REVIEW ARTICLES

Transferul transplacentar al autoanticorpilor antinucleari materni în lupusul neonatal – perspectiva obstetricianului

 Transplacental transfer of maternal antinuclear autoantibodies in neonatal lupus – the obstetrician’s perspective

First published: 20 decembrie 2021

Editorial Group: MEDICHUB MEDIA

DOI: 10.26416/ObsGin.69.4.2021.5778

Abstract

Neonatal lupus erythematosus (LEN) is an autoimmune disease caused by the transplacental transfer of autoantibodies to the fetus. An obstetrician can be confronted with mothers presenting antinuclear antibodies or fetal congenital heart block during pregnancy. The pathogenesis of neonatal lupus is not fully elucidated; however, the onset of the disease is related to the passive transplacental passage of maternal antinuclear autoantibodies, especially anti-Ro/SS-A and anti-La/SS-B. In this article, we provide a glimpse of the transport mechanism of maternal IgG class antibodies through the human placental and of the effects of transplacental transfer of maternal antinuclear autoantibodies in neonatal lupus. Good knowledge of pathogenic mechanisms and identifying patients at risk of neonatal lupus, with a complex interdisciplinary approach, may lead to better outcomes. 

Keywords
transplacental mechanism, anti-Ro/SS-A antibodies, anti-La/SS-B antibodies, neonatal lupus

Rezumat

Lupusul eritematos neonatal (LEN) este o boală autoimună cauzată de transferul transplacentar de autoanticorpi către făt. Un obstetrician poate fi confruntat cu gravide care prezintă anticorpi antinucleari sau poate diagnostica un bloc cardiac congenital fetal în timpul sarcinii. Patogenia lupusului neonatal nu este pe deplin elucidată; totuşi, debutul bolii este legat de trecerea transplacentară pasivă a autoanticorpilor antinucleari materni, în special anti-Ro/SS-A şi anti-La/SS-B. În acest articol, ne propunem să oferim o privire asupra mecanismului de transport al anticorpilor materni din clasa IgG prin placenta umană şi asupra efectelor transferului transplacentar al autoanticorpilor antinucleari materni în lupusul neonatal. O bună cunoaştere a mecanismelor patogene şi identificarea pacienţilor cu risc de lupus neonatal, asociate cu o abordare interdisciplinară complexă, pot duce la rezultate mai bune.

Introduction

Neonatal lupus erythematosus (LEN) is an autoim­mune disease caused by the transplacental transfer of autoantibodies to the fetus, resulting in characteristic clinical manifestations, mainly cardiac and cutaneous. The incidence rate of LEN is about 1 in 12,500 to 20,000 live births, being a little higher in females and premature babies(1). However, the true prevalence is not established due to a significant proportion of undetected cases.

The pathogenesis of neonatal lupus is not fully elucidated; however, the onset of the disease is related to the passive transplacental transfer of maternal antinuclear autoantibodies. This group of antibodies is mainly directed against several extractable nuclear antigens (ENAs): Sm, RNP, SS-A/Ro, SS-B/La, Scl-70, Jo-1, but those that have key roles in the pathogenesis of neonatal lupus are anti-Ro/SS-A, anti-La/SS-B and anti-RNP antibodies(2). Autoimmune congenital heart block (CHB) occurs in 2% of anti-Ro/SS-A-exposed pregnancies, and the recurrence rate is nine times higher in subsequent pregnancies(3).

In this article, we provide a glimpse of the transport mechanism of maternal IgG class antibodies through the human placental and of the effects of transplacental transfer of maternal antinuclear autoantibodies in neonatal lupus.

Transplacental transport of antinuclear autoantibodies and their pathogenic role

The onset of neonatal lupus is related to antibodies transferred from the mother and belonging to the class of immunoglobulins G, in which four subclasses are dif­ferentiated: IgG1, IgG2, IgG3 and IgG4. IgG antibodies are transported by transcytosis with the involvement of neonatal receptors for the Fc fragments of IgG antibodies, present on placental trophoblast cells. The neonatal Fc receptor (FcRn) is a heterodimeric glycoprotein with a structure similar to that of major histocompatibility complex (MHC) but, unlike MHC class I, FcRn is unable to bind peptides (antigens); however, it interacts with the Fc IgG and albumin. The FcRn-IgG interaction and the FcRn-albumin interaction take place in an acidic environment (optimal at a pH of 5-6.5) and not in a physiological environment. The initial hypothesis reported on the IgG transport was focused on the placenta, and later additional data on receptors were collected(4,5).

