Update On IgG4-mediated Autoimmune Diseases

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 21

Autoimmunity Reviews 19 (2020) 102646

Contents lists available at ScienceDirect

Autoimmunity Reviews
journal homepage: www.elsevier.com/locate/autrev

Update on IgG4-mediated autoimmune diseases: New insights and new T


family members
Inga Koneczny
Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, Währingergürtel 18-20, 1090 Vienna, Austria

ARTICLE INFO ABSTRACT

Keywords: Antibodies of IgG4 subclass are exceptional players of the immune system, as they are considered to be im-
IgG4 munologically inert and functionally monovalent, and as such may be part of classical tolerance mechanisms.
Autoimmunity IgG4 antibodies are found in a range of different diseases, including IgG4-related diseases, allergy, cancer,
Autoantibodies rheumatoid arthritis, helminth infection and IgG4 autoimmune diseases, where they may be pathogenic or
Pathogenic mechanisms
protective. IgG4 autoimmune diseases are an emerging new group of diseases that are characterized by pa-
Classification
thogenic, antigen-specific autoantibodies of IgG4 subclass, such as MuSK myasthenia gravis, pemphigus vulgaris
and thrombotic thrombocytopenic purpura. The list of IgG4 autoantigens is rapidly growing and to date contains
29 candidate antigens. Interestingly, IgG4 autoimmune diseases are restricted to four distinct organs: 1) the
central and peripheral nervous system, 2) the kidney, 3) the skin and mucous membranes and 4) the vascular
system and soluble antigens in the blood circulation. The pathogenicity of IgG4 can be validated using our
classification system, and is usually excerted by functional blocking of protein-protein interaction.

1. Introduction mortality rate in pemphigus [3]).


Most pathogenic mechanisms of autoantibodies rely on the effector
IgG4 autoimmune diseases (IgG4-AID) are an emerging group of function of the Fc region and can be called Fc-dependent (Fig. 2A).
autoimmune diseases that are characterized by the presence of auto- These include the recruitment of C1q and activation of the classical
antibodies of IgG4 subclass [1,2]. While the antigens in IgG4-AID are complement pathway and complement-dependent cytotoxicity (CDC),
located throughout the body, they are found accumulated in four major as seen e.g. in myasthenia gravis with autoantibodies to the acet-
organs (Fig. 1): 1) the predominant target organ with ten established ylcholine receptor (AChR-MG, reviewed by [12]). By-products of the
and proposed antigens is the central and peripheral nervous system complement cascade, C5a and C3a, may also recruit leukocytes that
(including the neuromuscular synapse), 2) six antigens are located in further inflammation and tissue damage. Leukocytes may interact via
the skin, 3) three (six if we also count intracellular antigens) are tar- activating Fcγ receptors with the Fc region of bound IgG and may lead
geting the kidney and 4) two antigens are located in the blood circu- to tissue damage via antibody-dependent cellular cytotoxicity (ADCC)
lation (seven if we also count ANCAs and autoantigens in APECED), e.g. by release of reactive oxygen species, perforin, granzymes, lipid
extending the number of autoantigens in proposed IgG4-AID from mediators or proinflammatory cytokines, as seen e.g. in neuromyelitis
fourteen to up to 29. Examples for IgG4-AID are well-known diseases optica spectrum disorder (NMOSD) [13,14]. Soluble antigen may be
such as pemphigus vulgaris and foliaceus, MuSK Myasthenia gravis or cross-linked by autoantibodies that bind multivalently, such as IgG or
thrombotic thrombocytopenic purpura. IgG4-AID are rare, with pre- IgM, forming immune complexes that may lead to microthrombosis or
valence below 5 per 10,000 (Table 1), and severe, with many forms further stimulate inflammation e.g. by activation of the complement
being fatal if left untreated or even with treatment (e.g. 5–30% system, as seen in systemic lupus erythematosus (SLE) [15]. Cross-

Abbreviations: ADAMTS13, A disintegrin and metalloproteinase with thrombospondin motifs 13; APECED, Autoimmune poly-
endocrinopathy–candidiasis–ectodermal dystrophy; AR, Aldose reductase; BP, Bullous pemphigoid; CIDP, Chronic inflammatory demyelinating polyneuropathy;
CNTN1, Contactin 1; Caspr1,2, Contactin associated protein1,2; Dsg1,3, Desmoglein 1, 3; DPPX, Dipeptidyl-peptidase-like protein 6; FAE, Fab-arm exchange;
GPIHBP1, Glycosylphosphatidylinositol-anchored high density lipoprotein–binding protein 1; GSH, Glutathione; IgLON5, IgLON family member 5; Lam332, Laminin
332; LGI1, Leucine-rich, glioma inactivated 1; Lrp4, Low density lipoprotein receptor-related protein 4; MBL, Mannose-binding lectin; MMP, Anti-laminin mucous
membrane pemphigoid; MuSK, Muscle-specific kinase; MG, Myasthenia gravis; MN, Membranous nephropathy; PLA2R, Phospholipase A2 receptor (PLA2R);
THSD7A, Thrombospondin type-1 domain- containing 7A; TTP, Thrombotic thrombocytopenic purpura; vWF, von Willebrand factor
E-mail address: inga.koneczny@meduniwien.ac.at.

https://doi.org/10.1016/j.autrev.2020.102646
Received 29 February 2020; Accepted 8 March 2020
Available online 13 August 2020
1568-9972/ © 2020 The Author. Published by Elsevier B.V. This is an open access article under the CC BY license
(http://creativecommons.org/licenses/BY/4.0/).
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

covalent interaction and that together form a heterotetramer (H2L2). At


the n-terminal end of the heavy and light chains, a variable region is
located (VH and VL), which recognizes and binds antigen. Specifically
the three hypervariable complementarity- determining regions
(CDR1–3) in the V-region ensure the high specificity and diversity of
the antibodies. Next to the V-region are one (light chain) or three
(heavy chain) constant domains, numbered from n-terminus to c-ter-
minus in ascending order (light chain: CL, heavy chain: CH1, CH2, CH3).
The constant regions mediate effector functions of the antibody. The
hinge region is located between the CH1 and the CH2, where the two
heavy chains are connected via disulphide bridges: two in IgG1 and
IgG4, four in IgG2 and eleven in IgG3 [32–34]. The hinge region allows
for structural flexibility of the antibody, which may assume different
conformations such as the Y-form, or the T-form that may grant in-
creased stability that withstands high pressure [35,36].
Functionally, IgG can also be divided in the “Fab” region which
consists of the CH1, VH, CL and VL, which binds the antigen, and the Fc
region, which consists of the CH2-CH3 and the hinge, and which med-
iates the antibody effector function. The term “Fab” and “Fc” derive
historically from the two products after IgG digestion with papain
(Fab= “Fragment, antigen binding”, Fc = Fragment, crystallized”, re-
viewed by [37]). There is over 90% sequence homology in the constant
domains of the four IgG subclasses, but single amino acid differences in
IgG4 (as compared to IgG1) have great impact on its structure and
Fig. 1. IgG4 autoantigens have been identified in four distinct organs: 1) the
function.
central and peripheral nervous system, 2) the skin and mucous membranes, 3)
the kidneys and 4) located in the blood circulation or associated with the
vascular system. 2.2. IgG4 is immunological inert

In the CH2 region, a serine instead of a proline at position 331


linking of transmembrane proteins may cause increased endocytosis
(“P331S”) changes the structure, which prevents binding of C1q and
and degradation of antigen from the cell surface, thus depleting the
leads to a loss of complement activation [38,39]. Complement activa-
protein from the cell surface, which can also be seen with auto-
tion can, however, not completely be dismissed as mechanism for IgG4,
antibodies against the AChR but also with antibodies against the NMDA
as there are two notable exceptions to the rule: 1) mutations in the Fc
receptor, glycine receptor and antibodies against other neurological
region of IgG4 led to the formation of artificial hexameric complexes
targets [16–20]. Phagocytes such as macrophages may also interact
via non-covalent interactions, and these could bind C1q and activate
with bound IgG via the FcγR and phagocytose the target cell, which can
the classical complement pathway [40]. While this experimental setup
be observed e.g. in autoimmune haemolytic anaemia [21]. A thorough
was quite artificial, another exception could be 2) IgG4 with hypoga-
review on pathogenic mechanisms of autoantibodies can be found here:
lactosylated side chains that could recruit mannose-binding lectins
[22].
(MBL) and activate complement via the lectin pathway. This me-
Due to structural differences in the Fc region, IgG4 is incompatible
chanism was suggested for PLA2R-IgG4 in membranous nephropathy
with these Fc-dependent effector mechanisms. Instead, IgG4 relies on
[41–46], rheumatoid arthritis [47] and IgA nephropathy [48].
Fc-independent mechanisms, which is a block of protein-protein inter-
Further single amino acid differences in the CH2 (L234F, P331S,
action with different consequences (Fig. 2B): direct blocking of receptor
A327G) reduce the interaction with activating Fcγ receptors [49–54]).
or enzyme function, as seen in thrombotic thrombocytopenic purpura
As a consequence, IgG4 is not able to activate immune cells. Taken
[23] or AChR-MG, stimulation of the receptor by binding as seen with
together, IgG4 cannot induce inflammatory processes via complement
anti-TSHR antibodies in Grave's disease [24,25], blocking of signal
or immune cells and is therefore immunologically inert.
transduction pathways as in MuSK-MG [26], and loss of cell-cell ad-
hesion that destabilizes the tissue architecture as seen in pemphigus
2.3. The IgG4 hinge region is flexible
[27].
IgG4 is considered as immunologically inert and upregulated after
The sequence differences between IgG1 and IgG4 have structural
chronic or strong antigen stimulation [28–31]. Therefore it can be
consequences. Noncovalent interaction of the CH3 regions are strongly
present in addition to antibodies of other Ig classes and IgG subclasses,
reduced by a single amino acid change in the CH3 region (R409K)
where it has a protective (rather than a pathogenic) function. In con-
[55,56] and another amino acid replacement in the hinge (P228S) leads
sequence it is essential for diseases associated with IgG4 to determine
to greater stereometric flexibility. Indeed IgG4 is not only found in the
whether IgG4 is pathogenic or protective. Our recently developed
Y- and T-conformation but also occurs in a novel conformation termed
classification system allows to determine IgG4 pathogenicity [1]. In this
λ-shape. Here one of the Fab arms is located near the Fc region and the
study we will review the key characteristics of IgG4 and examine es-
other Fab arm is stretched out [57]. Moreover, this flexibility also al-
tablished and proposed IgG4 autoimmune diseases for IgG4 patho-
lows for the generation of intrachain disulphide bridges instead of the
genicity.
interchain disulphide bridges that normally connect the two heavy
chains [58–60].
2. Structure and function of IgG4
2.4. The mechanism of Fab-arm exchange
2.1. IgG structure
The loss of covalent connections between the heavy chains, together
IgG consists of two heavy chains (H) with 50 kDa and two light with reduced binding of the CH3 regions leads to a dissociation of IgG4
chains (L) with 25 kDa that are connected via covalent and non- into two half-molecules consisting each of a heavy and a light chain.

2
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Table 1
Prevalence of IgG4 autoimmune diseases.
Antigenic target of IgG4 Associated disease Prevalence (per 10,000) Reference/Source Orpha number

Class I disease: pathogenicity of IgG4 proven


MuSK MuSK Myasthenia gravis 0.02 [4,5] ORPHA:589
CNTN1 CIDP < 0.014 (less than 5% of CIDP which as a prevalence [6,7] ORPHA:2932
of 0.28)
NF155 Peripheral neuropathies, CIDP < 0.014 (less than 5% of CIDP which as a prevalence [6,7] ORPHA:2932
of 0.28)
Desmoglein 1 Pemphigus foliaceus 0.1–0.95 [8,9] ORPHA:79481
Desmoglein 3 Pemphigus vulgaris 1–5 Orpha.net ORPHA:704
ADAMTS13 Thrombotic thrombocytopenic purpura 1–5 Orpha.net ORPHA:93585

Class II disease: pathogenicity of IgG4 very likely


CASPR2 PNS and CNS diseases N/A N/A ORPHA:276402
LGI1 Limbic encephalitis N/A N/A ORPHA:163908
PLA2R Membranous nephropathy 0.1 Orpha.net ORPHA:97560
THSD7A Membranous nephropathy 0.003 Orpha.net ORPHA:97560
CASPR1 CIDP < 0.014 (less than 5% of CIDP which as a prevalence [6,7] ORPHA:2932
of 0,28)
GPIHBP1 GPIHBP1 autoantibody syndrome Only few patients described [10,11] Not yet registered
Lam 332 Mucous membrane pemphigoid < 0.246 [8] ORPHA:46486

Class III disease: pathogenicity of IgG4 unclear


Type IV Collagen Goodpasture syndrome 0.001–0.009 Orpha.net ORPHA:375
IgLON5 IgLON5-parasomnia < 0.001 Orpha.net ORPHA:420789
DPPX DPPX-encephalitis < 0.001 Orpha.net ORPHA:329341
Neurofascin 140/186 CIDP < 0.014 (less than 5% of CIDP [6,7] ORPHA:2932
αenolase, SOD2, AR Membranous nephropathy N/A N/A ORPHA:97560
ANCA Granulomatosis with polyangiitis 0.1–0.9 Orpha.net ORPHA:900
Bulloid pemphigoid antigen 180/230 Bullous pemphigoid 1–5 Orpha.net ORPHA:703
IFN I, IL-17A, IL-17F and IL-22 APECED 0.01–0.09 Orpha.net ORPHA:3453
P200 (laminin γ1) Anti-laminin γ1/Anti-P200 < 0.01 Orpha.net ORPHA:454710
pemphigoid

The half-molecules can then recombine randomly to form chimeric, bi- 3. IgG4 in health and disease
specific antibodies that may cross-link two different antigens and that
may also possess two different light chains. This process is termed “Fab- Due to its immunological inertness and inability to exert Fc-de-
arm exchange” and may occur continuously under reducing conditions pendent antibody effector mechanisms, it is thought that IgG4 plays a
(Fig. 3), which may also have led to its comparison to “square-dancing” role in anti-inflammatory tolerance mechanisms [87]. By competitive
of antibody half-molecules [61–68]. According to our calculations, we binding and blocking of epitopes on autoantigens, IgG4 prevents
estimate that up to 99% of IgG4 is bi-specific, and we could also observe binding of other antibody classes or subclasses and thus neutralizes
bi-specificity in IgG4 autoantibodies against MuSK [68,69]. The bi- their effector mechanism (Fig. 4) [30,31,72,88–93]. The protective role
specificity has the consequence that IgG4 is not able to cross-link an- of IgG4 has been demonstrated by passive transfer experiments with
tigen and therefore cannot induce antibody-induced endocytosis or purified IgG4, e.g. in the context of bee venom allergy and myasthenia
form antigen-antibody immune complexes [66,70,71]. IgG4 can instead gravis [66,94,95].
prevent immune precipitation by IgG1 antibodies [63,72]. Yet IgG4 has been associated with a range of diseases (Fig. 5). In
addition to the IgG4-AID, IgG4 may play a role in cancer, allergies,
rheumatoid arthritis, helminth infection, systemic lupus erythematosus
2.5. Fc-Fc interactions and rheumatoid arthritis and IgG4-related diseases (IgG4-RLD). The
loss of antibody effector function due to neutralization of antigen by
Interestingly, IgG4 has yet another unique function, as it was found IgG4 is thought to be protective within the scope of allergy, as antigen-
that a dissociation of the heavy chains of IgG4 was associated with the specific IgG4 levels were increased in allergy patients [96] and IgG4
binding to other IgG via Fc-Fc interactions if the binding partner is levels rise after allergen immunotherapy [97–99].
immobilized on a solid phase [73–76]. This interaction with other IgG In the context of malignancies, stimulating a class switch towards
may lead to the aggregation of IgG4, or the formation of IgG4-con- IgG4 of anti-tumour antibodies may be the tumour cell's way to escape
taining immune complexes as observed in IgG4-RLD, rheumatoid ar- immune surveillance and survive and is associated with unfavourable
thritis and membranous nephropathy [77–79], but it is unclear whether prognosis, e.g. in melanoma, extrahepatic cholangiocarcinoma, pan-
this plays a role for IgG4 pathogenicity in IgG4-AID. creatic cancer or glioblastoma [100].
In parasitic infection, IgG4 was suspected to play a role in the Similarly, helminths are parasites that aim to evade the host im-
regulation of complement activation [80–83]. A recent study found that mune system, while the host organism tries to destroy the parasite,
the Fc of IgG4 interacts with other IgG FCs and reduces the affinity of which led to an arms race between parasite and host that shaped our
IgG1 and IgG2 (but not IgG3) for C1q binding, which was restored after immune system [101,102]. To evade the host immune system, hel-
IgG4 depletion from the patient plasma [84]. Taken together with the minths stimulate (among other things) the production of IL-10 and
ability to break up immune complexes, this is another active me- Tregs [103], which inhibits T-cell immunity [104] and induce the class-
chanism for anti-inflammatory action, in addition to the passive me- switch of anti-worm antibodies towards IgG4 [105,106]. As a “by-
chanism of immunological inertness. The following excellent reviews stander effect”, the immune modulation by helminth parasites may
are suggested for a more in-depth understanding of the structure, protect the host from allergies and autoimmune diseases. This is also
function and regulation of IgG4 [1,70,85–87]. part of the “hygiene hypothesis” which sees causality between the rise
of autoinflammatory and allergy conditions in the Western world with

3
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Fig. 2. Effector mechanisms of antibodies. A) Fc-


dependent antibody effector mechanisms include
complement activation, formation of immune com-
plexes, leukocyte recruitment and activation of im-
mune cells via activating FcγR and phagocytosis,
leading to inflammation and tissue damage, and
cross-linking of transmembrane proteins leading to
increased turnover and antigen depletion, often
dramatically affecting functional tissue architecture.
B) Fc-independent mechanisms include receptor sti-
mulation, and mechanisms mediated by blocking of
protein-protein interaction in trans, e.g. interrupting
cell-cell interaction or block of ligand or substrate
binding to enzymes and receptors, or in cis, leading
to interruption of signal transduction pathways.

the increase of hygienic standards and reduced parasite infections of the same machinery (Treg-IL10-IgG4) could be responsible for pa-
[101,102,107,108]. Thus, whether anti-worm IgG4 is protective or not thogenicity, and may present similar targets for new therapies.
in helminth infection depends on the perspective and circumstances, IgG4 subclass antibodies are present in rheumatoid arthritis, as they
since it certainly protects the worm. The study of helminth infection is comprise a large fraction of the anti-citrullinated protein antibodies
interesting also for the field of IgG4-AID, since in both conditions parts (ACPA) [109,110]. ACPA-IgG4 have also a different Fc-glycan profile

4
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Fig. 3. Fab-arm exchange is a continuous process. Under reducing conditions


the two IgG half-molecules may lose the covalent connection via interchain
disulphide bridges in the hinge region, as intrachain and interchain disulphide
bridges exist in a dynamic equilibrium. The resulting IgG4 half-molecules dis-
sociate and may stochastically recombine to form intact bispecific IgG4, which
may again dissociate to half-molecules for the duration of reducing conditions.

than IgG1-ACPA [111], and it is possible that the differential glycosy-


lation and IgG subclass may affect effector- and regulatory function of
the antibodies. Interestingly, a positive treatment response in RA pa-
tients was associated with decreased levels of ACPA-IgG4 [112,113].
Furthermore, in RA and SLE, F(ab)2 fragments of IgG4 subclass anti-
bodies may become targets of anti-hinge antibodies (AHA), leading to
formation of large immune complexes that may induce inflammation
and tissue damage or reconstitution of complement activation by the Fc
of the AHA [114–116]. Nevertheless, the relevance of the IgG4 subclass
in RA is not understood yet.
Similarly, the role of IgG4 in IgG4-RLD is unclear [117]. IgG4-RLD
is a collective term for a group of diseases that may affect different
organs and are associated with IgG4, such as IgG4-related pancreatitis
[118,119], IgG4-related periaortitis/periarteritis [120], IgG4-related
lung disease [121] or IgG4-related hypophysitis [122]. Hallmark
characteristics are high levels of serum IgG4 [123], fibrosis and tume-
factive lesions in different target organs with IgG4+ plasma cell in-
Fig. 4. IgG4 is immunologically inert and may dampen immune reactions. IgG4
filtrates [124,125]. Notably, these characteristics are not observed in
competes for antigen binding with other antibody (sub-)classes that may exert
IgG4-AID, which is why we believe them to be two distinct groups of
Fc-dependent effector mechanisms, therefore effectively inhibiting antibody
diseases. There are also few known antigen-specific IgG4 auto- effector mechanisms. IgG4 is bispecific, which allows only for monovalent
antibodies in IgG4-RLD, with unclear evidence regarding the patho- antigen binding, thereby preventing the formation of antibody-antigen immune
genicity of IgG4, which is discussed in detail in this excellent review: complexes in solution, and the cross-linking and endocytosis of transmembrane
[117]. Indeed there is some evidence for a protective role of IgG4. proteins. Furthermore, IgG4 does not bind to C1q and has reduced affinity for
Shiokawa et al. showed that passive transfer of isolated IgG1 from activating FcγR, thereby preventing complement activation or immune cell
patients with a pancreatic form of IgG4-RLD (and to a degree also IgG4) mediated mechanisms such as antibody dependent cellular cytotoxicity
was pathogenic in neonatal male Balb/c mice, but that co-transfer of (ADCC).
IgG1 and IgG4 significantly inhibited the pathogenic damage [126].
Furthermore, autoantibodies of IgG1 and IgG4 subclass to annexin-A11, correlated with increased plasma galectin-3 levels and increase of IgG4
a calcium-dependent phospholipid-binding protein, were identified in levels [128] and it was hypothesized that these antibodies may be as-
patients with IgG4-RLD targeting the pancreas, bile duct and salivary sociated with tissue fibrosis [117]. There are further studies needed to
glands. Two binding epitopes were shared among patients and it was determine the role of IgG4 in IgG4-RLD. Recently a new animal model
suggested that IgG4 antibodies block the binding of IgG1 antibodies of IgG4-RLD was proposed, here a mutation of an adaptor protein in-
[127]. Still, annexin A11 is an intracellular protein, making it an un- volved in T-cell receptor signalling, Linker for activation of T cell (LAT),
likely target for pathogenic autoantibodies as a primary cause of dis- induced increased expression of the mouse-equivalent of human IgG4,
ease, and it is also found in a range of other autoimmune diseases such which is mouse IgG1, in combination with multiple organ tissue lesions
as SLE. A recent study identified another autoantibody target in IgG4- similar to those observed in patients with IgG4-RLD [129]. It would be
RLD, namely galectin-3, the autoantibodies were of IgG4 and IgE sub- interesting to study if autoantigen-specific antibodies are present in this
class and found in 28% of the patient cohort. Notably galectin-3-IgG4 mouse model, and whether passive transfer of IgG to wildtype animals

5
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Fig. 5. Diseases associated with IgG4. IgG4 is found in a range of diseases, in


which it may have different roles. IgG4 has immune regulative properties and
by such is protective in the context of allergy, but may instead be harmful in the
context of cancer and helminth infection, where the host immune system is
prevented from attacking tumour cells or parasitic worms. In rheumatoid ar-
thritis, IgG4 may become a target of autoantibodies itself that target the IgG4
hinge region (so called anti-hinge antibodies, or AHAs). The role of IgG4 in
IgG4-related diseases is not understood. In IgG4 autoimmune diseases, antigen-
specific IgG4 are directly pathogenic by blocking mechanisms.

also induces IgG4-RLD-like symptoms. Taken together, it is not yet clear


what the role of IgG4 is in IgG4-RLD, but in many IgG4-AID there is
evidence (or at least a strong suspicion) that antigen-specific IgG4 is a
pathogenic entity [1].

