234
Views
27
CrossRef citations to date
0
Altmetric
Review

Animal models of myasthenia gravis: utility and limitations

, , &
Pages 53-64 | Published online: 04 Mar 2016

Abstract

Myasthenia gravis (MG) is a chronic autoimmune disease caused by the immune attack of the neuromuscular junction. Antibodies directed against the acetylcholine receptor (AChR) induce receptor degradation, complement cascade activation, and postsynaptic membrane destruction, resulting in functional reduction in AChR availability. Besides anti-AChR antibodies, other autoantibodies are known to play pathogenic roles in MG. The experimental autoimmune MG (EAMG) models have been of great help over the years in understanding the pathophysiological role of specific autoantibodies and T helper lymphocytes and in suggesting new therapies for prevention and modulation of the ongoing disease. EAMG can be induced in mice and rats of susceptible strains that show clinical symptoms mimicking the human disease. EAMG models are helpful for studying both the muscle and the immune compartments to evaluate new treatment perspectives. In this review, we concentrate on recent findings on EAMG models, focusing on their utility and limitations.

Myasthenia gravis

Acquired myasthenia gravis (MG) is a B-cell-mediated T-cell-dependent autoimmune disease, which is characterized by impairment of the neuromuscular junction (NMJ) transmission and caused by specific autoantibodies (auto-Abs) directed against the acetylcholine receptor (AChR) on the postsynaptic membrane of skeletal muscle cells.Citation1Citation3 The majority of AChR antibodies recognize an extracellular domain of the receptor, defined as main immunogenic region, localized between residues 67 and 76 of the α-subunit of the receptor.Citation4 The development of anti-AChR auto-Abs is apparently due to the breakdown of self-tolerance in the thymus,Citation5Citation7 with activation of AChR-specific CD4+ T helper (Th) cells and production of proinflammatory cytokines, leading to the synthesis of high-affinity antibodiesCitation8,Citation9 and chemokines contributing to autoimmunity maintenance.Citation10 MG responds to the clinical criteria of antibody-mediated autoimmune diseases,Citation11 implying the presence of auto-Abs in patients,Citation1,Citation12,Citation13 which specifically interact with the target antigen forming immune complexesCitation2,Citation14 and induce an experimental model, when injected in recipient animals, reproducing the feature of the disease (passive transfer).Citation15,Citation16 Other criteria imply that the immunization with the specific antigen produces an experimental model that is clinically similar to the diseaseCitation17 and that the reduction in circulating antibody levels ameliorates the disease.Citation18,Citation19 AChR-specific auto-Abs induce complement activation and damage of the NMJ with increased degradation of AChR. The direct binding of auto-Abs (IgG1 and IgG3 subtypes) activates the complement cascade, leading to the formation of the membrane attack complex (MAC) and consequently to the lysis of the muscle cell.Citation14 The destruction of the postsynaptic membrane results in a morphological alteration, with decreased number of functional AChRs and sodium channels.Citation20 The formation of immune complexes induces endocytosis-mediated internalization of AChR, which is not compensated by ex novo synthesis, and increases lysosomal degradation of AChR, reducing its availability on the postsynaptic membrane.Citation14,Citation21 Further impairment of AChR function can also be derived from the physical interaction of a subset of polyclonal anti-AChR auto-Abs to the specific acetylcholine binding sites on the receptor.Citation22

Experimental autoimmune myasthenia gravis

The first report on an experimental model of MG was published >30 years ago,Citation17 showing that rabbits immunized with AChR, purified from the Electrophorus electricus electric organ, developed MG-like symptoms. Later on, many animal studies confirmed that an autoimmune response was occurring in MG patients against muscle AChR and that anti-AChR antibodies were responsible for the structural and functional damage of the NMJ. Over the years, experimental autoimmune MG (EAMG) has represented an excellent model to investigate the pathogenic mechanisms underlying the human disease and to evaluate the efficacy of new immunotherapies.Citation23

MG and its animal models share several immunopathological featuresCitation23 such as the presence of anti-AChR antibodies in serum, IgGs and complement component deposition at the NMJ, major histocompatibility complex class II-restricted presentation of AChR epitopes, and involvement of Th cells in the production of B-cell antibody,Citation24 and several clinical features, such as muscle weakness and fatigability, decremented response after repetitive nerve stimulation, increased curare sensitivity, and temporary improvement of muscle strength following treatment with anticholinesterase drugs ().Citation24

Table 1 Immunopathological similarities and differences between MG and its experimental model

Although EAMG can be induced in various animal species, most of the experimental models are established in rats and mice, mainly due to the high incidence of clinical EAMG signs.Citation25 The course of EAMG is evaluated by monitoring the loss of body weight and muscular strength of the immunized animals. Myasthenic symptoms, assessed after exercise, include tremor, hunched posture, muscle weakness, and fatigue and are given in detail in . In susceptible rat strains, EAMG is induced by active immunization with Torpedo californica AChR (TAChR)Citation25 or with a rat AChR epitope capable of breaking immunological tolerance (amino acids [aa] 97–116 of the α-subunit).Citation26,Citation27 EAMG can also be induced by passive transfer of anti-AChR antibodies,Citation1,Citation4 which is the simplest protocol for studying the pathogenic effects of auto-Abs in vivo.

Table 2 Clinical evaluation of EAMG symptoms

Active EAMG

Mice would represent the ideal model for the development of the experimental disease due to the availability of transgenic, knockout, and mutant mice that are optimal for the investigation of the biological mechanisms at the basis of MG pathogenesis.Citation24,Citation28 Indeed, EAMG has been intensively studied in mice to better understand the factors that are involved in the disease pathogenesis and to investigate their potential modulation and regulation. Highly susceptible murine strains are C57Bl/6, SJL, and AKR, where 50%–70% of animals developed myasthenic symptoms induced by TAChR immunization, which are different from the poorly susceptible BALB/c and SWR strains.Citation28,Citation29 EAMG in the mouse is routinely induced by immunization with purified AChR (20 g) in complete Freund’s adjuvant (CFA) followed by two or three boosts with AChR (20 μg) in incomplete Freund’s adjuvant. This procedure triggers the production of antibodies to both foreign AChR and self-AChR,Citation25,Citation30 and myasthenic symptoms typically appear 7–14 days after the last injection.Citation24,Citation28 Due to the several immunization boosts required to induce this model, it is relatively difficult to define the appropriate time windows for preventive and/or therapeutic approaches in mice.

Various inbred rat strains have been tested for the induction of active EAMG via immunization with TAChR, with more severe clinical features compared with those observed in murine models. The strain most employed is the Lewis rat, which exhibits clinical manifestations most similar to those of human MG.Citation31 EAMG in Lewis rats is generally induced via single immunization with purified AChR (20 μg) in CFA, prompting the production of antibodies to foreign AChR, which similar to the mouse model are able to cross-react with the self-AChR.Citation25,Citation30 Two different disease phases can be clinically distinguished. The first acute transient phase begins ~7 days postimmunization and is characterized by the synthesis of anti-AChR antibodies (IgM type), which leads to complement depositions on muscle membrane, extensive phagocytic invasion at the NMJ, and destruction of the postsynaptic membrane. The cellular invasion decreases the AChR content of a rapid muscle, which is followed (after 2–3 days) by an abnormal increase in the AChR content likely due to the formation of extrajunctional AChR.Citation30 The second progressive chronic phase begins ~28 days postimmunizationCitation25 and is characterized by the production of a larger amount of antibodies (IgG type) and complement deposition at the postsynaptic membrane, which thus appears flat, due to lack in junctional folds. In this phase, there are no phagocytic cells, and the reduction in skeletal muscle AChR content is such that it is reduced to one-third compared with that of healthy animals. Importantly, this phase reflects the clinical course of the human disease.