The mechanism of transport of maternal IgG class antibodies through the human placental syncytiotrophoblast begins with the absorption of IgG into the cell by liquid-phase endocytosis. Then, in the acidic endosomal environment, IgG molecules are bound to FcRn and are thus protected against degradation by lysosomal enzymes. FcRn-IgG complexes are transferred inside the chorionic villous stroma, while IgG molecules not bound to FcRn are directed to the degradation pathway. FcRn releases IgG into the chorionic villous stroma (pH 7.4) and, after IgG dissociation, the FcRn receptor returns to the surface of the apical syncytiotrophoblast. As a result of this mechanism, fetal serum antibodies can reach slightly higher concentrations than maternal serum, as it is the case with the IgG1 subclass, while for the IgG2 subclass the fetal serum antibody concentration may be lower, and the levels of IgG3 and IgG4 in maternal and fetal serum are equal(6).

Anti-SSA autoantibodies are anti-Sjögren’s-syndrome-related antigen A autoantibodies, also known as anti-Ro, similar names including anti-SSA/Ro, anti-Ro/SSA, anti-SS-A/Ro, and anti-Ro/SS-A. Anti-Ro antibodies transferred via placenta are involved in the pathogenic mechanisms of the CHB. These autoantibodies can link cross-reactive epitopes represented by calcium regulatory molecules, such as L and T type ion channels, in the fetal heart(7). Altered calcium homeostasis and cardiac cell apoptosis are pathogenic consequences.

Anti-Ro60 and anti-La autoantibodies can bind similar antigens on the surface of apoptotic cells, this triggering an inflammatory process. This stage is probably associated with the prolongation of AV conduction(8). Depending on the presence of fetal susceptibility genes, local inflammation may be resolved with the normalization of fetal AV conduction or may be amplified and persist, leading to processes of fibrosis and calcification, with the development of permanent fetal AV block. HLA-DRB1*04 and HLA-Cw*05 alleles are associated with a higher risk of CHB, while DRB1*13 and Cw*06 appear to have a protective effect(9).

In addition to the inflammatory process generated by transplacental anti-Ro autoantibodies, apoptotic cardiomyocytes, complement C4 fraction deposition, calcification and fibrosis processes(10,11) and macro­phage activation with the release of proinflammatory and profibrotic cytokines such as TNFa and TGFb are also involved in the pathogenic mechanisms(12,13).

Siglec-1-positive macrophages are detected in fetal heart lesions with CHB(14), the expression of the sialic acid binding immunoglobulin-like lectin Siglec-1 being increased by type 1 interferon which upregulates Ro52 and stimulates apoptosis(15).

Another possible cardiological complication related to maternal anti-SS-A antibodies is endocardial fibroelastosis, its severe consequence being represented by dilated cardiomyopathy(2).

Anti-SS-A antibodies – also named anti-Ro after the patient’s name (Rose) in which they were initially detected – are directed against four different antigens, each consisting of a complex of low-molecular-weight RNA (micro-RNA) and proteins, with a molecular weight of 45, 52, 54 and 60 kDa, respectively. Only antibodies to SS-A/Ro52 and SS-A/Ro60 molecules are used in current medical practice, although older studies do not distinguish between Ro52 and Ro60 antibodies, defining them together as anti-SS-A/Ro(2,16).

Ro60 is described as an RNA-binding protein(17), while the Ro52 protein is a ubiquitin E3 ligase, which targets cytosolic virus-antibody complexes(18).

Anti-Ro52 antibodies are directed against the Ro52 antigen belonging to the tripartite motif protein TRIM21 receptor family and E3 ubiquitin ligase family. It has a role in the ubiquitination of proteins, initiating proinflammatory activities and cellular apoptosis(19-22).

After it is stimulated by interferon and binds to TLR (Toll receptors), the TRIM21 receptor also interacts with the transcription factors for the interferon regulatory factor. Substrates reported for Ro52 replication include IRF3, IRF5, IRF7 and IRF8, and these transcription factors regulate the level of type 1 interferon and cytokine production(19).

In addition, Ro52/TRIM21 may regulate T cell activation or proliferation, and overexpression of this receptor may increase the IL-2 synthesis(23).

There is a relationship between anti-Ro52 antibodies and late-onset systemic lupus erythematosus, with photo­sensitivity and hematological disorders(24).

