biological activity of the pre-existing IL-2 by formation of immune complexes [121]. Increased levels of IL-2 was also found to expand Tregs while preventing the induction of Th17 during EAE development [122]. However, NK cells have different effects during the early stages of EAE, and possibly MS, compared to the late stages. In the early stages NK cells were found to protect the CNS whereas NK cells were found to kill neural stem cells (NSCs) during the late stages of EAE, as a result of reduced expression of Qa-1 on NSCs [120, 123].

5.4 NKT cells

NKT cells are unique T lymphocytes that express NK cell lineage markers, and act as a bridge between the innate and adaptive immune system. NKT cells account for a small percentage of lymphocytes, but have profound immunomodulatory roles in a variety of diseases [124]. There are two categories of NKT cells, type I and type II. Type I NKT cells, also known as invariant NKT cells (iNKT cells), express a semi-invariant Vα24-Jα18 (Vα14-Jα18 in mice), paired with a restricted range of β chains, that recognises α-galactosylceramide (α-GalCer) presented by CD1d [125, 126]. Type II NKT cells use TCRα and β chains that are reactive to a broad range of antigens, but do not recognise α-Galcer [127].

Nonobese diabetic (NOD) mice are susceptible to MOG-induced EAE. However, if NKT cells are increased either by transgenesis or adoptive transfer, the mice show protection from disease [128]. EAE protection has been correlated with inhibition of Ag-specific IFN-γ production in the spleen, modulating the encephalitogenic Th1 response [128]. There is conflicting evidence as to the effects of deletion of NKT cells on EAE. Some studies resulted in no effect on disease [129], with other studies showing disease exacerbation in CD1d-deficient and Jα18-deficient mice [130, 131]. Activation of type I NKT cells by α-GalCer has been shown to improve EAE outcome. These improvements arise by indirectly enhancing Th2 response and reducing the Th1 response, or potentiating the differential of immunosuppressive myeloid cells [131, 132, 133, 134]. However conflicting studies showed that high doses of α-GalCer could worsen EAE by directly enhancing Th17 and Th1 differentiation through phosphorylation of STAT3 and activation of NK-κB [135].

NKT cells from MS patients have been reported to have an increased production of cytokines. IL-4 production was increased by CD4 NKT cell clones in RRMS compared to other MS progression types, causing significant Th2 bias [136]. However, NKT cells in progressive MS patients displayed proinflammatory profiles [137]. It has also been suggested that the current available drugs for MS treatment may function through NKT cell targeting. A large reduction of type I NKT cells in peripheral blood was associated with remission of MS [136]. Type 1 interferon-β (T1IFN-β), a popular disease modifying therapy (DMT) for RRMS treatment, has been noted to promote expansion and functionality of type I NKT cells in vitro and to prevent disease in in vivo models of MS [138]. Research indicates a diverse role for NKT cells in MS pathology due to cytokine production.

5.5 Monocytes and macrophages

Besides imbalances in cytokine levels in the CNS and cerebrospinal fluid (CSF), immune imbalances also occur in the blood of MS patients, as reflected by altered levels of cytokines and cytokine producing cells (reviewed in [139]). The cause of these imbalances are thought to be due to circulating monocytes, with monocytes and macrophages influencing early MS, mediating both pro and anti-inflammatory responses [140, 141].

Surface expression of CD14 and CD16 are used to distinguish three distinct monocyte subsets: classical (CD14++CD16), intermediate (CD14++CD16+) and nonclassical (CD14+ CD16++) [142]. Monocytes and macrophages perform the key functions of antigen presentation and co-stimulation vital to the body’s immune response, with important roles in T and B cell activation and differentiation via the CD40-CD154 interaction (reviewed in [143]). Macrophages are primarily derived from blood borne monocytes, are present at sites of active demyelination in MS, and are assumed to be a part of the demyelinating process [144]. These inflammatory cells produce a range of toxic oxygen metabolites which mediate host tissue destruction. During MS progression, there is a significant expansion of the CD16+ monocyte population, which can primarily be attributed to nonclassical monocytes [145]. Depletion of these nonclassical monocytes may be an alternative to T and B cell depletion with the advantage of leaving the major classical monocyte population untouched. Selective subset depletion of monocytes may also supplement existing therapies to increase efficacy [145].

Advertisement

6. Conclusions

Multiple sclerosis is a complex autoimmune disease. Due to the many cell types involved in pathogenesis of the disease, therapeutics and treatments are often broad ranged and relatively inefficient. Further studies are necessary to uncover the genetic and environmental triggers leading to aberrant cellular toxicity and its role in MS pathogenesis. Discovering these and the related pathways will potentially lead to more targeted therapeutics and the elimination of not only MS but other autoimmune and neurological diseases in the future.

Advertisement

Acknowledgments

AMLW is supported by an Australian Government Research Training Program Stipend and MAJ was supported by an MS Research Australia/NHMRC Research Betty Cuthbert Fellowship. Project funds were obtained from Multiple Sclerosis Research Australia (MSRA), Lions’ club, Australia, and Australian Health Research Alliance-Women’s Health Research Translation Network (WHRTN).

