#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Early Mechanisms of Pathobiology Are Revealed by Transcriptional Temporal Dynamics in Hippocampal CA1 Neurons of Prion Infected Mice


Prion diseases typically have long pre-clinical incubation periods during which time the infectious prion particle and infectivity steadily propagate in the brain. Abnormal neuritic sprouting and synaptic deficits are apparent during pre-clinical disease, however, gross neuronal loss is not detected until the onset of the clinical phase. The molecular events that accompany early neuronal damage and ultimately conclude with neuronal death remain obscure. In this study, we used laser capture microdissection to isolate hippocampal CA1 neurons and determined their pre-clinical transcriptional response during infection. We found that gene expression within these neurons is dynamic and characterized by distinct phases of activity. We found that a major cluster of genes is altered during pre-clinical disease after which expression either returns to basal levels, or alternatively undergoes a direct reversal during clinical disease. Strikingly, we show that this cluster contains a signature highly reminiscent of synaptic N-methyl-D-aspartic acid (NMDA) receptor signaling and the activation of neuroprotective pathways. Additionally, genes involved in neuronal projection and dendrite development were also altered throughout the disease, culminating in a general decline of gene expression for synaptic proteins. Similarly, deregulated miRNAs such as miR-132-3p, miR-124a-3p, miR-16-5p, miR-26a-5p, miR-29a-3p and miR-140-5p follow concomitant patterns of expression. This is the first in depth genomic study describing the pre-clinical response of hippocampal neurons to early prion replication. Our findings suggest that prion replication results in the persistent stimulation of a programmed response that is mediated, at least in part, by synaptic NMDA receptor activity that initially promotes cell survival and neurite remodelling. However, this response is terminated prior to the onset of clinical symptoms in the infected hippocampus, seemingly pointing to a critical juncture in the disease. Manipulation of these early neuroprotective pathways may redress the balance between degeneration and survival, providing a potential inroad for treatment.


Vyšlo v časopise: Early Mechanisms of Pathobiology Are Revealed by Transcriptional Temporal Dynamics in Hippocampal CA1 Neurons of Prion Infected Mice. PLoS Pathog 8(11): e32767. doi:10.1371/journal.ppat.1003002
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003002

Souhrn

Prion diseases typically have long pre-clinical incubation periods during which time the infectious prion particle and infectivity steadily propagate in the brain. Abnormal neuritic sprouting and synaptic deficits are apparent during pre-clinical disease, however, gross neuronal loss is not detected until the onset of the clinical phase. The molecular events that accompany early neuronal damage and ultimately conclude with neuronal death remain obscure. In this study, we used laser capture microdissection to isolate hippocampal CA1 neurons and determined their pre-clinical transcriptional response during infection. We found that gene expression within these neurons is dynamic and characterized by distinct phases of activity. We found that a major cluster of genes is altered during pre-clinical disease after which expression either returns to basal levels, or alternatively undergoes a direct reversal during clinical disease. Strikingly, we show that this cluster contains a signature highly reminiscent of synaptic N-methyl-D-aspartic acid (NMDA) receptor signaling and the activation of neuroprotective pathways. Additionally, genes involved in neuronal projection and dendrite development were also altered throughout the disease, culminating in a general decline of gene expression for synaptic proteins. Similarly, deregulated miRNAs such as miR-132-3p, miR-124a-3p, miR-16-5p, miR-26a-5p, miR-29a-3p and miR-140-5p follow concomitant patterns of expression. This is the first in depth genomic study describing the pre-clinical response of hippocampal neurons to early prion replication. Our findings suggest that prion replication results in the persistent stimulation of a programmed response that is mediated, at least in part, by synaptic NMDA receptor activity that initially promotes cell survival and neurite remodelling. However, this response is terminated prior to the onset of clinical symptoms in the infected hippocampus, seemingly pointing to a critical juncture in the disease. Manipulation of these early neuroprotective pathways may redress the balance between degeneration and survival, providing a potential inroad for treatment.


Zdroje

1. JeffreyM, HallidayWG, BellJ, JohnstonAR, MacLeodNK, et al. (2000) Synapse loss associated with abnormal PrP precedes neuronal degeneration in the scrapie-infected murine hippocampus. Neuropathol Appl Neurobiol 26: 41–54.

2. CunninghamC, DeaconR, WellsH, BocheD, WatersS, et al. (2003) Synaptic changes characterize early behavioural signs in the ME7 model of murine prion disease. Eur J Neurosci 17: 2147–2155.

3. SiskovaZ, PageA, O'ConnorV, PerryVH (2009) Degenerating synaptic boutons in prion disease: Microglia activation without synaptic stripping. Am J Pathol 175: 1610–1621.

4. MorenoJA, RadfordH, PerettiD, SteinertJR, VerityN, et al. (2012) Sustained translational repression by eIF2alpha-P mediates prion neurodegeneration. Nature 485: 507–511.

5. HwangD, LeeIY, YooH, GehlenborgN, ChoJH, et al. (2009) A systems approach to prion disease. Mol Syst Biol 5: 252.

6. KimHO, SnyderGP, BlazeyTM, RaceRE, ChesebroB, et al. (2008) Prion disease induced alterations in gene expression in spleen and brain prior to clinical symptoms. Adv Appl Bioinform Chem 1: 29–50.

