You are on page 1of 14

Clinical Immunology 237 (2022) 108976

Contents lists available at ScienceDirect

Clinical Immunology
journal homepage: www.elsevier.com/locate/yclim

Review Article

Revisiting immunological and clinical aspects of membranous nephropathy


Israel Nieto-Gañán a, 1, *, Ignacio Iturrieta-Zuazo a, b, 1, Claudia Rita a, Ángela Carrasco-Sayalero a
a
Immunology Department, Hospital Universitario Ramón y Cajal, Madrid, Spain
b
Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Spain

A R T I C L E I N F O A B S T R A C T

Keywords: Idiopathic or primary membranous nephropathy (IMN) is one of the most frequent causes of nephrotic syndrome
Glomerulonephritis in adults and the elderly. It is characterized by a thickening of the wall of the glomerular capillaries due to the
Membranous nephropaty presence of immune complex deposits. 85% of membranous nephropathy cases are classified as primary or
Anti-PLA2R
idiopathic (IMN). The rest are of secondary origin (SMN), caused by autoimmune conditions or malignant tumors
Thrombospondin type I domain-containing 7A
as lung cancer, colon and melanomas. It is an organ-specific autoimmune disease in which the complement
Immunopathogenesis
Diagnosis system plays an important role with the formation of the membrane attack complex (MAC; C5b-9), which
Epidemiology produces an alteration of the podocyte structure. The antigen responsible for 70-80% of IMN is a podocyte
Treatment protein called M-type phospholipase A2 receptor (PLA2R). More recently, another podocyte antigen has been
identified, the “Thrombospondin type-1 domain-containing 7A” (THSD7A), which is responsible for 10% of the
cases of negative IMN for anti- PLA2R.

1. Introduction It is an organ-specific autoimmune disease in which the complement


system plays an important pathogenic role after the formation of im­
The term glomerulonephritis (GN) is used to designate diseases that mune complexes, when antibodies bind directly to antigens on the
affect the structure and function of the glomerulus, although the other podocyte membrane. After the formation of the membrane attack
structures of the nephron may later be involved. We speak of primary complex (MAC; C5b-9) there is alteration of the podocyte structure and
GN when the renal involvement is not the consequence of a more general distortion of its slit diaphragms, causing the appearance of massive
disease and the clinical manifestations are restricted to the kidney. proteinuria [1].
Conversely, we talk of secondary GN when is due to a systemic disease. In recent years, it has been possible to identify podocyte antigens
Idiopathic or primary membranous nephropathy (IMN) constitutes responsible for most cases of IMN. The antigen responsible for 70-80% of
one of the most frequent causes of nephrotic syndrome in adults and the IMN is a podocyte protein called M-type phospholipase A2 receptor
elderly, while it is rare in children and adolescents. It is characterized by (PLA2R). The antibodies formed against this protein are mainly of the
a uniform and diffuse thickening of the wall of the glomerular capil­ IgG4 isotype [2]. The mechanisms that trigger these autoimmunity
laries, without associated cell proliferation. This thickening is due to the processes are unknown. Recent studies have shown a genetic predispo­
presence of immune complex deposits along the capillary wall. It usually sition to the disease associated with certain HLA alleles and the genes
progresses slowly. 85% of membranous nephropathy cases are classified that encode PLA2R [3]. More recently, another podocyte antigen has
as primary (IMN), that is, the cause of the disorder is idiopathic or un­ been identified, the “Thrombospondin type-1 domain-containing 7A”
known. The rest are of secondary origin (SMN), caused by autoimmune (THSD7A) against which a similar autoimmune process is triggered and
conditions (systemic lupus erythematosus, infections (syphilis, malaria which is responsible for 10% of the cases of negative IMN for anti-
and hepatitis B), drugs (captopril and NSAIDs), inorganic salts or ma­ PLA2R [4].
lignant tumors as lung cancer, colon and melanomas. The determination of anti-PLA2R has represented a great advance in

Abbreviations: BSA, Bovine serum albumin; GN, Glomerulonephritis; GBM, Glomerular Basement Membrane; ESRD, end-stage renal disease; MAC, Membrane
Attack Complex; MN, Membranous Nephropathy; PLA2R, M-type phospholipase A2 receptor; IMN, Primary Membranous nephropathy; THSD7A, Thrombospondin
type-1 domain-containing 7A.
* Corresponding author.
E-mail address: Israel.ganan@salud.madrid.org (I. Nieto-Gañán).
1
These authors contributed equally.

https://doi.org/10.1016/j.clim.2022.108976
Received 3 September 2021; Received in revised form 16 February 2022; Accepted 5 March 2022
Available online 9 March 2022
1521-6616/© 2022 Elsevier Inc. All rights reserved.
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

the rapid differential diagnosis of MN. Several authors consider that the nephrotic range proteinuria [16] but approximately 60% of untreated
positivity of anti-PLA2R is always diagnostic of IMN, even though other patients will end up developing progressive loss of renal function[17].
conditions coexist in the patient (for example, tumors or infectious The remainder develops non-progressive chronic kidney disease. In
diseases) that could theoretically be responsible for the process. The addition, in 30-40% of cases, it leads to end-stage renal disease (ESRD)
detection of anti-THSD7A requires a deeper search for a tumour process within 5–15 years [8]. Around 10 to 50% of patients have high blood
in MN patients, since several cases of MN anti-THSD7A positive with pressure, which has been linked to the development of chronic kidney
associated tumour have been published [5]. failure [18]. Other known risk factors are age, male sex, and decreased
Next we will address the epidemiology, clinical aspects, immuno­ glomerular filtration rate at diagnosis [7].
pathogenesis, diagnosis and treatment of this disease.
4. Immunopathogenesis
2. Epidemiology
MN is a typical organ-specific autoimmune disease that affects the
MN is the most common cause of idiopathic nephrotic syndrome in kidney glomerulus, being the main target the glomerular cells (podo­
nondiabetic caucasian adults over 40 years worldwide [6]. Although cytes). It involves the formation of subepithelial immunocomplexes
discrepancies are observed between the different series studied, it is deposits that result in complement activation and podocyte damage
accepted that it represents around 30% of the cases, increasing to 40% in [19–21].
the elderly [7]. However, studies in the pediatric population show that it IMN is characterized by light microscopy by thickening of the
is a rare entity, being responsible for 4-5% of cases of infantile nephrotic glomerular basement membrane (GBM) with spike formation. Immu­
syndrome [8]. In addition, MN is one of the leading causes of primary nofluorescence shows fine granular deposits of IgG, complement C3, and
glomerulonephritis, occupying the second or third position according to terminal complement complex C5b-9. Finally, electron microscope re­
the series studied [9]. Furthermore, is the main glomerulopathy in terms veals a subepithelial electron-dense deposits (Fig. 1).
of recurrence after kidney transplantation (about 40% of patients)[10]. The immunopathogenesis of IMN is multifactorial, involving pre­
Globally, the estimated incidence rate is about 1,2/100.000 per year, disposing environmental and genetic factors [22,23]. Neither is clear
with a mean age between 45 and 60, progressively increasing with age why the disease is restricted to the kidney, as PLA2R and other antigens
[6]. Significant differences have been found by ethnicity, being more characteristic of the disease are expressed in multiple organs [24]. It has
common in Caucasians, followed by Asians, and less frequent in Blacks been suggested that differences in glycosilation or other post-
and Hispanics [10]. Some authors have also found differences between translational modifications or the facility of autoantibodies to reach
genders, being more frequent in men than in women (2:1) [9]. the antigen could influence in the injury to specific organs [25,26].
One of the main challenges understands how some patients presents
3. Clinical aspects spontaneous remission while others lead to kidney failure. In this regard
it has been seen that patients with IMN had lower percentage of regu­
IMN is renal limited and his autoimmune nature has been known for latory CD4+ T cells at baseline compared with healthy controls [27].
some years, after the identification of IgG autoantibodies, predomi­ There is less data about patients with spontaneous remissions.
nately IgG4, deposited in a podocyte receptor (phospholipase A2 re­ So knowledge of the immunopathogenesis of IMN and its etiology in
ceptor, PLA2R) in approximately 75% of cases [11]. More recently, a each individual is essential to understand the clinical evolution of the
second podocyte antigen called thrombospondin type 1 domain con­ disease and to evaluate the most appropriate treatment in each case.
taining 7A (THSD7A) was identified in a smaller number of patients with
IMN [4]. In this respect, some authors consider that the term idiopathic 4.1. Main experimental models in MN
MN should be replaced by autoimmune MN [12].
In secondary MN, kidney damage occurs due to the formation of Two models have contributed mainly to our knowledge of the
immune complexes at the glomerular level in the context of systemic pathogenesis of IMN: Heymann nephritis model and Cationic Bovine
diseases or exposure to substances or microorganisms, which are serum albumin (BSA) model.
responsible for the antigens involved in the formation of these immune First experiments on Heymann nephritis [28–30] were performed
complexes. There are numerous entities that are associated and can about 50 years ago in rats. In these studies, it was concluded that the
trigger a secondary MN: systemic diseases (systemic lupus erythemato­ disease was caused by the deposition of subepithelial immunocomplexes
sus, sarcoid, thyroiditis, diabetes and rheumatoid arthritis), tumors consisting of the binding of circulating antibodies to antigens that are
(lung, colon and prostate)., infections (hepatitis B and C and HIV), drugs part of the podocyte membrane. Kerjaschhki and Farquhar (1982, 1983)
(d-penicillamide, gold salts and non-steroidal anti-inflammatories) detected that target protein in rats was megalin, belonging to the low
[10,13]. density lipoprotein receptor superfamily, specifically in a epitope
Clinically, most patients with nephrotic syndrome present: protein­ located in a small glycosilated N-terminal fragment [31].
uria above 3.0g/24h, edema, hypoalbuminemia, hyperlipidemia, The binding of circulating anti-megalin antibodies to surface megalin
normal or slightly altered kidney function and normal blood pressure induces complement activation with generation of the membrane attack
[14], which facilitates its early diagnosis. Non-nephrotic proteinuria is complex (MAC). This leads to the podocyte injury through structural and
detected in approximately 15% of cases and the diagnosis can, unfor­ functional changes as calcium influx, oxidative injury, production of
tunately, be delayed due to the absence of symptoms. For some authors arachidonic acid metabolites, endoplasmic reticulum stress, cell cycle
that implies that the incidence is probably underestimated. Other less dysregulation, disruption of the actin cytoskeleton, and alterations in
frequent signs may include arterial hypertension, lipiduria, hypona­ the ubiquitin-proteasome system. The loss of the cytoskeleton structure
tremia, microhematuria and anemia. [7]. induces loss and displacement of slit diaphragm structures, leading to a
Complications are typically those derived from a nephrotic syn­ profound non-selective proteinuria. The injured podocyte also produces
drome: Thrombophilia, or hypercoagulability disorders (in some cases, new extracellular material assembled between the immune deposits,
the first manifestation of the disease may be a vein thrombosis or even a giving rise to the characteristic “spikes” and GBM thickening [32,33]
pulmonary embolism as a consequence of this [7], increased suscepti­ (Fig. 2).
bility to infections, pulmonary edema, acute kidney failure, Hypothy­ However, although megalin is also expressed in human podocytes
roidism, Hypocalcaemia, Vitamin D deficiency and malnutrition.[15]. [34], deposits of megalin and circulating antibodies against it have not
The clinical course is variable, one-third of patients will have a been described in patients with IMN.
spontaneous remission, particularly, in those patients who never have An alternative model was described in rabbit by Border et al. [24] by

2
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

Fig. 1. Different microscopy images on IMN. A- Glomerulus showing the typical “spikes” of basement membrane projecting from the outer surface of the glomerular
basement membrane (arrows) when stained with silver-methenamine (×40). (Provided by Dr. Charles Alpers, Department of Pathology, University of Washington,
Seattle, WA.). B- Immunofluorescence microscopy in IMN. Finely granular staining for IgG, predominately IgG4, present uniformly in a subepithelial distribution in
all glomeruli (×40). C-Electron microscopy image showing subepithelial electron-dense deposits in a patient with IMN (yellow arrows). Adapted from https://www.
kidneypathology.com/English_version/Membranous_GN.html. D-Glomerular deposition of antipPLA2r antibodies by immunoperoxidase in renal biopsy; 200x.

