• 沒有找到結果。

FoxP2 acts as a regulator in neuronal subtype specification

During development, neurons are instructed to migrate and send projections to specific regions in the brain. A fundamental question is how cells differentiate into specific neuronal subtypes. The subdivision of the GE can give rise to different subtypes of neurons by the expression of transcription factors. The LGE gives rise to not only cortical GABAergic interneurons but also striatal projection neurons and the MGE gives rise to many cortical GABAergic interneurons (Sussel et al., 1999). Several recent studies have shown that the GE regional identities are maintained through cross-regulatory interactions by transcription factors (Tucker et al., 2008) For example, homeobox transcription factors Dlx1 and Dlx2 are expressed by progenitors within the ventral telencephalon, regulate the fate and migration of ventral precursors, and promote GABAergic interneuron differentiation (Maqdelena et al., 2007). Another

transcription factor Mash1 has a role in the specification of neuronal

35

identity in different regions of the nervous system and can induce expression of markers of GABAergic neurons in the cerebral cortex (Fode et al. 2000). Moreover, Shh signaling is centrally involved in the patterning of the nervous system. It has been shown that expression of Shh is required for the specification of neuronal identity (Ericson et al., 1996) and GABAergic populations such as interneurons derived from the MGE also depend on Shh-signaling (Nery et al., 2001). Many

Shh-dependent homeodomain transcription factors are involved in ventral patterning in GE (Corbin et al., 2003). Previous study has demonstrated that Gli family of transcriptional regulators – Gli1, Gli2 are collectively necessary and sufficient for all aspects of intracellular Shh-responses involved in ventral forebrain patterning (Matise et al., 1998). Another study shows that the Shh-mediated induction of Nkx2-1 expression initiates MGE development (Xu et al., 2005). However, the mechanisms controlling the segregation of LGE and MGE neuronal populations

remain poorly understood. Previous reports have shown that transcription factors are thought to play a major role in regulating the specification of neuronal subtype. For example, one previous study showed that FoxP2 was expressed by LGE-derived neurons and absent from MGE-derived interneurons (Takahashi et al., 2009). Therefore, FoxP2 may be important to specify GE-derived neuronal subtypes. Using loss-of-function

approaches, my experiments demonstrate that loss of FoxP2 expression results in a reduction of LGE-derived medium spiny neurons. In contrast, MGE-derived interneurons are increased. These results suggest that in the absence of FoxP2, LGE progenitors are specified to a more MGE fate.

36

This transformation leads to a dramatic change in the number of GABAergic cortical interneurons, striatal interneurons and projection neurons derived from the MGE and LGE, respectively (Sussel et al., 1999). Furthermore, other factors may be involved in transcriptional regulation of neuronal subtype specification. I believe that evolution has selected distinct regulatory networks of gene expression for the cell types derived from each of the two GE. For example, Nkx2-1, unlike FoxP2, is expressed in the MGE and is critical for the development of the

mammalian forebrain (Gulacsi et al., 2006). Also, previous studies have revealed that Nkx2-1 regulates the specification of MGE-derived cells during early patterning of the telencephalon (Corbin et al., 2003).

Interestingly, FoxP2 inhibits Nkx2-1 transcription activity in alveolar cells (Zhou et al., 2008). It is likely that FoxP2 interacts with other transcription factors to regulate neuronal subtype specification. Taken together, it implies that these two transcription factors FoxP2and Nkx2-1 may regulate the specification of neuronal fates in the ventral forebrain.

Overall, in this study, I investigate the effects and mechanisms of FoxP2 in embryonic neurogenesis. Therefore, the next step is to investigate whether FoxP2 and Shh/Nkx2-1 together control the

specification of GABAergic neurons and potential interactions between FoxP2 and Shh/Nkx2-1 in the embryonic brain.

37

5. References

Bertrand N, Castro DS, Guillemot F (2002) Proneural genes and the specification of neural cell types. Nat Rev Neurosci 3:517-530.

