#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Mechanistically Distinct Mouse Models for -Associated Retinopathy


Cone-rod homeobox (CRX) protein is a “paired-like” homeodomain transcription factor that is essential for regulating rod and cone photoreceptor transcription. Mutations in human CRX are associated with the dominant retinopathies Retinitis Pigmentosa (RP), Cone-Rod Dystrophy (CoRD) and Leber Congenital Amaurosis (LCA), with variable severity. Heterozygous Crx Knock-Out (KO) mice (“+/−”) have normal vision as adults and fail to model the dominant human disease. To investigate how different mutant CRX proteins produce distinct disease pathologies, we generated two Crx Knock-IN (K-IN) mouse models: CrxE168d2 (“E168d2”) and CrxR90W (“R90W”). E168d2 mice carry a frameshift mutation in the CRX activation domain, Glu168del2, which is associated with severe dominant CoRD or LCA in humans. R90W mice carry a substitution mutation in the CRX homeodomain, Arg90Trp, which is associated with dominant mild late-onset CoRD and recessive LCA. As seen in human patients, heterozygous E168d2 (“E168d2/+”) but not R90W (“R90W/+”) mice show severely impaired retinal function, while mice homozygous for either mutation are blind and undergo rapid photoreceptor degeneration. E168d2/+ mice also display abnormal rod/cone morphology, greater impairment of CRX target gene expression than R90W/+ or +/− mice, and undergo progressive photoreceptor degeneration. Surprisingly, E168d2/+ mice express more mutant CRX protein than wild-type CRX. E168d2neo/+, a subline of E168d2 with reduced mutant allele expression, displays a much milder retinal phenotype, demonstrating the impact of Crx expression level on disease severity. Both CRX[E168d2] and CRX[R90W] proteins fail to activate transcription in vitro, but CRX[E168d2] interferes more strongly with the function of wild type (WT) CRX, supporting an antimorphic mechanism. E168d2 and R90W are mechanistically distinct mouse models for CRX-associated disease that will allow the elucidation of molecular mechanisms and testing of novel therapeutic approaches for different forms of CRX-associated disease.


Vyšlo v časopise: Mechanistically Distinct Mouse Models for -Associated Retinopathy. PLoS Genet 10(2): e32767. doi:10.1371/journal.pgen.1004111
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.pgen.1004111

Souhrn

Cone-rod homeobox (CRX) protein is a “paired-like” homeodomain transcription factor that is essential for regulating rod and cone photoreceptor transcription. Mutations in human CRX are associated with the dominant retinopathies Retinitis Pigmentosa (RP), Cone-Rod Dystrophy (CoRD) and Leber Congenital Amaurosis (LCA), with variable severity. Heterozygous Crx Knock-Out (KO) mice (“+/−”) have normal vision as adults and fail to model the dominant human disease. To investigate how different mutant CRX proteins produce distinct disease pathologies, we generated two Crx Knock-IN (K-IN) mouse models: CrxE168d2 (“E168d2”) and CrxR90W (“R90W”). E168d2 mice carry a frameshift mutation in the CRX activation domain, Glu168del2, which is associated with severe dominant CoRD or LCA in humans. R90W mice carry a substitution mutation in the CRX homeodomain, Arg90Trp, which is associated with dominant mild late-onset CoRD and recessive LCA. As seen in human patients, heterozygous E168d2 (“E168d2/+”) but not R90W (“R90W/+”) mice show severely impaired retinal function, while mice homozygous for either mutation are blind and undergo rapid photoreceptor degeneration. E168d2/+ mice also display abnormal rod/cone morphology, greater impairment of CRX target gene expression than R90W/+ or +/− mice, and undergo progressive photoreceptor degeneration. Surprisingly, E168d2/+ mice express more mutant CRX protein than wild-type CRX. E168d2neo/+, a subline of E168d2 with reduced mutant allele expression, displays a much milder retinal phenotype, demonstrating the impact of Crx expression level on disease severity. Both CRX[E168d2] and CRX[R90W] proteins fail to activate transcription in vitro, but CRX[E168d2] interferes more strongly with the function of wild type (WT) CRX, supporting an antimorphic mechanism. E168d2 and R90W are mechanistically distinct mouse models for CRX-associated disease that will allow the elucidation of molecular mechanisms and testing of novel therapeutic approaches for different forms of CRX-associated disease.