Fig. 2. Scheme of Heymann nephritis model of MN. (A) The normal podocyte structure is maintained by a actin cytoskeleton that also anchor the foot processes to
the GBM through adhesion complexes and cell-matrix adhesion molecules. The normal filtration slit diaphragm forms a barrier to albumin and is linked to the actin
cytoskeleton via a complex of proteins. (B) In the Heymann nephritis model antibody binding to megalin activates complement leading to formation of the MAC. This
disorganizes the dissolution of the actin cytoskeleton, which displaces the filtration slit diaphragms allowing free passage of albumin into the urine. Also affects cell-
matrix adhesion and cause flattening of the podocyte foot processes. Adapted from Cybulsky et al. (2005).

injection of cationized BSA as exogenous antigen testing the hypothesis 4.2. Endogenous antigen
that only antigen with positive charge influence in the immunocomplex
deposition, since the glomerular membrane is negatively charged. The relevance of these experimental models above described in
human pathology was confirmed by the identification of the endogenous
antigen neutral endopeptidase (NEP) and phospolipase A2 receptor
(PLA2R) serving as target of circulating antibodies in the neonatal and

3
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

adulthood respectively. In addition, a new antigen involved in the dis­ (CysR, or ricin B) domain, a single fibronectin type-2 (FnII) domain,
ease has recently been discovered, THSD7A. Finally, it is probably that and eight to ten C-type lectin-like domains (CTLD). Their cyto­
new target antigens for the disease may be discovered in the future. plasmic domains contain motifs that enable constitutive endocytic
Complement activation by immunocomplex deposits has been consid­ recycling in clathrin-coated pits [50] (Fig. 3A).
ered the main cause of podocyte injury in IMN [33]. However, it cannot All of them recirculate between the plasma membrane and the
be ruled out that the binding of the autoantibodies to their respective endosomes and present at least two pH dependent configurations: an
antigens interrupts the biological function of the latter, also contributing extended configuration (N-terminal cysteine-rich domain pointing
to the development of the disease. outwards from the cell surface) and a bent configuration (N-terminal
domain folds back to interact with C-type lectin-like domains at the
- Neutral endopeptidase antigen (NEP): Was described in a neonate with middle of the structure). Although more studies in molecular
respiratory distress and acute renal failure followed by proteinuria modeling of PLA2R are needed, this seems important because the
and hypertension, being MN the final diagnosis [35]. This rare dis­ target epitope for circulating antibodies might be accessible in only
ease called “maternal-fetal alloimmunization with antenatal MN” is one of these configurations [51]. In fact, this conformational change
due to the presence of maternal antibodies that cross the placenta seems necessary to allow binding at physiologic pH and release in the
and bind to NEP in fetal glomerular podocytes [35,36]. NEP appears more acidic pH of endosomes and lysosomes, before return to the cell
in the S-shaped body (glomerulus precursors) and persists in the surface. However, biological function of this is unknow [52].
mature kidney [37,38]. However, in the above described case, Anti-PLA2R antibodies bind mainly with the CysR domain of
mother lacked NEP despite healthy condition; so antibodies pro­ PLA2R receptor [52,53], although CTLD1 and CTLD7 also can be
duction could have occurred after exposition to the paternal NEP on ligand points[53]. In fact, patients with autoantibodies that bind to
syncytiotrophoblastic cells [35]. the three domains had worst evolution than patients with antiPLA2
Lack of NEP appears to be influenced by genetic factors. NEP is ab that only join to the CysR domain [54].
encoded by the MME gene (membrane metallo-endopeptidase; also The binding of anti-PLA2 antibodies to the PLA2 receptor located
called EC 3.4.24.11, neprilysin, enkephalinase, CD10, CALLA) in the podocyte foot processes activate the complement with for­
composed of 24 exons on the chromosome 3. All immunized mothers mation of the MAC that causes the injury of the podocyte with
lacked NEP enzymatic activity by truncating mutations located in disorganization of the actin skeleton as well as of the cellular adhe­
exons 7 (in all cases described) and 15 (only in one case) [39–41]. sion molecules (Fig. 4).
However, severity of the renal disease was variable, depending on Binding of anti-PLA2R antibodies to PLA2R receptor on podocytes
the mother`s antibody response. It has been described that only the could alter receptor function. In normal conditions, binding of sol­
presence of anti-NEP IgG1 isotype is responsible for the development uble PLA2 (sPLA2) to its receptor has both positive and negative
of the disease [39–41]. These autoantibodies can inhibit the enzy­ functions (Table 1).
matic activity causing persistence of vasoactive peptides responsible
for the ischemic injury [41]. Renal function usually improves in post- - Thrombospondin type I domain-containing 7A: THSD7A was identified
natal life when contact with maternal IgG circulating is stopped, by western-blotting and mass spectrometry as a 250-kDa protein
reason why many cases of NEP-MN go undetected. [59]. It is not a member of the mannose receptor family as PLA2R. It
In the cases described, anti-NEP antibodies of the IgG1 and IgG4 is a large transmembrane glycoprotein characterized by the presence
subtypes have been found, so that while in cases where anti-NEP of multiple thrombospondin type-1 repeats (TSRs), which alternate
igG4 are produced, a rapid regression of the symptoms is observed, between group 1 and group 2 differing in the position of three
in those cases in which complement-fixing anti-NEP IgG1 is devel­ intradomain disulphide bonds [60] (Fig. 3B). Like PLA2R, also
oped, a severe disease requiring peritoneal dialysis and even kidney activate an IgG4 response in 3-5% of IMN patients. Unfortunately, so
transplantation is produced [41]. In any case, screening of families is far few studies have focused on the role of THSD7A in MN. THSD7A
essential because this disease could lead to chronic renal failure in participates in cell adhesion [61], being present in foot processes
adulthood with needed renal transplant and subsequent pregnancies near of slit diaphragms [62]. Binding of anti-THSD7A causes fibre
would have high risk for the fetus [42]. NEP is involved in the disorganization and cell dissociation and apoptosis [63]. Unlike pa­
catabolism of regulatory peptides with vasoactive properties and also tients with IMN associated with anti-PLA2R antibodies, MN associ­
participates in the blocking of peptide signalling events at the cell ated with anti-THSD7A has been shown to have an increased risk of
surface [43]. Injurious effects of anti-NEP antibody might be related developing tumors [64,65], specially of prostate, breast, renal and
to blockade of enzymatic activity that affects glomerular hemody­ colorectal, where THSD7A is over expressed [65].
namic and endothelial permeability [35,41]. - Other antigens: Recently, studies have appeared that describe new
- PLA2R antigen: Is the main autoantigen in adult IMN [11]. Type-M antigens that could be involved in the development of MN. Lecithin-
phospholipase A2 receptor (PLA2R) was identified by mass spec­ cholesterol acetyltransferase (LCAT) is an enzyme involved in
trometry as a 185kD protein present in normal human glomerular cholesterol metabolism which has been found in immunocomplexes
podocytes. IMN involves the formation of subepithelial deposits of with anti-LCAT antibodies on the walls of the glomerular capillaries
circulating anti-PLA2R IgG4 autoantibodies binding to podocyte of MN patients [66]. On the other hand, proteomic studies have
PLA2R in about 70%-80% of patients [11]. It has been seen a path­ allowed the detection in glomerular podocytes of antibodies against
ogenic function of these antibodies by early recurrence of MN in antigens such as superoxide dismutase 2, aldose reductase and alpha
kidney transplant recipients with circulating anti-PLA2R autoanti­ enolase, which are not present in cell surface of healthy glomerulus
bodies. However, some patients with high antibodies titers do not [67,68].
relapse. Also, it is unknown if anti-PLA2R antibodies start the disease
or are preceded of others factors. Thus, further studies are necessary PLA2R and THSD7A may represent about 85% of IMN. However, the
to find out the specific role of antibodies in the pathogenesis of the presence of false negatives has also been suggested when detecting an­
disease [44–47]. tibodies in the early stages of the disease [69]. In any case, the remaining
PLA2R is a member of the mannose receptor family as well as the 15% may be due to other antigens not yet described [70]. Possible
cation-dependent mannose-6-phosphate receptor, the C-type reasons why these new antigens have not been discovered were still
mannose receptor 2, the dendritic cell receptor DEC-205, and the picked up by Beck. [70] (Table 2).
avian yolk sac IgGY receptor FcRY [48,49]. To finish, studies that jointly compare the presence and absence of
All are transmembrane proteins with N-terminal cysteine rich the different antigens against which antibody production has been

4
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

Fig. 3. Structure of PLA2R and THSD7A. These receptors are two type I transmembrane glycoprotein. A) PLA2R: composed of a large extracellular portion which
consists of an N-terminal cysteine-rich region (CRD), a fibronectin-like type II domain (FNII), a tandem repeat of eight C-type lectin domains (CTLD), a trans­
membrane domain (TMD), and a short intracellular C-terminal domain (CD). In red are indicated typical variants associated with MN. The blue loop shows that in the
bent configuration, the-N terminal domain not binds with C-type lectin-like domains. B) THSD7A: composed of a large extracellular portion which consists of
thrombospondin type-1 repeats (TSRs), which tend to alternate between group 1 and group 2 TSRs, differing only in the positions of the three intradomain disulfide
bonds. GBM; Glomerular basement membrane.

Fig. 4. Anti-phospholipase A2 receptor antibodies and idiopathic membranous nephropathy in a peripheral capillary from a normal glomerulus. The basement
membrane separates endothelium from podocyte. A) Phospholipase A2 receptors (PLA2R) are localized on podocyte foot processes. B) In idiopathic membranous
nephropathy, autoantibodies bind to PLA2R. C) This activates complements with the formation of MAC and podocyte injury, with disociation of actin cytoskeleton.

