Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

A repertoire of 124 potential autoantigens for autoimmune kidney diseases identified by dermatan sulfate affinity enrichment of kidney tissue proteins

  • Wei Zhang,

    Roles Data curation

    Affiliation Department of Gastroenterology, Affiliated Hospital of Guizhou Medical University, Guizhou, China

  • Jung-hyun Rho,

    Roles Methodology, Writing – review & editing

    Affiliation MP Biomedicals, Auckland, New Zealand

  • Michael W. Roehrl,

    Roles Data curation, Writing – review & editing

    Affiliation Curandis, Scarsdale, New York, United States of America

  • Michael H. Roehrl ,

    Roles Conceptualization, Investigation, Supervision, Writing – review & editing

    julia@curandis.com (JYW); roehrlm@mskcc.org (MHR)

    Affiliation Department of Pathology and Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York City, New York, United States of America

  • Julia Y. Wang

    Roles Conceptualization, Data curation, Formal analysis, Funding acquisition, Investigation, Methodology, Project administration, Resources, Supervision, Writing – original draft, Writing – review & editing

    julia@curandis.com (JYW); roehrlm@mskcc.org (MHR)

    Affiliation Curandis, Scarsdale, New York, United States of America

Abstract

Autoantigens are the molecular targets in autoimmune diseases. They are a cohort of seemingly unrelated self-molecules present in different parts of the body, yet they can trigger a similar chain of autoimmune responses such as autoantibody production. We previously reported that dermatan sulfate (DS) can bind self-molecules of dying cells to stimulate autoreactive CD5+ B cells to produce autoantibodies. The formation of autoantigen-DS complexes converts the normally non-antigenic self-molecules to none-self antigens, and thus DS-affinity represents a common underlying biochemical property for autoantigens. This study sought to apply this property to identify potential autoantigens in the kidney. Total proteins were extracted from mouse kidney tissues and loaded onto DS-Sepharose resins. Proteins without affinity were washed off the resins, whereas those with increasing DS-affinity were eluted with step gradients of increasing salt strength. Fractions with strong and moderate DS-affinity were sequenced by mass spectrometry and yielded 25 and 99 proteins, respectively. An extensive literature search was conducted to validate whether these had been previously reported as autoantigens. Of the 124 proteins, 79 were reported autoantigens, and 19 out of 25 of the strong-DS-binding ones were well-known autoantigens. Moreover, these proteins largely fell into the two most common autoantibody categories in autoimmune kidney diseases, including 40 ANA (anti-nuclear autoantibodies) and 25 GBM (glomerular basement membrane) autoantigens. In summary, this study compiles a large repertoire of potential autoantigens for autoimmune kidney diseases. This autoantigen-ome sheds light on the molecular etiology of autoimmunity and further supports our hypothesis DS-autoantigen complexes as a unifying principle of autoantigenicity.

Introduction

Autoimmune diseases are results of aberrant autoimmune responses. Immune defenses are normally generated against invading pathogens to protect the body. However, due to unclear mechanisms, the immune system sometimes deviates from its norm and produces undesirable autoimmune responses against the body itself. Autoimmune attacks can come from self-reactive cells and/or autoantibodies produced by autoreactive cells. Under normal circumstances, self-molecules in the body are non-antigenic, i.e., unable to trigger defensive immune reactions. It is puzzling how and why a self-molecule becomes an auto-antigenic trigger and/or target of autoimmune response. It is even more puzzling why, among the tens of thousands of molecules in the body, a cohort of only several hundred of seemingly unrelated molecules can trigger autoimmune reactions, e.g., production of autoantibodies.

In previous studies, we demonstrated that some molecules of dying cells have affinity for dermatan sulfate (DS), and that these molecules can form macromolecular complexes with DS to co-stimulate autoreactive CD5+ B cells to secrete autoantibodies [1]. Furthermore, we demonstrated that molecules with affinity for DS have a high propensity to be autoantigens (autoAgs) [2]. We thus proposed a unifying principle of autoantigenicity that explains how different molecules can become autoantigenic by means of a shared biochemical property. To gain further support, we have been testing whether autoantigens can be uncovered from specific tissues by enrichment with DS-affinity.

In this study, we applied the DS-affinity enrichment strategy to define the repertoire of possible autoantigens, i.e., the “autoantigen-ome”, in autoimmune kidney diseases. Although autoimmune attacks can happen in many parts of the body, they become especially serious if the kidneys are involved. Patients with autoimmune kidney diseases may develop glomerulonephritis. Glomeruli are made up of tiny blood vessels and help filter blood and remove excess fluids. When glomeruli are damaged, the kidneys stop working properly, which then leads to renal failure. Autoimmune renal diseases include lupus nephritis, Goodpasture syndrome or anti-GBM (glomerular basement membrane) disease, ANCA-associated vasculitis, and other rare diseases. They are largely defined by the kidney sub-location affected by the autoantibodies or immune cells. By identifying possible autoantigens in the kidney, we hope to gain a better understanding of the pathophysiology of these diseases.

Materials and methods

DS-Sepharose resin synthesis

DS-Sepharose resins were prepared by coupling dermatan sulfate (DS; Sigma-Aldrich) to EAH Sepharose 4B resins (GE Healthcare). Sepharose resins (20 mL) were washed with distilled water and 0.5 M NaCl and then mixed with 100 mg of DS dissolved in 10 mL of 0.1 M MES buffer (pH 5.0). N-ethyl-N-(3-dimethylaminopropyl) carbodiimide hydrochloride (Sigma-Aldrich) was added to a final concentration of 0.1 M. The reaction proceeded at 25°C for 24 hours with end-over-end rotation. After the first 60 minutes, the pH of the reaction mixture was readjusted to 5.0. After the coupling, the resins were washed three times, each time with a low pH buffer (0.1 M acetate, 0.5 M NaCl, pH 5.0) and a high pH buffer (0.1 M Tris, 0.5 M NaCl, pH 8.0). The washed DS-Sepharose resins were suspended in a 10 mM phosphate buffer (pH 7.4) and packed into a C16/20 column (GE Healthcare). The column was equilibrated with 10 mM phosphate buffer before use.