Zdroje

1. ChenS, WangQL, NieZ, SunH, LennonG, et al. (1997) Crx, a novel Otx-like paired-homeodomain protein, binds to and transactivates photoreceptor cell-specific genes. Neuron 19(5): 1017–30.

2. FurukawaT, MorrowEM, CepkoCL (1997) Crx, a novel otx-like homeobox gene, shows photoreceptor-specific expression and regulates photoreceptor differentiation. Cell 91: 531–541.

3. FurukawaT, MorrowEM, LiT, DavisFC, CepkoCL (1999) Retinopathy and attenuated circadian entrainment in Crx-deficient mice. Nat Genet 23(4): 466–470.

4. ChauKY, ChenS, ZackDJ, OnoSJ (2000) Functional domains of the cone-rod homeobox (CRX) transcription factor. The Journal of biological chemistry 275(47): 37264–70.

5. KimuraA, SinghD, WawrousekE, KikuchiM, NakamuraM, et al. (2000) Both PCE-1/RX and OTX/CRX Interactions Are Necessary for Photoreceptor-specific Gene Expression. J Biol Chem 275: 1152–1160.

6. LangmannT, LaiCCL, WeigeltK, TamBM, Warneke-WittstockR, et al. (2008) CRX controls retinal expression of the X-linked juvenile retinoschisis (RS1) gene. Nucleic acids research 36(20): 6523–34.

7. PengGH, ChenS (2007) Crx activates opsin transcription by recruiting HAT-containing co-activators and promoting histone acetylation. Hum Mol Genet 16(20): 2433–2452.

8. HennigAK, PengGH, ChenS (2008) Regulation of photoreceptor gene expression by Crx-associated transcription factor network. Brain Res 1192: 114–133.

9. PengGH, ChenS (2011) Active opsin loci adopt intrachromosomal loops that depend on the photoreceptor transcription factor network. Proc Natl Acad Sci USA 108(43): 17821–6.

10. FeiY, HughesTE (2000) Nuclear trafficking of photoreceptor protein crx: the targeting sequence and pathologic implications. Investigative ophthalmology & visual science 41(10): 2849–56.

11. MittonKP, SwainPK, ChenS, XuS, ZackDJ, et al. (2000) The leucine zipper of NRL interacts with the CRX homeodomain. A possible mechanism of transcriptional synergy in rhodopsin regulation. J Biol Chem 275(38): 29794–9.

12. NicholsLL, AlurRP, BoobalanE, SergeevYV, CarusoRC, et al. (2010) Two novel CRX mutant proteins causing autosomal dominant Leber congenital amaurosis interact differently with NRL. Human mutation 31(6): E1472–83.

13. PengGH, AhmadO, AhmadF, LiuJ, ChenS (2005) The photoreceptor-specific nuclear receptor Nr2e3 interacts with Crx and exerts opposing effects on the transcription of rod versus cone genes. Hum Mol Genet 14: 747–764.

14. RoduitR, EscherP, SchorderetDF (2009) Mutations in the DNA-binding domain of NR2E3 affect in vivo dimerization and interaction with CRX. PLoS ONE 4(10): e7379.

15. SanyalS, JansenHG (1981) Absence of receptor outer segments in the retina of rds mutant mice. Neurosci Lett 21: 23–26.

16. HumphriesMH, RancourtD, FarrarGJ, KennaP, HazelM, et al. (1997) Retinopathy induced in mice by targeted disruption of the rhodopsin gene. Nature Genet 15: 216–219.

17. MorrowEM, FurukawaT, RaviolaE, CepkoCL (2005) Synaptogenesis and outer segment formation are perturbed in the neural retina of Crx mutant mice. BMC Neurosci 6: 5.