5
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

Table 1 serum and urine of patients with MN compared with other renal patients
Functions of sPLA2-PLA2R binding (adapted from [50,55–58]). [93,94].TNFα seems to be involved in dyscohesion at the podocytes’ slit
Functions of sPLA2-PLA2R binding membrane with disorganization of actin filaments [95].
Nephrin (NPHS1) is a podocyte protein joining the slit diaphragm to
Positive Negative
modulators of actin, maintaining glomerular permeability [96] (Huber
Activation of the mitogen-activated Endocytosis of sPLA2 protecting cells and Benzing, 2005). Several polymorphisms in NPHS1 loci have been
protein kinase (MAPK) cascade. of enzymatic activities.
Production of lipid mediators Cell degradation
associated with susceptibility and remission to IMN [97].
Release of arachidonic acid in bone Also there is a relation between the 4G allele of Plasminogen acti­
marrow-derived mast cells vator inhibitor type (PAI1) and renal deterioration and increased car­
ROS production diovascular problems in MN patients [98]. This allele is associated with
Activation of DNA-damage pathways
increased transcriptional activity and therefore with high PAI1 levels
[99].
Some polymorphisms in the IL-6 and STAT4 [100,101] and Toll-like
Table 2 receptor 9 (TLR9) [102] genes appear to be related to increased prob­
Possible reasons why the remaining MN autoantigens have not been discovered ability of IMN.
yet (adapted from [70]).
All these data seem to indicate that a combination of several genetic
Rare antigens factors participate in the onset of the disease as well as in the control of
Non-glomerular antigen derived from circulation from other tissue or environment. glomerular permeability and the processes of inflammation and fibrosis.
Neo-antigen not expressed in normal kidney
Antigen that is difficult to extract from glomeruli
Humoral epitope not detectable with current available techniques 4.4. Exogenous antigen
Non-IgG4 autoantibody that difficult detection
BSA can contribute to the development of MN in about 2% of chil­
dren aged <5 years[103], since in some of these patients with IMN have
described in IMN patients are required to determine their usefulness in
been found high serum titres of anti-BSA antibodies (IgG1 and IgG4).
monitoring the disease.
Apparently, the BSA detected in children migrates in the basic pH region
while in adults it does in neutral pH ranges. Cationic BSA could be
4.3. Risk allele variants for idiopathic MN formed by differences in food processing and intestinal microbiota
respect to the adult [104,105]. Cow’s milk is the major source of BSA in
Some reports have suggested a genetic predisposition to IMN young children. All this suggests that some IMNs can be caused by
[22,25,26,71,72], although is not a typical Mendelian hereditary dis­ exogenous food antigens, being fundamental their identification to
ease. Related with this, IMN seem associated in Caucasoid with HLA- remove them from the diet instead of using immunosuppression in the
B*08 HLA-DRB1*03 haplotype as well some HLA-DQA1 alleles and treatment of kidney diseases. This has been explained by a dysregulation
other genes coded in HLA class II region of chromosome 6 as TAP and of the IL-13 route, leading to production of IgE specific antibodies of
HLA-DM [73–79]. Regards to HLA, recent genome-wide association food components[106]. It should be noted that BSA immunocomplexes
studies (GWAS) employed single-nucleotide polymorphisms (SNPs) have only been detected in those MN patients without anti-PLA2R an­
revealed high association of the 6p21 HLA-DQA1 and 2q24 PLA2R1 loci tibodies [89].
in IMN patients, showing an additive effect when 4 risk alleles are Alloimmune reactions also can happen in enzyme replacement
present increased the OR about 80-fold [3]. This association was therapy (ERT) [107–109], arising alloantibodies with or without clinical
confirmed in European [80,81] and Asian populations [82,83]. relevance. A few cases of MN by ERT have been reported, although the
Taking into account the linkage disequilibrium existing in the HLA prevalence is probably underestimated. Two of these patients with
genes, HLA haplotypes DRB1*03:01 DRB3*02:02 DQA1*05:01 Pompe and mucopolysacaccharidosis type VI who were treated respec­
DQB1*02:01 [51,80,84,85], DRB1*15:01 DRB5*01:01 DQA1*01:02 tively with recombinant human alpha-glucosidase (rhGAA) [110] and
DQB1*06:02[51,86–88] and MHC 8.1 (HLA-A*01 HLA-B*08 HLA-C*07) recombinant arylsulfatase B (rhASB) [111], developing MN. In these
[85] seem to confer risk of IMN. cases, given the need of patients to receive recombinant lysosomal en­
Several hypotheses have been proposed to explain the role of HLA in zymes, it is advisable to monitor their proteinuria to additionally pro­
the development of the disease. First, it is possible a direct interaction vide immunosuppressive therapy when necessary and thus avoid a
between antigen binding sites on HLA-DQA1 molecules and specific relapse of kidney disease.
PLA2R derived peptides. Second, mimicry between microorganisms or On the other hand, Hepatitis, Helicobacter pylori and tumor antigens
other exogenous antigens and some variants of PLA2R could activate the as well as thyroglobulin, and DNA-containing material have been found
production of anti-PLA2R antibodies in patients with some HLA-DQA1 in subepithelial deposits in patients with secondary MN [112–115].
alleles which predispose to autoimmune diseases [89]. Finally, a rare form of IMN is characterized by deposits of mono­
It has been suggested that variants that affect the coding and non- clonal IgG1 (rarely IgG3) in the outer layer of basement membranes
coding regions of PLA2R1 could influence the degree of expression of [116]. These immunoglobulines are not apparently directed against any
PLA2R as well as the conformation of binding epitopes with antibodies, glomerular structure, although their precipitation in the subepithelial
triggering autoimmune events [90]. However, only sequences in 5’ space induces the development of MN [117].
intronic regions have been related to MN [90], maybe by alteration in Taking all this into account, it seems that in the future new antigens
the expression level or in transcription process. may be found in MN patients.
Using predictive algorithms, sequences in CTLD1 and CTLD7 do­
mains of PLA2R receptor have showed high binding affinity with HLA 4.5. Role of complement in MN
mollecules [87]. However, more sequence-based methods and cell and
molecular analyses are needed to understand how variants of PLA2R1 Complement activation is normally regulated to avoid autoimmune
and HLA-DQA1 loci interact increasing predisposition to IMN. events by several complement regulatory proteins (CRPs).
Other genes have also been related with predisposition to IMN. The In podocytes, the surface-bound regulatory proteins CR1 (C3b re­
tumor necrosis factor (TNF) allele G308A and the TNFd microsatellite ceptor), membrane cofactor protein (MCP, CD46), and decay-
polymorphisms are strongly associated with occurrence and initiation of accelerating factor (DAF, CD55) act at C3 and C5 convertases level. In
MN by increased TNF production[91,92], whose levels have risen in the addition, formation of membrane attack complex (MAC, C5b-9) is

6
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

prevented by plasma clusterin and vitronectin and the podocyte- proteasome system and autophagy [135–137]. However, continuous
expressed CD59. Their regulatory capacity can be overcome by the de­ ER stress is cytotoxic by activation of pro-apoptotic pathways [138].
posit of immunocomplex in the subepithelial area of podocytes
[118,119] or several polymorphisms of CRPs [120–122] (Fig. 5). 5. Diagnostics
Unlike the secondary MN where the classic pathway of complement
plays an important role, IMN is characterized by IgG4 deposits which First assessment of a patient with a nephrotic syndrome suggestive of
not activate this pathway [123,124], being the lectin and alternative MN must include detailed medical history and patient examination.
pathways involved in the MAC formation. In fact, characteristics C1q Analytical test should include serological markers of systemic autoim­
deposits of the classical pathway are generally not seen on immuno­ munity such as antinuclear antibodies, anti-dsDNA, anti-ribonucleo­
fluorescence of IMN patients. Further, mannan-binding lectin (MBL) has proteins, anti-Smith, antiRo(SSA), anti-La(SSB), anti-topoisomerase I
been identified in the glomeruli of many patients with IMN [125], which (Scl-70), anti-centromere, anti-Jo-1, anti-cardiolipin, anti-ß2-glycopro­
seems bind to anti PLA2R IgG4 antibodies because they lack galactose tein I antibodies, thyroid autoimmunity and complement values. These
[33]. are complementary tests that help diagnose primary membranous ne­
However, there are exceptions. About 5% of PLA2R-associated IMN phropathy. [139,140].
patients lack antibodies of IgG4 subclass. Besides, neonatal alloimmune Proteinuria and serum creatinine are considered the gold-standard
MN is produced by maternal IgG1. [47,126]. clinical biomarkers to risk-stratify MN patients. Moreover, urinary
Complement activation and MAC formation by immune complex markers of renal tubular damage, such as Beta2 microglobulin, N-acetyl-
deposits participate in direct podocyte injury and proteinuria [99,101]. β-D-glucosaminidase (NAG), retinol-binding protein (RBP), kidney
Presence of MAC in low concentration activates several mediators that injury molecule 1 (KIM-1) and neutrophil gelatinase-associated lipocalin
modify the metabolism of podocytes, the structure of the cytoskeleton, (NGAL) have been also proposed to risk-stratify patients with MN.
the location of nephrine, the formation of extracellular matrix as well as However, the levels of these biomarkers seem to not correlate with the
the integrity of DNA [32,127,128]. Also causes upregulation of NADPH severity of the disease [141].
oxidoreductase with formation of reactive oxygen species which injury Currently, the diagnosis of MN essentially relies on the clinical fea­
several components of podocyte and glomerular basement membrane tures and pathological findings after renal biopsy. However, biopsy is an
components. Related with this, podocyte produce new extracellular invasive procedure with some disadvantages, such as risk of bleeding,
matrix that accumulate between immune deposits originating the secondary infection and elevated cost [142]; nevertheless, staining of
typical “spikes” observed by silver staining [89]. renal tissue is more sensitive, since serum antibodies can become
Simultaneously podocytes active mechanisms to limit the injury and negative spontaneously, or when patients are treated with immuno­
promote recovery of complement-dependent cytotoxicity [129–131]. suppression. Moreover, findings from renal biopsies are helpful not only
One of these mechanisms is based on endocytosis of the MAC and its for confirming the diagnosis of MN, but also allowing an estimation of
subsequent elimination in the urine, being a biomarker of kidney dam­ tissue damage, i.e. tubular atrophy and interstitial fibrosis or glomerular
age [132]. sclerosis, as well as diagnosis of lesions not related to MN [140].
Besides, the accumulation of misfolded proteins in the endoplasmic Nevertheless, antibody detection in serum of nephrotic patients has been
reticulum (ER) leads to ER stress [133,134]. Various studies suggest that shown to be extremely specific for the diagnosis of MN; these serological
complement react against these proteins together with the ubiquitin- tests are now commercially available and is increasingly being

Fig. 5. Summary of complement activation and regulation in membranous nephropathy. Complement can be activated by classical, lectin, or alternative pathways.
While the classical pathway is activated in secondary forms, lectin and alternative pathways play an important role in primary MN. All three pathways converge to C5
convertase formation. Regulatory proteins (complement receptor-1 [CR1], decay accelerating factor [DAF], membrane cofactor protein [MCP]) control de formation
of C5 convertase. Cleavage of C5 produces the chemotactic factor C5a and C5b which combines with C6, C7, C8, and multiple C9 molecules to form the membrane
attack complex (MAC). C5b-9 formation is regulated by cell-bound CD59. Clusterin and vitronectin prevent insertion of MAC in the membrane

7
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

implemented into clinical practice. The reported sensitivities and spec­ exclusive, however, rare cases of dual positivity have been described
ificities are highly variable depending on the method used, patient [154,155]. The detection of circulating autoantibodies have been
population studied, and cut-off levels that are used to define a positive possible by western blot and IIF, which provide only semi-quantitative
result [143]. titers. In 2019, a novel ELISA was reported as an useful tool in the
diagnosis of MN patients for THSD7A-associated disease, to carefully
5.1. Serological detection techniques monitor disease activity and response to treatment during follow-up,
and predict clinical outcome[154].
Anti-PLA2R was first identified in patient sera by performing western
blot (WB) using protein extracts from normal human glomeruli, which 5.1.3. Other potentially pathogenic antibodies
were electrophoresed under non-reducing conditions (SDS-PAGE). Other proteins postulated that act as autoantigens in MN include
However, this method has limited use in clinical practice because it is anti–manganese superoxide dismutase (SOD2), aldose reductase (AR),
time-consuming and has a complex standardization[144,145]. In 2011, cationic bovine serum albumin and alpha-enolase in cases of childhood
a recombinant cell-based indirect immunofluorescence (IIF) assay for MN [103]. Anti-alpha-enolase antibodies have been known to be present
detection of anti-PLA2R antibodies was developed [146]. Although this in the serum of patients with primary and secondary MN (nearly 70%)
assay is relatively inexpensive and easy to perform, it is not well suited but they are not detected within the subepithelial deposits [156,157].
to high throughput laboratories and can be troubled by subjective Antibodies directed against SOD2 and AR, have also been detected in the
interpretation [147,148]. serum and tissue of glomeruli in biopsy samples from patients with MN
Years later, a group developed an in-house enzyme-linked immu­ [67]. These cytoplasmic antigens are not usually accessible to circu­
nosorbent assay (ELISA) which included as solid-phase an extracellular lating antibodies under normal conditions. However, when oxidative
domain of PLA2R produced by recombinant techniques [149]. ELISA stress occurs, they can migrate to the cell membrane and serve as targets
may become a preferred method in many clinical laboratories because it for circulating antibodies. The activation of complement leading could
provides quantitative results, is not prone to subjective interpretation, be a cause of oxidative stress in podocytes [158]. The initial lesions
and is well-suited for high sample volumes [143]. induced by the immune complexes involving PLA2R may induce
Recently, a novel quantitative assay was reported. This is an oxidative stress responsible for the novo membrane expression of pro­
observer-independent immunoassay on an addressable laser bead teins, which are physiologically cytoplasmic, leading to the formation of
immunoassay (ALBIA) platform (Luminex technology) that employed new autoantibodies. Their pathogenic role in the initiation and/or
cell lysates bearing the full-length recombinant human protein to reli­ maintenance of the disease is still hypothetical [12].
ably detect anti-PLA2R antibodies in the serum of IMN patients. This
immunoassay is capable of test multiple targets of other nephrotoxic 5.2. Histological features of MN
antibodies or immunological markers in a single assay and requires only
small serum sample volumes of 2–20 μL [147,148]. A comparison be­ Renal biopsy is still the gold standard for the diagnosis of MN. A
tween the CBA-IFA, ELISA and ALBIA platforms, showed similar ca­ granular positivity for IgG along the glomerular basement membrane
pacity across the different tests to detect anti-PLA2R autoantibodies and electrondense deposits restricted to the subepithelial space of the
[148]. glomerular filtration barrier are the characteristic morphologic features.
The diagnosis of PLA2R-associated MN can be made by staining for the
5.1.1. Anti PLA2R antibodies PLA2R antigen in the renal tissue [7,140,155,159] (Fig. 1).
PLA2R autoantibodies has proved to be a valuable biomarker for the Tissue testing for PLA2R antigen can been performed by immuno­
diagnosis of IMN, with a high specificity. However, it is important to histochemistry (IHC) on paraffin-embedded tissue, IIF on paraffin-
remember that they are also found in a very small proportion of sera embedded tissue and IIF on frozen[11,140,160]. Subepithelial deposits
from patients with secondary MN. Anti-PLA2R antibodies are not only in secondary forms of MN are almost always PLA2R-negative.[143,161].
useful for diagnosis; a positive test may be used in conjunction with Generally, a strong association between glomerular PLA2R staining and
clinical features to indicate a need for immunosuppressive therapy and circulating anti-PLA2R antibodies is found [150], particularly when
the autoantibody titers used to monitor patients during therapy [148]. It autoantibody levels are measured at the same time of the biopsy
is useful because circulating anti-PLA2R antibodies reflect the immu­ assessment. However, glomerular PLA2R staining is not considered a
nological activity of disease and have been shown to disappear before diagnostic test for active disease, since the positivity of glomerular
clinical remission of nephrotic syndrome and to reappear in the circu­ PLA2R staining with undetectable circulating anti-PLA2R autoanti­
lation before clinical relapse [147]. bodies is unlikely and may reflect an immunologically inactive disease
Anti-PLA2R antibodies prevalence can vary by geographical regions. as a positive PLA2R antigen can persist for weeks or months after
This antibodies are positive in approximately 70 to 80% of patients with remission [141,162].
IMN in Europe[150], EEUU[11], and Asia, with the exception of Japan IgG subclass analysis in the tissue indicates that IgG subclass patterns
where the prevalence of these antibodies is lower than in other Asian may give some information about pathogenesis and prognosis. [11,163].
countries (about 50% of patients with IMN) [151]. Immunohistological studies have shown that IgG4 is the predominant
After a kidney transplantation, anti-PLA2R antibodies are excep­ IgG subclass in the glomerular immune deposits in IMN, leading to the
tionally detected in “de novo” cases as compared with recurrent MN in assumption that IgG4 antibodies may induce disease. In contrast, the
kidney transplant patients (8% versus 83%)[152]. Nearly 50% of cases absence of IgG4 and the presence of IgG1 and IgG2 in renal biopsies are
of recurrent MN on the kidney graft are associated with the presence of considered to be characteristic for secondary, malignancy-associated
anti-PLA2R antibodies before the trasplantation. The detection or MN [164,165].
persistence of anti-PLA2R in kidney transplant patients has been asso­
ciated with an increased risk of loss of graft function [153]. 6. Treatment

