197
Views
4
CrossRef citations to date
0
Altmetric
Original Article

Semaphorin-3A, semaphorin-7A gene single nucleotide polymorphisms, and systemic lupus erythematosus susceptibility

, , , , , , & show all
Pages 161-167 | Received 06 Apr 2019, Accepted 08 Jul 2019, Published online: 08 Aug 2019

References

  • Buyon JP. Systemic lupus erythematosus. In: Klippel JH, Stone JH, Crofford LJ, White PH, editors. Primer on the rheumatic diseases. New York (NY): Springer; 2008. p. 303–338.
  • Correa Freitas E, Evelyn Karnopp T, de Souza Silva JM, et al. Vitamin D supplementation ameliorates arthritis but does not alleviates renal injury in pristane-induced lupus model. Autoimmunity 2019;52:69–77.
  • Crispín J, Hedrich C, Tsokos G. Gene-function studies in systemic lupus erythematosus. Nat Rev Rheumatol. 2013;9:476–484.
  • Roth L, Koncina E, Satkauskas S, et al. The many faces of semaphorins: from development to pathology. Cell Mol Life Sci. 2009;66:649–666.
  • Wang LL, Zhang Y, Fan Y, et al. SEMA3A rs7804122 polymorphism is associated with Hirschsprung disease in the Northeastern region of China. Birth Defects Res A Clin Mol Teratol. 2012;94:91–95.
  • Suzuki K, Kumanogoh A, Kikutani H. Semaphorins and their receptors in immune cell interactions. Nat Immunol. 2008;9:17–23.
  • Liu LN, Li XM, Ye DQ, et al. Emerging role of semaphorin-3A in autoimmune diseases. Inflammopharmacology. 2018;26:655–665.
  • Lepelletier Y, Moura IC, Hadj-Slimane R, et al. Immunosuppressive role of semaphorin-3A on T cell proliferation is mediated by inhibition of actin cytoskeleton reorganization. Eur J Immunol. 2006;36:1782–1793.
  • Moretti S, Procopio A, Lazzarini R, et al. Semaphorin3A signaling controls Fas (CD95)-mediated apoptosis by promoting Fas translocation into lipid rafts. Blood. 2008;111:2290–2299.
  • Vadasz Z, Toubi E. Semaphorins: their dual role in regulating immune-mediated diseases. Clin Rev Allergy Immunol. 2014;47:17–25.
  • Gao H, Guo Q, Zou YD, et al. Expression and clinical significance of semaphorin 3A in serum and monouclear cells in patients with systemic lupus erythematous. Natl Med J China. 2017;97:370–374.
  • Takagawa SN, Kumagai K, Nagashima Y, et al. Decreased semaphorin3A expression correlates with disease activity and histological features of rheumatoid arthritis. BMC Musculoskelet Disord. 2013;14:40.
  • Rezaeepoor M, Shapoori S, Ganjalikhani-hakemi M, et al. Decreased expression of Sema3A, an immune modulator, in blood sample of multiple sclerosis patients. Gene. 2017;610:59–63.
  • Vadasz Z, Rimar D, Toubi E. A6.14 Semaphorin 3A, an immunoregulator and potential biomarker for disease severity in systemic sclerosis. Ann Rheum Dis. 2015;74:A61.61–A61.
  • Lange C, Liehr T, Goen M, et al. New eukaryotic semaphorins with close homology to semaphorins of DNA viruses. Genomics. 1998;51:340–350.
  • Mine T, Harada K, Matsumoto T, et al. CDw108 expression during T-cell development. Tissue Antigens. 2000;55:429–436.
  • Suzuki K, Okuno T, Yamamoto M, et al. Semaphorin 7A initiates T-cell-mediated inflammatory responses through alpha1beta1 integrin. Nature. 2007;446:680–684.
  • Mizui M, Kumanogoh A, Kikutani H. Immune semaphorins: novel features of neural guidance molecules. J Clin Immunol. 2009;29:1–11.
  • Holmes S, Downs AM, Fosberry A, et al. Sema7A is a potent monocyte stimulator. Scand J Immunol. 2002;56:270–275.
  • Czopik A, Bynoe M, Palm N, et al. Semaphorin 7A is a negative regulator of T cell responses. Immunity. 2006;24:591–600.
  • Xie J, Wang H. Semaphorin 7A as a potential immune regulator and promising therapeutic target in rheumatoid arthritis. Arthritis Res Ther. 2017;19:10.
  • Gutiérrez-Franco A, Eixarch H, Costa C, et al. Semaphorin 7A as a potential therapeutic target for multiple sclerosis. Mol Neurobiol. 2017;54:4820–4831.
  • Eixarch H, Gutiérrez-Franco A, Montalban X, et al. Semaphorins 3A and 7A: potential immune and neuroregenerative targets in multiple sclerosis. Trends Mol Med. 2013;19:157–164.
  • Kremer D, Hartung H, Küry P. Targeting semaphorins in MS as a treatment strategy to promote remyelination: a tale of mice, rats and men. Mult Scler. 2015;21:1616–1617.
  • Cantó E, Tintoré M, Villar L, et al. Validation of semaphorin 7A and ala-β-his-dipeptidase as biomarkers associated with the conversion from clinically isolated syndrome to multiple sclerosis. J Neuroinflammation. 2014;11:181.
  • Hochberg M. Updating the American College of Rheumatology revised criteria for the classification of systemic lupus erythematosus. Arthritis Rheum. 1997;40:1725.
