fbpx
Wikipedia

Anti-dsDNA antibodies

Anti-double stranded DNA (Anti-dsDNA) antibodies are a group of anti-nuclear antibodies (ANA) the target antigen of which is double stranded DNA. Blood tests such as enzyme-linked immunosorbent assay (ELISA) and immunofluorescence are routinely performed to detect anti-dsDNA antibodies in diagnostic laboratories. They are highly diagnostic of systemic lupus erythematosus (SLE) and are implicated in the pathogenesis of lupus nephritis.[1][2]

Immunofluorescence staining pattern of anti-dsDNA antibodies on HEp-20-10 cells (left), Crithidia luciliae (centre) and rat liver (right)

Discovery edit

The first evidence for antinuclear antibodies arose in 1948 when Hargraves, Richmond and Morton discovered the LE cell.[3] These abnormal cells, which are found in the bone marrow of persons who have SLE are categorised as polymorphonuclear leukocytes with phagocytosed whole nuclei.[4] Subsequently, in 1957, antibodies to dsDNA were the first autoantibodies to be identified in patients with SLE.[5]

Antibody production edit

Although the exact mechanism of the generation of dsDNA antibodies is still unknown, it is likely that extracellular DNA is one cause of an immune response against dsDNA. There is a great deal of evidence supporting the idea that dead or dying cells are one major source of this extracellular DNA.[6] Apoptosis is the highly organised process of programmed cell death in which the cell degrades the nuclear DNA and signals for phagocytosis. In people with SLE and other autoimmune disorders this process is thought to be defective, causing either an increase in cell death and/or a decrease in the rate of dead cell clearance.[7]

There is a higher rate of apoptosis in people with SLE and various changes in genes and proteins have been implicated in the defects in apoptosis. These include increased levels of soluble Fas and bcl-2 and polymorphisms in the programmed cell death 1 and runt-related transcription factor X1.[7]

Blebs on apoptotic cells contain nearly all the autoantigens found in SLE, and phagocytes bind these apoptotic cells and phagocytose them. If this process is defective, these autoantigens can be released into the circulation allowing an immune response. Serum amyloid P component is a protein that is thought to aid in the clearance of chromatin produced by apoptotic cells and deficiencies in this protein have been shown (in mice) to cause spontaneous formation of ANA. Autoantigens present on the blebs of apoptotic cells are also prone to modification, which can increase their immunogenicity.[7][8]

Upon release of nuclear proteins and chromatin, antigen presenting cells, such as dendritic cells and macrophages, display these antigens to T helper cells. Although the details of this process are still controversial, evidence shows that to produce an immune response, DNA must activate an antigen presenting cell to produce type 1 interferons. This cytokine serves to induce maturation of plasmacytoid dendritic cells (PDCs) so that they can display their antigens to T helper cells. The mechanism in which eukaryotic DNA activates these cells is still as yet unclear; however, immunogenic CpG sequences have been found to either activate PDCs or act as adjuvant in the response to eukaryotic DNA. CpG motif DNA acts via the pattern recognition receptor, toll-like receptor 9, found highly expressed in PDCs and B cells. The T helper cells then activate B cells, which are also in the presence of these antigens, causing the production of autoantibodies.[6][9][10][11]

Anti-dsDNA antibodies can also be produced through infection via a mechanism known as molecular mimicry. Upon exposure to pneumococcal polysaccharides, cross reactive antibodies between dsDNA and pneumococcal polysaccharides are produced in lupus.[12] Epstein–Barr virus is also known to induce dsDNA antibodies, as seen after immunisation of animals with EBNA-1 epitopes.[13]

Anti-dsDNA antibodies might also be created secondary to the production of antibodies to other proteins within the nucleosome. Mice that have T cells directed towards the nucleosome can elicit a response to other antigens such as dsDNA and histone via a mechanism known as antigen spreading. This effect can also occur after an infection causes the production of autoantibodies to other structures within the nucleus.[13][14]

Role in disease edit

SLE edit

Anti-dsDNA antibodies are incredibly specific for SLE, with studies quoting nearly 100%, and are therefore used in the diagnosis of SLE. Higher titres of anti-dsDNA antibodies are more suggestive of SLE and lower titres can be found in people without the disease. In contrast to the high specificity, estimates of 25–85% have been observed for the sensitivity of anti-dsDNA in SLE. Therefore, presence of anti-dsDNA antibodies are suggestive of SLE, however an absence of the antibodies does not rule out the disease.[1]

The levels of circulating anti-dsDNA antibodies fluctuate with disease activity in SLE. Increases in titres of the antibodies can coincide with, or even precede an increase of disease activity. For this reason titres are serially monitored by clinicians to assess disease progression. Titres are monitored more often in cases of more active lupus than that of less active lupus at intervals of 1–3 months and 6–12 months, respectively.[1]