Kidney protein extraction

Kidneys were obtained from 5-month-old BALB/cJ mice (Jackson Laboratory). A total of 20 mice were killed with CO2, their blood was removed through heart puncture, and their kidneys were collected immediately. The kidneys were cleaned by rinsing with phosphate buffered saline (PBS, pH 7.2) twice and then stored at 4°C for 1 hour, -20°C for 2 hours, and then -80°C until further processing. Thawed kidneys were cut to small pieces and pressed through a cell strainer (Fisher Scientific). To remove red blood cells (RBC), kidney tissue was mixed with 10 mL of RBC lysis buffer for 10 seconds. After centrifugation for 5 minutes, the supernatant was discarded. The tissue was mixed with 40 mL of RIPA lysis buffer (Sigma-Aldrich) and 4 tablets of protease inhibitor (complete protease inhibitor cocktail, Sigma-Aldrich). The tissue mixture was sonicated for 10 minutes, or until all tissue pieces appeared dissolved. The mixture was centrifuged at 13,300 rpm for 20 minutes, and the supernatant that contained all the soluble kidney proteins was collected. Protein concentration was measured by the RC DC protein assay (Bio-Rad).

DS-affinity fractionation

Fractionation of kidney proteins was carried out by FPLC using a Biologic Duo-Flow System (Bio-Rad). Kidney proteins were loaded into the DS-Sepharose column in 10 mM phosphate buffer at a rate of 1 mL/min. The column was washed with 20 mL of 10 mM phosphate buffer and then 40 mL of 10 mM phosphate with 0.2 M NaCl to remove unbound proteins. Proteins bound to DS resins with moderate-to-strong affinity were eluted with a step-gradient of 0.4 M, 0.6 M, and 1.0 M NaCl in a 10 mM phosphate buffer, with each step being 40 mL. Elution was monitored by UV and conductivity detectors. All bound fractions were collected. Fractions were concentrated and desalted in Vivaspin centrifugal concentrators (MWCO 10 kDa, Sigma-Aldrich). Concentrated proteins were reconstituted in a 10 mM phosphate buffer for further analysis.

Protein sequencing by mass spectrometry

Fractionated proteins with different affinity to DS were separated on 1D SDS PAGE in 4–12% NuPAGE Novex Bis-Tris gels (Invitrogen). Based on protein band intensity, the protein lane containing proteins eluting at 0.4 M NaCl was cut into three sections, containing top, middle, and bottom bands. The lanes containing proteins eluting at 0.6 M and 1.0 M NaCl were each cut into two sections, containing top and bottom bands, respectively. Gel sections were transferred into 1-mL tubes, cut into tiny pieces, dehydrated with acetonitrile, and then dried in a speed-vac. Proteins in gel pieces were then rehydrated in 50 mM NH4HCO3 and digested with 12.5 ng/μL modified sequencing-grade trypsin (Promega) at 4°C overnight.

Mass spectrometric sequencing was performed at the Taplin Biological Mass Spectrometry Facility (Harvard Medical School, Boston, USA). Tryptic peptides were separated on a nano-scale C18 HPLC capillary column and analyzed after electrospray ionization in an LTQ linear ion-trap mass spectrometer (Thermo Fisher Scientific). Peptide sequences and protein identities were assigned by matching protein or translated nucleotide databases with the measured fragmentation pattern using Sequest software. Peptides were required to be fully tryptic peptides with XCorr values of at least 1.5 (+1 ion), 1.5 (+2 ion), or 3.0 (+3 ion). All data were manually inspected. Only proteins with at least two peptide matches were considered confidently identified.

Results and discussion

Fractionation of kidney proteins by DS affinity

Proteins extracted from mouse kidneys were initially separated into fractions according to their strength of binding to DS. This was carried out by loading the kidney proteins onto DS-Sepharose columns to allow binding to take place. Proteins that did not bind to DS resins were washed off the column first with the loading buffer (10 mM phosphate, pH 7.4) and then with 0.2 M NaCl. Afterwards, proteins that remained bound to DS were sequentially eluted from the column with 0.4 M, 0.6 M, and 1.0 M NaCl in 10mM phosphate (pH 7.4). Elution was monitored for presence of proteins by 280 nm absorbance, and chromatographic fractions that contained proteins at each salt strength were pooled, desalted, and concentrated. Final protein content and size distribution of each pool were assessed with 1D SDS PAGE gels.

The majority of kidney proteins did not bind to DS, as most proteins flowed through or eluted at the 0.2 M NaCl washing steps. As the NaCl step gradient elution proceeded, the amount of proteins eluting at increasing ionic strength became smaller and smaller. The flow-through and non-DS-binding proteins were not further analyzed. Proteins eluting at 0.4 M, 0.6 M, and 1.0 M NaCl were sequenced by LC-MS/MS, yielding 124 proteins (Table 1).

Proteins eluting at 1.0 M NaCl ionic strength were considered as having strong-DS affinity, whereas those eluteing at 0.6 M and 0.4 M were considered as having moderate and weak DS-affinity, respectively. Theoretical isoelectric points of all proteins were calculated using the pI compute tool of the ExPASy portal of the Swiss Institute of Bioinformatics (web.expasy.org/compute_pi/). These proteins span a wide range of pI values, ranging from very basic to acidic. Despite the fact that DS is acidic, there is no clear correlation between DS affinity strength and calculated pI values of these proteins.

Proteins with strong DS affinity

Proteins eluting off the DS-Sepharose column at 1.0 M NaCl were considered as having strong DS affinity. From the 1.0 M elution, 25 proteins were identified by MS sequencing (Table 1). Among these 25 proteins, 13 were DNA-/RNA-associated, 5 were related to cytoskeleton/extracellular matrix, 5 were related to cell stress and/or apoptosis, and 2 were miscellaneous. Literature searches revealed that at least 18 (72%) of these proteins have previously been identified as target autoantigens for autoantibodies.

Histones, the proteins that organize DNA into nucleosomes, are the most prominent autoantigens identified with strong-DS affinity. Particularly, H4, two isoforms of H2A, and two isoforms of H2B were found in the 1.0 M elution. Two isoforms of H1 were also identified, but in the weak DS affinity fraction. Top1 (DNA topoisomerase 1), an enzyme that controls and alters the topology of DNA during transcription, was also identified by strong DS affinity. All six of these DNA-binding proteins are known autoantigens (see references in Table 1). In particular, antibodies to histones and nucleosomes are central in the pathogenesis of lupus nephritis.