5.1.2. THSD7A antibodies MN have been described as a large spectrum of disease severity and
In 2014, anti-thrombospondin type 1 domain-containing 7A shows cases of spontaneous remission without therapy, being difficult to
(THSD7A) was identified as a second autoantigen for another group of predict its progression is, which explains the therapeutic uncertainties
MN patients. [4,10]. These antibodies were noted to be positive in 3%– that still persist today. In less than 30% of the cases, the disease pro­
5% of patients with IMN who were anti-PLA2R negative, with higher gresses slowly towards severe renal insufficiency despite optimal sup­
prevalence in women [4]. PLA2R and THSD7A use to be mutually portive care combined with classical immunosuppressive therapy

8
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

[147,166–168]. who present thrombotic risk factors such as obesity, genetic predispo­
Today it is accepted that the type of treatment must be adapted to the sition, heart failure, and Prophylactic anticoagulant therapy should be
the risk of progressive loss of kidney function of each patient, based in based on an estimate of the risk of thrombotic events and the risk of
clinical and laboratory data (GFR (glomerular filtration rate; protein­ bleeding complications [169].
uria, response to renin-angiotensin-aldosterone system inhibitors On the other hand, forms that persist over time with nephrotic syn­
(ACEi), serum albumin, anti-PLA2R antibodies levels, etc.) and can be drome without deterioration of renal function will require immuno­
summarized into 4 categories: low, moderate, highy and very high risk suppressive therapy, being predictive factors of progression to the end-
[169]. A brief outline of the different treatment guidelines is shown in stage kidney disease (ESKD) a nephrotic persistent proteinuria for more
Fig. 6. than 6 months, a decrease in glomerular filtration rate at the time of
General considerations for treating patients with primary MN diagnosis, uncontrolled arterial hypertension and the presence of
include that forms with the possibility of spontaneous remission (non interstitial fibrosis and tubular atrophy seen on a renal biopsy
nephrotic proteinuria (<3.0 g/day), high serum creatinine level or un­ [147,169,174,175].
controlled arterial hypertension [170]) should receive supportive Severe forms with progressive deterioration of kidney function, in­
treatment, consisting of renoprotective treatment as drugs that inhibit cludes patients with a progressive deterioration of renal function (in­
the renin-angiotensin-aldosterone system (ACEs), control of arterial crease in creatinine> 30% of the baseline value during the first 6-12
hypertension, dyslipidaemia, excess weight, and other cardiovascular months of evolution), complications caused by nephrotic syndrome (i.e.
risk factors [169] in appropriate doses to maintain a Blood Pressure pulmonary thromboembolism) or with extreme hypoalbuminemia (less
<130/80 mmHg and exert a sustained antiproteinuric effect. As re­ 1.5-2 g/dl) and persistent massive edema that does not respond to
flected in various studies [171,172] these patients have a good evolution combinations of diuretics. In these patients, the use of anticalcineurinics
without immunosuppression. Induction of partial remission (proteinuria is not recommended due to their nephrotoxicity. Furthermore, there are
less than 3.5 g/24h) is associated with significantly higher renal survival hardly any studies on the efficacy of treatment with RTX and MMF,
than non-remission [173]. Therefore, although the optimal goal would among others. Therefore, the most widely used therapeutic option is the
be to achieve a complete remission, obtaining a partial remission can be Ponticelli scheme[166,176]. In fact, in a study comparing the efficacy of
considered acceptable. the Ponticelli scheme, cyclosporine, and supportive treatment, the first
In accordance with the KDIGO guidelines [169], Immunosuppressive group showed statistically significantly better results [177].
therapy should be restricted to patients considered at risk for progressive Below is a brief description of the main immunosuppressive treat­
kidney injury, cases that maintain nephrotic proteinuria after an ments used:
observation period of at least 6 months and provided that proteinuria Corticosteroids plus alkylating agents (cyclophosphamide or chlor­
does not tend to decrease. Patients with advanced chronic renal failure ambucil): The combination of corticosteroids and cyclophosphamide or
(glomerular filtration rate <30 ml/min/1.73m2) should not follow chlorambucil, administered alternatively for 6 months, starting with
immunosuppressive treatment either, given the limited benefit it can corticosteroids, constitutes the Ponticelli scheme. The rate of complete
provide in these cases. Patients should follow a diet without salt and or partial remissions reaches 70-80% of cases [168,178], although side
with diuretics to reduce edema and allow a normal life. Treatment with effects can be serious, especially when chlorambucil is used. That is why
ACE inhibitors, in addition to treating hypertension, reduces protein­ cyclophosphamide is currently used as an alkylating agent. Some au­
uria, facilitating possible spontaneous remission. Regarding anticoagu­ thors prefer the simultaneous use of both treatments [166,176],
lant treatments, the KDIGO guidelines recommend their use in patients although it is unknown which regimen is more effective. What is known

Fig. 6. Risk-based treatment of Membranous Nephrophathy according to KDIGO 2021 guideline (adapted from [169] (RTX: Rituximab; CNI: Calcineurin inhibitor).

