文档库 最新最全的文档下载
当前位置:文档库 › 用化合物C能快速高效的将hESCs和hiPSCs诱导为神经祖细胞

用化合物C能快速高效的将hESCs和hiPSCs诱导为神经祖细胞

用化合物C能快速高效的将hESCs和hiPSCs诱导为神经祖细胞
用化合物C能快速高效的将hESCs和hiPSCs诱导为神经祖细胞

Author contributions: J.Z.: Conception and design, manuscript writing, collection and assembly of data, data analysis and interpretation; P.S.: Collection and assembly of data, data analysis and interpretation; D.L.: Collection and assembly of data, data analysis and interpretation; S.T.: Collection and assembly of data, data analysis and interpretation; E.D.: Conception and design, data analysis and interpretation, financial support; F.W.: Conception and design, financial support, manuscript writing, final approval of manuscript

*Corresponding author: Department of Cell and Developmental Biology and Institute for Genomic Biology, University of Illinois at Urbana-Champaign, 601 S. Goodwin Ave, Urbana, IL -61801, USA. Tel.: 217-333-5972; Fax: 217-244-1648; E -mail:feiwang@https://www.wendangku.net/doc/9518740365.html,; Received May 07, 2010; S TEM C ELLS ?

E MBRYONIC S TEM C ELLS /I NDUCED P LURIPOTENT S TEM C ELLS

High-Efficiency Induction of Neural Conversion in hESCs and hiPSCs with a Single Chemical Inhibitor of TGF-E Superfamily Receptors

Jiaxi Zhou 1,2, Pei Su 1,2, Dong Li 1,2, Stephanie Tsang 1,2, Enkui Duan 3, Fei Wang 1,2,*

1

Department of Cell and Developmental Biology and 2Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA. 3State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences (CAS), Beijing 100101, China

Key words. human embryonic stem cells x neural conversion x compound C x TGF-E superfamily receptors x induce

pluripotent stem cells

A BSTRACT

Chemical compounds have emerged as powerful tools for modulating embryonic stem cell (ESC) functions and deriving induced pluripotent stem cells (iPSCs), but documentation of compound-induced efficient directed differentiation in human ESC (hESCs) and human iPSC (hiPSCs) is limited. By screening a collection of chemical compounds, we identified compound C (also denoted as dorsomorphin), a protein kinase inhibitor, as a potent regulator of hESC and hiPSC fate decisions. Compound C suppresses mesoderm, endoderm and trophoectoderm differentiation and induces rapid and high-efficiency neural conversion in both hESCs and hiPSCs (88.7% and 70.4%, respectively). Interestingly, compound C is ineffective in inducing neural conversion in mouse ESCs (mESCs). Large-scale kinase assay revealed that compound C targets at least seven TGF-E superfamily

receptors, including both type I and type II receptors, and thereby blocks both the Activin and BMP signaling pathways in hESCs. Dual inhibition of Activin and BMP signaling accounts for the effects of compound C on hESC differentiation and neural conversion. We also identified muscle segment homeobox gene 2 (MSX2) as a downstream target gene of compound C and a key signaling intermediate of the BMP pathway in hESCs. Our findings provide a single-step cost-effective method for efficient derivation of neural progenitor cells in adherent culture from human pluripotent stem cells. T herefore, it will be uniquely suitable for the production of neural progenitor cells in large scale and should facilitate the use of stem cells in drug screening and regenerative medicine and study of early human neural development. I NTRODUCTION

Human embryonic stem cells (hE SCs) and human induced pluripotent stem cells (hiPSCs) reprogrammed from somatic cells can differentiate into multiple cell types, allowing them to be utilized for biomedical research, drug discovery and cell-based therapies [1, 2]. A deeper understanding of the signaling mechanisms and pathways that maintain

pluripotency and induce direct differentiation of hESCs and iPSCs will shed light on early human development and facilitate the applications of these powerful cellular systems.

Several developmentally important signals have been extensively studied in hESCs. For example, basic FGF (bFGF) supports long-term undifferentiated growth of hESCs [3]. The TGF-? superfamily, comprising TGF-?, Activin,

Nodal and bone morphogenesis proteins (BMPs), has diverse roles in hESCs [4, 5]. TGF-

?/Activin/Nodal was shown to co-operate with FGF signaling to maintain pluripotency of h SCs by controlling NANOG expression. Activation of BMP signaling in hE SCs induces mesoderm and trophoectoderm activities depending on the duration of activation [6-8], while activation of Activin/Nodal pathway can trigger endoderm differentiation [9]. Conversely, inhibition of Activin/Nodal and BMP signaling, alone or in combination, promotes neuroectoderm specification [10-12].

Chemical compounds can be used to control cellular processes and have been applied successfully to the modulation of fate decisions

of stem cells including hE SCs and hiPSCs. Chemical compounds can be synthesized at a large scale and great purity, offer temporal and tunable control of molecular and cellular functions, and has a low cost. E arlier studies showed that the ROCK inhibitor Y-27632 promotes survival of dissociated hE SCs, while the small-molecule valproic acid (VPA) improves the efficiency of hiPSC derivation [13, 14]. Recently, cell-permeable small molecules that direct differentiation of hE SCs into the endoderm or early pancreatic lineage were indentified [15, 16]. Despite these encouraging advances, there is limited documentation about using chemical compounds to efficiently induce differentiation to other lineages such as neuroectoderm in hESCs and hiPSCs.

Here, we report the identification and use of a single chemical compound, compound C/ dorsomorphin, for suppressing endoderm, mesoderm and trophoectoderm differentiation of human pluripotent stem cells and for rapid and highly efficient neural conversion from hE SCs and hiPSCs in defined adherent cultures. We also show that compound C targets at least seven type I and type II TGF-E superfamily receptors and blocks both the Activin and the BMP signaling pathway, which account for compound

C's ability to induce high-efficiency neural conversion.

M ATERIALS AND M ETHODS

Cell Lines and Cell Culture

H1 and H9 hE SCs (WiCell Research Institute)

were routinely maintained under feeder conditions as described [1]. The culture medium

consists of DME M/F12 with 20% knockout

serum replacement (KSR), 1 mM glutamine, 1%

non-essential amino acid, 0.1 mM ?-mercaptoethanol and 4 ng/ml bFGF. KSR is a

defined supplement.For feeder-free cultures,

cells were cultured on plates coated with

Matrigel (BD Biosciences) in the presence of conditioned medium (CM) from mouse embryonic fibroblasts (ME Fs), which replaces

the ME F feeders. Fresh medium was added

every day. For neural conversion of hE SCs,

mTeSR medium (StemCell Technologies) and

DMEM/F12 medium with 20% knockout serum replacement (KSR), 1 mM glutamine, 1% non-

essential amino acid and 0.1 mM ?-mercaptoethanol were used. mTeSR is a defined

feeder-independent medium. The hiPSC line

RND5 (passage 8-10) was from ArunA Biomedical, Inc (Athens, GA) and was maintained under the same conditions as H1 and

H9 hE SCs. Derivation and validation of the

hiPSCs are described: https://www.wendangku.net/doc/9518740365.html,/GeneTargeting/

viPS%20Lentiviral%20Vector%20Kit/. NTera-2 (American Type Culture Collection, ATCC)

cells were cultured according to ATCC recommendations. The medium consists of 10%

fetal bovine serum (FBS, ATCC). For low-

density single-cell culture, cells were dissociated

into single cells with 0.025% trypsin-0.02%

E DTA and plated at the density of 10,000

cells/cm2. W4 mouse E SC (mE SC) line was

from Taconic and was cultured as described

[17].

Western Blotting

For the analysis of proteins except phospho-

Smad2/3, cells (5×106) were lysed directly with

200P l laemmli sample buffer (Biorad). For

detection of phospho-Smad2/3, nuclear fraction

of hESCs was isolated with a NEPER? Nuclear

and Cytoplasmic

E

xtraction Kit (Pierce).

Samples (25 P g) were analyzed by western blot. Dilutions for various antibodies were described in Table S4. The blots were developed using SuperSignal West Pico Chemiluminescent Substrate (Pierce), and signals were quantified with Image J.

EB Formation Assay

H1 or H9 hESCs (5×106

/well) cultured on MEFs

were dissociated as clusters with dispase (1 mg/mL), plated onto Ultra-Low Attachment 6-well plates (Corning) and grown in embryoid body (E B) medium (DME M/F12, 20% FBS, 1 mM glutamine, 1 mM 1% non-essential amino acid and 0.1 mM ?-mercaptoethanol). To change

medium, E Bs and medium were gently transferred into a 15-ml tube and left at RT for 5

min to allow the EBs to sink to the bottom. The medium was gently removed and replaced by fresh medium. E Bs were collected at different time points for further analysis.

Induction of Neural Differentiation

For neural differentiation, H1 or H9 hE SCs were cultured on Matrigel with mTeSR medium to eliminate variations introduced by the ME F feeders. hE SCs were gently dissociated into single-cell suspension with 1 mg/mL accutase for 5–7 min at 37oC. The dissociated cells were plated on Matrigel at high density (106/cm 2) and cultured to confluency in mTeSR medium before

induction of differentiation. For compound C-induced neural conversion, mTeSR medium was

replaced with DMEM/F12 containing 20% KSR,

1 mM glutamine, 1% non-essential amino acid and 0.1 mM ?-mercaptoethanol. This condition was used for all treatment groups. Medium was replaced every day. For early neural patterning, SHH (100 ng/mL) was incubated with

compound C-induced neural progenitor cells for 5 d. For late neural terminal differentiation/maturation, SHH-treated cells were gently dissociated into small clusters with dispase and plated on laminin-coated plates. Cells were cultured in neural differentiation medium, which consists of neural basal medium, 1x N2 supplement, purmorphamine (1 μM), ascorbic acid (200 μM), BDNF (20 ng/mL), GDNF (20 ng/mL), SHH (100 ng/mL) and FGF8 (100 ng/mL), for 2 weeks before collected for analysis.

Luciferase Assay

The ID-120-luc reporter construct for BMP signaling activity, kindly provided by Dr. Renhe Xu (University of Connecticut), was described [18]. To measure the reporter activity, H1 hESCs were plated on Matrigel-coated 24-well plates

before transfection. The ID-120-luc plasmid was co-transfected into cells with the Rellina plasmid

(Promega), and 24 h after transfection, medium was replaced with hESC growth medium without

bFGF. BMP-4 (10 ng/mL) in combination with different concentrations of compound C was

administered to cells. The cells were lysed 24 h after the addition of BMP-4 and compound C by using lysis buffer provided with a dual-luciferase assay kit (Promega). The luciferase activity was

measured with LUMIstar plate reader (BMG Labtech). The luciferase-based Activin reporter plasmid (pARE-GL3, [19]) was kindly provided by Dr. Yisrael Sidis (Partners Healthcare). To measure the reporter activity, H1 hESCs was co-transfected with 0.25 μg pARE -GL3, 0.25 μg FAST-1 (forkhead activin signal transducer-1), and Rellina plasmid. Activin A (100 ng/mL) and various concentrations of SB421542 or compound C were added 24 h after transfection.