In the rat, it was proven that active EAMG can also be induced via immunization with a synthetic peptide, corresponding to the immunogenic region 97–116 of rat AChR α-subunit (R97–116) in CFA (50 μg of peptide), followed by a second immunization boost of R97–116 (50 μg) in incomplete Freund’s adjuvant 30 days after the first immunization.Citation26 The onset of EAMG manifestations appears 2 weeks after the booster injection.Citation26 When compared with TAChR-induced EAMG, R97–116-induced EAMG shows a different time course, which is characterized by a slower progression over time and a slightly wider clinical inhomogeneity among immunized animals.Citation26 Due to the considerably better feasibility of working with a peptide of the rat AChR instead of the whole AChR extracted from T. californica, several recently published studies on new therapeutic strategies have been performed in the R97–116 experimental model.Citation32Citation35

Passive transfer of EAMG

EAMG can be also induced by passive transfer of auto-Abs via two distinct mechanisms: either with daily injections into healthy recipient animals of serum IgG fraction isolated from MG patientsCitation15 or with anti-AChR antibodies purified from AChR-immunized donor animals in chronic EAMG.Citation1 Alternatively, passive EAMG can be induced via administration of monoclonal antibodies (IgG1 or IgG2a) which are directed to the AChR α-subunit, either derived from AChR-immunized animalsCitation36 or cell line culture supernatants,Citation37 which trigger EAMG symptoms in the recipient animals within 24 hours. This EAMG induction protocol has proved to be the perfect model not only for characterizing the immunopathogenesis of AChR-MG and for testing the pathogenicity of other antigen-targeted auto-Abs but also for evaluating the therapeutic potentials of drugs specifically aimed to reduce auto-Ab pathogenic effects.Citation38

Adoptive transfer of EAMG

EAMG can be induced via the transplantation of human tissues or cells in severe combined immunodeficiency (SCID) mice, lacking mature B- and T-cells and tolerating xenografts.Citation39,Citation40 Published studies show that SCID mice engrafted with thymus tissue fragments of MG patients produce human anti-mouse AChR antibodies 1–2 weeks after transplantation, demonstrating that a myasthenic thymus contains all the cellular components required for producing auto-Abs and maintaining their synthesis for at least 11 weeks after transplantation.Citation39 Similarly, SCID mice injected with peripheral blood lymphocytes, derived from MG patients, show the typical signs of the human disease, which is characterized by circulating anti-AChR antibodies and human IgG deposits at the NMJ, and demonstrate that only CD4+ T-cells, and not CD8+ T-cells, are necessary for the pathogenesis of the disease.Citation40 Finally, clinical manifestations of MG are also observed in AChR-immunized SCID mice and simultaneously injected with peripheral blood lymphocytes isolated from healthy controls.Citation41

Newly emerged auto-antigens in MG and new EAMG models

The majority of patients with generalized MG (85%) and with ocular MG (50%) develop antibodies against AChR, usually belonging to IgG1 and IgG3 isotypes; these auto-Abs can be detected by the standard radioimmunoassay method.Citation1 In ~40% of MG patients without anti-AChR antibodies (AChR-negative MG), antibodies directed to a postsynaptic muscle-specific tyrosine kinase (MuSK) can be detected,Citation42 predominantly of the IgG4 type.Citation43,Citation44 The clinical disease, which is characterized by bulbar and facial muscle weakness and extreme fatigue, can be difficult to treat in an effective manner. MuSK auto-Abs affect the NMJ dispersing AChR clusters. Indeed, MuSK together with neural agrin, low-density lipoprotein receptor-related protein 4 (LRP4), downstream of tyrosine kinase 7, and rapsyn is crucial in stabilizing postsynaptic AChRs clusters.Citation45 Experimental animals actively immunized with MuSK (active MuSK EAMG) develop MuSK auto-Abs and muscle weakness, which are accompanied by reduced postsynaptic AChR numbers, decremented amplitudes of endplate potentials, and failure of neuromuscular transmission.Citation46Citation48 Although MuSK immunization stimulates the production of all antibody isotypes, noncomplement-fixing IgG1, the mouse analog of human IgG4, is the dominant anti-MuSK Ig isotype in both sera and NMJs.Citation47 Moreover, MuSK-immunized mice sera and supernatants of cultured lymph node cells show high levels of interleukin (IL)-4 and IL-10, suggesting a role for Th2-type cells in the activation of anti-MuSK IgG1.Citation49 Similar results have been found in mice injected with IgG from MG patients positive for MuSK auto-Ab (passive transfer of MuSK EAMG).Citation9,Citation45,Citation51,Citation52 Thanks to these models, besides enlightening the mechanisms by which MuSK antibodies disrupt synaptic function at the NMJ, clues for the pathogenesis of IgG4-related diseases have been given, which might in turn be of great value for developing specific therapies.Citation47,Citation52,Citation53

Moreover, recent studies in MG patients double negative for anti-AChR and anti-MuSK have identified auto-Abs against LRP4, an agrin receptor critical for NMJ formation.Citation54Citation58 LRP4 auto-Abs have been demonstrated to be pathogenic;Citation59,Citation60 indeed, mice immunized with the extracellular domain of LRP4 produce anti-LRP4 antibodies and show MG-like symptoms.Citation60 Moreover, mouse anti-LRP4 antibodies inhibit agrin-induced MuSK activation and AChR clustering, thus showing potential pathophysiological mechanisms. Indeed, passive transfer experiments confirmed the pathogenicity of LRP4 antibodies and demonstrated that LRP4, which contributes to NMJ maintenance, is an auto-antigen in MG.Citation60

EAMG models for the investigation of therapeutic approaches

The main aim of experimental MG is to understand the pathological mechanisms of the disease and to investigate potential new therapiesCitation16 in order to flank or replace the actual immunosuppressive therapies. Indeed, current conventional therapies for MG are not effective in a proportion of patients, and immunosuppressive drugs induce numerous side effects; hence, new approaches are necessary to suppress antigen-specific immune cells and reduce the undesired effects usually observed following the inhibition of the whole immune system in MG patients.

The recently studied new interventions on the EAMG model may be subdivided into five major classes on the basis of their general mechanism of action: 1) treatments to induce peripheral tolerance, via tolerizing agents or immunomodulating cellular delivery; 2) treatments to induce immunomodulation via biological agents, such as cytokines or probiotics; 3) newly developed pharmacological approaches; 4) inhibitors of complement activity; and 5) molecular biology approaches such as RNA/microRNA interference. The following sections serve as a general excursus on those approaches, which are also schematically summarized in .

Table 3 Therapeutic approaches investigated via EAMG

Induction of peripheral tolerance

The most supported pathogenetic hypothesis for MG induction is the loss of self-tolerance in the thymus,Citation7,Citation61 which induces the production of AChR-specific auto-reactive CD4+ T-cell and consequently anti-AChR auto-Abs. The development of EAMG seems to be caused by a disruption of T-cell subset balance, which is characterized by an increase in Th1/Th17 cells and a decrease in Th2/regulatory T-cells (Tregs).Citation62 The immune response is normally kept under control by a peripheral immune surveillance system, which deletes self-reactive T-cells escaped from thymic selection. This immune surveillance is maintained in a steady state by the balance between different CD4+ T-cell subsets, breaking that balance leads to failure of immune surveillance.Citation63 Therefore, the goal of some therapeutic strategies is the induction of peripheral tolerance and reestablishment of the balance between Th1/Th2/Th17/Treg cell responses.