7. SorensenG, MedinaS, ParchaliukD, PhillipsonC, RobertsonC, et al. (2008) Comprehensive transcriptional profiling of prion infection in mouse models reveals networks of responsive genes. BMC Genomics 9: 114.

8. XiangW, HummelM, MittereggerG, PaceC, WindlO, et al. (2007) Transcriptome analysis reveals altered cholesterol metabolism during the neurodegeneration in mouse scrapie model. J Neurochem 102: 834–847.

9. SkinnerPJ, AbbassiH, ChesebroB, RaceRE, ReillyC, et al. (2006) Gene expression alterations in brains of mice infected with three strains of scrapie. BMC Genomics 7: 114.

10. BrownAR, RebusS, McKimmieCS, RobertsonK, WilliamsA, et al. (2005) Gene expression profiling of the pre-clinical scrapie-infected hippocampus. Biochem Biophys Res Commun 334: 86–95.

11. BoothS, BowmanC, BaumgartnerR, DolenkoB, SorensenG, et al. (2004) Molecular classification of scrapie strains in mice using gene expression profiling. Biochem Biophys Res Commun 325: 1339–1345.

12. RiemerC, NeidholdS, BurwinkelM, SchwarzA, SchultzJ, et al. (2004) Gene expression profiling of scrapie-infected brain tissue. Biochem Biophys Res Commun 323: 556–564.

13. XiangW, WindlO, WunschG, DugasM, KohlmannA, et al. (2004) Identification of differentially expressed genes in scrapie-infected mouse brains by using global gene expression technology. J Virol 78: 11051–11060.

14. PietersenCY, LimMP, MaceyL, WooTU, SonntagKC (2011) Neuronal type-specific gene expression profiling and laser-capture microdissection. Methods Mol Biol 755: 327–343.

15. RossnerMJ, HirrlingerJ, WichertSP, BoehmC, NewrzellaD, et al. (2006) Global transcriptome analysis of genetically identified neurons in the adult cortex. J Neurosci 26: 9956–9966.

16. ChungCY, SeoH, SonntagKC, BrooksA, LinL, et al. (2005) Cell type-specific gene expression of midbrain dopaminergic neurons reveals molecules involved in their vulnerability and protection. Hum Mol Genet 14: 1709–1725.

17. KammeF, SalungaR, YuJ, TranDT, ZhuJ, et al. (2003) Single-cell microarray analysis in hippocampus CA1: Demonstration and validation of cellular heterogeneity. J Neurosci 23: 3607–3615.

18. AmbrosV, LeeRC, LavanwayA, WilliamsPT, JewellD (2003) MicroRNAs and other tiny endogenous RNAs in C. elegans. Curr Biol 13: 807–818.

19. BartelDP (2004) MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 116: 281–297.

20. Barbato C, Ruberti F. (2012). MicroRNA Regulation of Neuronal Differentiation and Plasticity. In: Mallick B, Ghosh Z, editors. Regulatory RNAs Basics, Methods and Applications. Springer Berlin Heidelberg. Pp. 175–195.

21. SonntagKC (2010) MicroRNAs and deregulated gene expression networks in neurodegeneration. Brain Res 1338: 48–57 10.1016/j.brainres.2010.03.106.

22. Majer, A., Boese, A.S., Booth, S.A., 2012. The role of microRNAs in neurodegenerative diseases: Implications for early detection and treatment. In: Mallick B, Ghosh Z, editors. Regulatory RNAs Basics, Methods and Applications. Springer Berlin Heidelberg. Pp. 443–473.

23. CuellarTL, DavisTH, NelsonPT, LoebGB, HarfeBD, et al. (2008) Dicer loss in striatal neurons produces behavioral and neuroanatomical phenotypes in the absence of neurodegeneration. Proc Natl Acad Sci U S A 105: 5614–5619.

24. DamianiD, AlexanderJJ, O'RourkeJR, McManusM, JadhavAP, et al. (2008) Dicer inactivation leads to progressive functional and structural degeneration of the mouse retina. J Neurosci 28: 4878–4887.

25. DavisTH, CuellarTL, KochSM, BarkerAJ, HarfeBD, et al. (2008) Conditional loss of dicer disrupts cellular and tissue morphogenesis in the cortex and hippocampus. J Neurosci 28: 4322–4330.

26. HaramatiS, ChapnikE, SztainbergY, EilamR, ZwangR, et al. (2010) miRNA malfunction causes spinal motor neuron disease. Proc Natl Acad Sci U S A 107: 13111–13116.

27. KaimalV, BardesEE, TabarSC, JeggaAG, AronowBJ (2010) ToppCluster: A multiple gene list feature analyzer for comparative enrichment clustering and network-based dissection of biological systems. Nucleic Acids Res 38: W96–102.

28. ClineMS, SmootM, CeramiE, KuchinskyA, LandysN, et al. (2007) Integration of biological networks and gene expression data using cytoscape. Nat Protoc 2: 2366–2382.

29. SmootM, OnoK, IdekerT, MaereS (2011) PiNGO: A cytoscape plugin to find candidate genes in biological networks. Bioinformatics 27: 1030–1031.

30. KimAH, ReimersM, MaherB, WilliamsonV, McMichaelO, et al. (2010) MicroRNA expression profiling in the prefrontal cortex of individuals affected with schizophrenia and bipolar disorders. Schizophr Res 124: 183–191.