The luciferase activity was measure as described above.Additional experimental procedures and associated references are available in the Supporting Information. R ESULTS Compound C Modulates Differentiation Activities in hESCs

We recently screened a collection of chemical compounds, identified and reported mTOR as a key positive regulator of pluripotency [17]. In the current study, we screened for compounds that prevent hE SC differentiation. We cultured two hE SC lines with distinct genotypes [WA01 (H1), XY; WA09 (H9), XX] [1]in the absence

of bFGF and conditioned medium (CM) produced by mouse embryonic fibroblasts (ME Fs), both required to support hE SC self-renewal and pluripotency [1, 20]. Human E SCs grown without bFGF and CM continued to

proliferate at a slower rate, but began to differentiate from the center of the colonies and gradually lost the expression of pluripotency markers OCT-4 and SOX2 as described [17] (Fig. 1A). We applied the collection of chemical compounds and inhibitors (Table S1) to hE SCs cultured in the absence of bFGF and CM. We found that compound C (1 P M), a kinase inhibitor, supported the growth of hE SCs as compact colonies, reminiscent of control cells cultured with bFGF and CM (Fig. S1A). In addition to preventing the differentiation morphology, compound C greatly attenuated the reduction of OCT-4, SOX2 and NANOG proteins (Fig. 1A, Fig. S1B). The supportive effects of compound C on OCT-4, NANOG and SOX2 expression were further confirmed at the transcription level: the mRNA levels of POU5F1(the gene encoding OCT-4), SOX2 and NANOG in compound C-treated hESCs were significantly higher than in cells without the treatment (Fig. 1B). Interestingly, the rescuing effect of compound C on NANOG expression was weaker than POU5F1 and SOX2. Furthermore, compound C markedly prevented the differentiation morphology and down-regulation of pluripotency markers in a pluripotent human embryonal carcinoma cell line, NTera-2 cells (Supporting Information) (Fig. S1C and S1D), suggesting a conserved effect in human pluripotent stem cells.

Compound C profoundly influenced the differentiation activities in h E

SCs. It significantly reduced the expression of markers for mesoderm (MESP1,T and MIXL1),

endoderm (GATA4,GATA6) and trophoectoderm

(CDX2 and CGB7) in H1 cells cultivated in the absence of bFGF and CM (Fig. 1C), but caused the expression of neural ectoderm markers SOX1

and NEUROD1to moderately increase (Fig. 1C).

The effect of compound C on h E

SC differentiation was further verified with the embryoid body (EB) formation assay. During EB formation, expression of differentiation markers was consistently up-regulated in untreated control cells (Fig. 1D and Fig. S1E) [17]. In this assay, compound C treatment markedly prevented the up-regulation of markers for endoderm, mesoderm and trophoectoderm, but enhanced the expression of neuroectoderm markers after E B induction (Fig. 1D and Fig. S1E). Compound C treatment had no effects on the size or the number of EBs (Fig. S1F). High-Efficiency Induction of Neural Conversion by Compound C

Because compound C potently suppressed endoderm, mesoderm and trophoectoderm but enhanced neural ectoderm activities in hE SCs, we asked whether it could be applied to the induction of neural conversion. To test this, we developed differentiation conditions for hE SCs using adherent cell culture, which is more consistent and easier to manipulate than the EB condition. Cells dissociated by accutase (1 mg/mL) were plated on Matrigel and continuously cultured to confluency in mTeSR medium, which could eliminate variations introduced by the ME F feeders. Differentiation was initialized by replacing mTeSR with DM E

M/F12 medium containing knock-out serum replacement (KSR) (but lacking bFGF and CM) and compound C (1 μM). Another small-molecule inhibitor, SB431542, which inhibits type I TGF-E receptors and reportedly promotes neural conversion in hESCs [12], was used with or without compound C. We used PAX6, a neural progenitor marker, to monitor early neural conversion. Human E SCs, when

plated as single cells in the absence of bFGF and CM, began to exhibit differentiation morphology by d 3, and the differentiation was more apparent

by d 5~d 6 with the majority of cells spread out and losing the expression of OCT-4 (Fig. 2A and Fig. S2A). Interestingly, cells treated with compound C grew as a compact uniform

monolayer, in contrast to untreated cells, which often showed areas with disassociated cells and cell clumps (Fig. S2A). Immunofluorescence studies revealed that PAX6 protein was nearly

uniformly expressed in compound C-treated cells (Fig. 2A, middle panel), whereas much fewer untreated cells exhibited anti-PAX6 fluorescence (Fig. 2A, upper panel). Other early neural progenitor markers such as SOX1 and SOX2 were also highly expressed in compound C-treated cells (Fig. 2A, middle panel). Western blot analysis confirmed increased expression of PAX6 and SOX2 proteins (Fig. 2B). Furthermore, western blot analysis also showed that expression of Nestin protein, a neural stem cell marker, was elevated in compound C-treated cells (Fig. 2B). In contrast, addition of SB431542 caused only a slight increase of PAX6 protein and failed to induce further increase of PAX6, Nestin and SOX2 proteins in compound C-treated cells (Fig. 2A and 2B). The expression of pluripotency marker OCT-4 was almost undetectable 7 d after differentiation induction (data not shown). In keeping with the increased level of protein, a gradual increase of PAX6mRNA was also observed during the differentiation process, which lasted for at least 10 d (Fig. S2B).

To determine the fraction of PAX6+

cells, we analyzed the differentiated cells by flow cytometry 7 d to 10 d after compound C treatment. Remarkably, the treatment yielded a high percentage of PAX6+ cells (88.7±2.5%, Fig. 2C and Fig. S2C). There was no significant difference between d 7 to d 10 cells (data not shown). Interestingly, the dual treatment of compound C and SB431542 failed to increase the percentage of PAX6+ cells (80.3%±10.3%) (Fig. 2C and Fig. S2C). Furthermore, while Noggin treatment alone moderately increased the percentage of PAX6+

cells (36.9±1.6%), combined treatment of SB431542 and Noggin significantly increased the percentage (84.7±3.9%) (Fig. 2C and Fig. S2C), consistent with a previous report [10]. The percentage of PAX6+ cells acquired with compound C was slightly higher than with combined treatment of Noggin and SB431542 (Fig. 2C). Thus, the use of compound C, mTeSR medium and KSR-containing DME M/F12 medium enabled us to develop a single-step cost-effective procedure for high-efficiency neural conversion in hE SCs in defined adherent cultures.

Compound C also induced high-efficiency neural conversion in hiPSCs. We used hiPSCs derived from human fetal lung fibroblasts (IMR-90) for neural induction by following the same procedure developed for h E

SCs. Immunofluorescence and western blot analyses revealed a drastic increase of PAX6 and SOX2 proteins in cells treated with compound C (1 P M, 7 d) (Fig. 2D and 2E ). Similar to hE SCs, compound C treatment of hiPSCs induced a high population of PAX6+ cells (70.4%±5.5%) (Fig. 2C). These results demonstrate that compound C alone induces high-efficiency neural conversion in hE SCs and hiPSCs and could be used as a powerful tool in large-scale generation of neural progenitors for therapeutic applications.

The terminal differentiation potential of PAX6+cells derived with compound C was confirmed. Immunofluorescence of Neuron specific class ????-tubulin (TUJ1), a pan-neuronal marker, was nearly uniformly expressed in cells after 2-week

neural maturation (Fig. 2F, I). In contrast, TUJ1 immunofluorescence was not detected in undifferentiated hESCs or compound C-induced PAX6+cells (data not shown). In addition, a fraction of the cells also expressed Nur-related factor 1 (NURR1), a dopaminegic neuron marker (Fig. 2F, II). Thus, the PAX6+neural progenitors derived by the use of compound C have the potential to undergo terminal differentiation and to produce neuronal subtypes. It was reported that compound C/dorsomorphin

can promote cardiomyogenesis in mouse E SCs (mESCs) [21], prompting us to examine whether it could exert the same effect in hE SCs. However, we found that compound C failed to induce cardiomyogenesis in h E

SCs (Supplementary Text; Fig. S3). In addition, treatment of mESCs with compound C failed to induce early neural conversion (Supplementary Text; Fig. S3). The differential effects of

compound C in mESCs and hESCs might be due to their differences in species, origin of

development or other undefined characteristics. Some known signaling pathways mediating self-renewal and differentiation of mE SCs are not conserved in hESCs [22, 23].

Compound C Targets Several Families of Protein Kinases

Compound C is widely used as a ATP-competitive inhibitor of AMP-activated kinase (AMPK), a multimeric protein complex that regulates cellular and organismal metabolism [24, 25], but recently its specificity has been questioned [26, 27]. In keeping with this, compound C has been shown to also target the type I BMP receptors (ALK2, ALK3 and ALK6) and likely other protein kinases [27, 28]. To gain a broader, more comprehensive view of the potential targets of compound C, we assessed its effects on a panel of kinases (402) in a large-scale in-vitro kinase assay, as described [29]. This assay identified far more targets of compound C than previously recognized (Fig. 3A and Table S2). Notably, compound C robustly inhibited many other members of TGF-

E superfamily receptors in addition to ALK2, ALK3 and ALK6 and of the SNF/AMPK family kinases in addition to AMPK D1 and D2 (Fig. 3A). Furthermore, compound C potently inhibited PDG

F receptors (PDGFR D and PDGFR E), VEGFR receptors [VEGFR1 (FLT1) and VEGFR2 (KDR)], FLT3 and KIT (Fig. 3A). To determine if AMPK inhibition was responsible for the effects of compound C on hESCs, we abolished the function of AMPK by depleting the ?1 and ?2 subunits. Surprisingly, depletion of either ?1 or ?2 subunit failed to support the undifferentiated growth and pluripotency of hE SCs and instead markedly decreased the levels of SOX2 protein (Fig. S4). Furthermore, AICAR, an AMPK activator, failed to rescue the phenotypic changes caused by compound C in hE SCs. Based on these results, we concluded that AMPK inhibition was not responsible for the effect of compound C. Inhibition of PDGF receptors in hE SCs also failed to mimic the effects of compound C. Treatment of hESCs with PDGF receptor kinase inhibitor V slightly decreased cell proliferation (data not shown).

Compound C Inhibits Both Activin And BMP Signaling in hESCs

Two type II receptors, ActRIIA and ActRIIB, mediate Activin signaling [30] and were markedly inhibited by compound C in vitro (Fig. 3A), leading us to assess whether compound C targeted this pathway in hE SCs. We found that compound C inhibited the activity of a luciferase-based Activin reporter system (pARE-GL3, [19]) in a dose-dependent manner (Fig. 3B). At a dose that effectively modulated hESC differentiation (1 P M), compound C consistently reduced the Activin reporter activity triggered by the addition of Activin A (100 ng/mL), to a similar extent to low concentration of SB431542 (250 nM) (Fig. 3B), which inhibits type I receptors including ALK4, ALK5 and ALK7 and consequently blocks the Activin signaling pathway, without affecting BMP signaling [31].

A higher dose of compound C (10 P M) was required to completely prevent the Activin reporter activity. In keeping with reduced reporter activity, compound C (1 P M) significantly attenuated the increase in the nuclear phospho-Smad2/3 levels in h

E

SCs induced by Activin A (Fig. 3C). Thus, compound C potently inhibited the Activin pathway in hE SCs. Interestingly, inhibition of Activin signaling by compound C was observed in mouse pulmonary artery smooth muscle cells (PASMCs) at higher concentrations [28]. The variability might be attributed to the differences in cell types or other undefined characteristics.

Compound C also blocked BMP signaling in hE SCs. Compound C inhibited a luciferase-based inhibitor of DNA binding (ID) reporter activity (Fig. S5A). It reduced BMP-4-induced activation (phosphorylation) of Smad1/5/8 in hE SCs (Fig. S5B). Thus, compound C potently inhibited the BMP-4 signaling pathway in hE SCs. These results are consistent with the

ability of compound C to inhibit both type I (ALK2, ALK3 and ALK6) and type II BMP receptors (ActRIIA and ActRIIB) in vitro. The effects of compound C on BMP signaling in hE SCs confirm and extend previous studies in other model systems [28].

Compound C Prevents Differentiation Activities Induced By Activin A and BMP-4

It has been well established that activation of Activin signaling triggers endoderm activities in E SCs [9, 32]. Furthermore, Activin A can directly act to down-regulate PAX6 expression [33]. In RPMI medium containing low serum, Activin A induces efficient differentiation of hE SCs to definitive endoderm [9]. We asked whether compound C blocked Activin A - induced endoderm activities. hE SCs exhibited reduced survival in RPMI medium after the addition of compound C, prompting us to use a custom mTeSR medium instead, which lacks bFGF and TGF-E and thus causes cells to differentiate. Compound C did not cause apoptosis of cells cultured in the custom mTeSR medium. As expected, the addition of Activin A (100 ng/mL) induced up-regulation of endoderm markers including SOX17,GSC, and FOXA2 in hESCs (Fig. 3D). The presence of compound C

(1 μM) reduced the up-regulation (Fig. 3D).