The RNA-associated proteins identified in the strong DS affinity fraction are RBM8a (RNA-binding protein 8A), Sf3b3 (splicing factor 3B subunit 3), Prpf19 (pre-mRNA processing factor 19), Rmpl2 (39S ribosomal protein L2), Rpl18 (60S ribosomal protein L18), Mrps27 (28S ribosomal protein S27), and ELAV-like protein 1 (HuR). Elval1, also known as Hu antigen R, binds to and stabilizes mRNAs. HuR autoantibodies are a serologic hallmark of paraneoplastic encephalomyelitis [20]. Ribosomal P proteins are classical autoantigens, and autoantibodies to various other ribosomal proteins (such as L2, L5, L7, L12, L23a, S13, and S19) are reported but the heterogeneity awaits further characterization [21]. The specific L18 and Mrps27 ribosomal proteins identified here have not been reported as autoantigens. Autoantigen Sf3b1 (splicing factor 3B subunit 1) has been reported [11], but Sf3b3 identified here has not been reported as an autoantigen. Although similar RNA-binding proteins have been reported as autoantigens, RBM8a and Prpf19 have not been described as autoantigens.

DLST (dihydrolipopyllysine-residue succinyltransferase) is the E2 component of the 2-oxoglutarate dehydrogenase complex. It is mainly located in mitochondria. However, a small fraction localizes to the nucleus and participates in histone succinylation [72]. The RNA-binding autoantigen HuR is reported to affect the transcript of DLST [73]. DLST has been reported as an autoantigen [52].

All five proteins in the basement membrane or extracellular matrix family, including myosin 9, actin, lamins A and B2, and Lrp2 are known autoantigens (Table 1). Lrp2 (low-density lipoprotein receptor-related protein 2, also known as megalin) is found in the 1.0, 0.6, and 0.4 M elutions, suggesting its abundance in kidney tissue. Lrp2 is a multi-ligand endocytic receptor found in different tissues, but primarily in absorptive cells such as in the kidney. Its membranous expression is mainly seen in renal tubules and the parathyroid gland. Autoantibodies to Lrp2 have been found in patients with rheumatoid arthritis, systemic lupus erythematosus, Behçet disease, systemic sclerosis, and osteoarthritis [74]. In particular relation to kidney diseases, anti-Lrp2 and Lrp2 have been found in patients with kidney anti-brush border disease [25]. Immune deposits of anti-Lrp2 antibodies have been detected in the tubular basement membrane of affected kidneys.

Three heat shock proteins, Hsp5, Hspa1l, and Hsp90ab1, were identified in the strong DS affinity fraction (Table 1). HSPs are a group of stress-induced proteins and are thought to act as scavengers to trap abnormal proteins and protect stressed cells. HSPs share conserved structural motives, and autoantibodies to HSPs are frequently found in various autoimmune diseases [4446].

Prdx1 (peroxiredoxin-1) plays a role in cell protection against oxidative stress by detoxifying peroxides and acting as a sensor of H2O2-mediated signaling events. Prdx1 was identified in the strong DS affinity fraction and is a reported autoantigen [47].

Agmat (agmatinase, also named agmatine ureohydrolase) was seen in the strong DS affinity fraction. Agmat has its highest expression level in the cortex of the kidney. Immunochemical studies have revealed that Agmat is expressed in the tubular epithelial cells of a normal kidney, but its expression is altered in renal cell carcinoma [75]. A role of Agmat in autoimmunity has not yet been described.

Atg9a (autophagy-related protein 9A) showed strong DS affinity. Atg9a is involved in autophagy and cytoplasm-to-vacuole transport vesicle formation. Autophagy is an intracellular degradation pathway. Metabolic stress can trigger the rearrangement of the 14-3-3ζ interactome to favor interaction with the core autophagy regulator Atg9A, resulting in enhanced phagosome production [76]. A number of 14-3-3 proteins were found in the weak DS affinity fraction (Table 1). It was recently reported that annexin A2 is an autophagy modulator that regulates autophagosome formation by enabling appropriate ATG9A trafficking from endosomes to autophagosomes via actin [77]. Annexin A2 was identified in the weak DS affinity fraction, whereas actin was identified in the strong DS affinity fraction. Although 14-3-3, Anxa2, and actin have been reported as autoantigens, the role of Atg9a in autoimmunity is still unclear.

Proteins with medium DS affinity

From the fractions eluting at 0.6 M NaCl from the DS-affinity columns, 7 proteins were identified by MS, but 3 of them had already been present in the 1.0 M fraction. Therefore, only 4 new proteins were found (Table 1). The 3 proteins found in both the 1.0 M and 0.6 M fractions were Hspa5, Sf3b3, and Lrp2. The 4 new proteins were Prpf8, Atp1a1, Cpt2, and HMW-MAA.

Atp1a1 (Na+/K+-transporting ATPase subunit alpha 1) was found in the medium DS affinity fraction, whereas its two other subunits (Atp1a3 and Atp1b1) were found in the weak DS affinity fraction (Table 1). Atp1a1 (also called sodium pump subunit alpha-1) is the catalytic component of the active enzyme, catalyzing hydrolysis of ATP coupled with the exchange of Na+ and K+ ions across the plasma membrane. Mutations in the Atp1a1 gene causes dominant Charcot-Marie-Tooth disease 2DD [78]. Although autoantibodies to Atp1a1 have not been reported, an autoantibody to the beta-1 subunit has been described [56].

Cpt2 (carnitine O-palmitoyltransferase 2) is involved in the fatty acid-oxidation pathway, which is part of lipid metabolism. C2 deficiency caused by mutations in the Cpt2 gene is an autosomal recessive disorder of mitochondrial long-chain fatty acid oxidation, characterized by recurrent myoglobinuria, muscle pain, stiffness, and rhabdomyolysis. Myoglobinuria can cause kidney failure and death.

HMW-MAA (high molecular weight melanoma-associated protein) is also known as the melanoma chondroitin sulfate proteoglycan. Its expression is restricted in normal tissues but found in a large percentage of melanomas, and it has been used as a target for the immunotherapy of melanoma. Autoantibodies to this autoantigen were found in melanoma patients [32]. HMW-MAA is also a marker of activated pericytes [79]. Pericytes are contractile cells that wrap around the endothelial cells that line the capillaries and venules throughout the body. A deficiency of pericytes in the central nervous system can cause the blood-brain barrier to break down. Radiolabeled mAb immunoreactivity studies showed that HMW-MAA is highly expressed in the kidney and liver [80]. Possible autoimmune responses to HMW-MAA expressed in kidney microvascular pericytes may provide an important clue to renal capillary damage.