9
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

is that the exclusive use of corticosteroids is not effective[179]. actually in the treatment of MN [207]. In the future, treatment protocols
Calcineurin inhibitor (CNI or anticalcineurinics): The use of cyclo­ for MN patients will probably be individualized based on the level of
sporine[180] and tacrolimus [181–186] has been shown effective in anti-PLA2R1 antibodies and proteinuria [208].
MN, inducing complete or partial remission in more than 70-80% of
cases. Usually, the initial dose of cyclosporine is 3.5-5.0 mg/kg/d, 7. Conclusions and future directions
accompanied by steroids, while that of tacrolimus is 0.05-0.075 mg/kg/
d without steroids [180]. The doses of both drugs are adjusted according The description of the autoimmune nature of multiple cases of IMN
to the blood levels. The recommended duration of treatment with has meant an important change in the therapeutic approach, diagnosis
anticalcineurinics is 12-18 months with gradual withdrawal. In addition and monitoring of this pathology. In this respect, the discovery of new
to their immunosuppressive effect, they also have a direct anti­ autoantibodies and their epitopes could well pave the way for a better
proteinuric effect on the podocyte thanks to the interaction with syn­ understanding of the immunopathogenesis of IMN. Therapies based on
aptopodin [187]. The main problem generated in the treatment of MN protreosome inhibitors and the use of new monoclonal antibodies tar­
with anticalcineurinics is the high rate of relapse (close to 50%) after geting B cells may likewise have a place in future treatment.
withdrawal of the drug, which is usually associated with high protein­
uria. Some studies have shown that the administration of rituximab Funding
before starting tacrolimus withdrawal can reduce the number of relapses
[188]. Studies are underway to evaluate the efficacy of this sequential “This research received no external funding”
therapy with tacrolimus-rituximab. The response to anticalcineurinics is
more effective with lower proteinuria and better renal function at the
Ethical approval
beginning of treatment.
Rituximab: There are case series studies in which the use of rituximab
Not required.
(RTX) induces complete or partial remission of the nephrotic syndrome
in 50% -60% of cases [189–191] with good tolerance. There is no
consensus on the optimal regimen of RTX to treat MN. The doses used Declaration of competing interest
vary between 375 mg/m2/week for four consecutive weeks or 1 or 2
doses of 1 g [192]. Anti-PLA2R1 antibodies levels are useful for the “The authors declare no conflict of interest.”
evaluation of rituximab efficacy. No correlation has been seen between
the degree of CD20+ lymphocyte depletion and the occurrence of re­
References
missions or relapses [193,194]. In a study called GENRITUX [190], no
short-term differences were observed between treatment with rituximab [1] M. Schulze, et al., Elevated urinary excretion of the C5b-9 complex in
plus supportive treatment and only conservative treatment. However, in membranous nephropathy, Kidney Int. 40 (3) (Sep. 1991) 533–538.
the long term, the group treated with rituximab presented a greater [2] R.J. Glassock, Human idiopathic membranous nephropathy — a mystery solved?
N. Engl. J. Med. 361 (1) (2009) 81–83.
increase in serum albumin, a more marked decrease in the anti-PLA2R [3] H.C. Stanescu, et al., Risk HLA-DQA1 and PLA(2)R1 alleles in idiopathic
titer, and a greater degree of remission. This has been seen in other membranous nephropathy, N. Engl. J. Med. 364 (7) (Feb. 2011) 616–626.
studies as well[44,153,195]. Therefore, it appears that the use of rit­ [4] N.M. Tomas, et al., Thrombospondin type-1 domain-containing 7A in idiopathic
membranous nephropathy, N. Engl. J. Med. 371 (24) (Dec. 2014) 2277–2287.
uximab in patients with MN is slow to take effect. A new protocol based [5] E. Hoxha, et al., An indirect immunofluorescence method facilitates detection of
on the association of RTX and cyclosporine for 6 months, followed by a thrombospondin type 1 domain containing 7A specific antibodies in membranous
second cycle of RTX and tapering of cyclosporine during 18 months as a nephropathy, J. Am. Soc. Nephrol. 28 (2) (2017) 520–531.
[6] A. Mcgrogan, C.F.M. Franssen, C.S. De Vries, The Incidence of Primary
maintenance phase, demonstrated that proteinuria decreased by 80% at
Glomerulonephritis Worldwide: A Systematic Review of the Literature, November
6 months, although complete remission only was observed in 54% of 2010, 2011, pp. 414–430.
patients at 12 months [196]. Some studies suggest that rituximab may [7] W.G. Couser, Primary membranous nephropathy, Clin. J. Am. Soc. Nephrol. 12
(6) (Jun. 2017) 983–997.
be an effective option in patients with recurrence of MN or MN de novo
[8] W.L. Lai, et al., Membranous nephropathy: a review on the pathogenesis,
after kidney transplant [197]. Although it is safe, several cases of acute diagnosis, and treatment, J. Formos. Med. Assoc. 114 (2) (Feb. 2015) 102–111.
non ischemic cardiomyopathy [198] and progressive urticarial derma­ [9] J.M. Francis, L.H.J. Beck, D.J. Salant, Membranous nephropathy: a journey from
titis [199] related to the RTX administration have been described. bench to bedside, Am. J. kidney Dis. Off. J. Natl. Kidney Found. 68 (1) (Jul. 2016)
138–147.
Mycophenolate (MMF): Existing studies with this drug show con­ [10] K. Keri, S. Blumenthal, T. Chongkrairatanakul, V. Kulkarni, L. Beck, Primary
flicting results. Thus, there are studies that demonstrate its efficacy membranous nephropathy: comprehensive review and historical perspective,
[200] and others in which no differences are observed with patients with Postgrad. Med. J. 95 (1119) (2019).
[11] L.H.J. Beck, et al., M-type phospholipase A2 receptor as target antigen in
supportive treatment [201]. MMF significantly reduced proteinuria but idiopathic membranous nephropathy, N. Engl. J. Med. 361 (1) (Jul. 2009) 11–21.
had no significant effect on the induction of complete remission and was [12] D.C. Cattran, P.E. Brenchley, Membranous nephropathy: integrating basic science
also associated with an increased risk of relapse[176,202]. MMF may be into improved clinical management, Kidney Int. 91 (3) (Mar. 2017) 566–574.
[13] C. Lefaucheur, et al., Membranous nephropathy and cancer: Epidemiologic
beneficial when associated with steroids [169]. Other studies suggest evidence and determinants of high-risk cancer association, Kidney Int. 70 (8)
that the use of MMF may be useful in patients with resistance to other (Oct. 2006) 1510–1517.
therapies although further studies are needed to confirm this [203]. [14] R.J. Glassock, Diagnosis and natural course of membranous nephropathy, Semin.
Nephrol. 23 (4) (Jul. 2003) 324–332.
Adrenocorticotropic hormone (ACTH): Subcutaneous administration
[15] X.-D. Zhang, et al., Clinical implications of pathological features of primary
of synthetic ACTH has shown some efficacy in patients with MN and membranous nephropathy, BMC Nephrol. 19 (1) (Aug. 2018) 215.
nephrotic syndrome in several case series [204,205]. Their long-term [16] N. Polanco, et al., Spontaneous Remission of Nephrotic Syndrome in Idiopathic
Membranous Nephropathy, 2010, pp. 697–704.
efficacies as well as its side effects are unknown.
[17] P. Ronco, H. Debiec, Pathophysiological advances in membranous nephropathy:
Proteasome inhibitor: A novel therapy based on proteasome inhibitor time for a shift in patient’s care, Lancet (London, England) 385 (9981) (May
(bortezomib, 4 doses of 1.3 mg/m over 2 weeks) efficiently reduced 2015) 1983–1992.
proteinuria in a case of recurrent MN after renal transplantation that had [18] W. Lu, S. Gong, J. Li, Y. Wang, Clinicopathological features and prognosis in
patients with idiopathic membranous nephropathy with hypertension, Exp. Ther.
been only partially responsive to RTX [206]. In addition to rituximab, Med. 19 (4) (Apr. 2020) 2615–2621.
the development of new monoclonal antibodies targeting B cells such as [19] D. Kerjaschki, Pathogenetic concepts of membranous glomerulopathy (MGN),
ofatumumab and belimumab as well as new treatments such as borte­ J. Nephrol. 13 (Suppl. 3) (2000) S96–100.
[20] R.J. Glassock, The pathogenesis of idiopathic membranous nephropathy: a 50-
zomib and eculizumab provides a variety of useful therapies with the year odyssey, Am. J. kidney Dis. Off. J. Natl. Kidney Found. 56 (1) (Jul. 2010)
potential to replace the nonspecific and toxic immunosuppressants used 157–167.

10
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

[21] P. Ronco, H. Debiec, Antigen identification in membranous nephropathy moves [52] M. Fresquet, et al., Identification of a major epitope recognized by PLA2R
toward targeted monitoring and new therapy, J. Am. Soc. Nephrol. 21 (4) (Apr. autoantibodies in primary membranous nephropathy, J. Am. Soc. Nephrol. 26 (2)
2010) 564–569. (Feb. 2015) 302–313.
[22] A. Vangelista, R. Tazzari, V. Bonomini, Idiopathic membranous nephropathy in 2 [53] B. Seitz-Polski, et al., Epitope spreading of autoantibody response to PLA2R
twin brothers, Nephron 50 (1) (1988) 79–80. Switzerland. associates with poor prognosis in membranous nephropathy, J. Am. Soc. Nephrol.
[23] D. Bockenhauer, et al., Familial membranous nephropathy: an X-linked genetic 27 (5) (May 2016) 1517–1533.
susceptibility? Nephron. Clin. Pract. 108 (1) (2008) c10–c15. [54] B. Seitz-Polski, et al., Cross-reactivity of anti-PLA2R1 autoantibodies to rabbit and
[24] W.A. Border, H.J. Ward, E.S. Kamil, A.H. Cohen, Induction of membranous mouse PLA2R1 antigens and development of two novel ELISAs with different
nephropathy in rabbits by administration of an exogenous cationic antigen, diagnostic performances in idiopathic membranous nephropathy, Biochimie 118