Advertisement

Conflict of interest

The authors declare no conflict of interest.

References

  1. 1. Walton C, King R, Rechtman L, Kaye W, Leray E, Marrie RA, et al. Rising prevalence of multiple sclerosis worldwide: Insights from the Atlas of MS. Multiple Sclerosis Journal. 2020;26(14):1816-1821
  2. 2. Weiner HL. Multiple sclerosis is an inflammatory T-cell–mediated autoimmune disease. Archives of Neurology. 2004;61(10):1613-1615
  3. 3. McDonald WI, Compston A, Edan G, Goodkin D, Hartung HP, Lublin FD, et al. Recommended diagnostic criteria for multiple sclerosis: Guidelines from the International Panel on the diagnosis of multiple sclerosis. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society. 2001;50(1):121-127
  4. 4. Pender MP, Greer JM. Immunology of multiple sclerosis. Current Allergy and Asthma Reports. 2007;7(4):285-292
  5. 5. Cottrell D, Kremenchutzky M, Rice G, Koopman W, Hader W, Baskerville J, et al. The natural history of multiple sclerosis: A geographically based study: 5. The clinical features and natural history of primary progressive multiple sclerosis. Brain. 1999;122(4):625-639
  6. 6. Tremlett H, Paty D, Devonshire V. The natural history of primary progressive MS in British Columbia, Canada. Neurology. 2005;65(12):1919-1923
  7. 7. Goudarzi MH, Eadie MJ, Hollingworth SA. Disease modifying therapies for relapsing-remitting multiple sclerosis: Use and costs in Australia (1996-2019). Multiple Sclerosis and Related Disorders. 2021;50:102835
  8. 8. Confavreux C, Vukusic S, Moreau T, Adeleine P. Relapses and progression of disability in multiple sclerosis. New England Journal of Medicine. 2000;343(20):1430-1438
  9. 9. Confavreux C, Vukusic S. Natural history of multiple sclerosis: A unifying concept. Brain. 2006;129(3):606-616
  10. 10. Kurtzke JF. Epidemiology in multiple sclerosis: A pilgrim’s progress. Brain. 2013;136(9):2904-2917
  11. 11. Wang Y, Marling SJ, Zhu JG, Severson KS, DeLuca HF. Development of experimental autoimmune encephalomyelitis (EAE) in mice requires vitamin D and the vitamin D receptor. Proceedings of the National Academy of Sciences. 2012;109(22):8501-8504
  12. 12. Haghmorad D, Yazdanpanah E, Jadid Tavaf M, Zargarani S, Soltanmohammadi A, Mahmoudi MB, et al. Prevention and treatment of experimental autoimmune encephalomyelitis induced mice with 1, 25-dihydroxyvitamin D3. Neurological Research. 2019;41(10):943-957
  13. 13. Amezcua L, Conti DV, Liu L, Ledezma K, Langer-Gould AM. Place of birth, age of immigration, and disability in Hispanics with multiple sclerosis. Multiple Sclerosis and Related Disorders. 2015;4(1):25-30
  14. 14. Hammond S, English D, McLeod J. The age-range of risk of developing multiple sclerosis: Evidence from a migrant population in Australia. Brain. 2000;123(5):968-974
  15. 15. Levin LI, Munger KL, O'Reilly EJ, Falk KI, Ascherio A. Primary infection with the Epstein-Barr virus and risk of multiple sclerosis. Annals of Neurology. 2010;67(6):824-830
  16. 16. Pender MP. The essential role of Epstein-Barr virus in the pathogenesis of multiple sclerosis. The Neuroscientist. 2011;17(4):351-367
  17. 17. Correale J, Farez MF. Smoking worsens multiple sclerosis prognosis: Two different pathways are involved. Journal of Neuroimmunology. 2015;281:23-34
  18. 18. Sadovnick AD, Baird PA, Ward RH, Optiz JM, Reynolds JF. Multiple sclerosis. Updated risks for relatives. American Journal of Medical Genetics. 1988;29(3):533-541
  19. 19. Sadovnick A, Armstrong H, Rice G, Bulman D, Hashimoto L, Party D, et al. A population-based study of multiple sclerosis in twins: Update. Annals of Neurology. 1993;33(3):281-285
  20. 20. Consortium IMSG. Risk alleles for multiple sclerosis identified by a genomewide study. New England Journal of Medicine. 2007;357(9):851-862
  21. 21. Consortium IMSG. IL12A, MPHOSPH9/CDK2AP1 and RGS1 are novel multiple sclerosis susceptibility loci. Genes and Immunity. 2010;11(5):397
  22. 22. Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, et al. Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature. 2011;476(7359):214
  23. 23. Consortium*† IMSG, ANZgene, IIBDGC, WTCCC2. Multiple sclerosis genomic map implicates peripheral immune cells and microglia in susceptibility. Science. 2019;365(6460):eaav7188