Proteins with weak DS affinity

From fractions eluting at 0.4 M NaCl, 116 proteins were initially identified. However, 21 of them had also been detected in fractions eluted with 0.6 M or 1.0 M NaCl. After excluding the redundancies, 95 new proteins were discovered in the 0.4 M elution, and are these are regarded as having weak DS affinity (Table 1).

Among these 95 proteins, 26 are attributed to the ANA family, 19 to the basement membrane/extracellular matrix family, 7 to the cell stress/apoptosis family, and the rest (43) are miscellaneous. Of the 95 proteins, 52 (55%) are known autoantigens based on specific autoantibodies reported in literature (see references in Table 1).

Miscellaneous proteins that were verified as autoantigens by literature search include protein disulfide isomerase, calreticulin, calnexin, transglutaminase 2, neutral alpha-glucosidase, proteasome protein, transitional endoplasmic reticulum ATPase, serpin B5, cytochrome P450 reductase, cytochrome c oxidase, branched-chain alpha-keto acid dehydrogenase, and aminopeptidase (see references in Table 1).

Of particular interest are the 14 proteasome proteins identified in the weak DS affinity fraction. TGM2 autoantibodies are widely used in the diagnostics of celiac disease. It has come to be appreciated that these autoantibodies, and also the TGM2-specific B cells, might play an active role in the pathogenesis of celiac disease. TGM2 catalyzes the cross-linking of proteins and the conjugation of polyamines to proteins. It is believed to play a role in the pathogenesis of several diseases, including celiac sprue, neurodegenerative diseases, and certain types of cancer.

A few proteins that play significant roles in diseases, but against which autoantibodies have not been reported, include Ggt1 (gamma-glutamyltransferase 1 or glutathione hydrolase 1 proenzyme) and CDH16 (cadherin 16). Ggt1 is expressed in tissues involved in adsorption and secretion, particularly in the kidney. Gamma-glutamyltransferase is found in the renal microvascular compartment [81]. It is speculated that Ggt1 may contribute to the etiology of diabetes. However, the role of Ggt1 in autoimmunity has not yet been established. CDH16 was identified in the fraction with weak DS affinity. It belongs to the cadherin superfamily and is expressed in the kidney and the thyroid gland. Although CDH16 has not been described as an autoantigen, autoantibodies to vascular endothelial cadherin are reported to be a marker of endothelial dysfunction in autoimmune diseases [59].

Among autoimmune diseases of the kidney, lupus nephritis is the most common. Systemic lupus erythematosus (SLE) is a frequent trigger of nephritis. About two-thirds of patients with lupus develop inflammation in the glomeruli. ANA, particular anti-nucleosome autoantibodies, are serological markers of SLE. Nucleosomes are highly organized functional subunits of chromatin, consisting of ~2 turns of dsDNA wrapped around an octamer of 2 molecules of H2A, H2B, H3, and H4. Nucleosomes are joined by linker DNA associated with H1. In our study, we have identified different isoforms of H4, H2A, H2B, and H1 (Table 1). Anti-nucleosome antibodies have been shown to be a prognostic marker for SLE with renal involvement.

Goodpasture syndrome, also known as anti-glomerular basement membrane (anti-GBM) disease, is a prototype of autoimmune disease [82]. It is a rare disease in which autoantibodies attack the kidneys and lungs, particularly the glomerular and/or pulmonary small vessels or capillaries. The patients develop specific autoantibodies against basement membrane antigens. These autoantibodies deposit in the basement membrane and form immune complexes that activate the classical complement pathway. Subsequently, neutrophil-dependent inflammation occurs resulting in glomerulonephritis and/or alveolitis. Ultimately, chronic kidney disease and kidney failure result. The GBM is an unusually thick extracellular matrix, consisting of laminin, collagen, heparan sulfate proteoglycan, and other components. Thus far, the only characterized GBM autoantigen is type IV collagen [83]. In our current study, we identified 22 proteins potentially associated with GBM, including lamin, actin, spectrin, tropomyosin, tubulin, vimentin, myosin, and agrin. Except for clathrin, all others (21/22) have been reported as autoantigens in various diseases (see references in Table 1). It is possible that the 22 proteins identified here will provide additional diagnostic markers for anti-GBM disease.

Anti-neutrophil cytoplasmic antibodies (ANCA)-associated vasculitis is another autoimmune disease where abnormal autoantibodies attack small blood vessels in different parts of the body, including the kidney. Myeloperoxidase (MPO) and proteinase 3 (PR3), two ANCA antigens, are pathogenic players in ANCA vasculitis and have diagnostic value in renal vasculitis. However, MPO and PR3 are produced by infiltrating neutrophils in inflammation, but not by the normal vascular components of the kidney, which explains why these two autoantigens were not identified from the kidney tissue in this study.

Conclusion

This study used the DS (dermatan sulfate)-affinity enrichment strategy and identified 96 proteins that are either verified or yet-to-be-confirmed autoantigens from kidney tissue. In particular, among these autoantigens, 22 proteins belong to the ANA (anti-nuclear antibodies) family, and 22 are related to the GBM or extracellular matrix, with ANA and GMB being two of the most characteristic autoantigens in autoimmune kidney diseases. In addition, this study identified 12 potential autoantigens that are related to apoptosis and/or cell stress, supporting the notion that apoptotic cells are an origin of autoantigens. Overall, our results strongly support a hypothesis for a unifying autoantigenicity principle, i.e., that self-molecules with affinity to DS have a high propensity to become autoantigens [1, 2]. The binding of DS to self-molecules may convert singular self-molecules (that are intrinsically non-antigenic) to non-self-complexes (that become antigenic), thereby inducing an autoimmune response. Our study demonstrates that our thesis about an underlying common property of autoantigens can be applied to identify known and to discover new renal autoantigens. A unifying physicochemical principle of autoantigenicity may eventually lead to therapeutic innovation by understanding the molecular etiology of autoimmunity.

Acknowledgments

We thank the Taplin Biological Mass Spectrometry facility at Harvard Medical School for expert help with protein sequencing.