18. LiveseyFJ, FurukawaT, SteffenMA, ChurchGM, CepkoCL (2000) Microarray analysis of the transcriptional network controlled by the photoreceptor homeobox gene Crx. Curr Biol 10(6): 301–310.

19. HsiauTH, DiaconuC, MyersCA, LeeJ, CepkoCL, et al. (2007) The cis-regulatory logic of the mammalian photoreceptor transcriptional network. PLoS ONE 2(7): 2e643.

20. BlackshawS, FraioliRE, FurukawaT, CepkoCL (2001) Comprehensive analysis of photoreceptor gene expression and the identification of candidate retinal disease genes. Cell 107(5): 579–589.

21. CorboJC, LawrenceKA, KarlstetterM, MyersCA, AbdelazizM, et al. (2010) CRX ChIP-seq reveals the cis-regulatory architecture of mouse photoreceptors. Genome Research 20(11): 1512–1525.

22. RivoltaC, BersonEL, DryjaTP (2001) Dominant Leber congenital amaurosis, cone-rod degeneration, and retinitis pigmentosa caused by mutant versions of the transcription factor CRX. Human mutation 18(6): 488–98.

23. SohockiMM, SullivanLS, Mintz-HittnerHA, BirchD, HeckenlivelyJR, et al. (1998) A range of clinical phenotypes associated with mutations in CRX, a photoreceptor transcription-factor gene. Am J Hum Genet 63(5): 1307–1315.

24. JacobsonSG, CideciyanAV, HuangY, HannaDB, FreundCL, et al. (1998) Retinal degenerations with truncation mutations in the cone-rod homeobox (CRX) gene. Invest Ophthalmol Vis Sci 39(12): 2417–2426.

25. HaneinS, PerraultI, GerberS, TanguyG, BarbetF, et al. (2004) Leber congenital amaurosis: comprehensive survey of the genetic heterogeneity, refinement of the clinical definition, and genotype-phenotype correlations as a strategy for molecular diagnosis. Human mutation 23: 306–317.

26. DharmarajSR, SilvaER, Pina aL, LiYY, YangJM, et al. (2000) Mutational analysis and clinical correlation in Leber congenital amaurosis. Ophthalmic genetics 21: 135–150.

27. GalvinJA, FishmanGA, StoneEM, KoenekoopRK (2005) Evaluation of genotype-phenotype associations in Leber Congenital Amaurosis. Retina 25(7): 919–29.

28. PerraultI, HaneinS, GerberS, BarbetF, DufierJL, et al. (2003) Evidence of autosomal dominant Leber congenital amaurosis (LCA) underlain by a CRX heterozygous null allele. J Med Genet 40(7): e90.

29. NakamuraM, ItoS, MiyakeY (1998) Novel De Novo Mutation in CRX Gene in a Japanese Patient with Leber Congenital Amaurosis. American Journal of Ophthalmology 24: 465–467.

30. ZhangQ, LiS, GuoX, GuoL, XiaoX, et al. (2001) Screening for CRX gene mutations in Chinese patients with Leber congenital amaurosis and mutational phenotype. Ophthalmic Genet 22: 89–96.

31. LoteryAJ, NamperumalsamyP, JacobsonSG, WeleberRG, FishmanGA, et al. (2000) Mutation analysis of 3 genes in patients with Leber congenital amaurosis. Archives of ophthalmology 118(4): 538–43.

32. WangP, GuoX, ZhangQ (2007) Further evidence of autosomal-dominant Leber congenital amaurosis caused by heterozygous CRX mutation. Graefe's archive for clinical and experimental ophthalmology 245: 1401–1402.

33. HuangL, XiaoX, LiS, JiaX, WangP, et al. (2012) CRX variants in cone-rod dystrophy and mutation overview. Biochemical and biophysical research communications 426(4): 498–503.

34. TzekovRT, LiuY, SohockiMM, ZackDJ, DaigerSP, et al. (2001) Autosomal dominant retinal degeneration and bone loss in patients with a 12-bp deletion in the CRX gene. Investigative ophthalmology & visual science 42: 1319–1327.