E xpectedly, SB431542, at a dose commonly used to inhibit Activin signaling (10 P M) [31], nearly completely prevented the up-regulation of the endoderm markers (Fig. 3D). In contrast, Noggin, a secreted protein that antagonizes BMP signaling, exerted little effects (Fig. 3D). Thus, compound C decreased Activin A-induced endoderm activities in hE SCs, separately from its actions on BMP receptors.

BMP-4 induced mesoderm or trophoectoderm activities in hE SCs, depending upon the length of stimulation [7, 8]. We thus treated hE SCs with BMP-4 (20 ng/mL) for 24 h and 6 d, with or without compound C (1 μM). Short-term BMP-4 stimulation (24 h) induced a rapid up-regulation of mesoderm markers, including T, MESP1and MIXL1 (Fig.S5C). Compound C nearly completely abolished the up-regulation (Fig. S5C). In addition, compound C prevented the up-regulation of CDX2 and down-regulation

of pluripotency markers in hE SCs after long-term exposure to BMP-4 (Supporting Information) (Fig. S5D and S5

E

). Thus, compound C inhibited BMP-4-induced mesoderm and trophoectoderm activities in hESCs.

The large-scale in vitro kinase assay showed that compound C targeted all three type I and two type II BMP receptors, which might explain its potent inhibition of BMP signaling activities in hESCs. To test this possibility, we compared the effects of depleting a single or multiple BMP receptors. Indeed, depletion of ALK2, ALK3 or ALK6 alone failed to affect BMP4-induced signals (Fig. S6A). In contrast, depletion of all three receptors nearly abolished BMP-4-induced signals including Smad1/5/8 phosphorylation, and up-regulation of the early BMP-4 responsive gene ID1and the differentiation markers T, MESP1 and CDX2 (Fig. S6B). Moreover, addition of compound C to the receptor-depleted cells caused no additional effects on Smad1/5/8 phosphorylation (Fig. 3E). Depletion of two type

I receptors also attenuated up-regulation of target genes, although the effects were less prominent than those from depleting all three (Fig. S6B). Furthermore, in cells induced to spontaneously differentiate, mRNA levels of mesoderm and trophoectoderm differentiation markers T and CGB7were reduced in cells with triple depletion, whereas the decrease of the pluripotency and neuroectoderm markers such as POU5F1,NANOG and SOX1was attenuated (Fig. S6C). These results suggest that inhibition

of at least two receptors is necessary for robust inhibition of BMP signals. Together, the ability

to target multiple TGF-E family receptors, dual inhibition of Activin and BMP signaling and suppression of Activin- and BMP-induced endoderm, mesoderm and trophoectoderm activities provide mechanistic explanations for the strong effects of compound C in hESCs.

MSX2 as A Compound C's Target Gene and a Key Intermediate of BMP Signaling in hESCs To identify potential target genes and signaling pathway(s) of compound C in hE SCs, we used Affymetrix-based whole-genome microarrays to compare global gene-expression profiles of differentiating hE SCs (induced by bFGF and CM removal) with and without compound C treatment. Focusing on early times (12 and 24 h) after treatment, we sought to identify early responsive gene(s) and/or signaling pathway(s). Microarray analyses of cells at 12 and 24 h identified a small number of genes with 1.5-fold altered expression (Table S3). Interestingly, a subset of transcription factors, including ID2, muscle segment homeobox gene 2 (MSX2) and distal-less homeobox 2 (DLX2), were down-regulated 12 and 24 h after compound C treatment (Table S3, highlighted in yellow). Among those, IDs are shown as downstream genes of the BMP signaling in hE SCs and mE SCs [7, 34]. Real-time PCR confirmed and extended these findings: transcription of the ID family members (ID1-ID3) and MSX family members (MSX1 and MSX2) were markedly suppressed after compound C treatment (Fig. 4A).

Despite the established roles of MSX family transcription factors in development, their functions have not been depicted in hESCs. We explored their potential functions in mediating BMP signaling pathway. BMP-4 stimulation rapidly up-regulated the mRNA levels of MSX1 and MSX2 in hE SCs (as early as 30 min post-stimulation) (Fig. 4B). Concomitant to the increase in mRNA, MSX2 protein levels were also elevated after BMP-4 stimulation (Fig. 4C). In contrast, BMP-4 stimulation failed to alter levels of MSX1 protein, and in addition, mRNA levels of MSX1were significantly lower than those of MSX2 in hESCs.

Depleting MSX2 prevented the morphological changes induced by BMP-4 in hESCs (Fig. 4D). Western blot analysis showed that MSX2 depletion also attenuated the down-regulation of OCT-4 and SOX2 (Fig. 4E). Interestingly, the rescuing effect of MSX2 depletion on OCT-4 expression was more prominent than SOX2. In addition, down-regulation of mRNA levels of POU5F1, SOX2 and NANOG was much slower

in MSX2-depleted cells than control cells after BMP-4 stimulation. These results indicate that MSX2 acts as a key component of BMP signaling in hE SCs. Future experiments will dissect the potential role of the DLX family members in regulating BMP signals or other signaling pathways, pluripotency and differentiation in hESCs.

D ISCUSSION

Developing conditions and factors for efficient directed differentiation of human pluripotent stem cells such as hESCs and hiPSCs is essential

for their potential applications for the study of early human development, drug discovery and the treatment of diseases. Two strategies have been explored. One is to manipulate developmental pathways via the use of growth factors and their antagonists, and the other is to identify chemical compounds to direct the differentiation activities. The benefits of utilizing compounds to control stem cell fate include the elimination of animal products, better temporal and tunable control, ease of production and significant cost reduction for materials. Despite these benefits, there is no documentation to date

for compounds that induce efficient differentiation from hESCs and hiPSCs to neural progenitors. Here we identified compound C, which potently suppresses mesoderm, endoderm and trophoectoderm activities and can be applied

to high-efficiency neural conversion in hE SCs and hiPSCs. Our study provides the first example for the efficient derivation of neural progenitor cells from human pluripotent stem cells with a single chemical compound.

We found that compound C can induce nearly 90% of hE SCs to form neuroectoderm in adherent cultures. Why is this compound so potent in inducing neural conversion? Our results suggest the following mechanisms. Compound C inhibits at least seven type I and

type II TGF-E superfamily receptors, including two Activin receptors and five BMP receptors, thus effectively preventing Activin and BMP signals. Indeed, our experiments show that depletion of at least two BMP receptors is necessary for robust inhibition of BMP-induced signaling activities. Furthermore, dual inhibition

of the BMP and the Activin pathway is necessary for efficient neural conversion in hESCs. In this scenario, inhibition of Activin and BMP signaling suppresses endoderm, mesoderm and trophoectoderm activities, thereby

facilitating h

E SC differentiation into the

neuroectoderm lineage. This notion is supported by our observation and a recent report that inhibition of BMP and Activin signaling alone improves the efficiency of neural induction, but combined inhibition (with Noggin and SB431542) leads to high-efficiency neural conversion (Fig. 2) [10]. Therefore, our results identified TGF-E superfamily receptors and the Activin and BMP signaling pathways as the main biological targets in hESCs. The ability to target multiple TGF-E family receptors, dual inhibition of Activin and BMP signaling and suppression of Activin- and BMP-induced endoderm, mesoderm and trophoectoderm activities could account for the effects of compound C on differentiation and neural conversion in hESCs.

Neural progenitors and more specified functional neural subtypes were derived previously from hESCs by using EBs as the initial step [35, 36]. While the EB-based procedures are effective and flexible in producing desired differentiated cell types, they often involve multiple steps, produce multiple differentiation lineages and require subsequent isolation/enrichment of specific differentiated cells. These limitations could hinder the acquisition of efficient neural conversion and the generation of large-scale neural precursors. In our study, we applied a single chemical compound to hESCs and hiPSCs cultivated in adherent cultures and achieved rapid, high-efficiency neural conversion in a single step. Compared with the Noggin - based protocol [10], our method produces a slightly higher efficacy of induction and also offers some other potential benefits in the long term. First, compound C costs significantly less than Noggin, thereby markedly reducing the cost for deriving neural progenitor cells from hESCs and hiPSCs, especially on a large-scale. Second, utilization of the defined medium mTeSR [37] prior to the induction of neural conversion eliminates the variability introduced by the ME Fs or ME F-CM. Furthermore, xeno-free TeSR2 and KSR media can be used if more clinically-compatible culture conditions are desired. Third, compound C can be conveniently delivered and removed and is therefore highly suited for therapeutic applications. Thus, our method is highly suitable for large-scale efficient derivation of potentially therapy-grade neural precursors from hE SCs and hiPSCs and should facilitate the utilization of these cells for the study of early human neural development, drug discovery, tissue repair and regeneration. Compound C had long been used as a specific inhibitor against AMPK [24, 25]. It was recently found to inhibit the type I BMP receptors ALK2, ALK3 and ALK6 and induce dorsalization in zebrafish (thus denoted as dorsomorphin) [28]. Our large-scale proteomic analysis confirmed these known targets but discovered far more additional targets, including many other members of the TGF-E superfamily receptors, of the SNF/AMPK family and PDGF receptors. This approach served as a starting point, enabling us to identify the targets and pathways of compound C in hE SCs. Our findings might explain the variability in previous studies with compound C and could provide a platform for further analysis of its potential targets in various model systems. They also suggest that experimentation with kinase inhibitors should be evaluated critically under the specific cellular and biological context. As such, our results outline a strategy that may be utilized in future studies with kinase inhibitors.

A CKNOWLEDGMENTS

We thank Dr. Renhe Xu for providing the ID-120-luc reporter construct and Dr. Yisrael Sidis for providing the pARE-GL3 construct, Jenny Drnevich for analyzing the microarray data and members of the Wang lab for helpful discussions. Support was provided by NIH (GM-83812; to F.W.), the Illinois Regenerative Medicine Institute (IDPH 2006-05516; to F.W.), NSF CARE E R award (0953267; to F.W.), the Beckman award from the University of Illinois and the National Natural Science Foundation of China (30728022; to F.W. and E.D.). Disclosure of Potential Conflicts of Interest The authors indicate no potential conflicts of interest.

R EFERENCES

1. Thomson JA, Itskovitz-E ldor J, Shapiro SS, et al.

mbryonic stem cell lines derived from human blastocysts. Science. 1998;282:1145-1147.

2. Yamanaka S. A fresh look at iPS cells. Cell.

2009;137:13-17.

3. Levenstein ME, Ludwig TE, Xu RH, et al. Basic

fibroblast growth factor support of human embryonic stem cell self-renewal. Stem Cells. 2006;24:568-574. 4. Vallier L, Alexander M, Pedersen RA. Activin/Nodal

and FGF pathways cooperate to maintain pluripotency of human embryonic stem cells. J Cell Sci.

2005;118:4495-4509.

5. Xu RH, Sampsell-Barron TL, Gu F, et al. NANOG is a

direct target of TGFbeta/activin-mediated SMAD signaling in human ESCs. Cell Stem Cell. 2008;3:196-206.

6. Chadwick K, Wang L, Li L, et al. Cytokines and BMP-4

promote hematopoietic differentiation of human embryonic stem cells. Blood. 2003;102:906-915.

7. Xu RH, Chen X, Li DS, et al. BMP4 initiates human

embryonic stem cell differentiation to trophoblast. Nat Biotechnol. 2002;20:1261-1264.

