efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells

5
NATURE BIOTECHNOLOGY VOL 18 JUNE 2000 http://biotech.nature.com 675 TECHNICAL REPORTS Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells Sang-Hun Lee 1† , Nadya Lumelsky 1† , Lorenz Studer 1,2 , Jonathan M. Auerbach 1 , and Ron D. McKay 1 * 1 Laboratory of Molecular Biology, NINDS, NIH, Bethesda, MD 20892. 2 Current address: Laboratory of Stem Cell and Tumor Biology, Neurosurgery and Cellular Biochemistry and Biophysics, Memorial Sloan Kettering Cancer Center, New York, NY, 10021. Both authors contributed equally to this work. *Corresponding author ([email protected]). Received 15 April, 2000; accepted 24 April, 2000 Embryonic stem (ES) cells are clonal cell lines derived from the inner cell mass of the developing blastocyst that can proliferate extensively in vitro and are capable of adopting all the cell fates in a developing embryo. Clinical interest in the use of ES cells has been stimulated by studies showing that isolated human cells with ES properties from the inner cell mass 1, 2 or developing germ cells 3 can provide a source of somatic precursors. Previous studies have defined in vitro conditions for promoting the development of spe- cific somatic fates, specifically, hematopoietic, mesodermal, and neurectodermal 4–7 . In this study, we present a method for obtain- ing dopaminergic (DA) and serotonergic neurons in high yield from mouse ES cells in vitro. Furthermore, we demonstrate that the ES cells can be obtained in unlimited numbers and that these neuron types are generated efficiently. We generated CNS progeni- tor populations from ES cells, expanded these cells and promoted their differentation into dopaminergic and serotonergic neurons in the presence of mitogen and specific signaling molecules. The differentation and maturation of neuronal cells was completed after mitogen withdrawal from the growth medium. This experi- mental system provides a powerful tool for analyzing the molecu- lar mechanisms controlling the functions of these neu- rons in vitro and in vivo, and potentially for under- standing and treating neurodegenerative and psychi- atric diseases. To understand the developmental transitions of stem cells and fulfill their clinical promise, it must be possible to control the differentiation of ES cells into specific cell fates. Neurons, astrocytes, and oligodendrocytes have been derived from ES cells 7–10 . The neurotransmitters, dopamine and serotonin, have important functions in the etiology and treatment of neurodegenerative 11 and psychiatric diseases 12–15 . The neurons that synthesize these transmitters are generated by similar developmen- tal signals around the boundary between the midbrain and the hindbrain. To assess the developmental progression of ES cells in vitro, we examined the appearance of CNS- and mid- brain-specific gene products in a modification of our previous work where CNS stem cells were derived from ES cells 10 . The differentiation involves several steps: the generation of embryoid bodies (EBs; stage 2), the use of a defined medium to select for CNS stem cells (stage 3), the proliferation of CNS stem cells in the presence of mitogen, basic fibroblast growth factor (bFGF; stage 4), and the differentiation of the stem cells by removal of the mitogen (stage 5; Fig. 1A). The successful conversion of ES cells into midbrain dopamin- ergic (DA) neurons in vitro should be dependent on the induction of the same genes that are expressed in CNS stem cells and neurons in vivo. The OTX homeobox genes (OTX1 and OTX2) are widely expressed at early stages of neurectoderm differentiation 16 . OTX2 is expressed throughout the epiblast and subsequently restricted to anterior neurectoderm, where it is required for development of the forebrain and midbrain. The homolog OTX1 is first expressed in the neurectoderm in the dorsal telencephalon, and interactions between these two OTX genes are thought to specify the develop- ment of the midbrain and hindbrain 17 . OTX2 is expressed in the undifferentiated (stage 1) ES cells and present at lower levels at stage 2 and 3 of differentiation. OTX1 is not expressed at high lev- els until stage 3 (Fig. 1B). Several genes (Pax2, Pax 5, Wnt1, En1, Nurr1) have been identi- fied that control differentiation of dopaminergic and serotonergic neurons in the midbrain and hindbrain 18,19 . PCR analysis detected expression of these genes at stages 3 and 4 of ES cell differentiation. After stage 4, the bFGF-expanded cells are morphologically uniform and express nestin, an intermediate filament protein characteristic of CNS stem cells (Fig. 2A) 20,21 . These results suggest that ES cells show a progressive restriction to mesencephalic and metencephalic CNS stem cells. Identification of neurons synthesizing neurotransmitters expected for midbrain and hindbrain fates would lend support to this conclusion. Tyrosine hydroxylase (TH) is the rate-limiting enzyme for the biosynthesis of dopamine and a marker of ventral midbrain neurons. No TH expression was detected at stages 1–4 of ES cell culture (data not shown). However, the stage 4 cells may be primed for induction of TH, but still require time and appropriate conditions to express differentiated neuronal features. Differentiation (stage 5) was promoted by using conditions known to promote neuronal differentiation from the proliferating precur- sor state 22–24 . TuJ1, an antibody directed against the neuron-specific β-III tubulin bound many cells with a clear neuronal morphology (Fig 2B). Of the total cell population, 71.9 ± 6.9% were TUJ1 + , and 6.9% ± 1.5% of the TUJ1 + cells were also TH + (these numbers are the average ± s.e.m. of three independent experiments, n > 40 microscopic fields, for a total containing 15.7 × 10 3 neurons). The Figure 1. ES cells progressively differentiate into mesencephalic stem cells. (A) General scheme of ES cell culture. (B) Differentiated ES cells express genes characteristic of midbrain fates. Semi-quantitative RT-PCR analysis of the early CNS, mesencephalic, and midbrain-specific regulatory genes at different stages of ES cell culture. Numbers at the top of the panel designate stages of culture defined in (A). Generation of DA neuronal populations from undifferentiated ES cells Expand undifferentiated ES cells population on gelatin-coated tissue culture surface in ES cell medium in the presence of LIF (Stage 1) Generate EBs in suspension cultures for 4 days in ES cell medium (Stage 2) Select nestin -positive cells for 8 days in ITSFn medium from EBs plated on tissue culture surface (Stage 3) Expand nestin -positive cells for 6 days in N2 medium containing bFGF/laminin (Stage 4) Induce differentiation of the expanded neuronal precursor cells by withdrawing bFGF from N2 medium containing laminin (Stage 5) ß-actin Pax2 Nurr1 Nestin Pax5 Wnt1 En1 Midbrain DA neuronal-specific mesencephalic 1 2 3 4 Otx1 Otx2 Early CNS A B © 2000 Nature America Inc. • http://biotech.nature.com © 2000 Nature America Inc. • http://biotech.nature.com