  24. 24. Gresle MM, Jordan MA, Stankovich J, Spelman T, Johnson LJ, Laverick L, et al. Multiple sclerosis risk variants regulate gene expression in innate and adaptive immune cells. Life science alliance. 2020;3(7):1-11
  25. 25. Palacios R, Goni J, Martinez-Forero I, Iranzo J, Sepulcre J, Melero I, et al. A network analysis of the human T-cell activation gene network identifies JAGGED1 as a therapeutic target for autoimmune diseases. PloS One. 2007;2(11):e1222
  26. 26. Jordan MA, Baxter AG. In: Rose NR, Mackay IR, editors. The Autoimmune Diseases. Boston, MA, USA: Elsevier; 2020. pp. 383-418
  27. 27. Rose NR. Autoimmune disease: Reflections and projections. In: Rose NR. Mackay IR, editors. The Autoimmune Diseases. Boston, MA, USA: Elsevier; 2020. pp. 3-8
  28. 28. Harari A, Enders FB, Cellerai C, Bart P-A, Pantaleo G. Distinct profiles of cytotoxic granules in memory T cells correlate with function, differentiation stage, and antigen exposure. Journal of Virology. 2009;83(7):2862-2871
  29. 29. Zahavi D, AlDeghaither D, O’Connell A, Weiner LM. Enhancing antibody-dependent cell-mediated cytotoxicity: A strategy for improving antibody-based immunotherapy. Antibody Therapeutics. 2018;1(1):7-12
  30. 30. Kaufmann SH. Immunology's foundation: The 100-year anniversary of the Nobel Prize to Paul Ehrlich and Elie Metchnikoff. Nature Immunology. 2008;9(7):705-712
  31. 31. Hedges JF, Lubick KJ, Jutila MA. γδ T cells respond directly to pathogen-associated molecular patterns. The Journal of Immunology. 2005;174(10):6045-6053
  32. 32. Weiner HL. A shift from adaptive to innate immunity: A potential mechanism of disease progression in multiple sclerosis. Journal of Neurology. 2008;255(1):3-11
  33. 33. Gandhi R, Laroni A, Weiner HL. Role of the innate immune system in the pathogenesis of multiple sclerosis. Journal of Neuroimmunology. 2010;221(1-2):7-14
  34. 34. Shin ML, Rus HG, Niculescu FI. Membrane attack by complement: Assembly and biology of terminal complement complexes. Biomembranes: A Multi-Volume Treatise. 1996;4:123-149
  35. 35. Weng N-p. Aging of the immune system: How much can the adaptive immune system adapt? Immunity. 2006;24(5):495-499
  36. 36. Bonilla FA, Oettgen HC. Adaptive immunity. Journal of Allergy and Clinical Immunology. 2010;125(2):S33-S40
  37. 37. Castellino F, Germain RN. Cooperation between CD4+ and CD8+ T cells: When, where, and how. Annual Review in Immunology. 2006;24:519-540
  38. 38. Zhu J, Jankovic D, Oler AJ, Wei G, Sharma S, Hu G, et al. The transcription factor T-bet is induced by multiple pathways and prevents an endogenous Th2 cell program during Th1 cell responses. Immunity. 2012;37(4):660-673
  39. 39. Röcken M, Saurat J-H, Hauser C. A common precursor for CD4+ T cells producing IL-2 or IL-4. The Journal of Immunology. 1992;148(4):1031-1036
  40. 40. Singh V, Mehrotra S, Agarwal S. The paradigm of Th1 and Th2 cytokines. Immunologic Research. 1999;20(3):147-161
  41. 41. Bettelli E, Carrier Y, Gao W, Korn T, Strom TB, Oukka M, et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature. 2006;441(7090):235-238
  42. 42. Veldhoen M, Hocking RJ, Atkins CJ, Locksley RM, Stockinger B. TGFβ in the context of an inflammatory cytokine milieu supports de novo differentiation of IL-17-producing T cells. Immunity. 2006;24(2):179-189
  43. 43. Kisielow P, Blüthmann H, Staerz UD, Steinmetz M, Von Boehmer H. Tolerance in T-cell-receptor transgenic mice involves deletion of nonmature CD4+ 8+ thymocytes. Nature. 1988;333(6175):742-746
  44. 44. Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+ CD25+ regulatory T cells. Nature Immunology. 2003;4(4):330-336
  45. 45. Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science. 2003;299(5609):1057-1061
  46. 46. Khattri R, Cox T, Yasayko S-A, Ramsdell F. An essential role for Scurfin in CD4+ CD25+ T regulatory cells. Nature Immunology. 2003;4(4):337-342
  47. 47. Blair PJ, Bultman SJ, Haas JC, Rouse BT, Wilkinson JE, Godfrey VL. CD4+ CD8-T cells are the effector cells in disease pathogenesis in the scurfy (sf) mouse. The Journal of Immunology. 1994;153(8):3764-3774
