Skip to main content

Part of the book series: Milestones in Drug Therapy ((MDT))

  • 969 Accesses

Abstract

Sjögrens syndrome (SS) is characterized by inflammation and dysfunction of the secretory organs. In the majority of patients the salivary and lachrymal glands are predominantly affected, although systemic symptoms are common. The pathogenesis of the disease is not well understood and to date there is no universally effective therapy available. The development of gene therapy and in particular local gene therapy applied to the salivary glands may be effective in the disease. Animal studies have shown that treatment with immunomodulatory molecules like interleukin 10 or vasoactive intestinal peptide can influence salivary function positively while changing the local inflammatory environment. Future research will have to show whether this approach is feasible in humans.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 169.00
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
Softcover Book
USD 219.99
Price excludes VAT (USA)
  • Compact, lightweight edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info
Hardcover Book
USD 219.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free shipping worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Preview

Unable to display preview. Download preview PDF.

Unable to display preview. Download preview PDF.

References

  1. Fox RI (2005) Sjögren’s syndrome. Lancet 366: 321–331

    Article  CAS  PubMed  Google Scholar 

  2. Vitali C, Bombardieri S, Jonsson R, Moutsopoulos HM, Alexander EL, Carsons SE, Daniels TE, Fox PC, Fox RI, Kassan SS et al. (2002) Classification criteria for Sjögren’s syndrome: a revised version of the European criteria proposed by the American-European Consensus Group. Ann Rheum Dis 61: 554–558

    Article  CAS  PubMed  Google Scholar 

  3. Williams PH, Cobb BL, Namjou B, Scofield RH, Sawalha AH, Harley JB (2007) Horizons in Sjögren’s syndrome genetics. Clin Rev Allergy Immunol 32: 201–209

    Article  CAS  PubMed  Google Scholar 

  4. Yamamoto K (2003) Pathogenesis of Sjögren’s syndrome. Autoimmun Rev 2: 13–18

    Article  PubMed  Google Scholar 

  5. Roescher N, Tak PP, Illei GG (2009) Cytokines in Sjögren’s syndrome. Oral Dis 15: 519–526

    Article  CAS  PubMed  Google Scholar 

  6. Daniels TE, Fox PC (1992) Salivary and oral components of Sjögren’s syndrome. Rheum Dis Clin North Am 18: 571–589

    CAS  PubMed  Google Scholar 

  7. Voulgarelis M, Dafni UG, Isenberg DA, Moutsopoulos HM (1999) Malignant lymphoma in primary Sjögren’s syndrome: a multicenter, retrospective, clinical study by the European Concerted Action on Sjögren’s Syndrome. Arthritis Rheum 42: 1765–1772

    Article  CAS  PubMed  Google Scholar 

  8. Tzioufas AG, Voulgarelis M (2007) Update on Sjögren’s syndrome autoimmune epithelitis: from classification to increased neoplasias. Best Pract Res Clin Rheumatol 21: 989–1010

    Article  CAS  PubMed  Google Scholar 

  9. Mariette X (1999) Lymphomas in patients with Sjögren’s syndrome: review of the literature and physiopathologic hypothesis. Leuk Lymphoma 33: 93–99

    CAS  PubMed  Google Scholar 

  10. Melvin JE, Yule D, Shuttleworth T, Begenisich T (2005) Regulation of fluid and electrolyte secretion in salivary gland acinar cells. Annu Rev Physiol 67: 445–469

    Article  CAS  PubMed  Google Scholar 

  11. Tsunawaki S, Nakamura S, Ohyama Y, Sasaki M, Ikebe-Hiroki A, Hiraki A, Kadena T, Kawamura E, Kumamaru W, Shinohara M et al. (2002) Possible function of salivary gland epithelial cells as nonprofessional antigen-presenting cells in the development of Sjögren’s syndrome. J Rheumatol 29: 1884–1896

    CAS  PubMed  Google Scholar 

  12. Dimitriou ID, Kapsogeorgou EK, Moutsopoulos HM, Manoussakis MN (2002) CD40 on salivary gland epithelial cells: high constitutive expression by cultured cells from Sjögren’s syndrome patients indicating their intrinsic activation. Clin Exp Immunol 127: 386–392