Upload: ron-d

Post on 21-Jul-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells

NATURE BIOTECHNOLOGY VOL 18 JUNE 2000 http://biotech.nature.com 675

TECHNICAL REPORTS

Efficient generation ofmidbrain and hindbrainneurons from mouseembryonic stem cellsSang-Hun Lee1†, Nadya Lumelsky1†, Lorenz Studer1,2, Jonathan M. Auerbach1, and Ron D. McKay1*

1Laboratory of Molecular Biology, NINDS, NIH, Bethesda, MD 20892.2Current address: Laboratory of Stem Cell and Tumor Biology, Neurosurgeryand Cellular Biochemistry and Biophysics, Memorial Sloan Kettering CancerCenter, New York, NY, 10021. † Both authors contributed equally to this work.*Corresponding author ([email protected]).

Received 15 April, 2000; accepted 24 April, 2000

Embryonic stem (ES) cells are clonal cell lines derived from theinner cell mass of the developing blastocyst that can proliferateextensively in vitro and are capable of adopting all the cell fates in adeveloping embryo. Clinical interest in the use of ES cells has beenstimulated by studies showing that isolated human cells with ESproperties from the inner cell mass1, 2 or developing germ cells3 canprovide a source of somatic precursors. Previous studies havedefined in vitro conditions for promoting the development of spe-cific somatic fates, specifically, hematopoietic, mesodermal, andneurectodermal4–7. In this study, we present a method for obtain-ing dopaminergic (DA) and serotonergic neurons in high yieldfrom mouse ES cells in vitro. Furthermore, we demonstrate thatthe ES cells can be obtained in unlimited numbers and that theseneuron types are generated efficiently. We generated CNS progeni-tor populations from ES cells, expanded these cells and promotedtheir differentation into dopaminergic and serotonergic neuronsin the presence of mitogen and specific signaling molecules. Thedifferentation and maturation of neuronal cells was completedafter mitogen withdrawal from the growth medium. This experi-mental system provides a powerful tool for analyzing the molecu-lar mechanisms controlling the functions of these neu-rons in vitro and in vivo, and potentially for under-standing and treating neurodegenerative and psychi-atric diseases.

To understand the developmental transitions of stemcells and fulfill their clinical promise, it must be possibleto control the differentiation of ES cells into specific cellfates. Neurons, astrocytes, and oligodendrocytes havebeen derived from ES cells7–10. The neurotransmitters,dopamine and serotonin, have important functions inthe etiology and treatment of neurodegenerative11 andpsychiatric diseases12–15. The neurons that synthesizethese transmitters are generated by similar developmen-tal signals around the boundary between the midbrainand the hindbrain.