Anti-dsDNA antibodies are highly associated with glomerulonephritis in SLE, although some patients with high titers of anti-dsDNA antibodies do not develop renal disease. This is most likely due to the fact that anti-dsDNA are a heterogeneous population, some of which have been found not to be pathogenic. Anti-dsDNA antibodies can be present in normal individuals, however these antibodies are usually low avidity IgM isotype. In contrast, pathogenic anti-dsDNA antibodies found in SLE are usually of IgG isotype and show high avidity for dsDNA.[15] One possible mechanism for anti-dsDNA and their role in nephritis is the formation of immune complexes that arise by indirect binding to DNA or nucleosomes that are adhered to the glomerular basement membrane (GBM). Another mechanism is direct binding of antibodies to GBM antigens such as C1q, nucleosomal proteins, heparin sulphate or laminin, which can initiate an inflammatory response by activating complement. They can also be internalised by certain molecules on the GBM cells and cause inflammatory cascades, proliferation and alteration of cellular functions.[2][16][17]

Rheumatoid arthritis edit

Patients with rheumatoid arthritis can develop anti-dsDNA antibodies, however they are usually treatment related. Anti-TNFα biological therapies, such as adalimumab, infliximab and etanercept, can often induce the production of anti-dsDNA antibodies. They are usually low avidity and are only detectable transiently after treatment. The presence of these antibodies can induce a lupus-like syndrome in some cases.[18][19]

Viral infection edit

Infection with viral pathogens can induce anti-dsDNA antibodies transiently. Human immunodeficiency virus, parvovirus B19 and BK virus are known to induce these antibodies.[20][21]

Other diseases edit

There is little evidence supporting the association between anti-dsDNA antibodies and other diseases. Occasionally the monoclonal proteins produced by myeloma patients can be anti-dsDNA. Also, some patients with type 1 autoimmune hepatitis produce anti-dsDNA antibodies.[22][23]

Detection and quantitation edit

A variety of assay formats can be used to detect and quantify anti-dsDNA antibodies but there is no 'gold standard' for diagnostic purposes and the concordance between different assays/methods is low.[24]

Farr assay edit

The Farr assay is used to quantify the amount of anti-dsDNA antibodies in serum. Ammonium sulphate is used to precipitate antigen-antibody complexes that form if the sera contains antibodies to dsDNA. The quantity of these antibodies is determined by using radioactively labelled dsDNA. Although this test is very specific, it is of little use in routine diagnostic laboratories due to its laboriousness and use of radioactive materials. The Farr assay is one of the only tests available that detects high avidity antibodies (along with Crithidia luciliae) and also has the ability to detect antibodies of any isotype.[15]

PEG edit

The polyethylene glycol (PEG) assay precipitates DNA-antibody complexes, similar to the Farr Assay. However, unlike the Farr Assay it does not dissociate the low avidity antibody complexes, allowing for the detection of both high and low avidity anti-dsDNA antibodies.[25]

Immunofluorescence edit

Animal Tissue edit

Animal tissue was the first substrate for immunofluorescent detection of antinuclear antibodies and has been in use since the late 1950s. Liver and kidney tissue sections from animals such as rats are used to identify anti-dsDNA antibodies. This substrate has largely been superseded by the use of HEp-2 cells.[1]

HEp-2 edit

Hep-2 cells, originally of laryngeal carcinoma origin, are actually a contamination of HeLa cells.[26] They are routinely used in the diagnosis of ANA in diagnostic laboratories. HEp-2 cells provide a greater ability to differentiate patterns of ANA than animal sections, due to the large nuclei and high mitotic rate of the cell line. Upon incubation with serum containing anti-dsDNA antibodies and fluorescent labelled secondary antibodies, homogeneous staining of interphase nuclei and condensed chromosomal staining of mitotic cells can be seen.[27]

Crithidia edit

Crithidia luciliae is a haemoflagellate protist with an organelle known as the kinetoplast. This organelle contains a high concentration of circular DNA with no recognisable nuclear antigens, allowing for the reliable detection of anti-dsDNA antibodies. The kinetoplast fluoresces if serum contains high avidity anti-dsDNA antibodies. This test has a higher specificity than EIA because it uses unprocessed DNA. Processed DNA can contain regions of ssDNA, allowing detection of anti-ssDNA antibodies, which can give false positive results.[1][28]

EIA edit

EIA (enzyme immunoassay) detects antibodies using a DNA-coated polystyrene microtitre plate. The DNA used in these assays is often recombinant dsDNA or from calf thymus extract.[29] Upon incubation with serum containing anti-dsDNA antibodies, the antibodies will bind to the DNA and can then be visualised using enzyme-linked secondary antibodies. This assay can be quantitative or semi-quantitative, allowing for estimations of the levels of anti-dsDNA antibodies. This test can produce false positives due to contamination of ssDNA from denatured dsDNA. EIA detects low and high avidity anti-dsDNA antibodies, increasing its sensitivity and reducing its specificity.[1]

Flow cytometry edit

Flow cytometry for the detection of ANA uses multiplexed polystyrene beads coated with multiple autoantigens, such as SSA, SSB, Sm, RNP, Scl-70, Jo-1, dsDNA, centromere B and histone. Serum is incubated with the beads and in the presence of anti-dsDNA antibodies, or any other ANA, the antibodies will bind and fluorescent labelled secondary antibodies will be used for detection. The beads are run through a flow cell which uses a laser to detect fluorescence.[30][31]