References

  1. 1. Wang JY, Lee J, Yan M, Rho JH, Roehrl MH. Dermatan sulfate interacts with dead cells and regulates CD5(+) B-cell fate: implications for a key role in autoimmunity. Am J Pathol. 2011;178(5):2168–76. PubMed Central PMCID: PMC3081202. pmid:21514431
  2. 2. Rho JH, Zhang W, Murali M, Roehrl MH, Wang JY. Human proteins with affinity for dermatan sulfate have the propensity to become autoantigens. Am J Pathol. 2011;178(5):2177–90. PubMed Central PMCID: PMC3081203. pmid:21514432
  3. 3. Bruschi M, Galetti M, Sinico RA, Moroni G, Bonanni A, Radice A, et al. Glomerular Autoimmune Multicomponents of Human Lupus Nephritis In Vivo (2): Planted Antigens. J Am Soc Nephrol. 2015;26(8):1905–24. PubMed Central PMCID: PMC4520170. pmid:25398787
  4. 4. Burlingame RW, Boey ML, Starkebaum G, Rubin RL. The central role of chromatin in autoimmune responses to histones and DNA in systemic lupus erythematosus. J Clin Invest. 1994;94(1):184–92. PubMed Central PMCID: PMC296296. pmid:8040259
  5. 5. Dieker J, Berden JH, Bakker M, Briand JP, Muller S, Voll R, et al. Autoantibodies against Modified Histone Peptides in SLE Patients Are Associated with Disease Activity and Lupus Nephritis. PLoS One. 2016;11(10):e0165373. PubMed Central PMCID: PMC5079581. pmid:27780265
  6. 6. Wesierska-Gadek J, Penner E, Lindner H, Hitchman E, Sauermann G. Autoantibodies against different histone H1 subtypes in systemic lupus erythematosus sera. Arthritis Rheum. 1990;33(8):1273–8. pmid:2390129
  7. 7. Qin Z, Lavingia B, Zou Y, Stastny P. Antibodies against nucleolin in recipients of organ transplants. Transplantation. 2011;92(7):829–35. pmid:21869741
  8. 8. Ivanova LV, Rudolph P, Kellner U, Jurgensen A, Tareeva IE, Alm P, et al. Expression of DNA topoisomerases in chronic proliferative kidney disease. Kidney Int. 2000;58(4):1603–12. pmid:11012894
  9. 9. Bischoff FR, Maier G, Tilz G, Ponstingl H. A 47-kDa human nuclear protein recognized by antikinetochore autoimmune sera is homologous with the protein encoded by RCC1, a gene implicated in onset of chromosome condensation. Proc Natl Acad Sci U S A. 1990;87(21):8617–21. PubMed Central PMCID: PMC55008. pmid:2236072
  10. 10. Furuta K, Hildebrandt B, Matsuoka S, Kiyosawa K, Reimer G, Luderschmidt C, et al. Immunological characterization of heterochromatin protein p25beta autoantibodies and relationship with centromere autoantibodies and pulmonary fibrosis in systemic scleroderma. J Mol Med (Berl). 1998;76(1):54–60. pmid:9462868
  11. 11. Hwang HM, Heo CK, Lee HJ, Kwak SS, Lim WH, Yoo JS, et al. Identification of anti-SF3B1 autoantibody as a diagnostic marker in patients with hepatocellular carcinoma. J Transl Med. 2018;16(1):177. PubMed Central PMCID: PMC6025833. pmid:29954402
  12. 12. Okano Y, Targoff IN, Oddis CV, Fujii T, Kuwana M, Tsuzaka K, et al. Anti-U5 small nuclear ribonucleoprotein (snRNP) antibodies: a rare anti-U snRNP specificity. Clin Immunol Immunopathol. 1996;81(1):41–7. pmid:8808640
  13. 13. Hassfeld W, Chan EK, Mathison DA, Portman D, Dreyfuss G, Steiner G, et al. Molecular definition of heterogeneous nuclear ribonucleoprotein R (hnRNP R) using autoimmune antibody: immunological relationship with hnRNP P. Nucleic Acids Res. 1998;26(2):439–45. PubMed Central PMCID: PMC147279. pmid:9421497
  14. 14. Yau WY, Shih HC, Tsai MH, Sheu JC, Chen CH, Chow LP. Autoantibody recognition of an N-terminal epitope of hnRNP L marks the risk for developing HBV-related hepatocellular carcinoma. J Proteomics. 2013;94:346–58. pmid:24125732
  15. 15. Katsumata Y, Kawaguchi Y, Baba S, Hattori S, Tahara K, Ito K, et al. Serum antibodies against the 70k polypeptides of the U1 ribonucleoprotein complex are associated with psychiatric syndromes in systemic lupus erythematosus: a retrospective study. Mod Rheumatol. 2013;23(1):71–80. pmid:22454191
  16. 16. Brahms H, Raymackers J, Union A, de Keyser F, Meheus L, Luhrmann R. The C-terminal RG dipeptide repeats of the spliceosomal Sm proteins D1 and D3 contain symmetrical dimethylarginines, which form a major B-cell epitope for anti-Sm autoantibodies. J Biol Chem. 2000;275(22):17122–9. pmid:10747894
  17. 17. Kubo M, Ihn H, Kuwana M, Asano Y, Tamaki T, Yamane K, et al. Anti-U5 snRNP antibody as a possible serological marker for scleroderma-polymyositis overlap. Rheumatology (Oxford). 2002;41(5):531–4. pmid:12011376.
  18. 18. Bradley SV, Oravecz-Wilson KI, Bougeard G, Mizukami I, Li L, Munaco AJ, et al. Serum antibodies to huntingtin interacting protein-1: a new blood test for prostate cancer. Cancer Res. 2005;65(10):4126–33. pmid:15899803
  19. 19. Imai H, Chan EK, Kiyosawa K, Fu XD, Tan EM. Novel nuclear autoantigen with splicing factor motifs identified with antibody from hepatocellular carcinoma. J Clin Invest. 1993;92(5):2419–26. PubMed Central PMCID: PMC288425. pmid:8227358
  20. 20. Nabors LB, Furneaux HM, King PH. HuR, a novel target of anti-Hu antibodies, is expressed in non-neural tissues. J Neuroimmunol. 1998;92(1–2):152–9. pmid:9916890