To assess the developmental progression of ES cells invitro, we examined the appearance of CNS- and mid-brain-specific gene products in a modification of ourprevious work where CNS stem cells were derived fromES cells10. The differentiation involves several steps: the

generation of embryoid bodies (EBs; stage 2), the use of a definedmedium to select for CNS stem cells (stage 3), the proliferation ofCNS stem cells in the presence of mitogen, basic fibroblast growthfactor (bFGF; stage 4), and the differentiation of the stem cells byremoval of the mitogen (stage 5; Fig. 1A).

The successful conversion of ES cells into midbrain dopamin-ergic (DA) neurons in vitro should be dependent on the inductionof the same genes that are expressed in CNS stem cells and neuronsin vivo. The OTX homeobox genes (OTX1 and OTX2) are widelyexpressed at early stages of neurectoderm differentiation16. OTX2is expressed throughout the epiblast and subsequently restricted toanterior neurectoderm, where it is required for development of theforebrain and midbrain. The homolog OTX1 is first expressed inthe neurectoderm in the dorsal telencephalon, and interactionsbetween these two OTX genes are thought to specify the develop-ment of the midbrain and hindbrain17. OTX2 is expressed in theundifferentiated (stage 1) ES cells and present at lower levels atstage 2 and 3 of differentiation. OTX1 is not expressed at high lev-els until stage 3 (Fig. 1B).

Several genes (Pax2, Pax 5, Wnt1, En1, Nurr1) have been identi-fied that control differentiation of dopaminergic and serotonergicneurons in the midbrain and hindbrain18,19. PCR analysis detectedexpression of these genes at stages 3 and 4 of ES cell differentiation.After stage 4, the bFGF-expanded cells are morphologically uniformand express nestin, an intermediate filament protein characteristicof CNS stem cells (Fig. 2A)20,21. These results suggest that ES cellsshow a progressive restriction to mesencephalic and metencephalicCNS stem cells.

Identification of neurons synthesizing neurotransmittersexpected for midbrain and hindbrain fates would lend support tothis conclusion. Tyrosine hydroxylase (TH) is the rate-limitingenzyme for the biosynthesis of dopamine and a marker of ventralmidbrain neurons. No TH expression was detected at stages 1–4 ofES cell culture (data not shown). However, the stage 4 cells may beprimed for induction of TH, but still require time and appropriateconditions to express differentiated neuronal features.Differentiation (stage 5) was promoted by using conditions knownto promote neuronal differentiation from the proliferating precur-sor state22–24. TuJ1, an antibody directed against the neuron-specificβ-III tubulin bound many cells with a clear neuronal morphology(Fig 2B). Of the total cell population, 71.9 ± 6.9% were TUJ1+, and6.9% ± 1.5% of the TUJ1+ cells were also TH+ (these numbers arethe average ± s.e.m. of three independent experiments, n > 40microscopic fields, for a total containing 15.7 × 103 neurons). The

Figure 1. ES cells progressively differentiate into mesencephalic stem cells. (A)General scheme of ES cell culture. (B) Differentiated ES cells express genescharacteristic of midbrain fates. Semi-quantitative RT-PCR analysis of the early CNS,mesencephalic, and midbrain-specific regulatory genes at different stages of ES cellculture. Numbers at the top of the panel designate stages of culture defined in (A).

Generation of DA neuronal populations from undifferentiated ES cells

Expand undifferentiated ES cells population on gelatin-coatedtissue culture surface in ES cell medium in the presence of LIF

(Stage 1)

Generate EBs in suspension cultures for 4 days in ES cell medium(Stage 2)

Select nestin-positive cells for 8 days in ITSFn medium from EBsplated on tissue culture surface (Stage 3)

Expand nestin-positive cells for 6 days in N2 medium containingbFGF/laminin (Stage 4)

Induce differentiation of the expanded neuronal precursor cells

by withdrawing bFGF from N2 medium containing laminin (Stage 5)

ß-actin

Pax2

Nurr1

Nestin

Pax5

Wnt1

En1

Midbrain DAneuronal-specific

mesencephalic

1 2 3 4

Otx1

Otx2

Early CNS

A B

© 2000 Nature America Inc. • http://biotech.nature.com©

200

0 N

atu

re A

mer

ica

Inc.

• h

ttp

://b

iote

ch.n

atu

re.c

om

Page 2: Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells

neuronal differentiation is as efficient as has been previouslyachieved from primary CNS stem cells22,25. This result suggests thatthe ES-derived nestin-positive population is dominated by CNSstem cells and differentiates into midbrain neurons.