Nasal administration to myasthenic rats of human recombinant fragments of the AChR α-subunit, including the whole extracellular domain of AChR (Hα1-210), induces tolerance to the AChR. Such treatment prevents the development and suppresses the progression of EAMG, inhibiting antigen-specific T-cell proliferative responses and reducing the levels of anti-AChR antibodies.Citation64,Citation65 Similar tolerization effect is achieved in EAMG rats, which is orally treated during the acute and chronic phase with a human recombinant extracellular domain of AChR α-subunit (Hα1-205), inducing a shift from Th1 to Th2 cell response and from IgG2 to IgG1 antibody isotypes.Citation65 Analogous evidence is observed after the oral administration of AChR epitopes.Citation66Citation68 Indeed, the nasal administration of the Tα146–162 synthetic peptide, corresponding to the immunodominant epitope of TAChR α-subunit, to mice immunized with TAChR reduces T-cell proliferative response to TAChR and Tα146–162 peptides, the production of pathogenic antibodies, and the loss of muscle AChR content, thus modulating ongoing disease.Citation68

Therapeutic potential of immunomodulating dendritic cells

Another therapeutic strategy, which is designed to suppress the antigen-specific response in MG, involves the cellular components that participate in the control of peripheral tolerance to the AChR. Dendritic cells (DCs) are specialized antigen-presenting cells that are able to initiate a primary immune response by activating naïve T-cells.Citation69 DCs first recognize and process antigens in the periphery, then migrate to lymphoid organs where they expose the processed peptides to naïve T-cells. In vitro, not only the maturation and function of DCs can be regulated in different pathways: the upregulation of costimulatory molecules (CD83, CD40, CD80, CD86) and major histocompatibility complex class II on DCs is essential to activate T-cells,Citation69 but also immature DCs can tolerize T-cells.Citation70,Citation71 Depending on their maturation and differentiation state, DCs can acquire either a tolerogenic or an immunogenic activity.Citation69 In the absence of inflammation, immature DCs control peripheral tolerance by promoting Treg differentiation; instead, inflammatory conditions provoke morphological and functional changes leading to mature DCs that are able to induce the activation of effector T-cells.Citation10,Citation72

Several published studies show that DC vaccine can induce tolerance and protect from autoimmune diseases. In this line, many therapeutic strategies aim to modulate in vitro DC maturation and differentiation with anti-inflammatory agents or growth factors. For instance, DC isolated from spleens of healthy rats and conditioned in vitro with transforming growth factor beta 1 (TGF-β1) can be arrested at their immature differentiation stage, and their administration to AChR-immunized rats reduces the severity of EAMG symptoms.Citation73 In addition, spleen-derived DCs exposed to IL-10 in vitro induce EAMG amelioration when injected intraperitoneally into AChR-immunized rats, due to the DC modulation of T- and B-cell responses.Citation74 Moreover, healthy animals injected with bone marrow DCs pulsed in vitro with AChR, and subsequently immunized with AChR, do not show clinical signs of EAMG, thus confirming the role of immature DCs in the control of peripheral tolerance.Citation75Citation77 Indeed, DCs engineered to present AChR epitopes can specifically target AChR-specific T-cells, resulting in the reduction in both AChR-T-cell responses and anti-AChR antibodies. Besides TGF-β and IL-10, also the treatment with granulocyte-macrophage colony-stimulating factor can suppress the development of EAMG manifestations when administered to mice before AChR immunization,Citation78 thanks to the activation of specific DC subpopulations and expansion of the Treg compartment.Citation78

Similarly, also DCs incubated with atorvastatin, a statin therapeutically employed for reducing cardiovascular diseases which is known to show a strong immunomodulatory activityCitation79,Citation80 especially inhibiting the maturation and function of antigen-presenting cells, acquire an immature tolerizing phenotype. Indeed, spleen-derived DCs extracted from ongoing EAMG rats can be in vitro tolerized by statin treatment and can improve clinical symptoms when injected into recipient EAMG rats, inducing an increase in CD4+CD25+ Tregs and Foxp3 expression, while decreasing lymphocyte proliferation and shifting cytokine profile from Th1/Th17- to Th2-type cytokines.Citation81,Citation82 Tolerized DC-induced Th cell shift is crucial for the amelioration of EAMG symptoms, indeed also bone-marrow-derived DCs, RelB silenced and pulsed with Tα146–162, are able to suppress EAMG progression in mice, by inducing a positive shift in favor of Th2/Treg responses.Citation83 Finally, the tolerizing function of DCs can be exploited even to induce specific killing of AChR-targeted effector T-cells, as in the “Guided Missile” strategy in which genetically engineered DCs simultaneously target and eliminate the individual’s unique AChR-specific T-cell repertoire, by presenting AChR epitopes and expressing Fas ligand.Citation84 Eventually, very recent data indicate that aside from the classical cell therapy, a tolerizing effect can be obtained in EAMG also via the delivery of exosomes produced by immature DCs.Citation85

Regulatory cells and suppressor cells as therapeutic approach

Another therapeutic approach acts directly on the immune cells devoted to T- and B-cell responses, that is, CD4+CD25+ Treg and myeloid-derived suppressor cells (MDSCs). Tregs arise in the thymus, which represent 5%–10% of CD4+ T-cells in the periphery and constitutively express CD25 molecule (IL-2 receptor α-chain). They play an essential role in the maintenance of peripheral tolerance, suppressing the proliferation and cytokine production of CD4+ effector T-cells.Citation86Citation88 Myasthenic patients often show a defect in Treg subset: the number of Tregs is reduced in the peripheral blood,Citation89,Citation90 while their suppressive function, but not their number, is altered in the thymus.Citation91,Citation92 Thus, the restoration or expansion of the Treg compartment can represent an important therapeutic tool for the disease. Tregs can be in vitro induced from CD4+ T-cells from spleens of healthy rats, which are stimulated with anti-CD3 and anti-CD28 antibodies in the presence of TGF-β and IL-2. Such induced CD4+CD25+ Tregs, sharing identical functional features with naturally occurring Tregs, can suppress clinical signs of EAMG in AChR-immunized rats.Citation93 Similarly, also naturally occurring Tregs, purified from spleens of healthy rats, can modulate EAMG progression when administered to AChR-immunized rats,Citation94 through the reduction in specific T-cell proliferation, decrease in pathogenic auto-Abs titer, and increase in muscle AChR content.Citation94

Besides Tregs, also MDSCs derived from myeloid progenitors have a therapeutic effect in ongoing EAMG. These cells, originally identified in tumors,Citation95 inhibit both innate and adaptive immunity,Citation96,Citation97 seemingly via antigen-specific immunosuppression in peripheral organs. Adoptive transfer of these MDSCs is able to reverse EAMG progression, specifically suppressing AChR-specific T-cell responses, decreasing serum anti-AChR IgGs, reducing complement activation at the NMJ, and also directly inhibiting B-cells through multiple mechanisms, including PGE2, inducible nitric oxide synthase, and arginase.Citation98

Other cell therapies for EAMG: mesenchymal stem cells and B10 cells

A further candidate cell therapy for human MG is represented by bone marrow stromal cells, which can modulate the functions of T- and B-cells, natural killer cells, and DCs. In particular, bone marrow stromal cells inhibit lymphocyte responses to different stimuli by the secretion of immunosuppressive factors.Citation99,Citation100 Indeed, stromal cells, derived from healthy rats, induce a strong reduction in disease severity when injected into EAMG rats at clinical onset. Such treatment results in the secretion of immunosuppressive factors, such as indoleamine 2,3-dioxygenase and TGF-β, thus suppressing both T- and B-cell responses to the immunizing antigen and production of modulating cytokine and decreasing Th1 and Th17 subsets while increasing Th2 and Treg subpopulations.Citation99,Citation100