31. CogswellJP, WardJ, TaylorIA, WatersM, ShiY, et al. (2008) Identification of miRNA changes in alzheimer's disease brain and CSF yields putative biomarkers and insights into disease pathways. J Alzheimers Dis 14: 27–41.

32. MestdaghP, FeysT, BernardN, GuentherS, ChenC, et al. (2008) High-throughput stem-loop RT-qPCR miRNA expression profiling using minute amounts of input RNA. Nucleic Acids Res 36: e143.

33. TangF, HajkovaP, BartonSC, LaoK, SuraniMA (2006) MicroRNA expression profiling of single whole embryonic stem cells. Nucleic Acids Res 34: e9.

34. LivakKJ, SchmittgenTD (2001) Analysis of relative gene expression data using real-time quantitative PCR and the 2(-delta delta C(T)) method. Methods 25: 402–408.

35. AbramoffMD, MagalhaesPJ, RamSJ (2004) “Image Processing with ImageJ”. Biophotonics International volume 11 (issue 7): 36–42.

36. ChenJ, BardesEE, AronowBJ, JeggaAG (2009) ToppGene suite for gene list enrichment analysis and candidate gene prioritization. Nucleic Acids Res 37: W305–11.

37. SchmuedLC, StowersCC, ScalletAC, XuL (2005) Fluoro-jade C results in ultra high resolution and contrast labeling of degenerating neurons. Brain Res 1035: 24–31.

38. Russelakis-CarneiroM, HetzC, MaundrellK, SotoC (2004) Prion replication alters the distribution of synaptophysin and caveolin 1 in neuronal lipid rafts. Am J Pathol 165: 1839–1848.

39. KyrkanidesS, OlschowkaJA, WilliamsJP, HansenJT, O'BanionMK (1999) TNF alpha and IL-1beta mediate intercellular adhesion molecule-1 induction via microglia-astrocyte interaction in CNS radiation injury. J Neuroimmunol 95: 95–106.

40. HughesMM, FieldRH, PerryVH, MurrayCL, CunninghamC (2010) Microglia in the degenerating brain are capable of phagocytosis of beads and of apoptotic cells, but do not efficiently remove PrPSc, even upon LPS stimulation. Glia 58: 2017–2030.

41. PerryVH, NicollJA, HolmesC (2010) Microglia in neurodegenerative disease. Nat Rev Neurol 6: 193–201.

42. MayrB, MontminyM (2001) Transcriptional regulation by the phosphorylation-dependent factor CREB. Nat Rev Mol Cell Biol 2: 599–609.

43. BelichenkoPV, BrownD, JeffreyM, FraserJR (2000) Dendritic and synaptic alterations of hippocampal pyramidal neurones in scrapie-infected mice. Neuropathol Appl Neurobiol 26: 143–149.

44. JohnstonAR, BlackC, FraserJ, MacLeodN (1997) Scrapie infection alters the membrane and synaptic properties of mouse hippocampal CA1 pyramidal neurones. J Physiol 500 (Pt 1) (Pt 1): 1–15.

45. ChitiZ, KnutsenOM, BetmouniS, GreeneJR (2006) An integrated, temporal study of the behavioural, electrophysiological and neuropathological consequences of murine prion disease. Neurobiol Dis 22: 363–373.

46. ZhangSJ, SteijaertMN, LauD, SchutzG, Delucinge-VivierC, et al. (2007) Decoding NMDA receptor signaling: Identification of genomic programs specifying neuronal survival and death. Neuron 53: 549–562.

47. ZhangSJ, ZouM, LuL, LauD, DitzelDA, et al. (2009) Nuclear calcium signaling controls expression of a large gene pool: Identification of a gene program for acquired neuroprotection induced by synaptic activity. PLoS Genet 5: e1000604.

48. LiST, JuJG (2012) Functional roles of synaptic and extrasynaptic NMDA receptors in physiological and pathological neuronal activities. Curr Drug Targets 13: 207–221.

49. SabaR, GoodmanCD, HuzarewichRL, RobertsonC, BoothSA (2008) A miRNA signature of prion induced neurodegeneration. PLoS One 3: e3652.

50. MorE, CabillyY, GoldshmitY, ZaltsH, ModaiS, et al. (2011) Species-specific microRNA roles elucidated following astrocyte activation. Nucleic Acids Res 39: 3710–3723.

51. ZovoilisA, AgbemenyahHY, Agis-BalboaRC, StillingRM, EdbauerD, et al. (2011) microRNA-34c is a novel target to treat dementias. EMBO J 30: 4299–4308.

52. LukiwWJ, DuaP, PogueAI, EickenC, HillJM (2011) Upregulation of micro RNA-146a (miRNA-146a), a marker for inflammatory neurodegeneration, in sporadic creutzfeldt-jakob disease (sCJD) and gerstmann-straussler-scheinker (GSS) syndrome. J Toxicol Environ Health A 74: 1460–1468.

53. RuscaN, MonticelliS (2011) MiR-146a in immunity and disease. Mol Biol Int 2011: 437301.

54. SabaR, GushueS, HuzarewichRL, ManguiatK, MedinaS, et al. (2012) MicroRNA 146a (miR-146a) is over-expressed during prion disease and modulates the innate immune response and the microglial activation state. PLoS One 7: e30832.

