ﺑﺎﺯﮔﺸﺖ ﺑﻪ ﺻﻔﺤﻪ ﻗﺒﻠﯽ
خرید پکیج
تعداد آیتم قابل مشاهده باقیمانده : 3 مورد
نسخه الکترونیک
medimedia.ir

IgA nephropathy: Pathogenesis

IgA nephropathy: Pathogenesis
Literature review current through: Jan 2024.
This topic last updated: Nov 16, 2023.

INTRODUCTION — IgA nephropathy (IgAN) is the most common cause of primary glomerulonephritis throughout most resource-abundant settings [1].

The pathogenesis of IgAN will be reviewed here. Other aspects of IgAN are discussed separately:

(See "IgA nephropathy: Clinical features and diagnosis".)

(See "IgA nephropathy: Treatment and prognosis".)

(See "IgA nephropathy: Recurrence after transplantation".)

(See "IgA vasculitis (Henoch-Schönlein purpura): Kidney manifestations".)

THE FOUR-HIT HYPOTHESIS — IgAN is an autoimmune disease resulting from dysregulation of mucosal-type IgA immune responses. The autoantigens are a specific set of IgA1 O-glycoforms displaying reduced O-linked galactosylation of the IgA1 hinge region. Excess levels of these O-glycoforms in the circulation result in the generation of hinge glycan-specific immunoglobulin G (IgG) and IgA autoantibodies in susceptible individuals.

The pathogenesis of IgAN is framed by the widely accepted "four-hit hypothesis," which postulates that a sequence of four events must occur for clinically significant disease to develop (figure 1):

Increased presence in the circulation of IgA1 molecules with reduced O-galactosylation of the IgA1 hinge region (also referred to as galactose-deficient IgA1 [Gd-IgA1])

Production of IgG and IgA autoantibodies that recognize Gd-IgA1

Formation of circulating immune complexes containing IgG and IgA autoantibodies bound to Gd-IgA1

Deposition of these circulating immune complexes in the glomerular mesangium, triggering kidney injury

Hit 1: Production of poorly glycosylated IgA1 (Gd-IgA1) — The "first hit" in the pathogenesis of IgAN is an increase in serum levels of a pool of circulating IgA molecules with special characteristics that promote mesangial deposition (figure 1).

IgA1 O-glycosylation — In humans and higher primates, IgA exists as two isoforms, IgA1 and IgA2. These isoforms can exist as monomers or polymers and are present in mucosal tissues and the circulation. IgA1 has an extended hinge region between the first and second constant domains that has between three and six O-glycans chains attached to serine or threonine residues at specific positions (figure 2). O-glycosylation of the IgA1 hinge region involves the addition of N-acetylgalactosamine (GalNAc) to a serine/threonine residue, followed by the addition of a galactose moiety to GalNAc. Additional modifications can occur via sialylation of the GalNAc or galactose moieties. These steps produce a combination of different IgA1 O-glycoforms with varying degrees of galactosylation and sialylation.

In patients with IgAN, there is an increase in circulating IgA1 that is polymeric and lacks terminal galactose moieties or GalNAc and galactose in its hinge region. This form of IgA1 is termed "poorly galactosylated IgA1" or "galactose-deficient IgA1" (Gd-IgA1). This increase in poorly galactosylated IgA1 O-glycoforms is found in serum IgA, as well as in IgA1 eluted from isolated kidney tissue [2-4]. Similar findings have been reported in children with either IgAN or IgA vasculitis (Henoch-Schönlein purpura) nephritis [5]. Gd-IgA1 is an O-glycosylated form of IgA1 produced at mucosal surfaces, and its increased presence in the serum and mesangium likely reflects a subtle dysregulation of the mucosal immune system in IgAN.

Gd-IgA1 has increased in vitro affinity for the extracellular matrix components fibronectin and type IV collagen [6], which may promote mesangial deposition.

Genetic control of IgA1 O-galactosylation — Genetic and epigenetic factors may play an important role in determining O-galactosylation of the IgA1 hinge region. As an example, high serum levels of Gd-IgA1 have been identified in 47 and 25 percent of first-degree relatives of patients with familial and sporadic IgAN, respectively [7]. The heritability of Gd-IgA1 among familial IgAN cohorts has been estimated between 54 and 76 percent. In a study of healthy monozygotic and dizygotic twin pairs, the heritability of serum levels of Gd-IgA1 was found to be as high as 80 percent [8]. Heritability of serum IgA levels was only 46 percent, suggesting that O-galactosylation of IgA1 is independent of serum IgA level.

Higher serum levels of Gd-IgA1 have been associated with a polymorphism in the gene encoding core 1 synthase, glycoprotein-N-acetylgalactosamine 3-beta-galactosyltransferase (C1GALT1), one of the enzymes responsible for O-galactosylation of IgA1 [9,10]. In one study of White European and Chinese populations, this association was not confined to patients with IgAN but was also identified in healthy subjects and patients with membranous nephropathy [9]. Although IgAN is more common in Chinese compared with White populations, a lower frequency of the C1GALT1 risk haplotype was observed in the Chinese cohort, which corresponded with lower serum levels of Gd-IgA1 among Chinese patients with IgAN compared with levels among White European patients. These findings may suggest that factors other than IgA1 O-galactosylation are likely to play an important role in the pathogenesis of IgAN among different ethnic populations.

MicroRNAs (miRNAs) are small, noncoding, single-stranded RNAs that regulate gene expression at the posttranscriptional level. Expression of the miRNA, miR-148b, has been found to be increased in leukocytes from patients with IgAN, and overexpression of miR-148b correlated with decreased expression of C1GAL-T1 and increased secretion of Gd-IgA1 [11]. The binding site for miR-148b on the messenger RNA of C1GALT1 corresponded to the C1GALT1 polymorphism that was associated with IgAN in previous studies.

Source and regulation of Gd-IgA1 production in IgAN — The source of Gd-IgA1 production in patients with IgAN remains under investigation. A potential mucosal origin of Gd-IgA1 is suggested by the following observations:

IgAN can present with gross (visible) hematuria that is concurrent with mucosal inflammation and infection (eg, an upper respiratory tract infection).

Mesangial and circulating Gd-IgA1 in patients with IgAN is primarily polymeric, which is normally produced at mucosal surfaces rather than in systemic immune sites.

The pattern of IgA1 hinge region O-galactosylation of Gd-IgA1 closely resembles that seen in IgA1 of mucosal origin and IgA1 directed against mucosally encountered antigens.

However, mucosal polymeric IgA plasma cell numbers are normal or even reduced and polymeric IgA antibody levels are not elevated in mucosal secretions in IgAN [12,13]. By contrast, there is an increase in polymeric IgA1 plasma cells in the bone marrow of patients with IgAN, and these are believed to be derived from mucosally primed B cells [13,14]. Systemic antigen challenge results in increased titers of circulating polymeric IgA1 antibodies (displaying a mucosal phenotype), which are thought, largely, to be derived from these dislocated, mucosally primed plasma cells [15-18]. In patients with IgAN, circulating GdIgA1-expressing B cells were identified to preferentially express lambda light chains, which are also found in increased quantities in mesangial deposits in IgAN, and also express mucosal (upper respiratory tract and gut) homing receptors [19]. Thus, the overproduction of polymeric "mucosal-type" IgA1 in the serum is likely, in part, to stem from an aberrant mucosal immune response resulting in mistrafficking of "mucosal" plasma cells to systemic sites due to defective homing during mucosal immune responses [20].

The two mucosal-associated lymphoid tissues (MALT) most studied in IgAN are the nasal-associated lymphoid tissue (NALT) and gut-associated lymphoid tissue (GALT). Which MALT sites contribute most significantly to Gd-IgA1 production remain unclear and are likely to vary between individuals.