Multiplex immunoassay (MIA) edit

Similar to the flow cytometry method of ANA detection, the MIA uses wells containing autoantigens and HEp-2 extract coated beads. The bead sets are coated with specific autoantigens and can be detected individually to allow identification of the particular autoantibody. Automated analysis of the well fluorescence allows for rapid detection of autoantibodies.[30][32]

Microarrays edit

Microarrays are a newly emerging method for the detection of ANA. Individual autoantigens are deposited in an array of dots onto a surface such as polystyrene. A single array could consist of hundreds of autoantigens for screening of multiple autoimmune diseases simultaneously. If anti-dsDNA antibodies are present, incubation of serum and the microarray allow for binding and the dots can then be visualised using a fluorescent labelled anti-IgG antibody.[33]

Therapeutics edit

As a result of the highly specific nature of antibodies, they can be engineered to target and bind key motifs. These motifs can be key features within the pathogenesis of particular diseases, for example human papillomavirus.[34]

References edit

  1. ^ a b c d e f Kavanaugh A, Tomar R, Reveille J, Solomon DH, Homburger HA (January 2000). "Guidelines for clinical use of the antinuclear antibody test and tests for specific autoantibodies to nuclear antigens. American College of Pathologists". Arch. Pathol. Lab. Med. 124 (1): 71–81. doi:10.5858/2000-124-0071-GFCUOT. PMID 10629135.
  2. ^ a b Mortensen ES, Fenton KA, Rekvig OP (February 2008). "Lupus nephritis: the central role of nucleosomes revealed". Am. J. Pathol. 172 (2): 275–83. doi:10.2353/ajpath.2008.070563. PMC 2312358. PMID 18187568.
  3. ^ Hargraves MM, Richmond H, Morton R (January 1948). "Presentation of two bone marrow elements; the tart cell and the L.E. cell". Proceedings of the Staff Meetings of the Mayo Clinic. 23 (2): 25–8. PMID 18921142.
  4. ^ Shao WH, Cohen PL (2011). "Disturbances of apoptotic cell clearance in systemic lupus erythematosus". Arthritis Research & Therapy. 13 (1): 202. doi:10.1186/ar3206. PMC 3157636. PMID 21371352.
  5. ^ Stollar BD (1989). "Immunochemistry of DNA". International Reviews of Immunology. 5 (1): 1–22. doi:10.3109/08830188909086987. PMID 2491157.
  6. ^ a b Su KY, Pisetsky DS (September 2009). "The role of extracellular DNA in autoimmunity in SLE". Scand. J. Immunol. 70 (3): 175–83. doi:10.1111/j.1365-3083.2009.02300.x. PMID 19703007. S2CID 205382203.
  7. ^ a b c Dieker JW, van der Vlag J, Berden JH (February 2004). "Deranged removal of apoptotic cells: its role in the genesis of lupus". Nephrol. Dial. Transplant. 19 (2): 282–5. doi:10.1093/ndt/gfg485. hdl:2066/58078. PMID 14736945.
  8. ^ Smeenk RJ (June 2000). "Antinuclear antibodies: cause of disease or caused by disease?". Rheumatology (Oxford). 39 (6): 581–4. doi:10.1093/rheumatology/39.6.581. PMID 10888701.
  9. ^ Graham KL, Utz PJ (September 2005). "Sources of autoantigens in systemic lupus erythematosus". Current Opinion in Rheumatology. 17 (5): 513–7. doi:10.1097/01.bor.0000171215.87993.6b. PMID 16093826. S2CID 18465332.
  10. ^ Marshak-Rothstein A (November 2006). "Toll-like receptors in systemic autoimmune disease". Nature Reviews Immunology. 6 (11): 823–35. doi:10.1038/nri1957. PMC 7097510. PMID 17063184.
  11. ^ Rekvig OP, Nossent JC (February 2003). "Anti-double-stranded DNA antibodies, nucleosomes, and systemic lupus erythematosus: a time for new paradigms?". Arthritis Rheum. 48 (2): 300–12. doi:10.1002/art.10739. PMID 12571837.
  12. ^ Blank M, Barzilai O, Shoenfeld Y (February 2007). "Molecular mimicry and auto-immunity". Clin Rev Allergy Immunol. 32 (1): 111–8. doi:10.1007/bf02686087. PMID 17426366. S2CID 20475334.
  13. ^ a b Poole BD, Scofield RH, Harley JB, James JA (February 2006). "Epstein-Barr virus and molecular mimicry in systemic lupus erythematosus". Autoimmunity. 39 (1): 63–70. doi:10.1080/08916930500484849. PMID 16455583. S2CID 9844130.
  14. ^ Berden JH (August 2003). "Lupus nephritis: consequence of disturbed removal of apoptotic cells?". Neth J Med. 61 (8): 233–8. PMID 14628957.