55. SanukiR, OnishiA, KoikeC, MuramatsuR, WatanabeS, et al. (2011) miR-124a is required for hippocampal axogenesis and retinal cone survival through Lhx2 suppression. Nat Neurosci 14: 1125–1134.

56. ConacoC, OttoS, HanJJ, MandelG (2006) Reciprocal actions of REST and a microRNA promote neuronal identity. Proc Natl Acad Sci U S A 103: 2422–2427.

57. KyeMJ, LiuT, LevySF, XuNL, GrovesBB, et al. (2007) Somatodendritic microRNAs identified by laser capture and multiplex RT-PCR. RNA 13: 1224–1234.

58. ParkCS, TangSJ (2009) Regulation of microRNA expression by induction of bidirectional synaptic plasticity. J Mol Neurosci 38: 50–56.

59. AronicaE, FluiterK, IyerA, ZuroloE, VreijlingJ, et al. (2010) Expression pattern of miR-146a, an inflammation-associated microRNA, in experimental and human temporal lobe epilepsy. Eur J Neurosci 31: 1100–1107.

60. LewisBP, BurgeCB, BartelDP (2005) Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 120: 15–20.

61. GrimsonA, FarhKK, JohnstonWK, Garrett-EngeleP, LimLP, et al. (2007) MicroRNA targeting specificity in mammals: Determinants beyond seed pairing. Mol Cell 27: 91–105.

62. FriedmanRC, FarhKK, BurgeCB, BartelDP (2009) Most mammalian mRNAs are conserved targets of microRNAs. Genome Res 19: 92–105.

63. BuelerH, AguzziA, SailerA, GreinerRA, AutenriedP, et al. (1993) Mice devoid of PrP are resistant to scrapie. Cell 73: 1339–1347.

64. BrandnerS, RaeberA, SailerA, BlattlerT, FischerM, et al. (1996) Normal host prion protein (PrPC) is required for scrapie spread within the central nervous system. Proc Natl Acad Sci U S A 93: 13148–13151.

65. BrownDR, SchmidtB, KretzschmarHA (1996) Role of microglia and host prion protein in neurotoxicity of a prion protein fragment. Nature 380: 345–347.

66. BrownD, BelichenkoP, SalesJ, JeffreyM, FraserJR (2001) Early loss of dendritic spines in murine scrapie revealed by confocal analysis. Neuroreport 12: 179–183.

67. FuhrmannM, MittereggerG, KretzschmarH, HermsJ (2007) Dendritic pathology in prion disease starts at the synaptic spine. J Neurosci 27: 6224–6233.

68. HeisekeA, AguibY, SchatzlHM (2010) Autophagy, prion infection and their mutual interactions. Curr Issues Mol Biol 12: 87–97.

69. BergerZ, RavikumarB, MenziesFM, OrozLG, UnderwoodBR, et al. (2006) Rapamycin alleviates toxicity of different aggregate-prone proteins. Hum Mol Genet 15: 433–442.

70. RubinszteinDC, DiFigliaM, HeintzN, NixonRA, QinZH, et al. (2005) Autophagy and its possible roles in nervous system diseases, damage and repair. Autophagy 1: 11–22.

71. IwataA, ChristiansonJC, BucciM, EllerbyLM, NukinaN, et al. (2005) Increased susceptibility of cytoplasmic over nuclear polyglutamine aggregates to autophagic degradation. Proc Natl Acad Sci U S A 102: 13135–13140.

72. DronM, BaillyY, BeringueV, HaeberleAM, GriffondB, et al. (2006) SCRG1, a potential marker of autophagy in transmissible spongiform encephalopathies. Autophagy 2: 58–60.

73. ClareR, KingVG, WirenfeldtM, VintersHV (2010) Synapse loss in dementias. J Neurosci Res 88: 2083–2090.

74. GongY, LippaCF (2010) Review: Disruption of the postsynaptic density in alzheimer's disease and other neurodegenerative dementias. Am J Alzheimers Dis Other Demen 25: 547–555.

75. ClintonJ, ForsythC, RoystonMC, RobertsGW (1993) Synaptic degeneration is the primary neuropathological feature in prion disease: A preliminary study. Neuroreport 4: 65–68.

76. ChiesaR, PiccardoP, BiasiniE, GhettiB, HarrisDA (2008) Aggregated, wild-type prion protein causes neurological dysfunction and synaptic abnormalities. J Neurosci 28: 13258–13267.

77. MallucciGR (2009) Prion neurodegeneration: Starts and stops at the synapse. Prion 3: 195–201.

78. DhawanJ, BenvenisteH, LuoZ, NawrockyM, SmithSD, et al. (2011) A new look at glutamate and ischemia: NMDA agonist improves long-term functional outcome in a rat model of stroke. Future Neurol 6: 823–834.

79. GhasemiM, SchachterSC (2011) The NMDA receptor complex as a therapeutic target in epilepsy: A review. Epilepsy Behav 22: 617–640.

80. GilmourG, DixS, FelliniL, GastambideF, PlathN, et al. (2012) NMDA receptors, cognition and schizophrenia–testing the validity of the NMDA receptor hypofunction hypothesis. Neuropharmacology 62: 1401–1412.

81. CollingeJ, WhittingtonMA, SidleKC, SmithCJ, PalmerMS, et al. (1994) Prion protein is necessary for normal synaptic function. Nature 370: 295–297.