8. Zhang P, Li J, Tan Z, et al. Short-term BMP-4 treatment

initiates mesoderm induction in human embryonic stem cells. Blood. 2008;111:1933-1941.

9. D'Amour KA, Agulnick AD, Eliazer S, et al. Efficient

differentiation of human embryonic stem cells to definitive endoderm. Nat Biotechnol. 2005;23:1534-1541.

10. Chambers SM, Fasano CA, Papapetrou E P, et al.

Highly efficient neural conversion of human E S and iPS cells by dual inhibition of SMAD signaling. Nat Biotechnol. 2009;27:275-280.

11. Pera MF, Andrade J, Houssami S, et al. Regulation of

human embryonic stem cell differentiation by BMP-2 and its antagonist noggin. J Cell Sci. 2004;117:1269-1280.

12. Smith JR, Vallier L, Lupo G, et al. Inhibition of

Activin/Nodal signaling promotes specification of human embryonic stem cells into neuroectoderm. Dev Biol. 2008;313:107-117. 13. Huangfu D, Osafune K, Maehr R, et al. Induction of

pluripotent stem cells from primary human fibroblasts with only Oct4 and Sox2. Nat Biotechnol.

2008;26:1269-1275.

14. Watanabe K, Ueno M, Kamiya D, et al. A ROCK

inhibitor permits survival of dissociated human embryonic stem cells. Nat Biotechnol. 2007;25:681-686.

15. Borowiak M, Maehr R, Chen S, et al. Small molecules

efficiently direct endodermal differentiation of mouse and human embryonic stem cells. Cell Stem Cell.

2009;4:348-358.

16. Chen S, Borowiak M, Fox JL, et al. A small molecule

that directs differentiation of human E SCs into the pancreatic lineage. Nat Chem Biol. 2009;5:258-265. 17. Zhou J, Su P, Wang L, et al. mTOR supports long-term

self-renewal and suppresses mesoderm and endoderm activities of human embryonic stem cells. Proc Natl Acad Sci U S A. 2009;106:7840-7845.

18. López-Rovira T CE, Massagué J, Rosa JL, Ventura F.

Direct binding of Smad1 and Smad4 to two distinct motifs mediates bone morphogenetic protein-specific transcriptional activation of Id1 gene. J Biol Chem.

2002;277:3176-3185.

19. Sidis Y, Tortoriello DV, Holmes WE, et al. Follistatin-

related protein and follistatin differentially neutralize endogenous vs. exogenous activin. E ndocrinology.

2002;143:1613-1624.

20. Xu C, Inokuma MS, Denham J, et al. Feeder-free

growth of undifferentiated human embryonic stem cells. Nat Biotechnol. 2001;19:971-974.

21. Hao J, Daleo MA, Murphy CK, et al. Dorsomorphin, a

selective small molecule inhibitor of BMP signaling, promotes cardiomyogenesis in embryonic stem cells.

PLoS One. 2008;3:e2904.

22. Daheron L, Opitz SL, Zaehres H, et al. LIF/STAT3

signaling fails to maintain self-renewal of human embryonic stem cells. Stem Cells. 2004;22:770-778. 23. Ying QL, Nichols J, Chambers I, et al. BMP induction

of Id proteins suppresses differentiation and sustains embryonic stem cell self-renewal in collaboration with STAT3. Cell. 2003;115:281-292.

24. DG H. AMP-activated/SNF1 protein kinases:

conserved guardians of cellular energy. Nat Rev Mol Cell Biol. 2007;8:774-785.

25. Zhou G, Myers R, Li Y, et al. Role of AMP-activated

protein kinase in mechanism of metformin action. J Clin Invest. 2001;108:1167-1174.

26. E merling BM VB, Tormos KV, Chandel NS.

Compound C inhibits hypoxic activation of HIF-1 independent of AMPK. FE BS Lett. 2007;581:5727-

5731.

27. Bain J, McLauchlan H, E lliott M, et al. The

specificities of protein kinase inhibitors: an update.

Biochem J. 2003;371:199-204.

28. Yu PB, Hong CC, Sachidanandan C, et al.

Dorsomorphin inhibits BMP signals required for embryogenesis and iron metabolism. Nat Chem Biol.

2008;4:33-41.

29. Fabian MA, Biggs WH, 3rd, Treiber DK, et al. A small

molecule-kinase interaction map for clinical kinase inhibitors. Nat Biotechnol. 2005;23:329-336.

30. Attisano L WJ. Signal transduction by the TGF-beta

superfamily. Science. 2002;296:1646-1647.

31. Inman GJ, Nicolas FJ, Callahan JF, et al. SB-431542 is

a potent and specific inhibitor of transforming growth

factor-beta superfamily type I activin receptor-like

kinase (ALK) receptors ALK4, ALK5, and ALK7. Mol Pharmacol. 2002;62:65-74.

32. Yasunaga M, Tada S, Torikai-Nishikawa S, et al.

Induction and monitoring of definitive and visceral endoderm differentiation of mouse E S cells. Nat Biotechnol. 2005;23:1542-1550.

33. Pituello F, Yamada G, Gruss P. Activin A inhibits Pax-

6 expression and perturbs cell differentiation in the

developing spinal cord in vitro. Proc Natl Acad Sci U S

A. 1995;92:6952-6956.

34. Hollnagel A OV, Heymer J, Rüther U, Nordheim A. Id

genes are direct targets of bone morphogenetic protein induction in embryonic stem cells. J Biol Chem.

1999;274:19838-19845.

35. Perrier AL, Tabar V, Barberi T, et al. Derivation of

midbrain dopamine neurons from human embryonic stem cells. Proc Natl Acad Sci U S A.

2004;101:12543-12548.

36. Zhang SC WM, Duncan ID, Brüstle O, Thomson JA.

In vitro differentiation of transplantable neural precursors from human embryonic stem cells. Nat Biotechnol. 2001;19:1129-1133.

37. Ludwig TE, Levenstein ME, Jones JM, et al.

Derivation of human embryonic stem cells in defined conditions. Nat Biotechnol. 2006;24:185-187.

See https://www.wendangku.net/doc/9518740365.html, for supporting information available online.

Fig. 1. Compound C suppresses mesoderm, endoderm and trophoectoderm differentiation in hESCs.

(A) Immunofluorescence studies of OCT-4, SOX2 and NANOG proteins (red) in H1 cell colonies cultured in complete growth medium (CM + bFGF), medium with KSR only, and medium with KSR and 1 P M compound C for 6 d. All three pluripotency markers were expressed in 92.5%±4.7% of the colonies (i.e., with CM and bFGF). Removal of CM and bFGF reduced the OCT-4-, SOX2- and NANOG-positive colonies to 8.1%±3.2%, and addition of compound C increased it to 88.4%±6.5%. Nuclei were stained with DAPI (blue). H9 cells showed similar response to compound C treatment. Dose-dependent experiments with compound C (100 nM, 200 nM, 500 nM, 1 P M and 2 P M) showed that 1 P M was necessary to give rise to the strong anti-differentiation effect. Scale bar, 200 μm. (B) mRNA levels of SOX2,POU5F1 and NANOG in H1 hE SCs cultured in aforementioned three conditions for 6 d, assessed by real-time PCR. All values were normalized to the level (=1) of mRNA in the cells treated with KSR alone. *, p<0.05; **, p<0.01. Cells treated with compound C were compared to control cells (i.e., with KSR only) treated with vehicle. ACTB (E-actin gene) was used as an internal control. (C) mRNA levels of differentiation markers in H1 hESCs cultured in medium with KSR only and medium with KSR and 1 P M compound C for 6 d, assessed by real-time PCR. All values were normalized to the level (=1) of mRNA in the cells treated with KSR alone. *, p<0.01; **, p<0.001. Cells treated with compound C were compared to control cells treated with vehicle. NE and TE denote neuroectoderm and trophoectoderm, respectively. (D) mRNA levels of control E Bs or compound C-treated EBs assessed by real-time PCR. Four separate experiments were conducted, and quantification of three replicates of a typical experiment is shown. E ach bar represents the mean ± SEM (error bars). All values were normalized to the mRNA level (=1) of in the control cells on day 0. *, p<0.05; **, p<0.01. EBs treated with compound C were compared to control EBs on day 4.

Fig. 2. Compound C induces high-efficiency neural conversion. (A) Immunofluorescence studies of PAX6 (left panel, red) and SOX1, (middle panel, green) and SOX2 (right panel, red) in differentiating H1 hE SCs. Cell nuclei were stained with DAPI (blue). In contrast to control cells, which exhibited minimal anti-PAX6 fluorescence, H1 cells treated with compound C alone (1 μM) or with compound C (1 P M) and SB431542 (50 nM) combined expressed high level of PAX6 after 7-d differentiation. SOX1 and SOX2 were also highly expressed in compound C or compound C and SB431542 treated cells. Bar, 200 P m. (B) Western blot analysis of PAX6, Nestin and SOX2 in H1 cells with three treatments. Cells were induced to differentiate for 7 d. A typical experiment from five separate experiments is shown. D-tubulin was a loading control. (C) The percentage of PAX6+ cells assessed by flow cytometry. H1 hE SCs treated with compound C alone or with compound C and SB431542 contained 88.7±2.5% and 80.3±10.3% (mean ± SEM) PAX6+ cells, respectively. In contrast, control cells and cells treated with SB431542 showed 16.9±1.9% and 17.7±0.4% PAX6+ cells. Treatment with Noggin (500 ng/mL) alone or combined with SB431542 gave rise to 36.9±1.6% and 84.7±3.9% PAX6+ cells, respectively. Similarly, compound C treatment of IMR-90-derived hiPSCs induced a high population of PAX6+ cells (70.3%±5.5%), in contrast to untreated cells (4.8%±2.1%). (D) Immunofluorescence studies of PAX6 (top panel, red) and SOX2 (bottom panel, red) in differentiating hiPSCs. Cell nuclei were stained with DAPI (blue). Cells were treated or not treated with 1 P M compound C for 7 d. scale bar, 50 μm. (E) Western blot analysis of PAX6 and SOX2 in hiPSCs treated or not treated with 1 P M compound C. Cells were induced to differentiate for 7 d. D-tubulin was a loading control. (F) Immunofluorescence studies of TUJ1 and NURR1 in terminally differentiated H1 hESCs. Anti-TUJ1 (?) and NURR1 (??) fluorescence (red) was observed in H1-derived differentiated cells after 2-w neural maturation. Nuclei were stained with DAPI (blue). Scale bar, 50 μm.

Fig. 3. Compound C targets the TGF-E superfamily receptors and blocks Activin and BMP signaling.

(A) Some of the kinases inhibited by 1 μM compound C and the degree of inhibition, as revealed by the in vitro kinase assay. The complete list of kinases examined is shown in Supplementary Table S2.

(B) Relative luciferase activity of the pARE-GL3 reporter in H1 cells with or without Activin A (100 ng/mL, 24 h), treated with different concentrations of SB421542 or compound C. Results from four separate experiments are shown as mean±SE M. *, p<0.05 ; **, P<0.01; ***, p<0.001. Cells with various treatments were compared to control (with Activin stimulation). Hint amount of Rellina plasmid was co-transfected as an internal control. (C) Western blot analysis of phosphorylated Smad2/3 and total Smad2/3 in the nuclear fraction of H1 cells treated with various concentrations of compound C and stimulated with Activin-A (100 ng/mL) for 24 h. A typical experiment from four separate experiments is shown. Total Smad2/3 was a loading control. Compound C (1 P M) reduced the nuclear p-Smad2/3 levels by approximately 25%. (D) mRNA levels of endoderm markers SOX17, FOXA2 and GSC in H1 cells with various treatments, assessed by real-time PCR. Cells were stimulated with Activin A (100 ng/mL) for 5 d. Four separate experiments were conducted, and quantification of three replicates of a typical experiment is shown. E ach bar represents the mean ± SEM (error bars). All values were normalized to the mRNA level (=1) of in the control cells. Activin and compound C treatments vs Activin alone were compared (*, p<0.05; **, p<0.01). (E) Western blot analysis of Smad1/5/8 phosphorylation under various conditions. Similar to 1 P M compound C treatment, depletions of ALK2, ALK3 and ALK6 altogether (denoted as Triple KD) attenuated the phosphorylation of Smad1/5/8 induced by BMP-4 (20 ng/mL, 30 min). Further addition of 1 P M compound C to cells lacking the three receptors exerted no detectable effects. A typical blot is shown in the bottom panel, and quantification of blots from four separate experiments is shown in the top panel.D-tubulin was a loading control. Y-axis represents relative intensities (measured with Image J) with values normalized to the signal (=100%) without BMP-4 or compound C treatment. E ach bar represents mean ± SE M (error bars). Student t tests compared data between cells treated with compound C vs control cells after BMP-4 addition (**, p<0.01).

