Skip to main content
Erschienen in: BMC Immunology 1/2019

Open Access 01.12.2019 | Research article

A comprehensive autoantigen-ome of autoimmune liver diseases identified from dermatan sulfate affinity enrichment of liver tissue proteins

verfasst von: Wei Zhang, Jung-hyun Rho, Michael H. Roehrl, Julia Y. Wang

Erschienen in: BMC Immunology | Ausgabe 1/2019

Abstract

Background

Autoimmune diseases result from aberrant immune attacks by the body itself. It is mysterious how autoantigens, a large cohort of seemingly unconnected molecules expressed in different parts of the body, can induce similar autoimmune responses. We have previously found that dermatan sulfate (DS) can form complexes with molecules of apoptotic cells and stimulate autoreactive CD5+ B cells to produce autoantibodies. Hence, autoantigenic molecules share a unique biochemical property in their affinity to DS. This study sought to further test this uniform principle of autoantigenicity.

Results

Proteomes were extracted from freshly collected mouse livers. They were loaded onto columns packed with DS-Sepharose resins. Proteins were eluted with step gradients of increasing salt strength. Proteins that bound to DS with weak, moderate, or strong affinity were eluted with 0.4, 0.6, and 1.0 M NaCl, respectively. After desalting, trypsin digestion, and gel electrophoresis, proteins were sequenced by mass spectrometry. To validate whether these proteins have been previously identified as autoantigens, an extensive literature search was conducted using the protein name or its alternative names as keywords. Of the 41 proteins identified from the strong DS-affinity fraction, 33 (80%) were verified autoantigens. Of the 46 proteins with moderate DS-affinity, 27 (59%) were verified autoantigens. Of the 125 proteins with weak DS-affinity, 44 (35%) were known autoantigens. Strikingly, these autoantigens fell into the classical autoantibody categories of autoimmune liver diseases: ANA (anti-nuclear autoantibodies), SMA (anti-smooth muscle autoantibodies), AMA (anti-mitochondrial autoantibodies), and LKM (liver-kidney microsomal autoantigens).

Conclusions

This study of DS-affinity enrichment of liver proteins establishes a comprehensive autoantigen-ome for autoimmune liver diseases, yielding 104 verified and 108 potential autoantigens. The liver autoantigen-ome sheds light on the molecular origins of autoimmune liver diseases and further supports the notion of a unifying biochemical principle of autoantigenicity.
Hinweise

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
AIH
Autoimmune hepatitis
AMA
Anti-mitochondrial autoantibodies
DS
Dermatan sulfate
LKM
Liver-kidney microsomal autoantigens
PBC
Primary biliary cirrhosis
PSC
Primary sclerosing cholangitis
SMA
Smooth muscle autoantibodies

Background

The etiology of autoimmune diseases in general has remained a biomedical mystery. It is not clear how and why some molecules or tissue components of the body become a self-target of the immune defense system, whereas most do not. In previous studies, we demonstrated that certain molecules from dying cells have affinity to 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 to DS have a high propensity to be autoantigens (autoAg) [2]. We proposed a uniform principle of autoantigenicity that explains how a vast variety of seemingly unrelated molecules can become autoantigenic by means of a shared biochemical property. In this study, we sought to test this principle and to define the repertoire of possible autoantigens, i.e., the autoantigen-ome, in autoimmune liver diseases.
Autoimmune diseases of the liver occur when the body’s own immune system attacks the liver [35]. These diseases have different clinical patterns with regard to degree of severity and clinical course, but they all share one important feature, i.e., the liver being the target of an aberrant autoimmune attack by autoantibodies and/or autoreactive cells. Autoimmune liver diseases are typically chronic conditions, and patients may experience persistent or recurrent autoimmune insults to the liver, often without overt symptoms. As the autoimmune attack persists, liver tissue scars and leads to hepatic fibrosis; and as fibrosis progresses to cirrhosis, liver function is compromised. Ultimately, end-stage liver disease and liver failure may ensue, requiring organ transplantation.
Among autoimmune diseases of the liver, autoimmune hepatitis (AIH) [3], primary biliary cirrhosis (PBC) [4], and primary sclerosing cholangitis (PSC) [5] are the most prominent. In AIH, the immune system attacks the hepatocytes and causes chronic inflammation of the liver. About 70% of AIH patients are female. In PBC, the autoimmune reaction is directed at small biliary ducts inside the liver. In PSC, autoimmunity targets the larger extrahepatic bile ducts. Characteristic morphological patterns are chronic inflammation and a hepatic pattern of injury with prominent plasma cells in AIH, destruction of small intrahepatic bile ducts and canals of Hering in PBC, and periductal fibrosis and inflammation of the larger bile ducts, often along with inflammatory bowel disease, in PSC. Although most liver autoimmune diseases fall into these three categories, overlaps and other syndromes also occur.
Autoimmune liver diseases are typically associated with several classes of autoantibodies, including ANA, AMA, anti-SMA/anti-F-actin, anti-LKM, and others [6, 7]. For AIH and PBC, testing for liver-related autoantibodies is a prerequisite for diagnosis. For PSC, autoantibodies are frequently present but their diagnostic value has not been established. When diagnosed at an early stage, autoimmune hepatitis can be controlled by daily doses of steroids and other medicines that suppress inflammation. However, these treatments only suppress or slow down the overactive immune system, but cannot cure the disease. Understanding the molecular origins of autoimmune liver diseases is therefore crucial to finding more effective therapies.

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 h with end-over-end rotation. After the first 60 min, 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 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.

Mouse liver protein extraction

Livers were obtained from 5-month-old BALB/cJ female mice (Jackson Laboratory, Bar Harbor, Maine). The mouse tissue use was approved by the Institutional Animal Care and Use Committee (IACUC) of Brigham and Women’s Hospital (Boston, MA). All animal care was provided according to institutional, local, state, and federal regulations at the Brigham and Women’s Hospital research animal facility. A total of 20 mice were killed with CO2, their blood was removed through heart puncture, and their livers were collected immediately. Livers were cleaned by rinsing with phosphate buffered saline (PBS, pH 7.2) twice and then stored at 4 °C for 1 h, -20 °C for 2 h, and then -80 °C until further processing. Thawed livers were cut to small pieces and pressed through a cell strainer (Fisher Scientific). To remove red blood cells, the liver tissue was mixed with 10 ml of RBC (red blood cell) lysis buffer for 10 s. After centrifugation for 5 min, the supernatant was discarded. The liver 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 min or until all tissue pieces appeared dissolved. The mixture was centrifuged at 13,300 rpm for 20 min, and the supernatant that contains total soluble liver proteins was collected. Protein concentration was measured by the RC DC protein assay (Bio-Rad).

DS-affinity fractionation

Pilot affinity fractionations were performed in small centrifuge tubes. Aliquots of 2 ml resin were centrifuged to remove the storage buffer, and 0.5 ml of extracted liver proteins was added. The tube was mixed by rotating end-over-end at room temperature for 1 h to allow sufficient binding of proteins to DS-resin. The resin slush was centrifuged, and the supernatant containing unbound proteins was removed. The resins were washed 4 times with 1 ml of 0.2 M NaCl in 10 mM phosphate buffer to further remove unbound or poorly bound proteins. Proteins bound to DS with weak affinity were released from the resins by 0.4 M NaCl in PBS by rotating end-over-end at room temperature for 30 min, and the supernatant containing weak-binding proteins was recovered by centrifugation. The high-affinity proteins still bound to DS-resin were obtained by boiling the resins with 0.2 ml of SDS-PAGE sample buffer. Proteins in each fraction were assessed by 1D gel electrophoresis.
After the pilot assessment, fractionation of larger quantities of liver proteins was carried out by FPLC using a Biologic Duo-Flow System (Bio-Rad). Liver 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 buffer A to remove unbound proteins. Proteins bound to DS resins with weak-to-strong affinity were eluted with a step-gradient of 0.2 M, 0.4 M, 0.6 M, and 1.0 M NaCl in 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 10 mM phosphate buffer for further analysis.

Protein sequencing by mass spectrometry

Fractionated proteins with different affinities 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 3 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 2 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 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 2 peptide matches were considered confidently identified.

Results

Fractionation of liver proteins by DS-affinity

Proteins extracted from mouse livers were separated into 4 fractions according to their strength of binding to DS: no-, weak-, medium-, and strong-affinity. This was carried out by loading the liver proteins onto DS-Sepharose columns to allow binding to take place. Proteins that did not bind to DS resins were washed off the column with the 10 mM phosphate loading buffer, followed by washing with 0.2 M NaCl and 10 mM phosphate buffer. Afterwards, proteins that had remained bound to DS were sequentially eluted from the column with 0.4 M, 0.6 M, and 1.0 M NaCl, designating these as weak-, medium-, and strong-affinity fractions, respectively. Elution was monitored for presence of proteins, and individual chromatographic fractions containing proteins at each of the salt strengths were pooled, desalted, and concentrated. Protein content and protein size distribution of the fractions were assessed with 1D SDS PAGE gels.
The majority of liver tissue proteins not binding to DS were observed in the flow-through, and non-specific binding proteins were further washed with 0.2 M NaCl. As the elution proceeded, the amount of proteins eluting at increasing ionic strength became smaller and smaller. Proteins eluting last off the column with 1.0 M NaCl had the highest affinity to DS but were also the relatively least abundant. The flow-through and 0.2 M NaCl washed proteins were not further analyzed. Proteins eluting at 0.4 M, 0.6 M, and 1.0 M salt were sequenced by LC-MS/MS, yielding 168, 68, and 41 identified protein entities, respectively. Some of the proteins were redundantly identified in 2 or 3 fractions and, when redundancies were excluded, the number of different uniquely identified proteins in the 0.4 M, 0.6 M, or 1.0 M fraction was found to be 125, 46, and 41, respectively.

Proteins with strong DS-affinity eluting at 1.0 M ionic strength

Proteins eluting off the DS-Sepharose column at 1.0 M NaCl were classified as having strong DS-affinity. From the 1.0 M elution, 41 proteins were identified by MS sequencing (Table 1). A literature search revealed that at least 33 (80.5%) of these have previously been identified as autoantigens for autoantibodies. Furthermore, these autoantigens were not randomly distributed in functional attribution but fell nicely into 5 classical categories of autoantibodies in autoimmune liver diseases: ANA (antinuclear autoantibodies), SMA (smooth muscle autoantibodies), AMA (anti-mitochondrial autoantibodies), LKM (liver-kidney microsomal autoantigens), and peroxisome (Table 1).
Table 1
Liver proteins with strong DS-affinity
a
  
ANA (antinuclear autoantigens)
Ref.
10
IPI00329998.3
H4
Histone H4
[8]
6
IPI00114642.4
Hist1h2bj
Histone H2B type 1-F/J/L
[9]
5
IPI00153400.2
H2afj
Histone H2A.J
[9]
5
IPI00111957.3
Hist1h2ba
Histone H2B type 1-A
[9]
4
IPI00137852.5
H2afy
Core histone macro-H2A.1
[10]
2
IPI00404590.1
H1f0
Histone H1.0
[11]
3
IPI00119220.1
Snrpd2
Small nuclear ribonucleoprotein Sm D2
[12]
2
IPI00114052.1
Snrpb
Small nuclear ribonucleoprotein-associated protein B
[13]
2
IPI00122350.4
Snrpa
U1 small nuclear ribonucleoprotein A
[14]
2
IPI00133955.1
Snrpe
Small nuclear ribonucleoprotein E
[15]
3
IPI00131988.1
Mrpl49
39S ribosomal protein L49, mitochondria
[16]
3
IPI00311236.1
Rpl7
60S ribosomal protein L7
[17]
2
IPI00122421.5
Rpl27
60S ribosomal protein L27
[18]
2
IPI00222549.6
Rpl30
60S ribosomal protein L30
[16]
3
IPI00124287.1
Pabpc1
Polyadenylate-binding protein
[19]
2
IPI00119959.1
Banf1
Barrier-to-autointegration factor
[20]
   
SMA (anti-smooth muscle autoantigens) / Cytoskeleton
 
6
IPI00753793.2
Spna2
Isoform 2 of Spectrin alpha chain
[21]
5
IPI00123181.4
Myh9
Myosin-9
[22]
2
IPI00109044.8
G15Rik
Myosin light chain, regulatory B-like
[23]
2
IPI00354819.5
Myl6
Isoform Smooth muscle of Myosin light chain 6
[24]
2
IPI00230435.1
Lmna
Isoform C2 of Lamin-A
[25]
   
AMA (anti-mitochondrial autoantigens)
 
13
IPI00319992.1
Hspa5
78 kDa glucose-regulated protein (Grp78)
[26]
13
IPI00114209.1
Glud1
Glutamate dehydrogenase 1, mitochondria
[27, 28]
11
IPI00131424.3
Cpt2
Carnitine O-palmitoyltransferase 2, mitochondria
 
