#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Identification of , a Locus Controlling Dominant Resistance to Autoimmune Orchitis, as Kinesin Family Member 1C


Experimental autoimmune orchitis (EAO), the principal model of non-infectious testicular inflammatory disease, can be induced in susceptible mouse strains by immunization with autologous testicular homogenate and appropriate adjuvants. As previously established, the genome of DBA/2J mice encodes genes that are capable of conferring dominant resistance to EAO, while the genome of BALB/cByJ mice does not and they are therefore susceptible to EAO. In a genome scan, we previously identified Orch3 as the major quantitative trait locus controlling dominant resistance to EAO and mapped it to chromosome 11. Here, by utilizing a forward genetic approach, we identified kinesin family member 1C (Kif1c) as a positional candidate for Orch3 and, using a transgenic approach, demonstrated that Kif1c is Orch3. Mechanistically, we showed that the resistant Kif1cD2 allele leads to a reduced antigen-specific T cell proliferative response as a consequence of decreased MHC class II expression by antigen presenting cells, and that the L578→P578 and S1027→P1027 polymorphisms distinguishing the BALB/cByJ and DBA/2J alleles, respectively, can play a role in transcriptional regulation. These findings may provide mechanistic insight into how polymorphism in other kinesins such as KIF21B and KIF5A influence susceptibility and resistance to human autoimmune diseases.


Vyšlo v časopise: Identification of , a Locus Controlling Dominant Resistance to Autoimmune Orchitis, as Kinesin Family Member 1C. PLoS Genet 8(12): e32767. doi:10.1371/journal.pgen.1003140
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1003140

Souhrn

Experimental autoimmune orchitis (EAO), the principal model of non-infectious testicular inflammatory disease, can be induced in susceptible mouse strains by immunization with autologous testicular homogenate and appropriate adjuvants. As previously established, the genome of DBA/2J mice encodes genes that are capable of conferring dominant resistance to EAO, while the genome of BALB/cByJ mice does not and they are therefore susceptible to EAO. In a genome scan, we previously identified Orch3 as the major quantitative trait locus controlling dominant resistance to EAO and mapped it to chromosome 11. Here, by utilizing a forward genetic approach, we identified kinesin family member 1C (Kif1c) as a positional candidate for Orch3 and, using a transgenic approach, demonstrated that Kif1c is Orch3. Mechanistically, we showed that the resistant Kif1cD2 allele leads to a reduced antigen-specific T cell proliferative response as a consequence of decreased MHC class II expression by antigen presenting cells, and that the L578→P578 and S1027→P1027 polymorphisms distinguishing the BALB/cByJ and DBA/2J alleles, respectively, can play a role in transcriptional regulation. These findings may provide mechanistic insight into how polymorphism in other kinesins such as KIF21B and KIF5A influence susceptibility and resistance to human autoimmune diseases.


Zdroje

1. Lustig L, Tung KSK (2006) The autoimmune diseases. Missouri: Elsevier-Academic Press. 841–848 p.

2. Tung KSK, Fusi F, Teuscher C (2002) Autoimmune disease of the speermatozoa, ovary and testis; USA: Routledge. 1031–1045 p.

3. KohnoS, MunozJA, WilliamsTM, TeuscherC, BernardCC, et al. (1983) Immunopathology of murine experimental allergic orchitis. J Immunol 130: 2675–2682.

4. TungKS, TeuscherC (1995) Mechanisms of autoimmune disease in the testis and ovary. Hum Reprod Update 1: 35–50.

5. YuleTD, TungKS (1993) Experimental autoimmune orchitis induced by testis and sperm antigen-specific T cell clones: an important pathogenic cytokine is tumor necrosis factor. Endocrinology 133: 1098–1107.

6. JacoboP, GuazzoneVA, Jarazo-DietrichS, TheasMS, LustigL (2009) Differential changes in CD4+ and CD8+ effector and regulatory T lymphocyte subsets in the testis of rats undergoing autoimmune orchitis. J Reprod Immunol 81: 44–54.

7. JacoboP, PerezCV, TheasMS, GuazzoneVA, LustigL (2011) CD4+ and CD8+ T cells producing Th1 and Th17 cytokines are involved in the pathogenesis of autoimmune orchitis. Reproduction 141: 249–258.

8. TeuscherC, BlankenhornEP, HickeyWF (1987) Differential susceptibility to actively induced experimental allergic encephalomyelitis and experimental allergic orchitis among BALB/c substrains. Cell Immunol 110: 294–304.

9. TeuscherC, SmithSM, GoldbergEH, ShearerGM, TungKS (1985) Experimental allergic orchitis in mice. I. Genetic control of susceptibility and resistance to induction of autoimmune orchitis. Immunogenetics 22: 323–333.

10. MeekerND, HickeyWF, KorngoldR, HansenWK, SudweeksJD, et al. (1995) Multiple loci govern the bone marrow-derived immunoregulatory mechanism controlling dominant resistance to autoimmune orchitis. Proc Natl Acad Sci U S A 92: 5684–5688.