Besnard V, Xu Y, Whitsett JA (2007) Sterol response element binding protein and thyroid transcription factor-1 (Nkx2.1) regulate Abca3 gene expression. Am J Physiol Lung Cell Mol Physiol

293:L1395-1405.

Britz O, Mattar P, Nguyen L, Langevin LM, Zimmer C, Alam S,

Guillemot F, Schuurmans C (2006) A role for proneural genes in the maturation of cortical progenitor cells. Cereb Cortex 16 Suppl 1:i138-151.

Butt SJ, Fuccillo M, Nery S, Noctor S, Kriegstein A, Corbin JG, Fishell G (2005) The temporal and spatial origins of cortical interneurons predict their physiological subtype. Neuron 48:591-604.

Butt SJ, Sousa VH, Fuccillo MV, Hjerling-Leffler J, Miyoshi G, Kimura S, Fishell G (2008) The requirement of Nkx2-1 in the temporal specification of cortical interneuron subtypes. Neuron 59:722-732.

Corbin JG, Rutlin M, Gaiano N, Fishell G (2003) Combinatorial function of the homeodomain proteins Nkx2.1 and Gsh2 in ventral

telencephalic patterning. Development 130:4895-4906.

Deisseroth K, Singla S, Toda H, Monje M, Palmer TD, Malenka RC (2004) Excitation-neurogenesis coupling in adult neural

stem/progenitor cells. Neuron 42:535-552.

Dessaud E, Yang LL, Hill K, Cox B, Ulloa F, Ribeiro A, Mynett A, Novitch BG, Briscoe J (2007) Interpretation of the sonic hedgehog

38

morphogen gradient by a temporal adaptation mechanism. Nature 450:717-720.

Dhruv NT, Tailby C, Sokol SH, Lennie P (2011) Multiple adaptable mechanisms early in the primate visual pathway. J Neurosci 31:15016-15025.

Du T, Xu Q, Ocbina PJ, Anderson SA (2008) NKX2.1 specifies cortical interneuron fate by activating Lhx6. Development 135:1559-1567.

Elias LA, Potter GB, Kriegstein AR (2008) A time and a place for nkx2-1 in interneuron specification and migration. Neuron 59:679-682.

Enard W, Gehre S, Hammerschmidt K, Holter SM, Blass T, Somel M, Bruckner MK, Schreiweis C, Winter C, Sohr R, Becker L, Wiebe V, Nickel B, Giger T, Muller U, Groszer M, Adler T, Aguilar A, Bolle I, Calzada-Wack J, Dalke C, Ehrhardt N, Favor J, Fuchs H,

Gailus-Durner V, Hans W, Holzlwimmer G, Javaheri A,

Kalaydjiev S, Kallnik M, Kling E, Kunder S, Mossbrugger I, Naton B, Racz I, Rathkolb B, Rozman J, Schrewe A, Busch DH, Graw J, Ivandic B, Klingenspor M, Klopstock T, Ollert M,

Quintanilla-Martinez L, Schulz H, Wolf E, Wurst W, Zimmer A, Fisher SE, Morgenstern R, Arendt T, de Angelis MH, Fischer J, Schwarz J, Paabo S (2009) A humanized version of Foxp2 affects cortico-basal ganglia circuits in mice. Cell 137:961-971.

Enard W, Przeworski M, Fisher SE, Lai CS, Wiebe V, Kitano T, Monaco AP, Paabo S (2002) Molecular evolution of FOXP2, a gene

involved in speech and language. Nature 418:869-872.

Englund C, Fink A, Lau C, Pham D, Daza RA, Bulfone A, Kowalczyk T,

39

Hevner RF (2005) Pax6, Tbr2, and Tbr1 are expressed sequentially by radial glia, intermediate progenitor cells, and postmitotic

neurons in developing neocortex. J Neurosci 25:247-251.

Eriksson C, Wictorin K (2003) Neuronal and glial differentiation within expanded glial cultures derived from the lateral and medial

ganglionic eminences. Exp Neurol 184:1058-1063.