6
IPI00133903.1
Hspa9
Stress-70 protein, mitochondria (Grp75)
[29]
3
IPI00111908.8
Cps1
Carbamoyl-phosphate synthase [ammonia], mitochondria
 
3
IPI00273146.1
Chdh
Choline dehydrogenase, mitochondria
 
2
IPI00129577.1
Aifm1
Apoptosis-inducing factor 1 (Aif, Pdcd8, programmed cell death protein 8), mitochondria
[30]
   
LKM (liver-kidney microsomal autoantigens)
 
3
IPI00111936.1
Ugt1a9
UDP-glucuronosyltransferase 1-9 (bilirubin-specific)
[31]
3
IPI00112322.2
Ugt2b5
UDP glucuronosyltransferase 2 family, polypeptide B5
[32]
2
IPI00230113.5
Cyb5
Microsomal cytochrome b5
[33]
2
IPI00131771.3
Cox6c
Cytochrome c oxidase subunit 6c
 
2
IPI00169666.3
Ugt2b34
UDP glucuronosyltransferase 2 family, polypeptide B3
[32]
2
IPI00230108.6
Pdia3
Protein disulfide-isomerase A
[34]
2
IPI00134746.5
Ass1
Argininosuccinate synthase
 
2
IPI00387289.3
Ces3
Carboxylesterase
[35]
   
Peroxisomal proteins
 
3
IPI00112549.1
Acsl1
Long-chain-fatty-acid-CoA ligase
 
2
IPI00312058.5
Cat
Catalase
[36, 37]
2
IPI00223367.5
Uox
Uricase
 
2
IPI00125325.1
Decr2
Peroxisomal 2,4-dienoyl-CoA reductase
 
2
IPI00125813.1
Dpp4
Dipeptidyl peptidase (CD26)
[38, 39]
aColumns left to right: Number of peptides identified for the protein by mass spectrometry; Protein ID; Gene name; Protein name; Reference (if any) reporting autoantibodies induced by the protein
Among the 41 proteins identified, 16 belong to the ANA family. They include 6 histones, 4 small nuclear ribonucleoproteins, 4 ribosomal proteins, and 2 others (polyadenylate-binding protein and barrier-to-autointegration factor). All of them are established ANA autoantigens (see references in Table 1). There are 5 proteins belonging to the SMA family, including spectrin alpha chain, myosin-9, myosin light chain, and lamin-A. All of them are known targets of autoantibodies (see references in Table 1). Among the AMA autoantigens, 7 were identified, with 4 having been verified as autoantigens, including Hspa5, Hspa9, glutamate dehydrogenase (Glud1), and apoptosis-inducing factor 1 (Aifm1). Autoantibodies to heat shock proteins (HSP) are widely found in autoimmune diseases as well as in numerous other diseases. Autoantibodies to HSP have been found in the circulation of various cancer patients, and are proposed as diagnostic and prognostic markers for various cancers such as breast cancer [40]. Autoantibodies to Glud1 have not been reported in humans but in mice and calves [27, 28]. Autoantibodies to Cpt2, Cps1, and Chdh could not be found in published reports. They could well represent autoantigens that have yet to be validated.
Autoantibodies to microsomal LKM antigens are associated with type 2 autoimmune hepatitis. Among proteins with high DS-affinity, 8 microsomal proteins were identified, including 3 of the UDG-glucuronosyltransferase family, 2 cytochromes, and 3 enzymes. Six of them (Ugt1a9, Ugt2b5, UgtCyb5, Cyb5, Pdia3, and Ces3) are known bona fide autoantigens (Table 1). Autoantibodies to Cox6c and Ass1 have not yet been described in literature. There are 5 proteins associated with the peroxisome in the 1.0 M elution fraction. Catalase and dipeptidyl peptidase (CD26, Dpp4) are reported autoantigens (Table 1). Autoantibodies to Acsl1, Uox, and Decr2 have not yet been reported.

Proteins with moderate DS-affinity eluting at 0.6 M ionic strength

From fractions eluting at 0.6 M salt from DS-affinity columns, 68 proteins were identified by MS, but 22 of them were also present in the 1.0 M fraction. Therefore, 46 unique proteins were found (Table 2). Similarly to those with strong DS-affinity as described above, these proteins fell nicely into 5 categories of well-known autoantibodies: ANA, SMA, AMA, LKM, and peroxisome.
Table 2
Liver proteins with moderate DS-affinity
a
  
ANA (antinuclear autoantigens)
Ref.
8
IPI00230730.4
Hist2h3b
Histone H3.2
[41]
4
IPI00223713.5
Hist1h1c
Histone H1.2
[42]
2
IPI00459318.1
Hist1h2bp
Putative uncharacterized protein, histone
[9]
2
IPI00136632.3
H2afy3
Histone H2A member Y3
[43]
2
IPI00320149.2
H2afv
Histone H2A member V
[9]
6
IPI00308706.4
Rpl5
60S ribosomal protein L5
 
5
IPI00127085.6
Rpl10a
60S ribosomal protein L10a
 
3
IPI00555113.2
Rpl18
60S ribosomal protein L18
 
3
IPI00138892.2
Uba52
Ubiquitin-60S ribosomal protein L40
 
2
IPI00122598.3
EG382723
Similar to ribosomal protein L10
 
5
IPI00339468.4
Dhx9
Isoform 2 of ATP-dependent RNA helicase, DNA helicase II
[44]
4
IPI00121596.3
Prpf8
Pre-mRNA-processing-splicing factor 8
 
3
IPI00109764.2
Top1
DNA topoisomerase 1 (Scl-70)
[45]
2
IPI00322749.3
Snrpd1
Small nuclear ribonucleoprotein Sm D1
[46]
2
IPI00120162.1
Csnk2a1
Casein kinase II subunit alpha
[17]
   
SMA (anti-smooth muscle autoantigens) / Cytoskeleton
 
6
IPI00400300.1
Lmna
Isoform C of Lamin-A
[47]
4
IPI00113886.1
Lmnb2
Isoform B3 of Lamin-B
[48]
2
IPI00126191.5
Lmnb2
Isoform B2 of Lamin-B
[49]
2
IPI00113824.1
Hspg2
Basement membrane-specific heparan sulfate proteoglycan core protein
[50]
   
AMA (anti-mitochondrial autoantigens)
 
5
IPI00331555.2
Bckdha
Branched chain keto acid dehydrogenase E1, alpha polypeptide (mitochondrion matrix)
[51]
5
IPI00111877.1
Ssbp1
Single-stranded DNA-binding protein, mitochondria
 
4
IPI00153144.3
Suox
Sulfite oxidase, mitochondria
[52]
3
IPI00133208.3
Hspa1l
Heat shock 70 kDa protein 1L
[53]
2
IPI00323357.3
Hspa8
Heat shock cognate 71 kDa protein
[54]
3
IPI00420718.4
Hmgcs2
Hydroxymethylglutaryl-CoA synthase, mitochondria
[27]
3
IPI00223092.5
Hadha
Trifunctional enzyme subunit alpha, mitochondria
 
3
IPI00135651.1
Slc25a13
Calcium-binding mitochondrial carrier protein Aralar2
 
3
IPI00111885.1
Uqcrc1
Cytochrome b-c1 complex subunit 1, mitochondria
[33]
3
IPI00132799.4
C1qbp
Complement component 1 q subcomponent binding protein
 
2
IPI00127841.3
Slc25a5
ADP/ATP translocase (mitochondrion inner membrane)
 
2
IPI00387379.1
Decr1
2,4-dienoyl-CoA reductase, mitochondria
 
   
LKM (liver-kidney microsomal autoantigens)
 
6
IPI00117914.3
Arg1
Arginase-1
[55]
6
IPI00115679.1
Ganab
Isoform 2 of Neutral alpha-glucosidase A
[56]
4
IPI00621548.2
Por
NADPH-cytochrome P450 reductase
[57]
4
IPI00134691.3
Ugt1a1
UDP-glucuronosyltransferase 1-1
[58]
2
IPI00110556.1
Cyp2e1
Cytochrome P450 2E1
[59, 60]
2
IPI00321644.3
Cyp2d26
Cytochrome P450 2D26 (mouse) (LKM1 human)
[31]
   
Peroxisomal proteins
 
6
IPI00127558.3
Acox1
Peroxisomal acyl-coenzyme A oxidase
 
6
IPI00127276.1
Ehhadh
Enoyl-Coenzyme A hydratase/3-hydroxyacyl CoA dehydrogenase (peroxisomal bifunctional enzyme)
[16]
2
IPI00331628.5
Hsd17b4
Peroxisomal multifunctional enzyme type (17 beta-hydroxysteroid dehydrogenase type 4)
[61]
   
Miscellaneous
 
4
IPI00115599.6
Hsd11b1
Corticosteroid 11-beta-dehydrogenase isozyme
 
3
IPI00117705.1
Ddost
Dolichyl-diphosphooligosaccharide-protein glycosyltransferase 48 kDa subunit
 
3
IPI00313236.3
Slc27a5
Bile acyl-CoA synthetase
 
2
IPI00309035.2
Rpn1
Dolichyl-diphosphooligosaccharide-protein glycosyltransferase subunit 1 (ribophorin 1)
 
2
IPI00127016.1
Hsd17b6
Hydroxysteroid 17-beta dehydrogenase
[62]
2
IPI00130985.1
Rdh7
Retinol dehydrogenase
 
aColumns left to right: Number of peptides identified for the protein by mass spectrometry; Protein ID; Gene name; Protein name; Reference (if any) reporting autoantibodies induced by the protein
There are 15 proteins belonging to the ANA family, including 5 histones and 5 ribosomal proteins (Table 2). Aside from these, other interesting autoantigens were identified. DNA topisomerase 1, the classical Scl-70 autoantigen, was identified [45]. Casein kinase II was also identified [17]. Dxh9 (ATP-dependent RNA helicase and DNA helicase II) may resemble the classical Ku antigen [44]. Prpf8, a pre-mRNA-processing-splicing factor, has not been reported as an autoantigen.
There are 4 proteins belonging to the SMA family. Aside from 3 isoforms of lamin, autoantigen Hspg2 (basement membrane-specific heparan sulfate proteoglycan core protein) was found in the 0.6 M elution. Among the 12 proteins in the AMA family, autoantibodies to 6 have been reported, including Bckdha, Suox, Hspa1l, Hmgcs2, Uqcrc1, and Hspa8. Although autoantibodies to C1q have been widely studied, anti-C1qbp (complement C1q subunit binding protein) has not yet been reported. Ssbp1, Hadha, Scl25a5, or Decr1 were not found in the literature as autoantigens.
From the 0.6 M elution, all 6 proteins identified in the LKM family are reported autoantigens (see references in Table 2). The classical cytochrome P450 antigens including LKM1 were identified. Other autoantigens identified are UDP-glucuronosyltransferase 1, Arg1, and Ganab. In addition to the above classical categories of autoantigens, 6 proteins identified in the 0.6 M elution are miscellaneous. Except for Rdh7 being a reported autoantigen, Hsd11b1, Ddost, Slc27a5, Rpn1, and Hsd17b6 autoantibodies remain to be characterized.