  48. 48. Lucchinetti C, Brück W, Parisi J, Scheithauer B, Rodriguez M, Lassmann H. Heterogeneity of multiple sclerosis lesions: Implications for the pathogenesis of demyelination. Annals of Neurology: Official Journal of the American Neurological Association and the Child Neurology Society. 2000;47(6):707-717
  49. 49. Wardemann H, Yurasov S, Schaefer A, Young JW, Meffre E, Nussenzweig MC. Predominant autoantibody production by early human B cell precursors. Science. 2003;301(5638):1374-1377
  50. 50. Rose NR, Bona C. Defining criteria for autoimmune diseases (Witebsky’s postulates revisited). Immunology Today. 1993;14(9):426-430
  51. 51. Zenewicz LA, Abraham C, Flavell RA, Cho JH. Unraveling the genetics of autoimmunity. Cell. 2010;140(6):791-797
  52. 52. Lerner A, Jeremias P, Matthias T. The world incidence and prevalence of autoimmune diseases is increasing. International Journal of Celiac Diseases. 2015;3(4):151-155
  53. 53. Jacobson DL, Gange SJ, Rose NR, Graham NM. Epidemiology and estimated population burden of selected autoimmune diseases in the United States. Clinical Immunology and Immunopathology. 1997;84(3):223-243
  54. 54. Vargas-Parada L. Research round-up: Autoimmune disease. Nature. 2021;595(7867):46-47
  55. 55. Shevach EM. Mechanisms of foxp3+ T regulatory cell-mediated suppression. Immunity. 2009;30(5):636-645
  56. 56. Tang Q , Bluestone JA. The Foxp3+ regulatory T cell: A jack of all trades, master of regulation. Nature Immunology. 2008;9(3):239-244
  57. 57. Coffman RL. Origins of the TH1-TH2 model: A personal perspective. Nature Immunology. 2006;7(6):539-541
  58. 58. Khader SA, Gaffen SL, Kolls JK. Th17 cells at the crossroads of innate and adaptive immunity against infectious diseases at the mucosa. Mucosal Immunology. 2009;2(5):403-411
  59. 59. Laan M, Cui Z-H, Hoshino H, Lötvall J, Sjöstrand M, Gruenert DC, et al. Neutrophil recruitment by human IL-17 via CXC chemokine release in the airways. The Journal of Immunology. 1999;162(4):2347-2352
  60. 60. Ye P, Rodriguez FH, Kanaly S, Stocking KL, Schurr J, Schwarzenberger P, et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. The Journal of Experimental Medicine. 2001;194(4):519-528
  61. 61. Romagnani S. Th1/th2 cells. Inflammatory Bowel Diseases. 1999;5(4):285-294
  62. 62. Kivisäkk P, Imitola J, Rasmussen S, Elyaman W, Zhu B, Ransohoff RM, et al. Localizing central nervous system immune surveillance: Meningeal antigen-presenting cells activate T cells during experimental autoimmune encephalomyelitis. Annals of Neurology. 2009;65(4):457-469
  63. 63. Brown DA, Sawchenko PE. Time course and distribution of inflammatory and neurodegenerative events suggest structural bases for the pathogenesis of experimental autoimmune encephalomyelitis. Journal of Comparative Neurology. 2007;502(2):236-260
  64. 64. Frischer JM, Bramow S, Dal-Bianco A, Lucchinetti CF, Rauschka H, Schmidbauer M, et al. The relation between inflammation and neurodegeneration in multiple sclerosis brains. Brain. 2009;132(5):1175-1189
  65. 65. Fogdell-Hahn A, Ligers A, Grønning M, Hillert J, Olerup O. Multiple sclerosis: A modifying influence of HLA class I genes in an HLA class II associated autoimmune disease. Tissue Antigens. 2000;55(2):140-148
  66. 66. Hohlfeld R. Biotechnological agents for the immunotherapy of multiple sclerosis. Principles, problems and perspectives. Brain: A Journal of Neurology. 1997;120(5):865-916
  67. 67. Herrera BM, Ebers GC. Progress in deciphering the genetics of multiple sclerosis. Current Opinion in Neurology. 2003;16(3):253-258
  68. 68. Kivisäkk P, Mahad DJ, Callahan MK, Trebst C, Tucky B, Wei T, et al. Human cerebrospinal fluid central memory CD4+ T cells: Evidence for trafficking through choroid plexus and meninges via P-selectin. Proceedings of the National Academy of Sciences. 2003;100(14):8389-8394
  69. 69. Harrington LE, Hatton RD, Mangan PR, Turner H, Murphy TL, Murphy KM, et al. Interleukin 17–producing CD4+ effector T cells develop via a lineage distinct from the T helper type 1 and 2 lineages. Nature Immunology. 2005;6(11):1123-1132
  70. 70. Panitch H, Haley A, Hirsch R, Johnson K. Exacerbations of multiple sclerosis in patients treated with gamma interferon. The Lancet. 1987;329(8538):893-895