  15. ^ a b Egner W (June 2000). "The use of laboratory tests in the diagnosis of SLE". J. Clin. Pathol. 53 (6): 424–32. doi:10.1136/jcp.53.6.424. PMC 1731203. PMID 10911799.
  16. ^ Mok CC, Lau CS (July 2003). "Pathogenesis of systemic lupus erythematosus". J. Clin. Pathol. 56 (7): 481–90. doi:10.1136/jcp.56.7.481. PMC 1769989. PMID 12835292.
  17. ^ Yung S, Chan TM (February 2008). "Anti-DNA antibodies in the pathogenesis of lupus nephritis--the emerging mechanisms". Autoimmun Rev. 7 (4): 317–21. doi:10.1016/j.autrev.2007.12.001. PMID 18295737.
  18. ^ Hanauer SB (September 1999). "Review article: safety of infliximab in clinical trials". Aliment. Pharmacol. Ther. 13 (Suppl 4): 16–22, discussion 38. doi:10.1046/j.1365-2036.1999.00027.x. PMID 10597335. S2CID 1642477.
  19. ^ Hyrich KL, Silman AJ, Watson KD, Symmons DP (December 2004). "Anti-tumour necrosis factor alpha therapy in rheumatoid arthritis: an update on safety". Ann. Rheum. Dis. 63 (12): 1538–43. doi:10.1136/ard.2004.024737. PMC 1754871. PMID 15242866.
  20. ^ Hansen KE, Arnason J, Bridges AJ (April 1998). "Autoantibodies and common viral illnesses". Semin. Arthritis Rheum. 27 (5): 263–71. doi:10.1016/s0049-0172(98)80047-4. PMID 9572708.
  21. ^ Reploeg MD, Storch GA, Clifford DB (July 2001). "Bk virus: a clinical review". Clin. Infect. Dis. 33 (2): 191–202. doi:10.1086/321813. PMID 11418879.
  22. ^ Isenberg DA, Manson JJ, Ehrenstein MR, Rahman A (July 2007). "Fifty years of anti-ds DNA antibodies: are we approaching journey's end?". Rheumatology (Oxford). 46 (7): 1052–6. doi:10.1093/rheumatology/kem112. PMID 17500073.
  23. ^ Maya R, Gershwin ME, Shoenfeld Y (February 2008). "Hepatitis B virus (HBV) and autoimmune disease". Clin Rev Allergy Immunol. 34 (1): 85–102. doi:10.1007/s12016-007-8013-6. PMID 18270862. S2CID 9324159.
  24. ^ Enocsson H; Sjöwall C; Wirestam L; Dahle C; Kastbom A; Rönnelid J; Wetterö J; Skogh T (2015). "Four Anti-dsDNA Antibody Assays in Relation to Systemic Lupus Erythematosus Disease Specificity and Activity". J Rheumatol. 42 (5): 817–25. doi:10.3899/jrheum.140677. PMID 25684763. S2CID 207570256.
  25. ^ Nossent JC, Huysen V, Smeenk RJ, Swaak AJ (September 1989). "Low avidity antibodies to dsDNA as a diagnostic tool". Ann. Rheum. Dis. 48 (9): 748–52. doi:10.1136/ard.48.9.748. PMC 1003868. PMID 2802796.
  26. ^ Lacroix M (January 2008). "Persistent use of "false" cell lines". Int. J. Cancer. 122 (1): 1–4. doi:10.1002/ijc.23233. PMID 17960586. S2CID 27432788.
  27. ^ Bradwell, A. R. (2003). Atlas of HEp-2 patterns and laboratory techniques. Birmingham: Binding Site. ISBN 0-7044-2437-1.
  28. ^ Slater NG, Cameron JS, Lessof MH (September 1976). "The Crithidia luciliae kinetoplast immunofluorescence test in systemic lupus erythematosus". Clin. Exp. Immunol. 25 (3): 480–6. PMC 1541410. PMID 786521.
  29. ^ Burnett, David; Crocker, John R. (1999). The Science of Laboratory Diagnosis. ISIS Medical Media. pp. 494–495. ISBN 1-899066-62-4.
  30. ^ a b Yu X, Schneiderhan-Marra N, Joos TO (2011). "[Protein microarrays and personalized medicine]". Ann. Biol. Clin. (Paris) (in French). 69 (1): 17–29. doi:10.1684/abc.2010.0512. PMID 21463992.
  31. ^ Avaniss-Aghajani E, Berzon S, Sarkissian A (May 2007). "Clinical value of multiplexed bead-based immunoassays for detection of autoantibodies to nuclear antigens". Clin. Vaccine Immunol. 14 (5): 505–9. doi:10.1128/CVI.00034-07. PMC 1865627. PMID 17376860.
  32. ^ Kumar Y, Bhatia A, Minz RW (2009). "Antinuclear antibodies and their detection methods in diagnosis of connective tissue diseases: a journey revisited". Diagn Pathol. 4: 1. doi:10.1186/1746-1596-4-1. PMC 2628865. PMID 19121207.
  33. ^ Hueber W, Utz PJ, Steinman L, Robinson WH (2002). "Autoantibody profiling for the study and treatment of autoimmune disease". Arthritis Res. 4 (5): 290–5. doi:10.1186/ar426. PMC 128938. PMID 12223102.
  34. ^ Cerutti ML, Centeno JM, Goldbaum FA, de Prat-Gay G (2001). "Generation of Sequence-specific, High Affinity Anti-DNA Antibodies". Journal of Biological Chemistry. 276 (16): 12766–12773. doi:10.1074/jbc.M100260200. hdl:11336/48130. PMID 11279040. S2CID 26068816.