Proteins with weak DS-affinity eluting at 0.4 M ionic strength

From fractions eluting at 0.4 M salt, 168 proteins were initially identified. Among these, 18 were also found in both 0.6 M and 1.0 M elution, including H2bj, H2afj, H2afy, Snrpb, Myh9, Lmna, Hspa9, Hspa5, Cps1, Cpt2, Glud1, Ugt2b5, Pdia3, Ass1, Acsl1, Cat, Uox, and Aif. Among the rest, 3 (Sm D2, Spna2, and Ces) were found also in the 1.0 M elution but in not 0.6 M elution, and 22 proteins were found also in the 0.6 M but not in the 1.0 M elution. After excluding these redundancies, 125 proteins were found only in the 0.4 M elution (Table 3).
Table 3
Liver proteins with weak DS-affinity
a
  
ANA (antinuclear autoantigens)
Ref.
19
IPI00122011.2
Sf3b3
Isoform 1 of Splicing factor 3B subunit
[63]
14
IPI00420807.3
Sfrs1
Isoform 1 of Splicing factor, arginine/serine-rich
 
2
IPI00153743.1
Sfrs7
Isoform 2 of Splicing factor, arginine/serine-rich
 
4
IPI00123604.4
Rpsa
40S ribosomal protein SA
 
3
IPI00469260.3
Eftud2
116 kDa U5 small nuclear ribonucleoprotein component
[64]
2
IPI00170008.1
Snrpa1
U2 small nuclear ribonucleoprotein A
[65]
2
IPI00114052.1
Snrpb
Small nuclear ribonucleoprotein-associated protein
[13]
2
IPI00119220.1
Snrpd2
Small nuclear ribonucleoprotein Sm D2
[66]
2
IPI00226073.2
Hnrnpf
Isoform 1 of Heterogeneous nuclear ribonucleoprotein F
 
2
IPI00109860.3
Rbm8a
Isoform 2 of RNA-binding protein 8
 
   
SMA (anti-smooth muscle autoantigens)/Cytoskeleton
 
19
IPI00123316.1
Tpm1
Isoform 1 of Tropomyosin alpha-1 chain
[67]
13
IPI00169707.2
Tpm3
Tropomyosin 3, gamm
[68]
9
IPI00230044.5
Tpm3
Isoform 2 of Tropomyosin alpha-3 chain
 
2
IPI00421223.3
Tpm4
Tropomyosin alpha-4 chain
[69]
13
IPI00118899.1
Actn4
Alpha-actinin
[70]
9
IPI00113539.2
Fn1
Fibronectin
[71]
7
IPI00110850.1
Actb
Actin, cytoplasmic
[72]
6
IPI00110827.1
Acta1
Actin, alpha skeletal muscle
[72]
7
IPI00265380.4
Myh8
Myosin-8
[73]
3
IPI00129404.1
Myh6
Myosin-6
 
3
IPI00114894.1
Myh11
Isoform 1 of Myosin-1
 
5
IPI00230394.5
Lmnb1
Lamin-B
[74]
4
IPI00121892.9
Spnb2
Isoform 2 of Spectrin beta chain
[75]
3
IPI00227299.6
Vim
Vimentin
[69]
2
IPI00109061.1
Tubb2b
Tubulin beta-2B chain
[76]
   
AMA (anti-mitochondrial autoantigens)
 
52
IPI00129526.1
Hsp90b1
Endoplasmin
[26]
9
IPI00229080.7
Hsp90ab1
MCG1823
 
4
IPI00330804.4
Hsp90aa1
Heat shock protein HSP 90-alpha
 
39
IPI00136213.5
Sardh
Sarcosine dehydrogenase, mitochondrial
 
31
IPI00468481.2
Atp5b
ATP synthase subunit beta, mitochondrial
[77]
21
IPI00130280.1
Atp5a1
ATP synthase subunit alpha, mitochondrial
 
27
IPI00471246.2
Ivd
Isovaleryl-CoA dehydrogenase, mitochondrial
 
17
IPI00134809.2
Dlst
Isoform 1 of Dihydrolipoyllysine-residue succinyltransferase component of 2-oxoglutarate dehydrogenase complex, mitochondrial
[78]
2
IPI00756386.1
Dhtkd1
Probable 2-oxoglutarate dehydrogenase E1 component DHKTD1, mitochondrial
 
13
IPI00331564.2
Dld
Dihydrolipoyl dehydrogenase
 
7
IPI00130535.1
Dbt
Lipoamide acyltransferase component of branched-chain alpha-keto acid dehydrogenase complex, mitochondrial
[78]
3
IPI00153660.4
Dlat
Dihydrolipoyllysine-residue acetyltransferase component of pyruvate dehydrogenase complex, mitochondrial
 
13
IPI00387491.1
Aass
Alpha-aminoadipic semialdehyde synthase, mitochondrial
 
13
IPI00468653.4
Pccb
Propionyl-CoA carboxylase beta chain, mitochondrial
 
12
IPI00330523.1
Pcca
Propionyl-CoA carboxylase alpha chain, mitochondrial
 
11
IPI00110843.3
Agmat
Agmatinase, mitochondrial
 
9
IPI00114710.2
Pcx
Pyruvate carboxylase, mitochondrial isoform 2
 
7
IPI00111218.1
Aldh2
Aldehyde dehydrogenase, mitochondrial
[27]
4
IPI00226430.2
Acaa2
3-Ketoacyl-CoA thiolase, mitochondrial
 
3
IPI00119766.1
Rdh16
Retinol dehydrogenase 1
 
3
IPI00405699.2
Aldh4a1
Delta-1-pyrroline-5-carboxylate dehydrogenase, mitochondrial
 
3
IPI00121309.2
Ndufs3
NADH dehydrogenase [ubiquinone] iron-sulfur protein 3, mitochondrial
 
3
IPI00753303.2
Npl22
Dihydrodipicolinate synthase-like, mitochondrial
 
3
IPI00169862.1
Coq9
Ubiquinone biosynthesis protein COQ9, mitochondrial
 
2
IPI00323592.2
Mdh2
Malate dehydrogenase, mitochondrial
 
2
IPI00121105.2
Hadh
Hydroxyacyl-coenzyme A dehydrogenase, mitochondrial
 
2
IPI00459725.2
Idh3a
Isoform 1 of Isocitrate dehydrogenase [NAD] subunit alpha, mitochondrial
[79]
2
IPI00133553.1
Mut
Methylmalonyl-CoA mutase, mitochondrial
 
2
IPI00115607.3
Hadhb
Trifunctional enzyme subunit beta, mitochondrial
 
2
IPI00130804.1
Ech1
Delta(3,5)-Delta(2,4)-dienoyl-CoA isomerase, mitochondrial
[80]
2
IPI00469195.2
Echdc2
Isoform 1 of Enoyl-CoA hydratase domain-containing protein 2, mitochondrial
 
2
IPI00314909.2
Agxt
Alanine-glyoxylate aminotransferase
 
2
IPI00226140.5
Maob
Amine oxidase [flavin-containing]
[81]
2
IPI00121440.4
Etfb
Electron transfer flavoprotein subunit beta
[82]
2
IPI00454008.1
Shmt2
Serine hydroxymethyltransferase
 
   
LKM (liver-kidney microsomal autoantigens)
 
54
IPI00309073.2
Mttp
Microsomal triglyceride transfer protein
 
33
IPI00153317.3
Aldh1l1
10-formyltetrahydrofolate dehydrogenase
 
10
IPI00111936.1
Ugt1a9
UDP-glucuronosyltransferase 1-9
[31]
8
IPI00762897.2
Ugcgl1
UDP-glucose:glycoprotein glucosyltransferase
 
3
IPI00127223.3
Ugt2b36
UDP-glucuronosyltransferase
 
3
IPI00114778.1
Cyp2c37
Cytochrome P450 2C37
[83]
3
IPI00131176.1
mt-Co2
Cytochrome c oxidase subunit 2
 
2
IPI00323908.1
Cyp2d10
Cytochrome P450 2D10
[83]
3
IPI00331322.3
Mgst1
Microsomal glutathione S-transferase
 
9
IPI00115867.4
Ces1
Liver carboxylesterase
[35]
   
Peroxisomal proteins
 
6
IPI00331596.6
Pecr
Peroxisomal trans-2-enoyl-CoA reductase
 
3
IPI00134870.3
Acox2
Peroxisomal acyl-coenzyme A oxidase
 
5
IPI00110719.1
Pipox
Peroxisomal sarcosine oxidase
[84]
2
IPI00130924.1
Slc27a2
Very long-chain acyl-CoA synthetaselow-den
 
2
IPI00121833.3
Acaa1a
Acaa1b 3-ketoacyl-CoA thiolase A, peroxisomal
 
2
IPI00111235.2
Aldh3a2
Fatty aldehyde dehydrogenase variant
 
   
Apoptosis
 
9
IPI00310240.4
Anxa6
Annexin A6 isoform
[85]
5
IPI00116498.1
Ywhaz
14-3-3 protein zeta/delta
[86]
5
IPI00118384.1
Ywhae
14-3-3 protein epsilon
[87]
5
IPI00230707.6
Ywhag
14-3-3 protein gamma
[87]
   
Proteasome
 
8
IPI00113845.1
Psmb1
Proteasome subunit beta type-1
[88]
4
IPI00119239.2
Psmb6
Proteasome subunit beta type-6
 
3
IPI00116712.1
Psmb8
Proteasome subunit beta type-8
 
2
IPI00128945.1
Psmb2
Proteasome subunit beta type-2
 
2
IPI00129512.3
Psmb4
Proteasome subunit beta type-4
 
2
IPI00136483.1
Psmb7
Proteasome subunit beta type-7
 
5
IPI00331644.5
Psma3
Proteasome subunit alpha type-3
[88]
4
IPI00109122.1
Psma8
Proteasome subunit alpha type-7-like
 
4
IPI00131845.1
Psma6
Proteasome subunit alpha type-6
 
4
IPI00420745.7
Psma2
Proteasome subunit alpha type-2
 
4
IPI00277001.4
Psma4
Proteasome subunit alpha type-4
[89]
   
Miscellaneous
 
68
IPI00123639.1
Calr
Calreticulin
[77]
67
IPI00271951.5
Pdia4
Protein disulfide isomerase A
 
61
IPI00122815.3
P4hb
Protein disulfide-isomerase
[90]
17
IPI00222496.3
Pdia6
Putative uncharacterized protein
 
2
IPI00163011.2
Txndc5
Thioredoxin domain-containing protein
 
20
IPI00119618.1
Canx
Calnexin
[91]
18
IPI00622235.5
Vcp
Transitional endoplasmic reticulum ATPase
[92]
14
IPI00125514.1
Entpd5
Ectonucleoside triphosphate diphosphohydrolase 5
 
11
IPI00475154.1
Rpn2
Dolichyl-diphosphooligosaccharide-protein glycosyltransferase subunit
 
2
IPI00309035.2
Rpn1
Dolichyl-diphosphooligosaccharide--protein glycosyltransferase subunit
 
11
IPI00112719.1
Alad
Delta-aminolevulinic acid dehydratase
 
10
IPI00115680.1
Prkcsh
Isoform 1 of Glucosidase 2 subunit beta
 
7
IPI00119063.2
Lrp1
Pro-low-density lipoprotein receptor-related protein (alpha-2-macroglobulin receptor, apolipoprotein E receptor)
 
2
IPI00624663.3
Pzp
Alpha-2-macroglobulin
 
7
IPI00135512.1
Cnpy2
Protein canopy homolog 2
 
6
IPI00316314.1
Hacl1
2-hydroxyacyl-CoA lyase
 
6
IPI00116254.1
Prdx4
Peroxiredoxin-4
[93]
6
IPI00125899.1
Ctnnb1
Catenin beta-1
 
4
IPI00112963.1
Ctnna1
Catenin alpha-1
[94]
5
IPI00113869.1
Bsg
Isoform 2 of Basigin (M6, EMMPRIN, TCSF, CD147)
 
4
IPI00126184.7
Gc
Vitamin D-binding protein
[95]
4
IPI00123342.4
Hyou1
Hypoxia up-regulated protein
 
3
IPI00130950.1
Bhmt
Betaine--homocysteine S-methyltransferase
[96]
3
IPI00134058.3
Erp44
Endoplasmic reticulum resident protein ERp44
 
3
IPI00387282.4
Aadac
Arylacetamide deacetylase
 
3
IPI00122346.2
Ssr4
Signal sequence receptor, delta
 
3
IPI00317740.5
Gnb2l1
Guanine nucleotide-binding protein subunit beta-2-like
 
3
IPI00319973.3
Pgrmc1
Membrane-associated progesterone receptor component
 
2
IPI00279218.1
Apeh
Isoform 2 of Acylamino-acid-releasing enzyme
 
2
IPI00323624.3
C3
Isoform Long of Complement C3
[97]
2
IPI00116432.1
Fmo1
Dimethylaniline monooxygenase [N-oxide-forming]
 
2
IPI00114044.1
Man2a1
Alpha-mannosidase
 
2
IPI00312018.6
Mlec
Malectin
 
2
IPI00115241.1
Mup4
Major urinary protein 4
 
aColumns left to right: Number of peptides identified for the protein by mass spectrometry; Protein ID; Gene name; Protein name; Reference (if any) reporting autoantibodies induced by the protein
As shown in Table 3, these 125 proteins fell naturally into 8 categories: ANA (10 proteins), SMA (15 proteins), AMA (35 proteins), LKM (10 proteins), peroxisome (6 proteins), apoptosis (4 proteins), proteasome (11 proteins), and miscellaneous (34 proteins).
The ANA autoantigens with weak DS-affinity are primarily isoforms of splicing factors and small nuclear ribonucleoproteins. Although autoantibodies to splicing factors have been reported [63], the exact isoforms identified here have not been reported. Anti-smooth muscle autoantigens identified in the 0.4 M NaCl elution included various forms of tropomyosin, actinin, fibronectin, actin, myosin, lamin, spectrin, and tubulin. Among the 15 identified here, 12 are bona fide autoantigens. Among the 35 proteins associated with mitochondria, 32 are enzymes, with 8/32 being reported autoantigens. These enzymes are from diverse families, e.g., dehydrogenases, synthases, acyltransferases, or carboxylases.
Similar to those identified in the 1.0 M and 0.6 M factions, the LKM autoantigens included members of cytochrome and UDP-glucuronosyltransferase families. In addition, 3 unrelated proteins, Mttp, Aldh1l1, and Ces1 were identified. Ces1 is a verified autoantigen. Six enzymes associated with the peroxisome were identified. Thus far, only peroxisomal sarcosine oxidase has been described to be an autoantigen. In addition to the above autoantigen categories, additional proteins were found associated with apoptosis and the proteasome (Table 3). Annexin A6 and 14–3-3 proteins are reported autoantigens. Members of the proteasome have also been reported as autoantigens.
The remaining 34 proteins could not easily be classified into particular categories. The majority of them are not yet characterized as autoantigens. However, some are reported autoantigens, such as calreticulin, calnexin, catenin, protein disulfide-isomerase, peroxiredoxin 4, vitamin D-binding protein, and complement C3 (Table 3).