The most significant clinical correlation of anti-Ro52 antibodies is with CHB. When analyzing the serological profile of mothers of infants with CHB, it was found that 95% of them have an increased titer of anti-Ro52 antibodies, and the frequency of Ro60 and La antibodies is 63% and 58%, respectively(25).

Anti-Ro52 antibodies are also found in other condi­tions, including interstitial lung disease(26), primary biliary cirrhosis and autoimmune hepatitis(27).

A sensitive and specific method for these autoantibodies detection is the enzyme-linked immunosorbent assay (ELISA), but the indirect immunofluorescence (IIF) method can also be used. Line-blot immunoassay (LIA) is also suitable for routine evaluation of autoantibodies to extractable nuclear antigens(16,28-31).

Antibodies to the anti-Ro52 p200 epitope (anti-amino acid antibodies in positions 200-239 of the Ro52 antigen) are most likely capable of causing fetal CHB and have been shown to be a high-risk factor for cardiac dam­age. ELISA may be used to detect antibodies binding the p200 peptide. In combination with fetal Doppler echocardiography, the determination of Ro52-p200 antibody levels may prove a valuable clinical tool to identify pregnancies where the risk for CHB is high, and allow the treatment before the condition has progressed into a complete AV block(32).

Anti-Ro60 antibodies are directed against Ro60, a protein with a molecular weight of 60 kD, consisting of two domains, one similar in structure to the von Willebrand factor and participating in cell adhesion, and the other being an alpha helix structure responsible for binding nucleotide acids(33).

The Ro60 antigen is a protein in the hY-RNA complex, with a role in RNA degradation(2,33)

Initially, molecular mimicry between nuclear antigen 1 of Epstein-Barr virus (EBNA1) and Ro60 protein was thought to be involved in the production of anti-Ro60 antibodies, but significant amino acid homologies were not found(2,34).

Subsequently, molecular mimicry with the Coxsackie virus protein was discussed in patients with Sjögren’s syndrome(35).

A new hypothesis revealed that the proteins of some commensal bacteria might contain epitopes that mimic regions of the human Ro60. A von Willebrand factor A (vWFA) peptide fraction is associated with the microorganism Capnocytophaga ochracea in the oral cavity and it is a potent activator of Ro60-reactive T cells. Another stimulating factor is the bacterium Escherichia coli which expresses vWFA(36).

These results indicate that commensal peptides can activate Ro60-reactive T cells(37).

Some species of human commensal bacteria that colonize the skin, mouth and intestines, namely Corynebacterium, Propionibacterium and Bacteroides, encode Ro60 orthologs with sequence similarities to human Ro60(38).

The salivary microbiome may be associated with the development of autoreactivity. The comparison of the von Willebrand factor domains present in human Ro60 with Lautropia mirabilis proteins also supports the molecular mimicry hypothesis(39).

Anti-SS-B antibodies, also called anti-La (anti-Lane antibodies) or, in combination, anti-La/SS-B, are targeted against the 48 kDa molecular weight phosphoprotein associated with RNA polymerase III(40).

They are present in patients with Sjögren’s syndrome and in patients with systemic lupus erythematosus (10-20%). In Sjögren’s syndrome, antibodies against La/SS-B are almost always present together with antibodies against Ro/SS-A(41).

The presence of anti-La antibodies in the absence of anti-Ro antibodies is very unusual, and cases of CHB associated with anti-La antibody positivity alone represent less than 1% of the incidence of autoimmune congenital heart blocks in the scientific literature(42)

Anti-RNP antibodies are directed against the RNP antigen, which belongs to a group of small nuclear ribonucleoproteins (snRNPs) that contain high uridine RNA (U-RNA) and various 70 kDa (U1) molecular weight base proteins, 33 kDa (protein A) and 22 kDa (protein C). Antibodies against nuclear ribonucleoprotein (nRNP) may be detected by indirect immunofluorescence (antigen-tissue substrates and HEp 2 cells). For a more accurate identification, ELISA and Western Blot methods are used.

Anti-U1-snRNP antibodies are detected in high titers in 95% to 100% of patients with mixed connective tissue disease. These autoantibodies may also be present in patients with systemic lupus erythematosus, rheumatoid arthritis or Sjögren’s syndrome, but in such patients they do not correlate with the disease activity(41).