Ericson J, Morton S, Kawakami A, Roelink H, Jessell TM (1996) Two critical periods of Sonic Hedgehog signaling required for the specification of motor neuron identity. Cell 1996-87:661–673.

Erlandsson A, Brannvall K, Gustafsdottir S, Westermark B,

Forsberg-Nilsson K (2006) Autocrine/paracrine platelet-derived growth factor regulates proliferation of neural progenitor cells.

Cancer Res 66:8042-8048.

Erlandsson A, Enarsson M, Forsberg-Nilsson K (2001) Immature neurons from CNS stem cells proliferate in response to platelet-derived growth factor. J Neurosci 21:3483-3491.

Farah MH, Olson JM, Sucic HB, Hume RI, Tapscott SJ, Turner DL (2000) Generation of neurons by transient expression of neural bHLH proteins in mammalian cells. Development 127:693-702.

Farries MA, Perkel DJ (2000) Electrophysiological properties of avian basal ganglia neurons recorded in vitro. J Neurophysiol

84:2502-2513.

Ferland RJ, Cherry TJ, Preware PO, Morrisey EE, Walsh CA (2003) Characterization of Foxp2 and Foxp1 mRNA and protein in the developing and mature brain. J Comp Neurol 460:266-279.

40

Fode C, Ma Q, Casarosa S, Ang SL, Anderson DJ, Guillemot F (2000) A role for neural determination genes in specifying the dorsoventral identity of telencephalic neurons. Genes Dev 14:67-80.

Fuccillo M, Rallu M, McMahon AP, Fishell G (2004) Temporal requirement for hedgehog signaling in ventral telencephalic patterning. Development 131:5031-5040.

Fuccillo M, Joyner AL, Fishell G (2006) Morphogen to mitogen: the multiple roles of hedgehog signalling in vertebrate neural development. Nat Rev Neurosci 7:772-783.

Fujita E, Tanabe Y, Shiota A, Ueda M, Suwa K, Momoi MY, Momoi T (2008) Ultrasonic vocalization impairment of Foxp2 (R552H) knockin mice related to speech-language disorder and abnormality of Purkinje cells. Proc Natl Acad Sci U S A 105:3117-3122.

Gulacsi A, Anderson SA (2006) Shh maintains Nkx2.1 in the MGE by a Gli3-independent mechanism. Cereb Cortex 16 Suppl 1:i89-95.

Hallam S, Singer E, Waring D, Jin Y (2000) The C. elegans NeuroD homolog cnd-1 functions in multiple aspects of motor neuron fate specification. Development 127:4239-4252.

Haesler S, Rochefort C, Georgi B, Licznerski P, Osten P, Scharff C (2007) Incomplete and inaccurate vocal imitation after knockdown of

FoxP2 in songbird basal ganglia nucleus Area X. PLoS Biol 5:e321.

Hevner RF, Shi L, Justice N, Hsueh Y, Sheng M, Smiga S, Bulfone A, Goffinet AM, Campagnoni AT, Rubenstein JL (2001) Tbr1 regulates differentiation of the preplate and layer 6. Neuron 29:353-366.

41

Hirschi KK, Rohovsky SA, D'Amore PA (1998) PDGF, TGF-beta, and heterotypic cell-cell interactions mediate endothelial cell-induced recruitment of 10T1/2 cells and their differentiation to a smooth muscle fate. J Cell Biol 141:805-814.

Hu JG, Wang YX, Wang HJ, Bao MS, Wang ZH, Ge X, Wang FC, Zhou JS, Lu HZ (2012) PDGF-AA mediates B104CM-induced

oligodendrocyte precursor cell differentiation of embryonic neural stem cells through Erk, PI3K, and p38 signaling. J Mol Neurosci 46:644-653.

Huang HP, Liu M, El-Hodiri HM, Chu K, Jamrich M, Tsai MJ (2000) Regulation of the pancreatic islet-specific gene BETA2 (neuroD) by neurogenin 3. Mol Cell Biol 20:3292-3307.