Nasal-associated lymphoid tissue – There is evidence that the tonsils may be a source of Gd-IgA1 [21,22]. A number of in vitro studies have demonstrated synthesis of polymeric Gd-IgA1 by tonsillar B cells, and there have been reports of a fall in serum levels of Gd-IgA1 following tonsillectomy [22]. Recognition of pathogen-associated molecular patterns (PAMPs) by toll-like receptors 7 and 9 (TLR7 and TLR9) has been shown to be important in promoting disease in a mouse model of IgAN and is present in the tonsillar tissue of patients with IgAN, supporting the involvement of NALT in the pathogenesis of IgAN [23,24]. However, there are differences in opinion as to the importance of the tonsils in IgAN, which could in part reflect variabilities in disease pathogenesis in different parts of the world. The data evaluating tonsillectomy as a therapy for IgAN are presented elsewhere. (See "IgA nephropathy: Treatment and prognosis", section on 'Adjunctive therapies'.)

Gut-associated lymphoid tissue – There has been increasing interest in a gut-kidney axis as being an important source of pathogenic IgA in IgAN, with important interplays between the GALT, the gut microbiome, and integrity of the intestinal barrier [25]. The gut produces the most IgA out of all mucosal sites [26]. A genome-wide association study (GWAS) highlighted a number of risk alleles at loci that are implicated in the maintenance of the intestinal barrier and regulation of the mucosal immune response to pathogens [27]. Patients with IgAN were found to have higher circulating levels of gut-homing regulatory B cells, memory B cells, and IgA-expressing memory B cells [28]. This area has become especially relevant with the development of GALT-directed therapies, such as targeted-release formulation budesonide. (See "IgA nephropathy: Treatment and prognosis", section on 'Other regimens'.)

Although the specific mechanisms that control IgA synthesis in the various immune sites are not well defined, systemic production of IgA appears to be under similar control mechanisms to those of IgG production [29], with major roles for B cell-activating factor (BAFF) and a proliferation-inducing ligand (APRIL). Both BAFF and APRIL are critical for mucosal B cell survival, maturation, proliferation, and IgA class switch recombination (and therefore in the production of Gd-IgA1). Serum BAFF and APRIL levels are increased in IgAN and correlate with disease severity [30,31]. Therapies that inhibit APRIL and/or BAFF are under investigation for the treatment of IgAN [32-34]. As with other immunoglobulin isotypes, type 2 T cell cytokines (interleukin [IL]-4, -5, and -6) also promote B cell class switching to IgA and subsequent proliferation and differentiation of IgA-producing cells. IgA production is also specifically and potently promoted by the cytokines IL-10 and transforming growth factor (TGF)–beta, which have suppressive effects on IgG production.

The control of mucosal IgA production is less well understood. BAFF and APRIL are highly expressed with the MALT. Furthermore, transgenic mice that overexpress BAFF demonstrated an IgAN-like disease with mesangial IgA deposition due to expansion of IgA-secreting plasma cells in the gut lamina propria [35]. Importantly, this was dependent upon the presence of gut commensal bacteria.

An imbalance of type 1 and type 2 T cell subsets has been proposed as an explanation for the dysregulated IgA responses seen in IgAN. In addition, there is provisional work suggesting that types 1 and 2 cytokines differentially affect IgA1 O-glycosylation [36].

Hit 2: Production of anti-Gd-IgA1 autoantibodies — The production of IgG and IgA autoantibodies that specifically recognize Gd-IgA1 represents the "second hit" of the four-hit hypothesis (figure 1).

Autoantibodies in IgAN recognize GalNac residues in the hinge region of Gd-IgA1 [37]. These Gd-IgA1-specific autoantibodies can be IgG or IgA, but IgG is the predominant isotype [38,39]. IgG autoantibodies against Gd-IgA1 contain a specific alanine to serine substitution in the heavy chain variable region that is critical for binding to Gd-IgA1 and appears to result from a somatic mutation that may be influenced by exposure to specific environmental antigens [37,40].

What triggers the production of anti-Gd-IgA1 autoantibodies in patients with IgAN is unclear, but genetic factors, in particular classical human leukocyte antigen (HLA) alleles within the major histocompatibility complex (MHC) locus, are likely to contribute. Healthy individuals can have equivalent levels of Gd-IgA1 to those with IgAN, without developing autoantibodies or kidney disease. (See 'Genetic susceptibility' below.)

Serum levels of Gd-IgA1-specific autoantibodies are variably reported to be associated with disease activity and progressive kidney disease in IgAN [4,38,41].

Hit 3: Formation of Gd-IgA1-containing circulating immune complexes — The formation of circulating immune complexes containing IgG or IgA autoantibodies bound to Gd-IgA1 represents the "third hit" of the four-hit hypothesis (figure 1).

The circulating immune complexes in patients with IgAN consist of IgG autoantibodies that are bound to polymeric Gd-IgA1 [42]. In addition to IgG, IgA, IgM, and complement pathway components can also be found in these circulating immune complexes [43]. Gd-IgA1 also has an increased tendency to self-aggregate, perhaps due to the different physiological environment of plasma compared with mucosal secretions.

Levels of circulating immune complexes in patients with IgAN have been shown to correlate with clinical and histologic activity, such as microscopic hematuria, episodes of macroscopic hematuria, and the percentage of glomeruli with crescents [44].

IgG antibodies against poorly O-galactosylated IgA1 contain a specific amino acid sequence, Y1CS3, in the heavy chain variable region that differs from a Y1CA3 sequence in similar isotype-matched healthy control patients. The S3 residue, which has been shown to be critical for binding to poorly galactosylated IgA1, is not observed in germline DNA and appears to be the result of a somatic mutation that may be influenced by exposure to specific environmental antigens [37,40].

Hit 4: Deposition of circulating immune complexes and kidney injury — Gd-IgA1-containing immune complexes are more likely to deposit in the mesangium [6,42]. Mesangial deposition of circulating immune complexes containing Gd-IgA1 and Gd-IgA1-specific IgG autoantibodies and activation of mesangial cells to induce kidney injury represent the "fourth hit" of the four-hit hypothesis (figure 1).

While IgG and complement components are often codeposited, IgA may be detected alone. In these cases, IgG may be deposited in small amounts that are undetectable by standard immunofluorescence techniques but may be observed with confocal microscopy [45].

Impaired IgA clearance — Impaired systemic clearance of IgA promotes IgA deposition in the mesangium. Persistent mesangial IgA accumulation occurs by one or both of two mechanisms: The rate of IgA deposition exceeds the mesangial clearance capacity, or the deposited IgA is resistant to mesangial clearance.

Systemic clearance – Alterations in systemic IgA and IgA–immune complex clearance mechanisms will facilitate their persistence in the serum. The liver plays an important role in IgA clearance from the circulation, and radiolabeled IgA clearance studies suggest reduced hepatic clearance in IgAN [46]. Reduced galactosylation of the IgA1 hinge region may affect its uptake by the asialoglycoprotein receptor on hepatocytes [47]. A second route of IgA clearance is through CD89, a fragment crystallizable (Fc) receptor for IgA expressed by myeloid and Kupffer cells [48]. IgAN is associated with downregulated CD89 expression on circulating myeloid cells, resulting in reduced clearance [49]. It has been proposed that this downregulation is a direct consequence of binding of Gd-IgA1-containing immune complexes with CD89 [49-51].

Mesangial clearance – Mesangial IgA deposition is not always associated with the development of glomerular inflammation. Furthermore, mesangial IgA deposition may be reversible, as suggested by the following observations:

Sequential biopsy studies of patients who underwent clinical remission were accompanied by disappearance of IgA deposits [52].

Mesangial IgA deposits disappear when kidneys with IgAN are inadvertently transplanted into recipients who originally did not have IgAN [53].