4. IgG4 classification system

In order to determine the pathogenicity of IgG4 in IgG4-AID, we


have recently proposed a new classification system (Box 1, [1]). The
classification system is based on the Witebsky postulates and observa-
tions from Naparstek and Plotz [130–132].
Box 1: Specific indicators for IgG4 pathogenicity [1]

1. Autoantibodies of IgG4 subclass against extracellular antigen are


present.
2. A pathogenic mechanism for IgG4 can be demonstrated in vitro.
IgG4 could be purified from patient serum, and the purity of isolated
IgG4 should be validated. Alternatively, cloned patient antibodies
from IgG4+ B-cells could be used.
Fig. 6. Pathogenic mechanisms of MuSK autoantibodies of IgG4 subclass in
3. Reproduction of disease in animals by passive transfer of purified
Myasthenia gravis. A) The neuromuscular junction. During neuromuscular
IgG4 from patients, monoclonal IgG or scFv (single chain variable
transmission, the neurotransmitter acetylcholine (ACh) is released into the sy-
region fragments) cloned from IgG4+ B-cells of patients. naptic cleft, where it binds to the acetylcholine receptor (AChR) at the post-
synaptic muscle membrane, causing the AChR to open, which leads to influx of
4.1. IgG4 autoantibodies with validated pathogenicity (class I) Na + ions and depolarisation of the membrane. The dense aggregation of
AChRs at the postsynaptic membrane is developed and maintained by the
In these diseases, IgG4 is proven to be pathogenic by fulfilling the function of the muscle-specific kinase (MuSK). The motoneuron releases neural
three classification criteria. Importantly, a passive transfer of purified agrin into the synaptic cleft, which binds to the agrin co-receptor, low-density
lipoprotein receptor-related protein 4 (Lrp4), causing an increased binding of
IgG4 could reproduce the disease in experimental animals. This means
Lrp4 to MuSK. This leads to autophosphorylation of MuSK, binding of the
that these diseases are definitely caused by pathogenic IgG4 auto-
adapter protein Dok-7 and to full activation of MuSK. MuSK then phosphor-
antibodies. ylates further down-stream proteins which leads to the clustering of AChR via
the scaffolding protein rapsyn. B) MuSK autoantibodies of IgG4 subclass bind to
4.1.1. MuSK MuSk and prevent Lrp4-MuSK interaction, therefore interrupting the agrin-
Patients with autoantibodies against the muscle-specific kinase Lrp4-MuSK-Dok-7 pathway, and leading to reduced densities of AChRs at the
(MuSK), a protein at the neuromuscular junction, develop MuSK neuromuscular junction and impaired neuromuscular transmission.
-myasthenia gravis [133]. MuSK is a key organizer of neuromuscular
junction development and maintenance. Upon release of the protein protein rapsyn [12] (Fig. 6A). MuSK antibodies are predominantly IgG4
agrin from the motoneuron, it binds to the agrin co-receptor Lrp4, [26,134] and are able to undergo Fab-arm exchange in vitro [69]. In-
which in turn binds and activates MuSK, which is a receptor tyrosine deed calculations suggest that up to 99% of MuSK antibodies are bis-
kinase that then becomes activated, phosphorylates downstream targets pecific in vivo. The MuSK antibodies recognize several domains of
and regulates the clustering of AChRs at the synapse via the scaffolding

6
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

MuSK, but they predominantly bind to the first Ig-like domain against CNTN1 to rats caused a conduction block in the absence of
[133–137]. This is interesting, because the first Ig-like domain also abnormal paranodal structure [162]. Notably IgG3 was not the only
contains two regions that are functionally important, one that is re- subclass present in the patients, also IgG1 and IgG4 were present at
quired for MuSK dimerization and a second one around Ile96 that is reduced levels, but IgG3 and C1q deposits were colocalized at the
required for the interaction between Lrp4 and MuSK [138], suggesting paranode. This suggests that complement-fixing IgG3 may play a role in
functional consequences of MuSK antibody binding. Indeed we could early disease stages, while non-complement fixing IgG4 may be re-
demonstrate that MuSK antibodies block the binding between Lrp4 and levant in the chronic phase by blocking.
MuSK (Fig. 6B) [26,137,139], which also led to a decreased MuSK ac- Interestingly a subclass switch from complement fixing IgG sub-
tivation and phosphorylation [139] and reduced AChR clustering at the classes to IgG4 was also observed in patients with anti- PLA2R anti-
neuromuscular synapse. While the blocking mechanism was exclusive bodies [43,163]. Perhaps a class switch from IgG1/3 to IgG4 is at work
for IgG4 subclass antibodies, IgG1–3 was also able to reduce agrin-in- in other IgG4-AID, but this will be challenging to prove, since many of
duced AChR clustering in vitro [26]. There may be another signalling these diseases are difficult to diagnose. By the time the autoantibodies
pathway that is blocked by MuSK antibodies, but we don't understand are tested, the subclass switch may already have happened.
the exact mechanism yet. We induced AChR clusters by overexpressing Last, but not least, the disease could be reproduced by passive
Dok-7, independent of agrin, and therefore independent of agrin-Lrp4- transfer of purified IgG4 to Lewis rats, leading to a loss of paranodal
MuSK interaction, and found that MuSK MG IgG and Fab fragments clusters and reduced motor nerve conduction [164], which makes it a
were able to disrupt these AChR clusters [26,140]. Perhaps the anti- class I IgG4-AID.
bodies induced conformational changes in MuSK that affected binding
of MuSK to Dok-7 or another protein. 4.1.3. Neurofascin 155
Interestingly, antibody valency also plays a role in pathogenicity of The binding partner of CNTN1 in the axo-glial complex, Neurofascin
MuSK autoantibodies. MuSK IgG4 is bispecific and binds monovalently, 155 (NF155, Fig. 7), is also targeted by IgG4 autoantibodies. NF155 is
and monovalent binding by Fab Fragments also blocked Lrp4-MuSK an adhesion protein expressed on the glia cells in the paranodal septate-
interaction and reduced agrin- induced AChR clusters [26]. Divalent like junctions of myelinated axons [165,166,167]. Autoantibodies to
recombinant MuSK antibodies cloned from MuSK MG B-cells had a NF155 (as to CNTN1 and perhaps also Caspr1) are thought to cause
different mechanism, they cross-linked MuSK and induced MuSK changes in the axo-glial complex that lead to peripheral neuropathies
phosphorylation and agrin-independent AChR clustering [141]. This [158,168–170], including chronic inflammatory demyelinating poly-
may present another pathogenic mechanism, perhaps for MuSK IgG1–3 neuropathy (CIDP). There are some differences though, as patients with
that may by this mechanism deplete AChRs from the NMJs and instead NF155 antibody positive CIDP have a distinct clinical phenotype that is
form ectopic AChR clusters. defined by a low-frequency and high-amplitude tremor with cerebellar
Another puzzling finding is that MuSK antibodies may affect a ret- features and IVIG unresponsiveness [171–174]. To date, at least two
rograde signal to the motoneuron, since presynaptic effects have been pathogenic mechanisms have been proposed for NF155-IgG4: 1) a
observed in MuSK MG patients and animal models. These include a blocking mechanism that prevents the interaction between CNTN1-/
reduction of the quantal release of acetylcholine and structural ab- Caspr1 and NF155 and disrupts the paranodal structure [158] and 2)
normalities in the NMJ architecture [142–146]. Lrp4 was found to induction of NF155 clustering and depletion from the cell surface by yet
contribute to a retrograde signalling to the motor neuron [147,148], unidentified mechanisms, with a clear absence of any blocking function
therefore it is possible that MuSK binding to Lrp4 is also involved in the [175].
retrograde signalling pathway, perhaps by anchoring Lrp4 at the NMJ. In the latter study, IgG4 was isolated from plasmapheresis material
MuSK antibodies may potentially also affect MuSK located in the brain. from three CIDP patients and used for studies in vitro in different cell
One study suggests that passive transfer of patient animals to mice led lines, as well as in vivo by passive transfer to postnatal and adult Lewis
to reduced exploring time in the novel object recognition test. This rats.
could have been caused by cognitive defects, though it cannot entirely The in vitro experiments showed no block of interaction between
be excluded that it was caused by muscle weakness [149]. It would NF155 and CNTN1/Caspr1 in transfected HEK293 cells, but instead
certainly be interesting to investigate potential effects of MuSK anti- induction of NF155 clustering. This is interesting, as it suggest a mul-
bodies on the brain. tivalent binding and cross-linking mechanism. In cell culture experi-
ments with Schwann cells, oligodendrocytes or transfected HEK293
4.1.2. CNTN1 cells, it was shown that NF155-IgG4 did not induce internalization of
Contactin-1 (CNTN1) is an adhesion molecule that are expressed on antigen as it would be found in cross-linking and endocytosis. And yet,
the surface of axons at the axoglial junction (Fig. 7). CNTN1 binds to- in the Lewis rats, NF155 was found to be depleted, and in the postnatal
gether with CNTN1-associated protein 1 (Caspr1) to the protein Neu- rats this was accompanied by impaired paranode formation and loss of
rofascin-155 (NF155) on the surface of oligodendroglia [150–153], and Caspr1 positive septate-like junctions. It is puzzling how NF155 could
all three proteins have been suggested as targets for IgG4 auto- be depleted if cross-linking and internalization is not a likely me-
antibodies. The CNTN1-Caspr1-NF155 complex is required to maintain chanism of action, due to the results of the in vitro experiments and the
paranode architecture and to maintain myelin insulation of the axon for bi-specific nature of IgG4. The authors hypothesize that antibody
nerve impulse propagation along myelinated axons. In patients with binding to the thrombin cleavage site in the third fibronectin type III
chronic inflammatory demyelinating polyradiculoneuropathy (CIDP), domain could induce proteolysis [176], or a reduced interaction with
autoantibodies of IgG1 and IgG4 subclass target CNTN1 that may alter an unknown binding partner may cause reduced anchoring of NF155 to
the paranode structure [154–157], and there is evidence that the IgG4 the paranodal junction. Also it is possible that NGF155-IgG4 induces
antibodies have a blocking mechanism that leads to a block of NF155- conformational changes that may lead to NF155 clustering and sub-
CNTN1 interaction [158]. Some CNTN1 antibody positive patients (as sequent breakdown of NF155. It is also thinkable that immobilized
well as NF186 positive patients) may also present with nephrotic syn- IgG4, via Fc-Fc interaction with other IgG4 or IgG1–3 could lead to
drome, or a “neuro-renal syndrome”. This may be explained by the cross-linking after all.
expression of CNTN1 and NF186 on podocytes [159,160], where au- Importantly, patient-like symptoms were induced by chronic in-
toantibodies might affect the structure and function of the glomeruli in trathecal infusion with patient IgG4 to adult Lewis rats, including gait
the kidney [161]. abnormalities, slower motoneuron conduction velocity, paraparesis and
IgG4 may also not be the sole contributor to pathogenicity, as complete tail paralysis in the absence of demyelination.
passive transfer of serum with predominant IgG3 subclass antibodies Taken together there may be different mechanisms at hand that are

7
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Fig. 7. The nodes of Ranvier in motoneuron axons are densely packed with IgG4 autoantigens. Schwann cells are providing electric insulation in motoneuron axons,
and together with the gaps, the nodes of Ranvier, allow for saltatory conduction, the propagation of action potentials, in the axon. To this end, the membranes of axon
and Schwann cell have to be tightly sealed. Many IgG4 autoantigens are, curiously, expressed at the juxtaparanodal junction, namely CNTN1, Caspr1, Caspr2, NF186,
NF140 (not shown), and NF155. It is unclear why this structure seems to be so susceptible to IgG4 autoantibodies.

not fully understood to date. Nevertheless, the Manso study clearly simultaneously [183,184]. The antibodies recognize epitopes in the
proves the pathogenic status of NF155-IgG4, as the disease could be EC1 and EC2 domain of Dsg1 and Dsg3 [185,186], and binding of the
reproduced by passive transfer experiments [175]. autoantibodies leads to a block of cell-cell adhesion and causes blis-
tering of the skin [187,188]. This pathogenic mechanism could be de-
4.1.4. ADAMTS13 monstrated in vitro, as antibodies induced the dissociation of cell sheets
A disintegrin and metalloproteinase with thrombospondin motifs 13 of cultured human keratinocytes [189,190] and human skin explants
(ADAMTS13) is the target of IgG4 autoantibodies in patients with [191]. In an endemic form of pemphigus, fogo selvagem, the main
thrombotic thrombocytopenic purpura (TTP). epitope was a 16-residue peptide in the EC1 domain of Dsg1
ADAMTS13 is located in the blood circulation, where it cleaves the (A129LNSMGQDLERPLELR144), and binding of Fab fragments disrupted
active, multimeric form of the von Willebrand factor (vWF, Fig. 8). the binding between Dsg1 and the desmosome protein desmocollin 1
ADAMTS13 binds to the vWF via its spacer domain, which is also the (Dsc1) [192]. Blocking of cell-cell adhesion is not the only mechanism
main epitope for the IgG4 autoantibodies, thus blocking the binding of at work, since the autoantibodies can also alter the signal transduction
the protease to its target. This leads to the accumulation of multimeric pathways that affect cytoskeleton rearrangement and cell adhesion in
vWF which also binds to platelets, forming microthrombi that lead to keratinocytes [191,193–199]. There is clinical evidence for the patho-
vascular occlusion [177,178]. The pathogenicity of the antibodies was genicity of IgG4 since the depletion of IgG4 from patient serum reduced
demonstrated in vitro, using IgG4 and IgG1 antibodies cloned from TTP the pathogenicity in vitro [200], and passive transfer of maternal anti-
patient B-cells. Both IgG4 and IgG1 were able to block ADAMTS13 bodies to the fetus induces a transient neonatal form of pemphigus
activity in vitro [179], and inhibition of ADAMTS13 was increased in [201]. Importantly, passive transfer of IgG4 subclass antibodies could
patients that had predominantly IgG4 subclass antibodies [180]. Fur- reproduce the disease in animals [185,202].
ther evidence comes from an association of IgG4 levels with relapse,
and some patients have only IgG4 subclass antibodies, which adds 4.2. IgG4 autoantibodies with non-validated pathogenicity (class II)
clinical evidence for the pathogenicity of IgG4 [181]. Disease could also
be reproduced by expression of patient derived (monovalent) scFv ex- IgG4-AIDs where pathogenicity of IgG4 has not yet been proven but
pressed in mice, though the subclass of the original patient antibodies is is otherwise strongly suspected belong to the class II IgG4-AIDs. Usually
unknown [182]. There is also an IgG subclass switch from IgG1 at the the antibodies have been demonstrated to be pathogenic by a blocking
first presentation of disease to IgG4 during (or shortly before) relapse, mechanism in vitro, but in vivo evidence by passive transfer of patient
with increased IgG levels in patients with the DR7-DQ2 and DR13-DQ6 IgG4 is still lacking.
haplotypes [180].
4.2.1. LGI1
4.1.5. Dsg1 and Dsg3 Patients with LGI1- associated limbic encephalitis have auto-
Antibodies to desmoglein 1 or desmoglein 3 (Dsg1,3) of IgG4 sub- antibodies to the neuronal antigen LGI1 and present with a range of
class are present in patients with pemphigus [27]. Dsg1 and Dsg3 are neurological dysfunction, specifically regarding memory and behaviour
transmembrane cell-adhesion proteins expressed in keratinocytes which is also accompanied by seizures [203–206]. LGI1 antibody po-
(Fig. 9). Dsg1 is expressed in the superficial layers of the epidermis, sitive patients have very characteristic faciobrachial dystonic seizures
while Dsg3 is expressed in the basal and parabasal layers of the skin. (short, unilateral seizures in the upper limb and face), subtle dyscog-
Pemphigus foliaceus is induced by Dsg1 antibodies, while pemphigus nitive or autonomic seizures, and generalized tonic–clonic seizures
vulgaris is induced by antibodies against Dsg3, or both Dsg1 and Dsg3 [207].

8
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Fig. 8. The pathogenic mechanism of ADAMTS13 autoantibodies in thrombotic thrombocytopenic purpura. ADAMTS13 is an enzyme located in the blood circu-
lation. It cleaves the active, multimeric form of the von Willebrand factor (vWF). IgG4 autoantibodies bind to the same region as vWF, block the binding of vWF to
ADAMTS13 and prevent cleavage of vWF. Multimeric vWF accumulates, binds platelets and forms microthrombi that and cause vascular occlusion.