Fig. 4. MSX2 as a target gene of compound C and a key intermediate of the BMP pathway. (A) mRNA levels of ID1,ID2,ID3,MSX1 and MSX2 in H1 hESCs induced to differentiate by the removal of bFGF and CM, with or without the treatment of 1 P M compound C, assessed by real-time PCR. Four separate experiments were conducted, and quantification of four replicates of a typical experiment is shown. Bars represent mean ± SE M (error bars). All values were normalized to the mRNA level in the control cells at 0 h. Cells treated with compound C were compared to control cells treated with vehicle. **, p<0.01; ***, p<0.001. (B) The mRNA level of MSX2 in H1 hE SCs after BMP-4 treatment (20 ng/mL) for various time points assessed by real-time PCR. (C) Western blot analysis of MSX2, OCT-4 and SOX2 in H1 cells after BMP-4 treatment (20 ng/mL) for various time points. (D) Phase-contrast images of H1 cells with MSX2 depletion 4 d after BMP-4 treatment (20 ng/mL). Two separate shRNAs were used to verify the effects of MSX2 depletion, while scramble shRNA was used as a control. Scale bar, 50 μm. ( Western blot analysis of MSX2, OCT-4 and SOX2 in H1 cells with MSX2 depletion 4 d after BMP-4 treatment (20 ng/mL). Two separate shRNAs (sh4849 and sh4850) were used to verify the effects of MSX2 depletion. D-tubulin was used as loading controls for (C) and (E).

神经干细胞的研究及其应用新进展

神经干细胞的研究及其应用新进展 [关键词] 神经干细胞研究 健康讯: 崔桂萍天津市脑系科中心医院 300060 1992 年, Reynolds 首次成功地从成年小鼠纹状体中分离出神经干细胞( neural stem cell, NSC ),于是“神经干细胞”这一概念被正式引入神经科学研究领域。可以总结为具有分化为神经元、星形细胞和少突胶质细胞的能力,能自我更新并足以提供大量脑组织细胞的细胞。不少文献中还提到神经祖细胞和神经前体细胞,目前认为,神经祖细胞是指比 NSC 更有明确发展方向的细胞,而神经前体细胞是指处于发育早期的增殖细胞,可指代 NSC 和神经祖细胞:与 NSC 相比,二者的分裂增殖能力较弱而分化能力较强,是有限增殖细胞,但三者均属 NSC 范畴。 1. NSC 的起源、存在部位及生物学特征中枢神经系统的发育起源于神经沟、神经嵴、神经管;研究发现, NSC 在神经管壁增殖,新生细胞呈放射状纤维迁移至脑的特定位置;主要存在于室管膜区,在成脑生发区以外的区域也广泛分布,即具有高度可塑性的神经前体细胞。现发现 NSC 的生物学特征为:( 1 )具有自我更新能力;( 2 )具有多向分化潜能,可分化为神经元、星形细胞和少突胶质细胞;( 3 )处于高度未分化状态;( 4 )终生具有增殖分化能力,在有损伤的局部环境信号变化的刺激下可以增殖分化。其中( 1 )和( 2 )是 NSC 的两个基本特征。 2. NSC 的基础研究进展 NSC 的增殖和分化调控是目前 NSC 研究的核心问题,最近的研究资料显示, NSC 的增殖、分化、迁移调控受多种相关因素的影响。神经递质神经递质作为细胞外环境的一员,不仅介导神经元之间和神经元与效应器之间的信号传递,还参与 NSC 的增殖和分化。这些神经递质包括谷氨酸( G1u )、 5- 羟色胺( 5-HT )、 GABA 、甘氨酸( G1y )、乙酰胆碱( Ach )一氧化氮( NO )、肾上腺素与性激素等。 G1u :在脑的发育过程中有高含量的 G1u 表达, Haydar 等发现, G1u 可以通过大鼠胚胎皮质 AMPA/KAR 的激活调节室周区前体细胞的增殖,但 GLU 对室管膜区( SZ )和室管膜下区( SVZ )体内细胞的影响是不同的,它可增加 SZ 细胞的增殖,减少 SVZ 细胞的增殖; GLU 还可促进神经元生长和分化。 5-HT :许多研究表明, 5-HT 在皮质发育、突触形成中起重要作用,抑制 5-HT 合成或选择性损伤 5-HT 神经元则引起齿状回及脑室下区神经元增殖活性下降, 5-HT 可促进胶质细胞分化和髓鞘形成。 GABA : GABA 是成体脑发育过程中主要

C3H1OT12细胞向神经元诱导分化的方法研究

C3H1O T1 2细胞向神经元诱导分化的方法研究 邓均 艾国平 王军平 徐辉 邹仲敏 闫国和 董世武 郝磊 冉新泽 粟永萍 【摘 要】 目的 探讨体外诱导间充质干细胞(m esenchym al stem cells,M SC s)系C3H1O T1 2细胞分化为神经元细胞的方法。 方法 取3月龄SD大鼠1只,体重200g,进行神经元细胞的原代培养及诱导上清液的收集,用细胞免疫组织化学方法进行鉴定。将C3H1O T1 2细胞进行培养,分别用Β巯基乙醇(A组)和神经元原代细胞上清液(B组)作为诱导剂,对C3H1O T1 2细胞进行诱导分化;对照组(C组)不加诱导剂进行培养。采用细胞免疫组织化学方法对分化的细胞进行鉴定。 结果 细胞免疫化学检测原代培养细胞为神经元,表达特异标志物:神经丝蛋白(neu rofilam en t p ro tein, N F)和神经元特异性烯醇化酶(neu ron specific eno lase,N SE)。A组C3H1O T1 2细胞经Β巯基乙醇诱导2h即表达N F和N SE,5h表达强度增强。B组C3H1O T1 2细胞经原代神经元培养上清液诱导,1d有N F和N SE表达,但表达强度比A 组弱。各组N SE表达阳性率和强度均高于N F。 结论 化学分子和微环境体液因素均可诱导M SC s向神经元细胞分化;为M SC s通过神经分化参与脑创伤的修复提供了理论依据。 【关键词】 间充质干细胞 分化 神经元细胞 诱导 中图分类号:Q813111 R329.2 文献标识码:A EXPER I M ENTAL INVESTIGATI ON ON CHARACTER ISTI CSOF C3H1OT1 2CELL IND UCED D IFFERENTI ATI ON INT O NEUR ON-L IKE CELL S D EN G J un,A I Guop ing,W A N G J unp ing,et a l.Institu te of Co m bined Inju ry,Colleg e of P reven tive M ed icine,T h ird M ilita ry M ed ica l U n iversity,Chong qing,400038,P.R.Ch ina.E2m a il:dj t mm u@to m. co m Corresp ond ing au thor:SU Y ongp ing,E2m a il:suyp2003@y ahoo.co https://www.wendangku.net/doc/9518740365.html, 【Abstract】 Objective To exp lo re the m ethod that can induce the m esenchym al stem cells(M SC s)to differen tiate in to the neu ron2like cells in v itro.M ethods T he neu ron2like cells w ere iso lated from an SD rat(age, 3mon th s;w eigh t,200g).T hey underw en t a p ri m ary cu ltu re;the induced liqu id supernatan t w as co llected,and w as iden tified by the cell i m m unoh istochem istry.T he C3H1O T1 2cells w ere cu ltu red,as an M SC s model,and they w ere induced in to differen tiati on byΒ2m ercap toethano l(Group A)and by the liqu id supernatan t of the neu ron2like p ri m ary cells(Group B),respectively.T he cells w ere cu ltu red w ithou t any inducti on w ere u sed as a con tro l(Group C). I mm unoh istochem istry w as u sed to iden tify the type of the cells.Results T he resu lt of the i m m unochem istry show ed that the cells undergo ing the p ri m ary cu ltu re exp ressed the neu rofilam en t p ro tein(N F)and the neu ron specific eno lase (N SE),and they w ere neu ron2like cells.Β2m ercap toethano l cou ld induce the C3H1O T1 2cells to exp ress N F and N SE at2h,and the exp ressi on in ten sity increased at5h.T he liqu id supernatan t of the p ri m arily2cu ltu red neu ron2like cells cou ld induce the C3H1O T1 2cells to exp ress N F and N SE at1d,bu t the exp ressi on in ten sity induced by the liqu id supernatan t w as w eaker than that induced byΒ2m ercap toethano l.T he po sitivity rate and the in ten sity exp ressi on of N SE w ere h igher than tho se of N F.Conclusion M SC s can differen tiate in to the neu ron2like cells byΒ2m ercap toethano l and the m icroenvironm en t humo ral facto r,w h ich can pave the w ay fo r a fu rther study of the differen tiati on of M SC s and the effect of the differen tiati on on the b rain traum a repair. 【Key words】 M esenchym al stem cells D ifferen tiati on N eu ron2like cells Induce Founda tion ite m:N ati onal Basic Science R esearch and D evelopm en t Gran ts(2005CB522605) 近年来,干细胞由于其多向分化潜能成为研究热点,也成为细胞工程、再生医学研究中的主要选择细 基金项目:国家重点基础研究发展计划(973)资助项目(2005CB522605) 作者单位:第三军医大学军事预防医学院复合伤研究所(重庆, 400038) 通讯作者:粟永萍,教授,博士导师,研究方向:干细胞参与创伤修复的机制研究,E2m ail:suyp2003@https://www.wendangku.net/doc/9518740365.html, 胞[1,2]。神经干细胞的发现为中枢神经细胞变性疾病和中枢神经系统损伤的移植治疗提供了有效的移植物,但由于目前很难获得足够的神经干细胞,因此寻找一种新的移植细胞来源,对神经科学领域基础与应用研究的发展至关重要[3,4]。研究证实,间充质干细胞(m esenchym al stem cells,M SC s)具有可塑性,可跨胚层分化,诱导分化为外胚层的神经元细胞[5,6]。而且M SC s易获取,易生长,有较强的增殖能力,可成为神

关于神经干细胞

.关于神经干细胞 定义是一类具有多向分化潜能, 能够自我复制, 在特定诱因下, 能够向神经元或神经胶质细胞分化的未分化细胞的总称。它是神经系统形成和发育的源泉。其主要功能是参与神经系统损伤修复或细胞凋亡的更新。 特点⑴自我更新:神经干细胞具有对称分裂及不对称分裂两种方式,从而保持干细胞库稳定。对称分裂由一个神经干细胞产生两个神经干细胞;在特定诱因下进行非对称分裂,会产生神经干细胞和神经胶质细胞(astrocyte,oligodendrocyte)。⑵多向分化潜能:神经干细胞可以向神经元、星形胶质细胞和少突胶质细胞分化,其分化与局部微环境(niche)密切相关。⑶低免疫源性:神经干细胞是未分化的原始细胞,不表达成熟的细胞抗原,可以不被免疫系统识别。⑷良好的组织融合性:可以与宿主(即接受神经干细胞移植的患者)的神经组织良好融合,并在宿主体内长期存活。 发现时间1992年,Reynodls等从成年小鼠脑纹状体中分离出能在体外不断分裂增殖,且具有多种分化潜能的细胞群,并正式提出了神经干细胞的概念,从而打破了认为神经细胞不能再生的传统理论。 产生区域神经干细胞主要产生于脑室周围的室管膜下区(SVZ,subvetricular zone)和海马齿状回的颗粒下区(SGZ,subgranular zone)。成人大脑中每天有3万个神经干细胞产生,按照从脑室周围的室管膜下区(SVZ)通过侧迁移流RMS(rostral migratory)最后到达嗅球 OB(olfactory bulb) 的方向移动。增殖时间为12~28天/代。 2.治疗机理与应用领域