  71. 71. Ferber IA, Brocke S, Taylor-Edwards C, Ridgway W, Dinisco C, Steinman L, et al. Mice with a disrupted IFN-gamma gene are susceptible to the induction of experimental autoimmune encephalomyelitis (EAE). The Journal of Immunology. 1996;156(1):5-7
  72. 72. Langrish CL, Chen Y, Blumenschein WM, Mattson J, Basham B, Sedgwick JD, et al. IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. The Journal of Experimental Medicine. 2005;201(2):233-240
  73. 73. O'Connor RA, Prendergast CT, Sabatos CA, Lau CW, Leech MD, Wraith DC, et al. Cutting edge: Th1 cells facilitate the entry of Th17 cells to the central nervous system during experimental autoimmune encephalomyelitis. The Journal of Immunology. 2008;181(6):3750-3754
  74. 74. Kroenke MA, Carlson TJ, Andjelkovic AV, Segal BM. IL-12–and IL-23–modulated T cells induce distinct types of EAE based on histology, CNS chemokine profile, and response to cytokine inhibition. The Journal of Experimental Medicine. 2008;205(7):1535-1541
  75. 75. Thakker P, Leach MW, Kuang W, Benoit SE, Leonard JP, Marusic S. IL-23 is critical in the induction but not in the effector phase of experimental autoimmune encephalomyelitis. The Journal of Immunology. 2007;178(4):2589-2598
  76. 76. Harbo H, Lie B, Sawcer S, Celius E, Dai KZ, Oturai A, et al. Genes in the HLA class I region may contribute to the HLA class II-associated genetic susceptibility to multiple sclerosis. Tissue Antigens. 2004;63(3):237-247
  77. 77. Booss J, Esiri MM, Tourtellotte WW, Mason DY. Immunohistological analysis of T lymphocyte subsets in the central nervous system in chronic progressive multiple sclerosis. Journal of the Neurological Sciences. 1983;62(1-3):219-232
  78. 78. Serafini B, Rosicarelli B, Magliozzi R, Stigliano E, Capello E, Mancardi GL, et al. Dendritic cells in multiple sclerosis lesions: Maturation stage, myelin uptake, and interaction with proliferating T cells. Journal of Neuropathology & Experimental Neurology. 2006;65(2):124-141
  79. 79. Chowdhury D, Lieberman J. Death by a thousand cuts: Granzyme pathways of programmed cell death. Annual Review in Immunology. 2008;26:389-420
  80. 80. Russell JH, Ley TJ. Lymphocyte-mediated cytotoxicity. Annual Review of Immunology. 2002;20(1):323-370
  81. 81. Raivich G, Jones LL, Kloss CU, Werner A, Neumann H, Kreutzberg GW. Immune surveillance in the injured nervous system: T-lymphocytes invade the axotomized mouse facial motor nucleus and aggregate around sites of neuronal degeneration. Journal of Neuroscience. 1998;18(15):5804-5816
  82. 82. Oliveira AL, Thams S, Lidman O, Piehl F, Hökfelt T, Kärre K, et al. A role for MHC class I molecules in synaptic plasticity and regeneration of neurons after axotomy. Proceedings of the National Academy of Sciences. 2004;101(51):17843-17848
  83. 83. Jurewicz A, Biddison WE, Antel JP. MHC class I-restricted lysis of human oligodendrocytes by myelin basic protein peptide-specific CD8 T lymphocytes. The Journal of Immunology. 1998;160(6):3056-3059
  84. 84. Pouly S, Becher B, Blain M, Antel JP. Interferon-γ modulates human oligodendrocyte susceptibility to Fas-mediated apoptosis. Journal of Neuropathology & Experimental Neurology. 2000;59(4):280-286
  85. 85. Rensing-Ehl A, Malipiero U, Irmler M, Tschopp J, Constam D, Fontana A. Neurons induced to express major histocompatibility complex class I antigen are killed via the perforin and not the Fas (APO-1/CD95) pathway. European Journal of Immunology. 1996;26(9):2271-2274
  86. 86. Neumann H, Schmidt H, Cavalie A, Jenne D, Wekerle H. Major histocompatibility complex (MHC) class I gene expression in single neurons of the central nervous system: Differential regulation by interferon (IFN)-γ and tumor necrosis factor (TNF)-α. The Journal of Experimental Medicine. 1997;185(2):305-316
  87. 87. Medana IM, Gallimore A, Oxenius A, Martinic MM, Wekerle H, Neumann H. MHC class I-restricted killing of neurons by virus-specific CD8+ T lymphocytes is effected through the Fas/FasL, but not the perforin pathway. European Journal of Immunology. 2000;30(12):3623-3633
  88. 88. Dörner T, Jacobi AM, Lipsky PE. B cells in autoimmunity. Arthritis Research & Therapy. 2009;11(5):1-11