anti, dsdna, antibodies, anti, double, stranded, anti, dsdna, antibodies, group, anti, nuclear, antibodies, target, antigen, which, double, stranded, blood, tests, such, enzyme, linked, immunosorbent, assay, elisa, immunofluorescence, routinely, performed, det. Anti double stranded DNA Anti dsDNA antibodies are a group of anti nuclear antibodies ANA the target antigen of which is double stranded DNA Blood tests such as enzyme linked immunosorbent assay ELISA and immunofluorescence are routinely performed to detect anti dsDNA antibodies in diagnostic laboratories They are highly diagnostic of systemic lupus erythematosus SLE and are implicated in the pathogenesis of lupus nephritis 1 2 Immunofluorescence staining pattern of anti dsDNA antibodies on HEp 20 10 cells left Crithidia luciliae centre and rat liver right Contents 1 Discovery 2 Antibody production 3 Role in disease 3 1 SLE 3 2 Rheumatoid arthritis 3 3 Viral infection 3 4 Other diseases 4 Detection and quantitation 4 1 Farr assay 4 2 PEG 4 3 Immunofluorescence 4 3 1 Animal Tissue 4 3 2 HEp 2 4 3 3 Crithidia 4 4 EIA 4 5 Flow cytometry 4 6 Multiplex immunoassay MIA 4 7 Microarrays 5 Therapeutics 6 ReferencesDiscovery editThe first evidence for antinuclear antibodies arose in 1948 when Hargraves Richmond and Morton discovered the LE cell 3 These abnormal cells which are found in the bone marrow of persons who have SLE are categorised as polymorphonuclear leukocytes with phagocytosed whole nuclei 4 Subsequently in 1957 antibodies to dsDNA were the first autoantibodies to be identified in patients with SLE 5 Antibody production editAlthough the exact mechanism of the generation of dsDNA antibodies is still unknown it is likely that extracellular DNA is one cause of an immune response against dsDNA There is a great deal of evidence supporting the idea that dead or dying cells are one major source of this extracellular DNA 6 Apoptosis is the highly organised process of programmed cell death in which the cell degrades the nuclear DNA and signals for phagocytosis In people with SLE and other autoimmune disorders this process is thought to be defective causing either an increase in cell death and or a decrease in the rate of dead cell clearance 7 There is a higher rate of apoptosis in people with SLE and various changes in genes and proteins have been implicated in the defects in apoptosis These include increased levels of soluble Fas and bcl 2 and polymorphisms in the programmed cell death 1 and runt related transcription factor X1 7 Blebs on apoptotic cells contain nearly all the autoantigens found in SLE and phagocytes bind these apoptotic cells and phagocytose them If this process is defective these autoantigens can be released into the circulation allowing an immune response Serum amyloid P component is a protein that is thought to aid in the clearance of chromatin produced by apoptotic cells and deficiencies in this protein have been shown in mice to cause spontaneous formation of ANA Autoantigens present on the blebs of apoptotic cells are also prone to modification which can increase their immunogenicity 7 8 Upon release of nuclear proteins and chromatin antigen presenting cells such as dendritic cells and macrophages display these antigens to T helper cells Although the details of this process are still controversial evidence shows that to produce an immune response DNA must activate an antigen presenting cell to produce type 1 interferons This cytokine serves to induce maturation of plasmacytoid dendritic cells PDCs so that they can display their antigens to T helper cells The mechanism in which eukaryotic DNA activates these cells is still as yet unclear however immunogenic CpG sequences have been found to either activate PDCs or act as adjuvant in the response to eukaryotic DNA CpG motif DNA acts via the pattern recognition receptor toll like receptor 9 found highly expressed in PDCs and B cells The T helper cells then activate B cells which are also in the presence of these antigens causing the production of autoantibodies 6 9 10 11 Anti dsDNA antibodies can also be produced through infection via a mechanism known as molecular mimicry Upon exposure to pneumococcal polysaccharides cross reactive antibodies between dsDNA and pneumococcal polysaccharides are produced in lupus 12 Epstein Barr virus is also known to induce dsDNA antibodies as seen after immunisation of animals with EBNA 1 epitopes 13 Anti dsDNA antibodies might also be created secondary to the production of antibodies to other proteins within the nucleosome Mice that have T cells directed towards the nucleosome can elicit a response to other antigens such as dsDNA and histone via a mechanism known as antigen spreading This effect can also occur after an infection causes the production of autoantibodies to other