82. MaglioLE, PerezMF, MartinsVR, BrentaniRR, RamirezOA (2004) Hippocampal synaptic plasticity in mice devoid of cellular prion protein. Brain Res Mol Brain Res 131: 58–64.

83. KhosravaniH, ZhangY, TsutsuiS, HameedS, AltierC, et al. (2008) Prion protein attenuates excitotoxicity by inhibiting NMDA receptors. J Cell Biol 181: 551–565.

84. Sanz-ClementeA, NicollRA, RocheKW (2012) Diversity in NMDA receptor composition: Many regulators, many consequences. Neuroscientist XX: 1–14.

85. Wittmann, M., Bengston, C.P., Bading, H., 2004. Extrasynaptic NMDA receptors: mediators of excitotoxic cell death. In: Krieglstein, J, Klumpp, S., editors. Pharmacology of Cerebral Ischemia. Stuttgart, Germany: Medpharm Scientific Publishers. pp. 253–266.

86. ChuC, AlapatD, WenX, TimoK, BursteinD, et al. (2004) Ectopic expression of murine diphosphoinositol polyphosphate phosphohydrolase 1 attenuates signaling through the ERK1/2 pathway. Cell Signal 16: 1045–1059.

87. AmadoroG, CiottiMT, CostanziM, CestariV, CalissanoP, et al. (2006) NMDA receptor mediates tau-induced neurotoxicity by calpain and ERK/MAPK activation. Proc Natl Acad Sci U S A 103: 2892–2897.

88. LiuY, WongTP, AartsM, RooyakkersA, LiuL, et al. (2007) NMDA receptor subunits have differential roles in mediating excitotoxic neuronal death both in vitro and in vivo. J Neurosci 27: 2846–2857.

89. GrocL, HeineM, CousinsSL, StephensonFA, LounisB, et al. (2006) NMDA receptor surface mobility depends on NR2A-2B subunits. Proc Natl Acad Sci U S A 103: 18769–18774.

90. TovarKR, SprouffskeK, WestbrookGL (2000) Fast NMDA receptor-mediated synaptic currents in neurons from mice lacking the epsilon2 (NR2B) subunit. J Neurophysiol 83: 616–620.

91. GuillaudL, SetouM, HirokawaN (2003) KIF17 dynamics and regulation of NR2B trafficking in hippocampal neurons. J Neurosci 23: 131–140.

92. von EngelhardtJ, CosereaI, PawlakV, FuchsEC, KohrG, et al. (2007) Excitotoxicity in vitro by NR2A- and NR2B-containing NMDA receptors. Neuropharmacology 53: 10–17.

93. MartelMA, WyllieDJ, HardinghamGE (2009) In developing hippocampal neurons, NR2B-containing N-methyl-D-aspartate receptors (NMDARs) can mediate signaling to neuronal survival and synaptic potentiation, as well as neuronal death. Neuroscience 158: 334–343.

94. ThomasCG, MillerAJ, WestbrookGL (2006) Synaptic and extrasynaptic NMDA receptor NR2 subunits in cultured hippocampal neurons. J Neurophysiol 95: 1727–1734.

95. PetraliaRS, WangYX, HuaF, YiZ, ZhouA, et al. (2010) Organization of NMDA receptors at extrasynaptic locations. Neuroscience 167: 68–87.

96. Kiss-TothE (2011) Tribbles: ‘puzzling’ regulators of cell signalling. Biochem Soc Trans 39: 684–687.

97. LauA, TymianskiM (2010) Glutamate receptors, neurotoxicity and neurodegeneration. Pflugers Arch 460: 525–542.

98. SalihDA, BrunetA (2008) FoxO transcription factors in the maintenance of cellular homeostasis during aging. Curr Opin Cell Biol 20: 126–136.

99. GalehdarZ, SwanP, FuerthB, CallaghanSM, ParkDS, et al. (2010) Neuronal apoptosis induced by endoplasmic reticulum stress is regulated by ATF4-CHOP-mediated induction of the bcl-2 homology 3-only member PUMA. J Neurosci 30: 16938–16948.

100. PeggionC, BertoliA, SorgatoMC (2011) Possible role for Ca2+ in the pathophysiology of the prion protein? Biofactors 37: 241–249.

101. WongK, QiuY, HyunW, NixonR, VanCleffJ, et al. (1996) Decreased receptor-mediated calcium response in prion-infected cells correlates with decreased membrane fluidity and IP3 release. Neurology 47: 741–750.

102. FlorioT, ThellungS, AmicoC, RobelloM, SalmonaM, et al. (1998) Prion protein fragment 106–126 induces apoptotic cell death and impairment of L-type voltage-sensitive calcium channel activity in the GH3 cell line. J Neurosci Res 54: 341–352.

103. BarrowPA, HolmgrenCD, TapperAJ, JefferysJG (1999) Intrinsic physiological and morphological properties of principal cells of the hippocampus and neocortex in hamsters infected with scrapie. Neurobiol Dis 6: 406–423.

104. FeigLA (2011) Regulation of neuronal function by ras-GRF exchange factors. Genes Cancer 2: 306–319.

105. TianX, GotohT, TsujiK, LoEH, HuangS, et al. (2004) Developmentally regulated role for ras-GRFs in coupling NMDA glutamate receptors to ras, erk and CREB. EMBO J 23: 1567–1575.

