DOI QR코드

DOI QR Code

A Brief Introduction to the Transduction of Neural Activity into Fos Signal

  • Chung, Leeyup (Dept. of Neurobiology, Duke University School of Medicine)
  • 투고 : 2015.04.01
  • 심사 : 2015.05.04
  • 발행 : 2015.06.30

초록

The immediate early gene c-fos has long been known as a molecular marker of neural activity. The neuron's activity is transformed into intracellular calcium influx through NMDA receptors and L-type voltage sensitive calcium channels. For the transcription of c-fos, neural activity should be strong enough to activate mitogen-activated protein kinase (MAPK) signaling pathway which shows low calcium sensitivity. Upon translation, the auto-inhibition by Fos protein regulates basal Fos expression. The pattern of external stimuli and the valence of the stimulus to the animal change Fos signal, thus the signal reflects learning and memory aspects. Understanding the features of multiple components regulating Fos signaling is necessary for the optimal generation and interpretation of Fos signal.

키워드

참고문헌

  1. Barth AL, Gerkin RC, Dean KL (2004) Alteration of neuronal firing properties after in vivo experience in a FosGFP transgenic mouse. Journal of Neuroscience 24:6466-6475. https://doi.org/10.1523/JNEUROSCI.4737-03.2004
  2. Berretta S, Robertson HA, Graybiel AM (1992) Dopamine and glutamate agonists stimulate neuron-specific expression of Fos-like protein in the striatum. Journal of Neurophysiology 68:767-777. https://doi.org/10.1152/jn.1992.68.3.767
  3. Berretta S, Parthasarathy HB, Graybiel AM (1997) Local release of GABAergic inhibition in the motor cortex induces immediate-early gene expression in indirect pathway neurons of the striatum. Journal of Neuroscience 17:4752-4763. https://doi.org/10.1523/JNEUROSCI.17-12-04752.1997
  4. Bourne AR, Mohan G, Stone MF, Pham MQ, Schultz CR, Meyerhoff JL, Lumley LA (2013) Olfactory cues increase avoidance behavior and induce Fos expression in the amygdala, hippocampus and prefrontal cortex of socially defeated mice. Behavioural Brain Research 256:188-196. https://doi.org/10.1016/j.bbr.2013.08.020
  5. Bramham CR, Worley PF, Moore MJ, Guzowski JF (2008) The immediate early gene arc/arg3.1: regulation, mechanisms, and function. Journal of Neuroscience 28:11760-11767. https://doi.org/10.1523/JNEUROSCI.3864-08.2008
  6. Cao VY, Ye Y, Mastwal S, Ren M, Coon M, Liu Q, Costa RM, Wang KH (2015) Motor learning consolidates Arc-expressing neuronal ensembles in secondary motor cortex. Neuron 86:1385-1392. https://doi.org/10.1016/j.neuron.2015.05.022
  7. Carrion AM, Link WA, Ledo F, Mellstrom B, Naranjo JR (1999) DREAM is a $Ca^{2+}$-regulated transcriptional repressor. Nature 398:80-84. https://doi.org/10.1038/18044
  8. Chaudhuri A, Zangenehpour S, Rahbar-Dehgan F, Ye F (2000) Molecular maps of neural activity and quiescence. Acta Neurobiologiae Experimentalis 60:403-410.
  9. Cohen S, Greenberg ME (2008) Communication between the synapse and the nucleus in neuronal development, plasticity, and disease. Annu Rev Cell Dev Biol 24:183-209. https://doi.org/10.1146/annurev.cellbio.24.110707.175235
  10. Cossell L, Iacaruso MF, Muir DR, Houlton R, Sader EN, Ko H, Hofer SB, Mrsic-Flogel TD (2015) Functional organization of excitatory synaptic strength in primary visual cortex. Nature 518:399-403. https://doi.org/10.1038/nature14182