A proposed pathway for mesangial clearance is through mesangial cell receptor–mediated endocytosis and catabolism of IgA deposits. A number of candidate receptors on the mesangial cell have been described [54-60], including the transferrin receptor (CD71), which binds IgA and polymeric Gd-IgA1 [59-61].

It is possible that impaired binding of IgA to mesangial cell receptor(s) could lead to defective mesangial IgA clearance and thereby contribute to IgA accumulation and the development of glomerular injury, in a positive feedback loop manner.

IgA1-induced activation of mesangial cells — Polymeric IgA1 elicits a phenotypic transformation in mesangial cells in vitro, with mesangial cell proliferation and secretion of extracellular matrix components [20]. Spleen tyrosine kinase (Syk) is expressed by mesangial cells, and, in vitro, Syk inhibition reduced the proliferative and proinflammatory effects of IgA immune complexes on mesangial cells [62]. Glomerular phospho-Syk expression was shown to be increased in IgAN.

There is increased expression of TGF-beta and components of the renin-angiotensin system in IgAN [63-66]. In addition, polymeric IgA appears to stimulate the production of a variety of proinflammatory and profibrotic molecules, such as IL-6 [20,67,68]. Expression of platelet-derived growth factor (PDGF) B and D chains, proteins known to be important in the pathogenesis of mesangioproliferative glomerulonephritis, are upregulated in glomeruli in IgAN and have been shown in mouse models to induce marked mesangial cell proliferation [69,70].

The production of inflammatory mediators by mesangial cells and podocytes in response to IgA-containing immune complex deposition results in the recruitment of inflammatory cells, predominantly macrophages. Endocapillary hypercellularity, observed as a marker of worse prognosis as part of the Oxford classification of IgAN, correlates with glomerular CD68 count, a marker of macrophages [71]. In addition, tubulointerstitial macrophage infiltration is a driver for tubulointerstitial inflammation and fibrosis in IgAN [72].

Codeposition of IgG may synergistically contribute to the development of a proinflammatory phenotype in mesangial cells, thereby influencing the degree of glomerular injury and clinical outcome [45,73-76]. (See "IgA nephropathy: Clinical features and diagnosis", section on 'Pathology'.)

Mesangial cell–podocyte crosstalk — Several studies support the influence of mesangial cell–derived soluble mediators (such as tumor necrosis factor [TNF]-alpha, TGF-beta, and platelet-activating factor) on podocyte phenotype (including expression of nephrin, ezrin, and podocin and TNF-alpha secretion) in IgAN [77-81]. A progressive loss of podocyte markers has been shown to occur early at sites of capsular adhesions and in capillary loops [82]. IgA-induced mesangial cell activation may influence not only local changes within the mesangium but also glomerulotubular communication and the development of interstitial damage in IgAN through alterations in podocyte function.

The loss of podocyte markers [82] and the early development of glomerular capsular adhesions [83] suggest the possibility that podocyte injury occurs in IgAN and there may be a podocytopathic variant of IgAN in which podocyte injury is the principal feature, possibly due to mesangial cell mediator release or even direct interaction between IgA immune complexes and podocytes [84,85].

Complement activation — Local complement activation appears to influence the extent of glomerular injury. C3 is codeposited with IgA in over 90 percent of patients with IgAN. Both the alternative and lectin pathways may be activated, leading to generation of the anaphylatoxins C3a and C5a and the membrane attack complex C5b-9, with subsequent promotion of inflammatory mediator and matrix protein production by mesangial cells [86-89]. (See "IgA nephropathy: Clinical features and diagnosis", section on 'Pathology'.)

Lectin pathway activation occurs in a subset of patients and is associated with worse kidney injury, highlighting the heterogeneous nature of this condition. In a biopsy series, one-fourth of patients displayed evidence of lectin pathway activation (as determined by glomerular deposition of mannose-binding lectin [MBL], L-ficolin, MBL-associated serine protease [MASP], and C4d), and these patients had increased proteinuria and kidney damage compared with those with no lectin pathway involvement [88]. In another study, approximately one-third of patients had evidence of lectin pathway activation (mesangial C4d deposition in the absence of the classical pathway component C1q), and these patients had a marked reduction in 10-year kidney survival [90]. MBL is able to bind to polymeric IgA, but not monomeric IgA, and its binding results in activation of C3 and C4 [91].

Other studies suggest that factor H, a regulator of the alternative pathway, and the complement factor H–related (CFHR) proteins play an important role in IgAN. Two GWAS identified that a single nucleotide polymorphism (SNP) in the complement factor H (CFH)/CFHR locus, resulting in deletion of CFHR-3 and CFHR-1, protects against the risk of developing IgAN [92,93]. CFHR-3 and CFHR-1 compete with the binding of factor H to C3, and their deletion results in uninhibited factor H-C3 binding and downregulation of the alternative pathway; conversely, an increase in their levels leads to increased alternative pathway activity. CFHR-1 levels have been shown to be increased in two cohorts of IgAN patients and associated with progressive disease [94,95].

There is also evidence for systemic activation of complement in a subpopulation of patients with IgAN, with elevated levels of split products of activated C3 reported [96].

C3 and MBL are synthesized locally by mesangial cells and podocytes [97]; it is possible that the binding of polymeric IgA to mesangial cells incites local complement activation independent of systemic complement activity. The contribution of this in situ complement synthesis and activation to progressive glomerular injury is unknown. (See "Mechanisms of immune injury of the glomerulus".)

The increasing ability to perform mass spectrometry–based proteomics in glomerular disease may be an interesting approach to further define components, including complement, within immune deposits and to delineate the intracellular signaling pathways involved in the pathogenesis of IgAN [98].

Downstream pathways of injury and fibrosis — There is clear evidence of early activation of the renin-angiotensin system in IgAN [64,99]. Use of angiotensin-converting enzyme (ACE) inhibitors or angiotensin receptor blockers (ARBs) is one of the cornerstones of management. (See "IgA nephropathy: Treatment and prognosis", section on 'Angiotensin inhibition'.)

There has been increased interest in the role of endothelin (ET)-1 in IgAN. ET-1 is a growth factor that acts via its two receptors, ETA and ETB receptor. It may have several deleterious effects in the kidney, including vasoconstriction, mesangial cell proliferation, podocyte disruption, production of extracellular matrix, inflammation, and fibrosis [100]. Increased ET-1 and ETB receptor staining was observed in patients with IgAN and significant proteinuria [101]. A specific ETA receptor antagonist had a protective effect against the development of histopathologic lesions and proteinuria in the ddY mouse model of IgAN [102]. Endothelin receptor antagonism is being evaluated in several clinical trials in IgAN. The dual endothelin angiotensin receptor antagonist sparsentan received conditional approval from the US Food and Drug Administration (FDA) for reduction of proteinuria in patients with IgAN at risk of rapid disease progression. (See "IgA nephropathy: Treatment and prognosis", section on 'Dual endothelin angiotensin receptor antagonists'.)

Pathways common to other forms of chronic kidney disease (CKD), such as those involving the mineralocorticoid receptor system, TGF-beta, and Wnt/beta-catenin signaling, are likely to play a role in promoting kidney fibrosis in progressive IgAN. Results are awaited from the Finerenone in Non-Diabetic CKD (FIND-CKD) trial, which will assess the efficacy and safety of finerenone, a nonsteroidal mineralocorticoid receptor antagonist, in addition to standard of care in delaying progression of nondiabetic CKD, and it is likely that a large cohort of patients with IgAN will be enrolled into this trial.

ENVIRONMENTAL FACTORS

Infections — The provocation of gross (visible) hematuria by mucosal infection in patients with IgAN has led to the view that mesangial IgA deposition may be triggered by infection (figure 1). Cytomegalovirus, Haemophilus parainfluenzae, Staphylococcus aureus, and toxoplasmosis have been implicated [103-109]. During the coronavirus 2019 (COVID-19) pandemic, flares of IgAN were reported with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection or vaccination, further supporting a link between mucosal infection and inflammation and IgA deposition in susceptible individuals [110].