神经干细胞的治疗机理 ⑴患病部位组织损伤后释放各种趋化因子,可以吸引神经干细胞聚集到损伤部位,并在局部微环境的作用下分化为不同种类的细胞,修复及补充损伤的神经细胞。 ⑵由于缺血、缺氧导致的血管内皮细胞、胶质细胞的损伤,使局部通透性增加,另外在多种黏附分子的作用下,神经干细胞可以透过血脑屏障,高浓度的聚集在损伤部位。 ⑶神经干细胞可以分泌多种神经营养因子,刺激原有神经元和神经胶质细胞,促进损伤细胞的修复。 ⑷神经干细胞可以增强神经突触之间的联系,建立新的神经环路,降低脑部氧化性压力。 神经干细胞的应用领域 神经干细胞主要应用于治疗中枢神经系统疾病,包括脑部和脊髓损伤的治疗。面前可以治疗的疾病包括脑瘫,脑膜炎后遗症, 脑发育不良脑, 中风(脑出血,脑梗塞)及后遗症, 脑外伤及脊髓损伤, 运动神经元病, 肌萎缩性侧索硬化症(ALS), 帕金森病, 脑萎缩, 共济失调, 癫痫, 多系统萎缩症(MSA), 老年性痴呆及血管性痴呆, 各种舞蹈症, 急性感染性多发性神经根炎(格林巴利氏病), 神经性耳聋, 面瘫及各类周围神经病。 目前有许多研究结果证明神经干细胞的分化潜能不仅仅局限于所属组织,在特定环境(niche)中,在一些细胞因子和蛋白的作用下,可以跨过神经系统而分化成其他类型的组织细胞,即具有横向分化潜能。如神经干细胞可被诱导分化为肌细胞和造血前体细胞。这无疑在理论上扩大了神经干细胞在今后的应用范围,使得更多用现今医学手段无法治愈的患者看到希望。 3.本公司的神经干细胞

实验14-细胞凋亡的诱导和检测

实验14 细胞凋亡的诱导和检测 20世纪60年代人们注意到细胞存在着两种不同形式的死亡方式:凋亡(apoptosis)和坏死(necrosis)。细胞坏死指病理情况下细胞的意外死亡,坏死过程细胞膜通透性增高,细胞肿胀,核碎裂,继而溶酶体、细胞膜破坏,细胞容物溢出,细胞坏死常引起炎症反应。 细胞凋亡apoptosis一词来源于古希腊语,意思是花瓣或树叶凋落,意味着生命走到了尽头,细胞到了一定时期会像树叶那样自然死亡。凋亡是细胞在一定生理或病理条件下遵守自身程序的主动死亡过程。凋亡时细胞皱缩,表面微绒毛消失,染色质凝集并呈新月形或块状靠近核膜边缘,继而核裂解,由细胞膜包裹着核碎片或其他细胞器形成小球状凋亡小体凸出于细胞表面,最后凋亡小体脱落被吞噬细胞或邻周细胞吞噬。凋亡过程中溶酶体及细胞膜保持完整,不引起炎症反应。细胞凋亡时的生化变化特征是核酸切酶被激活,染色体DNA被降解,断裂为50~300 kb长的DNA片段,再进一步断裂成180~200bp整倍数的寡核苷酸片断,在琼脂糖凝胶电泳上呈现“梯状”电泳图谱(DNA Ladder)。细胞凋亡在个体正常发育、紫稳态维持、免疫耐受形成、肿瘤监控和抵御各种外界因素干扰等方面都起着关键性的作用。 1.细胞凋亡的检测方法 凋亡细胞具有一些列不同于坏死细胞的形态特征和生化特征,据此可以鉴别细胞的死亡形式。细胞凋亡的机制十分复杂,一般采用多种方法综合加以判断,同时不同类型细胞的凋亡分析方法有所不同,方法选择依赖于具体的研究体系和研究目的(表?)。

形态学观察方法:利用各种染色法可观察到凋亡细胞的各种形态学特征: (1)DAPI时常用的一种与DNA结合的荧光染料。借助于DAPI染色,可以观察细胞核的形态变化。 (2)Giemsa染色法可以观察到染色质固缩、趋边、凋亡小体形成等形态。 (3)吖啶橙(AO)染色,荧光显微镜观察,活细胞核呈黄绿色荧光,胞质呈红色荧光。凋亡细胞核染色质呈黄绿色浓聚在核膜侧,可见细胞膜呈泡状膨出及凋亡小体。 (4)吖啶橙(A())/溴化乙啶(EB)复染可以更可靠地确定凋亡细胞的变化,AO只进入活细胞,正常细胞及处于凋亡早期的细胞核呈现绿色;EB只进入死细胞,将死细胞及凋亡晚期的细胞的核染成橙红色。 (5)台盼蓝染色对反映细胞膜的完整性,区别坏死细胞有一定的帮助,如果细胞膜不完整、破裂,台盼蓝染料进入细胞,细胞变蓝,即为坏死。如果细胞膜完整,细胞不为台盼蓝染色,则为正常细胞或凋亡细胞。使用透射电镜观察,可见凋亡细胞表面微绒毛消失,核染色质固缩、边集,常呈新月形,核膜皱褶,胞质紧实,细胞器集中,胞膜起泡或出“芽”及凋亡小体和凋亡小体被临近巨噬细胞吞噬现象。 (6)木精-伊红(HE)染色是经典的显示细胞核、细胞质的染色方法,染色结果清晰。发生凋亡的细胞经HE染色后,其细胞大小的变化及特征性细胞核的变化:染色质凝集、呈新月形或块状靠近核膜边缘,晚期核裂解、细胞膜包裹着核碎片“出芽”凸出于细胞表面形成凋亡小体等均可明显显示出来。 DNA凝胶电泳:细胞发生凋亡或坏死,其细胞DNA均发生断裂,细胞小分子 质量DNA片段增加,高分子DNA减少,胞质出现DNA片段。但凋亡细胞DNA断裂点均有规律的发生在核小体之间,出现180~200 bp DNA片段,而坏死细胞的DNA断裂点为无特征的杂乱片段,利用此特征可以确定群体细胞的死亡,并可与坏死细胞区别。

神经干细胞的应用前景及研究进展

神经干细胞的应用前景及研究进展 生科1301班李桐 1330170031 神经干细胞( neuralstem cells, NSCs)是重要的干细胞类型之一,是神经系统发育过程中保留下来的具有自我更新和多向分化潜能的原始细胞,可分化为神经元、星形胶质细胞、少突胶质细胞等多种类型的神经细胞。具有很多的特性,如自我更新、多潜能分化、迁移和播散、低免疫原性、良好的组织相容性、可长期存活等。目前神经干细胞的分离与体外培养已取得可喜的进展,有关神经干细胞的研究已经成为国内外神经科学领域的热点。 一、神经干细胞的生物学特性 19世纪80年代提出了神经干细胞的概念,它是指一类多潜能的干细胞,能够长期自我更新与复制,并具有分化形成神经元、星形胶质细胞的能力。神经干细胞的主要特征:未分化、缺乏分化标记、能自我更新并具有多种分化潜能。它并不是指特定的单一类型的细胞,而是具有相类似性质的细胞群。Gage将神经干细胞的特性进一步描绘为以下三点,可生成神经组织或来源于神经系统,具有自我更新能力,可通过不对称法、分裂产生新细胞。神经干细胞经过不对称分裂产生一个祖细胞和另一个干细胞,祖细胞只有有限的自我更新能力,并自主分化产生神经元细胞和成胶质细胞。神经干细胞是具有自我更新和具有多种潜能的母系神经细胞,它能分化成各种神经组织细胞表型,如神经元、星形胶质细胞和少突胶质细胞.并能自我更新产生新的神经干细胞,在神经发育和神经损伤中发挥作用。神经干细胞移植、迁移及分化与局部环境密切相关,这种特性为移植及移植后的结构重建和功能恢复提供了依据,为移植治疗不同疾病提供了局可能。 二、神经干细胞的应用前景 1.细胞移植以往脑内移植或神经组织移植研究进展缓慢,主要受到胚胎脑组织的来源、数量以及社会法律和伦理等方面的限制。神经干细胞的存在、分离和培养成功,尤其是神经干细胞系的建立可以无限地提供神经元和胶质细胞,解决了胎脑移植数量不足的问题,同时避免了伦理学方面的争论,为损伤后进行替代治疗提供了充足的材料。研究表明,干细胞不仅有很强的增殖能力,而且尚有潜在的迁移能力,这一点为治疗脑内因代谢障碍而引起的广泛细胞受损提供了理论依据,借助于它们的迁移能力,可以避免多点移植带来的附加损伤。另外,神经干细胞移植也为研究神经系统发育及可塑性的实验研究提供了观察手段,前文提及细胞因子参与调控神经元增殖和分化,通过移植的手段对这些因素的具体作用形式和机制进行探索,为进一步临床应用提供了理论基础。 2.基因治疗目前诱导干细胞向具有合成某些特异性递质能力的神经元分化尚未找到成熟的方法,利用基因工程修饰体外培养的干细胞是这一领域的又一重大进展;另外已经发现许多细胞因子可以调节发育期甚至成熟神经系统的可塑性和结构的完整性,将编码这些递质或因子的基因导入干细胞,移植后可以在局部表达,同时达到细胞替代和基因治疗的作用。 3.自体干细胞分化诱导移植免疫至今为止仍是器官或组织移植的首要问题。前文提到已经证明成年动物或人脑内、脊髓内存在着具有多向分化潜能的干细胞,那么使人们很容易想到通过自体干细胞诱导来完成损伤的修复。中枢神经系统损伤后,首先反应的是胶质细胞,在某些因子的作用下快速分裂增殖,形成胶质瘢。其实在这个过程中也有干细胞的参与,可不幸的是大多数干细胞增殖后分化为胶