35. KoenekoopRK, LoyerM, DembinskaO, BeneishR (2002) Mutation report Visual improvement in Leber congenital amaurosis and the CRX genotype. Ophthalmic Genet 23: 49–60.

36. PaunescuK, PreisingMN, JankeB, WissingerB, LorenzB (2007) Genotype-phenotype correlation in a German family with a novel complex CRX mutation extending the open reading frame. Ophthalmology 114: 1348–1357.e1.

37. TzekovRT, SohockiMM, DaigerSP, BirchDG (2000) Visual phenotype in patients with Arg41Gln and ala196+1 bp mutations in the CRX gene. Ophthalmic genetics 21(2): 89–99.

38. WaliaS, FishmanGA, JacobsonSG, AlemanTS, KoenekoopRK, et al. (2010) Visual acuity in patients with Leber's congenital amaurosis and early childhood-onset retinitis pigmentosa. Ophthalmology 117: 1190–1198.

39. den HollanderAI, RoepmanR, KoenekoopRK, CremersFPM (2008) Leber congenital amaurosis: genes, proteins and disease mechanisms. Progress in retinal and eye research 27: 391–419.

40. FreundCL, Gregory-EvansCY, FurukawaT, PapaioannouM, LooserJ, et al. (1997) Cone-rod dystrophy due to mutations in a novel photoreceptor-specific homeobox gene (CRX) essential for maintenance of the photoreceptor. Cell 91(4): 543–553.

41. FreundC, WangQL, ChenS, MuskatB, WilesC, et al. (1998) De novo mutations in the CRX homeobox gene associated with Leber congenital amaurosis. Nature genetics 18(4): 311–312.

42. SwainPK, ChenS, WangQL, AffatigatoLM, CoatsCL, et al. (1997) Mutations in the cone-rod homeobox gene are associated with the cone-rod dystrophy photoreceptor degeneration. Neuron 19(6): 1329–36.

43. BergerW, Kloeckener-GruissemB, NeidhardtJ (2010) The molecular basis of human retinal and vitreoretinal diseases. Progress in retinal and eye research 29(5): 335–75.

44. SilvaE, YangJM, LiY, DarmarajS, OhS, et al. (2000) A CRX null mutation is associated with both Leber congenital amaurosis and a normal ocular phenotype. Invest Ophthalmol Vis Sci 41: 2076–2079.

45. SwaroopA, WangQL, WuW, CookJ, CoatsC, et al. (1999) Leber congenital amaurosis caused by a homozygous mutation (R90W) in the homeodomain of the retinal transcription factor CRX: direct evidence for the involvement of CRX in the development of photoreceptor function. Hum Mol Genet 8(2): 299–305.

46. TerrellD, XieB, WorkmanM, MahatoS, ZelhofA, et al. (2012) OTX2 and CRX rescue overlapping and photoreceptor-specific functions in the Drosophila eye. Developmental dynamics: an official publication of the American Association of Anatomists 241: 215–228.

47. ChenS, WangQL, XuS, LiuI, LiL, et al. (2002) Functional analysis of cone-rod homeobox (CRX) mutations associated with retinal dystrophy. Hum Mol Genet 11(8): 873–884.

48. HayashiS, LewisP, PevnyL, McMahonAP (2002) Efficient gene modulation in mouse epiblast using a Sox2Cre transgenic mouse strain. Mech Dev (Suppl 1): S97–S101.

49. NishidaA, FurukawaA, KoikeC, TanoY, AizawaS, et al. (2003) Homeobox gene controls retinal photoreceptor cell fate and pineal gland development. Nature Neuroscience 6(12): 1255–1263.

50. PeacheyNS, BallSL (2003) Electrophysiological analysis of visual function in mutant mice. Documenta ophthalmologica. Advances in ophthalmology 107(1): 13–36.

51. Carter-DawsonLD, LaVailMM (1979) Rods and Cones in the Mouse Retina. Journal of Comparative Neurology 188(2): 245–262.