We found that the method of EB formation and the compositionof growth medium exert a clear effect on the yield of TH+ neurons.The significant percentage of TH+ neurons among the differentiatedES cell progeny contrasts with previous studies in which no appre-ciable numbers of TH+ cells were observed10. The current procedurewas achieved by two modifications that optimized neuronal differ-entiation and TH expression. In this work ES cells were first dissoci-ated into single cells to obtain a more uniform procedure for EB for-mation. As a result of this modification the yield of nestin-positivecells, as determined after stage 3, was increased by 3.5-fold. The sec-ond modification was to use medium without HEPES during stages4 and 5 of culture. HEPES inhibits differentiation of TH+ neurons(Fig. 2C). This modification of the culture conditions leads to theefficient production of TH+ neurons.

Sonic hedgehog (SHH), FGF8, and ascorbic acid (AA)increased the yield of ES-derived TH+ neurons. FGF8 and SHHhave been previously shown to promote ventral midbrain fates inneural plate explant26, therefore we hypothesized that the additionof these factors at specific stages during an in vitro ES cell dif-ferentation might also increase the proportion of DA and seroton-ergic neurons in our culture system. As shown in Figure 3A, theSHH receptors (smoothened, Smo, and patched, Ptc), a down-stream transcription factor (Gli1), the FGF8 receptor (FGFR3),and the ligands (SHH and FGF8) are expressed during ES cell dif-ferentiation. Combined treatment with SHH/FGF8 during stage 4leads to a greater than twofold increase in the number of TH+ cells(Fig. 3B; 15.4 ± 2.4% of the TuJ1+ neurons; value of three indepen-dent experiments, n > 40 fields containing 15.7 × 103 neurons, P <0.01). Application of SHH/FGF8 at earlier (stage 2 or 3) or later(stage 5) stages had no effect on the number of TH+ neurons (datanot shown). When applied as single factors at stage 4, SHH andFGF8 were both significantly less effective than when added incombination.

The cAMP analog, dibutyryl cAMP, and AA have been previ-ously implicated in promoting DA neuron yield from primaryCNS cultures27,28. Whereas dibutyryl cAMP was ineffective aloneor in combination with SHH and FGF8, treatment with AA duringstage 5 led to a significant increase in TH+ cell yield (Fig. 3B). Thepercentage of ES-derived TH+ neurons after treatment withSHH/FGF8 during stage 4 and AA during stage 5 reached 33.9 ±5.5% of the neurons (n > 40 fields containing 4.2 × 103 neurons)

which corresponds to morethan 20% of the total cellpopulation. Importantly, wefound that the population ofTH+ cells was maintainedwhen stage 4 cells treated withSHH and FGF8 were passagedbefore differentiation.

To further characterize theES-derived TH+-cell populationwe performed double immuno-histochemistry for TH and (1) dopamine-β–hydroxylase(DBH), a marker of noradren-ergic neurons (2) γ-aminobu-tyric acid (GABA), a marker forinhibitory neurons, and (3)serotonin, a transmitter foundin many ventral hindbrain neu-rons. No coexpression of TH

with any of these markers was observed, suggesting that the TH+ cellsare DA neurons (data not shown).

The production of dopamine is a definitive measure of the iden-tity of a dopaminergic neuron. Reverse-phase high-performanceliquid chromatography (RP-HPLC) was used to measuredopamine release directly (Fig. 3C). The dopamine peak can bereadily separated and the sensitivity of the procedure repeatedlyconfirmed with control samples. Neurons were differentiated instage 5 for six days, and medium conditioned for 48 h was tested.Consistent with the increase in the number of TH+ neurons, thedopamine level was increased more than twofold in cultures treat-ed with SHH/FGF8/AA (n = 3, P < 0.05). Elevated potassium wasused to determine if dopamine could be released by depolarization.When Hank’s balanced salt solution (HBSS) was applied to the cellsfor 15 min, 165.7 ± 23.4 pg ml-1 dopamine was detected.SHH/FGF8/AA did not change dopamine levels. However,dopamine levels were elevated when the neurons were depolarizedwith elevated potassium (56 mM), and the medium collected after15 min (416.6 ± 7.2 pg ml-1 in the controls and 918.4 ± 123.2 pg ml-

1 of dopamine in the SHH/FGF8/AA-treated cultures). These datademonstrate that ES-derived TH+ cells secrete significant levels ofdopamine when depolarized.