Mothers with anti-Ro, anti-La antibodies or in some cases with anti-RNP antibodies may develop rheumatic diseases (systemic lupus erythematosus, Sjögren’s syndrome, rheumatoid arthritis), or have minimal symptoms (photosensitivity, Raynaud’s syndrome, myalgia, artalgia), or may be asymptomatic, being investigated after the onset of lupus in the newborn. The probability of developing lupus erythematosus within 10 years after the birth of a child with neonatal lupus was calculated at 18.6%, and the probability of developing Sjogren’s syndrome in such a patient is 27.9%(43).

Because most women with antibodies have children who do not develop neonatal lupus, it is believed that there are a number of factors, most likely genetic or environmental, necessary for the onset of neonatal lupus. A correlation was observed between the specificity and the level of antibodies from the mother crossing the placental barrier and the risk of developing fetal CHB(1,44).

Regarding the clinical manifestations, it is important to underline that some are reversible, such as skin, hematological, hepatic, neurological impairment(45-48), but some are irreversible, such as the heart lesions(49,50).

Cutaneous manifestations are photosensitivity and transient rash, erythematous-squamous or erythematous-annular lesions, which may occur from birth or in the first weeks of life. Periorbital erythema – called “raccoon’s eye” or “owl’s eye” – is more common(51).

From a histological point of view, the lesions are similar to those of subacute lupus, with hyperkeratosis, atrophy, basal degeneration and intracellular edema. Skin lesions persist during the elimination of circulating maternal antibodies and usually resolve spontaneously, without leaving scars, and therefore do not require drug treatment(52,53).

The spectrum of cardiac abnormalities includes conduction disorders (transient arrhythmias, grade I, II or III atrioventricular block) and cardiomyopathies (dilated cardiomyopathy, endocardial fibroelastosis)(54,55).

The specific heart disorder in neonatal lupus is the CHB, considered the most severe clinical manifesta­tion due to irreversibility and increased mortality and morbidity. A congenital atrioventricular block occurs between 18 and 24 weeks of gestation in the absence of other structural cardiac abnormalities, and may present as grade I, II and especially III AV block(37,56,57).

Autopsies data reveal areas of fibrosis and calcifications at the atrioventricular node, which is an additional argument for the irreversibility of heart damage in severe cases(59,60).

The negative prognostic factors in the evolution of CHB include: low gestational age (<20 weeks) at the onset of symptoms, generalized fetal edema, cardiomyopathy, fibroelastosis, decreased heart rate (<50 bpm) and impaired left ventricular function(61,62).

In utero exposure to maternal Ro/La autoantibodies may lead to fetal CHB, recent studies indicating interferon (IFN) activation in fibroblasts of the fetal heart. In both maternal and neonatal peripheral blood mononuclear cells, IFN-a production is induced by anti-Ro/La positive plasma.

Increased expression of IFN-regulated genes and elevated plasma IFN-a levels are revealed not only in anti-Ro/La positive women, but also in their newborns. The IFN scores of neonates (calculated using 13 IFN-regulated genes) born to mothers receiving immunomodulatory treatment are similar to those of controls, despite maternal persistent IFN activation(63).

Maternal autoantibodies biomarkers can identify women at a higher risk for delivering a child with CHB. The combination of anti-Ro52 and anti-Ro60 antibodies, especially in high levels, could recognize women at risk for fetal autoimmune CHB. Whether anti-p200 antibody should be tested as biomarker of CHB, over standard commercial ELISA, is still debated. The PR interval measurement by weekly fetal echocardiogram from 16 to at least 24 weeks of gestation is strongly recommended for CHB prenatal diagnosis. Some limitations are due to the difficult identification of first-degree atrioventricular block and to the possible occurrence of a complete block from a normal rhythm in only few days. Maternal pharmacological prophylaxis with hydroxychloroquine from the tenth week of gestation has an important role in preventing CHB in pregnant women with high risk for recurrent CHB(3).

Conclusions

The good knowledge of pathogenic mechanisms, the identification of patients at risk for neonatal lupus, the adequate screening for pregnant women with risk factors, together with an interdisciplinary collaboration, involving obstetricians, pediatricians, pediatric cardiologist and neonatologist, are important for the optimal care of these patients.   n

 

Conflict of interests: The authors declare no conflict of interests.