106. GasperiniR, Choi-LundbergD, ThompsonMJ, MitchellCB, FoaL (2009) Homer regulates calcium signalling in growth cone turning. Neural Dev 4: 29.

107. XiaoB, TuJC, WorleyPF (2000) Homer: A link between neural activity and glutamate receptor function. Curr Opin Neurobiol 10: 370–374.

108. BrakemanPR, LanahanAA, O'BrienR, RocheK, BarnesCA, et al. (1997) Homer: A protein that selectively binds metabotropic glutamate receptors. Nature 386: 284–288.

109. WilliamsC, Mehrian ShaiR, WuY, HsuYH, SitzerT, et al. (2009) Transcriptome analysis of synaptoneurosomes identifies neuroplasticity genes overexpressed in incipient alzheimer's disease. PLoS One 4: e4936.

110. AngoF, PinJP, TuJC, XiaoB, WorleyPF, et al. (2000) Dendritic and axonal targeting of type 5 metabotropic glutamate receptor is regulated by homer1 proteins and neuronal excitation. J Neurosci 20: 8710–8716.

111. AngoF, RobbeD, TuJC, XiaoB, WorleyPF, et al. (2002) Homer-dependent cell surface expression of metabotropic glutamate receptor type 5 in neurons. Mol Cell Neurosci 20: 323–329.

112. LuJ, HeltonTD, BlanpiedTA, RaczB, NewpherTM, et al. (2007) Postsynaptic positioning of endocytic zones and AMPA receptor cycling by physical coupling of dynamin-3 to homer. Neuron 55: 874–889.

113. JiY, LuY, YangF, ShenW, TangTT, et al. (2010) Acute and gradual increases in BDNF concentration elicit distinct signaling and functions in neurons. Nat Neurosci 13: 302–309.

114. KaramboulasC, SwedaniA, WardC, Al-MadhounAS, WiltonS, et al. (2006) HDAC activity regulates entry of mesoderm cells into the cardiac muscle lineage. J Cell Sci 119: 4305–4314.

115. SugoN, OshiroH, TakemuraM, KobayashiT, KohnoY, et al. (2010) Nucleocytoplasmic translocation of HDAC9 regulates gene expression and dendritic growth in developing cortical neurons. Eur J Neurosci 31: 1521–1532.

116. HasegawaH, KiyokawaE, TanakaS, NagashimaK, GotohN, et al. (1996) DOCK180, a major CRK-binding protein, alters cell morphology upon translocation to the cell membrane. Mol Cell Biol 16: 1770–1776.

117. LiX, GaoX, LiuG, XiongW, WuJ, et al. (2008) Netrin signal transduction and the guanine nucleotide exchange factor DOCK180 in attractive signaling. Nat Neurosci 11: 28–35.

118. XuNJ, HenkemeyerM (2009) Ephrin-B3 reverse signaling through Grb4 and cytoskeletal regulators mediates axon pruning. Nat Neurosci 12: 268–276.

119. KimJY, OhMH, BernardLP, MacaraIG, ZhangH (2011) The RhoG/ELMO1/Dock180 signaling module is required for spine morphogenesis in hippocampal neurons. J Biol Chem 286: 37615–37624.

120. RudolfR, BittinsCM, GerdesHH (2011) The role of myosin V in exocytosis and synaptic plasticity. J Neurochem 116: 177–191.

121. OsterweilE, WellsDG, MoosekerMS (2005) A role for myosin VI in postsynaptic structure and glutamate receptor endocytosis. J Cell Biol 168: 329–338.

122. MorrisSM, CooperJA (2001) Disabled-2 colocalizes with the LDLR in clathrin-coated pits and interacts with AP-2. Traffic 2: 111–123.

123. WuH, NashJE, ZamoranoP, GarnerCC (2002) Interaction of SAP97 with minus-end-directed actin motor myosin VI. implications for AMPA receptor trafficking. J Biol Chem 277: 30928–30934.

124. HuangCH, ChengJC, ChenJC, TsengCP (2007) Evaluation of the role of disabled-2 in nerve growth factor-mediated neurite outgrowth and cellular signalling. Cell Signal 19: 1339–1347.

125. WagnerW, BrenowitzSD, HammerJA3rd (2011) Myosin-va transports the endoplasmic reticulum into the dendritic spines of purkinje neurons. Nat Cell Biol 13: 40–48.

126. MakeyevEV, ZhangJ, CarrascoMA, ManiatisT (2007) The MicroRNA miR-124 promotes neuronal differentiation by triggering brain-specific alternative pre-mRNA splicing. Mol Cell 27: 435–448.

127. KrichevskyAM, SonntagKC, IsacsonO, KosikKS (2006) Specific microRNAs modulate embryonic stem cell-derived neurogenesis. Stem Cells 24: 857–864.

128. MaioranoNA, MallamaciA (2009) Promotion of embryonic cortico-cerebral neuronogenesis by miR-124. Neural Dev 4: 40.

129. VisvanathanJ, LeeS, LeeB, LeeJW, LeeSK (2007) The microRNA miR-124 antagonizes the anti-neural REST/SCP1 pathway during embryonic CNS development. Genes Dev 21: 744–749.

130. ChengLC, PastranaE, TavazoieM, DoetschF (2009) miR-124 regulates adult neurogenesis in the subventricular zone stem cell niche. Nat Neurosci 12: 399–408.