Jackson EL, Garcia-Verdugo JM, Gil-Perotin S, Roy M,

Quinones-Hinojosa A, VandenBerg S, Alvarez-Buylla A (2006) PDGFR alpha-positive B cells are neural stem cells in the adult SVZ that form glioma-like growths in response to increased PDGF signaling. Neuron 51:187-199.

Johansson CB, Momma S, Clarke DL, Risling M, Lendahl U, Frisen J (1999) Identification of a neural stem cell in the adult mammalian central nervous system. Cell 96:25-34.

Johe KK, Hazel TG, Muller T, Dugich-Djordjevic MM, McKay RD (1996) Single factors direct the differentiation of stem cells from the fetal and adult central nervous system. Genes Dev

10:3129-3140.

Kaminen N, Hannula-Jouppi K, Kestila M, Lahermo P, Muller K,

42

Kaaranen M, Myllyluoma B, Voutilainen A, Lyytinen H,

Nopola-Hemmi J, Kere J (2003) A genome scan for developmental dyslexia confirms linkage to chromosome 2p11 and suggests a new locus on 7q32. J Med Genet 40:340-345.

Konopka G, Bomar JM, Winden K, Coppola G, Jonsson ZO, Gao F, Peng S, Preuss TM, Wohlschlegel JA, Geschwind DH (2009)

Human-specific transcriptional regulation of CNS development genes by FOXP2. Nature 462:213-217.

Lai CS, Fisher SE, Hurst JA, Vargha-Khadem F, Monaco AP (2001) A forkhead-domain gene is mutated in a severe speech and language disorder. Nature 413:519-523.

Lai CS, Gerrelli D, Monaco AP, Fisher SE, Copp AJ (2003) FOXP2 expression during brain development coincides with adult sites of pathology in a severe speech and language disorder. Brain

126:2455-2462.

Laub F, Aldabe R, Friedrich V, Jr., Ohnishi S, Yoshida T, Ramirez F (2001) Developmental expression of mouse Kruppel-like

transcription factor KLF7 suggests a potential role in neurogenesis.

Dev Biol 233:305-318.

Lee JS, Wagoner-Johnson A, Murphy WL (2009) Modular peptide growth factors for substrate-mediated stem cell differentiation.

Angew Chem Int Ed Engl 48:6266-6269.

Liboi E, Pelosi E, Di Francesco P, Gallinari P, Petrini M, Sposi NM, Testa U, Rossi GB, Peschle C (1987) The EL2 rat fibroblasts line:

differential effects of growth factors (EGF, PDGF, FGF, TPA and

43

TGF beta) on cell proliferation and c-fos expression. Ann N Y Acad Sci 511:318-328.

Liu M, Pleasure SJ, Collins AE, Noebels JL, Naya FJ, Tsai MJ, Lowenstein DH (2000) Loss of BETA2/NeuroD leads to

malformation of the dentate gyrus and epilepsy. Proc Natl Acad Sci U S A 97:865-870.

Ma Q, Fode C, Guillemot F, Anderson DJ (1999) Neurogenin1 and neurogenin2 control two distinct waves of neurogenesis in developing dorsal root ganglia. Genes Dev 13:1717-1728.

Maden M (2002) Retinoid signalling in the development of the central nervous system. Nat Rev Neurosci 3:843-853.

Maqdelena AP, Gregory BP, David HR, John LR (2007) Dlx1 and Dlx2 control neuronal versus oligodendroglial cell fate acquisition in the developing forebrain. Neuron 55(3):417-433.

Matise MP, Epstein DJ, Park HL, Platt KA, Joyner AL (1998) Gli2 is required for induction of floor plate and adjacent cells, but not most ventral neurons in the mouse central nervous system. Development 125:2759–2770.

McBurney MW, Jones-Villeneuve EM, Edwards MK, Anderson PJ (1982) Control of muscle and neuronal differentiation in a cultured

embryonal carcinoma cell line. Nature 299:165-167.

Miller JE, Spiteri E, Condro MC, Dosumu-Johnson RT, Geschwind DH, White SA (2008) Birdsong decreases protein levels of FoxP2, a molecule required for human speech. J Neurophysiol

100:2015-2025.