structures within the nucleus 13 14 Role in disease editSLE edit Anti dsDNA antibodies are incredibly specific for SLE with studies quoting nearly 100 and are therefore used in the diagnosis of SLE Higher titres of anti dsDNA antibodies are more suggestive of SLE and lower titres can be found in people without the disease In contrast to the high specificity estimates of 25 85 have been observed for the sensitivity of anti dsDNA in SLE Therefore presence of anti dsDNA antibodies are suggestive of SLE however an absence of the antibodies does not rule out the disease 1 The levels of circulating anti dsDNA antibodies fluctuate with disease activity in SLE Increases in titres of the antibodies can coincide with or even precede an increase of disease activity For this reason titres are serially monitored by clinicians to assess disease progression Titres are monitored more often in cases of more active lupus than that of less active lupus at intervals of 1 3 months and 6 12 months respectively 1 Anti dsDNA antibodies are highly associated with glomerulonephritis in SLE although some patients with high titers of anti dsDNA antibodies do not develop renal disease This is most likely due to the fact that anti dsDNA are a heterogeneous population some of which have been found not to be pathogenic Anti dsDNA antibodies can be present in normal individuals however these antibodies are usually low avidity IgM isotype In contrast pathogenic anti dsDNA antibodies found in SLE are usually of IgG isotype and show high avidity for dsDNA 15 One possible mechanism for anti dsDNA and their role in nephritis is the formation of immune complexes that arise by indirect binding to DNA or nucleosomes that are adhered to the glomerular basement membrane GBM Another mechanism is direct binding of antibodies to GBM antigens such as C1q nucleosomal proteins heparin sulphate or laminin which can initiate an inflammatory response by activating complement They can also be internalised by certain molecules on the GBM cells and cause inflammatory cascades proliferation and alteration of cellular functions 2 16 17 Rheumatoid arthritis edit Patients with rheumatoid arthritis can develop anti dsDNA antibodies however they are usually treatment related Anti TNFa biological therapies such as adalimumab infliximab and etanercept can often induce the production of anti dsDNA antibodies They are usually low avidity and are only detectable transiently after treatment The presence of these antibodies can induce a lupus like syndrome in some cases 18 19 Viral infection edit Infection with viral pathogens can induce anti dsDNA antibodies transiently Human immunodeficiency virus parvovirus B19 and BK virus are known to induce these antibodies 20 21 Other diseases edit There is little evidence supporting the association between anti dsDNA antibodies and other diseases Occasionally the monoclonal proteins produced by myeloma patients can be anti dsDNA Also some patients with type 1 autoimmune hepatitis produce anti dsDNA antibodies 22 23 Detection and quantitation editA variety of assay formats can be used to detect and quantify anti dsDNA antibodies but there is no gold standard for diagnostic purposes and the concordance between different assays methods is low 24 Farr assay edit The Farr assay is used to quantify the amount of anti dsDNA antibodies in serum Ammonium sulphate is used to precipitate antigen antibody complexes that form if the sera contains antibodies to dsDNA The quantity of these antibodies is determined by using radioactively labelled dsDNA Although this test is very specific it is of little use in routine diagnostic laboratories due to its laboriousness and use of radioactive materials The Farr assay is one of the only tests available that detects high avidity antibodies along with Crithidia luciliae and also has the ability to detect antibodies of any isotype 15 PEG edit The polyethylene glycol PEG assay precipitates DNA antibody complexes similar to the Farr Assay However unlike the Farr Assay it does not dissociate the low avidity antibody complexes allowing for the detection of both high and low avidity anti dsDNA antibodies 25 Immunofluorescence edit Animal Tissue edit Animal tissue was the first substrate for immunofluorescent detection of antinuclear antibodies and has been in use since the late 1950s Liver and kidney tissue sections from animals such as rats are used to identify anti dsDNA antibodies This substrate has largely been superseded by the use of HEp 2 cells 1 HEp 2 edit Hep 2 cells originally of laryngeal carcinoma origin are actually a contamination of HeLa cells 26 They are routinely used in the diagnosis of ANA in diagnostic laboratories HEp 2 cells provide a greater ability to differentiate patterns of ANA than animal sections due to the