Bibliografie

  1. Wisuthsarewong W, Soongswang J, Chantorn R. Neonatal lupus erythematosus: clinical character, investigation, and outcome. Pediatr Dermatol. 2011 Mar-Apr;28(2):115-21.
  2. Gryka-Marton M, Szukiewicz D, Teliga-Czajkowska J, Olesinska M. An Overview of Neonatal Lupus with Anti-Ro Characteristics. Int J Mol Sci. 2021 Aug 27;22(17):9281.
  3. De Carolis S, Garufi C, Garufi E, et al. Autoimmune Congenital Heart Block: A Review of Biomarkers and Management of Pregnancy. Front Pediatr. 2020;8:607515.
  4. Brambell FW. The transmission of immunity from mother to young and the catabolism of immunoglobulins. Lancet. 1966 Nov 19;2(7473):1087-93. 
  5. Brambell FW, Hemmings WA, Morris IG. A theoretical model of gamma-globulin catabolism. Nature. 1964 Sep 26;203:1352-4.
  6. Kristoffersen EK. Placental Fc receptors and the transfer of maternal IgG. Transfus Med Rev. 2000 Jul;14(3):234-43.
  7. Xiao G-Q, Hu K, Boutjdir M. Direct Inhibition of Expressed Cardiac L- and T-Type Calcium Channels by IgG from Mothers Whose Children Have Congenital Heart Block. Circulation. 2001;103:1599–1604.
  8. Ambrosi A, Sonesson S-E, Wahren-Herlenius M. Molecular mechanisms of congenital heart block. Exp Cell Res. 2014;325:2–9.
  9. Meisgen S, Östberg T, Salomonsson S, Ding B, Eliasson H, Malarstig A, Alfredsson L, Klareskog L, Hamsten A, Olsson T, et al. The HLA locus contains novel foetal susceptibility alleles for congenital heart block with significant paternal influence. J Intern Med. 2014;275:640–651.
  10. Clancy RM, Kapur RP, Molad Y, Askanase AD, Buyon JP. Immunohistologic evidence supports apoptosis, IgG deposition, and novel macrophage/fibroblast crosstalk in the pathologic cascade leading to congenital heart block. Arthritis Rheum. 2004;50:173–182.
  11. Llanos C, Friedman DM, Saxena A, Izmirly P, Tseng C-E, Dische R, Abellar RG, Halushka M, Clancy RM, Buyon J. Anatomical and pathological findings in hearts from fetuses and infants with cardiac manifestations of neonatal lupus. Rheumatology. 2012;51:1086–1092.
  12. Miranda-Carús M-E, Askanase AD, Clancy RM, Di Donato F, Chou T-M, Libera MR, Chan EKL, Buyon JP. Anti-SSA/Ro and Anti-SSB/La Autoantibodies Bind the Surface of Apoptotic Fetal Cardiocytes and Promote Secretion of TNF-α by Macrophages. J Immunol. 2000;165:5345–5351. 
  13. Clancy RM, Askanase AD, Kapur RP, Chiopelas E, Azar N, Miranda-Carus ME, Buyon JP. Transdifferentiation of cardiac fibroblasts, a fetal factor in anti-SSA/Ro-SSB/La antibody-mediated congenital heart block. J Immunol. 2002;169:2156–2163.
  14. Clancy RM, Halushka M, Rasmussen S, Lhakhang T, Chang M, Buyon J. Siglec-1 Macrophages and the Contribution of IFN to the Development of Autoimmune Congenital Heart Block. J Immunol. 2018;202:48–55.
  15. Strandberg L, Ambrosi A, Espinosa A, Ottosson L, Eloranta M-L, Zhou W, Elfving A, Greenfield E, Kuchroo VK, Wahren-Herlenius M. Interferon-α Induces Up-regulation and Nuclear Translocation of the Ro52 Autoantigen as Detected by a Panel of Novel Ro52-specific Monoclonal Antibodies. J Clin Immunol. 2007;28:220–231.
  16. Defendenti C, Atzeni F, Spina MF, Grosso S, Cereda A, Guercilena G, Bollani S, Saibeni S, Puttini PS. Clinical and laboratory aspects of Ro/SSA-52 autoantibodies. Autoimmun Rev. 2011 Jan;10(3):150-4.
  17. Wolin SL, Steitz JA. The Ro small cytoplasmic ribonucleoproteins: identification of the antigenic protein and its binding site on the Ro RNAs. Proc Natl Acad Sci USA. 1984 Apr;81(7):1996-2000.