44

Muzio L, Di Benedetto B, Stoykova A, Boncinelli E, Gruss P, Mallamaci A (2002) Emx2 and Pax6 control regionalization of the

pre-neuronogenic cortical primordium. Cereb Cortex 12:129-139.

Nadarajah B, Brunstrom JE, Grutzendler J, Wong RO, Pearlman AL (2001) Two modes of radial migration in early development of the cerebral cortex. Nat Neurosci 4:143-150.

Nery S, Wichterle H, Fishell G (2001) Sonic hedgehog contributes to oligodendrocyte specification in the mammalian forebrain.

Development 128:527–540.

Porter JA, Young KE, Beachy PA (1996) Cholesterol modification of hedgehog signaling proteins in animal development. Science 274:255-259.

Przybyla LM, Voldman J (2012) Attenuation of extrinsic signaling reveals the importance of matrix remodeling on maintenance of embryonic stem cell self-renewal. Proc Natl Acad Sci U S A.

Qian X, Shen Q, Goderie SK, He W, Capela A, Davis AA, Temple S (2000) Timing of CNS cell generation: a programmed sequence of neuron and glial cell production from isolated murine cortical stem cells. Neuron 28:69-80.

Rallu M, Machold R, Gaiano N, Corbin JG, McMahon AP, Fishell G (2002) Dorsoventral patterning is established in the telencephalon of mutants lacking both Gli3 and Hedgehog signaling.

Development 129:4963-4974.

Reimers-Kipping S, Hevers W, Paabo S, Enard W (2011) Humanized Foxp2 specifically affects cortico-basal ganglia circuits.

Neuroscience 175:75-84.

Riccio O, Murthy S, Szabo G, Vutskits L, Kiss JZ, Vitalis T, Lebrand C, Dayer AG (2012) New pool of cortical interneuron precursors in

45

the early postnatal dorsal white matter. Cereb Cortex 22:86-98.

Rochefort C, He X, Scotto-Lomassese S, Scharff C (2007)

Recruitment of FoxP2-expressing neurons to area X varies during song development. DevNeurobiol 67: 809–817.

Romo P, Madigan MC, Provis JM, Cullen KM (2011) Differential effects of TGF-beta and FGF-2 on in vitro proliferation and migration of primate retinal endothelial and Muller cells. Acta Ophthalmol 89:e263-268.

Ross SE, Greenberg ME, Stiles CD (2003) Basic helix-loop-helix factors in cortical development. Neuron 39:13-25.

Ruhnke M, Ungefroren H, Zehle G, Bader M, Kremer B, Fandrich F (2003) Long-term culture and differentiation of rat embryonic stem cell-like cells into neuronal, glial, endothelial, and hepatic lineages.

Stem Cells 21:428-436.

Sanders EJ, Harvey S (2008) Peptide hormones as developmental growth and differentiation factors. Dev Dyn 237:1537-1552.

Schulz SB, Haesler S, Scharff C, Rochefort C (2010) Knockdown of FoxP2 alters spine density in Area X of the zebra finch. Genes Brain Behav 9:732-740.

Schuurmans C, Armant O, Nieto M, Stenman JM, Britz O, Klenin N, Brown C, Langevin LM, Seibt J, Tang H, Cunningham JM, Dyck R, Walsh C, Campbell K, Polleux F, Guillemot F (2004)

Sequential phases of cortical specification involve

Neurogenin-dependent and -independent pathways. EMBO J 23:2892-2902.

46

Schwab MH, Bartholomae A, Heimrich B, Feldmeyer D,

Druffel-Augustin S, Goebbels S, Naya FJ, Zhao S, Frotscher M, Tsai MJ, Nave KA (2000) Neuronal basic helix-loop-helix proteins (NEX and BETA2/Neuro D) regulate terminal granule cell

differentiation in the hippocampus. J Neurosci 20:3714-3724.