11. RazaviR, ChanY, AfifiyanFN, LiuXJ, WanX, et al. (2006) TRPV1+ sensory neurons control beta cell stress and islet inflammation in autoimmune diabetes. Cell 127: 1123–1135.

12. MacMickingJ, XieQW, NathanC (1997) Nitric oxide and macrophage function. Annu Rev Immunol 15: 323–350.

13. NagyG, KonczA, TelaricoT, FernandezD, ErsekB, et al. (2010) Central role of nitric oxide in the pathogenesis of rheumatoid arthritis and systemic lupus erythematosus. Arthritis Res Ther 12: 210.

14. BoydenED, DietrichWF (2006) Nalp1b controls mouse macrophage susceptibility to anthrax lethal toxin. Nat Genet 38: 240–244.

15. KoppP, LammersR, AepfelbacherM, WoehlkeG, RudelT, et al. (2006) The kinesin KIF1C and microtubule plus ends regulate podosome dynamics in macrophages. Mol Biol Cell 17: 2811–2823.

16. WubboltsR, Fernandez-BorjaM, JordensI, ReitsE, DusseljeeS, et al. (1999) Opposing motor activities of dynein and kinesin determine retention and transport of MHC class II-containing compartments. J Cell Sci 112 ((Pt 6))

785–795.

17. WattersJW, DewarK, LehoczkyJ, BoyartchukV, DietrichWF (2001) Kif1C, a kinesin-like motor protein, mediates mouse macrophage resistance to anthrax lethal factor. Curr Biol 11: 1503–1511.

18. DornerC, CiossekT, MullerS, MollerPH, UllrichA, et al. (1998) Characterization of KIF1C, a new kinesin-like protein involved in vesicle transport from the Golgi apparatus to the endoplasmic reticulum. J Biol Chem 273: 20267–20275.

19. KirchnerJ, SeilerS, FuchsS, SchliwaM (1999) Functional anatomy of the kinesin molecule in vivo. Embo J 18: 4404–4413.

20. SchlagerMA, KapiteinLC, GrigorievI, BurzynskiGM, WulfPS, et al. (2010) Pericentrosomal targeting of Rab6 secretory vesicles by Bicaudal-D-related protein 1 (BICDR-1) regulates neuritogenesis. Embo J 29: 1637–1651.

21. MachoB, BrancorsiniS, FimiaGM, SetouM, HirokawaN, et al. (2002) CREM-dependent transcription in male germ cells controlled by a kinesin. Science 298: 2388–2390.

22. ChennathukuzhiV, MoralesCR, El-AlfyM, HechtNB (2003) The kinesin KIF17b and RNA-binding protein TB-RBP transport specific cAMP-responsive element modulator-regulated mRNAs in male germ cells. Proc Natl Acad Sci U S A 100: 15566–15571.

23. HedrichCM, RauenT, TsokosGC (2011) cAMP-responsive element modulator (CREM)alpha protein signaling mediates epigenetic remodeling of the human interleukin-2 gene: implications in systemic lupus erythematosus. J Biol Chem 286: 43429–43436.

24. SnoekM, JansenM, OlavesenMG, CampbellRD, TeuscherC, et al. (1993) Three Hsp70 genes are located in the C4-H-2D region: possible candidates for the Orch-1 locus. Genomics 15: 350–356.

25. TeuscherC, GasserDL, WoodwardSR, HickeyWF (1990) Experimental allergic orchitis in mice. VI. Recombinations within the H-2S/H-2D interval define the map position of the H-2-associated locus controlling disease susceptibility. Immunogenetics 32: 337–344.

26. ButterfieldRJ, SudweeksJD, BlankenhornEP, KorngoldR, MariniJC, et al. (1998) New genetic loci that control susceptibility and symptoms of experimental allergic encephalomyelitis in inbred mice. J Immunol 161: 1860–1867.

27. ButterfieldRJ, BlankenhornEP, RoperRJ, ZacharyJF, DoergeRW, et al. (2000) Identification of genetic loci controlling the characteristics and severity of brain and spinal cord lesions in experimental allergic encephalomyelitis. Am J Pathol 157: 637–645.

28. TeuscherC, HickeyWF, GraferCM, TungKS (1998) A common immunoregulatory locus controls susceptibility to actively induced experimental allergic encephalomyelitis and experimental allergic orchitis in BALB/c mice. J Immunol 160: 2751–2756.

29. ShawMH, BoyartchukV, WongS, KaraghiosoffM, RagimbeauJ, et al. (2003) A natural mutation in the Tyk2 pseudokinase domain underlies altered susceptibility of B10.Q/J mice to infection and autoimmunity. Proc Natl Acad Sci U S A 100: 11594–11599.

30. SpachKM, NoubadeR, McElvanyB, HickeyWF, BlankenhornEP, et al. (2009) A single nucleotide polymorphism in Tyk2 controls susceptibility to experimental allergic encephalomyelitis. J Immunol 182: 7776–7783.

31. WandstratA, WakelandE (2001) The genetics of complex autoimmune diseases: non-MHC susceptibility genes. Nat Immunol 2: 802–809.

32. GabrilovichDI, NagarajS (2009) Myeloid-derived suppressor cells as regulators of the immune system. Nat Rev Immunol 9: 162–174.