Electrophysiology was used to determine if ES-derived neuronswere functionally active. When current was injected into individualpatch-clamped neurons, sustained trains of action potentials, char-acteristic of mature neurons, were routinely observed (n = 54; Fig.4A). The cells also responded to direct application of the neurotrans-mitters GABA and glutamate (Fig. 4B, C). After 13 days of differenti-ation at stage 5, the neurons showed spontaneous activity that wasalmost completely blocked by tetrodotoxin (TTX), an inhibitor ofaction potentials (Fig. 4D, E). Biocytin labeling demonstrated thatneurons exhibiting TTX-sensitive spontaneous activity were TH+

(Fig. 4F, G). This demonstrates that TH+ cells receive synaptic inputand suggests that this system can be used to define synaptic controlof dopamine release.

In addition to specification of midbrain DA precursors, SHH isalso important for the generation of hindbrain serotonergic neu-rons26. As shown in Figure 5, in untreated cultures 0.8 ± 0.1% (n =56, fields containing 6.2 × 103 neurons) of all TuJ1+ neurons wereserotonin-positive. However, addition of SHH/FGF8 during stage 4increased the serotonergic population by 14-fold (11.0 ± 0.5%, n =56 fields containing 6.2 × 103 neurons, P < 0.01 of all TuJ1+ cells).Serotonin and TH were not coexpressed. Interestingly, application ofSHH alone promoted serotonergic fates to an extent similar to thecombined treatment (Fig. 5B). These results are in agreement with

676 NATURE BIOTECHNOLOGY VOL 18 JUNE 2000 http://biotech.nature.com

TECHNICAL REPORTS

Figure 2. Mesencephalic precursors differentiate into TH+ neurons. (A) ES cell progeny at stage 4 express nestin,an intermediate filament protein expressed in CNS stem cells but not in neurons and glia (scale bar = 20 µm). (B) ES cell progeny at stage 5 differentiate into TH+ neurons. Removal of the mitogen initiates differentiation ofthe nestin-positive cells into neurons recognized by the antibody TuJ1 that binds to a neuron-specific tubulin. Asubset of the neurons expresses the enzyme tyrosine hydroxylase (TH), which is required for the production ofcatecholamine neurotransmitters (scale bar = 20 µm). (C) The effect of HEPES buffer on differentiation of TH+

cells. N3FL is a medium used for ES culture10.

TuJ1 / TH Nestin

After expansion with bFGF(stage 4)

After differentiation by withdrawal of bFGF (stage 5)

bb c

0

5

10

15

20

25

30

TH

+ c

ells

/ w

ell (

x103

)

N2 N3FL N3FL- HEPES

+ NaHCO3

N2- NaHCO3+ HEPES

A B C

© 2000 Nature America Inc. • http://biotech.nature.com©

200

0 N

atu

re A

mer

ica

Inc.

• h

ttp

://b

iote

ch.n

atu

re.c

om

Page 3: Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells

NATURE BIOTECHNOLOGY VOL 18 JUNE 2000 http://biotech.nature.com 677

findings in neural plate explant cultures suggesting that SHH can actalone to specify the precursors for serotonergic neurons 26. The effi-cient induction of serotonergic neurons by SHH suggests that thesedifferentiation conditions support hindbrain serotonergic fates.

It has been shown that ES cells can differentiate into neurons andglia in vitro7,10 and in vivo8,9,29. However, no systematic derivation ofmidbrain and hindbrain neurons from ES cells has been reported. Herewe found that catecholaminergic and serotonergic neurons can be effi-ciently derived from ES cells. HEPES buffer addition during stage 4 andstage 5 was incompatible with DA differentiation of ES cells. At presentthe mechanism of this inhibitory effect is not known. SHH, FGF8, andAA also enhance the differentiation to dopaminergic and serotonergicfates. Less than 5% of the neurons express TH when mesencephaliccells are directly differentiated without expansion in the precursorstate30. When E12 rat mesencephalic precursors were first expanded inbFGF, 25% of the neurons were TH+ (ref. 24). This ES cell system givesaccess to different stages of development leading to >30% of TH+ neu-rons, the highest yield ever obtained either in vitro or in vivo.

The development of cell therapy is a potential advantage of thedirected differentiation of ES cells into DA neurons. Although laterprecursors such as CNS stem cells can also be expanded and differen-tiated into DA neurons, these have only a limited ability to expand invitro and to adopt a dopaminergic fate; moreover, these cells do notproduce more DA neurons in response to SHH and FGF8 treatment(our unpublished observation). The ES cells, on the other hand, pro-liferate without limit, respond to signaling molecules, and are easilyaccessible to genetic manipulation. We anticipate that the efficiencyof the system described here can be further improved by generatingES cell lines expressing regulatory genes such as Nurr1 known tocontrol commitment to DA fate in vivo31. We also expect that thepurification of a homogeneous midbrain population can beachieved by genetic methods. Although studies in Parkinsonianrodents are needed to further assess the function and safety of EScell-derived DA neurons in vivo, our data demonstrate that the neu-rons derived from ES cells produce dopamine, respond to neuro-transmitters, and exhibit spontaneous synaptic activity.