In addition, IL-10–competent B-cells, known as B10, characterized by the expression of CD5 and high CD1 (CD1dhiCD5+) can prevent or suppress EAMG, either indirectly through low-dose granulocyte-macrophage colony-stimulating factor administration, which increases the number of circulating B10 cells, or directly by adoptive transfer of CD1dhiCD5+ B-cells. B10 cells alter T-cell cytokine profile, downregulate mature DC markers, and expand Treg compartment, while directly blocking B-cell proliferation and auto-Ab production in an IL-10–dependent manner.Citation101

Immunomodulation through biological agents

Besides tolerization induction and cell suppression, direct modulation of key immunological factors can be pivotal in the therapy of autoimmunity. Indeed, cytokines and costimulatory molecules are important in autoimmune pathogenesis, as shown in EAMG rats treated with antibodies either to proinflammatory, costimulatory factors or to chemokines, which suppress the disease but acting via different complementing mechanisms. For instance, the stimulatory molecule CD40L, studied by EAMG experiments in CD40L knock-out (KO) mice or via anti-CD40L antibodies injection, is fundamental for T- and B-cell engagement, activation, and EAMG induction.Citation102 Similarly, EAMG experiments have proved the role of the pleiotropic inflammatory cytokine IL-6 in B/T-cell function and autoimmune reaction maintenance. Indeed, the administration of anti-IL-6 antibodies suppresses EAMG symptoms during both the acute and the chronic phase, thanks to an induced shift in favor of Tregs, instead of Th17 cells, accompanied by reduced numbers of B-cells.Citation103 Similarly to IL-6 blockade, also the inhibition of interferon γ inducible protein 10 (IP-10), a highly inducible chemoattractant for activated T-cells leads to immunomodulation and the attenuation of EAMG symptoms, when either anti-IP-10 antibodies or IP-10 receptor (CXCR3) antagonists are administered.Citation104,Citation105 Moreover, the cytokine IL-9 and IL-18 are crucial for EAMG development. Indeed, IL-9 neutralization via targeted antibody ameliorates the symptoms of EAMG, decreasing effector T-cells and altering humoral responses,Citation106 and IL-18 KO mice are resistant to the disease.Citation107 Similarly, anti-IL-18 antibodies suppress EAMG, increasing TGF-β levels while decreasing AChR-reactive Th1-type cellular responses.Citation108 An increased production of TGF-β is also observed when IL-2/anti-IL-2 mAb complexes are administered, which inhibit the development of EAMG, mediating the expansion of CD4+CD25+Foxp3+ Treg cells and the conversion of peripheral and circulating CD4+CD25 T-cells in Treg, leading to a shift of Th1/Th2 ratio in favor of a Th2 phenotype.Citation109 Aside from monoclonal antibody blockade, other strategies have been tested to efficiently inhibit proinflammatory cytokines in EAMG, such as the use of a specific caspase-1 inhibitor, which blocks caspase-1-mediated cleavage of both IL-1β and IL-18 precursors into their functional forms, thus ameliorating EAMG symptoms.Citation33

Finally, also less potent immunomodulatory agents, such as live probiotic bacteria administered orally, may have a beneficial role on EAMG symptoms, when given following a prophylactic schedule, through the generation of regulatory DCs that express increased levels of IL-10 and TGF-β and are able to convert CD4+ T-cells into CD4+Foxp3+ Treg.Citation110

Pharmacological immunotherapy

Besides approved current pharmacological therapies for the treatment of MG,Citation111 other emerging drugs, such as bortezomib and pixantrone (BBR2778) (PIX), show excellent efficacy in suppressing EAMG.Citation112Citation114 Bortezomib, an inhibitor of proteasomes, which depletes both short- and long-lived plasma cells, was shown to induce apoptosis in bone marrow cells and reduce the amount of plasma cells in EAMG rats, resulting in reduced anti-AChR auto-Ab titers, improved neuromuscular transmission, and decreased clinical symptoms.Citation112 Differently from bortezomib, PIX is an antineoplastic drug, which is structurally related to mitoxantrone. Both drugs are DNA intercalants and topoisomerase II inhibitors, but PIX is characterized by a reduced cardiotoxicity compared with mitoxantrone. When administrated to AChR-immunized rats via different treatment schedules, either preventive (before clinical onset) or therapeutic protocol (at overt clinical symptoms), PIX is able to suppress antigen-specific T-cell proliferative responses in a dose-dependent manner, reducing the levels of pathogenic antibodies and increasing muscle AChR content.Citation113 Interestingly, even if clinical symptoms could be improved only by repeated PIX administrations, allowing stable serum drug levels, a single administration is already able to suppress AChR-specific immune responses in primed rats, inhibiting only proliferating T-cells without impairing DC differentiation and B-cell viability.Citation114 Another pharmacological treatment recently tested in EAMG is the all-trans retinoic acid (ATRA), a vitamin A metabolite with diverse immunomodulatory actions, which is used therapeutically in the treatment of some autoimmune diseases. The study in the EAMG model allowed deeper inside in its mechanism of action, which is still unknown. Intraperitoneal injection of ATRA in EAMG rats ameliorated clinical symptoms, reduced total anti-AChR auto-Abs titers, and changed follicular T-cells levels, thus restoring the Th1/Th2/Th17/Treg balance. ATRA altered the Th cell distribution in EAMG animals resulting in a reduction in Th1/Th17/follicular T helper cells (Tfh) cells and an increase in Th2/Treg/regulatory follicular T cells (Tfr) cell types.Citation33 These results highlight the importance of EAMG in testing pharmacological drugs to assess their efficacy and to decipher their mechanism of action, offering new possibilities for the treatment of human MG.

Prevention of complement-mediated NMJ destruction

The role of complement at the level of the NMJ has been extensively studied in EAMG models. Indeed, complement activation plays an essential role in the destruction of the postsynaptic membrane (reviewed in the study by Tuzun and ChristadossCitation115). Although C3a and C5a promote inflammation by recruiting and activating phagocytic cells, C3b and C4b simultaneously lead to muscle membrane lysis.Citation116,Citation117 Depleting the complement cascade via treatment with cobra venom factor decreases the formation of anti-AChR antibodies/AChR complexes and ameliorates the acute phase of EAMG in rats.Citation118 More specifically, the effects of several complement components have been analyzed in various transgenic models. For instance, C5-deficient mice show a mild EAMG incidence and little decrease in muscle.Citation119 Besides, treatment with soluble recombinant form of human complement receptor 1 (sCR1) reduces EAMG severity.Citation38 The role of the MAC has also been studied in acute, passively transferred EAMG in Wistar rats, where administered anti-C6 Fab leads to the inhibition of MAC formation and suppression of EAMG clinical and electrophysiological signs.Citation32 Regulatory proteins that inhibit MAC formation (such as MIRL-CD59 that inhibits MAC assembly) and control the activation of the complement cascade (such as the decay-accelerating factor or CD55, which inactivates C3 and C5 convertase enzymes) represent crucial players in EAMG development, and their modulation has been a target of recent experimental approaches aimed at complement depletion.Citation117,Citation120Citation123 Pharmacological inhibition of the complement activation pathway is an alternative strategy to depleting approaches. Indeed, the administration of rEV576, a specific C5 complement component inhibitor, is able to reduce the severity of passive transfer of EAMG and the progression of acute experimental MG, reducing C9 deposits at the NMJ.Citation124 Another strategy is that of increasing the resistance of the NMJ to complement-mediated lysis. An increased interaction between rapsyn and AChR may stabilize the receptor molecules, conferring greater NMJ resistance, leading to minor AChR loss and muscle weakness in acute EAMG.Citation125 Thus, the overexpression of rapsyn may represent a further therapeutic option, but unfortunately the increased expression of rapsyn alone is not able to efficiently anchor the AChR to the postsynaptic membrane in chronic EAMG, once the destruction of the NMJ has already occurred.Citation126