  11. Covington HE, 3rd, Lobo MK, Maze I, Vialou V, Hyman JM, Zaman S, LaPlant Q, Mouzon E, Ghose S, Tamminga CA, Neve RL, Deisseroth K, Nestler EJ (2010) Antidepressant effect of optogenetic stimulation of the medial prefrontal cortex. Journal of Neuroscience 30:16082-16090. https://doi.org/10.1523/JNEUROSCI.1731-10.2010
  12. Cruz FC, Javier Rubio F, Hope BT (2014) Using c-fos to study neuronal ensembles in corticostriatal circuitry of addiction. Brain research, http://dx.doi.org/10.1016/j.brainres.2014.11.005
  13. Deisseroth K, Tsien RW (2002) Dynamic multiphosphorylation passwords for activity-dependent gene expression. Neuron 34:179-182. https://doi.org/10.1016/S0896-6273(02)00664-5
  14. Deisseroth K, Mermelstein PG, Xia H, Tsien RW (2003) Signaling from synapse to nucleus: the logic behind the mechanisms. Current Opinion in Neurobiology 13:354-365. https://doi.org/10.1016/S0959-4388(03)00076-X
  15. Derkach VA, Oh MC, Guire ES, Soderling TR (2007) Regulatory mechanisms of AMPA receptors in synaptic plasticity. Nature Reviews Neuroscience 8:101-113. https://doi.org/10.1038/nrn2055
  16. Durchdewald M, Angel P, Hess J (2009) The transcription factor Fos: a Janus-type regulator in health and disease. Histology and Histopathology 24:1451-1461.
  17. Ghosh A, Ginty DD, Bading H, Greenberg ME (1994) Calcium regulation of gene expression in neuronal cells. Journal of Neurobiology 25:294-303. https://doi.org/10.1002/neu.480250309
  18. Herdegen T, Leah JD (1998) Inducible and constitutive transcription factors in the mammalian nervous system: control of gene expression by Jun, Fos and Krox, and CREB/ATF proteins. Brain Research Reviews 28:370-490. https://doi.org/10.1016/S0165-0173(98)00018-6
  19. Hu JH, Park JM, Park S, Xiao B, Dehoff MH, Kim S, Hayashi T, Schwarz MK, Huganir RL, Seeburg PH, Linden DJ, Worley PF (2010) Homeostatic scaling requires group I mGluR activation mediated by Homer1a. Neuron 68:1128-1142. https://doi.org/10.1016/j.neuron.2010.11.008
  20. Kawashima T, Okuno H, Bito H (2014) A new era for functional labeling of neurons: activity-dependent promoters have come of age. Frontiers in Neural Circuits 8:37.
  21. Koya E, Cruz FC, Ator R, Golden SA, Hoffman AF, Lupica CR, Hope BT (2012) Silent synapses in selectively activated nucleus accumbens neurons following cocaine sensitization. Nature Neuroscience 15:1556-1562. https://doi.org/10.1038/nn.3232
  22. Krishnan V, Nestler EJ (2011) Animal models of depression: molecular perspectives. Current Topics in Behavioral Neurosciences 7:121-147. https://doi.org/10.1007/7854_2010_108
  23. Lau CG, Zukin RS (2007) NMDA receptor trafficking in synaptic plasticity and neuropsychiatric disorders. Nature Reviews Neuroscience 8:413-426.
  24. Liste I, Rozas G, Guerra MJ, Labandeira-Garcia JL (1995) Cortical stimulation induces Fos expression in striatal neurons via NMDA glutamate and dopamine receptors. Brain Research 700:1-12. https://doi.org/10.1016/0006-8993(95)00958-S
  25. Lucibello FC, Lowag C, Neuberg M, Muller R (1989) Trans-repression of the mouse c-fos promoter: a novel mechanism of Fos-mediated trans-regulation. Cell 59:999-1007. https://doi.org/10.1016/0092-8674(89)90756-3
  26. Lyons MR, West AE (2011) Mechanisms of specificity in neuronal activity-regulated gene transcription. Progress in Neurobiology 94:259-295. https://doi.org/10.1016/j.pneurobio.2011.05.003
  27. Martinez M, Calvo-Torrent A, Herbert J (2002) Mapping brain response to social stress in rodents with c-fos expression: a review. Stress 5:3-13. https://doi.org/10.1080/102538902900012369