  89. 89. Aung LL, Balashov KE. Decreased Dicer expression is linked to increased expression of co-stimulatory molecule CD80 on B cells in multiple sclerosis. Multiple Sclerosis Journal. 2015;21(9):1131-1138
  90. 90. Barr TA, Shen P, Brown S, Lampropoulou V, Roch T, Lawrie S, et al. B cell depletion therapy ameliorates autoimmune disease through ablation of IL-6–producing B cells. Journal of Experimental Medicine. 2012;209(5):1001-1010
  91. 91. Li R, Rezk A, Miyazaki Y, Hilgenberg E, Touil H, Shen P, et al. Proinflammatory GM-CSF–producing B cells in multiple sclerosis and B cell depletion therapy. Science Translational Medicine. 2015;7(310):ra166
  92. 92. Kamburova EG, Koenen HJ, Borgman KJ, Ten Berge I, Joosten I, Hilbrands LB. A single dose of rituximab does not deplete B cells in secondary lymphoid organs but alters phenotype and function. American Journal of Transplantation. 2013;13(6):1503-1511
  93. 93. Bielekova B, Catalfamo M, Reichert-Scrivner S, Packer A, Cerna M, Waldmann TA, et al. Regulatory CD56bright natural killer cells mediate immunomodulatory effects of IL-2Rα-targeted therapy (daclizumab) in multiple sclerosis. Proceedings of the National Academy of Sciences. 2006;103(15):5941-5946
  94. 94. Giovannoni G, Gold R, Selmaj K, Havrdova E, Montalban X, Radue E-W, et al. Daclizumab high-yield process in relapsing-remitting multiple sclerosis (SELECTION): A multicentre, randomised, double-blind extension trial. The Lancet Neurology. 2014;13(5):472-481
  95. 95. Kappos L, Wiendl H, Selmaj K, Arnold DL, Havrdova E, Boyko A, et al. Daclizumab HYP versus interferon beta-1a in relapsing multiple sclerosis. New England Journal of Medicine. 2015;373(15):1418-1428
  96. 96. Sun JC, Lanier LL. NK cell development, homeostasis and function: Parallels with CD8+ T cells. Nature Reviews Immunology. 2011;11(10):645-657
  97. 97. Freud AG, Becknell B, Roychowdhury S, Mao HC, Ferketich AK, Nuovo GJ, et al. A human CD34 (+) subset resides in lymph nodes and differentiates into CD56brightNatural killer cells. Immunity. 2005;22(3):295-304
  98. 98. Freud AG, Yokohama A, Becknell B, Lee MT, Mao HC, Ferketich AK, et al. Evidence for discrete stages of human natural killer cell differentiation in vivo. The Journal of Experimental Medicine. 2006;203(4):1033-1043
  99. 99. Eissens DN, Spanholtz J, Van Der Meer A, Van Cranenbroek B, Dolstra H, Kwekkeboom J, et al. Defining early human NK cell developmental stages in primary and secondary lymphoid tissues. PloS One. 2012;7(2):e30930
  100. 100. Scoville SD, Mundy-Bosse BL, Zhang MH, Chen L, Zhang X, Keller KA, et al. A progenitor cell expressing transcription factor RORγt generates all human innate lymphoid cell subsets. Immunity. 2016;44(5):1140-1150
  101. 101. Griffin JD, Hercend T, Beveridge R, Schlossman SF. Characterization of an antigen expressed by human natural killer cells. The Journal of Immunology. 1983;130(6):2947-2951
  102. 102. Lanier LL, Testi R, Bindl J, Phillips JH. Identity of Leu-19 (CD56) leukocyte differentiation antigen and neural cell adhesion molecule. The Journal of Experimental Medicine. 1989;169(6):2233-2238
  103. 103. Lanier LL, Le AM, Civin C, Loken M, Phillips J. The relationship of CD16 (Leu-11) and Leu-19 (NKH-1) antigen expression on human peripheral blood NK cells and cytotoxic T lymphocytes. The Journal of Immunology. 1986;136(12):4480-4486
  104. 104. Cooper MA, Fehniger TA, Turner SC, Chen KS, Ghaheri BA, Ghayur T, et al. Human natural killer cells: A unique innate immunoregulatory role for the CD56bright subset. Blood, The Journal of the American Society of Hematology. 2001;97(10):3146-3151
  105. 105. Ferlazzo G, Pack M, Thomas D, Paludan C, Schmid D, Strowig T, et al. Distinct roles of IL-12 and IL-15 in human natural killer cell activation by dendritic cells from secondary lymphoid organs. Proceedings of the National Academy of Sciences. 2004;101(47):16606-16611
  106. 106. Dalbeth N, Gundle R, Davies RJ, Lee YG, McMichael AJ, Callan MF. CD56bright NK cells are enriched at inflammatory sites and can engage with monocytes in a reciprocal program of activation. The Journal of Immunology. 2004;173(10):6418-6426