LGI1 is a neuronal antigen, which is expressed at synapses in the ADAM22 interaction, which is associated with reduced AMPAR levels
CNS as part of the voltage gated potassium channel complex (VGKC). It [213]. There is some evidence that LGI1 antibodies may also cross-link
is secreted by pre- and postsynaptic neurons and cross-links trans- and endocytose LGI1, with one in vitro study showing internalization of
membrane proteins from both sides of the synaptic cleft. LGI1 binds LGI1 in ADAM22-transfected HEK293T cells incubated with soluble
presynaptically to ADAM23 and postsynaptically to ADAM22. LGI1- LGI1 [214]. A limit of the study is the use of whole serum. These had
ADAM22 and -ADAM23 interaction is thought to regulate neuro- either predominant LGI1-IgG1 or LGI1-IgG4, but the complete IgG
transmitter receptor and ion channel levels at the synapse. subclass profile was not provided, therefore it is not entirely clear
The binding to ADAM22 is mediated by a hydrophobic pocket of the which antibody subclass was associated with this mechanism. A repeat
c-terminal epitempin-repeat (EPTP) domain of LGI1 which binds to the of the experiment with purified patient IgG4 and IgG1 would be ad-
metalloprotease-like domain of ADAM22 [208,209]. ADAM22 then visable.
binds the scaffolding protein PSD-95, and taken together the complex is At the presynaptic site, LGI1 is also involved in regulation of the
thought to regulate AMPA receptor levels [210]. AMPAR are ionotropic density of presynaptic Kv1.1 receptors at the axonal initial segment,
neurotransmitter receptors are required for excitatory synaptic trans- which regulates the action potential firing [215]. LGI1 binds ADAM23,
mission in many parts of the brain and play a role in long-term po- which in turn binds to and regulates expression levels of the presynaptic
tentiation, synaptic plasticity and memory. LGI1 expression is high in Kv1.1 [216]. Kv1.1 are shaker related voltage-gated potassium ion
the CA3 region in the hippocampus, which is important for episodic channels, that open during the repolarization phase and become se-
memory. In line of a neurotoxic effect of LGI1 autoantibodies, LGI1 lective ion channels for potassium ions. Interestingly, Kv1.1 are also the
patients suffer from memory deficits with focal atrophy in the CA3 target of autoantibodies in autoimmune neuromyotonia (Isaac's syn-
region [211]. The neurotoxic effect of LGI1 antibodies on hippocampal drome). The interaction between LGI1 and ADAM23 is therefore also
neurons could be observed in a recent study, where LGI1-IgG incuba- important to maintain functionality of the presynaptic site, and as it
tion on hippocampal neurons in cell culture was associated with turns out, may also be targeted by LGI1 antibodies. Passive transfer of
apoptosis and reduced calcium currents [212]. purified IgG from LGI1 patient serum to mice led to a block of LGI1-
At the postsynaptic site, the LGI1 antibodies cause a block between ADAM23 binding and reduced synaptic and overall expression levels of
LGI1 and ADAM22 binding. The antibodies bind to the same region as Kv1.1 and AMPAR at the mouse hippocampus [217]. Reduced Kv1.1
ADAM22, which is the EPTP domain, thereby effectively blocking LGI1- levels may also have caused the observed hyperexcitability in

9
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

and 3) it plays a role in nociceptive pain, as it regulates the excitability


of dorsal root ganglion cells by regulating the trafficking of Kv1 chan-
nels to the membrane [232]. The patients may have predominantly
IgG1 and IgG4 antibodies, and due to the different sites of expression, it
is thought that CASPR2 antibodies may have different pathogenic ef-
fects.
CASPR2 antibodies block CASPR2- TAG-1 interaction [233,234].
This mechanism is associated with IgG4 since IgG4-enriched CASPR2
antibodies were found to be blocking. In addition, an internalization of
CASPR2 was excluded since the antibodies in this study did not affect
cell surface expression of CASPR2 in hippocampal neurons in vitro
[233]. A recent study also verified the block of Caspr2-TAG1 interac-
tion, and observed an upregulation of Kv1.2 in hippocampal neurons as
a consequence of incubation with CASPR2 autoantibodies [235].
Another mechanism is an increased turnover/internalization of
CASPR2. In a passive transfer animal model using CASPR2-IgG, reduced
dendritic levels of CASPR2 were observed, likely induced by inter-
nalization of CASPR2. Here IgG was used that contained both IgG4 and
IgG1 subclass antibodies, and it is thought that the internalization may
be induced by IgG1 [236]. In addition, AMPAR levels were reduced at
the synapses in cortical neurons, and the cortical excitatory transmis-
sion was decreased. This may be due to a role of CASPR2 for regulation
of AMPAR trafficking that may have been interrupted by CASPR2 an-
tibodies. In another study, IgG from a patient with CASPR2-encephalitis
was injected intraperitoneally, to reproduce the transport of serum IgG
via LPS-breached blood brain barrier to the CSF [237]. Successful
Fig. 9. Blocking is a key pathogenic mechanism of IgG4 autoantibodies in transport to the brain was verified, as IgG deposits were located in the
pemphigus vulgaris. Desmosomes are cellular complexes that mediate cellular brain parenchyma, in addition to increased levels of activated microglia
adhesion. Desmogleins (Dsg) and desmocollins (Dsc) are part of this complex and astrocytes. There was otherwise no sign of an inflammatory re-
that mediate the cell-cell interaction. Keratinocytes of the skin and mucous sponse, and no reduction in CASPR2 expression. Importantly, the mice
membranes are targeted by IgG4 antibodies against Dsg1 or Dsg3, these have a displayed behavioural changes in social interactions and working-
direct blocking function and lead to a loss of cellular adhesion. This leads to memory defects. Unlike expectations of reduced CASPR2 levels as result
blistering of the skin and mucous membranes.
of increased internalization, an increase in cell surface expression of
CASPR2 was observed in these mice. An explanation could be that ex-
hippocampus preparations from the mouse, which was also previously pression of CASPR2 was upregulated by a compensatory mechanism
shown with hippocampal CA3 pyramidal cells ex vivo [218]. The re- after initial internalization. This was reproduced in further in vitro ex-
duced AMPAR levels may have cause the defects in long-term po- periments with hippocampal neurons and CASPR2-transfected HEK293
tentiation that were observed in the mice, as well as severe, but re- cells that showed internalization of patient IgG to a small degree, to-
versible, memory deficits. These are in line with the memory deficits of gether with higher surface CASPR2 expression. The patient IgG was
LG1 patients. What was missing in the mouse model was the occurrence comprised of IgG1, 2 and 4 (72% IgG4, 22% IgG1, 6%IgG2, < 1%
of seizures, which are frequent in limbic encephalitis and in LGI1 null IgG3), therefore it may be that IgG1 and IgG2 were responsible for the
mutant mice [219]. internalization.
The passive transfer experiments underline pathogenicity of LGI1 The reduction of CASPR2 levels is likely to have functional con-
autoantibodies, but what is still lacking is the evidence that IgG4 sub- sequences. In the PNS, CASPR2 antibodies could reduce CASPR2 levels
class antibodies mediate the pathogenic mechanisms. and therefore enhance dorsal root ganglion excitability via differential
regulation of Kv1 channel expression [232], which may interfere with
4.2.2. CASPR2 pain hypersensitivity regulation and cause neuropathic pain. Increased
Patients with antibodies against the neuronal antigen Contactin excitability is also a mechanism found in the CNS in patients with
associated protein 2 (CASPR2) are associated with a range of diseases limbic encephalitis, by reducing the inhibitory input by interneurons
such as neuromyotonia, Morvan's syndrome and limbic encephalitis [234].
[220–224]. CASPR2 antibodies are associated with many different A further effect could be on Purkinje cells in the cerebellum, as they
neurological symptoms, including neuropathic pain, sleep alterations, express CASPR2 at high levels. In the Giannoccaro study, a moderate
seizures, memory impairment, cognitive deficits, and psychosis cell loss was observed in the passive transfer animal model [237]. It is
[204,220,224–229]. The wide range of disorders and symptoms is due possible that CASPR2 antibodies may lead to neuronal cell loss, another
to the different roles of CASPR2 in the peripheral and central nervous supporting observation comes from a study that reported CASPR2 an-
system. CASPR2 is a neurexin-related cell-adhesion molecule and is tibodies in patients with cerebellar ataxia [238].
expressed in the peripheral and central nervous system, specifically in Taken together, autoantibodies against CASPR2 have heterogenous
the juxtaparanodal region of myelinated axons (Fig. 7), hippocampal effects in the peripheral and central nervous system, including blocking
GABAergic interneurons and in excitatory synapses in the cortex of protein-protein interaction and increased internalization, which in
[230,231](Fernandes, Santos et al., 2019). CASPR2 forms a complex turn may affect signal transduction and neuronal tissue architecture.
with transient axonal glycoprotein-1 (TAG-1, also named Contactin-2) What is still lacking is evidence for pathogenicity of the IgG4 subclass,
and shaker-type voltage-gated potassium channels Kv1.1 and Kv1.2, which could be obtained by passive transfer of purified patient IgG4.
and may be relevant for different processes that are essential for neu-
ronal activity: 4.2.3. GPIHBP1
1) it is a scaffolding protein for VGKCs, 2) it regulates the synaptic Autoantibodies against GPIHBP1 (glycosylphosphatidylinositol-an-
expression of AMPAR and thus synaptic plasticity in the visual cortex chored high density lipoprotein–binding protein 1) are known to cause

10
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

hypertriglyceridemia in the “GPIHBP1 autoantibody syndrome” [11]. [255,272]. Complement activation via the lectin pathway by hypo- or
GPIHBP1 is a protein expressed in capillary endothelial cells that is agalactosylated IgG is a topic of some controversy [273], and further
required for intravascular lipolysis. GPIHBP1 binds, stabilizes and studies are indicated to investigate the mechanisms of complement
transports lipoprotein lipase (LPL) to its site of action in the capillary activation in PLA2R positive MN.
lumen, where LPL is then required for lipolysis [239,240,241]. The Importantly, to prove pathogenicity of PLA2R autoantibodies, and
autoantibodies against GPIHBP1 block the binding to LPL [10,11], thus specifically IgG4, passive transfer animal models would be required,
leading to reduced LPL levels, impaired intravascular triglyceride pro- but unfortunately no appropriate animal model is available because
cessing, and severe hypertriglyceridemia (chylomicronemia). The IgG rodent podocytes do not express PLA2R [274].
subclass was only recently investigated in a single patient, and anti- One approach to overcome the lack of an animal model to study
bodies were found to be exclusively of the IgG4 subclass [242]. Since complement activation in MN was to immunize mice with a different
GPIHBP1 antibodies are pathogenic by a blocking mechanism, which is glomerular antigen, namely recombinant non-collagenous (NC1) do-
associated with IgG4 autoantibodies, it is likely that future studies will main of human α3(IV) collagen (rh-α3NC1), to induce the production
identify further patients with predominant IgG4 autoantibodies. of pathogenic anti-α3NC1 autoantibodies [275]. In these animals, a role
for the alternative pathway for complement activation is emerging,
4.2.4. PLA2R since knock-out of a protein from the alternative pathway, despite the
Patients with membranous nephropathy (MN) present clinically presence of subepithelial immune complexes with IgG deposition,
with proteinuria and nephrotic syndrome [243]. In up to 75% of pa- protected the mice from proteinuria, which is thought to be caused by
tients, autoantibodies against phospholipase A2 receptor (PLA2R), complement damage. It remains to be investigated whether the same
mostly of IgG4 subclass, can be observed [244–249] and patients with applies to human patients with MN and autoantibodies to PLA2R.
PLA2R antibodies have a more severe disease course than those without Taken together, several potential pathogenic mechanisms for IgG4
[250]. Not all PLA2R antibodies belong to the IgG4 subclass, there are have been proposed, including block of protein-protein interaction
cases that are negative for PLA2R-IgG4 [251]. Up to 5% of patients with between PLA2R and the At2-complex or binding to collagen IV, and
lupus nephritis patients have autoantibodies against PLA2R, but not of activation of complement via the lectin- and alternative pathway by
the IgG4 subclass [252]. One case with MN presented with IgG1 sub- hypogalactosylated IgG4 but require further investigation. Validation of
class antibodies against PLA2R and ADAMTS13-IgG1 [253]. It is pos- pathogenicity of PLA2R- IgG4 via passive transfer animal models is to
sible that initially IgG1 and IgG3 antibodies are produced in the acute date not possible due to the lack of PLA2R expression in rodents [274].
phase of disease, followed by a class switch towards IgG4 in the chronic The development of a PLA2R expressing animal model is therefore of
phase, with both antibodies contributing different disease mechanisms urgent priority to study the role of PLA2R-IgG4 autoantibodies.
to pathology [254,255].
PLA2R belongs to the mannose receptor protein family and is ex- 4.2.5. THSD7A
pressed in podocytes [256,257]. The role of PLA2R in the kidney is not In 3–5% of patients with MN, IgG4 subclass autoantibodies against
known, but recently it was discovered that PLA2R binds to the At2 thrombospondin type-1 domain containing 7A (THSD7A) are present.
complex. This heterotetrametric complex contains the proteins The THSD7A antibodies are predominantly of the IgG4 subclass, but
S100A10 and annexin 2A [258]. Interestingly the At2 complex is as- IgG1 can also be found. There is an association with cancer, as THSD7A
sociated with many important functions, such as the remodelling of the is expressed in different tumours, and increased frequency of malig-
actin cytoskeleton, membrane organization, endo- and exocytosis and nancy in patients with THSD7A positive MN [264,276]. THSD7A is a
ion channel conductance [259]. The binding epitope for the antibodies type-1 transmembrane protein, which is normally expressed at the basal
lies in the most n-terminal, cystein-rich region (CyR) [260,261], which surface of podocytes in the kidney. It is composed of 21 domains, of
is also the region where the At2-complex binds to PLA2R. Future ex- which 18 contain predicted B-cell epitope sites [277]. The antibodies
periments will show whether perhaps PLA2R antibodies block the may target a range of domains as they are polyreactive, but the primary
binding between PLA2R and the At2 complex. This could potentially target is the n-terminal region [278].
disturb the organization of the actin cytoskeleton and affect tight The function of THSD7A has recently been unravelled, it is a foot
junction assembly, which is important for the podocyte function, and process protein that may interact with extracellular matrix proteins in
disruptions of which have been observed in patients with MN [258]. To the glomerular basement membrane and play a role in cell adhesion,
date, this has not yet been investigated, but another blocking me- regulation of cell dynamics and, importantly, slit diaphragm stabiliza-
chanism was proposed to affect cell adhesion of podocytes to collagen tion in podocytes [279], and patient antibodies were found to be de-
IV [262]. Controversially, in another study PLA2R did not bind to posited at the slit diaphragm in a passive transfer mouse model [279].
collagen I or IV [263,264]. This is in line with observations in patient biopsies, where deposits of
Interestingly, PLA2R autoantibodies may be one rare exception IgG and complement are found at the glomerular basement membrane
from the rule that IgG4 does not activate complement. In patient [280–284]. One hypothesis is that THSD7A-IgG4 may disrupt the glo-
biopsies, immune deposits containing IgG, also PLA2R-IgG4, PLA2R merular filtration barrier by loss of cell-cell adhesion at the slit dia-
and complement were found below the basal surface of podocytes in the phragm by blocking THSD7A interactions, or by directly affecting its
kidney [255,265–267]. While it is to be expected that, especially during function, which may cause destabilization of the dynamics of the foot
the acute phase of disease, IgG1 and IgG3 may fix complement by the processes and the slit diaphragm and causing proteinuria [279].
classical pathway [268], IgG4 may also activate complement in the There is supporting evidence that the antibodies may affect THSD7A
chronic phase via the lectin or alternative pathway [41–45]. It is function, as experiments on primary murine podocyte cells and primary
thought that IgG4 with galactose deficient side-chains binds to man- glomerular epithelial cells in vitro show an effect of THSD7A antibodies
nose-binding lectins (MBL) and ficolins [42,46,47,79,269], that then on focal adhesion, including cytoskeletal rearrangements, formation of
bind acetylated glycans and MBL-associated serine proteases (MASPs) stress fibres and increased focal adhesion signalling [285,286]. There
that lead to activation of C4 and the formation of the membrane-attack are also findings that support a blocking mechanism, as THSD7A anti-
complex (MAC) [41]. In line with this hypothesis is the observation that bodies induced the detachment of THSD7A expressing HEK293 cells
C1q is usually absent from these deposits [255], while C4 and MBL are [285]. These mechanisms are thought to be independent of comple-
present, and MBL levels measured in patient biopsies in Japanese MN ment-activation, as disease could be reproduced in passive transfer
patients correlated with IgG4 [270]. Furthermore, complement may animal models using anti-THSD7A antibodies that were produced in
also be activated via the alternative pathway, since IgG4, C3, C4 and rabbits [286], or with affinity purified THSD7A patient antibodies
C5b-9 were found in patients deficient for MBL [271] and ficolin-3 [285]. Still, complement activation may contribute to pathology.

11
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Passive transfer of patient serum to mice led to late complement acti- progressive glomerulonephritis, with or without pulmonary haemor-
vation [285]. Renal biopsies showed increased MBL and THSD7 rhage, which may be precipitated or reoccur after renal transplants
staining, and elevated levels of lectin pathway components were found [305,306]. The patients have autoantibodies that target the α3NC1
in patient serum and led to complement activation and reproduction of domain of collagen type IV in the glomerular basement membrane in
disease in mice [287]. the kidney. Importantly, passive transfer of patient IgG reproduced the
Taken together, THSD7A antibodies may be pathogenic by both a disease in animals [307]. To date it is known that a subgroup of patients
blocking mechanism and complement activation via the lectin pathway. has autoantibodies of the IgG4 subclass, and these patients are clinically
What is still lacking are experiments using purified IgG4 subclass an- described as predominantly female, smokers, presenting with severe
tibodies as well as a passive transfer of patient IgG4 to experimental lung engagement and relatively mild or no renal disease
animals to demonstrate pathogenicity of IgG4. [305,308–310]. IgG4 autoantibodies were temporal occurring and re-
occurring with lung haemorrhage [308,309], but it is not known if and
4.2.6. Laminin 332 how they contribute to the pathology. Furthermore, these patients also
Patients with anti-laminin mucous membrane pemphigoid (MMP, may have IgG1–3 subclass antibodies that could contribute to the dis-
formerly known as anti-epiligrin cicatricial pemphigoid, AECP) suffer ease [308,311]. Studies on purified patient IgG4 are required to de-
from blistering of mucosal membranes, mainly the oral mucosa, but termine whether these are pathogenic.
also of conjunctival, nasal, anogenital, pharyngeal, laryngeal and oe-
sophageal mucosa, and occasional also from blistering of the skin 4.4.4. Α-enolase, SOD2 and AR
[288]. In 20% of cases the patients have autoantibodies against laminin In patients with membranous nephropathy, IgG4 and IgG1 anti-
332 (Lam 332, previously named laminin 5, epiligrin, kalinin, nicein, or bodies against α-enolase, manganese superoxide dismutase (SOD2) and
BM600) [288–292]. In some patients IgG4 was predominant [293], and aldose reductase (AR) can be detected. Α-enolase is expressed in tubular
serum levels of Lam 332 antibodies was found to correlate with disease kidney cells and in liver epithelia, where it works as a catalyst of the
activity [288]. Interestingly, approximately 30% of anti-laminin-MMP dehydration of 2-phosphoglycerate to phosphoenolpyruvate in glyco-
patients also present with associated malignancy, mostly solid tumours lysis. Anti-α-enolase, anti-SOD2 and anti-AR antibodies were found,
[294–298]. together with the antigen and C5b-9, in laser capture micro-dissected
Lam 332 is located in the skin in the epidermal basement mem- glomeruli of MN patients and in the serum [312,313]. The problem
brane, where it plays a role in keratinocyte adhesion [299,300], and a with these antigens is, that they are primarily located intracellularly
recent study found that Lam 332 antibodies led to the detachment of (e.g. as shown in the human protein atlas, https://www.proteinatlas.
keratinocytes in vitro, suggesting a blocking mechanism similar to org/), which would be a reason to exclude them as targets in IgG4-AID.
Dsg1/3 antibodies in pemphigus [301]. Furthermore, passive transfer There are, however, studies that suggest cell surface expression of these
of total IgG from patients with predominant IgG4 to SCID mice with targets [313], and it was even found that cell-surface expressed α
human skin grafts led to the development of non-inflammatory skin -enolase in endothelial cells acts as a plasminogen receptor [314].
blisters in the human skin [302]. This supports a non-inflammatory Therefore it is thinkable that cell-surface expressed α-enolase, AR or
antibody mechanism by blocking [301], as it would be expected by SOD2 may be a targets for IgG4 antibodies, but it is still equally possible
IgG4. However, further studies are required to pinpoint the patho- that these are intracellularly expressed antigens, and that the IgG4
genicity to the IgG4 subclass, both in vitro and in vivo. antibodies were produced as a by-product, after an initial tissue damage
by another antibody (e.g. PLA2R) that opens up the cell membrane and
4.3. IgG4 autoantibodies with unclear contribution to pathogenicity (Class exposed intracellular antigen to immune cells, which may have led to
III) the production of antibodies against intracellular targets. This is likely,
since PLA2R-IgG4 were present in patient serum simultaneously with
Class III IgG4-AIDs diseases where a) not enough data is present to antibodies against α -enolase, SOD2 and AR [249,315]. Therefore it
estimate the role of IgG4 for pathogenicity, or b) the pathogenicity of cannot be excluded that these antibodies were produced secondary to
IgG4 is unlikely, because a Fc-dependent pathogenic mechanism is PLA2R (or THSD7A, or other antibodies), and would have only re-
present, e.g. complement activation, or other, complex mechanisms are levance as biomarker, similar to autoantibodies against intracellular
at work, which make it unlikely that IgG4 is a key pathogenic player. titin in patients with AChR myasthenia gravis [316]. It is not clear if
these antibodies have a pathogenic mechanism of their own, and if they
4.4. Class IIIa IgG4 autoimmune diseases are the primary pathogenic entity. In vitro studies on the pathogenic
action and passive transfer experiments with purified IgG4 should shed
4.4.1. Caspr1 light on the role of anti- α -enolase, anti-AR and anti-SOD2 antibodies.
There are only few publications on patients with CIDP that have
IgG4 subclass antibodies against Caspr1, which is another protein lo- 4.4.5. P200
cated at the paranode of motoneurons (Fig. 7) or complexed Caspr1/ Patients with antibodies against p200 at the dermal-epidermal-
CNTN1 [154,155,303]. Overall there is not enough data available yet to junction develop an autoimmune subepidermal blistering disease
determine pathogenicity of Caspr1-IgG4. named anti-p200 pemphigoid [317,318]. The patients are usually be-
tween 50 and 70 years of age with a mean age of onset of 65.5 years
4.4.2. NF 140 and NF 186 [319], but disease may also occur in children [320]. Clinically the pa-
In very few patients with CIDP, neurofascin −140 and −186 tients present with tense skin blisters, in approximately 50% of cases
(NF140, NF186) which are also expressed at the paranodes of moto- also with urticarial plaques, and the histology shows subepidermal
neurons (Fig. 7) were found as the main targets of autoantibodies. IgG blistering, often with mild to dense inflammatory infiltrates (neu-
subclasses were described in one study, in which four were pre- trophils, eosinophils) in the upper dermis. P200 is located within the
dominantly of IgG4 and one IgG3 [303], and in one case file additional lower lamina lucida of the basement membrane zone, outside the
antibodies against CNTN1 were present [304]. To date there is not hemidesmosomes and the antibodies were found to be of IgG4 subclass
enough information to determine the pathogenic status of NF140- and [321,322]. In up to 20% of cases, other autoantibodies were present,
NF186-IgG4. including BP180, BP230, lam332 and type VII collagen [319], and one
patient with p200 antibodies also had additional antibodies to lam332
4.4.3. Type IV collagen and CNTN1 [323]. This overlap suggests that intra- and intermolecular
Patients with Goodpasture syndrome present with rapidly epitope spreading events may lead to diversification of the antibodies to