Discussion

Under normal physiologic conditions, the immune system is designed to protect from infection and disease through intricate mechanisms that distinguish self from non-self. It is a mystery why and how the immune system is mistakenly triggered to attack the body’s self. Autoimmune responses are causally linked to autoantibodies, autoreactive cells, or both. Despite advances in our understanding of the many facets of autoimmunity, the underlying molecular and cellular mechanisms that trigger autoimmunity remain largely unknown.
We are intrigued by the question why and how a vast number of diverse, seemingly functionally disconnected proteins in different parts of the body and with diverse structures and biological functions can all induce a converging autoimmune response, i.e., the production of autoantibodies by autoreactive B cells. Based on our previous studies [1, 2], we concluded that autoantigens share a common biochemical property in their binding affinity to dermatan sulfate (DS), also called chondroitin sulfate B, a glycosaminoglycan-type mucopolysaccharide found mostly in skin but also in blood vessels, heart valves, tendons, lungs, and other tissues. DS can directly bind molecules released from dying cells or other sources and form macromolecular DS-autoantigen complexes, and such complexes, in turn, can stimulate autoreactive B cells through simultaneous engagement of multiple signaling molecules on the B cell surface to induce an activated B cell response. To further characterize our proposed “unifying principle of autoantigenicity” based on DS-affinity as a shared physicochemical property of autoantigens, we tested whether we could identify autoantigens from a specific parenchymal organ, and whether autoantigens showed preferential intrinsic biochemical propensity for high DS-affinity.
Autoimmune liver diseases result from the immune system mistakenly attacking hepatocytes or cholangiocytes in the liver [35]. Patients with these chronic conditions are usually initially rather asymptomatic, and autoantibody serology tests are often necessary to clarify the diagnosis [6, 7]. For example, while routine blood tests for liver enzymes can reveal patterns of hepatitis, further autoantibody tests are needed to diagnose autoimmune hepatitis. Autoantibody tests also help distinguish autoimmune hepatitis from other liver diseases, such as viral hepatitis or metabolic diseases such as Wilson disease.
Common autoimmune liver diseases include autoimmune hepatitis (AIH) [3], primary biliary cirrhosis (PBC) [4], and primary sclerosing cholangitis (PSC) [5]. An autoimmune liver disease panel (a series of tests that detect autoantibodies to common autoantigens associated with these diseases) include anti-liver-kidney microsomal antibodies (LKM), anti-mitochondrial antibodies (AMA), anti-nuclear antibodies (ANA), and anti-smooth muscle antibodies (SMA). AIH is further classified to two types, type I is defined by positive ANA and SMA, whereas type 2 is associated with anti-KLM autoantibodies. ANA occur in a wide variety of systemic autoimmune diseases, such as systemic lupus erythematosus, rheumatoid arthritis, Sjögren syndrome, and systemic sclerosis. Lupus hepatitis is regarded as a distinct manifestation of SLE [98]. The identification of 41 confirmed or putative ANA autoantigens from liver tissue uncovered by our study may perhaps explain the overlap autoantibody profile and clinical manifestations between lupus and AIH. AMA are hallmark diagnostic markers for PBC. In PBC, the targets are small bile ducts, but the prototypic serologic response is the production of a multilineage immune response to mitochondrial autoantigens. AMA are detected in 90–95% of PBC patients, although their presence is extremely low in the general population (varying between 0.16 and 1%) [99]. More than 60 autoantibodies have been detected in patients with PBC [99]. In our current study, we identified 54 verified and putative autoantigens associated with mitochondria.
Based on all of our observation so far, we find that autoantigens with the strongest DS affinity are typically DNA- and RNA-binding proteins. Other autoantigens largely display moderate to weak DS affinity. However, it should be noted that our definition of DS binding strength is arbitrary, with DS-autoAg complexes dissociable at 1.0, 0.6, and 0.4 M ionic strength defined as strongly, moderately, and weakly binding, respectively. All of these DS-binding proteins would be expected to remain in complexed forms with DS under physiologic conditions. For example, cytochrome P450 2D6 (CYP2D6) is the major autoantigen of LKM1 autoantibodies [100], but its mouse homologues (Cyp2d26 and Cyp2d10) were found to possess only moderate to weak DS affinity (Tables 2 and 3). As another example, PDC-E2 is a major autoantigen in PBC patients, but several components of the PDC (pyruvate dehydrogenase complex) were only identified in the weak but not the strong DS affinity fraction of this study (Table 3). Hence, these results suggest that proteins only need to exhibit some (sufficient) DS affinity to become potentially autoantigenic. It is also possible that in toto weakly DS-binding proteins may contain fragment epitopes with strong DS affinity, and such epitopes could determine the autoantigenicity of the protein.
The liver is the largest internal organ, the largest gland of the human body, and also the largest reservoir of human proteins. The liver serves hundreds of physiological functions, including removal of toxic substance, storage of glycogen, decomposition of red blood cells, production of bile and hormones, and synthesis of plasma proteins. Transcriptome analysis shows that 59% (n = 11,553) of all human proteins (n = 19,613) are expressed in the liver (The Human Protein Atlas). It should be noted that our DS-affinity approach provided a significant enrichment of liver protein autoantigens, yielding only a little over 200 proteins (i.e., around 1% of the total human proteome) as bona fide verified or potential autoantigens.

Conclusions

Our study of DS-affinity enrichment of the liver proteome produced a comprehensive autoantigen-ome that includes 104 bona fide autoantigens and 108 potential autoantigens for autoimmune liver diseases. These autoantigens fell into the classical categories of autoantibodies for autoimmune liver diseases. Our study provides further support to a model in which DS-affinity is a distinct biochemical property of proteins that can become autoantigens, whereas proteins that lack DS-affinity have a much lower propensity to be targets of autoimmunity (Fig. 1). These results may help in the further characterization of autoantigenic molecules and thus point to new innovative directions in autoimmunity research.

Acknowledgements

We thank the Taplin Biological Mass Spectrometry facility at Harvard Medical School for expert help with protein sequencing. We thank Michael W. Roehrl for editing the manuscript.
The mouse tissue use was approved by the Institutional Animal Care and Use Committee (IACUC) of Brigham and Women’s Hospital (Boston, MA). All animal care was provided according to institutional, local, state, and federal regulations at the Brigham and Women’s Hospital research animal facility.
Not applicable.