J. Clin. Invest. 69 (2) (Feb. 1982) 451–461. (Nov. 2015) 104–115.
[25] K. Sato, et al., Idiopathic membranous nephropathy in two brothers, Nephron 46 [55] P. Ancian, G. Lambeau, M. Lazdunski, Multifunctional activity of the extracellular
(2) (1987) 174–178. domain of the M-type (180 kDa) membrane receptor for secretory phospholipases
[26] I. Elshihabi, C.I. Kaye, A. Brzowski, Membranous nephropathy in two human A2, Biochemistry 34 (40) (Oct. 1995) 13146–13151.
leukocyte antigen-identical brothers, J. Pediatr. 123 (6) (Dec. 1993) 940–942. [56] K. Hanasaki, H. Arita, Phospholipase A2 receptor: a regulator of biological
[27] M. Rosenzwajg, et al., B- and T-cell subpopulations in patients with severe functions of secretory phospholipase A2, Prostaglandins Other Lipid Mediat.
idiopathic membranous nephropathy may predict an early response to rituximab, 68–69 (Aug. 2002) 71–82.
Kidney Int. 92 (1) (Jul. 2017) 227–237. [57] G. Lambeau, M.H. Gelb, Biochemistry and physiology of mammalian secreted
[28] W. Heymann, D.B. Hackel, S. Harwood, S.G. Wilson, J.L. Hunter, Production of phospholipases A2, Annu. Rev. Biochem. 77 (2008) 495–520.
nephrotic syndrome in rats by Freund’s adjuvants and rat kidney suspensions, [58] A. Augert, C. Payré, Y. de Launoit, J. Gil, G. Lambeau, D. Bernard, The M-type
Proc. Soc. Exp. Biol. Med. Soc. Exp. Biol. Med. (New York, N.Y.) 100 (4) (Apr. receptor PLA2R regulates senescence through the p53 pathway, EMBO Rep. 10
1959) 660–664. (3) (Mar. 2009) 271–277.
[29] B.J. Van Damme, G.J. Fleuren, W.W. Bakker, R.L. Vernier, P.J. Hoedemaeker, [59] A. Kronbichler, J. Oh, B. Meijers, G. Mayer, J. Il Shin, Recent progress in
Experimental glomerulonephritis in the rat induced by antibodies directed deciphering the etiopathogenesis of primary membranous nephropathy, Biomed.
against tubular antigens. V. Fixed glomerular antigens in the pathogenesis of Res. Int. 2017 (2017) 1936372.
heterologous immune complex glomerulonephritis, Lab. Investig. 38 (4) (Apr. [60] K. Tan, et al., Crystal structure of the TSP-1 type 1 repeats: a novel layered fold
1978) 502–510. and its biological implication, J. Cell Biol. 159 (2) (Oct. 2002) 373–382.
[30] W.G. Couser, D.R. Steinmuller, M.M. Stilmant, D.J. Salant, L.M. Lowenstein, [61] C.-H. Wang, et al., Thrombospondin type I domain containing 7A (THSD7A)
Experimental glomerulonephritis in the isolated perfused rat kidney, J. Clin. mediates endothelial cell migration and tube formation, J. Cell. Physiol. 222 (3)
Invest. 62 (6) (Dec. 1978) 1275–1287. (Mar. 2010) 685–694.
[31] P. Shah, A. Tramontano, S.P. Makker, Intramolecular epitope spreading in [62] M. Gödel, F. Grahammer, T.B. Huber, Thrombospondin type-1 domain-containing
Heymann nephritis, J. Am. Soc. Nephrol. 18 (12) (Dec. 2007) 3060–3066. 7A in idiopathic membranous nephropathy, N. Engl. J. Med. 372 (11) (Mar.
[32] A.V. Cybulsky, R.J. Quigg, D.J. Salant, Experimental membranous nephropathy 2015). United States, p. 1073.
redux, Am. J. Physiol. Ren. Physiol. 289 (4) (Oct. 2005) F660–F671. [63] N.M. Tomas, et al., Autoantibodies against thrombospondin type 1 domain-
[33] S.D. Ma H, L.H. Beck, Membranous nephropathy-associated anti-phospholipase containing 7A induce membranous nephropathy, J. Clin. Invest. 126 (7) (Jul.
A2 receptor IgG4 autoantibodies activate the lectin comple- ment pathway, 2016) 2519–2532.
J. Am. Soc. Nephrol. 22 (62A) (2011) p. [Abstract]. [64] E. Hoxha, T. Wiech, P.R. Stahl, G. Zahner, N.M. Tomas, C. Meyer-Schwesinger,
[34] T. Prabakaran, et al., Receptor-mediated endocytosis of α-galactosidase a in U. Wenzel, M. Janneck, O.M. Steinmetz, U. Panzer, S. Harendza, A mechanism for
human podocytes in fabry disease, PLoS One 6 (9) (Sep. 2011), e25065. cancer-associated membranous nephropathy, N. Engl. J. Med. 374 (2016)
[35] H. Debiec, et al., Antenatal membranous glomerulonephritis due to anti–neutral 1995–1996.
endopeptidase antibodies, N. Engl. J. Med. 346 (26) (Jun. 2002) 2053–2060. [65] P.R. Stahl, E. Hoxha, T. Wiech, C. Schröder, R. Simon, R.A.K. Stahl, THSD7A
[36] P. Ronco, H. Debiec, V. Guigonis, Mechanisms of disease: alloimmunization in expression in human cancer, Genes Chromosom. Cancer 56 (4) (Apr. 2017)
renal diseases, Nat. Clin. Pract. Nephrol. 2 (7) (Jul. 2006) 388–397. 314–327.
[37] J.L. Platt, T.W. LeBien, A.F. Michael, Stages of renal ontogenesis identified by [66] S. Takahashi, et al., Nephrotic syndrome caused by immune-mediated acquired
monoclonal antibodies reactive with lymphohemopoietic differentiation antigens, LCAT deficiency, J. Am. Soc. Nephrol. 24 (8) (Jul. 2013) 1305–1312.
J. Exp. Med. 157 (1) (Jan. 1983) 155–172. [67] M. Prunotto, et al., Autoimmunity in Membranous Nephropathy Targets Aldose
[38] M.-D. Appay, F. Mounier, M.-C. Gubler, M. Rouchon, A. Beziau, M. Reductase and SOD2, 2010, pp. 507–519.
D. Kazatchkine, Ontogenesis of the glomerular C3b receptor (CR1) in fetal human [68] M. Bruschi, et al., Direct characterization of target podocyte antigens and auto-
kidney, Clin. Immunol. Immunopathol. 37 (1) (1985) 103–113. antibodies in human membranous glomerulonephritis: alfa-enolase and
[39] H. Debiec, et al., Role of truncating mutations in MME gene in fetomaternal borderline antigens, J. Proteome 74 (10) (Sep. 2011) 2008–2017.
alloimmunisation and antenatal glomerulopathies, Lancet (London, England) 364 [69] M.S. Ryan, A.A. Satoskar, G.M. Nadasdy, S.V. Brodsky, J.A. Hemminger,
(9441) (Oct. 2004) 1252–1259. T. Nadasdy, Phospholipase A2 receptor staining is absent in many kidney biopsies
[40] P. Ronco, H. Debiec, Molecular pathomechanisms of membranous nephropathy: with early-stage membranous glomerulonephritis, Kidney Int. 89 (6) (Jun. 2016)
from Heymann nephritis to alloimmunization, J. Am. Soc. Nephrol. 16 (5) (May 1402–1403. United States.
2005) 1205–1213. [70] L.H.J. Beck, PLA2R and THSD7A: disparate paths to the same disease? J. Am. Soc.
[41] M. Vivarelli, et al., Genetic homogeneity but IgG subclass-dependent clinical Nephrol. 28 (9) (Sep. 2017) 2579–2589.
variability of alloimmune membranous nephropathy with anti-neutral [71] C.D. Short, J. Feehally, R. Gokal, N.P. Mallick, Familial membranous
endopeptidase antibodies, Kidney Int. 87 (3) (Mar. 2015) 602–609. nephropathy, Br. Med. J. (Clin. Res. Ed.) 289 (6457) (Dec. 1984), pp. 1500 LP –
[42] J.L. Nortier, et al., Neonatal disease in neutral endopeptidase alloimmunization: 1500.
lessons for immunological monitoring, Pediatr. Nephrol. 21 (10) (Oct. 2006) [72] D. Vasmant, K. Murnaghan, A. Bensman, J.Y. Muller, B. Mougenot, Familial
1399–1405. idiopathic membranous glomerulonephritis, Int. J. Pediatr. Nephrol. 5 (4) (Dec.
[43] A.J. Turner, R.E. Isaac, D. Coates, The neprilysin (NEP) family of zinc 1984) 193–196.
metalloendopeptidases: genomics and function, Bioessays 23 (3) (Mar. 2001) [73] P.T. Klouda, et al., Strong association between idiopathic membranous
261–269. nephropathy and HLA-DRW3, Lancet (London, England) 2 (8146) (Oct. 1979)
[44] R. Stahl, E. Hoxha, K. Fechner, PLA2R autoantibodies and recurrent membranous 770–771.
nephropathy after transplantation, N. Engl. J. Med. 363 (5) (Jul 2010) 496–498. [74] J.C. Le Petit, B. Laurent, F.C. Berthoux, HLA-DR3 and idiopathic membranous
United States. nephritis (IMN) association, Tissue Antigens 20 (3) (Sep. 1982) 227–228.
[45] C.D. Blosser, R. Ayalon, R. Nair, C. Thomas, L.H.J. Beck, Very early recurrence of [75] F.C. Berthoux, et al., Immunogenetics and immunopathology of human primary
anti-Phospholipase A2 receptor-positive membranous nephropathy after membranous glomerulonephritis: HLA-A, B, DR antigens; functional activity of
transplantation, Am. J. Transplant. Off. J. Am. Soc. Transplant. Am. Soc. splenic macrophage Fc-receptors and peripheral blood T-lymphocyte
Transplant Surg. 12 (6) (Jun. 2012) 1637–1642. subpopulations, Clin. Nephrol. 22 (1) (Jul. 1984) 15–20.
[46] H. Debiec, et al., Autoantibodies specific for the phospholipase A2 receptor in [76] R.W. Vaughan, A.G. Demaine, K.I. Welsh, A DQA1 allele is strongly associated
recurrent and De Novo membranous nephropathy, Am. J. Transplant. Off. J. Am. with idiopathic membranous nephropathy, Tissue Antigens 34 (5) (Nov. 1989)
Soc. Transplant. Am. Soc. Transplant Surg. 11 (10) (Oct. 2011) 2144–2152. 261–269.
[47] H. Debiec, et al., Recurrent membranous nephropathy in an allograft caused by [77] P.A. Dyer, C.D. Short, E.A. Clarke, N.P. Mallick, HLA antigen and gene
IgG3κ targeting the PLA2 receptor, J. Am. Soc. Nephrol. 23 (12) (Dec. 2012) polymorphisms and haplotypes established by family studies in membranous
1949–1954. nephropathy, Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. - Eur.
[48] L. East, C.M. Isacke, The mannose receptor family, Biochim. Biophys. Acta 1572 Ren. Assoc. 7 (Suppl. 1) (1992) 42–47.
(2–3) (Sep. 2002) 364–386. [78] S.H. Sacks, C. Warner, R.D. Campbell, I. Dunham, Molecular mapping of the HLA
[49] O. Llorca, Extended and bent conformations of the mannose receptor family, Cell. class II region in HLA-DR3 associated idiopathic membranous nephropathy,
Mol. Life Sci. 65 (9) (May 2008) 1302–1310. Kidney Int. Suppl. 39 (Jan. 1993) S13–S19.
[50] E. Zvaritch, G. Lambeau, M. Lazdunski, Endocytic properties of the M-type 180- [79] D. Chevrier, et al., Idiopathic and secondary membranous nephropathy and
kDa receptor for secretory phospholipases A2, J. Biol. Chem. 271 (1) (Jan. 1996) polymorphism at TAP1 and HLA-DMA loci, Tissue Antigens 50 (2) (Aug. 1997)
250–257. 164–169.
[51] A. Behnert, et al., An anti-phospholipase A2 receptor quantitative immunoassay [80] D. Kanigicherla, et al., Anti-PLA2R antibodies measured by ELISA predict long-
and epitope analysis in membranous nephropathy reveals different antigenic term outcome in a prevalent population of patients with idiopathic membranous
domains of the receptor, PLoS One 8 (4) (Apr. 2013), e61669. nephropathy, Kidney Int. 83 (5) (May 2013) 940–948.