The central finding of this study is that midbrain and hindbrainneurons can be generated in unlimited numbers from ES cells. Forevery 3 × 106 of ES cells, we obtain 21 × 106 neurons and 7 × 106 DAneurons. Numerically, two to three TH+ cells are harvested at stage 5for every undifferentiated ES cell plated at stage 1. This representsthe sum total of all parameters of proliferation, cell death, and selec-tive differentiation throughout the five culture stages. There are 3 × 104 DA neurons in the rat substantia nigra, and 103 grafted DAcells lead to behavioral recovery in a rat model of Parkinson’s dis-ease25. Genetic manipulation may now be used to obtain pure popu-lations of the DA neuron suitable for grafting and other experimentsrelevant to the etiology and therapy of Parkinson’s disease.

The dopamine- and serotonin-synthesizing neurons are two ven-tral fates generated in vivo, anterior and posterior of the isthmic orga-nizer, a source of signaling molecules that control the differentiation

TECHNICAL REPORTS

Figure 3. The effect of external signals on the yield of neurons that are TH+ and secrete dopamine. (A) Expression of genes in the SHH and FGF8signal pathways. Semiquantitative RT-PCR analysis at different stages of cell culture. Numbers at top of the panel designate st ages of theculture; see Figure 1A. (B) The yield of TH+ neurons is expressed as a percentage of TuJ1+ neurons. SHH (500 ng ml-1), FGF8 (100 ng ml-1), cAMP(1 mM), and AA (200 µM) were added at different stages of ES cell development, as shown. (C) The effect of treatment with SHH, FGF8, and AAon maturation of DA neurons as measured by dopamine release. The RP-HPLC determination of dopamine concentration is shown in medium(N2) conditioned for 48 h (left), in HBSS conditioned for 15 min (center), and in HBSS + 56 mM KCl conditioned for 15 min (right).

Figure 4. Synaptic properties of ES-derived TH+ neurons. (A) Actionpotential spiking behavior. Following 13 days of differentiation (stage 5),depolarization causes the cell to fire an initial action potential followed bya few others at low frequency. With increasing amounts of depolarizationthe neuron will fire a train of action potentials at a higher frequency. Thisbehavior was common in the cultures and is characteristic of matureneurons. (B) GABA application to the dendrites of a neuron leads to aninward current. (C) Glutamate application to the dendrites of a neuronleads to an inward current. (D) Spontaneous activity in TH+ neuronsrecorded in voltage clamp mode. (E) Tetrodotoxin (TTX) blocksspontaneous activity in TH+ neurons. The activity in the same cell as shownin (D) was diminished when action potentials were blocked with 1 µM TTX.Note that the TTX blocks almost all spontaneous activity, indicating thatmost of the activity is synaptic release of transmitter evoked byspontaneous action potential firing in presynaptic neurons. (F, G) The cellrecorded in (D) and (E) is TH+. The cell tracer biocytin was introducedthrough the recording electrode, and confocal images are presentedshowing the colocalization of TH (F) and biocytin (G) (Scale bars = 20 µm).

F G

% T

H(+

)/T

uJ1(

+)

0

10

20

30

40

DuringDifferentiation

(Stage 5)

SHH

Smo

Ptc

Gli1

FGFR3

FGF8b

SHHsignaling

FGF8signaling

_-actin

1 2 3 4

During Expansion(Stage 4)

SHH&FGF8

SHH&FGF8- - -

SHH&FGF8

- AA-

SHH FGF8

SHH&FGF8

cAMP AA cAMP- -

-

DA

in S

uper

nata

nt (

pg/m

l)

200

400

600

800

1000

1200

0

48hr conditionmedium

15Õ HBSS 15Õ HBSS/KCl

No treatmentSHH+FGF8+AA

A B C

AB

C

D E

© 2000 Nature America Inc. • http://biotech.nature.com©

200

0 N

atu

re A

mer

ica

Inc.

• h

ttp

://b

iote

ch.n

atu

re.c

om

Page 4: Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells

midbrain and hindbrain patterning32. Our findings show that underthis protocol almost half of the neurons generated can adopt a ventralmid/hindbrain fate. This will facilitate understanding the mecha-nisms controlling early steps in the differentiation of these neuronsthat have previously been difficult to access. For example, at presentin vivo and explant systems are not well suited to define if SHH andFGF8 act through inductive or selective mechanisms, but clonalanalysis in the dissociated ES cells system will answer this question. Aswe demonstrate, this system is also well suited to analyze mechanismsof neuronal differentiation, function, and survival.