MicroRNA interference as future gene therapy

MicroRNAs have been shown to act as regulators of gene expression and play an important role in immune homeostasis and autoimmunity susceptibility.Citation127 Indeed, it has been recently shown that miR-146a is upregulated in activated B-cells in response to rat AChR 97–116 peptide, and this upregulation can be attenuated by miR-146a-specific antagonist.Citation128 Consequently, miR-146a systemic silencing ameliorates EAMG symptoms in mice via B-cell blocking, including decreased production of anti-R97–116 antibodies, class switching, reduced numbers of plasma cells and memory B-cells and B-1 cells.Citation128 Similar data were published regarding another miRNA, miR-155, that is upregulated in AChR-stimulated B-cells.Citation129 The systemic delivery of a miR-155 inhibitor conjugated to anti-CD20 single-chain antibody impairs B-cell signaling and reduces EAMG autoimmune reaction in mice.Citation129 Conversely, it was recently demonstrated that another miRNA, miR-145, is downregulated in peripheral monocytes from EAMG rats, especially in CD4+ T-cells, and its in vitro upregulation in a DC-T-cell coculture setup suppresses Th 17 cell response.Citation130 Finally, the administration of lentiviral miR-145 during ongoing EAMG decreased the severity of symptoms and production of IL-17.Citation130 Altogether, these results provide insights into the role of miRNA in EAMG pathogenesis and open a new prospective for EAMG/MG gene therapy.

Limitations of the animal model

As discussed so far, the EAMG model has been extensively used to analyze various aspects of MG pathology and experimental therapies. Nevertheless, there are limitations in using this animal model. For instance, EAMG can be easily affected by the induction procedure, despite the publication of several detailed guidelines which should help obtaining high standard disease models.Citation38,Citation131,Citation132 Indeed, the chosen experimental parameters and procedures affect the disease time course, incidence, and severity. For example, strong EAMG clinics in susceptible strains, or using potent adjuvants, mean unbearable animal suffering and increased number of animal deaths, which in turn damage the statistical power of the results. On the other side, mild EAMG scores are scarcely effective in demonstrating beneficial treatment effects.Citation131

Moreover, despite faithfully reproducing many aspects of the human pathology, the experimental model still presents several discrepancies with the human disease (), such as the absence of a spontaneous disease in experimental animals, accounting for a strongly different genetic background. Besides, the role of the thymus as the main site for initiating, sustaining, and maintaining the diseaseCitation7,Citation61,Citation133 has so far not been paralleled in the animal models, despite only few old reportsCitation6,Citation134,Citation135 and very recent data indicating a pathogenic role of thymic epithelial cells and DCs in the myasthenic rat, in contributing to developing an active inflammatory milieu.Citation10

Eventually, the most relevant limitation of the animal model is being an animal model. Mice and rats are bred in controlled facilities and experiments are always performed on syngeneic animals. There is very little or no involvement of genetic drift or even environmental exposure. In the human disease, instead, environmental factors, such as viral or microbial agents, play a pivotal role in the pathogenesis of autoimmunity.Citation61,Citation133

Finally, in an ever more sensible and ethically correct research environment, we must bear in mind that, when possible, alternative strategies must always be pursued. EAMG has been fundamental in discovering the pathogenic mechanisms of MG and in developing several therapeutic strategies, at the expense of suffering animals. Researchers must now make an effort to set up and create alternative methodologies such as complex cellular cultures for validating pathogenic hypothesisCitation10 and new treatments.Citation136

Conclusion

MG is a rare chronic autoimmune disease involving different compartments of the organism: the immune system and the NMJ. In vitro models, which are able to fully represent complex pathologies where more tissues and systems are involved, are not yet available but more effort should be given in order to obtain complex in vitro setups mimicking the autoimmune inflammatory milieu. The EAMG model allows the investigation of both the muscle compartment and the immune system, focusing on the pathogenic mechanisms and the clinical outcome. Thus, EAMG is an essential tool for understanding pathogenic mechanisms and investigating new therapies, which may later be translated to clinical trials.

Unfortunately, several treatment strategies effective in EAMG failed when transferred to the human disease. Indeed, EAMG has a less complex pathogenesis compared with the human disease, as it is performed on syngeneic animals that bred in controlled conditions, especially in terms of genetic predisposition and environmental factors such as the exposure to viral antigens. Nevertheless, EAMG has been of great value first in understanding the pathogenic mechanisms of auto-Abs and in proving the efficacy of both pharmacological treatments and cell therapy strategies with results that encourage the investigation of the human disease. A similar approach is necessary for any new immunosuppressive or immunomodulating compound of potential interest. Always considering Russell and Burch’s 3R rule for the replacement of experimental animal procedures with alternative methods, the reduction in the number of used animals, and refinement of the animal conditions,Citation137 we cannot forget that complex diseases, of which MG is a prototypical representative, need to be addressed with preclinical research in order to obtain more efficient therapies. Despite the evident differences between EAMG and MG, most importantly the axiom that the former cannot spontaneously arise in laboratory animals, the experimental approach remains an unavoidable and irreplaceable method to discover new efficient therapies.

Acknowledgments

We regret that due to space constraints, we could not include the essential work of many investigators and colleagues. We acknowledge the Italian Ministry of Health Grant PE-2011-02346818 for financial support.

Disclosure

The authors report no conflicts of interest in this work.