11
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

[81] G. Bullich, et al., HLA-DQA1 and PLA2R1 polymorphisms and risk of idiopathic [111] H. Debiec, et al., Allo-immune membranous nephropathy and recombinant aryl
membranous nephropathy, Clin. J. Am. Soc. Nephrol. 9 (2) (Feb. 2014) 335–343. sulfatase replacement therapy: a need for tolerance induction therapy, J. Am. Soc.
[82] Y.-H. Liu, et al., Association of phospholipase A2 receptor 1 polymorphisms with Nephrol. 25 (4) (Apr. 2014) 675–680.
idiopathic membranous nephropathy in Chinese patients in Taiwan, J. Biomed. [112] S.C. Jordan, B. Buckingham, R. Sakai, D. Olson, Studies of immune-complex
Sci. 17 (1) (Oct. 2010) 81. glomerulonephritis mediated by human thyroglobulin, N. Engl. J. Med. 304 (20)
[83] S. Kim, et al., Single nucleotide polymorphisms in the phospholipase A2 receptor (May 1981) 1212–1215.
gene are associated with genetic susceptibility to idiopathic membranous [113] W.H. Hörl, D. Kerjaschki, Membranous glomerulonephritis (MGN), J. Nephrol. 13
nephropathy, Nephron. Clin. Pract. 117 (3) (2011) c253–c258. (4) (2000) 291–316.
[84] P. Ruggenenti, et al., Anti-phospholipase A2 receptor antibody titer predicts post- [114] R. Bhimma, H.M. Coovadia, Hepatitis B virus-associated nephropathy, Am. J.
rituximab outcome of membranous nephropathy, J. Am. Soc. Nephrol. 26 (10) Nephrol. 24 (2) (2004) 198–211.
(Oct. 2015) 2545–2558. [115] K. Nakahara, et al., Membranous nephropathy associated with chronic hepatitis B
[85] P. Sekula, et al., Genetic risk variants for membranous nephropathy: extension of occurring in a short period after acute hepatitis B virus infection, Intern. Med. 49
and association with other chronic kidney disease aetiologies, Nephrol. Dial. (5) (2010) 383–388.
Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. - Eur. Ren. Assoc. 32 (2) (Feb. [116] E. Guiard, et al., Patterns of noncryoglobulinemic glomerulonephritis with
2017) 325–332. monoclonal Ig deposits: correlation with IgG subclass and response to rituximab,
[86] W.-B. Le, et al., HLA-DRB1*15:01 and HLA-DRB3*02:02 in PLA2R-related Clin. J. Am. Soc. Nephrol. 6 (7) (Jul. 2011) 1609–1616.
membranous nephropathy, J. Am. Soc. Nephrol. 28 (5) (May 2017) 1642–1650. [117] S. de Seigneux, et al., Immunoglobulin deposition disease with a membranous
[87] Z. Cui, et al., MHC class II risk alleles and amino acid residues in idiopathic pattern and a circulating monoclonal immunoglobulin G with charge-dependent
membranous nephropathy, J. Am. Soc. Nephrol. 28 (5) (May 2017) 1651–1664. aggregation properties, Am. J. Kidney Dis. Off. J. Natl. Kidney Found. 56 (1) (Jul.
[88] M. Thiri, et al., High-density association mapping and interaction analysis of 2010) 117–121.
PLA2R1 and HLA regions with idiopathic membranous nephropathy in Japanese, [118] H.C. Morgan BP, Chapter 3 - Regulation in the activation pathways, in: Regulation
Sci. Rep. 6 (Dec. 2016) 38189. in the activation pathways, Academic Press, London, 1999, pp. 41–136. In:
[89] H. Debiec, P. Ronco, Immunopathogenesis of membranous nephropathy: an Complement regulatory proteins. Academic, San Diego, B. P. Morgan and C. L. B.
update, Semin. Immunopathol. 36 (4) (Jul. 2014) 381–397. T.-C. R. P. Harris, Eds.
[90] M.J.H. Coenen, et al., Phospholipase A2 receptor (PLA2R1) sequence variants in [119] A. Naik, S. Sharma, R.J. Quigg, Complement regulation in renal disease models,
idiopathic membranous nephropathy, J. Am. Soc. Nephrol. 24 (4) (Mar. 2013) Semin. Nephrol. 33 (6) (Nov. 2013) 575–585.
677–683. [120] C. Loirat, V. Frémeaux-Bacchi, Atypical hemolytic uremic syndrome, Orphanet J.
[91] C. Bantis, et al., Tumor necrosis factor-alpha gene G-308A polymorphism is a risk Rare Dis. 6 (Sep. 2011) 60.
factor for the development of membranous glomerulonephritis, Am. J. Nephrol. [121] A.E. Hughes, N. Orr, H. Esfandiary, M. Diaz-Torres, T. Goodship,
26 (1) (2006) 12–15. U. Chakravarthy, A common CFH haplotype, with deletion of CFHR1 and CFHR3,
[92] D. Thibaudin, et al., TNFA2 and d2 alleles of the tumor necrosis factor alpha gene is associated with lower risk of age-related macular degeneration, Nat. Genet. 38
polymorphism are associated with onset/occurrence of idiopathic membranous (10) (Oct. 2006) 1173–1177.
nephropathy, Kidney Int. 71 (5) (Mar. 2007) 431–437. [122] B. Gold, et al., Variation in factor B (BF) and complement component 2 (C2) genes
[93] E. Honkanen, E. von Willebrand, A.M. Teppo, T. Törnroth, C. Grönhagen-Riska, is associated with age-related macular degeneration, Nat. Genet. 38 (4) (Apr.
Adhesion molecules and urinary tumor necrosis factor-alpha in idiopathic 2006) 458–462.
membranous glomerulonephritis, Kidney Int. 53 (4) (Apr. 1998) 909–917. [123] A.T.M.G. Tiebosch, J. Wolters, P.F.M. Frederik, T.W. van der Wiel, E. Zeppenfeldt,
[94] T.J. Neale, et al., Tumor necrosis factor-alpha is expressed by glomerular visceral P.J. van Breda Vriesman, Epidemiology of idiopathic glomerular disease: a
epithelial cells in human membranous nephropathy, Am. J. Pathol. 146 (6) (Jun. prospective study, Kidney Int. 32 (1) (1987) 112–116.
1995) 1444–1454. [124] R.C. Aalberse, J. Schuurman, IgG4 breaking the rules, Immunology 105 (1) (Jan.
[95] S. Tabibzadeh, et al., TNF-α induces dyscohesion of epithelial cells. Association 2002) 9–19.
with disassembly of actin filaments, Endocrine 3 (8) (Aug. 1995) 549–556. [125] K. Lhotta, R. Würzner, P. König, Glomerular deposition of mannose-binding lectin
[96] T.B. Huber, T. Benzing, The slit diaphragm: a signaling platform to regulate in human glomerulonephritis, Nephrol. Dial. Transplant. Off. Publ. Eur. Dial.
podocyte function, Curr. Opin. Nephrol. Hypertens. 14 (3) (May 2005) 211–216. Transpl. Assoc. – Eur. Ren. Assoc. 14 (4) (Apr. 1999) 881–886.
[97] W.-Y. Lo, et al., Association between genetic polymorphisms of the NPHS1 gene [126] J.M. Hofstra, et al., Antiphospholipase A 2 Receptor Antibody Titer and Subclass
and membranous glomerulonephritis in the Taiwanese population, Clin. Chim. in Idiopathic Membranous Nephropathy, 2012, pp. 1735–1743.
Acta 411 (9–10) (May 2010) 714–718. [127] M. Nangaku, S.J. Shankland, W.G. Couser, Cellular response to injury in
[98] C.-H. Chen, et al., Impact of plasminogen activator inhibitor-1 gene membranous nephropathy, J. Am. Soc. Nephrol. 16 (5) (May 2005) 1195–1204.
polymorphisms on primary membranous nephropathy, Nephrol. Dial. Transplant. [128] P.N. Cunningham, R.J. Quigg, Contrasting roles of complement activation and its
Off. Publ. Eur. Dial. Transpl. Assoc. - Eur. Ren. Assoc. 23 (10) (Oct. 2008) regulation in membranous nephropathy, J. Am. Soc. Nephrol. 16 (5) (May 2005)
3166–3173. 1214–1222.
[99] Y. Luo, C. Wang, H. Tu, Impact of the 4G/5G polymorphism in the plasminogen [129] C.A. Tegla, et al., Membrane attack by complement: the assembly and biology of
activator inhibitor-1 gene on primary nephrotic syndrome, Mol. Med. Rep. 9 (3) terminal complement complexes, Immunol. Res. 51 (1) (Oct. 2011) 45–60.
(Mar. 2014) 894–898. [130] O. Bohana-Kashtan, L. Ziporen, N. Donin, S. Kraus, Z. Fishelson, Cell signals
[100] S.-Y. Chen, C.-H. Chen, Y.-C. Huang, H.-M. Chuang, M.-M. Lo, F.-J. Tsai, Effect of transduced by complement, Mol. Immunol. 41 (6–7) (Jul. 2004) 583–597.
IL-6 C-572G polymorphism on idiopathic membranous nephropathy risk in a Han [131] O. Moskovich, Z. Fishelson, Live cell imaging of outward and inward vesiculation
Chinese population, Ren. Fail. 32 (10) (2010) 1172–1176. induced by the complement c5b-9 complex, J. Biol. Chem. 282 (41) (Oct. 2007)
[101] S.-Y. Chen, et al., Association of STAT4 polymorphisms with susceptibility to 29977–29986.
primary membranous glomerulonephritis and renal failure, Clin. Chim. Acta 412 [132] S.P. Kon, et al., Urinary C5b-9 excretion and clinical course in idiopathic human
(21–22) (Oct. 2011) 1899–1904. membranous nephropathy, Kidney Int. 48 (6) (Dec. 1995) 1953–1958.
[102] Y.-T. Chen, et al., Toll-like receptor 9 SNPs are susceptible to the development [133] A.V. Cybulsky, Endoplasmic reticulum stress in proteinuric kidney disease,
and progression of membranous glomerulonephritis: 27 years follow-up in Kidney Int. 77 (3) (Feb. 2010) 187–193.
Taiwan, Ren. Fail. 35 (10) (2013) 1370–1375. [134] A.V. Cybulsky, The intersecting roles of endoplasmic reticulum stress, ubiquitin-
[103] H. Debiec, et al., Early-childhood membranous nephropathy due to cationic proteasome system, and autophagy in the pathogenesis of proteinuric kidney
bovine serum albumin, N. Engl. J. Med. 364 (22) (Jun. 2011) 2101–2110. disease, Kidney Int. 84 (1) (Jul. 2013) 25–33.
[104] S.K. Sathe, S.S. Teuber, K.H. Roux, Effects of food processing on the stability of [135] L. Wang, et al., Autophagy can repair endoplasmic reticulum stress damage of the
food allergens, Biotechnol. Adv. 23 (6) (Sep. 2005) 423–429. passive Heymann nephritis model as revealed by proteomics analysis, J. Proteome
[105] C. Sanchez, S. Frémont, Conséquences des traitements thermiques et de la 75 (13) (Jul. 2012) 3866–3876.
formulation sur la structure et l ’ allergénicité des protéines alimentaires [136] C. Meyer-Schwesinger, et al., A new role for the neuronal ubiquitin C-terminal
Consequences of heat treatment and processing of food on the structure and hydrolase-L1 (UCH-L1) in podocyte process formation and podocyte injury in
allergenicity of component proteins, vol. 43, 2003, pp. 13–20. human glomerulopathies, J. Pathol. 217 (3) (Feb. 2009) 452–464.
[106] M. Abdel-Hafez, M. Shimada, P.Y. Lee, R.J. Johnson, E.H. Garin, Idiopathic [137] C. Meyer-Schwesinger, et al., Ubiquitin C-terminal hydrolase-l1 activity induces
nephrotic syndrome and atopy: is there a common link? Am. J. kidney Dis. Off. J. polyubiquitin accumulation in podocytes and increases proteinuria in rat
Natl. Kidney Found. 54 (5) (Nov. 2009) 945–953. membranous nephropathy, Am. J. Pathol. 178 (5) (May 2011) 2044–2057.
[107] S.M. Richards, Immunologic considerations for enzyme replacement therapy in [138] C. Hetz, The unfolded protein response: controlling cell fate decisions under ER
the treatment of lysosomal storage disorders, Clin. Appl. Immunol. Rev. 2 (4) stress and beyond, Nat. Rev. Mol. Cell Biol. 13 (2) (Jan. 2012) 89–102.
(2002) 241–253. [139] A. Radice, et al., Diagnostic specificity of autoantibodies to M-type phospholipase
[108] D.A. Brooks, Immune response to enzyme replacement therapy in lysosomal A2 receptor (PLA2R) in differentiating idiopathic membranous nephropathy
storage disorder patients and animal models, Mol. Genet. Metab. 68 (2) (Oct. (IMN) from secondary forms and other glomerular diseases, J. Nephrol. 31 (2)
1999) 268–275. (2018) 271–278.
[109] E. Koren, L.A. Zuckerman, A.R. Mire-Sluis, Immune responses to therapeutic [140] T. Wiech, R.A.K. Stahl, E. Hoxha, Diagnostic role of renal biopsy in PLA(2)R1-
proteins in humans–clinical significance, assessment and prediction, Curr. Pharm. antibody-positive patients with nephrotic syndrome, Mod. Pathol. Off. J. United
Biotechnol. 3 (4) (Dec. 2002) 349–360. States Can. Acad. Pathol. Inc 32 (9) (Sep. 2019) 1320–1328.
[110] T.E. Hunley, et al., Nephrotic syndrome complicating alpha-glucosidase [141] P. Cravedi, M. Jarque, A. Angeletti, À. Favà, C. Cantarelli, O. Bestard, Immune-
replacement therapy for Pompe disease, Pediatrics 114 (4) (Oct. 2004) monitoring disease activity in primary membranous nephropathy, Front. Med. 6
e532–e535. (2019) 241.