131. ArvanitisDN, JungasT, BeharA, DavyA (2010) Ephrin-B1 reverse signaling controls a posttranscriptional feedback mechanism via miR-124. Mol Cell Biol 30: 2508–2517.

132. PasqualeEB (2008) Eph-ephrin bidirectional signaling in physiology and disease. Cell 133: 38–52.

133. WaymanGA, DavareM, AndoH, FortinD, VarlamovaO, et al. (2008) An activity-regulated microRNA controls dendritic plasticity by down-regulating p250GAP. Proc Natl Acad Sci U S A 105: 9093–9098.

134. EdbauerD, NeilsonJR, FosterKA, WangCF, SeeburgDP, et al. (2010) Regulation of synaptic structure and function by FMRP-associated microRNAs miR-125b and miR-132. Neuron 65: 373–384.

135. GoleyED, WelchMD (2006) The ARP2/3 complex: An actin nucleator comes of age. Nat Rev Mol Cell Biol 7: 713–726.

136. LippiG, SteinertJR, MarczyloEL, D'OroS, FioreR, et al. (2011) Targeting of the Arpc3 actin nucleation factor by miR-29a/b regulates dendritic spine morphology. J Cell Biol 194: 889–904.

137. LiuW, LiuC, ZhuJ, ShuP, YinB, et al. (2012) MicroRNA-16 targets amyloid precursor protein to potentially modulate alzheimer's-associated pathogenesis in SAMP8 mice. Neurobiol Aging 33: 522–534.

138. ShiL, KoML, KoGY (2009) Rhythmic expression of microRNA-26a regulates the L-type voltage-gated calcium channel alpha1C subunit in chicken cone photoreceptors. J Biol Chem 284: 25791–25803.

139. ViaderA, ChangLW, FahrnerT, NagarajanR, MilbrandtJ (2011) MicroRNAs modulate schwann cell response to nerve injury by reinforcing transcriptional silencing of dedifferentiation-related genes. J Neurosci 31: 17358–17369.

140. MontagJ, HittR, OpitzL, Schulz-SchaefferWJ, HunsmannG, et al. (2009) Upregulation of miRNA hsa-miR-342-3p in experimental and idiopathic prion disease. Mol Neurodegener 4: 36.

141. KieblerMA, BassellGJ (2006) Neuronal RNA granules: Movers and makers. Neuron 51: 685–690.

142. VoN, KleinME, VarlamovaO, KellerDM, YamamotoT, et al. (2005) A cAMP-response element binding protein-induced microRNA regulates neuronal morphogenesis. Proc Natl Acad Sci U S A 102: 16426–16431.

143. SiegelG, ObernostererG, FioreR, OehmenM, BickerS, et al. (2009) A functional screen implicates microRNA-138-dependent regulation of the depalmitoylation enzyme APT1 in dendritic spine morphogenesis. Nat Cell Biol 11: 705–716.

144. SchrattGM, TuebingF, NighEA, KaneCG, SabatiniME, et al. (2006) A brain-specific microRNA regulates dendritic spine development. Nature 439: 283–289.

145. WaymanGA, KaechS, GrantWF, DavareM, ImpeyS, et al. (2004) Regulation of axonal extension and growth cone motility by calmodulin-dependent protein kinase I. J Neurosci 24: 3786–3794.

146. FioreR, KhudayberdievS, ChristensenM, SiegelG, FlavellSW, et al. (2009) Mef2-mediated transcription of the miR379-410 cluster regulates activity-dependent dendritogenesis by fine-tuning Pumilio2 protein levels. EMBO J 28: 697–710.

147. ImajoM, NishidaE (2010) Human tribbles homolog 1 functions as a negative regulator of retinoic acid receptor. Genes Cells 15: 1089–1097.

148. Kiss-TothE, BagstaffSM, SungHY, JozsaV, DempseyC, et al. (2004) Human tribbles, a protein family controlling mitogen-activated protein kinase cascades. J Biol Chem 279: 42703–42708.

149. MoriM, BurgessDL, GefridesLA, ForemanPJ, OpfermanJT, et al. (2004) Expression of apoptosis inhibitor protein Mcl1 linked to neuroprotection in CNS neurons. Cell Death Differ 11: 1223–1233.

150. GrabruckerAM, SchmeisserMJ, SchoenM, BoeckersTM (2011) Postsynaptic ProSAP/Shank scaffolds in the cross-hair of synaptopathies. Trends Cell Biol 21: 594–603.

151. WendholtD, SpilkerC, SchmittA, DolnikA, SmallaKH, et al. (2006) ProSAP-interacting protein 1 (ProSAPiP1), a novel protein of the postsynaptic density that links the spine-associated rap-gap (SPAR) to the scaffolding protein ProSAP2/Shank3. J Biol Chem 281: 13805–13816.

152. BordenLA, SmithKE, GustafsonEL, BranchekTA, WeinshankRL (1995) Cloning and expression of a betaine/GABA transporter from human brain. J Neurochem 64: 977–984.

153. CalellaAM, NerlovC, LopezRG, SciarrettaC, von Bohlen und HalbachO, et al. (2007) Neurotrophin/Trk receptor signaling mediates C/EBPalpha, -beta and NeuroD recruitment to immediate-early gene promoters in neuronal cells and requires C/EBPs to induce immediate-early gene transcription. Neural Dev 2: 4.