  18. Ben-Chetrit E, Chan EK, Sullivan KF, Tan EM. A 52-kD protein is a novel component of the SS-A/Ro antigenic particle. J Exp Med. 1988 May 1;167(5):1560-71.
  19. Oke V, Wahren-Herlenius M. The immunobiology of Ro52 (TRIM21) in autoimmunity: a critical review. J Autoimmun. 2012 Aug;39(1-2):77-82.
  20. Zhang Z, Bao M, Lu N, Weng L, Yuan B, Liu YJ. The E3 ubiquitin ligase TRIM21 negatively regulates the innate immune response to intracellular double-stranded DNA. Nat Immunol. 2013 Feb;14(2):172-8.
  21. Liu Y, Xu M, Min X, Wu K, Zhang T, Li K, Xiao S, Xia Y. TWEAK/Fn14 Activation Participates in Ro52-Mediated Photosensitization in Cutaneous Lupus Erythematosus. Front Immunol. 2017 May 31;8:651.
  22. Espinosa A, Dardalhon V, Brauner S, Ambrosi A, Higgs R, Quintana FJ, Sjöstrand M, Eloranta ML, Ní Gabhann J, Winqvist O, Sundelin B, Jefferies CA, Rozell B, Kuchroo VK, Wahren-Herlenius M. Loss of the lupus autoantigen Ro52/Trim21 induces tissue inflammation and systemic autoimmunity by disregulating the IL-23-Th17 pathway. J Exp Med. 2009 Aug 3;206(8):1661-71.
  23. Ishii T, Ohnuma K, Murakami A, Takasawa N, Yamochi T, Iwata S, Uchiyama M, Dang NH, Tanaka H, Morimoto C. SS-A/Ro52, an autoantigen involved in CD28-mediated IL-2 production. J Immunol. 2003 Apr 1;170(7):3653-61.
  24. Yoshimi R, Ueda A, Ozato K, Ishigatsubo Y. Clinical and pathological roles of Ro/SSA autoantibody system. Clin Dev Immunol. 2012;2012:606195.
  25. Salomonsson S, Dzikaite V, Zeffer E, Eliasson H, Ambrosi A, Bergman G, Fernlund E, Theander E, Ohman A, Rydberg A, Skogh T, Wållberg-Jonsson S, Elfving A, Fored M, Ekbom A, Lundström U, Mellander M, Winqvist O, Sonesson SE, Gadler F, Jonzon A, Wahren-Herlenius M. A population-based investigation of the autoantibody profile in mothers of children with atrioventricular block. Scand J Immunol. 2011 Nov;74(5):511-7.
  26. Ghillani P, André C, Toly C, Rouquette AM, Bengoufa D, Nicaise P, Goulvestre C, Gleizes A, Dragon-Durey MA, Alyanakian MA, Chretien P, Chollet-Martin S, Musset L, Weill B, Johanet C. Clinical significance of anti-Ro52 (TRIM21) antibodies non-associated with anti-SSA 60kDa antibodies: results of a multicentric study. Autoimmun Rev. 2011 Jul;10(9):509-13.
  27. Montano-Loza AJ, Shums Z, Norman GL, Czaja AJ. Prognostic implications of antibodies to Ro/SSA and soluble liver antigen in type 1 autoimmune hepatitis. Liver Int. 2012 Jan;32(1):85-92.
  28. Grim A, Komosińska-vassev K, Olczyk P. Different types of autoantibodies in rheumatic diseases and methods of their determination. Diagn Lab. 2015;51:235–250. 
  29. Damoiseaux J, Boesten K, Giesen J, Austen J, Tervaert JW. Evaluation of a novel line-blot immunoassay for the detection of antibodies to extractable nuclear antigens. Ann N Y Acad Sci. 2005 Jun;1050:340-7. 
  30. Villalta D, Bizzaro N, Tonutti E, Visentin D, Manoni F, Piazza A, Toffolo L, Rizzotti P, Clemen P, Pradella M, Bassetti D, Tozzoli R. Detection of anti-ENA autoantibodies in patients with systemic connective tissue diseases. Analytical variability and diagnostic sensitivity of 4 methods. Recenti Prog Med. 1999 Nov;90(11):579-84.
  31. Maclachlan D, Vogt P, Wu X, Rose L, Tyndall A, Hasler P. Comparison between line immunoassay (LIA) and enzyme-linked immunosorbent assay (ELISA) for the determination of antibodies to extractable nuclear antigenes (ENA) with reference to other laboratory results and clinical features. 
  32. Z Rheumatol. 2002 Oct;61(5):534-44.