33. GeissmannF, ManzMG, JungS, SiewekeMH, MeradM, et al. (2010) Development of monocytes, macrophages, and dendritic cells. Science 327: 656–661.

34. FuW, WojtkiewiczG, WeisslederR, BenoistC, MathisD (2012) Early window of diabetes determinism in NOD mice, dependent on the complement receptor CRIg, identified by noninvasive imaging. Nat Immunol 13: 361–368.

35. Moline-VelazquezV, CuervoH, Vila-Del SolV, OrtegaMC, ClementeD, et al. (2011) Myeloid-derived suppressor cells limit the inflammation by promoting T lymphocyte apoptosis in the spinal cord of a murine model of multiple sclerosis. Brain Pathol 21: 678–691.

36. WeberMS, Prod'hommeT, YoussefS, DunnSE, RundleCD, et al. (2007) Type II monocytes modulate T cell-mediated central nervous system autoimmune disease. Nat Med 13: 935–943.

37. RicherMJ, LavalleeDJ, ShaninaI, HorwitzMS (2012) Immunomodulation of antigen presenting cells promotes natural regulatory T cells that prevent autoimmune diabetes in NOD mice. PLoS ONE 7: e31153 doi:10.1371/journal.pone.0031153.

38. BernasconiP, CappellettiC, NavoneF, NessiV, BaggiF, et al. (2008) The kinesin superfamily motor protein KIF4 is associated with immune cell activation in idiopathic inflammatory myopathies. J Neuropathol Exp Neurol 67: 624–632.

39. StagiM, GorlovoyP, LarionovS, TakahashiK, NeumannH (2006) Unloading kinesin transported cargoes from the tubulin track via the inflammatory c-Jun N-terminal kinase pathway. Faseb J 20: 2573–2575.

40. McCauleyJL, ZuvichRL, BeechamAL, De JagerPL, KonidariI, et al. (2010) Comprehensive follow-up of the first genome-wide association study of multiple sclerosis identifies KIF21B and TMEM39A as susceptibility loci. Hum Mol Genet 19: 953–962.

41. AlcinaA, VandenbroeckK, OtaeguiD, SaizA, GonzalezJR, et al. (2010) The autoimmune disease-associated KIF5A, CD226 and SH2B3 gene variants confer susceptibility for multiple sclerosis. Genes Immun 11: 439–445.

42. HirokawaN, NodaY, TanakaY, NiwaS (2009) Kinesin superfamily motor proteins and intracellular transport. Nat Rev Mol Cell Biol 10: 682–696.

43. NakagawaT, TanakaY, MatsuokaE, KondoS, OkadaY, et al. (1997) Identification and classification of 16 new kinesin superfamily (KIF) proteins in mouse genome. Proc Natl Acad Sci U S A 94: 9654–9659.

44. NakajimaK, TakeiY, TanakaY, NakagawaT, NakataT, et al. (2002) Molecular motor KIF1C is not essential for mouse survival and motor-dependent retrograde Golgi apparatus-to-endoplasmic reticulum transport. Mol Cell Biol 22: 866–873.

45. McAllisterRD, SinghY, du BoisWD, PotterM, BoehmT, et al. (2003) Susceptibility to anthrax lethal toxin is controlled by three linked quantitative trait loci. Am J Pathol 163: 1735–1741.

46. BoilleeS, YamanakaK, LobsigerCS, CopelandNG, JenkinsNA, et al. (2006) Onset and progression in inherited ALS determined by motor neurons and microglia. Science 312: 1389–1392.

47. WildinRS, GarvinAM, PawarS, LewisDB, AbrahamKM, et al. (1991) Developmental regulation of lck gene expression in T lymphocytes. J Exp Med 173: 383–393.

48. NoubadeR, MilliganG, ZacharyJF, BlankenhornEP, del RioR, et al. (2007) Histamine receptor H1 is required for TCR-mediated p38 MAPK activation and optimal IFN-gamma production in mice. J Clin Invest 117: 3507–3518.

49. Affymetrix (2005–2006) GeneChip Expression Analysis Technical Manual.

50. BolstadBM, IrizarryRA, AstrandM, SpeedTP (2003) A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics 19: 185–193.

51. IrizarryRA, BolstadBM, CollinF, CopeLM, HobbsB, et al. (2003) Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res 31: e15.

52. BenjaminiY, DraiD, ElmerG, KafkafiN, GolaniI (2001) Controlling the false discovery rate in behavior genetics research. Behav Brain Res 125: 279–284.

53. PoppRA, BailiffEG, SkowLC, WhitneyJB3rd (1982) The primary structure of genetic variants of mouse hemoglobin. Biochem Genet 20: 199–208.

54. WattersJW, DietrichWF (2001) Genetic, physical, and transcript map of the Ltxs1 region of mouse chromosome 11. Genomics 73: 223–231.

55. RoderickTH, HuttonJJ, RuddleFH (1970) Linkage of esterase-3 and rex on linkage group VII of the mouse. J Hered 61: 278–279.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2012 Číslo 12
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#