As an example, in a clinical study from Japan, kidney biopsy specimens from 116 patients with IgAN and 122 patients with other types of kidney disease were examined for the presence of S. aureus antigen in the glomeruli [104]. Although antigen was not detected in non-IgA disease, 68 percent of specimens from patients with IgAN had S. aureus cell envelope antigen localized with IgA antibody in the glomeruli. Another report from Japan suggested that, at least in that region, the inciting event may be pharyngeal colonization with H. parainfluenzae [106].

While a number of different exogenous antigens have been identified in glomerular deposits in IgAN, this observation is not consistently reported across all studies. As a result, it seems more probable that the development of IgAN is a consequence of a dysregulated mucosal IgA immune response rather than a specific IgA response to a specific antigen (infecting organism). Support for this view comes from genetic studies that identified susceptibility loci in genes that influence the immune response to intestinal pathogens [27].

Exogenous mucosal antigens — It has also been suggested that IgAN may result from a dysregulated mucosal IgA response to food antigens, in view of rare associations between IgAN and celiac and inflammatory bowel disease [25,111]. Mucosal IgA responses to several oral antigens, including food and vaccine antigens, are increased in patients with IgAN compared with responses in healthy individuals [112]. There is, however, no evidence for a relationship between any particular dietary antigen and risk of development of IgAN in humans. Furthermore, exclusion diets, outside of those for patients with proven celiac disease, do not improve outcomes in IgAN. Dietary restriction of gluten was studied in a clinical trial in patients with IgAN without celiac disease [113]. Although levels of proteinuria temporarily reduced (which could have been a nonspecific effect), there was no overall effect on clinical outcomes. (See "IgA nephropathy: Clinical features and diagnosis", section on 'Associated conditions'.)

Gut microbiome — There is great interest in the role of the mucosal microbiome in IgAN. The mucosal microbiome is in continuous communication with the mucosal-associated lymphoid tissues (MALT), modulating mucosal IgA responses to maintain a mucosal microenvironment that is amenable to the microbiota.

In vitro data have shown that pathogen-associated molecular patterns (PAMPs), by binding to pattern recognition receptors (PRRs) on mucosal lymphocytes, are capable of directly influencing not only IgA secretion by mucosal lymphocytes but also the expression of O-glycosyltransferases and the extent of IgA1 O-glycosylation. It is therefore possible that mucosal dysbiosis may be one of the factors responsible for dysregulated mucosal IgA synthesis and increases in circulating galactose-deficient IgA1 (Gd-IgA1) in IgAN. Indeed, the spontaneous development of IgA glomerular deposits in BAFF-overexpressing transgenic mice was dependent upon the presence of gut commensal bacteria [35]. In a separate rodent model of IgAN, mesangial IgA deposits were significantly reduced when mice were treated with broad spectrum antibiotics that rendered the gut sterile [114]. In a subsequent study using this model, fecal microbiota transplantation (FMT) was performed from healthy human subjects, those with nonprogressive IgAN, and those with progressive IgAN [115]. Microbiota from patients with progressive IgAN induced an increase in serum BAFF and Gd-IgA1 levels, while microbiota from healthy subjects led to a temporary reduction in albuminuria.

In a small cross-sectional study of the gut microbiome in IgAN, patients with progressive disease had a reduced gut microbial diversity compared with nonprogressors and healthy subjects [116]. Differences have also been reported in tonsillar crypt, salivary, and subgingival microbiomes in IgAN [117-119]. Case reports in which FMT has been performed in patients with IgAN have reported a clinical response, in terms of reductions in proteinuria [120,121].

GENETIC SUSCEPTIBILITY — Although IgAN is considered a sporadic disease, rare familial cases have been described in the United States (in eastern Kentucky) and elsewhere [122-128]. This may reflect an inherited susceptibility to develop mesangial glomerulonephritis. One study in Italy, for example, studied 269 asymptomatic, first-degree relatives of patients with IgAN [124]. Persistent microscopic hematuria was found in 42 patients (15.6 percent); biopsy confirmed the presence of IgAN in four of these patients.

Familial IgAN and susceptibility to sporadic IgAN are most likely due to variants in multiple loci involving both the major histocompatibility complex (MHC) and non-MHC susceptibility alleles [27,92,128-142]. As study sizes increase, more genetic risk alleles are being identified, and from available genome-wide association studies (GWAS), these risk alleles are most commonly associated with biological pathways and processes associated with mucosal immunity, maintenance of mucosal integrity, and pathways common to many other autoimmune diseases [27].

The frequency of subclinical IgAN in supposedly "normal" control populations means that all genetic studies thus far reported are examining clinical expression and not susceptibility to disease.

Genetic study of IgAN is further complicated by the uncertainty as to whether IgAN is truly a single entity and the presence of subclinical IgAN in apparently normal control populations. Taken together, available genetic studies suggest that IgAN is a genetically heterogeneous entity that does not have classic Mendelian inheritance attributable to a single gene locus but is a complex polygenic disease probably involving both MHC and non-MHC susceptibility alleles.

SUMMARY

Overview – IgA nephropathy (IgAN) is an autoimmune disease resulting from dysregulation of mucosal-type IgA immune responses. The autoantigens are a specific set of IgA1 O-glycoforms displaying reduced O-linked galactosylation of the IgA1 hinge region, referred to as galactose-deficient IgA1 (Gd-IgA1). Excess levels of these O-glycoforms in the circulation produced by B cells (or plasma cells) in mucosal and/or systemic sites (eg, bone marrow) lead to the generation of hinge glycan-specific immunoglobulin G (IgG) and IgA autoantibodies by B cells in susceptible individuals, and formation of immune complexes that subsequently deposit within the glomerular mesangium. (See 'The four-hit hypothesis' above.)

Four-Hit Hypothesis – The pathogenesis of IgAN is framed by the "four-hit hypothesis," which postulates that a sequence of four events must occur for clinically significant disease to develop (figure 1):

Hit 1 – Increased presence in the circulation of IgA1 molecules with reduced O-galactosylation of the IgA1 hinge region (Gd-IgA1). (See 'Hit 1: Production of poorly glycosylated IgA1 (Gd-IgA1)' above.)

Hit 2 – Production of IgG and IgA autoantibodies that recognize Gd-IgA1. (See 'Hit 2: Production of anti-Gd-IgA1 autoantibodies' above.)

Hit 3 – Formation of circulating immune complexes containing IgG and IgA autoantibodies bound to Gd-IgA1. (See 'Hit 3: Formation of Gd-IgA1-containing circulating immune complexes' above.)

Hit 4 – Deposition of these circulating immune complexes in the glomerular mesangium, triggering kidney injury. (See 'Hit 4: Deposition of circulating immune complexes and kidney injury' above.)

Environmental factors – Certain environmental factors may also contribute to the pathogenesis of IgAN:

Infections – The provocation of gross (visible) hematuria by mucosal infection in patients with IgAN has led to the view that mesangial IgA deposition may be triggered by infection. Cytomegalovirus, Haemophilus parainfluenzae, Staphylococcus aureus, and toxoplasmosis have been implicated. (See 'Infections' above.)

Exogenous mucosal antigens – IgAN may result from a dysregulated mucosal IgA response to food antigens (and possibly respiratory tract antigens), in view of rare associations between IgAN and celiac and inflammatory bowel disease. Mucosal IgA responses to several oral antigens, including those contained within foods and vaccines, are increased in patients with IgAN compared with responses in healthy individuals; however, there is no evidence for a relationship between any particular dietary antigen and risk of development of IgAN. (See 'Exogenous mucosal antigens' above.)