  21. 21. Absi M, La Vergne JP, Marzouki A, Giraud F, Rigal D, Reboud AM, et al. Heterogeneity of ribosomal autoantibodies from human, murine and canine connective tissue diseases. Immunol Lett. 1989;23(1):35–41. pmid:2606513
  22. 22. Mooney CJ, Dunphy EJ, Stone B, McNeel DG. Identification of autoantibodies elicited in a patient with prostate cancer presenting as dermatomyositis. Int J Urol. 2006;13(3):211–7. pmid:16643611
  23. 23. Chen X, Fu S, Chen F, Chen H, Chen Z. Identification of tumor-associated antigens in human hepatocellular carcinoma by autoantibodies. Oncol Rep. 2008;20(4):979–85. pmid:18813843
  24. 24. Arbuckle MR, McClain MT, Rubertone MV, Scofield RH, Dennis GJ, James JA, et al. Development of autoantibodies before the clinical onset of systemic lupus erythematosus. N Engl J Med. 2003;349(16):1526–33. pmid:14561795
  25. 25. Larsen CP, Trivin-Avillach C, Coles P, Collins AB, Merchant M, Ma H, et al. LDL Receptor-Related Protein 2 (Megalin) as a Target Antigen in Human Kidney Anti-Brush Border Antibody Disease. J Am Soc Nephrol. 2018;29(2):644–53. PubMed Central PMCID: PMC5791069. pmid:29074737
  26. 26. von Muhlen CA, Chan EK, Peebles CL, Imai H, Kiyosawa K, Tan EM. Non-muscle myosin as target antigen for human autoantibodies in patients with hepatitis C virus-associated chronic liver diseases. Clin Exp Immunol. 1995;100(1):67–74. PubMed Central PMCID: PMC1534265. pmid:7697925
  27. 27. Musante L, Candiano G, Bruschi M, Santucci L, Carnemolla B, Orecchia P, et al. Circulating anti-actin and anti-ATP synthase antibodies identify a sub-set of patients with idiopathic nephrotic syndrome. Clin Exp Immunol. 2005;141(3):491–9. PubMed Central PMCID: PMC1809461. pmid:16045739
  28. 28. Hanrotel-Saliou C, Segalen I, Le Meur Y, Youinou P, Renaudineau Y. Glomerular antibodies in lupus nephritis. Clin Rev Allergy Immunol. 2011;40(3):151–8. pmid:20414746
  29. 29. Brito J, Biamonti G, Caporali R, Montecucco C. Autoantibodies to human nuclear lamin B2 protein. Epitope specificity in different autoimmune diseases. J Immunol. 1994;153(5):2268–77. pmid:7519647
  30. 30. Collard JF, Senecal JL, Raymond Y. Differential accessibility of the tail domain of nuclear lamin A in interphase and mitotic cells. Biochem Biophys Res Commun. 1990;173(1):363–9. pmid:2256928
  31. 31. von Mikecz A, Konstantinov K, Buchwald DS, Gerace L, Tan EM. High frequency of autoantibodies to insoluble cellular antigens in patients with chronic fatigue syndrome. Arthritis Rheum. 1997;40(2):295–305. pmid:9041942
  32. 32. Dummer R, Mittelman A, Fanizzi FP, Lucchese G, Willers J, Kanduc D. Non-self-discrimination as a driving concept in the identification of an immunodominant HMW-MAA epitopic peptide sequence by autoantibodies from melanoma cancer patients. Int J Cancer. 2004;111(5):720–6. pmid:15252841
  33. 33. Zaninoni A, Vercellati C, Imperiali FG, Marcello AP, Fattizzo B, Fermo E, et al. Detection of red blood cell antibodies in mitogen-stimulated cultures from patients with hereditary spherocytosis. Transfusion. 2015;55(12):2930–8. pmid:26259504
  34. 34. Kimura A, Sakurai T, Yamada M, Koumura A, Hayashi Y, Tanaka Y, et al. Anti-endothelial cell antibodies in patients with cerebral small vessel disease. Curr Neurovasc Res. 2012;9(4):296–301. pmid:23030505
  35. 35. Geng X, Biancone L, Dai HH, Lin JJ, Yoshizaki N, Dasgupta A, et al. Tropomyosin isoforms in intestinal mucosa: production of autoantibodies to tropomyosin isoforms in ulcerative colitis. Gastroenterology. 1998;114(5):912–22. pmid:9558279
  36. 36. Gajbhiye R, Sonawani A, Khan S, Suryawanshi A, Kadam S, Warty N, et al. Identification and validation of novel serum markers for early diagnosis of endometriosis. Hum Reprod. 2012;27(2):408–17. pmid:22158085
  37. 37. Manson JJ, Mills K, Jury E, Mason L, D'Cruz DP, Ni L, et al. Pathogenic autoantibodies from patients with lupus nephritis cause reduced tyrosine phosphorylation of podocyte proteins, including tubulin. Lupus Sci Med. 2014;1(1):e000013. PubMed Central PMCID: PMC4225730. pmid:25396061
  38. 38. Suzuki K, Nagao T, Itabashi M, Hamano Y, Sugamata R, Yamazaki Y, et al. A novel autoantibody against moesin in the serum of patients with MPO-ANCA-associated vasculitis. Nephrol Dial Transplant. 2014;29(6):1168–77. pmid:24319012
  39. 39. Wieczorek M, Czernik A. Paraneoplastic pemphigus: a short review. Clin Cosmet Investig Dermatol. 2016;9:291–5. PubMed Central PMCID: PMC5042195. pmid:27729809
  40. 40. Li FJ, Surolia R, Li H, Wang Z, Kulkarni T, Liu G, et al. Autoimmunity to Vimentin Is Associated with Outcomes of Patients with Idiopathic Pulmonary Fibrosis. J Immunol. 2017;199(5):1596–605. PubMed Central PMCID: PMC5563167. pmid:28754682
  41. 41. Zhang B, Shen C, Bealmear B, Ragheb S, Xiong WC, Lewis RA, et al. Autoantibodies to agrin in myasthenia gravis patients. PLoS One. 2014;9(3):e91816. PubMed Central PMCID: PMC3954737. pmid:24632822
  42. 42. Wakui H, Imai H, Kobayashi R, Itoh H, Notoya T, Yoshida K, et al. Autoantibody against erythrocyte protein 4.1 in a patient with autoimmune hemolytic anemia. Blood. 1988;72(2):408–12. pmid:3401589