牙髓干细胞向神经细胞方向的诱导分化实验

万方数据

万方数据

华西口腔医学杂志第篮卷第4期2007年8月 ———— 一一里型!11些地婴坐堂皇!型些!臣!!!堑盟型些g:!婴!:塑!: 物经琼脂糖凝胶电泳分折,可见一条约380bp大小 的扩增产物条带,与GFAP预期片段大小基本一致。 内参对照GAPDH也可见900bp的清晰条带(图4)。 2.2免疫细胞化学染色1+Ma妇2:G8A9和诱导细胞,3:cFA’耳口束诱导细胞4: 诱导细胞抗黑∑世氍e鲞掣肌岬:、”冒:I鬻翟:詈:黧鬻:…。,。。呈 3),对照组这两种抗体染色均呈阴性。一 阳性表选 囤2坤经诱导4d盾牙髓干细胞抗GFAP的阳性表远ABc×200 ,培2h椰ve8叩哪l…m曲tlbod)∞cFAP缸deⅡklFLIlpst…eⅡsⅡeated诵thI)叫口ljnducbverr?刚iumRⅡ4dHYs ABCH200 幽3砷经诱导4d后,牙髓T细胞抗N虬的|5H性表述ABc×200 F咱3Pogid…砷…m们【ha埘bodyt0NsE0fden词pU如“Le…dkh即L出wJⅡIJ?em“1ndlmd…l。出Ilrt_h4davs ABC×Ⅻ 2_31w—Pcl{检测 咀未诱导细胞为对照,诱导细胞的RT—PcR产3讨论 成体干细胞的横向分化潜能或称为可塑性是指其不仅能分化为来源组织的各种细胞类型,而且具有分化为其他胚层来源的成熟细胞的潜能H。自从1997年有关成体干细胞可塑性的系列研究=l挂道后,可塑性研究引起生命科学领域的极大关注,对干细胞的研究产生重要影响。而这其中研究最多、可塑性最强的是骨髓基质干细胞(boncmcsenchymalstemcells-BMscs),可以诱导分化为骨髂肌细胞、心肌细胞、内皮细胞、肝细胞、胆管上皮细胞、肺上皮细胞、肠上皮细胞、皮肤上皮细胞、神经细胞、脂肪细胞等,其中最引八注目的是其神经方向的分化潜能。 Editis等”研究表明小胶质细胞、星形胶质细胞是来源于骨髓的一种前体细胞。而Brazelton等‘啦过绿色荧光蛋白基因转染示踪研究发现,在脑内微环境下,BMscs能分化为神经元样细胞、星形胶质样细胞,除表达相应神经细胞表型特点外,尚有神经元样功能。而在体外,削DMsO、BHA、B—ME等作为主要诱导剂,可诱导成年大鼠和人的BMSCs分化为神经元和神经胶质细胞,诱导后细胞在形态上出现类似于神经元样突起,表达NsE、nestin等神经细胞特异性标志㈣。此外,表皮生长|盘|子或脑源性神经生长因子与维甲酸或胎鼠的中脑、纹状体的悬液也可体外诱导BMscs分化为幼稚的神经兀和神经胶 质细胞口。可见体外不同培养条件下都可诱导BMscs 万方数据

神经干细胞体外诱导分化为胆碱能神经元的研究进展.

神经干细胞体外诱导分化为胆碱能神经元的研究进展 硕士研究生孙晓静导师孙莉 传统的观点认为,“中枢神经细胞不可再生”,然而自1992年Reynolds等从成年鼠脑纹状体和海马中首次分离出了神经干细胞(NSC),且它与多能干细胞一样具有自我更新、分裂增殖、多向分化的潜能,此后人们开始深入了解神经干细胞。Erikson等于1998年也证实了人脑中同样存在神经干细胞,随后的研究发现成年脑中神经干细胞主要存在于侧脑室外侧壁脑室下区和海马齿状回中。随着近年来对中枢神经系统的损伤或病变部位实行细胞替代或移植治疗的研究深入,为中枢神经系统损伤、神经系统变性疾病或神经退行性疾病等的治疗提供了新的方向。 阿尔茨海默病(AD)是老年人常见的一种慢性进行性神经系统变性疾病。长期以来认为前脑胆碱能投射系统(尤其是Meynert基底核)胆碱能神经元变性、细胞数量减少等导致AD认知功能不可逆的减退。目前就其病因有多种学说:如基因学说(APP基因[ 1 ]、早老蛋白基因[ 2 ] 、ApoE基因[ 3 ] )、胆碱能假说、雌激素水平下降学说[4] 、氧化应激学说[ 5 ] 、铝中毒学说、炎症学说[6 ] 、神经营养因子学说[7 ] 等。此外,AD还可能与年龄、种族、社会心理因素及各种疾病史等多种因素有关。 综上所述, 多因素参与AD 发病, 但目前为止确切的发病机制尚不清楚。其治疗方法包括对于轻度和中度AD 予以乙酰胆碱酯酶(AChE)抑制剂以提高认知功能, 以及N-甲基-D-天冬氨酸(NMDA)拮

抗剂对重度AD 的治疗[8],其他药物如脑循环改善药物剂、AChE 抑制剂、M1 胆碱受体激动剂、乙酰胆碱释放促进剂、神经生长因子、抗氧化药物和抗β-淀粉样药物等。这些治疗手段虽能减轻AD 的症状, 但无法补充大脑皮层和海马大量丢失的神经细胞,因而对中、晚期AD 患者的疗效有限。神经干细胞(NSCs)的广泛研究及重大进展为AD 的治疗提供了一个更有前景的治疗策略,NSC移植治疗AD等退行性疾病成为近年来的研究热点。然而不管是诱导、促进内源性NSCs增殖分化,产生相应的神经细胞代替缺损变性的细胞,还是体外培养需要的NSCs移植入体内,如何诱导NSCs定向分化为胆碱能神经元都是一个至关重要的问题。 神经干细胞增殖、分化相关因素 神经干细胞的增殖、迁移、分化与多种因素相关。目前的研究认为决定神经干细胞定向分化的机制有两种:一种是细胞自身基因调控;另一种是外来因素调控,主要为各类细胞因子家族,如神经营养因子、生长因子、细胞黏附因子等均可影响神经细胞的分化,其影响机制各不相同。不同的细胞因子诱导分化出的神经元不同。研究最多有生长因子如:表皮生长因子(EGF)家族、碱性成纤维细胞生长因子(FGF)家族、β-转导生长因子(β-TGF)超家族、神经营养因子、脑源性生长因子及化学诱导剂及药物等。 一、生长因子 碱性成纤维生长因子(FGF)在神经干细胞增殖的早期阶段发挥促有丝分裂的作用, 使神经干组胞获得对另一作用更强的促有丝分裂

神经母细胞瘤

中文名:神经母细胞瘤 英文名:neuroblastoma 别名:成神经细胞瘤 目录 1概述 2流行病学 3病因 4实验室检查 5其它辅助检查 6临床表现 展开 目录 1概述 2流行病学 3病因 4实验室检查 5其它辅助检查 6临床表现 7并发症 8诊断 9治疗 10预后 概述 神经母细胞瘤(neuroblastomaNB)从原始神经嵴细胞演化而来,交感神经链、肾上腺髓质是最常见的原发部位不同年龄、肿瘤发生部位及不同的组织分化程度使其生物特性及临床表现有很大差异部分可自然消退或转化 成良性肿瘤,但另一部分病人却又十分难治,预后不良鶒。在过去的30 年中,婴儿型或早期NB预后有了明显的改善,但大年龄晚期病人预后仍 然十分恶劣在NB中有许多因素可影响预后,年龄和分期仍然是最重要的 因素健康搜索。 流行病学 NB是儿童最常见的颅外实体瘤,占所有儿童肿瘤的8%~10%,一些高发 地区如法国、以色列瑞士、新西兰等的年发病率达11/100万(0~15岁),美国为25/100万,中国和印度的报道低于5/100万。

病因 属胚胎性肿瘤,多位于大脑半球。 发病机制: NB来自起源于神经嵴的原始多能交感神经细胞,形态为蓝色小圆细胞。从神经嵴移行后细胞的分化程度、类型及移行部位形成不同的交感神经系统正常组织,包括脊髓交感神经节、肾上腺嗜铬细胞健康搜索。 NB组织学亚型与交感神经系统的正常分化模型相一致。经典的病理分类 将NB分成3型即神经母细胞瘤神经节母细胞瘤、神经节细胞瘤,这3个 类型反应了NB的分化、成熟过程。典型的NB由一致的小细胞组成,约 15%~50%的病例,母细胞周围有嗜酸性神经纤维网。 另一种完全分化的、良性NB为神经节细胞瘤,由成熟的节细胞神经纤维 网及Schwann细胞组成神经节母细胞瘤介于前两者之间含有神经母细胞 和节细胞混杂成分。 Shimada分类结合年龄将病理分成4个亚型,临床分成2组。4个亚型即 包括NB(Schwannin少基质型);GNB混合型(基质丰富型);GN成熟型和(3NB 结节型(包括少基质型和基质丰富型)。前3型代表了NB的成熟过程,而 最后一型则为多克隆性。对NB而言,细胞分化分为3级,包括未分化、 分化不良分化型;细胞的有丝分裂指数(MKI)也分为低、中、高3级。 Shimada分类综合肿瘤细胞的分化程度、有丝分裂指数和年龄将NB分为 临床预后良好组(FH)和预后不良组(UFH): 1.FH包括以下各类 1)NB,MKI为低中度,年龄<1.5岁 (2)分化型NB,MKI为低度,年龄1.5~5岁。 (3)GNB混合型。 (4)GN。 2.UFH包括 (1)NBMKI高级。 (2)NBMKI为中级年龄1.5~5岁 (3)未分化或分化健康搜索不良型NB,年龄1.5~5岁。 (4)所有>5岁的NB。 (5)GNB结节型。在病理上除HE染色外,可进一步做免疫组化电镜检查来 与其他小圆细胞肿瘤相鉴别,NB时神经特异性酯酶(NSE)阳性电镜下可见

牙髓干细胞向神经细胞方向的诱导分化实验

牙髓干细胞向神经细胞方向的诱导分化实验 贺慧霞;金岩;史俊南;罗玉庆;周艳妮;彭智;许玉和 【期刊名称】《华西口腔医学杂志》 【年(卷),期】2007(025)004 【摘要】目的探讨克隆化培养分离的人牙髓干细胞是否具有向神经细胞方向分化的潜能,并确定其诱导条件.方法克隆化培养的人牙髓干细胞预诱导24 h,然后换含一定浓度二甲基亚砜(DMSO)、丁羟基茴香醚(BHA)、forskolin、β-巯基乙醇(β-ME)和氢化可的松(hydrocortisone)的联合诱导液连续诱导4 d,对诱导细胞进行形态学观察,神经胶质酸性蛋白(GFAP)、非特异性酯酶(NSE)免疫组化染色和GFAP mRNA RT-PCR检测.同时,以未诱导细胞为对照.结果诱导12 h 时细胞形态开始改变,24 h时分化为较为典型的神经细胞样细胞,继续诱导分化细胞数量增多;诱导细胞表达神经元细胞特异性标志NSE和GFAP蛋白;RT-PCR 检测诱导细胞表达GFAP mRNA,而未诱导细胞均无上述改变和表达.结论人牙髓干细胞在一定的诱导条件下可横向分化为神经元样细胞. 【总页数】4页(331-334) 【关键词】牙髓干细胞;诱导;分化 【作者】贺慧霞;金岩;史俊南;罗玉庆;周艳妮;彭智;许玉和 【作者单位】第四军医大学口腔医院病理科,组织工程中心,陕西,西安,710032;武警甘肃总队医院,口腔科,甘肃,兰州,730050;第四军医大学口腔医院病理科,组织工程中心,陕西,西安,710032;第四军医大学口腔医院,牙体牙髓科,陕西,西安,710032;兰州军区临潼疗养院三区,门诊部,陕西,西安,710600;武警甘肃总队医院,口腔科,甘肃,兰州,730050;武警甘肃总队医院,口腔科,甘肃,兰州,730050;武