    Article  CAS  PubMed  Google Scholar 

  13. Matsumura R, Umemiya K, Goto T, Nakazawa T, Kagami M, Tomioka H, Tanabe E, Sugiyama T, Sueishi M (2001) Glandular and extraglandular expression of costimulatory molecules in patients with Sjögren’s syndrome. Ann Rheum Dis 60: 473–482

    Article  CAS  PubMed  Google Scholar 

  14. Moutsopoulos HM (1994) Sjögren’s syndrome: autoimmune epithelitis. Clin Immunol Immunopathol 72: 162–165

    Article  CAS  PubMed  Google Scholar 

  15. Humphreys-Beher MG, Brayer J, Yamachika S, Peck AB, Jonsson R (1999) An alternative perspective to the immune response in autoimmune exocrinopathy: induction of functional quiescence rather than destructive autoaggression. Scand J Immunol 49: 7–10

    Article  CAS  PubMed  Google Scholar 

  16. Fox PC, Speight PM (1996) Current concepts of autoimmune exocrinopathy: immunologic mechanisms in the salivary pathology of Sjögren’s syndrome. Crit Rev Oral Biol Med 7: 144–158

    Article  CAS  PubMed  Google Scholar 

  17. Nakamura T, Matsui M, Uchida K, Futatsugi A, Kusakawa S, Matsumoto N, Nakamura K, Manabe T, Taketo MM, Mikoshiba K (2004) M(3) muscarinic acetylcholine receptor plays a critical role in parasympathetic control of salivation in mice. J Physiol 558: 561–575

    Article  CAS  PubMed  Google Scholar 

  18. Waterman SA, Gordon TP, Rischmueller M (2000) Inhibitory effects of muscarinic receptor autoantibodies on parasympathetic neurotransmission in Sjögren’s syndrome. Arthritis Rheum 43: 1647–1654

    Article  CAS  PubMed  Google Scholar 

  19. Cha S, Singson E, Cornelius J, Yagna JP, Knot HJ, Peck AB (2006) Muscarinic acetylcholine type-3 receptor desensitization due to chronic exposure to Sjögren’s syndrome-associated autoantibodies. J Rheumatol 33: 296–306

    CAS  PubMed  Google Scholar 

  20. Kovacs L, Marczinovits I, Gyorgy A, Toth GK, Dorgai L, Pal J, Molnar J, Pokorny G (2005) Clinical associations of autoantibodies to human muscarinic acetylcholine receptor 3(213–228) in primary Sjögren’s syndrome. Rheumatology (Oxford) 44: 1021–1025

    Article  CAS  Google Scholar 

  21. Dawson LJ, Allison HE, Stanbury J, Fitzgerald D, Smith PM (2004) Putative anti-muscarinic antibodies cannot be detected in patients with primary Sjögren’s syndrome using conventional immunological approaches. Rheumatology (Oxford) 43: 1488–1495

    Article  CAS  Google Scholar 

  22. Vivino FB, Al-Hashimi I, Khan Z, LeVeque FG, Salisbury PL 3rd, Tran-Johnson TK, Muscoplat CC, Trivedi M, Goldlust B, Gallagher SC (1999) Pilocarpine tablets for the treatment of dry mouth and dry eye symptoms in patients with Sjögren syndrome: a randomized, placebo-controlled, fixed-dose, multicenter trial. P92-01 Study Group. Arch Intern Med 159: 174–181

    Article  CAS  PubMed  Google Scholar 

  23. Drosos AA, Skopouli FN, Costopoulos JS, Papadimitriou CS, Moutsopoulos HM (1986) Cyclosporin A (CyA) in primary Sjögren’s syndrome: a double blind study. Ann Rheum Dis 45: 732–735

    Article  CAS  PubMed  Google Scholar 

  24. Skopouli FN, Jagiello P, Tsifetaki N, Moutsopoulos HM (1996) Methotrexate in primary Sjögren’s syndrome. Clin Exp Rheumatol 14: 555–558

    CAS  PubMed  Google Scholar 

  25. Zandbelt MM, van den Hoogen FH, de Wilde PC, van den Berg PJ, Schneider HG, van de Putte LB (2001) Reversibility of histological and immunohistological abnormalities in sublabial salivary gland biopsy specimens following treatment with corticosteroids in Sjögren’s syndrome. Ann Rheum Dis 60: 511–513

    Article  CAS  PubMed  Google Scholar 

  26. Fox PC, Datiles M, Atkinson JC, Macynski AA, Scott J, Fletcher D, Valdez IH, Kurrasch RH, Delapenha R, Jackson W (1993) Prednisone and piroxicam for treatment of primary Sjögren’s syndrome. Clin Exp Rheumatol 11: 149–156