  43. 43. Genevay S, Hayem G, Verpillat P, Meyer O. An eight year prospective study of outcome prediction by antiperinuclear factor and antikeratin antibodies at onset of rheumatoid arthritis. Ann Rheum Dis. 2002;61(8):734–6. PubMed Central PMCID: PMC1754185. pmid:12117683
  44. 44. Shimizu F, Schaller KL, Owens GP, Cotleur AC, Kellner D, Takeshita Y, et al. Glucose-regulated protein 78 autoantibody associates with blood-brain barrier disruption in neuromyelitis optica. Sci Transl Med. 2017;9(397). PubMed Central PMCID: PMC5585784. pmid:28679661
  45. 45. Chen M, Aosai F, Mun HS, Norose K, Hata H, Yano A. Anti-HSP70 autoantibody formation by B-1 cells in Toxoplasma gondii-infected mice. Infect Immun. 2000;68(9):4893–9. PubMed Central PMCID: PMC101692. pmid:10948102
  46. 46. Kenderov A, Minkova V, Mihailova D, Giltiay N, Kyurkchiev S, Kehayov I, et al. Lupus-specific kidney deposits of HSP90 are associated with altered IgG idiotypic interactions of anti-HSP90 autoantibodies. Clin Exp Immunol. 2002;129(1):169–76. PubMed Central PMCID: PMC1906416. pmid:12100037
  47. 47. Karasawa R, Ozaki S, Nishioka K, Kato T. Autoantibodies to peroxiredoxin I and IV in patients with systemic autoimmune diseases. Microbiol Immunol. 2005;49(1):57–65. pmid:15665454
  48. 48. Kistner A, Bigler MB, Glatz K, Egli SB, Baldin FS, Marquardsen FA, et al. Characteristics of autoantibodies targeting 14-3-3 proteins and their association with clinical features in newly diagnosed giant cell arteritis. Rheumatology (Oxford). 2017;56(5):829–34. pmid:28064210
  49. 49. van Beers-Tas MH, Marotta A, Boers M, Maksymowych WP, van Schaardenburg D. A prospective cohort study of 14-3-3eta in ACPA and/or RF-positive patients with arthralgia. Arthritis Res Ther. 2016;18:76. PubMed Central PMCID: PMC4818496. pmid:27037016
  50. 50. Caster DJ, Korte EA, Merchant ML, Klein JB, Wilkey DW, Rovin BH, et al. Autoantibodies targeting glomerular annexin A2 identify patients with proliferative lupus nephritis. Proteomics Clin Appl. 2015;9(11–12):1012–20. PubMed Central PMCID: PMC4690797. pmid:25824007
  51. 51. Li L, Chen SH, Yu CH, Li YM, Wang SQ. Identification of hepatocellular-carcinoma-associated antigens and autoantibodies by serological proteome analysis combined with protein microarray. J Proteome Res. 2008;7(2):611–20. pmid:18161940
  52. 52. Fregeau DR, Prindiville T, Coppel RL, Kaplan M, Dickson ER, Gershwin ME. Inhibition of alpha-ketoglutarate dehydrogenase activity by a distinct population of autoantibodies recognizing dihydrolipoamide succinyltransferase in primary biliary cirrhosis. Hepatology. 1990;11(6):975–81. pmid:2365294
  53. 53. Yeaman SJ, Fussey SP, Danner DJ, James OF, Mutimer DJ, Bassendine MF. Primary biliary cirrhosis: identification of two major M2 mitochondrial autoantigens. Lancet. 1988;1(8594):1067–70. pmid:2896910
  54. 54. Surh CD, Danner DJ, Ahmed A, Coppel RL, Mackay IR, Dickson ER, et al. Reactivity of primary biliary cirrhosis sera with a human fetal liver cDNA clone of branched-chain alpha-keto acid dehydrogenase dihydrolipoamide acyltransferase, the 52 kD mitochondrial autoantigen. Hepatology. 1989;9(1):63–8. pmid:2908870
  55. 55. Iwayama T, Leung PS, Coppel RL, Roche TE, Patel MS, Mizushima Y, et al. Specific reactivity of recombinant human PDC-E1 alpha in primary biliary cirrhosis. J Autoimmun. 1991;4(5):769–78. pmid:1797026
  56. 56. Jones CM, Toh BH, Pettitt JM, Martinelli TM, Humphris DC, Callaghan JM, et al. Monoclonal antibodies specific for the core protein of the beta-subunit of the gastric proton pump (H+/K+ ATPase). An autoantigen targetted in pernicious anaemia. Eur J Biochem. 1991;197(1):49–59. pmid:1707813
  57. 57. Aran A, Weiner K, Lin L, Finn LA, Greco MA, Peppard P, et al. Post-streptococcal auto-antibodies inhibit protein disulfide isomerase and are associated with insulin resistance. PLoS One. 2010;5(9):e12875. PubMed Central PMCID: PMC2944800. pmid:20886095
  58. 58. Kishore U, Sontheimer RD, Sastry KN, Zappi EG, Hughes GR, Khamashta MA, et al. The systemic lupus erythematosus (SLE) disease autoantigen-calreticulin can inhibit C1q association with immune complexes. Clin Exp Immunol. 1997;108(2):181–90. PubMed Central PMCID: PMC1904655. pmid:9158084
  59. 59. Bouillet L, Baudet AE, Deroux A, Sidibe A, Dumestre-Perard C, Mannic T, et al. Auto-antibodies to vascular endothelial cadherin in humans: association with autoimmune diseases. Lab Invest. 2013;93(11):1194–202. pmid:24061286
  60. 60. Weber CK, Haslbeck M, Englbrecht M, Sehnert B, Mielenz D, Graef D, et al. Antibodies to the endoplasmic reticulum-resident chaperones calnexin, BiP and Grp94 in patients with rheumatoid arthritis and systemic lupus erythematosus. Rheumatology (Oxford). 2010;49(12):2255–63. pmid:20716673
  61. 61. Kit Y, Starykovych M, Vajrychova M, Lenco J, Zastavna D, Stoika R. Detection of novel auto-antigens in patients with recurrent miscarriage: description of an approach and preliminary findings. Croat Med J. 2014;55(3):259–64. PubMed Central PMCID: PMC4049207. pmid:24891284
  62. 62. Sugimoto K, Hiwasa T, Shibuya K, Hirano S, Beppu M, Isose S, et al. Novel autoantibodies against the proteasome subunit PSMA7 in amyotrophic lateral sclerosis. J Neuroimmunol. 2018;325:54–60. pmid:30390597