52. ChiquetC, Dkhissi-BenyahyaO, ChounlamountriN, SzelA, DegripWJ, et al. (2002) Characterization of calbindin-positive cones in primates. Neuroscience 115(4): 1323–33.

53. RichKA, ZhanY, BlanksJC (1997) Migration and synaptogenesis of cone photoreceptors in the developing mouse retina. The Journal of comparative neurology 388(1): 47–63.

54. MieziewskaKE, Van VeenT, MurrayJM, AguirreGD (1991) Rod and cone specific domains in the interphotoreceptor matrix. The Journal of Comparative Neurology 308(3): 371–80.

55. SzélA, RöhlichP, CafféAR, Van VeenT (1996) Distribution of cone photoreceptors in the mammalian retina. Microscopy research and technique 35(6): 445–62.

56. AppleburyML, AntochMP, BaxterLC, ChunLL, FalkJD, et al. (2000) The murine cone photoreceptor: a single cone type expresses both S and M opsins with retinal spatial patterning. Neuron 27: 513–523.

57. FurukawaA, KoikeC, LippincottP, CepkoCL, FurukawaT (2002) The mouse Crx 5′-upstream transgene sequence directs cell-specific and developmentally regulated expression in retinal photoreceptor cells. The Journal of Neuroscience 22(5): 1640–7.

58. RobertsMR, HendricksonA, McGuireCR, RehTA (2005) Retinoid X receptor (gamma) is necessary to establish the S-opsin gradient in cone photoreceptors of the developing mouse retina. Investigative ophthalmology & visual science 46(8): 2897–904.

59. RobertsMR, SrinivasM, ForrestD, Morreale de EscobarG, RehTA (2006) Making the gradient: thyroid hormone regulates cone opsin expression in the developing mouse retina. Proc Natl Acad Sci USA 103(16): 6218–23.

60. Menotti-RaymondM, DeckmanKH, DavidVA, MyrkaloJ, O'BrienSJ, et al. (2010) Mutation discovered in a feline model of human congenital retinal blinding disease. Invest Ophthalmol Vis Sci 51(6): 2852–9.

61. CurtisR, BarnettK, LeonA (1987) An Early-Onset Retinal Dystrophy With Dominant Inheritance in the Abyssinian Cat. IOVS 28: 131–139.

62. OmoriY, KatohK, SatoS, MuranishiY, ChayaT, et al. (2011) Analysis of transcriptional regulatory pathways of photoreceptor genes by expression profiling of the Otx2-deficient retina. PloS one 6(5): e19685.

63. KoikeC, NishidaA, UenoS, SaitoH, SanukiR, et al. (2007) Functional roles of Otx2 transcription factor in postnatal mouse retinal development. Mol Cell Biol 27(23): 8318–8329.

64. LambaDA, GustJ, RehTA (2009) Transplantation of human embryonic stem cell-derived photoreceptors restores some visual function in Crx-deficient mice. Cell stem cell 4(1): 73–9.

65. ChaddertonN, Millington-WardS, PalfiA, O'ReillyM, TuohyG, et al. (2009) Improved retinal function in a mouse model of dominant retinitis pigmentosa following AAV-delivered gene therapy. Molecular therapy: the journal of the American Society of Gene Therapy 17(4): 593–9.

66. O'ReillyM, PalfiA, ChaddertonN, Millington-WardS, AderM, et al. (2007) RNA interference-mediated suppression and replacement of human rhodopsin in vivo. American journal of human genetics 81(1): 127–35.

67. WangX, XuS, RivoltaC, LiLY, PengGH, et al. (2002) Barrier to autointegration factor interacts with the cone-rod homeobox and represses its transactivation function. The Journal of Biological Chemistry 277(45): 43288–300.

68. ChenS, PengGH, WangX, SmithAC, GroteSK, et al. (2004) Interference of Crx-dependent transcription by ataxin-7 involves interaction between the glutamine regions and requires the ataxin-7 carboxy-terminal region for nuclear localization. Hum Molec Genet 13: 53–67.

Štítky
Genetika Reprodukčná medicína

Článok vyšiel v časopise

PLOS Genetics


2014 Číslo 2
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#