Competing interests

WZ and JHR were supported by the NIH during the course of this study and declare no competing interests. MP Biomedicals is the current employer of JHR and has neither relation to nor made contribution to the study and has not had any role in this research. MHR declares no competing interests. JYW is a co-founder and shareholder of Curandis and declares no other competing interests.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Literatur
1.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
3.
Zurück zum Zitat Terziroli Beretta-Piccoli B, Mieli-Vergani G, Vergani D. Autoimmune hepatitis: standard treatment and systematic review of alternative treatments. World J Gastroenterol. 2017;23(33):6030–48.PubMedPubMedCentralCrossRef Terziroli Beretta-Piccoli B, Mieli-Vergani G, Vergani D. Autoimmune hepatitis: standard treatment and systematic review of alternative treatments. World J Gastroenterol. 2017;23(33):6030–48.PubMedPubMedCentralCrossRef
4.
Zurück zum Zitat Trivedi HD, Lizaola B, Tapper EB, Bonder A. Primary biliary cholangitis: new treatments for an old disease. Frontline Gastroenterol. 2017;8(1):29–36.PubMedCrossRef Trivedi HD, Lizaola B, Tapper EB, Bonder A. Primary biliary cholangitis: new treatments for an old disease. Frontline Gastroenterol. 2017;8(1):29–36.PubMedCrossRef
5.
Zurück zum Zitat Arndtz K, Hirschfield GM. Primary sclerosing cholangitis and the management of uncertainty and complexity. Frontline Gastroenterol. 2017;8(4):260–6.PubMedPubMedCentralCrossRef Arndtz K, Hirschfield GM. Primary sclerosing cholangitis and the management of uncertainty and complexity. Frontline Gastroenterol. 2017;8(4):260–6.PubMedPubMedCentralCrossRef
6.
Zurück zum Zitat Sebode M, Weiler-Normann C, Liwinski T, Schramm C. Autoantibodies in autoimmune liver disease-clinical and diagnostic relevance. Front Immunol. 2018;9:609.PubMedPubMedCentralCrossRef Sebode M, Weiler-Normann C, Liwinski T, Schramm C. Autoantibodies in autoimmune liver disease-clinical and diagnostic relevance. Front Immunol. 2018;9:609.PubMedPubMedCentralCrossRef
8.
Zurück zum Zitat Dieker J, Berden JH, Bakker M, Briand JP, Muller S, Voll R, Sjowall C, Herrmann M, Hilbrands LB, van der Vlag J. Autoantibodies against modified histone peptides in SLE patients are associated with disease activity and lupus nephritis. PLoS One. 2016;11(10):e0165373.PubMedPubMedCentralCrossRef Dieker J, Berden JH, Bakker M, Briand JP, Muller S, Voll R, Sjowall C, Herrmann M, Hilbrands LB, van der Vlag J. Autoantibodies against modified histone peptides in SLE patients are associated with disease activity and lupus nephritis. PLoS One. 2016;11(10):e0165373.PubMedPubMedCentralCrossRef
9.
Zurück zum Zitat Burlingame RW, Rubin RL. Autoantibody to the nucleosome subunit (H2A-H2B)-DNA is an early and ubiquitous feature of lupus-like conditions. Mol Biol Rep. 1996;23(3–4):159–66.PubMedCrossRef Burlingame RW, Rubin RL. Autoantibody to the nucleosome subunit (H2A-H2B)-DNA is an early and ubiquitous feature of lupus-like conditions. Mol Biol Rep. 1996;23(3–4):159–66.PubMedCrossRef
10.
Zurück zum Zitat Kwon YS, Chung J, Shin GT, Lee SY, Jang YJ. Variable region genes of human monoclonal autoantibodies to histones H2A and H2B from a systemic lupus erythematosus patient. Mol Immunol. 2005;42(3):311–7.PubMedCrossRef Kwon YS, Chung J, Shin GT, Lee SY, Jang YJ. Variable region genes of human monoclonal autoantibodies to histones H2A and H2B from a systemic lupus erythematosus patient. Mol Immunol. 2005;42(3):311–7.PubMedCrossRef
11.
Zurück zum Zitat 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.PubMedCrossRef 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.PubMedCrossRef
12.
Zurück zum Zitat McClain MT, Ramsland PA, Kaufman KM, James JA. Anti-sm autoantibodies in systemic lupus target highly basic surface structures of complexed spliceosomal autoantigens. J Immunol. 2002;168(4):2054–62.PubMedCrossRef McClain MT, Ramsland PA, Kaufman KM, James JA. Anti-sm autoantibodies in systemic lupus target highly basic surface structures of complexed spliceosomal autoantigens. J Immunol. 2002;168(4):2054–62.PubMedCrossRef
13.
Zurück zum Zitat Wang H, Demirkan G, Bian X, Wallstrom G, Barker K, Karthikeyan K, Tang Y, Pasha SF, Leighton JA, Qiu J, et al. Identification of antibody against SNRPB, small nuclear Ribonucleoprotein-associated proteins B and B', as an autoantibody marker in Crohn's disease using an Immunoproteomics approach. J Crohns Colitis. 2017;11(7):848–56.PubMedCrossRef Wang H, Demirkan G, Bian X, Wallstrom G, Barker K, Karthikeyan K, Tang Y, Pasha SF, Leighton JA, Qiu J, et al. Identification of antibody against SNRPB, small nuclear Ribonucleoprotein-associated proteins B and B', as an autoantibody marker in Crohn's disease using an Immunoproteomics approach. J Crohns Colitis. 2017;11(7):848–56.PubMedCrossRef
14.
Zurück zum Zitat Mesa A, Somarelli JA, Wu W, Martinez L, Blom MB, Greidinger EL, Herrera RJ. Differential immunoglobulin class-mediated responses to components of the U1 small nuclear ribonucleoprotein particle in systemic lupus erythematosus and mixed connective tissue disease. Lupus. 2013;22(13):1371–81.PubMedCrossRef Mesa A, Somarelli JA, Wu W, Martinez L, Blom MB, Greidinger EL, Herrera RJ. Differential immunoglobulin class-mediated responses to components of the U1 small nuclear ribonucleoprotein particle in systemic lupus erythematosus and mixed connective tissue disease. Lupus. 2013;22(13):1371–81.PubMedCrossRef
15.
Zurück zum Zitat Reuter R, Rothe S, Habets W, Van Venrooij WJ, Luhrmann R. Autoantibody production against the U small nuclear ribonucleoprotein particle proteins E, F and G in patients with connective tissue diseases. Eur J Immunol. 1990;20(2):437–40.PubMedCrossRef Reuter R, Rothe S, Habets W, Van Venrooij WJ, Luhrmann R. Autoantibody production against the U small nuclear ribonucleoprotein particle proteins E, F and G in patients with connective tissue diseases. Eur J Immunol. 1990;20(2):437–40.PubMedCrossRef
16.
Zurück zum Zitat Absi M, La Vergne JP, Marzouki A, Giraud F, Rigal D, Reboud AM, Reboud JP, Monier JC. Heterogeneity of ribosomal autoantibodies from human, murine and canine connective tissue diseases. Immunol Lett. 1989;23(1):35–41.PubMedCrossRef Absi M, La Vergne JP, Marzouki A, Giraud F, Rigal D, Reboud AM, Reboud JP, Monier JC. Heterogeneity of ribosomal autoantibodies from human, murine and canine connective tissue diseases. Immunol Lett. 1989;23(1):35–41.PubMedCrossRef
17.
Zurück zum Zitat Neu E, von Mikecz AH, Hemmerich PH, Peter HH, Fricke M, Deicher H, Genth E, Krawinkel U. Autoantibodies against eukaryotic protein L7 in patients suffering from systemic lupus erythematosus and progressive systemic sclerosis: frequency and correlation with clinical, serological and genetic parameters. The SLE study group. Clin Exp Immunol. 1995;100(2):198–204.PubMedPubMedCentralCrossRef Neu E, von Mikecz AH, Hemmerich PH, Peter HH, Fricke M, Deicher H, Genth E, Krawinkel U. Autoantibodies against eukaryotic protein L7 in patients suffering from systemic lupus erythematosus and progressive systemic sclerosis: frequency and correlation with clinical, serological and genetic parameters. The SLE study group. Clin Exp Immunol. 1995;100(2):198–204.PubMedPubMedCentralCrossRef
18.
Zurück zum Zitat Perone D, Santos MA, Peixoto MS, Cicarelli RM. Trypanosoma cruzi: identification and characterization of a novel ribosomal protein L27 (TcrL27) that cross-reacts with an affinity-purified anti-Sm antibody. Parasitology. 2003;126(Pt 6):577–83.PubMed Perone D, Santos MA, Peixoto MS, Cicarelli RM. Trypanosoma cruzi: identification and characterization of a novel ribosomal protein L27 (TcrL27) that cross-reacts with an affinity-purified anti-Sm antibody. Parasitology. 2003;126(Pt 6):577–83.PubMed
19.
Zurück zum Zitat Becker A, Ludwig N, Keller A, Tackenberg B, Eienbroker C, Oertel WH, Fassbender K, Meese E, Ruprecht K. Myasthenia gravis: analysis of serum autoantibody reactivities to 1827 potential human autoantigens by protein macroarrays. PLoS One. 2013;8(3):e58095.PubMedPubMedCentralCrossRef Becker A, Ludwig N, Keller A, Tackenberg B, Eienbroker C, Oertel WH, Fassbender K, Meese E, Ruprecht K. Myasthenia gravis: analysis of serum autoantibody reactivities to 1827 potential human autoantigens by protein macroarrays. PLoS One. 2013;8(3):e58095.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Forne I, Carrascal M, Martinez-Lostao L, Abian J, Rodriguez-Sanchez JL, Juarez C. Identification of the autoantigen HB as the barrier-to-autointegration factor. J Biol Chem. 2003;278(50):50641–4.PubMedCrossRef Forne I, Carrascal M, Martinez-Lostao L, Abian J, Rodriguez-Sanchez JL, Juarez C. Identification of the autoantigen HB as the barrier-to-autointegration factor. J Biol Chem. 2003;278(50):50641–4.PubMedCrossRef
21.
Zurück zum Zitat Azizah MR, Azila MN, Zulkifli MN, Norita TY. The prevalence of antinuclear, anti-dsDNA, anti-Sm and anti-RNP antibodies in a group of healthy blood donors. Asian Pac J Allergy Immunol. 1996;14(2):125–8.PubMed Azizah MR, Azila MN, Zulkifli MN, Norita TY. The prevalence of antinuclear, anti-dsDNA, anti-Sm and anti-RNP antibodies in a group of healthy blood donors. Asian Pac J Allergy Immunol. 1996;14(2):125–8.PubMed
22.
Zurück zum Zitat Migliorini P, Baldini C, Rocchi V, Bombardieri S. Anti-Sm and anti-RNP antibodies. Autoimmunity. 2005;38(1):47–54.PubMedCrossRef Migliorini P, Baldini C, Rocchi V, Bombardieri S. Anti-Sm and anti-RNP antibodies. Autoimmunity. 2005;38(1):47–54.PubMedCrossRef
23.
Zurück zum Zitat Bledzhyants DA, Muratov RM, Movsesyan RR, Podlubnaya ZA. Autoantibodies to myosin light chains in the blood as early marker of myocardial injury after aortocoronary bypass surgery. Bull Exp Biol Med. 2007;144(2):241–5.PubMedCrossRef Bledzhyants DA, Muratov RM, Movsesyan RR, Podlubnaya ZA. Autoantibodies to myosin light chains in the blood as early marker of myocardial injury after aortocoronary bypass surgery. Bull Exp Biol Med. 2007;144(2):241–5.PubMedCrossRef
24.
Zurück zum Zitat Latif N, Baker CS, Dunn MJ, Rose ML, Brady P, Yacoub MH. Frequency and specificity of antiheart antibodies in patients with dilated cardiomyopathy detected using SDS-PAGE and western blotting. J Am Coll Cardiol. 1993;22(5):1378–84.PubMedCrossRef Latif N, Baker CS, Dunn MJ, Rose ML, Brady P, Yacoub MH. Frequency and specificity of antiheart antibodies in patients with dilated cardiomyopathy detected using SDS-PAGE and western blotting. J Am Coll Cardiol. 1993;22(5):1378–84.PubMedCrossRef
25.
Zurück zum Zitat Malka D, Pham BN, Courvalin JC, Corbic M, Pessayre D, Erlinger S. Acute hepatitis caused by alverine associated with anti-Lamin a and C autoantibodies. J Hepatol. 1997;27(2):399–403.PubMedCrossRef Malka D, Pham BN, Courvalin JC, Corbic M, Pessayre D, Erlinger S. Acute hepatitis caused by alverine associated with anti-Lamin a and C autoantibodies. J Hepatol. 1997;27(2):399–403.PubMedCrossRef
26.
Zurück zum Zitat Kadam KM, Mande PV, Gawas N, Ahire S, Khole LV. Autoantibodies to heat-shock protein, HSPA5, and epitope spreading: high-dose dexamethasone therapy rescues ovarian function in experimental autoimmune ovarian insufficiency mouse model. Am J Reprod Immunol. 2016;75(5):580–93.PubMedCrossRef Kadam KM, Mande PV, Gawas N, Ahire S, Khole LV. Autoantibodies to heat-shock protein, HSPA5, and epitope spreading: high-dose dexamethasone therapy rescues ovarian function in experimental autoimmune ovarian insufficiency mouse model. Am J Reprod Immunol. 2016;75(5):580–93.PubMedCrossRef
27.