  107. 107. Morandi B, Mortara L, Chiossone L, Accolla RS, Mingari MC, Moretta L, et al. Dendritic cell editing by activated natural killer cells results in a more protective cancer-specific immune response. PloS One. 2012;7(6):e39170
  108. 108. Juelke K, Killig M, Luetke-Eversloh M, Parente E, Gruen J, Morandi B, et al. CD62L expression identifies a unique subset of polyfunctional CD56dim NK cells. Blood, The Journal of the American Society of Hematology. 2010;116(8):1299-1307
  109. 109. Fauriat C, Long EO, Ljunggren H-G, Bryceson YT. Regulation of human NK-cell cytokine and chemokine production by target cell recognition. Blood, The Journal of the American Society of Hematology. 2010;115(11):2167-2176
  110. 110. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, et al. Activation of NK cells and T cells by NKG2D, a receptor for stress-inducible MICA. Science. 1999;285(5428):727-729
  111. 111. Bottino C, Moretta L, Moretta A. NK cell activating receptors and tumor recognition in humans. Immunobiology of Natural Killer Cell Receptors. 2006;298:175-182
  112. 112. Welte S, Kuttruff S, Waldhauer I, Steinle A. Mutual activation of natural killer cells and monocytes mediated by NKp80-AICL interaction. Nature Immunology. 2006;7(12):1334-1342
  113. 113. Diefenbach A, Hsia JK, Hsiung MYB, Raulet DH. A novel ligand for the NKG2D receptor activates NK cells and macrophages and induces tumor immunity. European Journal of Immunology. 2003;33(2):381-391
  114. 114. Hayakawa Y, Kelly JM, Westwood JA, Darcy PK, Diefenbach A, Raulet D, et al. Cutting edge: Tumor rejection mediated by NKG2D receptor-ligand interaction is dependent upon perforin. The Journal of Immunology. 2002;169(10):5377-5381
  115. 115. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL, et al. An activating immunoreceptor complex formed by NKG2D and DAP10. Science. 1999;285(5428):730-732
  116. 116. Gunesch JT, Dixon AL, Ebrahim TA, Berrien-Elliott MM, Tatineni S, Kumar T, et al. CD56 regulates human NK cell cytotoxicity through Pyk2. Elife. 2020;9:e57346
  117. 117. Saraste M, Irjala H, Airas L. Expansion of CD56Bright natural killer cells in the peripheral blood of multiple sclerosis patients treated with interferon-beta. Neurological Sciences. 2007;28(3):121-126
  118. 118. Lu L, Ikizawa K, Hu D, Werneck MB, Wucherpfennig KW, Cantor H. Regulation of activated CD4+ T cells by NK cells via the Qa-1–NKG2A inhibitory pathway. Immunity. 2007;26(5):593-604
  119. 119. Leavenworth JW, Schellack C, Kim H-J, Lu L, Spee P, Cantor H. Analysis of the cellular mechanism underlying inhibition of EAE after treatment with anti-NKG2A F (ab′) 2. Proceedings of the National Academy of Sciences. 2010;107(6):2562-2567
  120. 120. Hao J, Liu R, Piao W, Zhou Q , Vollmer TL, Campagnolo DI, et al. Central nervous system (CNS)–resident natural killer cells suppress Th17 responses and CNS autoimmune pathology. Journal of Experimental Medicine. 2010;207(9):1907-1921
  121. 121. Boyman O, Kovar M, Rubinstein MP, Surh CD, Sprent J. Selective stimulation of T cell subsets with antibody-cytokine immune complexes. Science. 2006;311(5769):1924-1927
  122. 122. Rouse M, Nagarkatti M, Nagarkatti PS. The role of IL-2 in the activation and expansion of regulatory T-cells and the development of experimental autoimmune encephalomyelitis. Immunobiology. 2013;218(4):674-682
  123. 123. Liu Q , Sanai N, Jin W-N, La Cava A, Van Kaer L, Shi F-D. Neural stem cells sustain natural killer cells that dictate recovery from brain inflammation. Nature Neuroscience. 2016;19(2):243-252
  124. 124. Taniguchi M, Seino K-i, Nakayama T. The NKT cell system: Bridging innate and acquired immunity. Nature Immunology. 2003;4(12):1164-1165
  125. 125. Kawano T, Cui J, Koezuka Y, Toura I, Kaneko Y, Motoki K, et al. CD1d-restricted and TCR-mediated activation of Vα14 NKT cells by glycosylceramides. Science. 1997;278(5343):1626-1629
  126. 126. Benlagha K, Weiss A, Beavis A, Teyton L, Bendelac A. In vivo identification of glycolipid antigen–specific T cells using fluorescent CD1d tetramers. The Journal of Experimental Medicine. 2000;191(11):1895-1904
  127. 127. Dellabona P, Padovan E, Casorati G, Brockhaus M, Lanzavecchia A. An invariant V alpha 24-J alpha Q/V beta 11 T cell receptor is expressed in all individuals by clonally expanded CD4-8-T cells. The Journal of Experimental Medicine. 1994;180(3):1171-1176