12
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

recognize a range of different antigens. Importantly, the precise epi- also reports of co-occurrence of ANCAs with PLA2R antibodies [349],
tope/antigen in p200 is not clear. P200 was identified from dermal but these were less frequently of the IgG4 subclass [350].
extracts as a protein of 200 kDa size and is an extracellular matrix It is clear that further systematic clinical studies are necessary,
glycoprotein [324]. Up to 90% of the sera also recognized laminin γ1, perhaps with the aim to develop more comprehensive guidelines to help
and it was initially thought that the main antigenic target had been improve diagnosis and to clarify if and how GPA and IgG4-RLD are
discovered [325,324,326]. In one patient, IgG4 anti-laminin γ1 titers associated, and to investigate the role of ANCAs in IgG4-RLD.
correlated with disease severity, but the patient also had anti-laminin It seems to me that the presence of ANCAs, that is, antigen-specific
332 antibodies [327]. Serum from patients with anti-p200 pemphigoid autoantibodies, is certainly somewhat more reminiscent of IgG4-AID
induced dermal-epidermal splitting in cryosections of human skin, but than IgG4-RLD, where antigen-specific IgG4 is rarely observed, on the
the effect was lost when the serum was depleted of antibodies that other hand the presence of fibrosis, elevated serum IgG4 and IgG4+
recognized the c-terminus of human laminin γ1 or laminin 111 or the plasma cell infiltrates are reminiscent of IgG4-RLD. At this point it is not
whole laminin γ1 chain. Furthermore, injection of rabbit derived anti- clear at all whether ANCA-IgG4 positive GPA is an IgG4-AID, an IgG4-
mouse laminin γ1 specific antibodies or active immunization did not RLD, neither or even both.
induce blistering in mice [328,329]. Therefore the role of laminin γ1
antibodies for pathogenicity is unclear, which suggests the presence of a
4.5.2. BP 180 and BP 230
second type of autoautoantibody in the serum. Patient serum was
In a subgroup of patients with bullous pemphigoid (BP), antibodies
clearly pathogenic in an ex vivo model but not when total IgG was
of IgG4 and IgG1 subclass against parts of the basal keratinocyte
depleted, and induced dermal-epidermal separation, suggesting an an-
hemidesmosomal proteins BP antigen 230 (BP 230) and BP antigen 180
tibody-mediated tissue damage [328]. However, we know almost
(BP 180) [351,352–356]. The role of IgG4 in BP is not clear. Auto-
nothing about the role of IgG4 autoantibodies for pathogenicity. The
antibodies bind to the basement membrane zone (BMZ) of the epi-
most important task would be to determine the precise epitope re-
dermis and fix complement, which is observed as C3 deposition at the
cognized by the pathogenic autoantibodies, and study if these are of
BMZ, and recruit granulocytes, and animal studies showed that pa-
IgG4 subclass as well, followed by passive transfer of purified IgG4 from
thogenicity depends on the presence of the Fc, the ability to activate
patient serum.
complement, the presence of neutrophil elastase or matrix metallo-
protease (MMP) and mast cells [357–359]. This pathogenic mechanism
4.5. Class IIIb IgG4 autoimmune diseases
is clearly dependent on IgG1 subclass antibodies, since IgG4 subclass
antibodies do not fix complement and do not recruit immune cells, and
4.5.1. ANCA
the pathogenicity is thought to be mediated solely by binding of the Fab
Antineutrophil cytoplasmic antibodies (ANCA) are present in pa-
fragment. A group of patients does not show complement depositions at
tients with small vessel vasculitis, collected under the term ANCA-as-
the BMZ [19], indicating a second pathogenic mechanism. IgG and F
sociated vasculitides (AAV). One of these AAVs is granulomatosis with
(ab)2 fragments (which bind divalently) from BP patients was found to
polyangiitis (GPA, also named Wegener's granulomatosis). GPA is an
internalize BP 180 by the macropinocytic pathway in keratinocyte
immune-mediated systemic necrotizing vasculitis often affecting the
cultures [360] and induce the detachment of the cells from the dish
upper respiratory tract, lung, and kidney [330]. The antibodies target
[361].
the proteinase 3 (PR3) and myeloperoxidase (MPO), which are ex-
These findings were reproduced in vivo, showing skin detachment,
pressed in neutrophils and monocytes. Interestingly ANCAs are of the
blister formation and loss of BP 180 in the lesions from neonatal BP 180
IgG1, IgG3 and IgG4 subclass, with different IgG subclass profiles in
-humanized, C3-deficient mice [362]. The increased internalization
different patients, some with predominant IgG4, IgG3 or IgG1
suggests also a mechanism of antibodies other than of the IgG4 sub-
[331,332]. It is thought that PR3-ANCA bind to PR3 on the surface of
class, but a detachment of cells by a blocking antibody of IgG4 subclass
neutrophils via the Fab, while simultaneous bind with the Fc to the
could be possible. There is to date no proof that IgG4 is pathogenic in
FcγRs and this leads to neutrophil activation and generation of reactive
the context of BP, indeed one study showed a protective role for IgG4
oxygen species. What is interesting is that isolated patient IgG4 ANCA
by blocking binding of pathogenic IgG1 and IgG3 in vivo [363]. Studies
were able to elicit the activation of neutrophils, despite the fact that
with purified antibody subclasses will be necessary to detect any pa-
IgG4 has reduced affinity for FcγR [331]. Perhaps here Fc-Fc interac-
thogenic role of IgG4.
tions of IgG4 with antibodies of other subclasses may have been in-
volved, or bi-specific IgG4 could cross-link PR3 with another surface
molecule that also led to the activation of neutrophils. A further study 4.5.3. DPPX
with chimeric IgG4-PR3-ANCA with similar results adds evidence for an A group of patients with encephalitis have IgG4 and IgG1 subclass
engagement with Fcγ receptors (FcγR; although not Fcγ RI) or other autoantibodies against dipeptidyl-peptidase-like protein 6 (DPPX).
surface molecules [333]. DPPX is a protein that regulates the expression of voltage-gated po-
According to the literature, it is clinically challenging to distinct tassium channels in the myenteric plexus, hippocampus, cerebellum
between GPA and IgG4-RLD, as GPA mimics IgG4-RLD, with high levels and striatum. Patients with DPPX autoantibodies usually have pro-
of IgG4+ plasma cells, sometimes elevated IgG4 serum levels and the dromal symptoms of diarrhea and substantial weight loss (around
presence of fibrosis [334–336] [334], but there are also cases pre- 20 kg), followed by an onset of neurological symptoms [364–366]. This
senting with both PR3-ANCA positive GPA and IgG4-RLD [337,338] or suggests a link between the brain and the “gut brain” in the context of
ANCA-positive IgG4-RLD in the absence of AAV [339–342]. This led to neuroimmunology. There is further evidence as patients with gastro-
the suggestion that there may be a new overlap syndrome between AAV intestinal diseases also have increased frequencies of antineuronal an-
and IgG4-RLD [343–346]. tibodies, though not against DPPX [367].
Another recent study found no overlap between GPA and IgG4-RLD, DPPX antibodies were found to induce a reversible loss of DPPX and
despite the presence of elevated serum IgG4 [347]. Interestingly a de- Kv4.2 from the cell surface of cultured enteric nervous system neurons,
crease of total IgG4 measured using IgG4:IgG1 RNA ratio was asso- and they also caused hyperexcitation in cultured hippocampal neurons
ciated with remission in patients with GPA [348], putting an emphasis [365,366]. However, the pathogenic mechanism of cross-linking and
on the relevance of IgG4 for this disease, though this study did not look endocytosis of antigen is associated with IgG1–3 antibodies, not with
at ANCA specific IgG4 and the patients selection criteria included that IgG4. This suggests that IgG1–3 and not IgG4 are the pathogenic anti-
elevated (PR3)-ANCA had to be present at one point in the patient's bodies. Unless further evidence turns up, DPPX encephalitis may not be
history, not specifically at the time of the study. In addition, there are an IgG4-AID.

13
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

4.5.4. IgLON5 must add intriguing new clues to the mechanisms of autoimmunization.
Similarly, a pathogenic mechanism for IgG1, but not IgG4, has been
demonstrated in IgLON5 parasomnia. IGLON family member 5 5. Conclusion
(IgLON5) is a glucosyl-phosphatidyl-inositol anchored protein located
in the CNS, and that may mediate membrane stabilization. Anti-IgLON5 IgG4-AID are rare and severe diseases that are hallmarked by the
antibodies are associated with IgLON5 parasomnia which is a severe presence of antigen-specific autoantibodies of the IgG4 subclass. In
autoimmune disease characterized by abnormal sleep behaviour. contrast to IgG4-RLD the autoantibodies have specific antigenic targets
Interestingly, there is also a tauopathy in these patients, which suggests and are directly pathogenic by blocking protein-protein interactions.
a very unusual connection between autoimmunity and neurodegen- IgG4 autoantibodies target antigens that are located in four distinct
eration [368–371]. The IgLON5-IgG1 antibodies have been demon- organs: 1) the central and peripheral nervous system, 2) the kidney, 3)
strated to induce irreversible cross-linking and endocytosis of antigen the skin and mucous membranes and 4) the vascular system including
[18]. The loss of IgLON5 is thought to destabilize the cytoskeleton, soluble antigens in the blood circulation. In this review we could
which may also affect microtubule stability and hyperphosphorylation identify six class I (verified) and seven class II (likely) IgG4 autoimmune
of microtubuli-associated tau protein, which might cause the tauopathy diseases, and identified 16 antigens for class III IgG4 autoimmune dis-
[368]. Importantly, it was demonstrated that the cross-linking and eases, with insufficient data or evidence suggesting other pathogenic
endocytosis depends on divalent binding, as IgG4 or Fabs were not able players than IgG4.
to internalize IgLON5 [18], which emphasizes the relevance of valency
for pathogenicity. Declaration of Competing Interest

4.5.5. Cytokines None.


Autoimmune polyendocrinopathy–candidiasis–ectodermal dys-
trophy (APECED) is a severe autoimmune syndrome caused by muta- Acknowledgements
tions in the autoimmune regulator (AIRE) gene [372]. AIRE is a tran-
scription factor that induces the ectopic expression of peripheral tissue We thank Nick Willcox for scientific advice and feedback on
self-antigens by thymic medullary epithelial cells that negatively select APECED. Figures were created using BioRender Software. We also
autoreactive T-cells and so help to maintain self-tolerance. Therefore, thank Wolfgang Bauer for scientific advice. Inga Koneczny is funded by
mutations in AIRE lead to severe autoimmunity, initially directed a Hertha Firnberg project (project number T996-B30) by the Austrian
against unusual targets – in infancy or childhood – such as the Th17/ Science Fund (FWF). T996-B30
Th22 system, causing chronic mucocutaneous candidiasis (CMC), the
parathyroids, the adrenal cortex and/or the conjunctiva, nails or teeth. References
That may be followed by more typical autoimmune conditions such as
vitiligo, alopecia, thyroid disease or type I diabetes. Usually at diag- [1] Koneczny I. A new classification system for IgG4 autoantibodies. Front Immunol
nosis, most patients have high titres of neutralizing autoantibodies 2018;9.
against cytokines, such as type I interferon (IFN I), interleukin (IL)-17A, [2] Huijbers MG, Querol LA, Niks EH, Plomp JJ, van der Maarel SM, Graus F, et al. The
expanding field of IgG4-mediated neurological autoimmune disorders. Eur J
IL-17F and IL-22 [373–375]. Neurol 2015;22:1151–61.
While anti-IFN I antibodies may protect against further auto- [3] Kridin K, Sagi SZ, Bergman R. Mortality and cause of death in patients with
immunity [373], those against IL-17A/F and IL-22 correlate with re- pemphigus. Acta Derm Venereol 2017;97:607–11.
[4] Niks EH, Kuks JB, Verschuuren JJ. Epidemiology of myasthenia gravis with anti-
duced numbers of Th17/ Th22 cells and with CMC. These cytokine muscle specific kinase antibodies in the Netherlands. J Neurol Neurosurg
antibodies are predominantly of IgG1 and IgG4 subclass, sometimes Psychiatry 2007;78:417–8.
exclusively” Th2-type” IgG4s [375]. Patients with thymomas show in- [5] Carr AS, Cardwell CR, McCarron PO, McConville J. A systematic review of popu-
lation based epidemiological studies in myasthenia gravis. BMC Neurol
triguing (a) serological similarities and (b) clinical differences: (a) high 2010;10:46.
titres of antibodies neutralizing IFN-Is (≥70%), and/or IL-17 s or IL-22, [6] Broers MC, Bunschoten C, Nieboer D, Lingsma HF, Jacobs BC. Incidence and
again correlating with CMC (< 5%; [375]); (b) endocrine auto- prevalence of chronic inflammatory demyelinating Polyradiculoneuropathy: a
systematic review and meta-analysis. Neuroepidemiology 2019;52:161–72.
immunity is infrequent (< 5%) whereas myasthenia gravis is common
[7] Cortese A, Lombardi R, Briani C, Callegari I, Benedetti L, Manganelli F, et al.
(≥30%). The obvious connecting thread with APECED is that the Antibodies to neurofascin, contactin-1, and contactin-associated protein 1 in CIDP:
neoplastic epithelial cells in most thymomas fail to express AIRE, but Clinical relevance of IgG isotype. Neurol Neuroimmunol Neuroinflam 2020;7.
nevertheless generate numerous developing T cells in a disorganised [8] Hubner F, Recke A, Zillikens D, Linder R, Schmidt E. Prevalence and age dis-
tribution of pemphigus and pemphigoid diseases in Germany. J Invest Dermatol
environment where failure to tolerize seems likely (Nick Willcox, per- 2016;136:2495–8.
sonal communication). [9] Schmidt E, Kasperkiewicz M, Joly P. Pemphigus. Lancet 2019;394:882–94.
Since APECED is a very complex disease, it would be interesting to [10] Hu X, Dallinga-Thie GM, Hovingh GK, Chang SY, Sandoval NP, Dang TLP, et al.
GPIHBP1 autoantibodies in a patient with unexplained chylomicronemia. J Clin
study pathogenic and protective functions of IgG4 anti-cytokine anti- Lipidol 2017;11:964–71.
bodies by passively transferring purified patient IgG4 to healthy ex- [11] Beigneux AP, Miyashita K, Ploug M, Blom DJ, Ai M, Linton MF, et al.
perimental animals. To date, that has only been undertaken with one Autoantibodies against GPIHBP1 as a cause of hypertriglyceridemia. N Engl J Med
2017;376:1647–58.
anti-IL-22 IgG1 antibody cloned from an APECED patient, which im- [12] Koneczny I, Herbst R. Myasthenia gravis: Pathogenic effects of autoantibodies on
paired fungal clearance [376]. In a mouse model with Aire deficiency, neuromuscular architecture, cells 8. 2019.
autoreactive B-cells appeared important mainly for priming T-cells ra- [13] Duan T, Smith AJ, Verkman AS. Complement-independent bystander injury in
AQP4-IgG seropositive neuromyelitis optica produced by antibody-dependent
ther than producing pathogenic antibodies [377]. However, there is a
cellular cytotoxicity. Acta Neuropathol Commun 2019;7:112.
new rat model for APECED that reflects more of the patients' symptoms [14] Diny NL, Rose NR, Cihakova D. Eosinophils in autoimmune diseases. Front
and serology, with Th2-type IgG1s and IgMs and IgAs, against cytokines Immunol 2017;8:484.
[15] Wener MH. Immune Complexes in Systemic Lupus Erythematosus. In: Lahita RG,
[378]. It would be interesting to study the contribution of the different
editor. Systemic Lupus Erythematosus. 5th ed.Academic Press; 2011. p. 321–38.
antibody subclasses in these rats by passively transferring their cloned [16] Drachman DB, Angus CW, Adams RN, Michelson JD, Hoffman GJ. Myasthenic
IgGs to normal animals. Whether these thymus-associated autoimmune antibodies cross-link acetylcholine receptors to accelerate degradation. N Engl J
syndromes should be added to the list of’IgG4 autoimmune diseases' is Med 1978;298:1116–22.
[17] Heinemann S, Bevan S, Kullberg R, Lindstrom J, Rice J. Modulation of acet-
debatable, as the proportion of IgG4 to total autoantibody varies so ylcholine receptor by antibody against the receptor. Proc Natl Acad Sci U S A
much between patients, and the pathogenic potential of any of the 1977;74:3090–4.
antibodies has yet to be definitively established. In the meantime, they [18] Sabater L, Planaguma J, Dalmau J, Graus F. Cellular investigations with human