154. ItoH, MorishitaR, ShinodaT, IwamotoI, SudoK, et al. (2010) Dysbindin-1, WAVE2 and abi-1 form a complex that regulates dendritic spine formation. Mol Psychiatry 15: 976–986.

155. HardingHP, ZhangY, ScheunerD, ChenJJ, KaufmanRJ, et al. (2009) Ppp1r15 gene knockout reveals an essential role for translation initiation factor 2 alpha (eIF2alpha) dephosphorylation in mammalian development. Proc Natl Acad Sci U S A 106: 1832–1837.

156. YanoH, NinanI, ZhangH, MilnerTA, ArancioO, et al. (2006) BDNF-mediated neurotransmission relies upon a myosin VI motor complex. Nat Neurosci 9: 1009–1018.

157. MoriguchiS, OomuraY, ShiodaN, HanF, HoriN, et al. (2011) Ca2+/calmodulin-dependent protein kinase II and protein kinase C activities mediate extracellular glucose-regulated hippocampal synaptic efficacy. Mol Cell Neurosci 46: 101–107.

158. LeeY, AhnC, HanJ, ChoiH, KimJ, et al. (2003) The nuclear RNase III drosha initiates microRNA processing. Nature 425: 415–419.

159. SunX, WuY, ChenB, ZhangZ, ZhouW, et al. (2011) Regulator of calcineurin 1 (RCAN1) facilitates neuronal apoptosis through caspase-3 activation. J Biol Chem 286: 9049–9062.

160. AkhtarMW, KimMS, AdachiM, MorrisMJ, QiX, et al. (2012) In vivo analysis of MEF2 transcription factors in synapse regulation and neuronal survival. PLoS One 7: e34863.

161. WangX, SheH, MaoZ (2009) Phosphorylation of neuronal survival factor MEF2D by glycogen synthase kinase 3beta in neuronal apoptosis. J Biol Chem 284: 32619–32626.

162. TsurutaF, GreenEM, RoussetM, DolmetschRE (2009) PIKfyve regulates CaV1.2 degradation and prevents excitotoxic cell death. J Cell Biol 187: 279–294.

163. ChongZZ, LiF, MaieseK (2005) Oxidative stress in the brain: Novel cellular targets that govern survival during neurodegenerative disease. Prog Neurobiol 75: 207–246.

164. RiccioA, AhnS, DavenportCM, BlendyJA, GintyDD (1999) Mediation by a CREB family transcription factor of NGF-dependent survival of sympathetic neurons. Science 286: 2358–2361.

165. FinkbeinerS (2000) CREB couples neurotrophin signals to survival messages. Neuron 25: 11–14.

166. LonzeBE, GintyDD (2002) Function and regulation of CREB family transcription factors in the nervous system. Neuron 35: 605–623.

167. WuHY, HudryE, HashimotoT, KuchibhotlaK, RozkalneA, et al. (2010) Amyloid beta induces the morphological neurodegenerative triad of spine loss, dendritic simplification, and neuritic dystrophies through calcineurin activation. J Neurosci 30: 2636–2649.

168. Spires-JonesTL, KayK, MatsoukaR, RozkalneA, BetenskyRA, et al. (2011) Calcineurin inhibition with systemic FK506 treatment increases dendritic branching and dendritic spine density in healthy adult mouse brain. Neurosci Lett 487: 260–263.

169. LeoneDP, SrinivasanK, BrakebuschC, McConnellSK (2010) The rho GTPase Rac1 is required for proliferation and survival of progenitors in the developing forebrain. Dev Neurobiol 70: 659–678.

170. LinsemanDA, LoucksFA (2008) Diverse roles of rho family GTPases in neuronal development, survival, and death. Front Biosci 13: 657–676.

171. AndersonKA, KaneCD (1998) Ca2+/calmodulin-dependent protein kinase IV and calcium signaling. Biometals 11: 331–343.

172. HansenMR, BokJ, DevaiahAK, ZhaXM, GreenSH (2003) Ca2+/calmodulin-dependent protein kinases II and IV both promote survival but differ in their effects on axon growth in spiral ganglion neurons. J Neurosci Res 72: 169–184.

173. AntoniFA (2000) Molecular diversity of cyclic AMP signalling. Front Neuroendocrinol 21: 103–132.

174. AntoniFA, SosunovAA, HaunsoA, PatersonJM, SimpsonJ (2003) Short-term plasticity of cyclic adenosine 3′,5′-monophosphate signaling in anterior pituitary corticotrope cells: The role of adenylyl cyclase isotypes. Mol Endocrinol 17: 692–703.

175. MoosmangS, HaiderN, KlugbauerN, AdelsbergerH, LangwieserN, et al. (2005) Role of hippocampal Cav1.2 Ca2+ channels in NMDA receptor-independent synaptic plasticity and spatial memory. J Neurosci 25: 9883–9892.

176. LuJ, NozumiM, TakeuchiK, AbeH, IgarashiM (2011) Expression and function of neuronal growth-associated proteins (nGAPs) in PC12 cells. Neurosci Res 70: 85–90.

177. FjorbackAW, MullerHK, WiborgO (2009) Membrane glycoprotein M6B interacts with the human serotonin transporter. J Mol Neurosci 37: 191–200.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2012 Číslo 11
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#