Segal RA, Takahashi H, McKay RD (1992) Changes in neurotrophin responsiveness during the development of cerebellar granule neurons. Neuron 9:1041-1052.

Shimamura K, Martinez S, Puelles L, Rubenstein JL (1997) Patterns of gene expression in the neural plate and neural tube subdivide the embryonic forebrain into transverse and longitudinal domains. Dev Neurosci 19:88-96.

Spiteri E, Konopka G, Coppola G, Bomar J, Oldham M, Ou J, Vernes SC, Fisher SE, Ren B, Geschwind DH (2007) Identification of the

transcriptional targets of FOXP2, a gene linked to speech and language, in developing human brain. Am J Hum Genet 81:1144-1157.

Stoykova A, Treichel D, Hallonet M, Gruss P (2000) Pax6 modulates the dorsoventral patterning of the mammalian telencephalon. J

Neurosci 20:8042-8050.

Sun J, Wang DA, Jain RK, Carie A, Paquette S, Ennis E, Blaskovich MA, Baldini L, Coppola D, Hamilton AD, Sebti SM (2005) Inhibiting angiogenesis and tumorigenesis by a synthetic molecule that blocks binding of both VEGF and PDGF to their receptors. Oncogene 24:4701-4709.

47

Sussel L, Marin O, Kimura S, Rubenstein JL (1999) Loss of Nkx2.1 homeobox gene function results in a ventral to dorsal molecular respecification within the basal telencephalon: evidence for a transformation of the pallidum into the striatum. Development 126:3359-3370.

Takahashi H, Takahashi K, Liu FC (2009) FOXP genes, neural

development, speech and language disorders. Adv Exp Med Biol 665:117-129.

Taupin P, Gage FH (2002) Adult neurogenesis and neural stem cells of the central nervous system in mammals. J Neurosci Res

69:745-749.

Teramitsu I, Kudo LC, London SE, Geschwind DH, White SA (2004) Parallel FoxP1 and FoxP2 expression in songbird and human brain predicts functional interaction. J Neurosci 24:3152-3163.

Teramitsu I, White SA (2006) FoxP2 regulation during undirected singing in adult songbirds. J Neurosci 26:7390-7394.

Toresson H, Mata de Urquiza A, Fagerstrom C, Perlmann T, Campbell K (1999) Retinoids are produced by glia in the lateral ganglionic eminence and regulate striatal neuron differentiation. Development 126:1317-1326.

Tucker ES, Segall S, Gopalakrishna D, Wu Y, Vernon M, Polleux F, Lamantia AS (2008) Molecular specification and patterning of progenitor cells in the lateral and medial ganglionic eminences. J Neurosci 28:9504-9518.

Vitalis T, Rossier J (2011) New insights into cortical interneurons

48

development and classification: contribution of developmental studies. Dev Neurobiol 71:34-44.

Wang TW, Zhang H, Parent JM (2005) Retinoic acid regulates postnatal neurogenesis in the murine subventricular zone-olfactory bulb pathway. Development 132:2721-2732.

White SA, Fisher SE, Geschwind DH, Scharff C, Holy TE (2006) Singing mice, songbirds, and more: models for FOXP2 function and dysfunction in human speech and language. J Neurosci 26:10376-10379.

Wiens KM, Lee HL, Shimada H, Metcalf AE, Chao MY, Lien CL (2010) Platelet-derived growth factor receptor beta is critical for zebrafish intersegmental vessel formation. PLoS One 5:e11324.

Williams BP, Park JK, Alberta JA, Muhlebach SG, Hwang GY, Roberts TM, Stiles CD (1997) A PDGF-regulated immediate early gene response initiates neuronal differentiation in ventricular zone progenitor cells. Neuron 18:553-562.

Wilson SW, Houart C (2004) Early steps in the development of the forebrain. Dev Cell 6:167-181.

Xie J, Aszterbaum M, Zhang X, Bonifas JM, Zachary C, Epstein E, McCormick F (2001) A role of PDGFRalpha in basal cell

carcinoma proliferation. Proc Natl Acad Sci U S A 98:9255-9259.