  33. Strandberg L, Winqvist O, Sonesson SE, Mohseni S, Salomonsson S, Bremme K, Buyon JP, Julkunen H, Wahren-Herlenius M. Antibodies to amino acid 200-239 (p200) of Ro52 as serological markers for the risk of developing congenital heart block. Clin Exp Immunol. 2008 Oct;154(1):30-7.
  34. Wolin SL, Reinisch KM. The Ro 60 kDa autoantigen comes into focus: interpreting epitope mapping experiments based on structure. Autoimmun Rev. 2006 Jul;5(6):367-72.
  35. McClain MT, Heinlen LD, Dennis GJ, Roebuck J, Harley JB, James JA. Early events in lupus humoral autoimmunity suggest initiation through molecular mimicry. Nat Med. 2004;11(1):85–89.
  36. Stathopoulou EA, Routsias J, Stea EA, Moutsopoulos HM, Tzioufas AG. Cross-reaction between antibodies to the major epitope of Ro60 kD autoantigen and a homologous peptide of Coxsackie virus 2B protein. Clin Exp Immunol. 2005;141:148–154.
  37. Szymula A, Rosenthal J, Szczerba BM, Bagavant H, Fu SM, Deshmukh US. T cell epitope mimicry between Sjogren’s syndrome Antigen A (SSA)/Ro60 and oral, gut, skin and vaginal bacteria. Clin Immunol. 2014;152:1–9.
  38. Buyon JP, Hiebert R, Copel J, Craft J, Friedman D, Katholi M, Lee LA, Provost TT, Reichlin M, Rider L, Rupel A, Saleeb S, Weston WL, Skovron ML. Autoimmune-associated congenital heart block: demographics, mortality, morbidity and recurrence rates obtained from a national neonatal lupus registry. J Am Coll Cardiol. 1998 Jun;31(7):1658-66.
  39. Greiling TM, Dehner C, Chen X, Hughes K, Iñiguez AJ, Boccitto M, Ruiz DZ, Renfroe SC, Vieira SM, Ruff WE, et al. Commensal orthologs of the human autoantigen Ro60 as triggers of autoimmunity in lupus. Sci Transl Med. 2018;10:eaan2306.
  40. Clancy RM, Marion MC, Ainsworth HC, Blaser MJ, Chang M, Howard TD, Izmirly PM, Lacher C, Masson M, Robins K, Buyon JP, Langefeld CD. Salivary dysbiosis and the clinical spectrum in anti-Ro positive mothers of children with neonatal lupus. J Autoimmun. 2020 Feb;107:102354.
  41. Routsias JG, Tzioufas AG. Sjögren’s syndrome – study of autoantigens and autoantibodies. Clin Rev Allergy Immunol. 2007 Jun;32(3):238-51.
  42. Cozzani E, Drosera M, Gasparini G, Parodi A. Serology of Lupus Erythematosus: Correlation between Immunopathological Features and Clinical Aspects. Autoimmune Dis. 2014;2014:321359.
  43. Brito-Zerón P, Izmirly PM, Ramos-Casals M, Buyon JP, Khamashta MA. The clinical spectrum of autoimmune congenital heart block. Nat Rev Rheumatol. 2015 May;11(5):301-12.
  44. Rivera TL, Izmirly PM, Birnbaum BK, Byrne P, Brauth JB, Katholi M, Kim MY, Fischer J, Clancy RM, Buyon JP. Disease progression in mothers of children enrolled in the Research Registry for Neonatal Lupus. Ann Rheum Dis. 2009 Jun;68(6):828-35.
  45. Buyon JP, Winchester RJ, Slade SG, Arnett F, Copel J, Friedman D, Lockshin MD. Identification of mothers at risk for congenital heart block and other neonatal lupus syndromes in their children. Comparison of enzyme-linked immunosorbent assay and immunoblot for measurement of anti-SS-A/Ro and anti-SS-B/La antibodies. Arthritis Rheum. 1993 Sep;36(9):1263-73.
  46. Zuppa AA, Riccardi R, Frezza S, Gallini F, Luciano RM, Alighieri G, Romagnoli C, De Carolis S. Neonatal lupus: Follow-up in infants with anti-SSA/Ro antibodies and review of the literature. Autoimmun Rev. 2017 Apr;16(4):427-432.
  47. Cimaz R, Spence DL, Hornberger L, Silverman ED. Incidence and spectrum of neonatal lupus erythematosus: a prospective study of infants born to mothers with anti-Ro autoantibodies. J Pediatr. 2003 Jun;142(6):678-83.