References

  • LindstromJMSeyboldMELennonVAAntibody to acetylcholine receptor in myasthenia gravis. Prevalence, clinical correlates, and diagnostic valueNeurology19762610541059988512
  • TrotterJLRingelSPCookJDMorphologic and immunologic studies in experimental autoimmune myasthenia gravis and myasthenia gravisNeurology1977271120112473154
  • MeriggioliMNMyasthenia gravis with anti-acetylcholine receptor antibodiesFront Neurol Neurosci2009269410819349707
  • TzartosSJMorelEEfthimiadisAFine antigenic specificities of antibodies in sera from patients with D-penicillamine-induced myasthenia gravisClin Exp Immunol19887480862464451
  • Newsom-DavisJWillcoxNCalderLThymus cells in myasthenia gravis selectively enhance production of anti-acetylcholine-receptor antibody by autologous blood lymphocytesN Engl J Med1981305131313187290153
  • MelmsALutherCStoeckleCThymus and myasthenia gravis: antigen processing in the human thymus and the consequences for the generation of autoreactive T cellsActa Neurol Scand Suppl2006183121316637920
  • CavalcantePLe PanseRBerrih-AkninSThe thymus in myasthenia gravis: site of “innate autoimmunity”?Muscle Nerve20114446748421922466
  • HoedemaekersACvan Breda VriesmanPJDe BaetsMHMyasthenia gravis as a prototype autoimmune receptor diseaseImmunol Res1997163413549439759
  • VincentALeiteMIFarrugiaMEMyasthenia gravis seronegative for acetylcholine receptor antibodiesAnn N Y Acad Sci20081132849218567857
  • CordiglieriCMaroldaRFranziSInnate immunity in myasthenia gravis thymus: pathogenic effects of Toll-like receptor 4 signaling on autoimmunityJ Autoimmun201452748924397961
  • DrachmanDBAutonomic “myasthenia”: the case for an autoimmune pathogenesisJ Clin Invest200311179779912639983
  • VincentAUnravelling the pathogenesis of myasthenia gravisNat Rev Immunol2002279780412360217
  • VincentADrachmanDBMyasthenia gravisAdv Neurol20028815918811908224
  • EngelAGLambertEHHowardFMImmune complexes (IgG and C3) at the motor end-plate in myasthenia gravis: ultrastructural and light microscopic localization and electrophysiologic correlationsMayo Clin Proc197752267280870771
  • ToykaKVBrachmanDBPestronkAMyasthenia gravis: passive transfer from man to mouseScience19751903973991179220
  • FuchsSArichaRReuveniDExperimental autoimmune myasthenia gravis (EAMG): from immunochemical characterization to therapeutic approachesJ Autoimmun201454515924970384
  • PatrickJLindstromJCulpBStudies on purified eel acetylcholine receptor and anti-acetylcholine receptor antibodyProc Natl Acad Sci U S A197370333433384128544
  • DauPCPlasmapheresis in idiopathic inflammatory myopathy. Experience with 35 patientsArch Neurol1981385445527271533
  • AntozziCGemmaMRegiBA short plasma exchange protocol is effective in severe myasthenia gravisJ Neurol19912381031071856735
  • Conti-FineBMMilaniMKaminskiHJMyasthenia gravis: past, present, and futureJ Clin Invest20061162843285417080188
  • EngelAGFumagalliGMechanisms of acetylcholine receptor loss from the neuromuscular junctionCiba Found Symp1982901972246923808
  • GomezCMRichmanDPAnti-acetylcholine receptor antibodies directed against the alpha-bungarotoxin binding site induce a unique form of experimental myastheniaProc Natl Acad Sci U S A198380408940936575398
  • BaggiFAntozziCToscaniCAcetylcholine receptor-induced experimental myasthenia gravis: what have we learned from animal models after three decades?Arch Immunol Ther Exp (Warsz)201260193022159475
  • ChristadossPPoussinMDengCAnimal models of myasthenia gravisClin Immunol200094758710637092
  • LinkHXiaoBGRat models as tool to develop new immunotherapiesImmunol Rev200118411712811918681
  • BaggiFAnnoniAUbialiFBreakdown of tolerance to a self-peptide of acetylcholine receptor alpha-subunit induces experimental myasthenia gravis in ratsJ Immunol20041722697270314764745
  • BaggiFAnnoniAUbialiFImmunization with rat-, but not Torpedo-derived 97-116 peptide of the AChR alpha-subunit induces experimental myasthenia gravis in Lewis ratAnn N Y Acad Sci200399839139414592903
  • BermanPWPatrickJExperimental myasthenia gravis. A murine systemJ Exp Med19801512042237350247
  • BermanPWPatrickJLinkage between the frequency of muscular weakness and loci that regulate immune responsiveness in murine experimental myasthenia gravisJ Exp Med19801525075206774045
  • LindstromJExperimental autoimmune myasthenia gravisJ Neurol Neurosurg Psychiatry1980435685766249892
  • BieseckerGGomezCMInhibition of acute passive transfer experimental autoimmune myasthenia gravis with Fab antibody to complement C6J Immunol1989142265426592703710
  • XieXMuLYaoXATRA alters humoral responses associated with amelioration of EAMG symptoms by balancing Tfh/Tfr helper cell profilesClin Immunol201314816217623773919
  • WangCCLiHZhangMCaspase-1 inhibitor ameliorates experimental autoimmune myasthenia gravis by innate dendric cell IL-1-IL-17 pathwayJ Neuroinflammation20151211826071315
  • LiNMuLWangJActivation of the adenosine A2A receptor attenuates experimental autoimmune myasthenia gravis severityEur J Immunol2012421140115122539289
  • LiNWangGYaoXAdenosine receptor expression in a rat model of experimental autoimmune myasthenia gravisCell Immunol201429021722525086239
  • TzartosSHochschwenderSVasquezPPassive transfer of experimental autoimmune myasthenia gravis by monoclonal antibodies to the main immunogenic region of the acetylcholine receptorJ Neuroimmunol1987151851943495549
  • PiddlesdenSJJiangSLevinJLSoluble complement receptor 1 (sCR1) protects against experimental autoimmune myasthenia gravisJ Neuroimmunol1996711731778982117
  • KusnerLLLosenMVincentAGuidelines for pre-clinical assessment of the acetylcholine receptor – specific passive transfer myasthenia gravis model-Recommendations for methods and experimental designsExp Neurol201527031025743217
  • SchonbeckSPadbergFHohlfeldRTransplantation of thymic autoimmune microenvironment to severe combined immunodeficiency mice. A new model of myasthenia gravisJ Clin Invest1992902452501634612
  • WangZYKarachunskiPIHowardJFJrMyasthenia in SCID mice grafted with myasthenic patient lymphocytes: role of CD4+ and CD8+ cellsNeurology19995248449710025776
  • MartinoGDuPontBLWollmannRLThe human-severe combined immunodeficiency myasthenic mouse model: a new approach for the study of myasthenia gravisAnn Neurol19933448568517679
  • HochWMcConvilleJHelmsSAuto-antibodies to the receptor tyrosine kinase MuSK in patients with myasthenia gravis without acetylcholine receptor antibodiesNat Med2001736536811231638
  • McConvilleJVincentADiseases of the neuromuscular junctionCurr Opin Pharmacol2002229630112020474
  • OhtaRMotomuraMAutoantibodies detected in acetylcholine receptor antibody-negative myasthenia gravisRinsho Byori20146225526024800501
  • GhazanfariNFernandezKJMurataYMuscle specific kinase: organiser of synaptic membrane domainsInt J Biochem Cell Biol20114329529820974278
  • JhaSXuKMarutaTMyasthenia gravis induced in mice by immunization with the recombinant extracellular domain of rat muscle-specific kinase (MuSK)J Neuroimmunol200617510711716697051
  • PhillipsWDChristadossPLosenMGuidelines for pre-clinical animal and cellular models of MuSK-myasthenia gravisExp Neurol2015270294025542979
  • BanJPhillipsWDMouse models of myasthenia gravisCurr Pharm Des2015212468248625777761
  • UlusoyCKimETuzunEPreferential production of IgG1, IL-4 and IL-10 in MuSK-immunized miceClin Immunol201415115516324589747