  63. 63. Becker A, Ludwig N, Keller A, Tackenberg B, Eienbroker C, Oertel WH, et al. Myasthenia gravis: analysis of serum autoantibody reactivities to 1827 potential human autoantigens by protein macroarrays. PLoS One. 2013;8(3):e58095. PubMed Central PMCID: PMC3587426. pmid:23483977
  64. 64. Miyachi K, Hirano Y, Horigome T, Mimori T, Miyakawa H, Onozuka Y, et al. Autoantibodies from primary biliary cirrhosis patients with anti-p95c antibodies bind to recombinant p97/VCP and inhibit in vitro nuclear envelope assembly. Clin Exp Immunol. 2004;136(3):568–73. PubMed Central PMCID: PMC1809050. pmid:15147362
  65. 65. Braus BK, Miller I, Kummer S, Kleinwort KJH, Hirmer S, Hauck SM, et al. Investigation of corneal autoantibodies in horses with immune mediated keratitis (IMMK). Vet Immunol Immunopathol. 2017;187:48–54. pmid:28494929
  66. 66. Kratz A, Harding MW, Craft J, Mackworth-Young CG, Handschumacher RE. Autoantibodies against cyclophilin in systemic lupus erythematosus and Lyme disease. Clin Exp Immunol. 1992;90(3):422–7. PubMed Central PMCID: PMC1554566. pmid:1458678
  67. 67. Shinoda M, Tanaka Y, Kuno T, Matsufuji T, Matsufuji S, Murakami Y, et al. High levels of autoantibodies against drug-metabolizing enzymes in SLA/LP-positive AIH-1 sera. Autoimmunity. 2004;37(6–7):473–80. pmid:15621574
  68. 68. Schmits R, Kubuschok B, Schuster S, Preuss KD, Pfreundschuh M. Analysis of the B cell repertoire against autoantigens in patients with giant cell arteritis and polymyalgia rheumatica. Clin Exp Immunol. 2002;127(2):379–85. PubMed Central PMCID: PMC1906335. pmid:11876765
  69. 69. Bremer HD, Landegren N, Sjoberg R, Hallgren A, Renneker S, Lattwein E, et al. ILF2 and ILF3 are autoantigens in canine systemic autoimmune disease. Sci Rep. 2018;8(1):4852. PubMed Central PMCID: PMC5859008. pmid:29556082
  70. 70. Xu X, Rahbar A, Omarsdottir S, Teng J, Nemeth A, Fischler B, et al. CD13 Autoantibodies Are Elevated in Sera From Mothers of Infants With Neonatal Cholestasis of Different Causes. J Pediatr Gastroenterol Nutr. 2017;64(1):76–82. pmid:27203397
  71. 71. Rauhavirta T, Hietikko M, Salmi T, Lindfors K. Transglutaminase 2 and Transglutaminase 2 Autoantibodies in Celiac Disease: a Review. Clin Rev Allergy Immunol. 2016. pmid:27263022
  72. 72. Wang Y, Guo YR, Liu K, Yin Z, Liu R, Xia Y, et al. KAT2A coupled with the alpha-KGDH complex acts as a histone H3 succinyltransferase. Nature. 2017;552(7684):273–7. PubMed Central PMCID: PMC5841452. pmid:29211711
  73. 73. Diaz-Munoz MD, Bell SE, Fairfax K, Monzon-Casanova E, Cunningham AF, Gonzalez-Porta M, et al. The RNA-binding protein HuR is essential for the B cell antibody response. Nat Immunol. 2015;16(4):415–25. PubMed Central PMCID: PMC4479220. pmid:25706746
  74. 74. Ooka S, Matsui T, Nishioka K, Kato T. Autoantibodies to low-density-lipoprotein-receptor-related protein 2 (LRP2) in systemic autoimmune diseases. Arthritis Res Ther. 2003;5(3):R174–80. PubMed Central PMCID: PMC165049.
  75. 75. Dallmann K, Junker H, Balabanov S, Zimmermann U, Giebel J, Walther R. Human agmatinase is diminished in the clear cell type of renal cell carcinoma. Int J Cancer. 2004;108(3):342–7. pmid:14648699
  76. 76. Weerasekara VK, Panek DJ, Broadbent DG, Mortenson JB, Mathis AD, Logan GN, et al. Metabolic-stress-induced rearrangement of the 14-3-3zeta interactome promotes autophagy via a ULK1- and AMPK-regulated 14-3-3zeta interaction with phosphorylated Atg9. Mol Cell Biol. 2014;34(24):4379–88. PubMed Central PMCID: PMC4248729. pmid:25266655
  77. 77. Moreau K, Ghislat G, Hochfeld W, Renna M, Zavodszky E, Runwal G, et al. Transcriptional regulation of Annexin A2 promotes starvation-induced autophagy. Nat Commun. 2015;6:8045. PubMed Central PMCID: PMC4560779. pmid:26289944
  78. 78. Lassuthova P, Rebelo AP, Ravenscroft G, Lamont PJ, Davis MR, Manganelli F, et al. Mutations in ATP1A1 Cause Dominant Charcot-Marie-Tooth Type 2. Am J Hum Genet. 2018;102(3):505–14. PubMed Central PMCID: PMC5985288. pmid:29499166
  79. 79. Rajkumar VS, Sundberg C, Abraham DJ, Rubin K, Black CM. Activation of microvascular pericytes in autoimmune Raynaud's phenomenon and systemic sclerosis. Arthritis Rheum. 1999;42(5):930–41. (199905)42:5<930::AID-ANR11>3.0.CO;2–1. pmid:10323448
  80. 80. Fawwaz RA, Wang TS, Estabrook A, Rosen JM, Hardy MA, Alderson PO, et al. Immunoreactivity and biodistribution of indium-111-labeled monoclonal antibody to a human high molecular weight-melanoma associated antigen. J Nucl Med. 1985;26(5):488–92. pmid:3989605
  81. 81. Dass PD, Misra RP, Welbourne TC. Presence of gamma-glutamyltransferase in the renal microvascular compartment. Can J Biochem. 1981;59(6):383–6. pmid:6117358
  82. 82. Hellmark T, Segelmark M. Diagnosis and classification of Goodpasture's disease (anti-GBM). J Autoimmun. 2014;48–49:108–12. pmid:24456936
  83. 83. McAdoo SP, Pusey CD. Anti-Glomerular Basement Membrane Disease. Clin J Am Soc Nephrol. 2017;12(7):1162–72. PubMed Central PMCID: PMC5498345. pmid:28515156