Zurück zum Zitat Toivola DM, Habtezion A, Misiorek JO, Zhang L, Nystrom JH, Sharpe O, Robinson WH, Kwan R, Omary MB. Absence of keratin 8 or 18 promotes antimitochondrial autoantibody formation in aging male mice. FASEB J. 2015;29(12):5081–9.PubMedPubMedCentralCrossRef Toivola DM, Habtezion A, Misiorek JO, Zhang L, Nystrom JH, Sharpe O, Robinson WH, Kwan R, Omary MB. Absence of keratin 8 or 18 promotes antimitochondrial autoantibody formation in aging male mice. FASEB J. 2015;29(12):5081–9.PubMedPubMedCentralCrossRef
28.
Zurück zum Zitat Mayasari N, Van Knegsel AT, de Vries RG, Kemp B, Parmentier HK. Natural autoantibodies in Bos taurus calves during the first twelve weeks of life. Vet Immunol Immunopathol. 2016;178:70–8.PubMedCrossRef Mayasari N, Van Knegsel AT, de Vries RG, Kemp B, Parmentier HK. Natural autoantibodies in Bos taurus calves during the first twelve weeks of life. Vet Immunol Immunopathol. 2016;178:70–8.PubMedCrossRef
29.
Zurück zum Zitat Okubo M, Yamamoto K, Kato T, Matsuura N, Nishimaki T, Kasukawa R, Ito K, Mizushima Y, Nishioka K. Detection and epitope analysis of autoantigen-reactive T cells to the U1-small nuclear ribonucleoprotein a protein in autoimmune disease patients. J Immunol. 1993;151(2):1108–15.PubMed Okubo M, Yamamoto K, Kato T, Matsuura N, Nishimaki T, Kasukawa R, Ito K, Mizushima Y, Nishioka K. Detection and epitope analysis of autoantigen-reactive T cells to the U1-small nuclear ribonucleoprotein a protein in autoimmune disease patients. J Immunol. 1993;151(2):1108–15.PubMed
30.
Zurück zum Zitat 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.PubMedCrossRef 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.PubMedCrossRef
31.
Zurück zum Zitat Manns MP, Obermayer-Straub P. Cytochromes P450 and uridine triphosphate-glucuronosyltransferases: model autoantigens to study drug-induced, virus-induced, and autoimmune liver disease. Hepatology. 1997;26(4):1054–66.PubMedCrossRef Manns MP, Obermayer-Straub P. Cytochromes P450 and uridine triphosphate-glucuronosyltransferases: model autoantigens to study drug-induced, virus-induced, and autoimmune liver disease. Hepatology. 1997;26(4):1054–66.PubMedCrossRef
32.
Zurück zum Zitat Obermayer-Straub P, Manns MP. Cytochromes P450 and UDP-glucuronosyl-transferases as hepatocellular autoantigens. Baillieres Clin Gastroenterol. 1996;10(3):501–32.PubMedCrossRef Obermayer-Straub P, Manns MP. Cytochromes P450 and UDP-glucuronosyl-transferases as hepatocellular autoantigens. Baillieres Clin Gastroenterol. 1996;10(3):501–32.PubMedCrossRef
33.
Zurück zum Zitat Hao S, Fu R, Wang H, Shao Z. Screening novel autoantigens targeted by serum IgG autoantibodies in immunorelated pancytopenia by SEREX. Int J Hematol. 2017;106(5):622–30.PubMedCrossRef Hao S, Fu R, Wang H, Shao Z. Screening novel autoantigens targeted by serum IgG autoantibodies in immunorelated pancytopenia by SEREX. Int J Hematol. 2017;106(5):622–30.PubMedCrossRef
34.
Zurück zum Zitat Edassery SL, Shatavi SV, Kunkel JP, Hauer C, Brucker C, Penumatsa K, Yu Y, Dias JA, Luborsky JL. Autoantigens in ovarian autoimmunity associated with unexplained infertility and premature ovarian failure. Fertil Steril. 2010;94(7):2636–41.PubMedPubMedCentralCrossRef Edassery SL, Shatavi SV, Kunkel JP, Hauer C, Brucker C, Penumatsa K, Yu Y, Dias JA, Luborsky JL. Autoantigens in ovarian autoimmunity associated with unexplained infertility and premature ovarian failure. Fertil Steril. 2010;94(7):2636–41.PubMedPubMedCentralCrossRef
35.
Zurück zum Zitat Smith GC, Kenna JG, Harrison DJ, Tew D, Wolf CR. Autoantibodies to hepatic microsomal carboxylesterase in halothane hepatitis. Lancet. 1993;342(8877):963–4.PubMedCrossRef Smith GC, Kenna JG, Harrison DJ, Tew D, Wolf CR. Autoantibodies to hepatic microsomal carboxylesterase in halothane hepatitis. Lancet. 1993;342(8877):963–4.PubMedCrossRef
36.
Zurück zum Zitat Miura H, Tobe T, Nakano Y. Analysis of epitope regions for autoantibodies in catalase. Immunol Investig. 2010;39(8):796–806.CrossRef Miura H, Tobe T, Nakano Y. Analysis of epitope regions for autoantibodies in catalase. Immunol Investig. 2010;39(8):796–806.CrossRef
37.
Zurück zum Zitat D'Souza A, Kurien BT, Rodgers R, Shenoi J, Kurono S, Matsumoto H, Hensley K, Nath SK, Scofield RH. Detection of catalase as a major protein target of the lipid peroxidation product 4-HNE and the lack of its genetic association as a risk factor in SLE. BMC Med Genet. 2008;9:62.PubMedPubMedCentralCrossRef D'Souza A, Kurien BT, Rodgers R, Shenoi J, Kurono S, Matsumoto H, Hensley K, Nath SK, Scofield RH. Detection of catalase as a major protein target of the lipid peroxidation product 4-HNE and the lack of its genetic association as a risk factor in SLE. BMC Med Genet. 2008;9:62.PubMedPubMedCentralCrossRef
38.
Zurück zum Zitat Kubota T, Iizuka H, Bachovchin WW, Stollar BD. Dipeptidyl peptidase IV (DP IV) activity in serum and on lymphocytes of MRL/Mp-lpr/lpr mice correlates with disease onset. Clin Exp Immunol. 1994;96(2):292–6.PubMedPubMedCentralCrossRef Kubota T, Iizuka H, Bachovchin WW, Stollar BD. Dipeptidyl peptidase IV (DP IV) activity in serum and on lymphocytes of MRL/Mp-lpr/lpr mice correlates with disease onset. Clin Exp Immunol. 1994;96(2):292–6.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Vojdani A, Bazargan M, Vojdani E, Samadi J, Nourian AA, Eghbalieh N, Cooper EL. Heat shock protein and gliadin peptide promote development of peptidase antibodies in children with autism and patients with autoimmune disease. Clin Diagn Lab Immunol. 2004;11(3):515–24.PubMedPubMedCentral Vojdani A, Bazargan M, Vojdani E, Samadi J, Nourian AA, Eghbalieh N, Cooper EL. Heat shock protein and gliadin peptide promote development of peptidase antibodies in children with autism and patients with autoimmune disease. Clin Diagn Lab Immunol. 2004;11(3):515–24.PubMedPubMedCentral
40.
Zurück zum Zitat Shi L, Gehin T, Chevolot Y, Souteyrand E, Mange A, Solassol J, Laurenceau E. Anti-heat shock protein autoantibody profiling in breast cancer using customized protein microarray. Anal Bioanal Chem. 2016;408(5):1497–506.PubMedCrossRef Shi L, Gehin T, Chevolot Y, Souteyrand E, Mange A, Solassol J, Laurenceau E. Anti-heat shock protein autoantibody profiling in breast cancer using customized protein microarray. Anal Bioanal Chem. 2016;408(5):1497–506.PubMedCrossRef
41.
Zurück zum Zitat Chen M, Shirai M, Czaja AJ, Kurokohchi K, Arichi T, Arima K, Kodama T, Nishioka M. Characterization of anti-histone antibodies in patients with type 1 autoimmune hepatitis. J Gastroenterol Hepatol. 1998;13(5):483–9.PubMedCrossRef Chen M, Shirai M, Czaja AJ, Kurokohchi K, Arichi T, Arima K, Kodama T, Nishioka M. Characterization of anti-histone antibodies in patients with type 1 autoimmune hepatitis. J Gastroenterol Hepatol. 1998;13(5):483–9.PubMedCrossRef
42.
Zurück zum Zitat Konikoff F, Swissa M, Shoenfeld Y. Autoantibodies to histones and their subfractions in chronic liver diseases. Clin Immunol Immunopathol. 1989;51(1):77–82.PubMedCrossRef Konikoff F, Swissa M, Shoenfeld Y. Autoantibodies to histones and their subfractions in chronic liver diseases. Clin Immunol Immunopathol. 1989;51(1):77–82.PubMedCrossRef
43.
Zurück zum Zitat Khan MA, Dixit K, Uddin M, Malik A, Alam K. Role of peroxynitrite-modified H2A histone in the induction and progression of rheumatoid arthritis. Scand J Rheumatol. 2012;41(6):426–33.PubMedCrossRef Khan MA, Dixit K, Uddin M, Malik A, Alam K. Role of peroxynitrite-modified H2A histone in the induction and progression of rheumatoid arthritis. Scand J Rheumatol. 2012;41(6):426–33.PubMedCrossRef
44.
Zurück zum Zitat Xu Y, Liu AJ, Gao YX, Hu MG, Zhao GD, Zhao ZM, Liu R. Expression of Ku86 and presence of Ku86 antibody as biomarkers of hepatitis B virus related hepatocellular carcinoma. Dig Dis Sci. 2014;59(3):614–22.PubMedCrossRef Xu Y, Liu AJ, Gao YX, Hu MG, Zhao GD, Zhao ZM, Liu R. Expression of Ku86 and presence of Ku86 antibody as biomarkers of hepatitis B virus related hepatocellular carcinoma. Dig Dis Sci. 2014;59(3):614–22.PubMedCrossRef
45.
Zurück zum Zitat Mahler M, Silverman ED, Schulte-Pelkum J, Fritzler MJ. Anti-Scl-70 (topo-I) antibodies in SLE: myth or reality? Autoimmun Rev. 2010;9(11):756–60.PubMedCrossRef Mahler M, Silverman ED, Schulte-Pelkum J, Fritzler MJ. Anti-Scl-70 (topo-I) antibodies in SLE: myth or reality? Autoimmun Rev. 2010;9(11):756–60.PubMedCrossRef
46.
Zurück zum Zitat 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.PubMedCrossRef 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.PubMedCrossRef
47.
Zurück zum Zitat Lussiez V, Combe B, Graafland H, Rucheton M, Sany J. Anti-Sm and anti-RNP antibodies detected by immunoblotting in disseminated lupus erythematosus. Rev Rhum Mal Osteoartic. 1989;56(1):35–8.PubMed Lussiez V, Combe B, Graafland H, Rucheton M, Sany J. Anti-Sm and anti-RNP antibodies detected by immunoblotting in disseminated lupus erythematosus. Rev Rhum Mal Osteoartic. 1989;56(1):35–8.PubMed
48.
Zurück zum Zitat Mjelle JE, Rekvig OP, Fenton KA. Nucleosomes possess a high affinity for glomerular laminin and collagen IV and bind nephritogenic antibodies in murine lupus-like nephritis. Ann Rheum Dis. 2007;66(12):1661–8.PubMedPubMedCentralCrossRef Mjelle JE, Rekvig OP, Fenton KA. Nucleosomes possess a high affinity for glomerular laminin and collagen IV and bind nephritogenic antibodies in murine lupus-like nephritis. Ann Rheum Dis. 2007;66(12):1661–8.PubMedPubMedCentralCrossRef
49.
Zurück zum Zitat Meyer O, Kuntz D, Kahn MF, Bourgeois R, Haim T. Characterization by immunoblotting (western blot) of anti-RNP antibodies in 36 connective tissue disease patients of whom 26 had mixed connective tissue disease. Comparison with 28 lupus patients with anti-Sm antibodies. Rev Rhum Mal Osteoartic. 1989;56(1):39–44.PubMed Meyer O, Kuntz D, Kahn MF, Bourgeois R, Haim T. Characterization by immunoblotting (western blot) of anti-RNP antibodies in 36 connective tissue disease patients of whom 26 had mixed connective tissue disease. Comparison with 28 lupus patients with anti-Sm antibodies. Rev Rhum Mal Osteoartic. 1989;56(1):39–44.PubMed
50.
Zurück zum Zitat Fillit H, Shibata S, Sasaki T, Spiera H, Kerr LD, Blake M. Autoantibodies to the protein core of vascular basement membrane heparan sulfate proteoglycan in systemic lupus erythematosus. Autoimmunity. 1993;14(3):243–9.PubMedCrossRef Fillit H, Shibata S, Sasaki T, Spiera H, Kerr LD, Blake M. Autoantibodies to the protein core of vascular basement membrane heparan sulfate proteoglycan in systemic lupus erythematosus. Autoimmunity. 1993;14(3):243–9.PubMedCrossRef
51.
Zurück zum Zitat Kaburaki J, Stollar BD. Identification of human anti-DNA, anti-RNP, anti-SM, and anti-SS-A serum antibodies bearing the cross-reactive 16/6 idiotype. J Immunol. 1987;139(2):385–92.PubMed Kaburaki J, Stollar BD. Identification of human anti-DNA, anti-RNP, anti-SM, and anti-SS-A serum antibodies bearing the cross-reactive 16/6 idiotype. J Immunol. 1987;139(2):385–92.PubMed
52.
Zurück zum Zitat Preuss B, Berg C, Altenberend F, Gregor M, Stevanovic S, Klein R. Demonstration of autoantibodies to recombinant human sulphite oxidase in patients with chronic liver disorders and analysis of their clinical relevance. Clin Exp Immunol. 2007;150(2):312–21.PubMedPubMedCentralCrossRef Preuss B, Berg C, Altenberend F, Gregor M, Stevanovic S, Klein R. Demonstration of autoantibodies to recombinant human sulphite oxidase in patients with chronic liver disorders and analysis of their clinical relevance. Clin Exp Immunol. 2007;150(2):312–21.PubMedPubMedCentralCrossRef
53.
Zurück zum Zitat Zieve GW, Khusial PR. The anti-Sm immune response in autoimmunity and cell biology. Autoimmun Rev. 2003;2(5):235–40.PubMedCrossRef Zieve GW, Khusial PR. The anti-Sm immune response in autoimmunity and cell biology. Autoimmun Rev. 2003;2(5):235–40.PubMedCrossRef
54.