    CAS  PubMed  Google Scholar 

  27. Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP (2008) Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol Ther 117: 244–279

    Article  CAS  PubMed  Google Scholar 

  28. Mariette X, Ravaud P, Steinfeld S, Baron G, Goetz J, Hachulla E, Combe B, Puechal X, Pennec Y, Sauvezie B et al. (2004) Inefficacy of infliximab in primary Sjögren’s syndrome: results of the randomized, controlled Trial of Remicade in Primary Sjögren’s Syndrome (TRIPSS). Arthritis Rheum 50: 1270–1276

    Article  CAS  PubMed  Google Scholar 

  29. Niedermeyer J, Hoffmeyer F, Hertenstein B, Hoeper MM, Fabel H (2000) Treatment of lymphoproliferative disease with rituximab. Lancet 355: 499

    Article  CAS  PubMed  Google Scholar 

  30. Edwards JC, Szczepanski L, Szechinski J, Filipowicz-Sosnowska A, Emery P, Close DR, Stevens RM, Shaw T (2004) Efficacy of B-cell-targeted therapy with rituximab in patients with rheumatoid arthritis. N Engl J Med 350: 2572–2581

    Article  CAS  PubMed  Google Scholar 

  31. Arkfeld DG (2008) The potential utility of B cell-directed biologic therapy in autoimmune diseases. Rheumatol Int 28: 205–215

    Article  CAS  PubMed  Google Scholar 

  32. Pijpe J, van Imhoff GW, Spijkervet FK, Roodenburg JL, Wolbink GJ, Mansour K, Vissink A, Kallenberg CG, Bootsma H (2005) Rituximab treatment in patients with primary Sjögren’s syndrome: an open-label phase II study. Arthritis Rheum 52: 2740–2750

    Article  CAS  PubMed  Google Scholar 

  33. Meijer J, Vissink A, Meiners P, Spijkervet F, Kallenberg C, Bootsma H (2008 ACR/ARHP Scientific meeting) Rituximab treatment in primary Sjögren’s syndrome; a double-blind placebo controlled trial, San Francisco, CA, USA

    Google Scholar 

  34. Dass S, Bowman SJ, Vital EM, Ikeda K, Pease CT, Hamburger J, Richards A, Rauz S, Emery P (2008) Reduction of fatigue in Sjögren syndrome with rituximab: results of a randomised, doubleblind, placebo-controlled pilot study. Ann Rheum Dis 67: 1541–1544

    Article  CAS  PubMed  Google Scholar 

  35. Isaksen K, Jonsson R, Omdal R (2008) Anti-CD20 treatment in primary Sjögren’s syndrome. Scand J Immunol 68: 554–564

    Article  CAS  PubMed  Google Scholar 

  36. Moutsopoulos NM, Katsifis GE, Angelov N, Leakan RA, Sankar V, Pillemer S, Wahl SM (2008) Lack of efficacy of etanercept in Sjögren syndrome correlates with failed suppression of tumour necrosis factor alpha and systemic immune activation. Ann Rheum Dis 67: 1437–1443

    Article  CAS  PubMed  Google Scholar 

  37. Mavragani CP, Moutsopoulos HM (2007) Conventional therapy of Sjögren’s syndrome. Clin Rev Allergy Immunol 32: 284–291

    Article  CAS  PubMed  Google Scholar 

  38. Giarratana N, Penna G, Adorini L (2007) Animal models of spontaneous autoimmune disease: type 1 diabetes in the nonobese diabetic mouse. Methods Mol Biol 380: 285–311

    Article  CAS  PubMed  Google Scholar 

  39. Robinson CP, Yamachika S, Bounous DI, Brayer J, Jonsson R, Holmdahl R, Peck AB, Humphreys-Beher MG (1998) A novel NOD-derived murine model of primary Sjögren’s syndrome. Arthritis Rheum 41: 150–156

    Article  CAS  PubMed  Google Scholar 

  40. Lodde BM, Mineshiba F, Kok MR, Wang J, Zheng C, Schmidt M, Cotrim AP, Kriete M, Tak PP, Baum BJ (2006) NOD mouse model for Sjögren’s syndrome: lack of longitudinal stability. Oral Dis 12: 566–572