12
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

[142] B. Huang, et al., Phospholipase A2 receptor antibody IgG4 subclass improves [170] C. Ponticelli, R.J. Glassock, Glomerular diseases: membranous nephropathy–a
sensitivity and specificity in the diagnosis of idiopathic membranous modern view, Clin. J. Am. Soc. Nephrol. 9 (3) (Mar. 2014) 609–616.
nephropathy, Kidney Blood Press. Res. 44 (4) (2019) 848–857. [171] E. Hoxha, et al., Spontaneous remission of proteinuria is a frequent event in
[143] C. VanBeek, M. Haas, Anti-PLA2R-associated membranous nephropathy: A review phospholipase A2 receptor antibody-negative patients with membranous
with emphasis on diagnostic testing methods, Clin. Nephrol. 84 (1) (2015) 1–9. nephropathy, Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. - Eur.
[144] M. Tampoia, F. Migliucci, C. Villani, L. Abbracciavento, M. Rossini, Definition of a Ren. Assoc. 30 (11) (Nov. 2015) 1862–1869.
new cut-off for the anti-phospholipase A2 receptor (PLA2R) autoantibody [172] R. Dikow, et al., Optimal blood pressure control versus additional
immunoassay in patients affected by idiopathic membranous nephropathy, immunosuppressive therapy in idiopathic membranous nephropathy – a
J. Nephrol. 31 (6) (2018) 899–905. retrospective analysis, Clin. Nephrol. 72 (5) (Nov. 2009) 366–372.
[145] J.M. Hofstra, J.F. Wetzels, Anti-PLA₂R antibodies in membranous nephropathy: [173] S. Troyanov, C.A. Wall, J.A. Miller, J.W. Scholey, D.C. Cattran, Idiopathic
ready for routine clinical practice? Neth. J. Med. 70 (3) (Apr. 2012) 109–113. membranous nephropathy: definition and relevance of a partial remission, Kidney
[146] E. Hoxha, et al., And its clinical usefulness in patients with membranous Int. 66 (3) (Sep. 2004) 1199–1205.
glomerulonephritis, no. June, 2011, pp. 2526–2532. [174] H. Shiiki, et al., Prognosis and risk factors for idiopathic membranous
[147] A. Pozdzik, I. Brochériou, C. David, F. Touzani, J.M. Goujon, K.M. Wissing, nephropathy with nephrotic syndrome in Japan, Kidney Int. 65 (4) (Apr. 2004)
Membranous nephropathy and anti-podocytes antibodies: implications for the 1400–1407.
diagnostic workup and disease management, Biomed. Res. Int. (2018) 2018. [175] J. Xu, X. Hu, J. Xie, N. Chen, Management of membranous nephropathy in Asia,
[148] A. Behnert, M. Schiffer, J. Müller-Deile, L.H. Beck, M. Mahler, M.J. Fritzler, Kidney Dis. 1 (2) (2015) 119–125.
Antiphospholipase A2 receptor autoantibodies: a comparison of three different [176] A.J. Branten, P.W. du Buf-Vereijken, M. Vervloet, J.F. Wetzels, Mycophenolate
immunoassays for the diagnosis of idiopathic membranous nephropathy, mofetil in idiopathic membranous nephropathy: a clinical trial with comparison
J Immunol Res (2014) 2014. to a historic control group treated with cyclophosphamide, Am. J. kidney Dis. Off.
[149] J.M. Hofstra, et al., Antiphospholipase A 2 receptor antibody titer and subclass in J. Natl. Kidney Found. 50 (2) (Aug. 2007) 248–256.
idiopathic membranous nephropathy, J. Am. Soc. Nephrol. 23 (10) (2012) [177] A. Howman, et al., Immunosuppression for progressive membranous
1735–1743. nephropathy: a UK randomised controlled trial, Lancet 381 (9868) (Mar. 2013)
[150] E. Hoxha, et al., Enhanced expression of the M-type phospholipase A2 receptor in 744–751.
glomeruli correlates with serum receptor antibodies in primary membranous [178] C. Ponticelli, et al., A randomized study comparing methylprednisolone plus
nephropathy, Kidney Int. 82 (7) (2012) 797–804. chlorambucil versus methylprednisolone plus cyclophosphamide in idiopathic
[151] S. Akiyama, M. Akiyama, E. Imai, T. Ozaki, S. Matsuo, S. Maruyama, Prevalence membranous nephropathy, J. Am. Soc. Nephrol. 9 (3) (Mar. 1998) 444–450.
of anti-phospholipase A2 receptor antibodies in Japanese patients with [179] D.C. Cattran, et al., A randomized controlled trial of prednisone in patients with
membranous nephropathy, Clin. Exp. Nephrol. 19 (4) (2015) 653–660. idiopathic membranous nephropathy, N. Engl. J. Med. 320 (4) (Jan. 1989)
[152] H. Saito, Y. Hamasaki, A. Tojo, Y. Shintani, A. Shimizu, M. Nangaku, 210–215.
Phospholipase A2 receptor positive membranous nephropathy long after living [180] D.C. Cattran, et al., Cyclosporine in patients with steroid-resistant membranous
donor kidney transplantation between identical twins, Nephrology (Carlton) 20 nephropathy: a randomized trial, Kidney Int. 59 (4) (Apr. 2001) 1484–1490.
(Suppl. 2) (Jul. 2015) 101–104. [181] J. Caro, et al., Predictors of response and relapse in patients with idiopathic
[153] A. Grupper, L.D. Cornell, F.C. Fervenza, L.H.J. Beck, E. Lorenz, F.G. Cosio, membranous nephropathy treated with tacrolimus, Nephrol. Dial. Transplant.
Recurrent membranous nephropathy after kidney transplantation: treatment and Off. Publ. Eur. Dial. Transpl. Assoc. – Eur. Ren. Assoc. 30 (3) (Mar. 2015)
long-term implications, Transplantation 100 (12) (Dec. 2016) 2710–2716. 467–474.
[154] N. Jourde-Chiche, et al., Novel ELISA for thrombospondin type 1 domain- [182] J. Radhakrishnan, D.C. Cattran, The KDIGO practice guideline on
containing 7A autoantibodies in membranous nephropathy, Kidney Int. 95 (3) glomerulonephritis: Reading between the (guide)lines-application to the
(2019). individual patient, Kidney Int. 82 (8) (2012) 840–856.
[155] C.P. Larsen, L.N. Cossey, L.H. Beck, THSD7A staining of membranous [183] Y. Chen, et al., Immunosuppressive treatment for idiopathic membranous
glomerulopathy in clinical practice reveals cases with dual autoantibody nephropathy in adults with nephrotic syndrome, Cochrane Database Syst. Rev.
positivity, Mod. Pathol. 29 (4) (Apr. 2016) 421–426. 2014 (10) (Oct. 2014) CD004293.
[156] H. Wakui, H. Imai, A. Komatsuda, A.B. Miura, Circulating antibodies against [184] M. Praga, V. Barrio, G.F. Juárez, J. Luño, F. the G. E. D. E. E. D. E. L. A. N. M.
alpha-enolase in patients with primary membranous nephropathy (MN), Clin. (Members of the G. listed at the end of the paper), Tacrolimus monotherapy in
Exp. Immunol. 118 (3) (Dec. 1999) 445–450. membranous nephropathy: A randomized controlled trial, Kidney Int. 71 (9) (May
[157] Y. Kimura, et al., Circulating antibodies to α-enolase and phospholipase A(2) 2007) 924–930.
receptor and composition of glomerular deposits in Japanese patients with [185] R. Ramachandran, et al., Tacrolimus combined with corticosteroids versus
primary or secondary membranous nephropathy, Clin. Exp. Nephrol. 21 (1) (Feb. Modified Ponticelli regimen in treatment of idiopathic membranous nephropathy:
2017) 117–126. Randomized control trial, Nephrology (Carlton) 21 (2) (Feb. 2016) 139–146.
[158] H. Ma, D.G. Sandor, L.H.J. Beck, The role of complement in membranous [186] S. Ren, et al., Comparative effectiveness and tolerance of immunosuppressive
nephropathy, Semin. Nephrol. 33 (6) (Nov. 2013) 531–542. treatments for idiopathic membranous nephropathy: A network meta-analysis,
[159] C. Kodner, Diagnosis and management of nephrotic syndrome in adults, Am. Fam. PLoS One 12 (9) (2017), e0184398.
Physician 93 (6) (Mar. 2016) 479–485. [187] P.W. Mathieson, Proteinuria and Immunity — an Overstated Relationship?
[160] E. Hoxha, et al., An immunofluorescence test for phospholipase-A2-receptor N. Engl. J. Med. 359 (23) (Dec. 2008) 2492–2494.
antibodies and its clinical usefulness in patients with membranous [188] A. Segarra, et al., Successful treatment of membranous glomerulonephritis with
glomerulonephritis, Nephrol. Dial. Transplant. 26 (8) (2011) 2526–2532. rituximab in calcineurin inhibitor-dependent patients, Clin. J. Am. Soc. Nephrol.
[161] M. Dauvergne, et al., Membranous nephropathy associated with immunological 4 (6) (Jun. 2009) 1083–1088.
disorder-related liver disease A retrospective study of 10 cases, Med. (United [189] A.S. Bomback, V.K. Derebail, J.G. McGregor, A.V. Kshirsagar, R.J. Falk, P.
States) 94 (30) (2015). H. Nachman, Rituximab therapy for membranous nephropathy: a systematic
[162] B. Svobodova, E. Honsova, P. Ronco, V. Tesar, H. Debiec, Kidney biopsy is a review, Clin. J. Am. Soc. Nephrol. 4 (4) (Apr. 2009) 734–744.
sensitive tool for retrospective diagnosis of PLA2R-related membranous [190] K. Dahan, et al., Rituximab for severe membranous nephropathy: a 6-month trial
nephropathy, Nephrol. Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. - Eur. with extended follow-up, J. Am. Soc. Nephrol. 28 (1) (Jan. 2017) 348–358.
Ren. Assoc. 28 (7) (Jul. 2013) 1839–1844. [191] F.C. Fervenza, et al., A multicenter randomized controlled trial of rituximab
[163] W. Qin, et al., Anti-phospholipase A2 receptor antibody in membranous versus cyclosporine in the treatment of idiopathic membranous nephropathy
nephropathy, 2011, pp. 1137–1143. (MENTOR), Nephron 130 (3) (2015) 159–168.
[164] C.C. Huang, et al., IgG subclass staining in renal biopsies with membranous [192] P. Cravedi, Rituximab in membranous nephropathy: not all studies are created
glomerulonephritis indicates subclass switch during disease progression, Mod. equal, Nephron 135 (1) (2017) 46–50.
Pathol. an Off. J. United States Can. Acad. Pathol. Inc 26 (6) (Jun. 2013) [193] A. Aleš Rigler, et al., Rituximab for the treatment of membranous nephropathy: a
799–805. single-center experience, Clin. Nephrol. 88 (13) (2017) 27–31.
[165] W. Na, K. Yi, Y.S. Song, M.H. Park, Dissecting the relationships of IgG subclasses [194] G. Moroni, et al., Low-dose rituximab is poorly effective in patients with primary
and complements in membranous lupus nephritis and idiopathic membranous membranous nephropathy, Nephrol. Dial. Transplant. Off. Publ. Eur. Dial.
nephropathy, PLoS One 12 (3) (Mar. 2017), e0174501. Transpl. Assoc. - Eur. Ren. Assoc. 32 (10) (Oct. 2017) 1691–1696.
[166] A. Torres, et al., Conservative versus immunosuppressive treatment of patients [195] P. Cravedi, G. Remuzzi, P. Ruggenenti, Rituximab in primary membranous
with idiopathic membranous nephropathy, Kidney Int. 61 (1) (Jan. 2002) nephropathy: first-line therapy, why not? Nephron. Clin. Pract. 128 (3–4) (2014)
219–227. 261–269.
[167] J.A.J.G. van den Brand, J.M. Hofstra, J.F.M. Wetzels, Low-molecular-weight [196] M. Waldman, L.H.J. Beck, M. Braun, K. Wilkins, J.E. Balow, H.A. Austin 3rd,
proteins as prognostic markers in idiopathic membranous nephropathy, Clin. J. Membranous nephropathy: pilot study of a novel regimen combining cyclosporine
Am. Soc. Nephrol. 6 (12) (Dec. 2011) 2846–2853. and Rituximab, Kidney Int. Rep. 1 (2) (Jul. 2016) 73–84.
[168] V. Jha, et al., A randomized, controlled trial of steroids and cyclophosphamide in [197] B. Sprangers, et al., Beneficial effect of rituximab in the treatment of recurrent
adults with nephrotic syndrome caused by idiopathic membranous nephropathy, idiopathic membranous nephropathy after kidney transplantation, Clin. J. Am.
J. Am. Soc. Nephrol. 18 (6) (Jun. 2007) 1899–1904. Soc. Nephrol. 5 (5) (May 2010) 790–797.
[169] Kidney Disease: Improving Global Outcomes (KDIGO) Glomerular Diseases Work [198] W. Cheungpasitporn, S.L. Kopecky, U. Specks, K. Bharucha, F.C. Fervenza, Non-
Group, KDIGO 2021 clinical practice guideline for the management of glomerular ischemic cardiomyopathy after rituximab treatment for membranous
diseases, Kidney Int. 100 (4S) (Oct 2021) S1–S276, https://doi.org/10.1016/j. nephropathy, J. Ren. Inj. Prev. 6 (1) (Nov. 2016) 18–25.
kint.2021.05.021. PMID: 34556256. [199] R.C. Radhakrishnan, G. Basu, R.E. George, H. Parmar, V. Tamilarasi, Rituximab-
induced urticarial dermatitis during the treatment of membranous nephropathy,

13
I. Nieto-Gañán et al. Clinical Immunology 237 (2022) 108976

Saudi J. kidney Dis. Transplant. an Off. Publ. Saudi Cent. Organ Transplantation, [205] C. Ponticelli, et al., A randomized pilot trial comparing methylprednisolone plus a
Saudi Arab. 28 (3) (2017) 657–660. cytotoxic agent versus synthetic adrenocorticotropic hormone in idiopathic
[200] M.J. Choi, et al., Mycophenolate mofetil treatment for primary glomerular membranous nephropathy, Am. J. kidney Dis. Off. J. Natl. Kidney Found. 47 (2)
diseases, Kidney Int. 61 (3) (Mar. 2002) 1098–1114. (Feb. 2006) 233–240.
[201] B. Dussol, et al., Mycophenolate mofetil monotherapy in membranous [206] A. Barbari, et al., Bortezomib as a novel approach to early recurrent membranous
nephropathy: a 1-year randomized controlled trial, Am. J. kidney Dis. Off. J. Natl. glomerulonephritis after kidney transplant refractory to combined conventional
Kidney Found. 52 (4) (Oct. 2008) 699–705. rituximab therapy, Exp. Clin. Transplant. Off. J. Middle East Soc. Organ
[202] G. Xie, et al., Immunosuppressive treatment for nephrotic idiopathic membranous Transplant. 15 (3) (Jun. 2017) 350–354.
nephropathy: a meta-analysis based on Chinese adults, PLoS One 7 (9) (Sep. [207] G. La Manna, et al., Membranous glomerulonephritis (MGN), ongoing studies,
2012), e44330. G. Ital. di Nefrol. organo Uff. della Soc. Ital. di Nefrol. 34 (5) (Sep. 2017)
[203] A. Segarra, et al., Efficacy and safety of ‘rescue therapy’ with mycophenolate 113–118.
mofetil in resistant primary glomerulonephritis–a multicenter study, Nephrol. [208] P. Ruggenenti, F.C. Fervenza, G. Remuzzi, Treatment of membranous
Dial. Transplant. Off. Publ. Eur. Dial. Transpl. Assoc. - Eur. Ren. Assoc. 22 (5) nephropathy: time for a paradigm shift, Nat. Rev. Nephrol. 13 (9) (Sep. 2017)
(May 2007) 1351–1360. 563–579.
[204] T. Rauen, A. Michaelis, J. Floege, P.R. Mertens, Case series of idiopathic
membranous nephropathy with long-term beneficial effects of ACTH peptide 1-
24, Clin. Nephrol. 71 (6) (Jun. 2009) 637–642.

14

You might also like