Gut microbiome – There is great interest in the role of the gut mucosal microbiome in IgAN. The gut microbiome is in continuous communication with the gut-associated lymphoid tissues (GALT), modulating mucosal IgA responses to maintain a mucosal microenvironment that is amenable to the microbiota. It is possible that gut mucosal dysbiosis may be one of the factors responsible for dysregulated mucosal IgA synthesis and increases in circulating Gd-IgA1 in IgAN. (See 'Gut microbiome' above.)

Genetic susceptibility – Although IgAN is considered a sporadic disease, rare familial cases have been described in the United States (in eastern Kentucky) and elsewhere. Available genetic studies suggest that IgAN is a genetically heterogeneous entity that does not have classic Mendelian inheritance attributable to a single gene locus but is a complex polygenic disease probably involving both major histocompatibility complex (MHC) and non-MHC susceptibility alleles. (See 'Genetic susceptibility' above.)

ACKNOWLEDGMENT — The editorial staff at UpToDate would like to acknowledge John Feehally, DM, FRCP, who contributed to an earlier version of this topic review.

  1. Wyatt RJ, Julian BA. IgA nephropathy. N Engl J Med 2013; 368:2402.
  2. Hiki Y, Odani H, Takahashi M, et al. Mass spectrometry proves under-O-glycosylation of glomerular IgA1 in IgA nephropathy. Kidney Int 2001; 59:1077.
  3. Allen AC, Bailey EM, Brenchley PE, et al. Mesangial IgA1 in IgA nephropathy exhibits aberrant O-glycosylation: observations in three patients. Kidney Int 2001; 60:969.
  4. Berthoux F, Suzuki H, Thibaudin L, et al. Autoantibodies targeting galactose-deficient IgA1 associate with progression of IgA nephropathy. J Am Soc Nephrol 2012; 23:1579.
  5. Lau KK, Wyatt RJ, Moldoveanu Z, et al. Serum levels of galactose-deficient IgA in children with IgA nephropathy and Henoch-Schönlein purpura. Pediatr Nephrol 2007; 22:2067.
  6. Kokubo T, Hiki Y, Iwase H, et al. Protective role of IgA1 glycans against IgA1 self-aggregation and adhesion to extracellular matrix proteins. J Am Soc Nephrol 1998; 9:2048.
  7. Gharavi AG, Moldoveanu Z, Wyatt RJ, et al. Aberrant IgA1 glycosylation is inherited in familial and sporadic IgA nephropathy. J Am Soc Nephrol 2008; 19:1008.
  8. Lomax-Browne HJ, Visconti A, Pusey CD, et al. IgA1 Glycosylation Is Heritable in Healthy Twins. J Am Soc Nephrol 2017; 28:64.
  9. Gale DP, Molyneux K, Wimbury D, et al. Galactosylation of IgA1 Is Associated with Common Variation in C1GALT1. J Am Soc Nephrol 2017; 28:2158.
  10. Kiryluk K, Li Y, Moldoveanu Z, et al. GWAS for serum galactose-deficient IgA1 implicates critical genes of the O-glycosylation pathway. PLoS Genet 2017; 13:e1006609.
  11. Serino G, Sallustio F, Cox SN, et al. Abnormal miR-148b expression promotes aberrant glycosylation of IgA1 in IgA nephropathy. J Am Soc Nephrol 2012; 23:814.
  12. Harper SJ, Pringle JH, Wicks AC, et al. Expression of J chain mRNA in duodenal IgA plasma cells in IgA nephropathy. Kidney Int 1994; 45:836.
  13. Westberg NG, Baklien K, Schmekel B, et al. Quantitation of immunoglobulin-producing cells in small intestinal mucosa of patients with IgA nephropathy. Clin Immunol Immunopathol 1983; 26:442.
  14. Harper SJ, Allen AC, Pringle JH, Feehally J. Increased dimeric IgA producing B cells in the bone marrow in IgA nephropathy determined by in situ hybridisation for J chain mRNA. J Clin Pathol 1996; 49:38.
  15. van den Wall Bake AW, Daha MR, Evers-Schouten J, van Es LA. Serum IgA and the production of IgA by peripheral blood and bone marrow lymphocytes in patients with primary IgA nephropathy: evidence for the bone marrow as the source of mesangial IgA. Am J Kidney Dis 1988; 12:410.
  16. Barratt J, Bailey EM, Buck KS, et al. Exaggerated systemic antibody response to mucosal Helicobacter pylori infection in IgA nephropathy. Am J Kidney Dis 1999; 33:1049.
  17. de Fijter JW, Eijgenraam JW, Braam CA, et al. Deficient IgA1 immune response to nasal cholera toxin subunit B in primary IgA nephropathy. Kidney Int 1996; 50:952.
  18. Layward L, Allen AC, Hattersley JM, et al. Response to mucosal antigen challenge in IgA nephropathy. Exp Nephrol 1995; 3:300.
  19. Zachova K, Jemelkova J, Kosztyu P, et al. Galactose-Deficient IgA1 B cells in the Circulation of IgA Nephropathy Patients Carry Preferentially Lambda Light Chains and Mucosal Homing Receptors. J Am Soc Nephrol 2022; 33:908.
  20. Barratt J, Feehally J, Smith AC. Pathogenesis of IgA nephropathy. Semin Nephrol 2004; 24:197.
  21. Béné MC, Faure GC, Hurault de Ligny B, de March AK. Clinical involvement of the tonsillar immune system in IgA nephropathy. Acta Otolaryngol Suppl 2004; :10.
  22. Xie Y, Chen X, Nishi S, et al. Relationship between tonsils and IgA nephropathy as well as indications of tonsillectomy. Kidney Int 2004; 65:1135.
  23. Lee M, Suzuki H, Ogiwara K, et al. The nucleotide-sensing Toll-Like Receptor 9/Toll-Like Receptor 7 system is a potential therapeutic target for IgA nephropathy. Kidney Int 2023; 104:943.
  24. Muto M, Manfroi B, Suzuki H, et al. Toll-Like Receptor 9 Stimulation Induces Aberrant Expression of a Proliferation-Inducing Ligand by Tonsillar Germinal Center B Cells in IgA Nephropathy. J Am Soc Nephrol 2017; 28:1227.
  25. Coppo R. The Gut-Renal Connection in IgA Nephropathy. Semin Nephrol 2018; 38:504.
  26. Barratt J, Rovin BH, Cattran D, et al. Why Target the Gut to Treat IgA Nephropathy? Kidney Int Rep 2020; 5:1620.
  27. Kiryluk K, Li Y, Scolari F, et al. Discovery of new risk loci for IgA nephropathy implicates genes involved in immunity against intestinal pathogens. Nat Genet 2014; 46:1187.
  28. Sallustio F, Curci C, Chaoul N, et al. High levels of gut-homing immunoglobulin A+ B lymphocytes support the pathogenic role of intestinal mucosal hyperresponsiveness in immunoglobulin A nephropathy patients. Nephrol Dial Transplant 2021; 36:1765.
  29. Roodnat JI, de Fijter JW, van Kooten C, et al. Decreased IgA1 response after primary oral immunization with live typhoid vaccine in primary IgA nephropathy. Nephrol Dial Transplant 1999; 14:353.
  30. Xin G, Shi W, Xu LX, et al. Serum BAFF is elevated in patients with IgA nephropathy and associated with clinical and histopathological features. J Nephrol 2013; 26:683.
  31. Zhai YL, Zhu L, Shi SF, et al. Increased APRIL Expression Induces IgA1 Aberrant Glycosylation in IgA Nephropathy. Medicine (Baltimore) 2016; 95:e3099.
  32. Zhang X, Wang Y, Yarbrough J, et al. Safety, Pharmacokinetics, and Pharmacodynamics of Subcutaneous Sibeprenlimab in Healthy Participants. Clin Pharmacol Drug Dev 2023; 12:1211.