  • Romero-Diaz J, Isenberg D, Ramsey-Goldman R. Measures of adult systemic lupus erythematosus: updated version of British Isles Lupus Assessment Group (BILAG 2004), European Consensus Lupus Activity Measurements (ECLAM), Systemic Lupus Activity Measure, Revised (SLAM-R), Systemic Lupus Activity Questionnaire for Population Studies (SLAQ), Systemic Lupus Erythematosus Disease Activity Index 2000 (SLEDAI-2K), and Systemic Lupus International Collaborating Clinics/American College of Rheumatology Damage Index (SDI). Arthritis Care Res. 2011;63:20572.
  • Wu Y, Zhang F, Ma J, et al. Association of large intergenic noncoding RNA expression with disease activity and organ damage in systemic lupus erythematosus. Arthritis Res Ther. 2015;17:015–0632.
  • Aken BL, Achuthan P, Akanni W, et al. Ensembl 2017. Nucleic Acids Res. 2017;45:D635–D642.
  • Barrett JC, Fry B, Maller J, et al. Haploview: analysis and visualization of LD and haplotype maps. Bioinformatics. 2005;21:263–265.
  • Lee PH, Shatkay H. F-SNP: computationally predicted functional SNPs for disease association studies. Nucleic Acids Res. 2008;36:5.
  • Landu S, Everitt BS. A handbook of statistical analysis using SPSS. Boca Raton (FL): Chapman and Hall/CRC; 2004.
  • Li Z, Zhang Z, He Z, et al. A partition-ligation-combination-subdivision EM algorithm for haplotype inference with multiallelic markers: update of the SHEsis. Cell Res. 2009;19:519–523. http://analysis.bio-x.cn.
  • Zan H. Epigenetics in lupus. Autoimmunity. 2014;47:213–214.
  • Scarlato M, Ara J, Bannerman P, et al. Induction of neuropilins-1 and -2 and their ligands, Sema3A, Sema3F, and VEGF, during Wallerian degeneration in the peripheral nervous system. Exp Neurol. 2003;183:489–498.
  • Kawasaki T, Bekku Y, Suto F, et al. Requirement of neuropilin 1-mediated Sema3A signals in patterning of the sympathetic nervous system. Development. 2002;129:671–680.
  • Villa C, Venturelli E, Fenoglio C, et al. Candidate gene analysis of semaphorins in patients with Alzheimer’s disease. Neurol Sci. 2010;31:169–173.
  • Miao H, Soker S, Feiner L, et al. Neuropilin-1 mediates collapsin-1/semaphorin III inhibition of endothelial cell motility: functional competition of collapsin-1 and vascular endothelial growth factor-165. J Cell Biol. 1999;146:233–242.
  • Toubi ZVE. Semaphorins: their dual role in regulating immune-mediated diseases. Clin Rev Allerg Immunol. 2014;47:17–25.
  • Zahava Vadasz TH, Halasz K, Rosner I, et al. Semaphorin 3A is a marker for disease activity and a potential immunoregulator in systemic lupus erythematosus. Arthritis Res Ther. 2012;14:R146.
  • Kessel A, Peri R, Haj T, et al. IVIg attenuates TLR-9 activation in B cells from SLE patients. J Clin Immunol. 2011;31:30–38.
  • Perez SG, Malvar-Fernández B, Newsom SP, et al. A7.09 Class 3 semaphorins modulate the invasive capacity of rheumatoid arthritis fibroblast-like synoviocytes. Ann Rheum Dis. 2016;75:A59.1–A59.
  • Okuno T, Nakatsuji Y, Kumanogoh A. The role of immune semaphorins in multiple sclerosis. FEBS Lett. 2011;585:3829–3835.
  • Okubo M, Kimura T, Fujita Y, et al. Semaphorin 3A is expressed in human osteoarthritic cartilage and antagonizes vascular endothelial growth factor 165-promoted chondrocyte migration: an implication for chondrocyte cloning. Arthritis Rheum. 2011;63:3000–3009.
  • Rimar D, Nov Y, Rosner I, et al. Semaphorin 3A: an immunoregulator in systemic sclerosis. Rheumatol Int. 2015;35:1625–1630.
  • Black SA, Nelson AC, Gurule NJ, et al. Semaphorin 7a exerts pleiotropic effects to promote breast tumor progression. Oncogene. 2016;35:5170.
  • Delorme G, Saltel F, Bonnelye E, et al. Expression and function of semaphorin 7A in bone cells. Biol Cell. 2005;97:589–597.
  • Gan Y, Reilkoff R, Peng X, et al. Role of semaphorin 7a signaling in transforming growth factor β1-induced lung fibrosis and scleroderma-related interstitial lung disease. Arthritis Rheum. 2011;63:2484–2494.
  • Costa C, Martínez-Sáez E, Gutiérrez-Franco A, et al. Expression of semaphorin 3A, semaphorin 7A and their receptors in multiple sclerosis lesions. Mult Scler. 2015;21:1632–1643.
  • Wang LL, Fan Y, Zhou FH, et al. Semaphorin 3A expression in the colon of Hirschsprung disease. Birth Defects Res Part A Clin Mol Teratol. 2011;91:842–847.
  • Li Q, Zhang Z, Diao M, et al. Cumulative risk impact of RET, SEMA3, and NRG1 polymorphisms associated with Hirschsprung disease in Han Chinese. J Pediatr Gastroenterol Nutr. 2017;64:385–390.
  • Koh JM, Oh B, Lee JY, et al. Association study of semaphorin 7a (sema7a) polymorphisms with bone mineral density and fracture risk in postmenopausal Korean women. J Hum Genet. 2006;51:112–117.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.