large nuclei and high mitotic rate of the cell line Upon incubation with serum containing anti dsDNA antibodies and fluorescent labelled secondary antibodies homogeneous staining of interphase nuclei and condensed chromosomal staining of mitotic cells can be seen 27 Crithidia edit Crithidia luciliae is a haemoflagellate protist with an organelle known as the kinetoplast This organelle contains a high concentration of circular DNA with no recognisable nuclear antigens allowing for the reliable detection of anti dsDNA antibodies The kinetoplast fluoresces if serum contains high avidity anti dsDNA antibodies This test has a higher specificity than EIA because it uses unprocessed DNA Processed DNA can contain regions of ssDNA allowing detection of anti ssDNA antibodies which can give false positive results 1 28 EIA edit EIA enzyme immunoassay detects antibodies using a DNA coated polystyrene microtitre plate The DNA used in these assays is often recombinant dsDNA or from calf thymus extract 29 Upon incubation with serum containing anti dsDNA antibodies the antibodies will bind to the DNA and can then be visualised using enzyme linked secondary antibodies This assay can be quantitative or semi quantitative allowing for estimations of the levels of anti dsDNA antibodies This test can produce false positives due to contamination of ssDNA from denatured dsDNA EIA detects low and high avidity anti dsDNA antibodies increasing its sensitivity and reducing its specificity 1 Flow cytometry edit Flow cytometry for the detection of ANA uses multiplexed polystyrene beads coated with multiple autoantigens such as SSA SSB Sm RNP Scl 70 Jo 1 dsDNA centromere B and histone Serum is incubated with the beads and in the presence of anti dsDNA antibodies or any other ANA the antibodies will bind and fluorescent labelled secondary antibodies will be used for detection The beads are run through a flow cell which uses a laser to detect fluorescence 30 31 Multiplex immunoassay MIA edit Similar to the flow cytometry method of ANA detection the MIA uses wells containing autoantigens and HEp 2 extract coated beads The bead sets are coated with specific autoantigens and can be detected individually to allow identification of the particular autoantibody Automated analysis of the well fluorescence allows for rapid detection of autoantibodies 30 32 Microarrays edit Microarrays are a newly emerging method for the detection of ANA Individual autoantigens are deposited in an array of dots onto a surface such as polystyrene A single array could consist of hundreds of autoantigens for screening of multiple autoimmune diseases simultaneously If anti dsDNA antibodies are present incubation of serum and the microarray allow for binding and the dots can then be visualised using a fluorescent labelled anti IgG antibody 33 Therapeutics editAs a result of the highly specific nature of antibodies they can be engineered to target and bind key motifs These motifs can be key features within the pathogenesis of particular diseases for example human papillomavirus 34 References edit a b c d e f Kavanaugh A Tomar R Reveille J Solomon DH Homburger HA January 2000 Guidelines for clinical use of the antinuclear antibody test and tests for specific autoantibodies to nuclear antigens American College of Pathologists Arch Pathol Lab Med 124 1 71 81 doi 10 5858 2000 124 0071 GFCUOT PMID 10629135 a b Mortensen ES Fenton KA Rekvig OP February 2008 Lupus nephritis the central role of nucleosomes revealed Am J Pathol 172 2 275 83 doi 10 2353 ajpath 2008 070563 PMC 2312358 PMID 18187568 Hargraves MM Richmond H Morton R January 1948 Presentation of two bone marrow elements the tart cell and the L E cell Proceedings of the Staff Meetings of the Mayo Clinic 23 2 25 8 PMID 18921142 Shao WH Cohen PL 2011 Disturbances of apoptotic cell clearance in systemic lupus erythematosus Arthritis Research amp Therapy 13 1 202 doi 10 1186 ar3206 PMC 3157636 PMID 21371352 Stollar BD 1989 Immunochemistry of DNA International Reviews of Immunology 5 1 1 22 doi 10 3109 08830188909086987 PMID 2491157 a b Su KY Pisetsky DS September 2009 The role of extracellular DNA in autoimmunity in SLE Scand J Immunol 70 3 175 83 doi 10 1111 j 1365 3083 2009 02300 x PMID 19703007 S2CID 205382203 a b c Dieker JW van der Vlag J Berden JH February 2004 Deranged removal of apoptotic cells its role in the genesis of lupus Nephrol Dial Transplant 19 2 282 5 doi 10 1093 ndt gfg485 hdl 2066 58078 PMID 14736945 Smeenk RJ June 2000 Antinuclear antibodies cause of disease or caused by disease Rheumatology Oxford 39 6 581 4 doi 10 1093 rheumatology 39 6 581 PMID 10888701 Graham KL Utz PJ September 2005 Sources of autoantigens in systemic lupus erythematosus Current Opinion in Rheumatology 17 5 513 7 doi 10 1097 01 bor 0000171215 87993 6b PMID 16093826 