  33. Cheung CK, Barratt J, Carroll K, et al. Targeting APRIL in the Treatment of IgA Nephropathy. Clin J Am Soc Nephrol 2023.
  34. Mathur M, Barratt J, Chacko B, et al. A Phase 2 Trial of Sibeprenlimab in Patients with IgA Nephropathy. N Engl J Med 2023.
  35. McCarthy DD, Kujawa J, Wilson C, et al. Mice overexpressing BAFF develop a commensal flora-dependent, IgA-associated nephropathy. J Clin Invest 2011; 121:3991.
  36. Yamada K, Kobayashi N, Ikeda T, et al. Down-regulation of core 1 beta1,3-galactosyltransferase and Cosmc by Th2 cytokine alters O-glycosylation of IgA1. Nephrol Dial Transplant 2010; 25:3890.
  37. Suzuki H, Fan R, Zhang Z, et al. Aberrantly glycosylated IgA1 in IgA nephropathy patients is recognized by IgG antibodies with restricted heterogeneity. J Clin Invest 2009; 119:1668.
  38. Yanagawa H, Suzuki H, Suzuki Y, et al. A panel of serum biomarkers differentiates IgA nephropathy from other renal diseases. PLoS One 2014; 9:e98081.
  39. Placzek WJ, Yanagawa H, Makita Y, et al. Serum galactose-deficient-IgA1 and IgG autoantibodies correlate in patients with IgA nephropathy. PLoS One 2018; 13:e0190967.
  40. Huang ZQ, Raska M, Stewart TJ, et al. Somatic Mutations Modulate Autoantibodies against Galactose-Deficient IgA1 in IgA Nephropathy. J Am Soc Nephrol 2016; 27:3278.
  41. Zhao N, Hou P, Lv J, et al. The level of galactose-deficient IgA1 in the sera of patients with IgA nephropathy is associated with disease progression. Kidney Int 2012; 82:790.
  42. Tomana M, Novak J, Julian BA, et al. Circulating immune complexes in IgA nephropathy consist of IgA1 with galactose-deficient hinge region and antiglycan antibodies. J Clin Invest 1999; 104:73.
  43. Nakamura I, Iwase H, Ohba Y, et al. Quantitative analysis of IgA1 binding protein prepared from human serum by hypoglycosylated IgA1/Sepharose affinity chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 2002; 776:101.
  44. Coppo R, Basolo B, Martina G, et al. Circulating immune complexes containing IgA, IgG and IgM in patients with primary IgA nephropathy and with Henoch-Schoenlein nephritis. Correlation with clinical and histologic signs of activity. Clin Nephrol 1982; 18:230.
  45. Rizk DV, Saha MK, Hall S, et al. Glomerular Immunodeposits of Patients with IgA Nephropathy Are Enriched for IgG Autoantibodies Specific for Galactose-Deficient IgA1. J Am Soc Nephrol 2019; 30:2017.
  46. Roccatello D, Picciotto G, Coppo R, et al. The fate of aggregated immunoglobulin A injected in IgA nephropathy patients and healthy controls. Am J Kidney Dis 1991; 18:20.
  47. Leung JC, Tang SC, Chan DT, et al. Increased sialylation of polymeric lambda-IgA1 in patients with IgA nephropathy. J Clin Lab Anal 2002; 16:11.
  48. Monteiro RC, Van De Winkel JG. IgA Fc receptors. Annu Rev Immunol 2003; 21:177.
  49. Grossetête B, Launay P, Lehuen A, et al. Down-regulation of Fc alpha receptors on blood cells of IgA nephropathy patients: evidence for a negative regulatory role of serum IgA. Kidney Int 1998; 53:1321.
  50. van Zandbergen G, van Kooten C, Mohamad NK, et al. Reduced binding of immunoglobulin A (IgA) from patients with primary IgA nephropathy to the myeloid IgA Fc-receptor, CD89. Nephrol Dial Transplant 1998; 13:3058.
  51. Robert T, Berthelot L, Cambier A, et al. Molecular Insights into the Pathogenesis of IgA Nephropathy. Trends Mol Med 2015; 21:762.
  52. Hotta O, Furuta T, Chiba S, et al. Regression of IgA nephropathy: a repeat biopsy study. Am J Kidney Dis 2002; 39:493.
  53. Cuevas X, Lloveras J, Mir M, et al. Disappearance of mesangial IgA deposits from the kidneys of two donors after transplantation. Transplant Proc 1987; 19:2208.
  54. Haddad E, Moura IC, Arcos-Fajardo M, et al. Enhanced expression of the CD71 mesangial IgA1 receptor in Berger disease and Henoch-Schönlein nephritis: association between CD71 expression and IgA deposits. J Am Soc Nephrol 2003; 14:327.
  55. Gómez-Guerrero C, Duque N, Egido J. Mesangial cells possess an asialoglycoprotein receptor with affinity for human immunoglobulin A. J Am Soc Nephrol 1998; 9:568.
  56. McDonald KJ, Cameron AJ, Allen JM, Jardine AG. Expression of Fc alpha/mu receptor by human mesangial cells: a candidate receptor for immune complex deposition in IgA nephropathy. Biochem Biophys Res Commun 2002; 290:438.
  57. Barratt J, Greer MR, Pawluczyk IZ, et al. Identification of a novel Fcalpha receptor expressed by human mesangial cells. Kidney Int 2000; 57:1936.
  58. Gómez-Guerrero C, López-Armada MJ, González E, Egido J. Soluble IgA and IgG aggregates are catabolized by cultured rat mesangial cells and induce production of TNF-alpha and IL-6, and proliferation. J Immunol 1994; 153:5247.
  59. Moura IC, Centelles MN, Arcos-Fajardo M, et al. Identification of the transferrin receptor as a novel immunoglobulin (Ig)A1 receptor and its enhanced expression on mesangial cells in IgA nephropathy. J Exp Med 2001; 194:417.
  60. Moura IC, Arcos-Fajardo M, Sadaka C, et al. Glycosylation and size of IgA1 are essential for interaction with mesangial transferrin receptor in IgA nephropathy. J Am Soc Nephrol 2004; 15:622.
  61. Moura IC, Arcos-Fajardo M, Gdoura A, et al. Engagement of transferrin receptor by polymeric IgA1: evidence for a positive feedback loop involving increased receptor expression and mesangial cell proliferation in IgA nephropathy. J Am Soc Nephrol 2005; 16:2667.
  62. Kim MJ, McDaid JP, McAdoo SP, et al. Spleen tyrosine kinase is important in the production of proinflammatory cytokines and cell proliferation in human mesangial cells following stimulation with IgA1 isolated from IgA nephropathy patients. J Immunol 2012; 189:3751.
  63. Lai KN, Tang SC, Guh JY, et al. Polymeric IgA1 from patients with IgA nephropathy upregulates transforming growth factor-beta synthesis and signal transduction in human mesangial cells via the renin-angiotensin system. J Am Soc Nephrol 2003; 14:3127.
  64. Del Prete D, Gambaro G, Lupo A, et al. Precocious activation of genes of the renin-angiotensin system and the fibrogenic cascade in IgA glomerulonephritis. Kidney Int 2003; 64:149.
  65. Lai KN, Chan LY, Tang SC, et al. Mesangial expression of angiotensin II receptor in IgA nephropathy and its regulation by polymeric IgA1. Kidney Int 2004; 66:1403.
  66. Miyake-Ogawa C, Miyazaki M, Abe K, et al. Tissue-specific expression of renin-angiotensin system components in IgA nephropathy. Am J Nephrol 2005; 25:1.