Zurück zum Zitat Jaekel HP, Klopsch T, Benkenstein B, Grobe N, Baldauf A, Schoessler W, Werle E. Reactivities to the Sm autoantigenic complex and the synthetic SmD1-aa83-119 peptide in systemic lupus erythematosus and other autoimmune diseases. J Autoimmun. 2001;17(4):347–54.PubMedCrossRef Jaekel HP, Klopsch T, Benkenstein B, Grobe N, Baldauf A, Schoessler W, Werle E. Reactivities to the Sm autoantigenic complex and the synthetic SmD1-aa83-119 peptide in systemic lupus erythematosus and other autoimmune diseases. J Autoimmun. 2001;17(4):347–54.PubMedCrossRef
55.
Zurück zum Zitat Kimura M, Tatsumi KI, Tada H, Ikemoto M, Fukuda Y, Kaneko A, Kato M, Hidaka Y, Amino N. Enzyme immunoassay for autoantibodies to human liver-type arginase and its clinical application. Clin Chem. 2000;46(1):112–7.PubMed Kimura M, Tatsumi KI, Tada H, Ikemoto M, Fukuda Y, Kaneko A, Kato M, Hidaka Y, Amino N. Enzyme immunoassay for autoantibodies to human liver-type arginase and its clinical application. Clin Chem. 2000;46(1):112–7.PubMed
56.
Zurück zum Zitat 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.PubMedPubMedCentralCrossRef 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.PubMedPubMedCentralCrossRef
57.
Zurück zum Zitat Reuter R, Luhrmann R. Immunization of mice with purified U1 small nuclear ribonucleoprotein (RNP) induces a pattern of antibody specificities characteristic of the anti-Sm and anti-RNP autoimmune response of patients with lupus erythematosus, as measured by monoclonal antibodies. Proc Natl Acad Sci U S A. 1986;83(22):8689–93.PubMedPubMedCentralCrossRef Reuter R, Luhrmann R. Immunization of mice with purified U1 small nuclear ribonucleoprotein (RNP) induces a pattern of antibody specificities characteristic of the anti-Sm and anti-RNP autoimmune response of patients with lupus erythematosus, as measured by monoclonal antibodies. Proc Natl Acad Sci U S A. 1986;83(22):8689–93.PubMedPubMedCentralCrossRef
58.
Zurück zum Zitat Habets WJ, de Rooij DJ, Hoet MH, van de Putte LB, van Venrooij WJ. Quantitation of anti-RNP and anti-Sm antibodies in MCTD and SLE patients by immunoblotting. Clin Exp Immunol. 1985;59(2):457–66.PubMedPubMedCentral Habets WJ, de Rooij DJ, Hoet MH, van de Putte LB, van Venrooij WJ. Quantitation of anti-RNP and anti-Sm antibodies in MCTD and SLE patients by immunoblotting. Clin Exp Immunol. 1985;59(2):457–66.PubMedPubMedCentral
59.
Zurück zum Zitat Reuter R, Appel B, Bringmann P, Rinke J, Luhrmann R. 5′-terminal caps of snRNAs are reactive with antibodies specific for 2,2,7-trimethylguanosine in whole cells and nuclear matrices. Double-label immunofluorescent studies with anti-m3G antibodies and with anti-RNP and anti-Sm autoantibodies. Exp Cell Res. 1984;154(2):548–60.PubMedCrossRef Reuter R, Appel B, Bringmann P, Rinke J, Luhrmann R. 5′-terminal caps of snRNAs are reactive with antibodies specific for 2,2,7-trimethylguanosine in whole cells and nuclear matrices. Double-label immunofluorescent studies with anti-m3G antibodies and with anti-RNP and anti-Sm autoantibodies. Exp Cell Res. 1984;154(2):548–60.PubMedCrossRef
60.
Zurück zum Zitat Marti A, Matsuda M, Sihag RK, Eliceiri GL. The small nuclear ribonucleoproteins that react with anti-Sm and anti-RNP antibodies. Biochem Biophys Res Commun. 1984;123(1):345–51.PubMedCrossRef Marti A, Matsuda M, Sihag RK, Eliceiri GL. The small nuclear ribonucleoproteins that react with anti-Sm and anti-RNP antibodies. Biochem Biophys Res Commun. 1984;123(1):345–51.PubMedCrossRef
61.
Zurück zum Zitat Dinkel K, Rickert M, Moller G, Adamski J, Meinck HM, Richter W. Stiff-man syndrome: identification of 17 beta-hydroxysteroid dehydrogenase type 4 as a novel 80-kDa antineuronal antigen. J Neuroimmunol. 2002;130(1–2):184–93.PubMedCrossRef Dinkel K, Rickert M, Moller G, Adamski J, Meinck HM, Richter W. Stiff-man syndrome: identification of 17 beta-hydroxysteroid dehydrogenase type 4 as a novel 80-kDa antineuronal antigen. J Neuroimmunol. 2002;130(1–2):184–93.PubMedCrossRef
62.
Zurück zum Zitat Kumagai T, Uchida M, Okumura N, Kameko M, Katsuyama T. Kanai M: [enzyme-linked immunosorbent assay for the detection of immunoglobulin class-specific anti-RNP and anti-Sm antibodies]. Rinsho Byori. 1988;36(6):699–704.PubMed Kumagai T, Uchida M, Okumura N, Kameko M, Katsuyama T. Kanai M: [enzyme-linked immunosorbent assay for the detection of immunoglobulin class-specific anti-RNP and anti-Sm antibodies]. Rinsho Byori. 1988;36(6):699–704.PubMed
63.
Zurück zum Zitat Hwang HM, Heo CK, Lee HJ, Kwak SS, Lim WH, Yoo JS, Yu DY, Lim KJ, Kim JY, Cho EW. Identification of anti-SF3B1 autoantibody as a diagnostic marker in patients with hepatocellular carcinoma. J Transl Med. 2018;16(1):177.PubMedPubMedCentralCrossRef Hwang HM, Heo CK, Lee HJ, Kwak SS, Lim WH, Yoo JS, Yu DY, Lim KJ, Kim JY, Cho EW. Identification of anti-SF3B1 autoantibody as a diagnostic marker in patients with hepatocellular carcinoma. J Transl Med. 2018;16(1):177.PubMedPubMedCentralCrossRef
64.
Zurück zum Zitat Okano Y, Targoff IN, Oddis CV, Fujii T, Kuwana M, Tsuzaka K, Hirakata M, Mimori T, Craft J, Medsger TA Jr. Anti-U5 small nuclear ribonucleoprotein (snRNP) antibodies: a rare anti-U snRNP specificity. Clin Immunol Immunopathol. 1996;81(1):41–7.PubMedCrossRef Okano Y, Targoff IN, Oddis CV, Fujii T, Kuwana M, Tsuzaka K, Hirakata M, Mimori T, Craft J, Medsger TA Jr. Anti-U5 small nuclear ribonucleoprotein (snRNP) antibodies: a rare anti-U snRNP specificity. Clin Immunol Immunopathol. 1996;81(1):41–7.PubMedCrossRef
65.
Zurück zum Zitat Mimori T, Hinterberger M, Pettersson I, Steitz JA. Autoantibodies to the U2 small nuclear ribonucleoprotein in a patient with scleroderma-polymyositis overlap syndrome. J Biol Chem. 1984;259(1):560–5.PubMed Mimori T, Hinterberger M, Pettersson I, Steitz JA. Autoantibodies to the U2 small nuclear ribonucleoprotein in a patient with scleroderma-polymyositis overlap syndrome. J Biol Chem. 1984;259(1):560–5.PubMed
66.
Zurück zum Zitat Lehmeier T, Foulaki K, Luhrmann R. Evidence for three distinct D proteins, which react differentially with anti-Sm autoantibodies, in the cores of the major snRNPs U1, U2, U4/U6 and U5. Nucleic Acids Res. 1990;18(22):6475–84.PubMedPubMedCentralCrossRef Lehmeier T, Foulaki K, Luhrmann R. Evidence for three distinct D proteins, which react differentially with anti-Sm autoantibodies, in the cores of the major snRNPs U1, U2, U4/U6 and U5. Nucleic Acids Res. 1990;18(22):6475–84.PubMedPubMedCentralCrossRef
67.
Zurück zum Zitat Mahesh SP, Li Z, Buggage R, Mor F, Cohen IR, Chew EY, Nussenblatt RB. Alpha tropomyosin as a self-antigen in patients with Behcet's disease. Clin Exp Immunol. 2005;140(2):368–75.PubMedPubMedCentralCrossRef Mahesh SP, Li Z, Buggage R, Mor F, Cohen IR, Chew EY, Nussenblatt RB. Alpha tropomyosin as a self-antigen in patients with Behcet's disease. Clin Exp Immunol. 2005;140(2):368–75.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Gajbhiye R, Sonawani A, Khan S, Suryawanshi A, Kadam S, Warty N, Raut V, Khole V. Identification and validation of novel serum markers for early diagnosis of endometriosis. Hum Reprod. 2012;27(2):408–17.PubMedCrossRef Gajbhiye R, Sonawani A, Khan S, Suryawanshi A, Kadam S, Warty N, Raut V, Khole V. Identification and validation of novel serum markers for early diagnosis of endometriosis. Hum Reprod. 2012;27(2):408–17.PubMedCrossRef
69.
Zurück zum Zitat Kimura A, Sakurai T, Yamada M, Koumura A, Hayashi Y, Tanaka Y, Hozumi I, Ohtaki H, Chousa M, Takemura M, et al. Anti-endothelial cell antibodies in patients with cerebral small vessel disease. Curr Neurovasc Res. 2012;9(4):296–301.PubMedCrossRef Kimura A, Sakurai T, Yamada M, Koumura A, Hayashi Y, Tanaka Y, Hozumi I, Ohtaki H, Chousa M, Takemura M, et al. Anti-endothelial cell antibodies in patients with cerebral small vessel disease. Curr Neurovasc Res. 2012;9(4):296–301.PubMedCrossRef
70.
Zurück zum Zitat Renaudineau Y, Dalekos GN, Gueguen P, Zachou K, Youinou P. Anti-alpha-actinin antibodies cross-react with anti-ssDNA antibodies in active autoimmune hepatitis. Clin Rev Allergy Immunol. 2008;34(3):321–5.PubMedCrossRef Renaudineau Y, Dalekos GN, Gueguen P, Zachou K, Youinou P. Anti-alpha-actinin antibodies cross-react with anti-ssDNA antibodies in active autoimmune hepatitis. Clin Rev Allergy Immunol. 2008;34(3):321–5.PubMedCrossRef
71.
Zurück zum Zitat van Beers JJ, Willemze A, Stammen-Vogelzangs J, Drijfhout JW, Toes RE, Pruijn GJ. Anti-citrullinated fibronectin antibodies in rheumatoid arthritis are associated with human leukocyte antigen-DRB1 shared epitope alleles. Arthritis Res Ther. 2012;14(1):R35.PubMedPubMedCentralCrossRef van Beers JJ, Willemze A, Stammen-Vogelzangs J, Drijfhout JW, Toes RE, Pruijn GJ. Anti-citrullinated fibronectin antibodies in rheumatoid arthritis are associated with human leukocyte antigen-DRB1 shared epitope alleles. Arthritis Res Ther. 2012;14(1):R35.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Orth T, Gerken G, Kellner R, Meyer zum Buschenfelde KH, Mayet WJ. Actin is a target antigen of anti-neutrophil cytoplasmic antibodies (ANCA) in autoimmune hepatitis type-1. J Hepatol. 1997;26(1):37–47.PubMedCrossRef Orth T, Gerken G, Kellner R, Meyer zum Buschenfelde KH, Mayet WJ. Actin is a target antigen of anti-neutrophil cytoplasmic antibodies (ANCA) in autoimmune hepatitis type-1. J Hepatol. 1997;26(1):37–47.PubMedCrossRef
73.
Zurück zum Zitat Caforio AL, Grazzini M, Mann JM, Keeling PJ, Bottazzo GF, McKenna WJ, Schiaffino S. Identification of alpha- and beta-cardiac myosin heavy chain isoforms as major autoantigens in dilated cardiomyopathy. Circulation. 1992;85(5):1734–42.PubMedCrossRef Caforio AL, Grazzini M, Mann JM, Keeling PJ, Bottazzo GF, McKenna WJ, Schiaffino S. Identification of alpha- and beta-cardiac myosin heavy chain isoforms as major autoantigens in dilated cardiomyopathy. Circulation. 1992;85(5):1734–42.PubMedCrossRef
74.
Zurück zum Zitat Senecal JL, Rauch J, Grodzicky T, Raynauld JP, Uthman I, Nava A, Guimond M, Raymond Y. Strong association of autoantibodies to human nuclear Lamin B1 with lupus anticoagulant antibodies in systemic lupus erythematosus. Arthritis Rheum. 1999;42(7):1347–53.PubMedCrossRef Senecal JL, Rauch J, Grodzicky T, Raynauld JP, Uthman I, Nava A, Guimond M, Raymond Y. Strong association of autoantibodies to human nuclear Lamin B1 with lupus anticoagulant antibodies in systemic lupus erythematosus. Arthritis Rheum. 1999;42(7):1347–53.PubMedCrossRef
75.
Zurück zum Zitat Garbarz M, Dhermy D, Bournier O, Bezeaud A, Boivin P. Anti-spectrin in sera containing smooth muscle autoantibodies from patients with chronic active hepatitis. Clin Exp Immunol. 1981;43(1):87–93.PubMedPubMedCentral Garbarz M, Dhermy D, Bournier O, Bezeaud A, Boivin P. Anti-spectrin in sera containing smooth muscle autoantibodies from patients with chronic active hepatitis. Clin Exp Immunol. 1981;43(1):87–93.PubMedPubMedCentral
76.
Zurück zum Zitat Bansal D, Herbert F, Lim P, Deshpande P, Becavin C, Guiyedi V, de Maria I, Rousselle JC, Namane A, Jain R, et al. IgG autoantibody to brain beta tubulin III associated with cytokine cluster-II discriminate cerebral malaria in Central India. PLoS One. 2009;4(12):e8245.PubMedPubMedCentralCrossRef Bansal D, Herbert F, Lim P, Deshpande P, Becavin C, Guiyedi V, de Maria I, Rousselle JC, Namane A, Jain R, et al. IgG autoantibody to brain beta tubulin III associated with cytokine cluster-II discriminate cerebral malaria in Central India. PLoS One. 2009;4(12):e8245.PubMedPubMedCentralCrossRef
77.
Zurück zum Zitat Le Naour F, Brichory F, Misek DE, Brechot C, Hanash SM, Beretta L. A distinct repertoire of autoantibodies in hepatocellular carcinoma identified by proteomic analysis. Mol Cell Proteomics. 2002;1(3):197–203.PubMedCrossRef Le Naour F, Brichory F, Misek DE, Brechot C, Hanash SM, Beretta L. A distinct repertoire of autoantibodies in hepatocellular carcinoma identified by proteomic analysis. Mol Cell Proteomics. 2002;1(3):197–203.PubMedCrossRef