14
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

antibodies associated with the anti-IgLON5 syndrome. J Neuroinflammation phagocytosis. Eur J Immunol 1994;24:2542–7.
2016;13:226. [51] Canfield SM, Morrison SL. The binding affinity of human IgG for its high affinity fc
[19] Carvajal-Gonzalez A, Leite MI, Waters P, Woodhall M, Coutinho E, Balint B, et al. receptor is determined by multiple amino acids in the CH2 domain and is modu-
Glycine receptor antibodies in PERM and related syndromes: characteristics, lated by the hinge region. J Exp Med 1991;173:1483–91.
clinical features and outcomes. Brain 2014;137:2178–92. [52] Sondermann P, Huber R, Oosthuizen V, Jacob U. The 3.2-a crystal structure of the
[20] Hughes EG, Peng X, Gleichman AJ, Lai M, Zhou L, Tsou R, et al. Cellular and human IgG1 fc fragment-fc gammaRIII complex. Nature 2000;406:267–73.
synaptic mechanisms of anti-NMDA receptor encephalitis. J Neurosci [53] Shields RL, Namenuk AK, Hong K, Meng YG, Rae J, Briggs J, et al. High resolution
2010;30:5866–75. mapping of the binding site on human IgG1 for fc gamma RI, fc gamma RII, fc
[21] Barcellini W. New insights in the pathogenesis of autoimmune hemolytic Anemia. gamma RIII, and FcRn and design of IgG1 variants with improved binding to the fc
Transfusion Med Hemother 2015;42:287–93. gamma R. J Biol Chem 2001;276:6591–604.
[22] Ludwig RJ, Vanhoorelbeke K, Leypoldt F, Kaya Z, Bieber K, McLachlan SM, et al. [54] Radaev S, Motyka S, Fridman WH, Sautes-Fridman C, Sun PD. The structure of a
Mechanisms of autoantibody-induced pathology. Front Immunol 2017;8:603. human type III Fcgamma receptor in complex with fc. J Biol Chem
[23] Sadler JE. Pathophysiology of thrombotic thrombocytopenic purpura. Blood 2001;276:16469–77.
2017;130:1181–8. [55] Davies AM, Rispens T, den Bleker TH, McDonnell JM, Gould HJ, Aalberse RC, et al.
[24] Rapoport B, McLachlan SM. The thyrotropin receptor in Graves’ disease. Thyroid Crystal structure of the human IgG4 C(H)3 dimer reveals the role of Arg409 in the
2007;17:911–22. mechanism of fab-arm exchange. Mol Immunol 2013;54:1–7.
[25] Rapoport B, Chazenbalk GD, Jaume JC, McLachlan SM. The thyrotropin (TSH) [56] Labrijn AF, Rispens T, Meesters J, Rose RJ, den Bleker TH, Loverix S, et al. Species-
receptor: interaction with TSH and autoantibodies. Endocr Rev 1998;19:673–716. specific determinants in the IgG CH3 domain enable fab-arm exchange by affecting
[26] Koneczny I, Cossins J, Waters P, Beeson D, Vincent A. MuSK myasthenia gravis the noncovalent CH3-CH3 interaction strength. J Immunol 2011;187:3238–46.
IgG4 disrupts the interaction of LRP4 with MuSK but both IgG4 and IgG1-3 can [57] Blech M, Horer S, Kuhn AB, Kube S, Goddeke H, Kiefer H, et al. Structure of a
disperse preformed agrin-independent AChR clusters. PLoS One 2013;8:e80695. therapeutic full-length anti-NPRA IgG4 antibody: dissecting conformational di-
[27] Kasperkiewicz M, Ellebrecht CT, Takahashi H, Yamagami J, Zillikens D, Payne AS, versity. Biophys J 2019;116:1637–49.
et al. Pemphigus. Nat Rev Dis Primers 2017;3:17026. [58] Schuurman J, Perdok GJ, Gorter AD, Aalberse RC. The inter-heavy chain disulfide
[28] Lichtenstein LM, Holtzman NA, Burnett LS. A quantitative in vitro study of the bonds of IgG4 are in equilibrium with intra-chain disulfide bonds. Mol Immunol
chromatographic distribution and immunoglobulin characteristics of human 2001;38:1–8.
blocking antibody. J Immunol 1968;101:317–24. [59] Bloom JW, Madanat MS, Marriott D, Wong T, Chan SY. Intrachain disulfide bond
[29] Aalberse RC, van der Gaag R, van Leeuwen J. Serologic aspects of IgG4 antibodies. in the core hinge region of human IgG4. Protein Sci 1997;6:407–15.
I. Prolonged immunization results in an IgG4-restricted response. J Immunol [60] Angal S, King DJ, Bodmer MW, Turner A, Lawson AD, Roberts G, et al. A single
1983;130:722–6. amino acid substitution abolishes the heterogeneity of chimeric mouse/human
[30] Nakagawa T, Miyamoto T. The role of IgG4 as blocking antibodies in asthmatics (IgG4) antibody. Mol Immunol 1993;30:105–8.
and in bee keepers. Int Arch Allergy Appl Immunol 1985;77:204–5. [61] King DJ, Adair JR, Angal S, Low DC, Proudfoot KA, Lloyd JC, et al. Expression,
[31] Platts-Mills T, Vaughan J, Squillace S, Woodfolk J, Sporik R. Sensitisation, asthma, purification and characterization of a mouse-human chimeric antibody and chi-
and a modified Th2 response in children exposed to cat allergen: a population- meric Fab’ fragment. Biochem J 1992;281(Pt 2):317–23.
based cross-sectional study. Lancet 2001;357:752–6. [62] Margni RA, Binaghi RA. Nonprecipitating asymmetric antibodies. Annu Rev
[32] Pinck JR, Milstein C. Disulphide bridges of a human immunoglobulin G protein. Immunol 1988;6:535–54.
Nature 1967;216:941–2. [63] van der Zee JS, van Swieten P, Aalberse RC. Serologic aspects of IgG4 antibodies.
[33] Frangione B, Milstein C. Disulphide bridges of immunoglobin G-1 heavy chains. II. IgG4 antibodies form small, nonprecipitating immune complexes due to func-
Nature 1967;216:939–41. tional monovalency. J Immunol 1986;137:3566–71.
[34] Frangione B, Milstein C, Pink JR. Structural studies of immunoglobulin G. Nature [64] Petersen JG, Dorrington KJ. An in vitro system for studying the kinetics of inter-
1969;221:145–8. chain disulfide bond formation in immunoglobulin G. J Biol Chem
[35] Tian X, Vestergaard B, Thorolfsson M, Yang Z, Rasmussen HB, Langkilde AE. In- 1974;249:5633–41.
depth analysis of subclass-specific conformational preferences of IgG antibodies. [65] Colcher D, Milenic D, Roselli M, Raubitschek A, Yarranton G, King D, et al.
IUCrJ 2015;2:9–18. Characterization and biodistribution of recombinant and recombinant/chimeric
[36] Konig N, Paulus M, Julius K, Schulze J, Voetz M, Tolan M. Antibodies under constructs of monoclonal antibody B72.3. Cancer Res 1989;49:1738–45.
pressure: a small-angle X-ray scattering study of immunoglobulin G under high [66] van der Neut Kolfschoten M, Schuurman J, Losen M, Bleeker WK, Martinez-
hydrostatic pressure. Biophys Chem 2017;231:45–9. Martinez P, Vermeulen E, et al. Anti-inflammatory activity of human IgG4 anti-
[37] Schroeder Jr. HW, Cavacini L. Structure and function of immunoglobulins. J bodies by dynamic fab arm exchange. Science 2007;317:1554–7.
Allergy Clin Immunol 2010;125:S41–52. [67] Rispens T, Davies AM, Ooijevaar-de Heer P, Absalah S, Bende O, Sutton BJ, et al.
[38] Lu Y, Harding SE, Michaelsen TE, Longman E, Davis KG, Ortega A, et al. Solution Dynamics of inter-heavy chain interactions in human immunoglobulin G (IgG)
conformation of wild-type and mutant IgG3 and IgG4 immunoglobulins using subclasses studied by kinetic fab arm exchange. J Biol Chem 2014;289:6098–109.
crystallohydrodynamics: possible implications for complement activation. Biophys [68] Young E, Lock E, Ward DG, Cook A, Harding S, Wallis GL. Estimation of polyclonal
J 2007;93:3733–44. IgG4 hybrids in normal human serum. Immunology 2014;142:406–13.
[39] Abe Y, Gor J, Bracewell DG, Perkins SJ, Dalby PA. Masking of the fc region in [69] Koneczny I, Stevens JAA, De Rosa A, Huda S, Huijbers MG, Saxena A, et al. IgG4
human IgG4 by constrained X-ray scattering modelling: implications for antibody autoantibodies against muscle-specific kinase undergo fab-arm exchange in
function and therapy. Biochem J 2010;432:101–11. myasthenia gravis patients. J Autoimmun 2017;77:104–15.
[40] Diebolder CA, Beurskens FJ, de Jong RN, Koning RI, Strumane K, Lindorfer MA, [70] Aalberse RC, Stapel SO, Schuurman J, Rispens T. Immunoglobulin G4: an odd
et al. Complement is activated by IgG hexamers assembled at the cell surface. antibody. Clin Exp Allergy 2009;39:469–77.
Science 2014;343:1260–3. [71] Collins AM, Davies JM. Enhanced cell-binding by allergen multimers: how com-
[41] Lhotta K, Wurzner R, Konig P. Glomerular deposition of mannose-binding lectin in plex is it? Immunol Cell Biol 2013;91:115–7.
human glomerulonephritis. Nephrol Dial Transplant 1999;14:881–6. [72] van der Zee JS, van Swieten P, Aalberse RC. Inhibition of complement activation
[42] Yang Y, Wang C, Jin L, He F, Li C, Gao Q, et al. IgG4 anti-phospholipase A2 re- by IgG4 antibodies. Clin Exp Immunol 1986;64:415–22.
ceptor might activate lectin and alternative complement pathway meanwhile in [73] Davies AM, Rispens T, Ooijevaar-de Heer P, Gould HJ, Jefferis R, Aalberse RC,
idiopathic membranous nephropathy: an inspiration from a cross-sectional study. et al. Structural determinants of unique properties of human IgG4-fc. J Mol Biol
Immunol Res 2016;64:919–30. 2014;426:630–44.
[43] Segawa Y, Hisano S, Matsushita M, Fujita T, Hirose S, Takeshita M, et al. IgG [74] Rispens T, Ooievaar-De Heer P, Vermeulen E, Schuurman J, van der Neut
subclasses and complement pathway in segmental and global membranous ne- Kolfschoten M, Aalberse RC. Human IgG4 binds to IgG4 and conformationally
phropathy. Pediatr Nephrol 2010;25:1091–9. altered IgG1 via fc-fc interactions. J Immunol 2009;182:4275–81.
[44] Ma H, Beck LJ, Salant D. Membranous nephropathy-associated anti-phospholipase [75] Zack DJ, Stempniak M, Wong AL, Weisbart RH. Localization of an fc-binding re-
A2 receptor IgG4 autoantibodies activate the lectin complement pathway (ab- activity to the constant region of human IgG4. Implications for the pathogenesis of
stract). J Am Soc Nephrol 2011;22:62A. rheumatoid arthritis. J Immunol 1995;155:5057–63.
[45] Hayashi N, Okada K, Matsui Y, Fujimoto K, Adachi H, Yamaya H, et al. Glomerular [76] Kawa S, Kitahara K, Hamano H, Ozaki Y, Arakura N, Yoshizawa K, et al. A novel
mannose-binding lectin deposition in intrinsic antigen-related membranous ne- immunoglobulin-immunoglobulin interaction in autoimmunity. PLoS One
phropathy. Nephrol Dial Transplant 2017;33(5):832–40. 2008;3:e1637.
[46] Beck Jr. LH, Salant DJ. Membranous nephropathy: from models to man. J Clin [77] Cohen PL, Cheek RL, Hadler JA, Yount WJ, Eisenberg RA. The subclass distribution
Invest 2014;124:2307–14. of human IgG rheumatoid factor. J Immunol 1987;139:1466–71.
[47] Malhotra R, Wormald MR, Rudd PM, Fischer PB, Dwek RA, Sim RB. Glycosylation [78] Detlefsen S, Brasen JH, Zamboni G, Capelli P, Kloppel G. Deposition of comple-
changes of IgG associated with rheumatoid arthritis can activate complement via ment C3c, immunoglobulin (Ig)G4 and IgG at the basement membrane of pan-
the mannose-binding protein. Nat Med 1995;1:237–43. creatic ducts and acini in autoimmune pancreatitis. Histopathology
[48] Roos A, Bouwman LH, van Gijlswijk-Janssen DJ, Faber-Krol MC, Stahl GL, Daha 2010;57:825–35.
MR. Human IgA activates the complement system via the mannan-binding lectin [79] Ma H, Sandor DG, Beck Jr. LH. The role of complement in membranous nephro-
pathway. J Immunol 2001;167:2861–8. pathy. Semin Nephrol 2013;33:531–42.
[49] Tao MH, Smith RI, Morrison SL. Structural features of human immunoglobulin G [80] Rees-Roberts D, Mullen LM, Gounaris K, Selkirk ME. Inactivation of the comple-
that determine isotype-specific differences in complement activation. J Exp Med ment anaphylatoxin C5a by secreted products of parasitic nematodes. Int J
1993;178:661–7. Parasitol 2010;40:527–32.
[50] Brekke OH, Michaelsen TE, Aase A, Sandin RH, Sandlie I. Human IgG isotype- [81] Maizels RM, Yazdanbakhsh M. Immune regulation by helminth parasites: cellular
specific amino acid residues affecting complement-mediated cell lysis and and molecular mechanisms. Nat Rev Immunol 2003;3:733–44.

15
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

[82] Babu S, Blauvelt CP, Kumaraswami V, Nutman TB. Regulatory networks induced 2018;37:1783–90.
by live parasites impair both Th1 and Th2 pathways in patent lymphatic filariasis: [113] Bos WH, Bartelds GM, Vis M, van der Horst AR, Wolbink GJ, van de Stadt RJ, et al.
implications for parasite persistence. J Immunol 2006;176:3248–56. Preferential decrease in IgG4 anti-citrullinated protein antibodies during treat-
[83] Adjobimey T, Hoerauf A. Induction of immunoglobulin G4 in human filariasis: an ment with tumour necrosis factor blocking agents in patients with rheumatoid
indicator of immunoregulation. Ann Trop Med Parasitol 2010;104:455–64. arthritis. Ann Rheum Dis 2009;68:558–63.
[84] Prodjinotho UF, Hoerauf A, Adjobimey T. IgG4 antibodies from patients with [114] Pan Q, Lan Q, Peng Y, Cai J, Zheng J, Dickerson C, et al. Nature, functions, and
asymptomatic bancroftian filariasis inhibit the binding of IgG1 and IgG2 to C1q in clinical implications of IgG4 autoantibodies in systemic lupus erythematosus and
a fc-fc-dependent mechanism. Parasitol Res 2019;118:2957–68. rheumatoid arthritis. Discov Med 2017;23:169–74.
[85] Vidarsson G, Dekkers G, Rispens T. IgG subclasses and allotypes: from structure to [115] van de Stadt LA, de Vrieze H, Derksen NI, Brouwer M, Wouters D, van
effector functions. Front Immunol 2014;5:520. Schaardenburg D, et al. Antibodies to IgG4 hinge can be found in rheumatoid
[86] Nirula A, Glaser SM, Kalled SL, Taylor FR. What is IgG4? A review of the biology of arthritis patients during all stages of disease and may exacerbate chronic antibody-
a unique immunoglobulin subtype. Curr Opin Rheumatol 2011;23:119–24. mediated inflammation. Arthritis Rheum 2014;66:1133–40.
[87] Lighaam LC, Rispens T. The Immunobiology of immunoglobulin G4. Semin Liver [116] Falkenburg WJ, van Schaardenburg D, Ooijevaar-de Heer P, Tsang ASMW, Bultink
Dis 2016;36:200–15. IE, Voskuyl AE, et al. Anti-hinge antibodies recognize IgG subclass- and protease-
[88] Van der Zee S, Aalberse RC. IgG4 and hyposensitization. N Engl Reg Allergy Proc restricted Neoepitopes. J Immunol 2017;198:82–93.
1987;8:389–91. [117] Umehara H, Okazaki K, Kawano M, Tanaka Y. The front line of research into
[89] Margni RA, Perdigon G, Abatangelo C, Gentile T, Binaghi RA. Immunobiological immunoglobin G4-related disease - do autoantibodies cause immunoglobin G4-
behaviour of rabbit precipitating and non-precipitating (co-precipitating) anti- related disease? Mod Rheumatol 2019;29:214–8.
bodies. Immunology 1980;41:681–6. [118] Watanabe T, Minaga K, Kamata K, Kudo M, Strober W. Mechanistic insights into
[90] van Toorenenbergen AW, Aalberse RC. IgG4 and release of histamine from human autoimmune pancreatitis and IgG4-related disease. Trends Immunol
peripheral blood leukocytes. Int Arch Allergy Appl Immunol 1982;67:117–22. 2018;39:874–89.
[91] Aalberse RC, Dieges PH, Knul-Bretlova V, Vooren P, Aalbers M, van Leeuwen J. [119] Pagliari D, Cianci R, Rigante D. Autoimmune pancreatitis in children: the impact
IgG4 as a blocking antibody. Clin Rev Allergy 1983;1:289–302. of immune system in a challenging disease. Autoimmun Rev 2019;18:209–10.
[92] James LK, Bowen H, Calvert RA, Dodev TS, Shamji MH, Beavil AJ, et al. Allergen [120] Akiyama M, Kaneko Y, Takeuchi T. Characteristics and prognosis of IgG4-related
specificity of IgG(4)-expressing B cells in patients with grass pollen allergy un- periaortitis/periarteritis: a systematic literature review. Autoimmun Rev
dergoing immunotherapy. J Allergy Clin Immunol 2012;130:663–70. [e3]. 2019;18:102354.
[93] Santos AF, James LK, Bahnson HT, Shamji MH, Couto-Francisco NC, Islam S, et al. [121] Morales AT, Cignarella AG, Jabeen IS, Barkin JS, Mirsaeidi M. An update on IgG4-
IgG4 inhibits peanut-induced basophil and mast cell activation in peanut-tolerant related lung disease. Eur J Intern Med 2019;66:18–24.
children sensitized to peanut major allergens. J Allergy Clin Immunol [122] Romano A, Rigante D, Cipolla C. Autoimmune phenomena involving the pituitary
2015;135:1249–56. gland in children: new developing data about diagnosis and treatment.
[94] Kemeny DM, MacKenzie-Mills M, Harries MG, Youlten LJ, Lessof MH. Antibodies Autoimmun Rev 2019;18:102363.
to purified bee venom proteins and peptides. II. A detailed study of changes in IgE [123] Stone JH. IgG4-related disease: nomenclature, clinical features, and treatment.
and IgG antibodies to individual bee venom antigens. J Allergy Clin Immunol Semin Diagn Pathol 2012;29:177–90.
1983;72:376–85. [124] Umehara H, Okazaki K, Kawano M, Mimori T, Chiba T. How to diagnose IgG4-
[95] Kemeny DM, McKenzie-Mills M, Harries MG, Youlten LJ, Lessof MH. Changes in related disease. Ann Rheum Dis 2017;76(11):e46.
the levels of anti-phospholipase A2 and hyaluronidase antibodies during bee [125] Bozzalla Cassione E, Stone JH. IgG4-related disease. Curr Opin Rheumatol
venom immunotherapy. Monogr Allergy 1983;18:150–2. 2017;29:223–7.
[96] Resende RO, Ynoue LH, Miranda JS, de Almeida KC, Silva DAO, Sopelete MC, et al. [126] Shiokawa M, Kodama Y, Kuriyama K, Yoshimura K, Tomono T, Morita T, et al.
IgE, IgG1, and IgG4 reactivity to Dermatophagoides pteronyssinus glycosylated Pathogenicity of IgG in patients with IgG4-related disease. Gut 2016;65:1322–32.
extract in allergic patients. Biomed Res Int 2019;2019:9840890. [127] Hubers LM, Vos H, Schuurman AR, Erken R, Oude Elferink RP, Burgering B, et al.
[97] Subbarayal B, Schiller D, Mobs C, de Jong NW, Ebner C, Reider N, et al. Kinetics, Annexin A11 is targeted by IgG4 and IgG1 autoantibodies in IgG4-related disease.
cross-reactivity, and specificity of bet v 1-specific IgG4 antibodies induced by Gut 2018;67:728–35.
immunotherapy with birch pollen. Allergy 2013;68:1377–86. [128] Perugino CA, AlSalem SB, Mattoo H, Della-Torre E, Mahajan V, Ganesh G, et al.
[98] van Neerven RJ, Wikborg T, Lund G, Jacobsen B, Brinch-Nielsen A, Arnved J, et al. Identification of galectin-3 as an autoantigen in patients with IgG4-related disease.
Blocking antibodies induced by specific allergy vaccination prevent the activation J Allergy Clin Immunol 2019;143:736–45. [e6].
of CD4+ T cells by inhibiting serum-IgE-facilitated allergen presentation. J [129] Yamada K, Zuka M, Ito K, Mizuguchi K, Kakuchi Y, Onoe T, et al. LatY136F knock-
Immunol 1999;163:2944–52. in mouse model for human IgG4-related disease. PLoS One 2018;13:e0198417.
[99] Bodtger U, Ejrnaes AM, Hummelshoj L, Jacobi HH, Poulsen LK, Svenson M. Is [130] Witebsky E, Rose NR, Terplan K, Paine JR, Egan RW. Chronic thyroiditis and
immunotherapy-induced birch-pollen-specific IgG4 a marker for decreased al- autoimmunization. JAMA 1957;164:1439–47.
lergen-specific sensitivity? Int Arch Allergy Immunol 2005;136:340–6. [131] Rose NR, Bona C. Defining criteria for autoimmune diseases(Witebsky’s postulates
[100] Crescioli S, Correa I, Karagiannis P, Davies AM, Sutton BJ, Nestle FO, et al. IgG4 revisited). Immunol Today 1993;14:426–30.
characteristics and functions in Cancer immunity. Curr Allergy Asthma Rep [132] Naparstek Y, Plotz PH. The role of autoantibodies in autoimmune disease. Annu
2016;16:7. Rev Immunol 1993;11:79–104.
[101] Lopes F, Matisz C, Reyes JL, Jijon H, Al-Darmaki A, Kaplan GG, et al. Helminth [133] Hoch W, McConville J, Helms S, Newsom-Davis J, Melms A, Vincent A. Auto-
regulation of immunity: a three-pronged approach to treat colitis. Inflamm Bowel antibodies to the receptor tyrosine kinase MuSK in patients with myasthenia gravis
Dis 2016;22:2499–512. without acetylcholine receptor antibodies. Nat Med 2001;7:365–8.
[102] Maizels RM. Regulation of immunity and allergy by helminth parasites. Allergy [134] McConville J, Farrugia ME, Beeson D, Kishore U, Metcalfe R, Newsom-Davis J,
2019;75(3):524–34. et al. Detection and characterization of MuSK antibodies in seronegative myas-
[103] Elliott DE, Weinstock JV. Helminth-host immunological interactions: prevention thenia gravis. Ann Neurol 2004;55:580–4.
and control of immune-mediated diseases. Ann N Y Acad Sci 2012;1247:83–96. [135] Huijbers MG, Vink AF, Niks EH, Westhuis RH, van Zwet EW, de Meel RH, et al.
[104] Marple A, Wu W, Shah S, Zhao Y, Du P, Gause WC, et al. Cutting edge: helminth Longitudinal epitope mapping in MuSK myasthenia gravis: implications for disease
coinfection blocks effector differentiation of CD8 T cells through alternate host severity. J Neuroimmunol 2016;291:82–8.
Th2- and IL-10-mediated responses. J Immunol 2017;198:634–9. [136] Takamori M, Nakamura T, Motomura M. Antibodies against Wnt receptor of
[105] Boctor FN, Peter JB. IgG subclasses in human chronic schistosomiasis: over-pro- muscle-specific tyrosine kinase in myasthenia gravis. J Neuroimmunol
duction of schistosome-specific and non-specific IgG4. Clin Exp Immunol 2013;254:183–6.
1990;82:574–8. [137] Otsuka K, Ito M, Ohkawara B, Masuda A, Kawakami Y, Sahashi K, et al. Collagen Q
[106] Satoguina JS, Adjobimey T, Arndts K, Hoch J, Oldenburg J, Layland LE, et al. Tr1 and anti-MuSK autoantibody competitively suppress agrin/LRP4/MuSK signaling.
and naturally occurring regulatory T cells induce IgG4 in B cells through GITR/ Sci Rep 2015;5:13928.
GITR-L interaction, IL-10 and TGF-beta. Eur J Immunol 2008;38:3101–13. [138] Zhang W, Coldefy AS, Hubbard SR, Burden SJ. Agrin binds to the N-terminal re-
[107] Tang CL, Zou JN, Zhang RH, Liu ZM, Mao CL. Helminths protect against type 1 gion of Lrp4 protein and stimulates association between Lrp4 and the first im-
diabetes: effects and mechanisms. Parasitol Res 2019;118:1087–94. munoglobulin-like domain in muscle-specific kinase (MuSK). J Biol Chem
[108] Shen P, Fillatreau S. Suppressive functions of B cells in infectious diseases. Int 2011;286:40624–30.
Immunol 2015;27:513–9. [139] Huijbers MG, Zhang W, Klooster R, Niks EH, Friese MB, Straasheijm KR, et al.
[109] Chapuy-Regaud S, Nogueira L, Clavel C, Sebbag M, Vincent C, Serre G. IgG sub- MuSK IgG4 autoantibodies cause myasthenia gravis by inhibiting binding between
class distribution of the rheumatoid arthritis-specific autoantibodies to ci- MuSK and Lrp4. Proc Natl Acad Sci U S A 2013;110(51):20783–8.
trullinated fibrin. Clin Exp Immunol 2005;139:542–50. [140] Koneczny I. Potential mechanisms in MuSK myasthenia gravis. Book Potential
[110] Panza F, Pratesi F, Valoriani D, Migliorini P. Immunoglobulin G subclass profile of mechanisms in MuSK myasthenia gravis. Oxford University; 2014.
anticitrullinated peptide antibodies specific for Epstein Barr virus-derived and [141] Huijbers MG, Vergoossen DL, Fillie-Grijpma YE, van Es IE, Koning MT, Slot LM,
histone-derived citrullinated peptides. J Rheumatol 2014;41:407–8. et al. MuSK myasthenia gravis monoclonal antibodies: Valency dictates patho-
[111] Lundstrom SL, Fernandes-Cerqueira C, Ytterberg AJ, Ossipova E, Hensvold AH, genicity. Neurol Neuroimmunol Neuroinflam 2019;6:e547.
Jakobsson PJ, et al. IgG antibodies to cyclic citrullinated peptides exhibit profiles [142] Mori S, Kubo S, Akiyoshi T, Yamada S, Miyazaki T, Hotta H, et al. Antibodies
specific in terms of IgG subclasses, fc-glycans and a fab-peptide sequence. PLoS against muscle-specific kinase impair both presynaptic and postsynaptic functions
One 2014;9:e113924. in a murine model of myasthenia gravis. Am J Pathol 2012;180:798–810.
[112] Engelmann R, Nekarda S, Kuthning D, Kneitz C, Muller-Hilke B. Decreased IgG4 [143] Morsch M, Reddel SW, Ghazanfari N, Toyka KV, Phillips WD. Muscle specific ki-
ACPA levels in responders and increased CD1c(+) classical dendritic cells in non- nase autoantibodies cause synaptic failure through progressive wastage of post-
responders of patients with rheumatoid arthritis under therapy. Clin Rheumatol synaptic acetylcholine receptors. Exp Neurol 2012;237:286–95.