  67. van den Dobbelsteen ME, van der Woude FJ, Schroeijers WE, et al. Binding of dimeric and polymeric IgA to rat renal mesangial cells enhances the release of interleukin 6. Kidney Int 1994; 46:512.
  68. Horii Y, Iwano M, Hirata E, et al. Role of interleukin-6 in the progression of mesangial proliferative glomerulonephritis. Kidney Int Suppl 1993; 39:S71.
  69. Floege J, van Roeyen C, Boor P, Ostendorf T. The role of PDGF-D in mesangioproliferative glomerulonephritis. Contrib Nephrol 2007; 157:153.
  70. Terada Y, Yamada T, Nakashima O, et al. Expression of PDGF and PDGF receptor mRNA in glomeruli in IgA nephropathy. J Am Soc Nephrol 1997; 8:817.
  71. Soares MF, Genitsch V, Chakera A, et al. Relationship between renal CD68+ infiltrates and the Oxford Classification of IgA nephropathy. Histopathology 2019; 74:629.
  72. Pawluczyk IZA, Soares MSF, Barratt WA, et al. Macrophage interactions with collecting duct epithelial cells are capable of driving tubulointerstitial inflammation and fibrosis in immunoglobulin A nephropathy. Nephrol Dial Transplant 2020; 35:1865.
  73. Emancipator SN. IgA nephropathy: morphologic expression and pathogenesis. Am J Kidney Dis 1994; 23:451.
  74. Rauterberg EW, Lieberknecht HM, Wingen AM, Ritz E. Complement membrane attack (MAC) in idiopathic IgA-glomerulonephritis. Kidney Int 1987; 31:820.
  75. O'Donoghue DJ, Darvill A, Ballardie FW. Mesangial cell autoantigens in immunoglobulin A nephropathy and Henoch-Schönlein purpura. J Clin Invest 1991; 88:1522.
  76. Shin DH, Lim BJ, Han IM, et al. Glomerular IgG deposition predicts renal outcome in patients with IgA nephropathy. Mod Pathol 2016; 29:743.
  77. Lai KN, Leung JC, Chan LY, et al. Podocyte injury induced by mesangial-derived cytokines in IgA nephropathy. Nephrol Dial Transplant 2009; 24:62.
  78. Lai KN, Leung JC, Chan LY, et al. Activation of podocytes by mesangial-derived TNF-alpha: glomerulo-podocytic communication in IgA nephropathy. Am J Physiol Renal Physiol 2008; 294:F945.
  79. Ye ZC, Wang C, Tang Y, et al. Serum IgA1 from patients with IgA nephropathy up-regulates integrin-linked kinase synthesis and inhibits adhesive capacity in podocytes through indirect pathways. Clin Invest Med 2009; 32:E20.
  80. Wang C, Ye Z, Peng H, et al. Effect of aggregated immunoglobulin A1 from immunoglobulin A nephropathy patients on nephrin expression in podocytes. Nephrology (Carlton) 2009; 14:213.
  81. Coppo R, Fonsato V, Balegno S, et al. Aberrantly glycosylated IgA1 induces mesangial cells to produce platelet-activating factor that mediates nephrin loss in cultured podocytes. Kidney Int 2010; 77:417.
  82. Hill GS, Karoui KE, Karras A, et al. Focal segmental glomerulosclerosis plays a major role in the progression of IgA nephropathy. I. Immunohistochemical studies. Kidney Int 2011; 79:635.
  83. El Karoui K, Hill GS, Karras A, et al. Focal segmental glomerulosclerosis plays a major role in the progression of IgA nephropathy. II. Light microscopic and clinical studies. Kidney Int 2011; 79:643.
  84. Cook HT. Focal segmental glomerulosclerosis in IgA nephropathy: a result of primary podocyte injury? Kidney Int 2011; 79:581.
  85. Bellur SS, Lepeytre F, Vorobyeva O, et al. Evidence from the Oxford Classification cohort supports the clinical value of subclassification of focal segmental glomerulosclerosis in IgA nephropathy. Kidney Int 2017; 91:235.
  86. Maillard N, Wyatt RJ, Julian BA, et al. Current Understanding of the Role of Complement in IgA Nephropathy. J Am Soc Nephrol 2015; 26:1503.
  87. Hisano S, Matsushita M, Fujita T, et al. Mesangial IgA2 deposits and lectin pathway-mediated complement activation in IgA glomerulonephritis. Am J Kidney Dis 2001; 38:1082.
  88. Roos A, Rastaldi MP, Calvaresi N, et al. Glomerular activation of the lectin pathway of complement in IgA nephropathy is associated with more severe renal disease. J Am Soc Nephrol 2006; 17:1724.
  89. Matsuda M, Shikata K, Wada J, et al. Deposition of mannan binding protein and mannan binding protein-mediated complement activation in the glomeruli of patients with IgA nephropathy. Nephron 1998; 80:408.
  90. Espinosa M, Ortega R, Gómez-Carrasco JM, et al. Mesangial C4d deposition: a new prognostic factor in IgA nephropathy. Nephrol Dial Transplant 2009; 24:886.
  91. Roos A, Bouwman LH, van Gijlswijk-Janssen DJ, et al. Human IgA activates the complement system via the mannan-binding lectin pathway. J Immunol 2001; 167:2861.
  92. Gharavi AG, Kiryluk K, Choi M, et al. Genome-wide association study identifies susceptibility loci for IgA nephropathy. Nat Genet 2011; 43:321.
  93. Kiryluk K, Li Y, Sanna-Cherchi S, et al. Geographic differences in genetic susceptibility to IgA nephropathy: GWAS replication study and geospatial risk analysis. PLoS Genet 2012; 8:e1002765.
  94. Medjeral-Thomas NR, Lomax-Browne HJ, Beckwith H, et al. Circulating complement factor H-related proteins 1 and 5 correlate with disease activity in IgA nephropathy. Kidney Int 2017; 92:942.
  95. Tortajada A, Gutiérrez E, Goicoechea de Jorge E, et al. Elevated factor H-related protein 1 and factor H pathogenic variants decrease complement regulation in IgA nephropathy. Kidney Int 2017; 92:953.
  96. Seelen MA, Roos A, Daha MR. Role of complement in innate and autoimmunity. J Nephrol 2005; 18:642.
  97. Montinaro V, Gesualdo L, Ranieri E, et al. Renal cortical complement C3 gene expression in IgA nephropathy. J Am Soc Nephrol 1997; 8:415.
  98. Liu P, Lassén E, Nair V, et al. Transcriptomic and Proteomic Profiling Provides Insight into Mesangial Cell Function in IgA Nephropathy. J Am Soc Nephrol 2017; 28:2961.
  99. Nishiyama A, Konishi Y, Ohashi N, et al. Urinary angiotensinogen reflects the activity of intrarenal renin-angiotensin system in patients with IgA nephropathy. Nephrol Dial Transplant 2011; 26:170.
  100. Komers R, Plotkin H. Dual inhibition of renin-angiotensin-aldosterone system and endothelin-1 in treatment of chronic kidney disease. Am J Physiol Regul Integr Comp Physiol 2016; 310:R877.
  101. Lehrke I, Waldherr R, Ritz E, Wagner J. Renal endothelin-1 and endothelin receptor type B expression in glomerular diseases with proteinuria. J Am Soc Nephrol 2001; 12:2321.
  102. Nakamura T, Ebihara I, Fukui M, et al. Effect of a specific endothelin receptor A antagonist on glomerulonephritis of ddY mice with IgA nephropathy. Nephron 1996; 72:454.
  103. Sharmin S, Shimizu Y, Hagiwara M, et al. Staphylococcus aureus antigens induce IgA-type glomerulonephritis in Balb/c mice. J Nephrol 2004; 17:504.