  28. Morgan JI, Curran T (1991) Stimulus-transcription coupling in the nervous system: involvement of the inducible proto-oncogenes fos and jun. Annual Review of Neuroscience 14:421-451. https://doi.org/10.1146/annurev.ne.14.030191.002225
  29. Murphy LO, Smith S, Chen RH, Fingar DC, Blenis J (2002) Molecular interpretation of ERK signal duration by immediate early gene products. Nature Cell Biology 4:556-564. https://doi.org/10.1038/ncb822
  30. Murphy TH, Worley PF, Baraban JM (1991) L-type voltage-sensitive calcium channels mediate synaptic activation of immediate early genes. Neuron 7:625-635. https://doi.org/10.1016/0896-6273(91)90375-A
  31. Nestler EJ (2015) FosB: a transcriptional regulator of stress and antidepressant responses. European Journal of Pharmacology 753:66-72. https://doi.org/10.1016/j.ejphar.2014.10.034
  32. Parthasarathy HB, Graybiel AM (1997) Cortically driven immediate-early gene expression reflects modular influence of sensorimotor cortex on identified striatal neurons in the squirrel monkey. Journal of Neuroscience 17:2477-2491. https://doi.org/10.1523/JNEUROSCI.17-07-02477.1997
  33. Rajadhyaksha A, Barczak A, Macias W, Leveque JC, Lewis SE, Konradi C (1999) L-Type $Ca(^{2+})$ channels are essential for glutamate-mediated CREB phosphorylation and c-fos gene expression in striatal neurons. Journal of Neuroscience 19:6348-6359. https://doi.org/10.1523/JNEUROSCI.19-15-06348.1999
  34. Vanhoutte P, Barnier JV, Guibert B, Pages C, Besson MJ, Hipskind RA, Caboche J (1999) Glutamate induces phosphorylation of Elk-1 and CREB, along with c-fos activation, via an extracellular signal-regulated kinase-dependent pathway in brain slices. Molecular and Cellular Biology 19:136-146. https://doi.org/10.1128/MCB.19.1.136
  35. Vazdarjanova A, McNaughton BL, Barnes CA, Worley PF, Guzowski JF (2002) Experience-dependent coincident expression of the effector immediate-early genes arc and Homer 1a in hippocampal and neocortical neuronal networks. Journal of Neuroscience 22:10067-10071. https://doi.org/10.1523/JNEUROSCI.22-23-10067.2002
  36. Veyrac A, Besnard A, Caboche J, Davis S, Laroche S (2014) The transcription factor Zif268/Egr1, brain plasticity, and memory. Progress in Molecular Biology and Translational Science 122:89-129. https://doi.org/10.1016/B978-0-12-420170-5.00004-0
  37. Wang JQ, Fibuch EE, Mao L (2007) Regulation of mitogen-activated protein kinases by glutamate receptors. Journal of Neurochemistry 100:1-11. https://doi.org/10.1111/j.1471-4159.2006.04208.x
  38. Wang M, Perova Z, Arenkiel BR, Li B (2014) Synaptic modifications in the medial prefrontal cortex in susceptibility and resilience to stress. Journal of Neuroscience 34:7485-7492. https://doi.org/10.1523/JNEUROSCI.5294-13.2014
  39. Wu GY, Deisseroth K, Tsien RW (2001) Spaced stimuli stabilize MAPK pathway activation and its effects on dendritic morphology. Nature Neuroscience 4:151-158. https://doi.org/10.1038/83976
  40. Zangenehpour S, Chaudhuri A (2002) Differential induction and decay curves of c-fos and zif268 revealed through dual activity maps. Brain Research Molecular Brain Research 109:221-225. https://doi.org/10.1016/S0169-328X(02)00556-9
  41. Zhang J, Zhang D, McQuade JS, Behbehani M, Tsien JZ, Xu M (2002) c-fos regulates neuronal excitability and survival. Nature Genetics 30:416-420. https://doi.org/10.1038/ng859