    Article  CAS  PubMed  Google Scholar 

  41. Rott O, Fleischer B, Cash E (1994) Interleukin-10 prevents experimental allergic encephalomyelitis in rats. Eur J Immunol 24: 1434–1440

    Article  CAS  PubMed  Google Scholar 

  42. Bai XF, Zhu J, Zhang GX, Kaponides G, Hojeberg B, van der Meide PH, Link H (1997) IL-10 suppresses experimental autoimmune neuritis and down-regulates TH1-type immune responses. Clin Immunol Immunopathol 83: 117–126

    Article  CAS  PubMed  Google Scholar 

  43. Kok MR, Yamano S, Lodde BM, Wang J, Couwenhoven RI, Yakar S, Voutetakis A, Leroith D, Schmidt M, Afione S et al. (2003) Local adeno-associated virus-mediated interleukin 10 gene transfer has disease-modifying effects in a murine model of Sjögren’s syndrome. Hum Gene Ther 14: 1605–1618

    Article  CAS  PubMed  Google Scholar 

  44. Gonzalez-Rey E, Delgado M (2007) Vasoactive intestinal peptide and regulatory T-cell induction: a new mechanism and therapeutic potential for immune homeostasis. Trends Mol Med 13: 241–251

    Article  CAS  PubMed  Google Scholar 

  45. Lodde BM, Mineshiba F, Wang J, Cotrim AP, Afione S, Tak PP, Baum BJ (2006) Effect of human vasoactive intestinal peptide gene transfer in a murine model of Sjögren’s syndrome. Ann Rheum Dis 65: 195–200

    Article  CAS  PubMed  Google Scholar 

  46. Vosters JYH, Roescher N, Kok M, Tak P, Chiorini J (2009) Local expression of TNFR1:IgG can induce salivary gland dysfunction in a murine model of Sjögren’s Syndrome. Arthritis Res Ther 11: R189

    Article  PubMed  Google Scholar 

  47. Nedjai B, Hitman GA, Quillinan N, Coughlan RJ, Church L, McDermott MF, Turner MD (2009) Proinflammatory action of the antiinflammatory drug infliximab in tumor necrosis factor receptor-associated periodic syndrome. Arthritis Rheum 60: 619–625

    Article  CAS  PubMed  Google Scholar 

  48. Nguyen CQ, Cornelius JG, Cooper L, Neff J, Tao J, Lee BH, Peck AB (2008) Identification of possible candidate genes regulating Sjögren’s syndrome-associated autoimmunity: a potential role for TNFSF4 in autoimmune exocrinopathy. Arthritis Res Ther 10: R137

    Article  PubMed  Google Scholar 

  49. Sakai A, Sugawara Y, Kuroishi T, Sasano T, Sugawara S (2008) Identification of IL-18 and Th17 cells in salivary glands of patients with Sjögren’s syndrome, and amplification of IL-17-mediated secretion of inflammatory cytokines from salivary gland cells by IL-18. J Immunol 181: 2898–2906

    CAS  PubMed  Google Scholar 

  50. Oxholm P, Daniels TE, Bendtzen K (1992) Cytokine expression in labial salivary glands from patients with primary Sjögren’s syndrome. Autoimmunity 12: 185–191

    Article  CAS  PubMed  Google Scholar 

  51. van Woerkom JM, Kruize AA, Wenting-van Wijk MJ, Knol E, Bihari IC, Jacobs JW, Bijlsma JW, Lafeber FP, van Roon JA (2005) Salivary gland and peripheral blood T helper 1 and 2 cell activity in Sjögren’s syndrome compared with non-Sjögren’s sicca syndrome. Ann Rheum Dis 64: 1474–1479

    Article  PubMed  Google Scholar 

  52. Miceli-Richard C, Comets E, Loiseau P, Puechal X, Hachulla E, Mariette X (2007) Association of an IRF5 gene functional polymorphism with Sjögren’s syndrome. Arthritis Rheum 56: 3989–3994

    Article  CAS  PubMed  Google Scholar 

  53. Bave U, Nordmark G, Lovgren T, Ronnelid J, Cajander S, Eloranta ML, Alm GV, Ronnblom L (2005) Activation of the type I interferon system in primary Sjögren’s syndrome: a possible etiopathogenic mechanism. Arthritis Rheum 52: 1185–1195