  • ColeRNReddelSWGervasioOLAnti-MuSK patient antibodies disrupt the mouse neuromuscular junctionAnn Neurol20086378278918384168
  • ColeRNGhazanfariNNgoSTPatient autoantibodies deplete postsynaptic muscle-specific kinase leading to disassembly of the ACh receptor scaffold and myasthenia gravis in miceJ Physiol20105883217322920603331
  • MorschMReddelSWGhazanfariNPyridostigmine but not 3,4-diaminopyridine exacerbates ACh receptor loss and myasthenia induced in mice by muscle-specific kinase autoantibodyJ Physiol20135912747276223440963
  • MorschMReddelSWGhazanfariNMuscle specific kinase autoantibodies cause synaptic failure through progressive wastage of postsynaptic acetylcholine receptorsExp Neurol201223728629522789393
  • PevznerASchoserBPetersKAnti-LRP4 autoantibodies in AChR- and MuSK-antibody-negative myasthenia gravisJ Neurol201225942743521814823
  • ZhangBTzartosJSBelimeziMAutoantibodies to lipoprotein-related protein 4 in patients with double-seronegative myasthenia gravisArch Neurol20126944545122158716
  • ZongYZhangBGuSStructural basis of agrin-LRP4-MuSK signalingGenes Dev20122624725822302937
  • HiguchiOHamuroJMotomuraMAutoantibodies to low-density lipoprotein receptor-related protein 4 in myasthenia gravisAnn Neurol20116941842221387385
  • MotomuraMHiguchiOProgress of myasthenia gravis: discovery of Lrp4 antibodiesRinsho Shinkeigaku2012521303130523196599
  • BarikALuYSathyamurthyALRP4 is critical for neuromuscular junction maintenanceJ Neurosci201434138921390525319686
  • ShenCLuYZhangBAntibodies against low-density lipoprotein receptor-related protein 4 induce myasthenia gravisJ Clin Invest20131235190520224200689
  • CavalcantePCufiPMantegazzaREtiology of myasthenia gravis: innate immunity signature in pathological thymusAutoimmun Rev20131286387423535157
  • MuLSunBKongQDisequilibrium of T helper type 1, 2 and 17 cells and regulatory T cells during the development of experimental autoimmune myasthenia gravisImmunology2009128e826e83619740344
  • SchaffertHPelzASaxenaAIL-17-producing CD4(+) T cells contribute to the loss of B-cell tolerance in experimental autoimmune myasthenia gravisEur J Immunol2015451339134725676041
  • BarchanDSouroujonMCImSHAntigen-specific modulation of experimental myasthenia gravis: nasal tolerization with recombinant fragments of the human acetylcholine receptor alpha-subunitProc Natl Acad Sci U S A1999968086809110393952
  • ImSHBarchanDFuchsSSuppression of ongoing experimental myasthenia by oral treatment with an acetylcholine receptor recombinant fragmentJ Clin Invest19991041723173010606626
  • MaitiPKFefermanTImSHImmunosuppression of rat myasthenia gravis by oral administration of a syngeneic acetylcholine receptor fragmentJ Neuroimmunol200415211212015223243
  • OkumuraSMcIntoshKDrachmanDBOral administration of acetylcholine receptor: effects on experimental myasthenia gravisAnn Neurol1994367047137979216
  • BaggiFAndreettaFCaspaniEOral administration of an immunodominant T-cell epitope downregulates Th1/Th2 cytokines and prevents experimental myasthenia gravisJ Clin Invest19991041287129510545527
  • BanchereauJSteinmanRMDendritic cells and the control of immunityNature19983922452529521319
  • JonuleitHSchmittESteinbrinkKDendritic cells as a tool to induce anergic and regulatory T cellsTrends Immunol20012239440011429324
  • KruegerTWohlrabUKluckenMAutoantigen-specific protection of non-obese diabetic mice from cyclophosphamide-accelerated diabetes by vaccination with dendritic cellsDiabetologia2003461357136512928772
  • RoncaroloMGLevingsMKTraversariCDifferentiation of T regulatory cells by immature dendritic cellsJ Exp Med2001193F5F911208869
  • YarilinDDuanRHuangYMDendritic cells exposed in vitro to TGF-beta1 ameliorate experimental autoimmune myasthenia gravisClin Exp Immunol200212721421911876742
  • DuanRSAdikariSBHuangYMProtective potential of experimental autoimmune myasthenia gravis in Lewis rats by IL-10-modified dendritic cellsNeurobiol Dis20041646146715193302
  • XiaoBGDuanRSLinkHInduction of peripheral tolerance to experimental autoimmune myasthenia gravis by acetylcholine receptor-pulsed dendritic cellsCell Immunol2003223636912914759
  • XiaoBGHuangYMLinkHDendritic cell vaccine design: strategies for eliciting peripheral tolerance as therapy of autoimmune diseasesBioDrugs20031710311112641489
  • LiLSunSCaoXExperimental study on induction of tolerance to experimental autoimmune myasthenia gravis by immature dendritic cellsJ Huazhong Univ Sci Technolog Med Sci20052521521816116977
  • ShengJRLiLGaneshBBSuppression of experimental autoimmune myasthenia gravis by granulocyte-macrophage colony-stimulating factor is associated with an expansion of FoxP3+ regulatory T cellsJ Immunol20061775296530617015715
  • AdamsonPGreenwoodJHow do statins control neuroinflammation?Inflamm Res20035239940314520514
  • GreenwoodJWaltersCEPryceGLovastatin inhibits brain endothelial cell Rho-mediated lymphocyte migration and attenuates experimental autoimmune encephalomyelitisFASEB J20031790590712626426
  • LiXLLiuYCaoLLAtorvastatin-modified dendritic cells in vitro ameliorate experimental autoimmune myasthenia gravis by up-regulated Treg cells and shifted Th1/Th17 to Th2 cytokinesMol Cell Neurosci201356859523541702
  • LiHWangCCZhangMStatin-modified dendritic cells regulate humoral immunity in experimental autoimmune myasthenia gravisMol Cell Neurosci20156828429226311508
  • ZhangYYangHXiaoBEffect of RelB-silenced BMDC pulsed with Talpha146~162 on immunoreaction of T cells primed with TAChRZhong Nan Da Xue Xue Bao Yi Xue Ban201035384420130363
  • SunWAdamsRNMiagkovASpecific immunotherapy of experimental myasthenia gravis in vitro and in vivo: the Guided Missile strategyJ Neuroimmunol2012251253222769060
  • BuNWuHQZhangGLImmature dendritic cell exosomes suppress experimental autoimmune myasthenia gravisJ Neuroimmunol2015285717526198922
  • FehervariZSakaguchiSControl of Foxp3+ CD25+CD4+ regulatory cell activation and function by dendritic cellsInt Immunol2004161769178015520045
  • PiccirilloCAd’HennezelESgouroudisECD4+Foxp3+ regulatory T cells in the control of autoimmunity: in vivo veritasCurr Opin Immunol20082065566218926906
  • SakaguchiSNaturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responsesAnnu Rev Immunol20042253156215032588
  • BattagliaADi SchinoCFattorossiACirculating CD4+CD25+ T regulatory and natural killer T cells in patients with myasthenia gravis: a flow cytometry studyJ Biol Regul Homeost Agents200519546216178275
  • FattorossiABattagliaABuzzonettiACirculating and thymic CD4 CD25 T regulatory cells in myasthenia gravis: effect of immunosuppressive treatmentImmunology200511613414116108825
  • MatsuiNNakaneSSaitoFUndiminished regulatory T cells in the thymus of patients with myasthenia gravisNeurology20107481682020211905
  • BalandinaALecartSDartevellePFunctional defect of regulatory CD4 (+) CD25+ T cells in the thymus of patients with autoimmune myasthenia gravisBlood200510573574115454488
  • ArichaRFefermanTFuchsSEx vivo generated regulatory T cells modulate experimental autoimmune myasthenia gravisJ Immunol20081802132213918250419
  • NessiVNavaSRuoccoCNaturally occurring CD4+CD25+ regulatory T cells prevent but do not improve experimental myasthenia gravisJ Immunol20101855656566720881192
  • YoungMRAquinoSYoungMEDifferential induction of hematopoiesis and immune suppressor cells in the bone marrow versus in the spleen by Lewis lung carcinoma variantsJ Leukoc Biol1989452622732522492