16
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

[144] Selcen D, Fukuda T, Shen XM, Engel AG. Are MuSK antibodies the primary cause Neurofascin-155 antibodies. Tremor Other Hyperkinet Mov 2018;8:606.
of myasthenic symptoms? Neurology 2004;62:1945–50. [174] Briani C, Salvalaggio A, Ruiz M, Cacciavillani M, Rinaldi F, Callegari I, et al.
[145] Viegas S, Jacobson L, Waters P, Cossins J, Jacob S, Leite MI, et al. Passive and Tongue tremor in neurofascin-155 IgG4 seropositive chronic inflammatory poly-
active immunization models of MuSK-ab positive myasthenia: electrophysiological radiculoneuropathy. J Neuroimmunol 2019;330:178–80.
evidence for pre and postsynaptic defects. Exp Neurol 2012;234:506–12. [175] Manso C, Querol L, Lleixa C, Poncelet M, Mekaouche M, Vallat JM, et al. Anti-
[146] Niks EH, Kuks JB, Wokke JH, Veldman H, Bakker E, Verschuuren JJ, et al. Pre- and Neurofascin-155 IgG4 antibodies prevent paranodal complex formation in vivo. J
postsynaptic neuromuscular junction abnormalities in musk myasthenia. Muscle Clin Invest 2019;130.
Nerve 2010;42:283–8. [176] Dutta DJ, Woo DH, Lee PR, Pajevic S, Bukalo O, Huffman WC, et al. Regulation of
[147] Yumoto N, Kim N, Burden SJ. Lrp4 is a retrograde signal for presynaptic differ- myelin structure and conduction velocity by perinodal astrocytes. Proc Natl Acad
entiation at neuromuscular synapses. Nature 2012;489:438–42. Sci U S A 2018;115:11832–7.
[148] Wu H, Lu Y, Shen C, Patel N, Gan L, Xiong WC, et al. Distinct roles of muscle and [177] Thomas MR, de Groot R, Scully MA, Crawley JT. Pathogenicity of anti-ADAMTS13
motoneuron LRP4 in neuromuscular junction formation. Neuron 2012;75:94–107. autoantibodies in acquired thrombotic thrombocytopenic Purpura. Ebiomedicine
[149] Sabre L, Evoli A, Punga AR. Cognitive dysfunction in mice with passively induced 2015;2:942–52.
MuSK antibody seropositive myasthenia gravis. J Neurol Sci 2019;399:15–21. [178] Tsai HM, Lian EC. Antibodies to von Willebrand factor-cleaving protease in acute
[150] Faivre-Sarrailh C, Devaux JJ. Neuro-glial interactions at the nodes of Ranvier: thrombotic thrombocytopenic purpura. N Engl J Med 1998;339:1585–94.
implication in health and diseases. Front Cell Neurosci 2013;7:196. [179] Schaller M, Vogel M, Kentouche K, Lammle B, Kremer Hovinga JA. The splenic
[151] Peles E, Nativ M, Lustig M, Grumet M, Schilling J, Martinez R, et al. Identification autoimmune response to ADAMTS13 in thrombotic thrombocytopenic purpura
of a novel contactin-associated transmembrane receptor with multiple domains contains recurrent antigen-binding CDR3 motifs. Blood 2014;124:3469–79.
implicated in protein-protein interactions. EMBO J 1997;16:978–88. [180] Sinkovits G, Szilagyi A, Farkas P, Inotai D, Szilvasi A, Tordai A, et al.
[152] Rios JC, Melendez-Vasquez CV, Einheber S, Lustig M, Grumet M, Hemperly J, et al. Concentration and subclass distribution of anti-ADAMTS13 IgG autoantibodies in
Contactin-associated protein (Caspr) and contactin form a complex that is targeted different stages of acquired idiopathic thrombotic thrombocytopenic Purpura.
to the paranodal junctions during myelination. J Neurosci 2000;20:8354–64. Front Immunol 2018;9:1646.
[153] Charles P, Tait S, Faivre-Sarrailh C, Barbin G, Gunn-Moore F, Denisenko-Nehrbass [181] Kremer Hovinga JA, Coppo P, Lammle B, Moake JL, Miyata T, Vanhoorelbeke K.
N, et al. Neurofascin is a glial receptor for the paranodin/Caspr-contactin axonal Thrombotic thrombocytopenic purpura. Nat Rev Dis Primers 2017;3:17020.
complex at the axoglial junction. Curr Biol 2002;12:217–20. [182] Ostertag EM, Bdeir K, Kacir S, Thiboutot M, Gulendran G, Yunk L, et al.
[154] Doppler K, Appeltshauser L, Villmann C, Martin C, Peles E, Kramer HH, et al. Auto- ADAMTS13 autoantibodies cloned from patients with acquired thrombotic
antibodies to contactin-associated protein 1 (Caspr) in two patients with painful thrombocytopenic purpura: 2. Pathogenicity in an animal model. Transfusion
inflammatory neuropathy. Brain 2016;139:2617–30. 2016;56:1775–85.
[155] Querol L, Nogales-Gadea G, Rojas-Garcia R, Martinez-Hernandez E, Diaz-Manera [183] Stanley JR, Amagai M. Pemphigus, bullous impetigo, and the staphylococcal
J, Suarez-Calvet X, et al. Antibodies to contactin-1 in chronic inflammatory de- scalded-skin syndrome. N Engl J Med 2006;355:1800–10.
myelinating polyneuropathy. Ann Neurol 2012;3:370–80. [184] Amagai M. Desmoglein as a target in autoimmunity and infection. J Am Acad
[156] Miura Y, Devaux JJ, Fukami Y, Manso C, Belghazi M, Wong AH, et al. Contactin 1 Dermatol 2003;48:244–52.
IgG4 associates to chronic inflammatory demyelinating polyneuropathy with [185] Di Zenzo G, Di Lullo G, Corti D, Calabresi V, Sinistro A, Vanzetta F, et al.
sensory ataxia. Brain 2015;138:1484–91. Pemphigus autoantibodies generated through somatic mutations target the des-
[157] Doppler K, Appeltshauser L, Wilhelmi K, Villmann C, Dib-Hajj SD, Waxman SG, moglein-3 cis-interface. J Clin Invest 2012;122:3781–90.
et al. Destruction of paranodal architecture in inflammatory neuropathy with anti- [186] Li N, Aoki V, Hans-Filho G, Rivitti EA, Diaz LA. The role of intramolecular epitope
contactin-1 autoantibodies. J Neurol Neurosurg Psychiatry 2015;86:720–8. spreading in the pathogenesis of endemic pemphigus foliaceus (fogo selvagem). J
[158] Labasque M, Hivert B, Nogales-Gadea G, Querol L, Illa I, Faivre-Sarrailh C. Specific Exp Med 2003;197:1501–10.
contactin N-glycans are implicated in neurofascin binding and autoimmune tar- [187] Futei Y, Amagai M, Ishii K, Kuroda-Kinoshita K, Ohya K, Nishikawa T.
geting in peripheral neuropathies. J Biol Chem 2014;289:7907–18. Predominant IgG4 subclass in autoantibodies of pemphigus vulgaris and foliaceus.
[159] Sistani L, Rodriguez PQ, Hultenby K, Uhlen M, Betsholtz C, Jalanko H, et al. J Dermatol Sci 2001;26:55–61.
Neuronal proteins are novel components of podocyte major processes and their [188] Rock B, Martins CR, Theofilopoulos AN, Balderas RS, Anhalt GJ, Labib RS, et al.
expression in glomerular crescents supports their role in crescent formation. The pathogenic effect of IgG4 autoantibodies in endemic pemphigus foliaceus
Kidney Int 2013;83:63–71. (fogo selvagem). N Engl J Med 1989;320:1463–9.
[160] Reid RA, Bronson DD, Young KM, Hemperly JJ. Identification and characterization [189] Payne AS, Ishii K, Kacir S, Lin C, Li H, Hanakawa Y, et al. Genetic and functional
of the human cell adhesion molecule contactin. Brain Res Mol Brain Res characterization of human pemphigus vulgaris monoclonal autoantibodies iso-
1994;21:1–8. lated by phage display. J Clin Invest 2005;115:888–99.
[161] Taieb G, Le Quintrec M, Pialot A, Szwarc I, Perrochia H, Labauge P, et al. “neuro- [190] Ishii K, Harada R, Matsuo I, Shirakata Y, Hashimoto K, Amagai M. In vitro kera-
renal syndrome” related to anti-contactin-1 antibodies. Muscle Nerve tinocyte dissociation assay for evaluation of the pathogenicity of anti-desmoglein
2019;59:E19–21. 3 IgG autoantibodies in pemphigus vulgaris. J Invest Dermatol 2005;124:939–46.
[162] Doppler K, Schuster Y, Appeltshauser L, Biko L, Villmann C, Weishaupt A, et al. [191] Saito M, Stahley SN, Caughman CY, Mao X, Tucker DK, Payne AS, et al. Signaling
Anti-CNTN1 IgG3 induces acute conduction block and motor deficits in a passive dependent and independent mechanisms in pemphigus vulgaris blister formation.
transfer rat model. J Neuroinflammation 2019;16:73. PLoS One 2012;7:e50696.
[163] Kanigicherla D, Gummadova J, McKenzie EA, Roberts SA, Harris S, Nikam M, et al. [192] Evangelista F, Roth AJ, Prisayanh P, Temple BR, Li N, Qian Y, et al. Pathogenic
Anti-PLA2R antibodies measured by ELISA predict long-term outcome in a pre- IgG4 autoantibodies from endemic pemphigus foliaceus recognize a desmoglein-1
valent population of patients with idiopathic membranous nephropathy. Kidney conformational epitope. J Autoimmun 2018;89:171–85.
Int 2013;83:940–8. [193] Waschke J, Spindler V, Bruggeman P, Zillikens D, Schmidt G, Drenckhahn D.
[164] Manso C, Querol L, Mekaouche M, Illa I, Devaux JJ. Contactin-1 IgG4 antibodies Inhibition of rho a activity causes pemphigus skin blistering. J Cell Biol
cause paranode dismantling and conduction defects. Brain 2016;139:1700–12. 2006;175:721–7.
[165] Bhat MA, Rios JC, Lu Y, Garcia-Fresco GP, Ching W, Martin M St, et al. Axon-glia [194] Li N, Zhao M, Wang J, Liu Z, Diaz LA. Involvement of the apoptotic mechanism in
interactions and the domain organization of myelinated axons requires neurexin pemphigus foliaceus autoimmune injury of the skin. J Immunol 2009;182:711–7.
IV/Caspr/Paranodin. Neuron 2001;30:369–83. [195] Cipolla GA, Park JK, Lavker RM, Petzl-Erler ML. Crosstalk between signaling
[166] Boyle ME, Berglund EO, Murai KK, Weber L, Peles E, Ranscht B. Contactin or- pathways in pemphigus: a role for endoplasmic reticulum stress in p38 mitogen-
chestrates assembly of the septate-like junctions at the paranode in myelinated activated protein kinase activation? Front Immunol 2017;8:1022.
peripheral nerve. Neuron 2001;30:385–97. [196] Egu DT, Walter E, Spindler V, Waschke J. Inhibition of p38MAPK signaling pre-
[167] Sherman DL, Tait S, Melrose S, Johnson R, Zonta B, Court FA, et al. Neurofascins vents epidermal blistering and alterations of desmosome structure induced by
are required to establish axonal domains for saltatory conduction. Neuron pemphigus autoantibodies in human epidermis. Br J Dermatol 2017;6:1612–8.
2005;48:737–42. [197] Mavropoulos A, Orfanidou T, Liaskos C, Smyk DS, Spyrou V, Sakkas LI, et al. p38
[168] Ng JK, Malotka J, Kawakami N, Derfuss T, Khademi M, Olsson T, et al. Neurofascin MAPK signaling in pemphigus: implications for skin autoimmunity. Autoimmune
as a target for autoantibodies in peripheral neuropathies. Neurology Dis 2013;2013:728529.
2012;79:2241–8. [198] Spindler V, Vielmuth F, Schmidt E, Rubenstein DS, Waschke J. Protective en-
[169] Devaux JJ, Miura Y, Fukami Y, Inoue T, Manso C, Belghazi M, et al. Neurofascin- dogenous cyclic adenosine 5′-monophosphate signaling triggered by pemphigus
155 IgG4 in chronic inflammatory demyelinating polyneuropathy. Neurology autoantibodies. J Immunol 2010;185:6831–8.
2016;86:800–7. [199] Walter E, Vielmuth F, Rotkopf L, Sardy M, Horvath ON, Goebeler M, et al.
[170] Kadoya M, Kaida K, Koike H, Takazaki H, Ogata H, Moriguchi K, et al. IgG4 anti- Different signaling patterns contribute to loss of keratinocyte cohesion dependent
neurofascin155 antibodies in chronic inflammatory demyelinating poly- on autoantibody profile in pemphigus. Sci Rep 2017;7:3579.
radiculoneuropathy: clinical significance and diagnostic utility of a conventional [200] Funakoshi T, Lunardon L, Ellebrecht CT, Nagler AR, O’Leary CE, Payne AS.
assay. J Neuroimmunol 2016;301:16–22. Enrichment of total serum IgG4 in patients with pemphigus. Br J Dermatol
[171] Painous C, Lopez-Perez MA, Illa I, Querol L. Head and voice tremor improving 2012;167:1245–53.
with immunotherapy in an anti-NF155 positive CIDP patient. Ann Clin Transl [201] Zhao CY, Chiang YZ, Murrell DF. Neonatal autoimmune blistering disease: a sys-
Neurol 2018;5:499–501. tematic review. Pediatr Dermatol 2016;33:367–74.
[172] Querol L, Rojas-Garcia R, Diaz-Manera J, Barcena J, Pardo J, Ortega-Moreno A, [202] Tsunoda K, Ota T, Aoki M, Yamada T, Nagai T, Nakagawa T, et al. Induction of
et al. Rituximab in treatment-resistant CIDP with antibodies against paranodal pemphigus phenotype by a mouse monoclonal antibody against the amino-term-
proteins. Neurol Neuroimmunol Neuroinflamm 2015;2:e149. inal adhesive interface of desmoglein 3. J Immunol 2003;170:2170–8.
[173] Bailly L, Mongin M, Delorme C, Apartis E, Saheb S, Viala K, et al. Tremor asso- [203] Irani SR, Michell AW, Lang B, Pettingill P, Waters P, Johnson MR, et al.
ciated with chronic inflammatory demyelinating polyneuropathy and anti- Faciobrachial dystonic seizures precede Lgi1 antibody limbic encephalitis. Ann

17
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Neurol 2011;69:892–900. 2019;29:586–97.