  128. 128. Mars LT, Laloux V, Goude K, Desbois S, Saoudi A, Van Kaer L, et al. Cutting edge: Vα14-Jα281 NKT cells naturally regulate experimental autoimmune encephalomyelitis in nonobese diabetic mice. The Journal of Immunology. 2002;168(12):6007-6011
  129. 129. Oh SJ, Chung DH. Invariant NKT cells producing IL-4 or IL-10, but not IFN-γ, inhibit the Th1 response in experimental autoimmune encephalomyelitis, whereas none of these cells inhibits the Th17 response. The Journal of Immunology. 2011;186(12):6815-6821
  130. 130. Teige A, Teige I, Lavasani S, Bockermann R, Mondoc E, Holmdahl R, et al. CD1-dependent regulation of chronic central nervous system inflammation in experimental autoimmune encephalomyelitis. The Journal of Immunology. 2004;172(1):186-194
  131. 131. Furlan R, Bergami A, Cantarella D, Brambilla E, Taniguchi M, Dellabona P, et al. Activation of invariant NKT cells by αGalCer administration protects mice from MOG35-55-induced EAE: Critical roles for administration route and IFN-γ. European Journal of Immunology. 2003;33(7):1830-1838
  132. 132. Miyamoto K, Miyake S, Yamamura T. A synthetic glycolipid prevents autoimmune encephalomyelitis by inducing TH2 bias of natural killer T cells. Nature. 2001;413(6855):531-534
  133. 133. Parekh VV, Wu L, Olivares-Villagómez D, Wilson KT, Van Kaer L. Activated invariant NKT cells control central nervous system autoimmunity in a mechanism that involves myeloid-derived suppressor cells. The Journal of Immunology. 2013;190(5):1948-1960
  134. 134. Kojo S, Seino K-i, Harada M, Watarai H, Wakao H, Uchida T, et al. Induction of regulatory properties in dendritic cells by Vα14 NKT cells. The Journal of Immunology. 2005;175(6):3648-3655
  135. 135. Qian G, Qin X, Zang YQ , Ge B, Guo TB, Wan B, et al. High doses of α-galactosylceramide potentiate experimental autoimmune encephalomyelitis by directly enhancing Th17 response. Cell Research. 2010;20(4):480-491
  136. 136. Araki M, Kondo T, Gumperz JE, Brenner MB, Miyake S, Yamamura T. Th2 bias of CD4+ NKT cells derived from multiple sclerosis in remission. International Immunology. 2003;15(2):279-288
  137. 137. De Biasi S, Simone AM, Nasi M, Bianchini E, Ferraro D, Vitetta F, et al. iNKT cells in secondary progressive multiple sclerosis patients display pro-inflammatory profiles. Frontiers in Immunology. 2016;7:555
  138. 138. Freedman MS, Devonshire V, Duquette P, Giacomini PS, Giuliani F, Levin MC, et al. Treatment optimization in multiple sclerosis: Canadian MS Working Group recommendations. Canadian Journal of Neurological Sciences. 2020;47(4):437-455
  139. 139. Navikas V, Link H. Cytokines and the pathogenesis of multiple sclerosis. Journal of Neuroscience Research. 1996;45(4):322-333
  140. 140. Kouwenhoven M, Teleshova N, Özenci V, Press R, Link H. Monocytes in multiple sclerosis: Phenotype and cytokine profile. Journal of Neuroimmunology. 2001;112(1-2):197-205
  141. 141. Miron VE, Boyd A, Zhao J-W, Yuen TJ, Ruckh JM, Shadrach JL, et al. M2 microglia and macrophages drive oligodendrocyte differentiation during CNS remyelination. Nature Neuroscience. 2013;16(9):1211-1218
  142. 142. Ziegler-Heitbrock L, Ancuta P, Crowe S, Dalod M, Grau V, Hart DN, et al. Nomenclature of monocytes and dendritic cells in blood. Blood, The Journal of the American Society of Hematology. 2010;116(16):e74-e80
  143. 143. Chitnis T, Khoury SJ. Role of costimulatory pathways in the pathogenesis of multiple sclerosis and experimental autoimmune encephalomyelitis. Journal of Allergy and Clinical Immunology. 2003;112(5):837-849
  144. 144. Fisher M, Levine PH, Weiner BH, Vaudreuil CH, Natale A, Johnson MH, et al. Monocyte and polymorphonuclear leukocyte toxic oxygen metabolite production in multiple sclerosis. Inflammation. 1988;12(2):123-131
  145. 145. Gjelstrup MC, Stilund M, Petersen T, Møller HJ, Petersen EL, Christensen T. Subsets of activated monocytes and markers of inflammation in incipient and progressed multiple sclerosis. Immunology and Cell Biology. 2018;96(2):160-174

Written By

Annie M.L. Willson and Margaret A. Jordan

Submitted: 09 May 2022 Reviewed: 02 June 2022 Published: 29 June 2022