节细胞神经母细胞瘤

神经母细胞瘤 2005-12-11 23:10:52 作者:superneo 点击数:2241 神经母细胞瘤与神经节细胞瘤(neuroblastoma and ganglioneuroma)同属于交感神经系统肿瘤。神经母细胞瘤(成神经母细胞瘤)系由未分化的交感神经细胞所组成,具有高度恶性,虽然近年来已研究了“多种肿瘤生物化学诊断方法,对影响预后因素的认识亦有很大的提高,20余年来在治疗上虽有进展,但死亡率仍较高,主要原因是诊断过晚。神经节细胞瘤是由成熟细胞组成的良性肿瘤。 一、发病率 神经母细胞瘤据Gersos等报道10000出生活婴中有1例。神经母细胞瘤根据美国小儿死亡率统计,在出生到4岁间为1/10万,在5到9岁问为0.4/10万。年龄分布可见本肿瘤多见于婴儿,年龄最小者43日,50%在3岁前,其余50%亦多在5岁前,10岁后根罕见。男性多于女性。神经节细胞瘤多发生在儿童期或青年期。 曾有报道兄弟问及连续两代同患神经母细胞瘤者,但染色体核型研究并末证明有遗传性。神经母细胞瘤也观察到在某些疾病患者中多见,如神经纤维瘤(neurofibromatosis)及Becwith—Wiedemann综合征。神经母细胞瘤细胞基因研究曾证实几乎80%病例有异常,最常见的异常是染色体短臂缺失或再排列,这种情况并非是神经母细胞瘤所特有的。其次,最多见的改变部位是染色质。 病源 在胚胎早期,原始神经嵴产生交感神经元细胞(sympathogonia),后者移行到各部位而形成神经母细胞和肾上腺髓质的嗜铬母细胞,以后成熟为正常的交感神经节和肾上腺髓质。根据细胞的分化情况可形成正常组织或肿瘤。 凡是有交感神经元细胞的部位都可发生神经母细胞瘤,如颅内、眼眶内和颈后侧部,但均较少见,常见部位为胸脊柱旁,尤其是在腹膜后,偶尔亦发生于盆腔。病理 (一)神经母细胞瘤 1.大体检查神经母细胞瘤肉眼观之,在早期尚规则,随后发展为多节结,质地较硬,色泽灰紫,带有许多出血坏死区,甚至呈假囊肿状,组织脆弱,极易破裂。

细胞凋亡实验步骤及注意事 项

细胞凋亡实验步骤及注意事项 一、实验目的 1、掌屋凋亡细胞的形态特征 2、学会用荧光探针对细胞进行双标记来检测正常活细胞、凋亡细胞和坏死细胞的方法 二、实验原理 细胞死亡根据其性质、起源及生物学意义区分为凋亡和坏死两种不同类型。凋亡普遍存在于生命界,在生物个体和生存中起着非常重要的作用。它是细胞在一定生理条件下一系列顺序发生事件的组合,是细胞遵循一定规律自己结束生命的自主控制过程。细胞凋亡具有可鉴别的形态学和生物化学特征。 在形态上可见凋亡细胞与周围细胞脱离接触,细胞变园,细胞膜向内皱缩、胞浆浓缩、内质网扩张、细胞核固缩破裂呈团块状或新月状分布、内质网和细胞膜进一步融合将细胞分成多个完整包裹的凋亡小体,凋亡小体最后被吞噬细胞吞噬消化。在凋亡过程中细胞内容物并不释放到细胞外,不会影响其它细胞,因而不引起炎症反应。 在生物化学上,多数细胞凋亡的过程中,内源性核酸内切酶活化,活性增加。核DNA随机地在核小体的连接部位被酶切断,降解为180-200bp 或它的整倍数的各种片断。如果对核DNA进行琼脂糖电泳,可显示以180-200bp为基数的DNA ladder(梯状带纹)的特征。 相比之下,坏死是细胞处于剧烈损伤条件下发生的细胞死亡。细胞在坏死早期即丧失质膜完整性,各种细胞器膨胀,进而质膜崩解释放出其中的内容物,引起炎症反应,坏死过程中细胞核DNA虽也降解,但由于存在各种长度不等的DNA片断,不能形成梯状带纹,而呈弥散状。 一些温和的损伤刺激及一些抗肿瘤药物可诱导细胞凋亡,通常这些因素在诱导凋亡的同时,也可产生细胞坏死,这取决于损伤的剧烈程度和细胞本身对刺激的敏感程度。 三尖杉酯碱(HT)是我国自行研制的一种对急性粒细胞白血病,急性单核白血病等有良好疗效的抗肿瘤药物。研究表明HT在0.02~5μg/ml范围内作用2小时,即可诱导HL-60细胞凋亡,并表现出典型的凋亡特征。本实验用1μg/ml HT在体外诱导培养的HL-60细胞发生凋亡,同时也有少数细胞发生坏死。用Hoechst33342和碘化丙啶(propidium iodide,PI)对细胞进行双重染色,可以区别凋亡、坏死及正常细胞。细胞膜是一选择性的生物膜,一般的生物染料如PI等不能穿过质膜。当细胞坏死时,质膜不完整,PI就进入细胞内部,它可嵌入到DNA或RNA 中,使坏死细胞着色,凋亡细胞和活细胞不着色。而一些活细胞染料由于为亲脂性物质,可跨膜进入活细胞,因而可进行活细胞染色。

神经干细胞研究介绍

神经干细胞研究介绍 陈晓萍 程君奇 (浙江大学生命科学学院浙江省细胞与基因工程重点实验室浙江杭州310029) 摘要 神经干细胞研究是近年脑科学研究的热点,本文综述了神经干细胞的分离培养方法、脑内迁移路线、发育分化的影响因素以及可能的应用前景。 关键词 神经干细胞 分离 迁移 发育 分化 脑的结构与机能一直是生命科学的研究难题,它以极其错综复杂而又高度易变为特征,至今仍保持着极大的神秘性。近年来科学家对神经干细胞的研究是脑科学领域的重要成果之一,它突破了以往一直认为的成年动物神经细胞不能分裂再生的观念,为神经细胞的发育分化过程,也为神经系统疾病的治疗开辟了一条全新的途径。 1 神经干细胞的分离培养 神经干细胞(N eural stem cells,N SCS)是指具有如下特征的细胞:1)能形成神经组织;2)具有自我繁殖和自我更新能力;3)细胞分裂后能发生分化[1]。 胚胎时期是神经系统快速增长发育的阶段,在这时期脑内的许多部位都存在神经干细胞,这包括大脑皮质、纹状体、小脑等区域。成年后,脑细胞一般不再分裂增殖。以往曾一度认为成年动物神经细胞完全失去了这种能力,但近年科学家在高等哺乳动物(包括人)的脑室管膜下层等区域发现了仍具有增殖分化能力的神经干细胞。另外,在啮齿类动物主管学习记忆的海马区,也发现了神经干细胞的存在[2]。 成年动物脑内的神经干细胞仅仅是保存了能进行分裂增殖的潜能,通常情况下得不到足够的正面刺激信号,因而并不分裂增殖,而是处于静止状态。 从脑组织分离培养神经干细胞需要特殊的条件,目前多采用生长因子刺激和细胞克隆技术。具体有3种方法[3]:1)用无血清培养液将脑细胞分离,再加入具有丝裂原作用的生长因子如表皮生长因子或碱性成纤维生长因子,待原代克隆形成后挑选单个克隆机械分离继续进行亚克隆培养,也可采用单细胞克隆分离;2)用反转录病毒向脑细胞内导入原癌基因,如V2m yc和SV40大T抗原等,部分细胞可因此获得持续分裂的能力;3)从脑组织以外的部位,如胚胎干细胞,经过适当化学因子的诱导,使其定向分化为神经干细胞。 外加化学因子对于维持神经干细胞的分裂增殖能力是必须的。培养液中如撤去外加的化学因子,改用普通培养,神经细胞会很快发生分化,失去分裂增殖能力。 2 神经细胞的发育及脑内迁移路径 神经系统的发育源于胚胎早期的神经管和神经嵴[4],其中的中央管经发育形成脑室系统和脊髓中央管,管腔内表面覆盖的上皮细胞具有活跃的增殖和分化能力,是神经细胞发生的来源。成年后这个区域称为室管膜 室管膜下区。 内径1~2mm的完全闭合的呼吸管。据B landfo rd报道,这种呼吸管中衬有外套膜组织。上述蜗牛的夏眠能从1月持续至6月。同时具有裂口、裂沟和缝合线管的结构有利于气体的循环。在足部和外套膜肌肉运动下,可促使气体交流和循环,贝类学家F ischer曾对冬眠期间的盖罩大蜗牛进行了研究,业已证明其足部和外套膜从未停止过运动。 8)喇叭状口和壳壁上的穿孔 A lycaeinae的种类,其成体的壳口呈喇叭状,其后逐渐缩小成一口颈。在口颈近缝合线的壳壁上有穿孔。A ly caeus属的种类,如A2 ly caeus m ajor壳壁上的孔由内向外通入一覆盖在缝合线上的管。管的截面略呈三角形。此管在缝合线上,略弯曲,呈带状,长约6~7mm,管的末端是盲端,但常破碎,即使不破碎,该管仍可进行气体交换。据分析这个带状结构比通常的贝壳更具通透性。因种类不同,喇叭口的长度有差异,缝合线管内开口到口缘距离也有所不同。喇叭状的壳口可能是为了蜗牛在夏眠期间有一个较大的气室,这与无厣贝类具有的盖膜腔情况相似。由此可以推断A ly caeus和Pup ininae的一些种类缝合线管在内部的开口可能与肺呼吸孔靠得很近。 其他的一些陆生螺类,如R um ina d ecollata和C lausilia的一些种类,在夏眠期间往往胚螺层失去,然而可能也有利于呼吸。破损的一端由内脏分泌的膜封住,这比休眠时螺壳倒下,靠外套膜边缘呼吸更利于气体交换。 (BF) — 8 1 —生 物 学 通 报 2003年第38卷第2期

骨髓基质干细胞诱导分化为神经细胞的研究进展

骨髓基质干细胞诱导分化为神经细胞的研究进展骨髓基质干细胞具有取材简单、增殖速度快、培养过程中始终保持多向分 化的潜能等特点,已经成为干细胞研究领域的热点,是最好的组织工程种子细胞之一,还可以诱导分化为神经细胞。本文对骨髓基质干细胞诱导分化为神经细胞的研究进展作一综述。 [Abstract] Bone marrow stromal cells has derived simple,fast growth,the training process remains much to differentiate and other characteristics,has been become the hot spot in the study of stem cells,is one of the best seed cells of tissue engineering.It can also differentiate into neural cells.The article reviewes the progress of induction of bone marrow stromal cells to neural cells and its mechanisms. [Key words] Bone marrow stromal cells;Neural cells;Bone marrow 骨髓基质干细胞(bone marrow stromal cells,BMSCs)位于骨髓中,是具有自我复制和多向分化潜能的非造血干细胞[1]。由于BMSCs具有取材容易、培养简单、增殖能力强、可以传代并且不改变生物学特性以及没有伦理学及免疫排斥等优点而备受瞩目[2-3],BMSCs为治疗中枢神经系统疾病的一种理想供体细胞[4-6]。本文将BMSCs诱导分化为神经细胞的研究现状作一综述。 1 BMSCs定向分化为神经细胞的研究现状 1.1 体内定向分化 大量研究表明,BMSCs在体内可以分化为神经细胞。有学者将来源于人的BMSCs注入小鼠纹状体内,5~72 d后发现,在脑组织切片上有供体细胞存在,同时无明显炎症反应和排斥反应[7]。Mezey等[8]进行了雄性和雌性小鼠BMSCs 移植实验,将雄性小鼠的BMSCs植入雌性小鼠体内,一段时间后进行检测发现有携带Y染色体的神经细胞出现在雌性小鼠体内,提示雄性小鼠的BMSCs在雌性小鼠的体内分化成神经细胞。Guillermo等[9]将BrdU标记的大鼠BMSCs注入胎龄为15 d的大鼠胚胎的侧脑室内,在胎龄18、20 d时检测发现移植的细胞形成细胞团,2个月时检测仍有少量的移植细胞存活,表明BMSCs可以在脑的微环境中分化为神经细胞并存活较长时间。Chen等[10]将BMSCs应用于脑外伤动物,发现其能分化为神经细胞,并对神经功能的恢复有明显的促进作用。上述实验表明,BMSCs在体内可以迁移、存活、在相应部位的微环境影响下可分化为神经细胞,并能发挥一定的神经修复功能。 1.2 体外定向分化 能在体外分化为有较完整功能的神经细胞,是BMSCs作为神经细胞移植的来源细胞应用于临床的基础,很多学者在如何诱导BMSCs分化为神经细胞方面做了大量有益的探索,取得了较大进展,截止目前,诱导方法大致可归纳为以下

相关文档