[204] Irani SR, Alexander S, Waters P, Kleopa KA, Pettingill P, Zuliani L, et al. Antibodies [232] Dawes JM, Weir GA, Middleton SJ, Patel R, Chisholm KI, Pettingill P, et al.
to Kv1 potassium channel-complex proteins leucine-rich, glioma inactivated 1 Immune or Genetic-Mediated Disruption of CASPR2 Causes Pain Hypersensitivity
protein and contactin-associated protein-2 in limbic encephalitis, Morvan’s syn- Due to Enhanced Primary Afferent Excitability. Neuron 2018;97:806–22. (e10).
drome and acquired neuromyotonia. Brain 2010;133:2734–48. [233] Patterson KR, Dalmau J, Lancaster E. Mechanisms of Caspr2 antibodies in auto-
[205] Arino H, Armangue T, Petit-Pedrol M, Sabater L, Martinez-Hernandez E, Hara M, immune encephalitis and neuromyotonia. Ann Neurol 2018;83:40–51.
et al. Anti-LGI1-associated cognitive impairment: presentation and long-term [234] Pinatel D, Hivert B, Boucraut J, Saint-Martin M, Rogemond V, Zoupi L, et al.
outcome. Neurology 2016;87:759–65. Inhibitory axons are targeted in hippocampal cell culture by anti-Caspr2 auto-
[206] van Sonderen A, Thijs RD, Coenders EC, Jiskoot LC, Sanchez E, de Bruijn MA, et al. antibodies associated with limbic encephalitis. Front Cell Neurosci 2015;9:265.
Anti-LGI1 encephalitis: clinical syndrome and long-term follow-up. Neurology [235] Saint-Martin M, Pieters A, Dechelotte B, Malleval C, Pinatel D, Pascual O, et al.
2016;87:1449–56. Impact of anti-CASPR2 autoantibodies from patients with autoimmune en-
[207] Bastiaansen AEM, van Sonderen A, Titulaer MJ. Autoimmune encephalitis with cephalitis on CASPR2/TAG-1 interaction and Kv1 expression. J Autoimmun
anti-leucine-rich glioma-inactivated 1 or anti-contactin-associated protein-like 2 2019;103:102284.
antibodies (formerly called voltage-gated potassium channel-complex antibodies). [236] Fernandes D, Santos SD, Coutinho E, Whitt JL, Beltrao N, Rondao T, et al.
Curr Opin Neurol 2017;30:302–9. Disrupted AMPA receptor function upon genetic- or antibody-mediated loss of
[208] Fukata Y, Adesnik H, Iwanaga T, Bredt DS, Nicoll RA, Fukata M. Epilepsy-related autism-associated CASPR2. Cereb Cortex 2019;12:4919–31.
ligand/receptor complex LGI1 and ADAM22 regulate synaptic transmission. [237] Giannoccaro MP, Menassa DA, Jacobson L, Coutinho E, Prota G, Lang B, et al.
Science 2006;313:1792–5. Behaviour and neuropathology in mice injected with human contactin-associated
[209] Yamagata A, Miyazaki Y, Yokoi N, Shigematsu H, Sato Y, Goto-Ito S, et al. protein 2 antibodies. Brain 2019;142(7):2000–12.
Structural basis of epilepsy-related ligand-receptor complex LGI1-ADAM22. Nat [238] Becker EB, Zuliani L, Pettingill R, Lang B, Waters P, Dulneva A, et al. Contactin-
Commun 2018;9:1546. associated protein-2 antibodies in non-paraneoplastic cerebellar ataxia. J Neurol
[210] Lovero KL, Fukata Y, Granger AJ, Fukata M, Nicoll RA. The LGI1-ADAM22 protein Neurosurg Psychiatry 2012;83:437–40.
complex directs synapse maturation through regulation of PSD-95 function. Proc [239] Davies BS, Beigneux AP, Barnes 2nd RH, Tu Y, Gin P, Weinstein MM, et al.
Natl Acad Sci U S A 2015;112:E4129–37. GPIHBP1 is responsible for the entry of lipoprotein lipase into capillaries. Cell
[211] Miller TD, Chong TT, Aimola Davies AM, Ng TWC, Johnson MR, Irani SR, et al. Metab 2010;12:42–52.
Focal CA3 hippocampal subfield atrophy following LGI1 VGKC-complex antibody [240] Mysling S, Kristensen KK, Larsson M, Beigneux AP, Gardsvoll H, Fong LG, et al.
limbic encephalitis. Brain 2017;140:1212–9. The acidic domain of the endothelial membrane protein GPIHBP1 stabilizes li-
[212] Aysit-Altuncu N, Ulusoy C, Ozturk G, Tuzun E. Effect of LGI1 antibody-positive IgG poprotein lipase activity by preventing unfolding of its catalytic domain. eLife
on hippocampal neuron survival: a preliminary study. Neuroreport 2016;5:e12095.
2018;29:932–8. [241] Mysling S, Kristensen KK, Larsson M, Kovrov O, Bensadouen A, Jorgensen TJ, et al.
[213] Ohkawa T, Fukata Y, Yamasaki M, Miyazaki T, Yokoi N, Takashima H, et al. The angiopoietin-like protein ANGPTL4 catalyzes unfolding of the hydrolase do-
Autoantibodies to epilepsy-related LGI1 in limbic encephalitis neutralize LGI1- main in lipoprotein lipase and the endothelial membrane protein GPIHBP1
ADAM22 interaction and reduce synaptic AMPA receptors. J Neurosci counteracts this unfolding. eLife 2016;5.
2013;33:18161–74. [242] Eguchi J, Miyashita K, Fukamachi I, Nakajima K, Murakami M, Kawahara Y, et al.
[214] Thompson J, Bi M, Murchison AG, Makuch M, Bien CG, Chu K, et al. The im- GPIHBP1 autoantibody syndrome during interferon beta1a treatment. J Clin
portance of early immunotherapy in patients with faciobrachial dystonic seizures. Lipidol 2019;13:62–9.
Brain 2018;141:348–56. [243] Keri KC, Blumenthal S, Kulkarni V, Beck L, Chongkrairatanakul T. Primary
[215] Seagar M, Russier M, Caillard O, Maulet Y, Fronzaroli-Molinieres L, De San membranous nephropathy: comprehensive review and historical perspective.
Feliciano M, et al. LGI1 tunes intrinsic excitability by regulating the density of Postgrad Med J 2019;95:23–31.
axonal Kv1 channels. Proc Natl Acad Sci U S A 2017;114:7719–24. [244] Cattran DC, Brenchley PE. Membranous nephropathy: integrating basic science
[216] Lancaster E, Burnor E, Zhang J, Lancaster E. ADAM23 is a negative regulator of into improved clinical management. Kidney Int 2017;91:566–74.
Kv1.1/Kv1.4 potassium currents. Neurosci Lett 2019;704:159–63. [245] Doi T, Mayumi M, Kanatsu K, Suehiro F, Hamashima Y. Distribution of IgG sub-
[217] Petit-Pedrol M, Sell J, Planaguma J, Mannara F, Radosevic M, Haselmann H, et al. classes in membranous nephropathy. Clin Exp Immunol 1984;58:57–62.
LGI1 antibodies alter Kv1.1 and AMPA receptors changing synaptic excitability, [246] De Vriese AS, Glassock RJ, Nath KA, Sethi S, Fervenza FC. A proposal for a ser-
plasticity and memory. Brain 2018;141:3144–59. ology-based approach to membranous nephropathy. J Am Soc Nephrol
[218] Lalic T, Pettingill P, Vincent A, Capogna M. Human limbic encephalitis serum 2017;28:421–30.
enhances hippocampal mossy fiber-CA3 pyramidal cell synaptic transmission. [247] Beck Jr. LH, Bonegio RG, Lambeau G, Beck DM, Powell DW, Cummins TD, et al. M-
Epilepsia 2011;52:121–31. type phospholipase A2 receptor as target antigen in idiopathic membranous ne-
[219] Yu YE, Wen L, Silva J, Li Z, Head K, Sossey-Alaoui K, et al. Lgi1 null mutant mice phropathy. N Engl J Med 2009;361:11–21.
exhibit myoclonic seizures and CA1 neuronal hyperexcitability. Hum Mol Genet [248] Cheng G, Liu J, Gilbert A, Cao Y, An C, Lv Z, et al. Serum phospholipase A2 re-
2010;19:1702–11. ceptor antibodies and immunoglobulin G subtypes in adult idiopathic membra-
[220] Lancaster E, Huijbers MG, Bar V, Boronat A, Wong A, Martinez-Hernandez E, et al. nous nephropathy: clinical value assessment. Clin Chim Acta 2019;490:135–41.
Investigations of caspr2, an autoantigen of encephalitis and neuromyotonia. Ann [249] Murtas C, Bruschi M, Candiano G, Moroni G, Magistroni R, Magnano A, et al.
Neurol 2011;69:303–11. Coexistence of different circulating anti-podocyte antibodies in membranous ne-
[221] Vincent A, Irani SR. Caspr2 antibodies in patients with thymomas. J Thorac Oncol phropathy. Clin J Am Soc Nephrol 2012;7:1394–400.
2010;5:S277–80. [250] Zhang Q, Liu X, Zhang Z, Wu M, Huang B, Zhang Y, et al. A comparison of clinical
[222] Vale TC, Pedroso JL, Dutra LA, Azevedo L, Filho LH, Prado LB, et al. Morvan features between idiopathic membranous nephropathy patients with and without
syndrome as a paraneoplastic disorder of thymoma with anti-CASPR2 antibodies. serum antibody against phospholipase A2 receptor. Medicine (Baltimore)
Lancet 2017;389:1367–8. 2019;98:e17658.
[223] van Sonderen A, Petit-Pedrol M, Dalmau J, Titulaer MJ. The value of LGI1, Caspr2 [251] Hofstra JM, Debiec H, Short CD, Pelle T, Kleta R, Mathieson PW, et al.
and voltage-gated potassium channel antibodies in encephalitis. Nat Rev Neurol Antiphospholipase A2 receptor antibody titer and subclass in idiopathic mem-
2017;13:290–301. branous nephropathy. J Am Soc Nephrol 2012;23:1735–43.
[224] van Sonderen A, Arino H, Petit-Pedrol M, Leypoldt F, Kortvelyessy P, Wandinger [252] Garcia-Vives E, Sole C, Moline T, Alvarez-Rios AM, Vidal M, Agraz I, et al.
KP, et al. The clinical spectrum of Caspr2 antibody-associated disease. Neurology Antibodies to M-type phospholipase A2 receptor (PLA2R) in membranous lupus
2016;87:521–8. nephritis. Lupus 2019;28:396–405.
[225] Joubert B, Saint-Martin M, Noraz N, Picard G, Rogemond V, Ducray F, et al. [253] Al-Rabadi L, Quillen K, Shashar M, Al Marji C, Jaberi A, Chitalia V, et al.
Characterization of a subtype of autoimmune encephalitis with anti-Contactin- Concurrent presentation of thrombotic thrombocytopenic Purpura and membra-
associated protein-like 2 antibodies in the cerebrospinal fluid, prominent limbic nous nephropathy. Kidney Int Rep 2018;3:476–81.
symptoms, and seizures. JAMA Neurol. 2016;73:1115–24. [254] Huang CC, Lehman A, Albawardi A, Satoskar A, Brodsky S, Nadasdy G, et al. IgG
[226] Sunwoo JS, Lee ST, Byun JI, Moon J, Shin JW, Jeong DE, et al. Clinical mani- subclass staining in renal biopsies with membranous glomerulonephritis indicates
festations of patients with CASPR2 antibodies. J Neuroimmunol 2015;281:17–22. subclass switch during disease progression. Mod Pathol 2013;26:799–805.
[227] Klein CJ, Lennon VA, Aston PA, McKeon A, O’Toole O, Quek A, et al. Insights from [255] Salant DJ. Unmet challenges in membranous nephropathy. Curr Opin Nephrol
LGI1 and CASPR2 potassium channel complex autoantibody subtyping. JAMA Hypertens 2019;28:70–6.
Neurol 2013;70:229–34. [256] Lambeau G, Lazdunski M. Receptors for a growing family of secreted phospholi-
[228] Bien CG, Mirzadjanova Z, Baumgartner C, Onugoren MD, Grunwald T, Holtkamp pases A2. Trends Pharmacol Sci 1999;20:162–70.
M, et al. Anti-contactin-associated protein-2 encephalitis: relevance of antibody [257] Sukocheva O, Menschikowski M, Hagelgans A, Yarla NS, Siegert G, Reddanna P,
titres, presentation and outcome. Eur J Neurol 2017;24:175–86. et al. Current insights into functions of phospholipase A2 receptor in normal and
[229] Somers KJ, Lennon VA, Rundell JR, Pittock SJ, Drubach DA, Trenerry MR, et al. cancer cells: more questions than answers. Semin Cancer Biol 2019;56:116–27.
Psychiatric manifestations of voltage-gated potassium-channel complex auto- [258] Fresquet M, Jowitt TA, McKenzie EA, Ball MD, Randles MJ, Lennon R, et al. PLA2R
immunity. J Neuropsychiatry Clin Neurosci 2011;23:425–33. binds to the annexin A2-S100A10 complex in human podocytes. Sci Rep
[230] Poliak S, Gollan L, Martinez R, Custer A, Einheber S, Salzer JL, et al. Caspr2, a new 2017;7:6876.
member of the neurexin superfamily, is localized at the juxtaparanodes of mye- [259] Bharadwaj A, Bydoun M, Holloway R, Waisman D. Annexin A2 heterotetramer:
linated axons and associates with K+ channels. Neuron 1999;24:1037–47. structure and function. Int J Mol Sci 2013;14:6259–305.
[231] Scott R, Sanchez-Aguilera A, van Elst K, Lim L, Dehorter N, Bae SE, et al. Loss of [260] Fresquet M, Jowitt TA, Gummadova J, Collins R, O’Cualain R, McKenzie EA, et al.
Cntnap2 causes axonal excitability deficits, developmental delay in cortical mye- Identification of a major epitope recognized by PLA2R autoantibodies in primary
lination, and abnormal stereotyped motor behavior. Cerebral Cortex membranous nephropathy. J Am Soc Nephrol 2015;26:302–13.

18
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

[261] Kao L, Lam V, Waldman M, Glassock RJ, Zhu Q. Identification of the im- lectin complement pathway. Biosci Rep 2018;38.
munodominant epitope region in phospholipase A2 receptor-mediating autoanti- [288] Goletz S, Probst C, Komorowski L, Schlumberger W, Fechner K, van Beek N, et al.
body binding in idiopathic membranous nephropathy. J Am Soc Nephrol A sensitive and specific assay for the serological diagnosis of antilaminin 332
2015;26:291–301. mucous membrane pemphigoid. Br J Dermatol 2019;180:149–56.
[262] Skoberne A, Behnert A, Teng B, Fritzler MJ, Schiffer L, Pajek J, et al. Serum with [289] Verrando P, Blanchet-Bardon C, Pisani A, Thomas L, Cambazard F, Eady RA, et al.
phospholipase A2 receptor autoantibodies interferes with podocyte adhesion to Monoclonal antibody GB3 defines a widespread defect of several basement
collagen. Eur J Clin Invest 2014;44:753–65. membranes and a keratinocyte dysfunction in patients with lethal junctional
[263] Jurgensen HJ, Johansson K, Madsen DH, Porse A, Melander MC, Sorensen KR, epidermolysis bullosa. Lab Invest 1991;64:85–92.
et al. Complex determinants in specific members of the mannose receptor family [290] Giurdanella F, Nijenhuis AM, Diercks GFH, Jonkman MF, Pas HH. Keratinocyte
govern collagen endocytosis. J Biol Chem 2014;289:7935–47. footprint assay discriminates antilaminin-332 pemphigoid from all other forms of
[264] Beck Jr. LH. PLA2R and THSD7A: disparate paths to the same disease? J Am Soc pemphigoid diseases. Br J Dermatol 2019;2:373–81.
Nephrol 2017;28:2579–89. [291] Chan LS, Majmudar AA, Tran HH, Meier F, Schaumburg-Lever G, Chen M, et al.
[265] Chi JN, Lai TS, Wu CF, Fu TY, Chou YH, Chiu YL, et al. The relationship of anti- Laminin-6 and laminin-5 are recognized by autoantibodies in a subset of cicatricial
phospholipase A2 receptor antibody and C5a complement with disease activity pemphigoid. J Invest Dermatol 1997;108:848–53.
and short-term outcome in idiopathic membranous nephropathy. J Formos Med [292] Domloge-Hultsch N, Gammon WR, Briggaman RA, Gil SG, Carter WG, Yancey KB.
Assoc 2019;118:898–906. Epiligrin, the major human keratinocyte integrin ligand, is a target in both an
[266] Zhang MF, Cui Z, Zhang YM, Qu Z, Wang X, Wang F, et al. Clinical and prognostic acquired autoimmune and an inherited subepidermal blistering skin disease. J Clin
significance of glomerular C1q deposits in primary MN. Clin Chim Acta Invest 1992;90:1628–33.
2018;485:152–7. [293] Lazarova Z, Salato VK, Lanschuetzer CM, Janson M, Fairley JA, Yancey KB. IgG
[267] Hayashi N, Akiyama S, Okuyama H, Matsui Y, Adachi H, Yamaya H, et al. anti-laminin-332 autoantibodies are present in a subset of patients with mucous
Clinicopathological characteristics of M-type phospholipase A2 receptor (PLA2R)- membrane, but not bullous, pemphigoid. J Am Acad Dermatol 2008;58:951–8.
related membranous nephropathy in Japanese. Clin Exp Nephrol [294] Egan CA, Lazarova Z, Darling TN, Yee C, Cote T, Yancey KB. Anti-epiligrin cica-
2015;19:797–803. tricial pemphigoid and relative risk for cancer. Lancet 2001;357:1850–1.
[268] Debiec H, Hanoy M, Francois A, Guerrot D, Ferlicot S, Johanet C, et al. Recurrent [295] Matsushima S, Horiguchi Y, Honda T, Fujii S, Okano T, Tanabe M, et al. A case of
membranous nephropathy in an allograft caused by IgG3kappa targeting the PLA2 anti-epiligrin cicatricial pemphigoid associated with lung carcinoma and severe
receptor. J Am Soc Nephrol 2012;23:1949–54. laryngeal stenosis: review of Japanese cases and evaluation of risk for internal
[269] Haddad G, Kistler AD. An in vitro model of idiopathic membranous nephropathy malignancy. J Dermatol 2004;31:10–5.
reveals PLA2R- and complement-dependent pathways of podocyte injury. J Am [296] Leverkus M, Schmidt E, Lazarova Z, Brocker EB, Yancey KB, Zillikens D.
Soc Nephrol 2017;20:109A. Antiepiligrin cicatricial pemphigoid: an underdiagnosed entity within the spec-
[270] Hayashi N, Okada K, Matsui Y, Fujimoto K, Adachi H, Yamaya H, et al. Glomerular trum of scarring autoimmune subepidermal bullous diseases? Arch Dermatol
mannose-binding lectin deposition in intrinsic antigen-related membranous ne- 1999;135:1091–8.
phropathy. Nephrol Dial Transplant 2018;33:832–40. [297] Terra JB, Pas HH, Hertl M, Dikkers FG, Kamminga N, Jonkman MF.
[271] Bally S, Debiec H, Ponard D, Dijoud F, Rendu J, Faure J, et al. Phospholipase A2 Immunofluorescence serration pattern analysis as a diagnostic criterion in antil-
receptor-related membranous nephropathy and Mannan-binding lectin deficiency. aminin-332 mucous membrane pemphigoid: immunopathological findings and
J Am Soc Nephrol 2016;27:3539–44. clinical experience in 10 Dutch patients. Br J Dermatol 2011;165:815–22.
[272] Michalski M, Swierzko AS, Pagowska-Klimek I, Niemir ZI, Mazerant K, Domzalska- [298] La Placa M, Balestri R, Tartari F, Sechi A, Ferrara F, Loi C, et al. Mucous membrane
Popadiuk I, et al. Primary Ficolin-3 deficiency–is it associated with increased pemphigoid-associated malignancies: case series and a brief overview of the lit-
susceptibility to infections? Immunobiology 2015;220:711–3. erature. Dermatol Pract Concept 2019;9:119–25.
[273] Dekkers G, Rispens T, Vidarsson G. Novel concepts of altered immunoglobulin G [299] Carter WG, Ryan MC, Gahr PJ. Epiligrin, a new cell adhesion ligand for integrin
Galactosylation in autoimmune diseases. Front Immunol 2018;9:553. alpha 3 beta 1 in epithelial basement membranes. Cell 1991;65:599–610.
[274] Foster MH. Optimizing the translational value of animal models of glomerulone- [300] Rousselle P, Lunstrum GP, Keene DR, Burgeson RE. Kalinin: an epithelium-specific
phritis: insights from recent murine prototypes. Am J Physiol Renal Physiol basement membrane adhesion molecule that is a component of anchoring fila-
2016;311:F487–95. ments. J Cell Biol 1991;114:567–76.
[275] Luo W, Olaru F, Miner JH, Beck Jr. LH, van der Vlag J, Thurman JM, et al. [301] Koga H, Teye K, Ishii N, Nakama T. Detachment of keratinocytes at the basement
Alternative pathway is essential for glomerular complement activation and pro- membrane zone caused by inhibitory effect of the antibodies from sera of mucous
teinuria in a mouse model of membranous nephropathy. Front Immunol membrane pemphigoid patients. J Dermatol 2019;46:1046–8.
2018;9:1433. [302] Lazarova Z, Hsu R, Yee C, Yancey KB. Human anti-laminin 5 autoantibodies in-
[276] Hoxha E, Beck Jr. LH, Wiech T, Tomas NM, Probst C, Mindorf S, et al. An indirect duce subepidermal blisters in an experimental human skin graft model. J Invest
immunofluorescence method facilitates detection of thrombospondin type 1 do- Dermatol 2000;114:178–84.
main-containing 7A-specific antibodies in membranous nephropathy. J Am Soc [303] Delmont E, Manso C, Querol L, Cortese A, Berardinelli A, Lozza A, et al.
Nephrol 2017;28:520–31. Autoantibodies to nodal isoforms of neurofascin in chronic inflammatory de-
[277] Stoddard SV, Welsh CL, Palopoli MM, Stoddard SD, Aramandla MP, Patel RM, myelinating polyneuropathy. Brain 2017;140:1851–8.
et al. Structure and function insights garnered from in silico modeling of the [304] Lin HP, Ho KWD, Chuquilin M. Presence of both anti-contactin 1 and anti-neu-
thrombospondin type-1 domain-containing 7A antigen. Proteins 2019;87:136–45. rofascin 140 antibodies in a case of chronic inflammatory demyelinating poly-
[278] Seifert L, Hoxha E, Eichhoff AM, Zahner G, Dehde S, Reinhard L, et al. The most N- neuropathy. eNeurologicalSci 2018;13:38–9.
terminal region of THSD7A is the predominant target for autoimmunity in [305] Segelmark M, Hellmark T. Anti-glomerular basement membrane disease: An up-
THSD7A-associated membranous nephropathy. J Am Soc Nephrol date on subgroups, pathogenesis and therapies, Nephrol dial transplant. 2018.
2018;29:1536–48. [306] Thibaud V, Rioux-Leclercq N, Vigneau C, Morice S. Recurrence of Goodpasture
[279] Herwig J, Skuza S, Sachs W, Sachs M, Failla AV, Rune G, et al. Thrombospondin syndrome without circulating anti-glomerular basement membrane antibodies
type 1 domain-containing 7A localizes to the slit diaphragm and stabilizes mem- after kidney transplant, a case report. BMC Nephrol 2019;20:6.
brane dynamics of fully differentiated podocytes. J Am Soc Nephrol [307] Lerner RA, Glassock RJ, Dixon FJ. The role of anti-glomerular basement mem-
2019;30:824–39. brane antibody in the pathogenesis of human glomerulonephritis. J Exp Med
[280] Iwakura T, Ohashi N, Kato A, Baba S, Yasuda H. Prevalence of enhanced granular 1967;126:989–1004.
expression of thrombospondin Type-1 domain-containing 7A in the glomeruli of [308] Ohlsson S, Herlitz H, Lundberg S, Selga D, Molne J, Wieslander J, et al. Circulating
Japanese patients with idiopathic membranous nephropathy. PLoS One anti-glomerular basement membrane antibodies with predominance of subclass
2015;10:e0138841. IgG4 and false-negative immunoassay test results in anti-glomerular basement
[281] Tomas NM, Beck Jr. LH, Meyer-Schwesinger C, Seitz-Polski B, Ma H, Zahner G, membrane disease. Am J Kidney Dis 2014;63:289–93.
et al. Thrombospondin type-1 domain-containing 7A in idiopathic membranous [309] Sethi S, Lewin M, Lopez L, Lager D. Linear anti-glomerular basement membrane
nephropathy. N Engl J Med 2014;371:2277–87. IgG but no glomerular disease: Goodpasture’s syndrome restricted to the lung.
[282] Kronbichler A, Oh J, Meijers B, Mayer G, Shin JI. Recent Progress in deciphering Nephrol Dial Transplant 2007;22:1233–5.
the Etiopathogenesis of primary membranous nephropathy. Biomed Res Int [310] Cui Z, Zhao MH, Singh AK, Wang HY. Antiglomerular basement membrane disease
2017;2017:1936372. with normal renal function. Kidney Int 2007;72:1403–8.
[283] Wang J, Cui Z, Lu J, Probst C, Zhang YM, Wang X, et al. Circulating antibodies [311] Qu Z, Cui Z, Liu G, Zhao MH. The distribution of IgG subclass deposition on renal
against thrombospondin type-I domain-containing 7A in Chinese patients with tissues from patients with anti-glomerular basement membrane disease. BMC
idiopathic membranous nephropathy. Clin J Am Soc Nephrol 2017;12:1642–51. Immunol 2013;14:19.
[284] Hoxha E, Wiech T, Stahl PR, Zahner G, Tomas NM, Meyer-Schwesinger C, et al. A [312] Bruschi M, Carnevali ML, Murtas C, Candiano G, Petretto A, Prunotto M, et al.
mechanism for Cancer-associated membranous nephropathy. N Engl J Med Direct characterization of target podocyte antigens and auto-antibodies in human
2016;374:1995–6. membranous glomerulonephritis: alfa-enolase and borderline antigens. J
[285] Tomas NM, Hoxha E, Reinicke AT, Fester L, Helmchen U, Gerth J, et al. Proteomics 2011;74:2008–17.
Autoantibodies against thrombospondin type 1 domain-containing 7A induce [313] Prunotto M, Carnevali ML, Candiano G, Murtas C, Bruschi M, Corradini E, et al.
membranous nephropathy. J Clin Invest 2016;126:2519–32. Autoimmunity in membranous nephropathy targets aldose reductase and SOD2. J
[286] Tomas NM, Meyer-Schwesinger C, von Spiegel H, Kotb AM, Zahner G, Hoxha E, Am Soc Nephrol 2010;21:507–19.
et al. A heterologous model of thrombospondin type 1 domain-containing 7A-as- [314] Miles LA, Dahlberg CM, Plescia J, Felez J, Kato K, Plow EF. Role of cell-surface
sociated membranous nephropathy. J Am Soc Nephrol 2017;11:3262–77. lysines in plasminogen binding to cells: identification of alpha-enolase as a can-
[287] Wang Z, Wen L, Dou Y, Zhao Z. Human anti-thrombospondin type 1 domain- didate plasminogen receptor. Biochemistry 1991;30:1682–91.
containing 7A antibodies induce membranous nephropathy through activation of [315] Kimura Y, Miura N, Debiec H, Morita H, Yamada H, Banno S, et al. Circulating