Xu Q, Cobos I, De La Cruz E, Rubenstein JL, Anderson SA (2004) Origins of cortical interneuron subtypes. J Neurosci 24:2612-2622.

Xu Q, Guo L, Moore H, Waclaw RR, Campbell K, Anderson SA (2005) Sonic hedgehog signaling confers ventral telencephalic progenitors

49

with distinct cortical interneuron fates. Neuron 65:328–340.

Zhou B, Zhong Q, Minoo P, Li C, Ann DK, Frenkel B, Morrisey EE, Crandall ED, Borok Z (2008) Foxp2 inhibits Nkx2.1-mediated transcription of SP-C via interactions with the Nkx2.1

homeodomain. Am J Respir Cell Mol Biol 38:750-758.

50

51

Figure 1: Overexpression and knockdown of FoxP2 in primary cell culture by electroporation. To set up gain-of-function experiments, I overexpressed FoxP2 and GFP in E13.5 cortical progenitor cells and

cultured them for 2 days. Transfected cells were identified by GFP (green) staining and FoxP2 expressing cells were identified by FoxP2 (red)

staining. Arrows indicate GFP- and FoxP2-double positive cells;

arrowheads indicate GFP-positive but FoxP2-negative cells. The expression of FoxP2 in FoxP2 overexpression group was significantly increased (A, left two columns). For the loss-of-function experiments, I knocked down FoxP2 with shRNA against FoxP2 in GE progenitor cells and cultured them for 5 days. The expression of FoxP2 in the knockdown group was significantly decreased than in the control group (A, right two columns). For quantification anaylysis of US2 and FoxP2 groups,

numbers of FoxP2 and GFP double-positive cells were normalized to the numbers of total GFP-positive cells. n=3, ≧360 GFP-positive cells were counted in each group (B). For quantification anaylysis of UI4 and

shFoxP2 groups, n=3, ≧320 GFP-positive cells were counted in each group (C). Data are presented as means ± SEM. ** compared to the control group, p < 0.01 (t-test); scale bar: 50μm.

52

53

Figure 2: Test of knockdown efficiency of shFoxP2 constructs. To test the knockdown efficiency of three shFoxP2 constructs (shFoxP2-5, shFoxP2-6, shFoxP2-7), P19 cells were transfected with control vector, shFoxP2-5, shFoxP2-6, shFoxP2-7 along with GFP- and

FoxP2-expressing constructs and fixed 3 days after transfection (A).

Transfected cells were identified by GFP (green) staining and FoxP2 expressing cells were identified by FoxP2 (red) staining. Arrows indicate GFP- and FoxP2-double positive cells; arrowheads indicate GFP-positive but FoxP2-negative cells. More GFP-positive cells were FoxP2-negative in shFoxP2-6 group than in the control one (A). For quantification

anaylysis of (A), numbers of FoxP2 and GFP double-positive cells were normalized to the numbers of total GFP-positive cells. n=3, ≧197

GFP-positive cells were counted in each group (B). Western blot analysis

54

of shFoxP2 knockdown efficiency in P19 cells (C). Cells were lysed 3 days after transfection. Expression levels of β-tubulin (55 kDa) were used as loading controls. Quantification of FoxP2 protein levels was

determined as relative intensity of FoxP2 over β-tubulin. FoxP2

expression was reduced significantly in P19 cells transfected with two shRNA expression constructs (shFoxP2-6 or shFoxP2-7) (D). Data are presented as means ± SEM. * compared to the control group. **: p < 0.01;

*: p < 0.05 (t-test); scale bar: 50 μm.

55

56

Figure 3: FoxP2 is sufficient and necessary for neuronal differentiation in the embryonic telencephalon. To investigate whether FoxP2 regulats neuronal differentiation, I overexpressed FoxP2 and GFP in the cortical

Figure 3: FoxP2 is sufficient and necessary for neuronal differentiation in the embryonic telencephalon. To investigate whether FoxP2 regulats neuronal differentiation, I overexpressed FoxP2 and GFP in the cortical

相關文件