    Article  CAS  PubMed  Google Scholar 

  54. Hjelmervik TO, Petersen K, Jonassen I, Jonsson R, Bolstad AI (2005) Gene expression profiling of minor salivary glands clearly distinguishes primary Sjögren’s syndrome patients from healthy control subjects. Arthritis Rheum 52: 1534–1544

    Article  CAS  PubMed  Google Scholar 

  55. Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T, Jacques S, Ba N, Ittah M, Lepajolec C et al. (2006) Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of primary Sjögren’s syndrome. Proc Natl Acad Sci USA 103: 2770–2775

    Article  CAS  PubMed  Google Scholar 

  56. Wildenberg ME, van Helden-Meeuwsen CG, van de Merwe JP, Drexhage HA, Versnel MA (2008) Systemic increase in type I interferon activity in Sjögren’s syndrome: a putative role for plasmacytoid dendritic cells. Eur J Immunol 38: 2024–2033

    Article  CAS  PubMed  Google Scholar 

  57. Wu AJ, Chen ZJ, Baum BJ, Ambudkar IS (1996) Interferon-gamma induces persistent depletion of internal Ca2+ stores in a human salivary gland cell line. Am J Physiol 270: C514–521

    CAS  PubMed  Google Scholar 

  58. Daniels PJ, Gustafson SA, French D, Wang Y, DePond W, McArthur CP (2000) Interferon-mediated block in cell cycle and altered integrin expression in a differentiated salivary gland cell line (HSG) cultured on Matrigel. J Interferon Cytokine Res 20: 1101–1109

    Article  CAS  PubMed  Google Scholar 

  59. Wu AJ, Kurrasch RH, Katz J, Fox PC, Baum BJ, Atkinson JC (1994) Effect of tumor necrosis factor-alpha and interferon-gamma on the growth of a human salivary gland cell line. J Cell Physiol 161: 217–226

    Article  CAS  PubMed  Google Scholar 

  60. Szekanecz Z, Strieter RM, Kunkel SL, Koch AE (1998) Chemokines in rheumatoid arthritis. Springer Semin Immunopathol 20: 115–132

    Article  CAS  PubMed  Google Scholar 

  61. Vergunst CE, van de Sande MG, Lebre MC, Tak PP (2005) The role of chemokines in rheumatoid arthritis and osteoarthritis. Scand J Rheumatol 34: 415–425

    Article  CAS  PubMed  Google Scholar 

  62. Kimura H, Caturegli P (2007) Chemokine orchestration of autoimmune thyroiditis. Thyroid 17: 1005–1011

    Article  CAS  PubMed  Google Scholar 

  63. Barone F, Bombardieri M, Rosado MM, Morgan PR, Challacombe SJ, De Vita S, Carsetti R, Spencer J, Valesini G, Pitzalis C (2008) CXCL13, CCL21, and CXCL12 Expression in Salivary Glands of Patients with Sjögren’s Syndrome and MALT Lymphoma: Association with Reactive and Malignant Areas of Lymphoid Organization. J Immunol 180: 5130–5140

    CAS  PubMed  Google Scholar 

  64. Saito M, Ota Y, Ohashi H, Dei Y, Shimoyama K, Suzuki D, Hayashi H, Ogawa N (2007) CD40-CD40 ligand signal induces the intercellular adhesion molecule-1 expression through nuclear factor-kappa B p50 in cultured salivary gland epithelial cells from patients with Sjögren’s syndrome. Mod Rheumatol 17: 45–53

    Article  CAS  PubMed  Google Scholar 

  65. Vanbervliet B, Bendriss-Vermare N, Massacrier C, Homey B, de Bouteiller O, Briere F, Trinchieri G, Caux C (2003) The inducible CXCR3 ligands control plasmacytoid dendritic cell responsiveness to the constitutive chemokine stromal cell-derived factor 1 (SDF-1)/CXCL12. J Exp Med 198: 823–830

    Article  CAS  PubMed  Google Scholar 

  66. Schiff M, Keiserman M, Codding C, Songcharoen S, Berman A, Nayiager S, Saldate C, Li T, Aranda R, Becker JC et al. (2008) Efficacy and safety of abatacept or infliximab vs placebo in ATTEST: a phase III, multi-centre, randomised, double-blind, placebo-controlled study in patients with rheumatoid arthritis and an inadequate response to methotrexate. Ann Rheum Dis 67: 1096–1103