19
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

antibodies to alpha-enolase and phospholipase A2 receptor and composition of Case Rep 2017;20:92–4.
glomerular deposits in Japanese patients with primary or secondary membranous [343] Danlos FX, Rossi GM, Blockmans D, Emmi G, Kronbichler A, Durupt S, et al.
nephropathy. Clin Exp Nephrol 2017;21:117–26. Antineutrophil cytoplasmic antibody-associated vasculitides and IgG4-related
[316] Aarli JA. Myasthenia gravis in the elderly: is it different? Ann N Y Acad Sci disease: A new overlap syndrome. Autoimmun Rev 2017;16:1036–43.
2008;1132:238–43. [344] Perez Alamino R, Martinez C, Espinoza LR. IgG4-associated vasculitis. Curr
[317] Zillikens D, Kawahara Y, Ishiko A, Shimizu H, Mayer J, Rank CV, et al. A novel Rheumatol Rep 2013;15:348.
subepidermal blistering disease with autoantibodies to a 200-kDa antigen of the [345] Kawashima H, Utsugi A, Shibamiya A, Iida K, Mimura N, Ohashi H, et al.
basement membrane zone. J Invest Dermatol 1996;106:1333–8. Consideration concerning similarities and differences between ANCA-associated
[318] Chen KR, Shimizu S, Miyakawa S, Ishiko A, Shimizu H, Hashimoto T. Coexistence vasculitis and IgG-4-related diseases: case series and review of literature. Immunol
of psoriasis and an unusual IgG-mediated subepidermal bullous dermatosis: Res 2019;67:99–107.
identification of a novel 200-kDa lower lamina lucida target antigen. Br J [346] Goulabchand R, Delicque J, Gallo M, Le Quellec A, Guilpain P. Comment on the
Dermatol 1996;134:340–6. article entitled “Antineutrophil cytoplasmic antibody-associated vasculitides and
[319] Kridin K, Ahmed AR. Anti-p200 pemphigoid: a systematic review. Front Immunol IgG4-related disease: a new overlap syndrome”. Autoimmun Rev
2019;10:2466. 2017;16:1036–43. [Autoimmun Rev 17 (2018) 431–433].
[320] Patsatsi A, Meltzanidou P, Katafigiotis S, Sotiriadis D, Schmidt E, Lambropoulos A. [347] Yoo J, Ahn SS, Jung SM, Song JJ, Park YB, Lee SW. No overlap between IgG4-
Chronic bullous disease of childhood with IgG reactivity to p200 antigen. Int J related disease and microscopic polyangiitis and granulomatosis with polyangiitis
Dermatol 2017;56:773–5. despite elevated serum IgG4 at diagnosis: a retrospective monocentric study. Clin
[321] Zillikens D, Kawahara Y, Ishiko A, Shimizu H, Mayer J, Rank CV, et al. A novel Rheumatol 2019;38:1147–54.
subepidermal blistering disease with autoantibodies to a 200-kDa antigen of the [348] Al-Soudi A, Doorenspleet ME, Esveldt RE, Burgemeister LT, Hak AE, van den Born
basement membrane zone. J Invest Dermatol 1996;106:465–70. BJH, et al. IgG4:IgG RNA ratio differentiates active disease from remission in
[322] Shimanovich I, Hirako Y, Sitaru C, Hashimoto T, Brocker EB, Butt E, et al. The granulomatosis with polyangiitis: a new disease activity marker? A cross-sectional
autoantigen of anti-p200 pemphigoid is an acidic noncollagenous N-linked gly- and longitudinal study. Arthritis Res Ther 2019;21:43.
coprotein of the cutaneous basement membrane. J Invest Dermatol [349] Tominaga K, Uchida T, Imakiire T, Itoh K, Shimazaki H, Nakanishi K, et al. Anti-
2003;121:1402–8. neutrophil cytoplasmic antibody-associated glomerulonephritis with detection of
[323] Wald A, Schmidt E, Toberer F, Gutschalk A, Rentzsch K, Enk AH, et al. Overlap of myeloperoxidase and phospholipase A2 receptor in membranous nephropathy-
bullous, anti-Laminin-332, and anti-p200 pemphigoid with concomitant anti- lesions: report of two patients with microscopic polyangiitis. BMC Nephrol
Contactin-1-positive inflammatory polyneuropathy treated with intravenous im- 2018;19:120.
munoglobulins as a manifestation of epitope spreading. JAMA Dermatol [350] Zou R, Liu G, Cui Z, Chen M, Zhao MH. Clinical and immunologic characteristics of
2019;155:631–3. patients with ANCA-associated glomerulonephritis combined with membranous
[324] Dainichi T, Koga H, Tsuji T, Ishii N, Ohyama B, Ueda A, et al. From anti-p200 nephropathy: a retrospective cohort study in a single Chinese center. Medicine
pemphigoid to anti-laminin gamma1 pemphigoid. J Dermatol 2010;37:231–8. (Baltimore) 2015;94:e1472.
[325] Dainichi T, Kurono S, Ohyama B, Ishii N, Sanzen N, Hayashi M, et al. Anti-laminin [351] Stump M, Messingham KN, Fairley JA. Concurrent mucous membrane pemphigoid
gamma-1 pemphigoid. Proc Natl Acad Sci U S A 2009;106:2800–5. and membranous glomerulonephritis in a patient with autoantibodies targeting
[326] Groth S, Recke A, Vafia K, Ludwig RJ, Hashimoto T, Zillikens D, et al. the 1080 region of collagen XVII. Br J Dermatol 2019;4:835–6.
Development of a simple enzyme-linked immunosorbent assay for the detection of [352] Zheng M, Ujiie H, Iwata H, Muramatsu K, Yoshimoto N, Ito T, et al. Characteristics
autoantibodies in anti-p200 pemphigoid. Br J Dermatol 2011;164:76–82. of IgG subclasses and complement deposition in BP230-type bullous pemphigoid. J
[327] Monshi B, Groth S, Richter L, Schmidt E, Zillikens D, Rappersberger K. A long-term Eur Acad Dermatol Venereol 2019;33:595–600.
study of a patient with anti-p200 pemphigoid: correlation of autoantibody levels [353] Diaz LA, Ratrie 3rd H, Saunders WS, Futamura S, Squiquera HL, Anhalt GJ, et al.
with disease activity and an example of epitope spreading. Br J Dermatol Isolation of a human epidermal cDNA corresponding to the 180-kD autoantigen
2012;167:1179–83. recognized by bullous pemphigoid and herpes gestationis sera.
[328] Vafia K, Groth S, Beckmann T, Hirose M, Dworschak J, Recke A, et al. Immunolocalization of this protein to the hemidesmosome. J Clin Invest
Pathogenicity of autoantibodies in anti-p200 pemphigoid. PLoS One 1990;86:1088–94.
2012;7:e41769. [354] Stanley JR, Hawley-Nelson P, Yuspa SH, Shevach EM, Katz SI. Characterization of
[329] Koga H, Ishii N, Dainichi T, Tsuruta D, Hamada T, Ohata C, et al. An attempt to bullous pemphigoid antigen: a unique basement membrane protein of stratified
develop mouse model for anti-laminin gamma1 pemphigoid. J Dermatol Sci squamous epithelia. Cell 1981;24:897–903.
2013;70:108–15. [355] Sitaru C, Mihai S, Zillikens D. The relevance of the IgG subclass of autoantibodies
[330] Lamprecht P, Kerstein A, Klapa S, Schinke S, Karsten CM, Yu X, et al. Pathogenetic for blister induction in autoimmune bullous skin diseases. Arch Dermatol Res
and clinical aspects of anti-neutrophil cytoplasmic autoantibody-associated 2007;299:1–8.
Vasculitides. Front Immunol 2018;9:680. [356] Zhou XP, Liu B, Xu Q, Yang Y, He CX, Zuo YG, et al. Serum levels of im-
[331] Holland M, Hewins P, Goodall M, Adu D, Jefferis R, Savage CO. Anti-neutrophil munoglobulins G1 and G4 targeting the non-collagenous 16A domain of BP180
cytoplasm antibody IgG subclasses in Wegener’s granulomatosis: a possible pa- reflect bullous pemphigoid activity and predict bad prognosis. J Dermatol
thogenic role for the IgG4 subclass. Clin Exp Immunol 2004;138:183–92. 2016;43:141–8.
[332] Brouwer E, Tervaert JW, Horst G, Huitema MG, van der Giessen M, Limburg PC, [357] Liu Z, Giudice GJ, Swartz SJ, Fairley JA, Till GO, Troy JL, et al. The role of
et al. Predominance of IgG1 and IgG4 subclasses of anti-neutrophil cytoplasmic complement in experimental bullous pemphigoid. J Clin Invest 1995;95:1539–44.
autoantibodies (ANCA) in patients with Wegener’s granulomatosis and clinically [358] Chen R, Ning G, Zhao ML, Fleming MG, Diaz LA, Werb Z, et al. Mast cells play a
related disorders. Clin Exp Immunol 1991;83:379–86. key role in neutrophil recruitment in experimental bullous pemphigoid. J Clin
[333] Hussain A, Pankhurst T, Goodall M, Colman R, Jefferis R, Savage CO, et al. Invest 2001;108:1151–8.
Chimeric IgG4 PR3-ANCA induces selective inflammatory responses from neu- [359] Liu Z, Shapiro SD, Zhou X, Twining SS, Senior RM, Giudice GJ, et al. A critical role
trophils through engagement of Fcgamma receptors. Immunology for neutrophil elastase in experimental bullous pemphigoid. J Clin Invest
2009;128:236–44. 2000;105:113–23.
[334] Chang SY, Keogh K, Lewis JE, Ryu JH, Yi ES. Increased IgG4-positive plasma cells [360] Hiroyasu S, Ozawa T, Kobayashi H, Ishii M, Aoyama Y, Kitajima Y, et al. Bullous
in granulomatosis with Polyangiitis: a diagnostic pitfall of IgG4-related disease. Int pemphigoid IgG induces BP180 internalization via a macropinocytic pathway. Am
J Rheumatol 2012;2012:121702. J Pathol 2013;182:828–40.
[335] Bando H, Iguchi G, Fukuoka H, Taniguchi M, Kawano S, Saitoh M, et al. A diag- [361] Iwata H, Kamio N, Aoyama Y, Yamamoto Y, Hirako Y, Owaribe K, et al. IgG from
nostic pitfall in IgG4-related hypophysitis: infiltration of IgG4-positive cells in the patients with bullous pemphigoid depletes cultured keratinocytes of the 180-kDa
pituitary of granulomatosis with polyangiitis. Pituitary 2015;18:722–30. bullous pemphigoid antigen (type XVII collagen) and weakens cell attachment. J
[336] Kaushik P, Stone JH, Anderson JT, Dugar S, Mathew R, Nikolic B, et al. Medical Invest Dermatol 2009;129:919–26.
mirroring: granulomatosis with polyangiitis (formerly Wegener's) mimicking im- [362] Ujiie H, Sasaoka T, Izumi K, Nishie W, Shinkuma S, Natsuga K, et al. Bullous
munoglobulin-G4 related disease. Int J Rheum Dis 2018;21:885–9. pemphigoid autoantibodies directly induce blister formation without complement
[337] Della-Torre E, Lanzillotta M, Campochiaro C, Bozzalla E, Bozzolo E, Bandiera A, activation. J Immunol 2014;193:4415–28.
et al. Antineutrophil cytoplasmic antibody positivity in IgG4-related disease: a [363] Zuo Y, Evangelista F, Culton D, Guilabert A, Lin L, Li N, et al. IgG4 autoantibodies
case report and review of the literature. Medicine (Baltimore) 2016;95:e4633. are inhibitory in the autoimmune disease bullous pemphigoid. J Autoimmun
[338] Abbass K, Krug H. Granulomatosis with polyangiitis in a patient with biopsy- 2016;73:111–9.
proven IgG4-related pulmonary disease and coincident small cell lung cancer. BMJ [364] Boronat A, Gelfand JM, Gresa-Arribas N, Jeong HY, Walsh M, Roberts K, et al.
Case Rep 2019;12. Encephalitis and antibodies to dipeptidyl-peptidase-like protein-6, a subunit of
[339] Massey J. IgG4-related hypertrophic pachymeningitis coexpressing antineutrophil Kv4.2 potassium channels. Ann Neurol 2013;73:120–8.
cytoplasmic antibodies. Neurol Neuroimmunol Neuroinflam 2017;4:e341. [365] Hara M, Arino H, Petit-Pedrol M, Sabater L, Titulaer MJ, Martinez-Hernandez E,
[340] Watanabe T, Kanda M, Fukaya S, Ogawa Y, Akikawa K. Rapidly progressive glo- et al. DPPX antibody-associated encephalitis: Main syndrome and antibody effects.
merulonephritis caused by overlap syndrome of IgG4-related tubulointerstitial Neurology 2017;88:1340–8.
nephritis and myeloperoxidase-antineutrophil cytoplasmic antibody-associated [366] Piepgras J, Holtje M, Michel K, Li Q, Otto C, Drenckhahn C, et al. Anti-DPPX
necrotising glomerulonephritis. Clin Exp Rheumatol 2018;36(Suppl. 111):172–3. encephalitis: pathogenic effects of antibodies on gut and brain neurons. Neurology
[341] Su T, Yang L, Cui Z, Wang SX, Zhao MH. Concurrent IgG4-related tubulointerstitial 2015;85:890–7.
nephritis and IgG4 myeloperoxidase-anti-neutrophil cytoplasmic antibody positive [367] Lutt A, Michel K, Kruger D, Volz MS, Nassir M, Schulz E, et al. High prevalence and
crescentic glomerulonephritis: a case report. Medicine (Baltimore) 2017;96:e6707. functional effects of serum antineuronal antibodies in patients with gastro-
[342] Touge H, Tomita K, Yamasaki A, Shimizu E. A case of proteinase 3 anti-neutrophil intestinal disorders. Neurogastroenterol Motil 2018;30:e13292.
cytoplasmic antibody (PR3-ANCA) positive/IgG4-related lung disease. Respir Med [368] Gelpi E, Hoftberger R, Graus F, Ling H, Holton JL, Dawson T, et al.

20
I. Koneczny Autoimmunity Reviews 19 (2020) 102646

Neuropathological criteria of anti-IgLON5-related tauopathy. Acta Neuropathol 2016;166:582–95.


2016;132:531–43. [374] Kisand K, Boe Wolff AS, Podkrajsek KT, Tserel L, Link M, Kisand KV, et al. Chronic
[369] Cagnin A, Mariotto S, Fiorini M, Gaule M, Bonetto N, Tagliapietra M, et al. mucocutaneous candidiasis in APECED or thymoma patients correlates with au-
Microglial and neuronal TDP-43 pathology in anti-IgLON5-related Tauopathy. J toimmunity to Th17-associated cytokines. J Exp Med 2010;207:299–308.
Alzheimer’s Dis 2017;59:13–20. [375] Karner J, Pihlap M, Ranki A, Krohn K, Trebusak Podkrajsek K, Bratanic N, et al. IL-
[370] Sabater L, Gaig C, Gelpi E, Bataller L, Lewerenz J, Torres-Vega E, et al. A novel 6-specific autoantibodies among APECED and thymoma patients. Immun Inflamm
non-rapid-eye movement and rapid-eye-movement parasomnia with sleep Dis 2016;4:235–43.
breathing disorder associated with antibodies to IgLON5: a case series, char- [376] Bichele R, Karner J, Truusalu K, Smidt I, Mandar R, Conti HR, et al. IL-22 neu-
acterisation of the antigen, and post-mortem study. Lancet Neurol tralizing autoantibodies impair fungal clearance in murine oropharyngeal candi-
2014;13:575–86. diasis model. Eur J Immunol 2018;48:464–70.
[371] Schoberl F, Levin J, Remi J, Goldschagg N, Eren O, Okamura N, et al. IgLON5: a [377] Gavanescu I, Benoist C, Mathis D. B cells are required for Aire-deficient mice to
case with predominant cerebellar tau deposits and leptomeningeal inflammation. develop multi-organ autoinflammation: a therapeutic approach for APECED pa-
Neurology 2018;91:180–2. tients. Proc Natl Acad Sci U S A 2008;105:13009–14.
[372] Nagamine K, Peterson P, Scott HS, Kudoh J, Minoshima S, Heino M, et al. [378] Ossart J, Moreau A, Autrusseau E, Menoret S, Martin JC, Besnard M, et al.
Positional cloning of the APECED gene. Nat Genet 1997;17:393–8. Breakdown of immune tolerance in AIRE-deficient rats induces a severe auto-
[373] Meyer S, Woodward M, Hertel C, Vlaicu P, Haque Y, Karner J, et al. AIRE-deficient immune polyendocrinopathy-candidiasis-ectodermal dystrophy-like autoimmune
patients harbor unique high-affinity disease-ameliorating autoantibodies. Cell disease. J Immunol 2018;201:874–87.

21

You might also like