  • HansonEMClementsVKSinhaPMyeloid-derived suppressor cells down-regulate L-selectin expression on CD4+ and CD8+ T cellsJ Immunol200918393794419553533
  • Ostrand-RosenbergSSinhaPMyeloid-derived suppressor cells: linking inflammation and cancerJ Immunol20091824499450619342621
  • LiYTuZQianSMyeloid-derived suppressor cells as a potential therapy for experimental autoimmune myasthenia gravisJ Immunol20141932127213425057008
  • KongQFSunBBaiSSAdministration of bone marrow stromal cells ameliorates experimental autoimmune myasthenia gravis by altering the balance of Th1/Th2/Th17/Treg cell subsets through the secretion of TGF-betaJ Neuroimmunol2009207839119174310
  • KongQFSunBWangGYBM stromal cells ameliorate experimental autoimmune myasthenia gravis by altering the balance of Th cells through the secretion of IDOEur J Immunol20093980080919283707
  • ShengJRQuanSSolivenBCD1d(hi)CD5+ B cells expanded by GM-CSF in vivo suppress experimental autoimmune myasthenia gravisJ Immunol20141932669267725135828
  • ImSHBarchanDMaitiPKBlockade of CD40 ligand suppresses chronic experimental myasthenia gravis by down-regulation of Th1 differentiation and up-regulation of CTLA-4J Immunol20011666893689811359850
  • ArichaRMizrachiKFuchsSBlocking of IL-6 suppresses experimental autoimmune myasthenia gravisJ Autoimmun20113613514121193288
  • FefermanTArichaRMizrachiKSuppression of experimental autoimmune myasthenia gravis by inhibiting the signaling between IFN-gamma inducible protein 10 (IP-10) and its receptor CXCR3J Neuroimmunol2009209879519232748
  • FefermanTMaitiPKBerrih-AkninSOverexpression of IFN-induced protein 10 and its receptor CXCR3 in myasthenia gravisJ Immunol20051745324533115843529
  • YaoXKongQXieXNeutralization of interleukin-9 ameliorates symptoms of experimental autoimmune myasthenia gravis in rats by decreasing effector T cells and altering humoral responsesImmunology201414339640524850614
  • ShiFDTakedaKAkiraSIL-18 directs autoreactive T cells and promotes autodestruction in the central nervous system via induction of IFN-gamma by NK cellsJ Immunol20001653099310410975822
  • ImSHBarchanDMaitiPKSuppression of experimental myasthenia gravis, a B cell-mediated autoimmune disease, by blockade of IL-18FASEB J2001152140214811641240
  • LiuRZhouQLa CavaAExpansion of regulatory T cells via IL-2/anti-IL-2 mAb complexes suppresses experimental myastheniaEur J Immunol2010401577158920352624
  • ChaeCSKwonHKHwangJSProphylactic effect of probiotics on the development of experimental autoimmune myasthenia gravisPLoS One20127e5211923284891
  • MantegazzaRBonannoSCameraGCurrent and emerging therapies for the treatment of myasthenia gravisNeuropsychiatr Dis Treat2011715116021552317
  • GomezAMVrolixKMartinez-MartinezPProteasome inhibition with bortezomib depletes plasma cells and autoantibodies in experimental autoimmune myasthenia gravisJ Immunol20111862503251321239719
  • UbialiFNavaSNessiVPixantrone (BBR2778) reduces the severity of experimental autoimmune myasthenia gravis in Lewis ratsJ Immunol20081802696270318250482
  • MaroldaRRuoccoCCordiglieriCDifferential targeting of immune-cells by pixantrone in experimental myasthenia gravisJ Neuroimmunol2013258415023523328
  • TuzunEChristadossPComplement associated pathogenic mechanisms in myasthenia gravisAutoimmun Rev20131290491123537510
  • Chamberlain-BanoubJNealJWMizunoMComplement membrane attack is required for endplate damage and clinical disease in passive experimental myasthenia gravis in Lewis ratsClin Exp Immunol200614627828617034580
  • MorganBPChamberlain-BanoubJNealJWThe membrane attack pathway of complement drives pathology in passively induced experimental autoimmune myasthenia gravis in miceClin Exp Immunol200614629430217034582
  • LennonVASeyboldMELindstromJMRole of complement in the pathogenesis of experimental autoimmune myasthenia gravisJ Exp Med1978147973983206648
  • ChristadossPC5 gene influences the development of murine myasthenia gravisJ Immunol1988140258925923356901
  • KaminskiHJKusnerLLRichmondsCDeficiency of decay accelerating factor and CD59 leads to crisis in experimental myastheniaExp Neurol200620228729316859686
  • LinFKaminskiHJConti-FineBMMarkedly enhanced susceptibility to experimental autoimmune myasthenia gravis in the absence of decay-accelerating factor protectionJ Clin Invest20021101269127412417565
  • HudaRTuzunEChristadossPTargeting complement system to treat myasthenia gravisRev Neurosci20142557558324731953
  • SunLXingLZhangGHA new method to induce myasthenia gravis models and the protective effect of soluble decay accelerating factorsGenet Mol Res2015147782779226214459
  • SoltysJKusnerLLYoungANovel complement inhibitor limits severity of experimentally myasthenia gravisAnn Neurol200965677519194881
  • LosenMStassenMHMartinez-MartinezPIncreased expression of rapsyn in muscles prevents acetylcholine receptor loss in experimental autoimmune myasthenia gravisBrain20051282327233716150851
  • Martinez-MartinezPLosenMDuimelHOverexpression of rapsyn in rat muscle increases acetylcholine receptor levels in chronic experimental autoimmune myasthenia gravisAm J Pathol200717064465717255332
  • O’ConnellRMBaltimoreDMicroRNAs and hematopoietic cell developmentCurr Top Dev Biol20129914517422365738
  • ZhangJJiaGLiuQSilencing miR-146a influences B cells and ameliorates experimental autoimmune myasthenia gravisImmunology2015144566724962817
  • WangYZTianFFYanMDelivery of an miR155 inhibitor by anti-CD20 single-chain antibody into B cells reduces the acetylcholine receptor-specific autoantibodies and ameliorates experimental autoimmune myasthenia gravisClin Exp Immunol201417620722124387321
  • WangJZhengSXinNIdentification of novel MicroRNA signatures linked to experimental autoimmune myasthenia gravis pathogenesis: down-regulated miR-145 promotes pathogenetic Th17 cell responseJ Neuroimmune Pharmacol201381287130224043548
  • LosenMMartinez-MartinezPMolenaarPCStandardization of the experimental autoimmune myasthenia gravis (EAMG) model by immunization of rats with Torpedo californica acetylcholine receptors – recommendations for methods and experimental designsExp Neurol2015270182825796590
  • TuzunEBerrih-AkninSBrennerTGuidelines for standard preclinical experiments in the mouse model of myasthenia gravis induced by acetylcholine receptor immunizationExp Neurol2015270111725697844
  • CavalcantePBernasconiPMantegazzaRAutoimmune mechanisms in myasthenia gravisCurr Opin Neurol20122562162922941261
  • MelmsASchalkeBCKirchnerTThymus in myasthenia gravis. Isolation of T-lymphocyte lines specific for the nicotinic acetylcholine receptor from thymuses of myasthenic patientsJ Clin Invest1988819029082449461
  • MeinlEKlinkertWEWekerleHThe thymus in myasthenia gravis. Changes typical for the human disease are absent in experimental autoimmune myasthenia gravis of the Lewis ratAm J Pathol199113999510081951638
  • GomezAMWillcoxNVrolixKProteasome inhibition with bortezomib depletes plasma cells and specific autoantibody production in primary thymic cell cultures from early-onset myasthenia gravis patientsJ Immunol20141931055106324973445
  • RusselWMSBurchRLThe Principles of Humane Experimental TechniqueSpecial Edition UFAWLondonMethuen & Co1992
  • MaCGZhangGXXiaoBGLinkJOlssonTLinkHSuppression of experimental autoimmune myasthenia gravis by nasal administration of acetylcholine receptorJ Neuroimmunol199558151607537280
  • ZhangJMJiaGLiuQSilencing miR-146a influences B cells and ameliorates experimental autoimmune myasthenia gravisImmunology2014144566724962817