  48. Motta M, Rodriguez-Perez C, Tincani A, Lojacono A, Chirico G. Outcome of infants from mothers with anti-SSA/Ro antibodies. J Perinatol. 2007 May;27(5):278-83. 
  49. Boros CA, Spence D, Blaser S, Silverman ED. Hydrocephalus and macrocephaly: new manifestations of neonatal lupus erythematosus. Arthritis Rheum. 2007 Mar 15;57(2):261-6.
  50. Brucato A, Cimaz R, Caporali R, Ramoni V, Buyon J. Pregnancy outcomes in patients with autoimmune diseases and anti-Ro/SSA antibodies. Clin Rev Allergy Immunol. 2011 Feb;40(1):27-41. 
  51. Martínez-Sánchez N, Pérez-Pinto S, Robles-Marhuenda Á, Arnalich-Fernández F, Martín Cameán M, Hueso Zalvide E, Bartha JL. Obstetric and perinatal outcome in anti-Ro/SSA-positive pregnant women: a prospective cohort study. Immunol Res. 2017 Apr;65(2):487-494.
  52. Julkunen H, Eronen M. The rate of recurrence of isolated congenital heart block: a population-based study. Arthritis Rheum. 2001 Feb;44(2):487-8.
  53. Lee LA, Weston WL. Cutaneous lupus erythematosus during the neonatal and childhood periods. Lupus. 1997;6(2):132-8.
  54. Neiman AR, Lee LA, Weston WL, Buyon JP. Cutaneous manifestations of neonatal lupus without heart block: characteristics of mothers and children enrolled in a national registry. J Pediatr. 2000 Nov;137(5):674-80.
  55. Julkunen H, Kaaja R, Kurki P, Palosuo T, Friman C. Fetal outcome in women with primary Sjögren’s syndrome. A retrospective case-control study. Clin Exp Rheumatol. 1995 Jan-Feb;13(1):65-71.
  56. Askanase AD, Friedman DM, Copel J, Dische MR, Dubin A, Starc TJ, Katholi MC, Buyon JP. Spectrum and progression of conduction abnormalities in infants born to mothers with anti-SSA/Ro-SSB/La antibodies. Lupus. 2002;11(3):145-51.
  57. Vanoni F, Lava SAG, Fossali EF, Cavalli R, Simonetti GD, Bianchetti MG, Bozzini MA, Agostoni C, Milani GP. Neonatal Systemic Lupus Erythematosus Syndrome: A Comprehensive Review. Clin Rev Allergy Immunol. 2017 Dec;53(3):469-476.
  58. Teixeira AR, Rodrigues M, Guimarães H, Moura C, Brito I. Neonatal lupus – case series of a tertiary hospital. Acta Reumatol Port. 2017 Oct-Dec;42(4):318-323.
  59. Lee LA. Neonatal lupus erythematosus. J Invest Dermatol. 1993 Jan;100(1):9S-13S.
  60. Lev M, Silverman J, Fitzmaurice FM, Paul MH, Cassels DE, Miller RA. Lack of connection between the atria and the more peripheral conduction system in congenital atrioventricular block. Am J Cardiol. 1971 May;27(5):481-90.
  61. Ho SY, Esscher E, Anderson RH, Michaëlsson M. Anatomy of congenital complete heart block and relation to maternal anti-Ro antibodies. Am J Cardiol. 1986 Aug 1;58(3):291-4.
  62. Izmirly PM, Saxena A, Kim MY, Wang D, Sahl SK, Llanos C, Friedman D, Buyon JP. Maternal and fetal factors associated with mortality and morbidity in a multi-racial/ethnic registry of anti-SSA/Ro-associated cardiac neonatal lupus. Circulation. 2011 Nov 1;124(18):1927-35. 
  63. Eliasson H, Sonesson SE, Sharland G, Granath F, Simpson JM, Carvalho JS, Jicinska H, Tomek V, Dangel J, Zielinsky P, Respondek-Liberska M, Freund MW, Mellander M, Bartrons J, Gardiner HM; Fetal Working Group of the European Association of Pediatric Cardiology. Isolated atrioventricular block in the fetus: a retrospective, multinational, multicenter study of 175 patients. Circulation. 2011 Nov 1;124(18):1919-26.
  64. Hedlund M, Thorlacius GE, Ivanchenko M, et al. Type I IFN system activation in newborns exposed to Ro/SSA and La/SSB autoantibodies in utero. RMD Open. 2020;6(1):e000989.