    Article  CAS  PubMed  Google Scholar 

  67. Kremer JM, Genant HK, Moreland LW, Russell AS, Emery P, Abud-Mendoza C, Szechinski J, Li T, Teng J, Becker JC et al. (2008) Results of a two-year follow up study of patients with rheumatoid arthritis who received a combination of abatacept and methotrexate. Arthritis Rheum 58: 953–963

    Article  CAS  PubMed  Google Scholar 

  68. Leonardi C, Menter A, Hamilton T, Caro I, Xing B, Gottlieb AB (2008) Efalizumab: results of a 3-year continuous dosing study for the long-term control of psoriasis. Br J Dermatol 158: 1107–1116

    Article  CAS  PubMed  Google Scholar 

  69. Gupta AK, Cherman AM (2006) Efalizumab in the treatment of psoriasis. J Cutan Med Surg 10: 57–68

    PubMed  Google Scholar 

  70. Sharpe A (2005) Costimulation and regulation of autoimmunity and tolerance. J Pediatr Gastroenterol Nutr 40 Suppl 1: S20–21

    Google Scholar 

  71. Turkcapar N, Sak SD, Saatci M, Duman M, Olmez U (2005) Vasculitis and expression of vascular cell adhesion molecule-1, intercellular adhesion molecule-1, and E-selectin in salivary glands of patients with Sjögren’s syndrome. J Rheumatol 32: 1063–1070

    CAS  PubMed  Google Scholar 

  72. Balasa B, Krahl T, Patstone G, Lee J, Tisch R, McDevitt HO, Sarvetnick N (1997) CD40 ligand-CD40 interactions are necessary for the initiation of insulitis and diabetes in nonobese diabetic mice. J Immunol 159: 4620–4627

    CAS  PubMed  Google Scholar 

  73. Shan Z, Li J, Zheng C, Liu X, Fan Z, Zhang C, Goldsmith CM, Wellner RB, Baum BJ, Wang S (2005) Increased fluid secretion after adenoviral-mediated transfer of the human aquaporin-1 cDNA to irradiated miniature pig parotid glands. Mol Ther 11: 444–451

    Article  CAS  PubMed  Google Scholar 

  74. Effect of AdhAQP1 on salivary flow in patients treated with radiation for head and neck cancer (2009) Clinicaltrials.gov Identifier: NCT00372320

    Google Scholar 

  75. Cavill D, Waterman SA, Gordon TP (2004) Antibodies raised against the second extracellular loop of the human muscarinic M3 receptor mimic functional autoantibodies in Sjögren’s syndrome. Scand J Immunol 59: 261–266

    Article  CAS  PubMed  Google Scholar 

  76. Koo NY, Li J, Hwang SM, Choi SY, Lee SJ, Oh SB, Kim JS, Lee EB, Song YW, Park K (2008) Functional epitope of muscarinic type 3 receptor which interacts with autoantibodies from Sjögren’s syndrome patients. Rheumatology (Oxford) 47: 828–833

    Article  CAS  Google Scholar 

  77. Schegg V, Vogel M, Didichenko S, Stadler MB, Beleznay Z, Gadola S, Sengupta C, Stadler BM, Miescher SM (2008) Evidence that anti-muscarinic antibodies in Sjögren’s syndrome recognise both M3R and M1R. Biologicals 36: 213–222

    Article  CAS  PubMed  Google Scholar 

  78. Gao J, Cha S, Jonsson R, Opalko J, Peck AB (2004) Detection of anti-type 3 muscarinic acetylcholine receptor autoantibodies in the sera of Sjögren’s syndrome patients by use of a transfected cell line assay. Arthritis Rheum 50: 2615–2621

    Article  CAS  PubMed  Google Scholar 

  79. Dawson L, Tobin A, Smith P, Gordon T (2005) Antimuscarinic antibodies in Sjögren’s syndrome: where are we, and where are we going? Arthritis Rheum 52: 2984–2995

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2010 Springer Basel

About this chapter

Cite this chapter

Roescher, N., Tak, P.P., Chiorini, J.A. (2010). Gene therapy: Sjögren’s syndrome. In: Chernajovsky, Y., Robbins, P.D. (eds) Gene Therapy for Autoimmune and Inflammatory Diseases. Milestones in Drug Therapy. Springer, Basel. https://doi.org/10.1007/978-3-0346-0165-8_8

Download citation

Publish with us

Policies and ethics