78.
Zurück zum Zitat 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.PubMedCrossRef 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.PubMedCrossRef
79.
Zurück zum Zitat Carlsson L, Ronquist G, Nilsson BO, Larsson A. Dominant prostasome immunogens for sperm-agglutinating autoantibodies of infertile men. J Androl. 2004;25(5):699–705.PubMedCrossRef Carlsson L, Ronquist G, Nilsson BO, Larsson A. Dominant prostasome immunogens for sperm-agglutinating autoantibodies of infertile men. J Androl. 2004;25(5):699–705.PubMedCrossRef
80.
Zurück zum Zitat Dai L, Li J, Tsay JJ, Yie TA, Munger JS, Pass H, Rom WN, Tan EM, Zhang JY. Identification of autoantibodies to ECH1 and HNRNPA2B1 as potential biomarkers in the early detection of lung cancer. Oncoimmunology. 2017;6(5):e1310359.PubMedPubMedCentralCrossRef Dai L, Li J, Tsay JJ, Yie TA, Munger JS, Pass H, Rom WN, Tan EM, Zhang JY. Identification of autoantibodies to ECH1 and HNRNPA2B1 as potential biomarkers in the early detection of lung cancer. Oncoimmunology. 2017;6(5):e1310359.PubMedPubMedCentralCrossRef
81.
Zurück zum Zitat Pons C, Dansette PM, Gregeois J, Homberg JC, Billett EE, Mansuy D. Human anti-mitochondria autoantibodies appearing in iproniazid-induced immunoallergic hepatitis recognize human liver monoamine oxidase B. Biochem Biophys Res Commun. 1996;218(1):118–24.PubMedCrossRef Pons C, Dansette PM, Gregeois J, Homberg JC, Billett EE, Mansuy D. Human anti-mitochondria autoantibodies appearing in iproniazid-induced immunoallergic hepatitis recognize human liver monoamine oxidase B. Biochem Biophys Res Commun. 1996;218(1):118–24.PubMedCrossRef
82.
Zurück zum Zitat Kemp EH, Ridgway JN, Smith KA, Watson PF, Weetman AP. Autoantibodies to the flavoprotein subunit of succinate dehydrogenase: analysis of specificity in autoimmune thyroid disease. Clin Endocrinol. 2000;53(3):291–9.CrossRef Kemp EH, Ridgway JN, Smith KA, Watson PF, Weetman AP. Autoantibodies to the flavoprotein subunit of succinate dehydrogenase: analysis of specificity in autoimmune thyroid disease. Clin Endocrinol. 2000;53(3):291–9.CrossRef
83.
Zurück zum Zitat Loeper J, Descatoire V, Maurice M, Beaune P, Belghiti J, Houssin D, Ballet F, Feldmann G, Guengerich FP, Pessayre D. Cytochromes P-450 in human hepatocyte plasma membrane: recognition by several autoantibodies. Gastroenterology. 1993;104(1):203–16.PubMedCrossRef Loeper J, Descatoire V, Maurice M, Beaune P, Belghiti J, Houssin D, Ballet F, Feldmann G, Guengerich FP, Pessayre D. Cytochromes P-450 in human hepatocyte plasma membrane: recognition by several autoantibodies. Gastroenterology. 1993;104(1):203–16.PubMedCrossRef
84.
Zurück zum Zitat Cicek G, Vuorinen T, Stahle I, Stepanek P, Freudenberg N, Brandsch R. Coxsackievirus B3 infection induces anti-flavoprotein antibodies in mice. Clin Exp Immunol. 2000;122(3):404–9.PubMedPubMedCentralCrossRef Cicek G, Vuorinen T, Stahle I, Stepanek P, Freudenberg N, Brandsch R. Coxsackievirus B3 infection induces anti-flavoprotein antibodies in mice. Clin Exp Immunol. 2000;122(3):404–9.PubMedPubMedCentralCrossRef
85.
Zurück zum Zitat Seko Y, Matsumoto A, Fukuda T, Imai Y, Fujimura T, Taka H, Mineki R, Murayama K, Hirata Y, Nagai R. A case of neonatal lupus erythematosus presenting delayed dilated cardiomyopathy with circulating autoantibody to annexin A6. Int Heart J. 2007;48(3):407–15.PubMedCrossRef Seko Y, Matsumoto A, Fukuda T, Imai Y, Fujimura T, Taka H, Mineki R, Murayama K, Hirata Y, Nagai R. A case of neonatal lupus erythematosus presenting delayed dilated cardiomyopathy with circulating autoantibody to annexin A6. Int Heart J. 2007;48(3):407–15.PubMedCrossRef
86.
Zurück zum Zitat Chakravarti R, Gupta K, Swain M, Willard B, Scholtz J, Svensson LG, Roselli EE, Pettersson G, Johnston DR, Soltesz EG, et al. 14-3-3 in thoracic aortic aneurysms: identification of a novel autoantigen in large vessel Vasculitis. Arthritis Rheumatol. 2015;67(7):1913–21.PubMedPubMedCentralCrossRef Chakravarti R, Gupta K, Swain M, Willard B, Scholtz J, Svensson LG, Roselli EE, Pettersson G, Johnston DR, Soltesz EG, et al. 14-3-3 in thoracic aortic aneurysms: identification of a novel autoantigen in large vessel Vasculitis. Arthritis Rheumatol. 2015;67(7):1913–21.PubMedPubMedCentralCrossRef
87.
Zurück zum Zitat Kistner A, Bigler MB, Glatz K, Egli SB, Baldin FS, Marquardsen FA, Mehling M, Rentsch KM, Staub D, Aschwanden M, 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.PubMed Kistner A, Bigler MB, Glatz K, Egli SB, Baldin FS, Marquardsen FA, Mehling M, Rentsch KM, Staub D, Aschwanden M, 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.PubMed
88.
Zurück zum Zitat Feist E, Kuckelkorn U, Dorner T, Donitz H, Scheffler S, Hiepe F, Kloetzel PM, Burmester GR. Autoantibodies in primary Sjogren's syndrome are directed against proteasomal subunits of the alpha and beta type. Arthritis Rheum. 1999;42(4):697–702.PubMedCrossRef Feist E, Kuckelkorn U, Dorner T, Donitz H, Scheffler S, Hiepe F, Kloetzel PM, Burmester GR. Autoantibodies in primary Sjogren's syndrome are directed against proteasomal subunits of the alpha and beta type. Arthritis Rheum. 1999;42(4):697–702.PubMedCrossRef
89.
Zurück zum Zitat Feist E, Dorner T, Kuckelkorn U, Schmidtke G, Micheel B, Hiepe F, Burmester GR, Kloetzel PM. Proteasome alpha-type subunit C9 is a primary target of autoantibodies in sera of patients with myositis and systemic lupus erythematosus. J Exp Med. 1996;184(4):1313–8.PubMedCrossRef Feist E, Dorner T, Kuckelkorn U, Schmidtke G, Micheel B, Hiepe F, Burmester GR, Kloetzel PM. Proteasome alpha-type subunit C9 is a primary target of autoantibodies in sera of patients with myositis and systemic lupus erythematosus. J Exp Med. 1996;184(4):1313–8.PubMedCrossRef
90.
Zurück zum Zitat Fierabracci A, Saura F. Identification of a common autoantigenic epitope of protein disulfide isomerase, golgin-160 and voltage-gated potassium channel in type 1 diabetes. Diabetes Res Clin Pract. 2010;88(2):e14–6.PubMedCrossRef Fierabracci A, Saura F. Identification of a common autoantigenic epitope of protein disulfide isomerase, golgin-160 and voltage-gated potassium channel in type 1 diabetes. Diabetes Res Clin Pract. 2010;88(2):e14–6.PubMedCrossRef
91.
Zurück zum Zitat Weber CK, Haslbeck M, Englbrecht M, Sehnert B, Mielenz D, Graef D, Distler JH, Mueller RB, Burkhardt H, Schett G, 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.CrossRef Weber CK, Haslbeck M, Englbrecht M, Sehnert B, Mielenz D, Graef D, Distler JH, Mueller RB, Burkhardt H, Schett G, 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.CrossRef
92.
Zurück zum Zitat Miyachi K, Hosaka H, Nakamura N, Miyakawa H, Mimori T, Shibata M, Matsushima S, Chinoh H, Horigome T, Hankins RW, et al. Anti-p97/VCP antibodies: an autoantibody marker for a subset of primary biliary cirrhosis patients with milder disease? Scand J Immunol. 2006;63(5):376–82.PubMedCrossRef Miyachi K, Hosaka H, Nakamura N, Miyakawa H, Mimori T, Shibata M, Matsushima S, Chinoh H, Horigome T, Hankins RW, et al. Anti-p97/VCP antibodies: an autoantibody marker for a subset of primary biliary cirrhosis patients with milder disease? Scand J Immunol. 2006;63(5):376–82.PubMedCrossRef
93.
Zurück zum Zitat Kobayashi S, Hiwasa T, Arasawa T, Kagaya A, Ishii S, Shimada H, Ito M, Suzuki M, Kano M, Rahmutulla B, et al. Identification of specific and common diagnostic antibody markers for gastrointestinal cancers by SEREX screening using testis cDNA phage library. Oncotarget. 2018;9(26):18559–69.PubMedPubMedCentralCrossRef Kobayashi S, Hiwasa T, Arasawa T, Kagaya A, Ishii S, Shimada H, Ito M, Suzuki M, Kano M, Rahmutulla B, et al. Identification of specific and common diagnostic antibody markers for gastrointestinal cancers by SEREX screening using testis cDNA phage library. Oncotarget. 2018;9(26):18559–69.PubMedPubMedCentralCrossRef
94.
Zurück zum Zitat Liu M, Subramanian V, Christie C, Castro M, Mohanakumar T. Immune responses to self-antigens in asthma patients: clinical and immunopathological implications. Hum Immunol. 2012;73(5):511–6.PubMedPubMedCentralCrossRef Liu M, Subramanian V, Christie C, Castro M, Mohanakumar T. Immune responses to self-antigens in asthma patients: clinical and immunopathological implications. Hum Immunol. 2012;73(5):511–6.PubMedPubMedCentralCrossRef
95.
Zurück zum Zitat Papiha SS, Pal B. Gc (vitamin D binding protein) subtypes in rheumatoid arthritis. Hum Genet. 1985;70(3):278–80.PubMedCrossRef Papiha SS, Pal B. Gc (vitamin D binding protein) subtypes in rheumatoid arthritis. Hum Genet. 1985;70(3):278–80.PubMedCrossRef
96.
Zurück zum Zitat Van den Bergh K, Vercammen M, Regenass S, Derua R, Vermeersch P, Pokreisz P, Ocmant A, de Beeck KO, Janssens S, Waelkens E, et al. Betaine homocysteine methyl transferase 1, a novel auto-antigen associated with anti-Golgi immune reactivity. Clin Chim Acta. 2012;413(1–2):105–8.PubMedCrossRef Van den Bergh K, Vercammen M, Regenass S, Derua R, Vermeersch P, Pokreisz P, Ocmant A, de Beeck KO, Janssens S, Waelkens E, et al. Betaine homocysteine methyl transferase 1, a novel auto-antigen associated with anti-Golgi immune reactivity. Clin Chim Acta. 2012;413(1–2):105–8.PubMedCrossRef
97.
Zurück zum Zitat Vasilev VV, Noe R, Dragon-Durey MA, Chauvet S, Lazarov VJ, Deliyska BP, Fremeaux-Bacchi V, Dimitrov JD, Roumenina LT. Functional characterization of autoantibodies against complement component C3 in patients with lupus nephritis. J Biol Chem. 2015;290(42):25343–55.PubMedPubMedCentralCrossRef Vasilev VV, Noe R, Dragon-Durey MA, Chauvet S, Lazarov VJ, Deliyska BP, Fremeaux-Bacchi V, Dimitrov JD, Roumenina LT. Functional characterization of autoantibodies against complement component C3 in patients with lupus nephritis. J Biol Chem. 2015;290(42):25343–55.PubMedPubMedCentralCrossRef
98.
Zurück zum Zitat Arnett FC, Reichlin M. Lupus hepatitis: an under-recognized disease feature associated with autoantibodies to ribosomal P. Am J Med. 1995;99(5):465–72.PubMedCrossRef Arnett FC, Reichlin M. Lupus hepatitis: an under-recognized disease feature associated with autoantibodies to ribosomal P. Am J Med. 1995;99(5):465–72.PubMedCrossRef
99.
Zurück zum Zitat Yamagiwa S, Kamimura H, Takamura M, Aoyagi Y. Autoantibodies in primary biliary cirrhosis: recent progress in research on the pathogenetic and clinical significance. World J Gastroenterol. 2014;20(10):2606–12.PubMedPubMedCentralCrossRef Yamagiwa S, Kamimura H, Takamura M, Aoyagi Y. Autoantibodies in primary biliary cirrhosis: recent progress in research on the pathogenetic and clinical significance. World J Gastroenterol. 2014;20(10):2606–12.PubMedPubMedCentralCrossRef
100.
Zurück zum Zitat Ehser J, Holdener M, Christen S, Bayer M, Pfeilschifter JM, Hintermann E, Bogdanos D, Christen U. Molecular mimicry rather than identity breaks T-cell tolerance in the CYP2D6 mouse model for human autoimmune hepatitis. J Autoimmun. 2013;42:39–49.PubMedCrossRef Ehser J, Holdener M, Christen S, Bayer M, Pfeilschifter JM, Hintermann E, Bogdanos D, Christen U. Molecular mimicry rather than identity breaks T-cell tolerance in the CYP2D6 mouse model for human autoimmune hepatitis. J Autoimmun. 2013;42:39–49.PubMedCrossRef
Metadaten
Titel
A comprehensive autoantigen-ome of autoimmune liver diseases identified from dermatan sulfate affinity enrichment of liver tissue proteins
verfasst von
Wei Zhang
Jung-hyun Rho
Michael H. Roehrl
Julia Y. Wang
Publikationsdatum
01.12.2019
Verlag
BioMed Central
Erschienen in
BMC Immunology / Ausgabe 1/2019
Elektronische ISSN: 1471-2172
DOI
https://doi.org/10.1186/s12865-019-0304-1

Weitere Artikel der Ausgabe 1/2019

BMC Immunology 1/2019 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.