S2CID 18465332 Marshak Rothstein A November 2006 Toll like receptors in systemic autoimmune disease Nature Reviews Immunology 6 11 823 35 doi 10 1038 nri1957 PMC 7097510 PMID 17063184 Rekvig OP Nossent JC February 2003 Anti double stranded DNA antibodies nucleosomes and systemic lupus erythematosus a time for new paradigms Arthritis Rheum 48 2 300 12 doi 10 1002 art 10739 PMID 12571837 Blank M Barzilai O Shoenfeld Y February 2007 Molecular mimicry and auto immunity Clin Rev Allergy Immunol 32 1 111 8 doi 10 1007 bf02686087 PMID 17426366 S2CID 20475334 a b Poole BD Scofield RH Harley JB James JA February 2006 Epstein Barr virus and molecular mimicry in systemic lupus erythematosus Autoimmunity 39 1 63 70 doi 10 1080 08916930500484849 PMID 16455583 S2CID 9844130 Berden JH August 2003 Lupus nephritis consequence of disturbed removal of apoptotic cells Neth J Med 61 8 233 8 PMID 14628957 a b Egner W June 2000 The use of laboratory tests in the diagnosis of SLE J Clin Pathol 53 6 424 32 doi 10 1136 jcp 53 6 424 PMC 1731203 PMID 10911799 Mok CC Lau CS July 2003 Pathogenesis of systemic lupus erythematosus J Clin Pathol 56 7 481 90 doi 10 1136 jcp 56 7 481 PMC 1769989 PMID 12835292 Yung S Chan TM February 2008 Anti DNA antibodies in the pathogenesis of lupus nephritis the emerging mechanisms Autoimmun Rev 7 4 317 21 doi 10 1016 j autrev 2007 12 001 PMID 18295737 Hanauer SB September 1999 Review article safety of infliximab in clinical trials Aliment Pharmacol Ther 13 Suppl 4 16 22 discussion 38 doi 10 1046 j 1365 2036 1999 00027 x PMID 10597335 S2CID 1642477 Hyrich KL Silman AJ Watson KD Symmons DP December 2004 Anti tumour necrosis factor alpha therapy in rheumatoid arthritis an update on safety Ann Rheum Dis 63 12 1538 43 doi 10 1136 ard 2004 024737 PMC 1754871 PMID 15242866 Hansen KE Arnason J Bridges AJ April 1998 Autoantibodies and common viral illnesses Semin Arthritis Rheum 27 5 263 71 doi 10 1016 s0049 0172 98 80047 4 PMID 9572708 Reploeg MD Storch GA Clifford DB July 2001 Bk virus a clinical review Clin Infect Dis 33 2 191 202 doi 10 1086 321813 PMID 11418879 Isenberg DA Manson JJ Ehrenstein MR Rahman A July 2007 Fifty years of anti ds DNA antibodies are we approaching journey s end Rheumatology Oxford 46 7 1052 6 doi 10 1093 rheumatology kem112 PMID 17500073 Maya R Gershwin ME Shoenfeld Y February 2008 Hepatitis B virus HBV and autoimmune disease Clin Rev Allergy Immunol 34 1 85 102 doi 10 1007 s12016 007 8013 6 PMID 18270862 S2CID 9324159 Enocsson H Sjowall C Wirestam L Dahle C Kastbom A Ronnelid J Wettero J Skogh T 2015 Four Anti dsDNA Antibody Assays in Relation to Systemic Lupus Erythematosus Disease Specificity and Activity J Rheumatol 42 5 817 25 doi 10 3899 jrheum 140677 PMID 25684763 S2CID 207570256 Nossent JC Huysen V Smeenk RJ Swaak AJ September 1989 Low avidity antibodies to dsDNA as a diagnostic tool Ann Rheum Dis 48 9 748 52 doi 10 1136 ard 48 9 748 PMC 1003868 PMID 2802796 Lacroix M January 2008 Persistent use of false cell lines Int J Cancer 122 1 1 4 doi 10 1002 ijc 23233 PMID 17960586 S2CID 27432788 Bradwell A R 2003 Atlas of HEp 2 patterns and laboratory techniques Birmingham Binding Site ISBN 0 7044 2437 1 Slater NG Cameron JS Lessof MH September 1976 The Crithidia luciliae kinetoplast immunofluorescence test in systemic lupus erythematosus Clin Exp Immunol 25 3 480 6 PMC 1541410 PMID 786521 Burnett David Crocker John R 1999 The Science of Laboratory Diagnosis ISIS Medical Media pp 494 495 ISBN 1 899066 62 4 a b Yu X Schneiderhan Marra N Joos TO 2011 Protein microarrays and personalized medicine Ann Biol Clin Paris in French 69 1 17 29 doi 10 1684 abc 2010 0512 PMID 21463992 Avaniss Aghajani E Berzon S Sarkissian A May 2007 Clinical value of multiplexed bead based immunoassays for detection of autoantibodies to nuclear antigens Clin Vaccine Immunol 14 5 505 9 doi 10 1128 CVI 00034 07 PMC 1865627 PMID 17376860 Kumar Y Bhatia A Minz RW 2009 Antinuclear antibodies and their detection methods in diagnosis of connective tissue diseases a journey revisited Diagn Pathol 4 1 doi 10 1186 1746 1596 4 1 PMC 2628865 PMID 19121207 Hueber W Utz PJ Steinman L Robinson WH 2002 Autoantibody profiling for the study and treatment of autoimmune disease Arthritis Res 4 5 290 5 doi 10 1186 ar426 PMC 128938 PMID 12223102 Cerutti ML Centeno JM Goldbaum FA de Prat Gay G 2001 Generation of Sequence specific High Affinity Anti DNA Antibodies Journal of Biological Chemistry 276 16 12766 12773 doi 10 1074 jbc M100260200 hdl 11336 48130 PMID 11279040 S2CID 26068816 Retrieved from https en wikipedia org w index php title Anti dsDNA antibodies amp oldid 1213362204, wikipedia, wiki, book, books, library,

article

, read, download, free, free download, mp3, video, mp4, 3gp, jpg, jpeg, gif, png, picture, music, song, movie, book, game, games.