  104. Koyama A, Sharmin S, Sakurai H, et al. Staphylococcus aureus cell envelope antigen is a new candidate for the induction of IgA nephropathy. Kidney Int 2004; 66:121.
  105. Nasr SH, Markowitz GS, Whelan JD, et al. IgA-dominant acute poststaphylococcal glomerulonephritis complicating diabetic nephropathy. Hum Pathol 2003; 34:1235.
  106. Suzuki S, Nakatomi Y, Sato H, et al. Haemophilus parainfluenzae antigen and antibody in renal biopsy samples and serum of patients with IgA nephropathy. Lancet 1994; 343:12.
  107. Waldo FB, Britt WJ, Tomana M, et al. Non-specific mesangial staining with antibodies against cytomegalovirus in immunoglobulin-A nephropathy. Lancet 1989; 1:129.
  108. Park JS, Song JH, Yang WS, et al. Cytomegalovirus is not specifically associated with immunoglobulin A nephropathy. J Am Soc Nephrol 1994; 4:1623.
  109. Satoskar AA, Nadasdy G, Plaza JA, et al. Staphylococcus infection-associated glomerulonephritis mimicking IgA nephropathy. Clin J Am Soc Nephrol 2006; 1:1179.
  110. Canney M, Atiquzzaman M, Cunningham AM, et al. A Population-Based Analysis of the Risk of Glomerular Disease Relapse after COVID-19 Vaccination. J Am Soc Nephrol 2022; 33:2247.
  111. Ambruzs JM, Walker PD, Larsen CP. The histopathologic spectrum of kidney biopsies in patients with inflammatory bowel disease. Clin J Am Soc Nephrol 2014; 9:265.
  112. Selvaskandan H, Barratt J, Cheung CK. Immunological drivers of IgA nephropathy: Exploring the mucosa-kidney link. Int J Immunogenet 2022; 49:8.
  113. Coppo R, Roccatello D, Amore A, et al. Effects of a gluten-free diet in primary IgA nephropathy. Clin Nephrol 1990; 33:72.
  114. Chemouny JM, Gleeson PJ, Abbad L, et al. Modulation of the microbiota by oral antibiotics treats immunoglobulin A nephropathy in humanized mice. Nephrol Dial Transplant 2019; 34:1135.
  115. Lauriero G, Abbad L, Vacca M, et al. Fecal Microbiota Transplantation Modulates Renal Phenotype in the Humanized Mouse Model of IgA Nephropathy. Front Immunol 2021; 12:694787.
  116. De Angelis M, Montemurno E, Piccolo M, et al. Microbiota and metabolome associated with immunoglobulin A nephropathy (IgAN). PLoS One 2014; 9:e99006.
  117. Watanabe H, Goto S, Mori H, et al. Comprehensive microbiome analysis of tonsillar crypts in IgA nephropathy. Nephrol Dial Transplant 2017; 32:2072.
  118. Piccolo M, De Angelis M, Lauriero G, et al. Salivary Microbiota Associated with Immunoglobulin A Nephropathy. Microb Ecol 2015; 70:557.
  119. Cao Y, Qiao M, Tian Z, et al. Comparative Analyses of Subgingival Microbiome in Chronic Periodontitis Patients with and Without IgA Nephropathy by High Throughput 16S rRNA Sequencing. Cell Physiol Biochem 2018; 47:774.
  120. Zhao J, Bai M, Yang X, et al. Alleviation of refractory IgA nephropathy by intensive fecal microbiota transplantation: the first case reports. Ren Fail 2021; 43:928.
  121. Zhi W, Song W, Abdi Saed Y, et al. Fecal Capsule as a Therapeutic Strategy in IgA Nephropathy: A Brief Report. Front Med (Lausanne) 2022; 9:914250.
  122. Frimat L, Kessler M. Controversies concerning the importance of genetic polymorphism in IgA nephropathy. Nephrol Dial Transplant 2002; 17:542.
  123. Julian BA, Quiggins PA, Thompson JS, et al. Familial IgA nephropathy. Evidence of an inherited mechanism of disease. N Engl J Med 1985; 312:202.
  124. Schena FP. Immunogenetic aspects of primary IgA nephropathy. Kidney Int 1995; 48:1998.
  125. Scolari F, Amoroso A, Savoldi S, et al. Familial clustering of IgA nephropathy: further evidence in an Italian population. Am J Kidney Dis 1999; 33:857.
  126. Hsu SI, Ramirez SB, Winn MP, et al. Evidence for genetic factors in the development and progression of IgA nephropathy. Kidney Int 2000; 57:1818.
  127. Schena FP, Cerullo G, Rossini M, et al. Increased risk of end-stage renal disease in familial IgA nephropathy. J Am Soc Nephrol 2002; 13:453.
  128. Gharavi AG, Yan Y, Scolari F, et al. IgA nephropathy, the most common cause of glomerulonephritis, is linked to 6q22-23. Nat Genet 2000; 26:354.
  129. Feehally J, Farrall M, Boland A, et al. HLA has strongest association with IgA nephropathy in genome-wide analysis. J Am Soc Nephrol 2010; 21:1791.
  130. Yu XQ, Li M, Zhang H, et al. A genome-wide association study in Han Chinese identifies multiple susceptibility loci for IgA nephropathy. Nat Genet 2011; 44:178.
  131. Bisceglia L, Cerullo G, Forabosco P, et al. Genetic heterogeneity in Italian families with IgA nephropathy: suggestive linkage for two novel IgA nephropathy loci. Am J Hum Genet 2006; 79:1130.
  132. Paterson AD, Liu XQ, Wang K, et al. Genome-wide linkage scan of a large family with IgA nephropathy localizes a novel susceptibility locus to chromosome 2q36. J Am Soc Nephrol 2007; 18:2408.
  133. Karnib HH, Sanna-Cherchi S, Zalloua PA, et al. Characterization of a large Lebanese family segregating IgA nephropathy. Nephrol Dial Transplant 2007; 22:772.
  134. Liu XQ, Paterson AD, He N, et al. IL5RA and TNFRSF6B gene variants are associated with sporadic IgA nephropathy. J Am Soc Nephrol 2008; 19:1025.
  135. Miyata T, Nangaku M, Suzuki D, et al. A mesangium-predominant gene, megsin, is a new serpin upregulated in IgA nephropathy. J Clin Invest 1998; 102:828.
  136. Li YJ, Du Y, Li CX, et al. Family-based association study showing that immunoglobulin A nephropathy is associated with the polymorphisms 2093C and 2180T in the 3' untranslated region of the Megsin gene. J Am Soc Nephrol 2004; 15:1739.
  137. Xia Y, Li Y, Du Y, et al. Association of MEGSIN 2093C-2180T haplotype at the 3' untranslated region with disease severity and progression of IgA nephropathy. Nephrol Dial Transplant 2006; 21:1570.
  138. Xia YF, Huang S, Li X, et al. A family-based association study of megsin A23167G polymorphism with susceptibility and progression of IgA nephropathy in a Chinese population. Clin Nephrol 2006; 65:153.
  139. Suzuki D, Miyata T, Nangaku M, et al. Expression of megsin mRNA, a novel mesangium-predominant gene, in the renal tissues of various glomerular diseases. J Am Soc Nephrol 1999; 10:2606.
  140. Hsu SI. Racial and genetic factors in IgA nephropathy. Semin Nephrol 2008; 28:48.
  141. Li GS, Zhang H, Lv JC, et al. Variants of C1GALT1 gene are associated with the genetic susceptibility to IgA nephropathy. Kidney Int 2007; 71:448.
  142. Pirulli D, Crovella S, Ulivi S, et al. Genetic variant of C1GalT1 contributes to the susceptibility to IgA nephropathy. J Nephrol 2009; 22:152.
Topic 3095 Version 34.0

References

آیا می خواهید مدیلیب را به صفحه اصلی خود اضافه کنید؟