Experimental protocolMaintenance of undifferentiated ES cells (R1, E14.1, B5—all three lines of EScells behaved similarly), EB formation, and selection of nestin-positive cellswere carried out as described10 with modifications. Undifferentiated (stage 1)ES cells were grown on gelatin-coated tissue culture plates in the presence of1,400 U ml-1 of leukemia inhibitory factor (LIF; GIBCO/BRL, Grand Island,NY) in ES cell medium consisting of knockout Dulbecco’s minimal essentialmedium (DMEM; GIBCO/BRL) supplemented with 15% FCS, 100 mMMEM nonessential amino acids, 0.55 mM 2-mercaptoethanol, L-glutamine,and antibiotics (all from GIBCO/BRL). To induce EB formation (stage 2), thecells were dissociated into a single-cell suspension by 0.05% trypsin and0.04% EDTA in PBS and plated onto nonadherent bacterial culture dishes at adensity of 2–2.5 × 104 cells cm-2 in the medium described above. The EBs wereformed for four days and then plated onto adhesive tissue culture surface inthe ES cell medium. After 24 h of culture, selection of nestin-positive cells(stage 3) was initiated by replacing the ES cell medium by serum-freeInsulin/Transferrin/Selenium/Fibronectin (ITSFn) medium10.

After 6–10 days of selection, cell expansion (stage 4) was initiated.Specifically, the cells were dissociated by 0.05% trypsin/0.04% EDTA, andplated on tissue culture plastic or glass coverslips at a concentration of 1.5–2 ×105 cells cm-2 in N2 medium modified according to Johe and colleagues22, andsupplemented with 1 µg ml-1 of laminin and 10 ng ml-1 of bFGF (R&DSystems, Minneapolis, MN) in the presence or absence of murine N-terminalfragment of SHH (500 ng ml-1) and murine FGF8 isoform b (100 ng ml-1; bothfrom R&D Systems). Before cell plating, dishes and coverslips were precoated

with polyornithine (15 mg ml-1) and laminin (1 µg ml-1, both from BectonDickinson Labware, Bedford, MA). Nestin-positive cells were expanded for sixdays. The medium was changed every two days. Differentiation (stage 5) wasinduced by removal of bFGF. The differentiation medium consisted of N2medium supplemented with laminin (1 mg ml-1) in the presence or absence ofcAMP (1 µM), AA (200 µM, both from Sigma, St. Louis, MO). The cells wereincubated under differentiation conditions for 6–15 days.

Immunocytochemistry. Cells were fixed in 4% paraformaldehyde/0.15%picric acid in PBS. Immunocytochemistry was carried out using standardprotocols. Antibodies and dilutions were as follows: TH polyclonal 1:200(Pel-Freeze, Rogers, AR) or TH monoclonal 1:1000 (Sigma), β-tubulin typeIII (TuJ1) monoclonal 1:500 (Babco, Richmond, CA), GABA polyclonal1:1000 (Sigma), DBH polyclonal 1:100 (Protos Biotech Corp., New York,NY), or DBH polyclonal 1:100 (Pharmingen, San Diego, CA), serotoninpolyclonal 1:4000 (Sigma), and fluorescently labeled secondary antibodies(Jackson Immunoresearch Laboratories, West Grove, PA).

RNA extraction and RT-PCR analysis. Total cellular RNA was preparedusing RNAeasy total RNA purification kit (Qiagen, Valencia, CA) followed bytreatment with RNase-free RQ DNase (Promega Corp., Madison, WI). ForcDNA synthesis, random hexamer primers (Gibco/BRL) were used to primereverse transcriptase (RT) reactions. Using this method it was possible to usethe same RT reaction (cDNA) for PCR amplification with different sets ofgene-specific primers. The cDNA synthesis was carried out using Moloneymurine leukemia virus (M-MLV) Superscript II reverse transcriptase(Gibco/BRL) following the manufacturer's instructions. To analyze relativeexpression of different mRNAs, the amount of cDNA was normalized basedon the signal from ubiquitously expressed actin mRNA. Levels of neuralmRNAs at different stages of ES cell culture was compared to that in theundifferentiated ES cells. The PCR was carried out using standard protocolswith Taq polymerase (Boehringer-Mannheim, Indianapolis, IN). Cyclingparameters were as follows, denaturation at 94º C for 30sec, annealing at 58-61º C for 1min depending on the primer, and elongation at 72º C for 1 min.The number of cycles varied between 25 and 35, depending on the particularmRNA abundance. The number of cycles and the amount of cDNA was cho-sen in such a way as to select PCR conditions on the linear portion of thereaction curve avoiding “saturation effects” of PCR. The identity of the PCRproducts was confirmed by sequencing. Primer sequences (forward andreverse), and the length of the amplified products were as follows:

actin (ATGGATGACGATATCGCTG, ATGAGGTAGTCTGTCAGGT, 569);nestin (GGAGTGTCGCTTAGAGGTGC,TCCAGAAAGCCAAGAGAA-GC,327); Nurr1 (TGAAGAGAGCGGAGAAGGAGATC, TCTGGAGTTAAG-AAATCGGAGCTG, 255); Gli1 (TCCACAGGCATACAGGATCA,TGCAAC-CTTCTTGCTCACAC,462); Smoothened (Smo) (CTGAGAGTGCCA-GAAAAGGG, TCATCATGCTGGAGAACTCG, 370); Patched (Ptc) (CCTC-CTTTACGGTGGACAAA, ATCAACTCCTCCTGCCAATG, 272); Wnt1(ACCTGTTGACGGATTCCAAG,TCATGAGGAAGCGTAGGTCC,462); Otx1(GCTGTTCGCAAAGACTCGCTAC, ATGGCTCTGGCACTGATACGGATG,425); Otx2 (CCATGACCTATA CTCAGGCTTCAGG, GAAGCTCCATATCC-CTGGGTGGAAAG, 211); Pax2 (CCAAAGTGGTGGACAAGATTGCC,GGGATAGGAAGGACGCTCAAAGAC, 545); Pax5 (CAGATGTAGTCCGC-CAAAGGATAG, ATGCCACTGATGGAGTATGAGGAGCC, 451); FGFR3(ATCCTCGGGAGATGACGAAGAC, GGATGCTGCCAAACTTGTTCTC,326); Fgf8 (CATGTGAGGGACCAGAGCC, GTAGTTGTTCTCCAGC-AGGATC, 312); En1 (TCAAGACTGACTCACAGCAACCCC, CTTTGTCCT-GAACCGTGGTGGTAG, 381); Shh (GGAAGATCACAAGAAACTCCGAAC,GGATGCGAGCTTTGGATTCATAG, 354).

Reverse-phase HPLC. Dopamine levels were determined in conditionedmedium and in HBSS stabilized with orthophosphoric acid (7.5%)/metabisul-fate (0.22 mg ml-1), and stored at -80°C until analysis. Aluminum absorptionand HPLC analysis of dopamine have been described25,33.

Electrophysiology. For electrophysiological recordings, cells grown on 12mm glass coverslips were transferred to recording medium containing 130mM NaCl, 4 mM KCl, 2 mM CaCl2, 1 mM MgCl2, 10 mM HEPES, 10 mMglucose (pH 7.35, 325 mOsm). Patch pipettes were filled with 110 mM potas-sium gluconate, 20 mM KCl, 2 mM Mg-ATP, 10 mM sodium phosphocrea-tine, 1.0 mM EGTA, 0.3 mM GTP-Tris, and 20 mM HEPES (pH 7.25, 320mOsm). To label the recorded cells, biocytin (0.2%) was added to the intra-cellular medium. Recordings, biocytin labeling, and confocal imaging wereperformed as described34.

Cell counting and statistics. Uniform random sampling procedure wereused for cell counts and quantified using the fractionator technique32.Statistical comparisons were made by ANOVA with post hoc Dunnett test

678 NATURE BIOTECHNOLOGY VOL 18 JUNE 2000 http://biotech.nature.com

TECHNICAL REPORTS

Figure 5. Characterization of serotonin-positive neurons. (A) Immunostaining shows serotonin-positive neurons. Serotoninand TH-positive neurons are shown by immunostaining culturestreated at stage 4 in the absence (left) or presence (right) of SHH (500ng ml-1) and FGF8 (100 ng ml-1). The yield of serotonergic neuronsover the untreated controls in the presence of the growth factors(scale bars = 20 µm). (B) The yield of serotonin-positive neurons(expressed as a percentage of all TuJ1+ neurons) is shown for cellstreated with different growth factor combinations at stage 4. In thiscase, AA was not used at stage 5.

0

2

4

6

8

10

12

14

control SHH+FGF8 SHH FGF8

TH / Ser

No treatment SHH+FGF8

% S

erot

onin

(+)/

TuJ

1(+

)A

B

© 2000 Nature America Inc. • http://biotech.nature.com©

200

0 N

atu

re A

mer

ica

Inc.

• h

ttp

://b

iote

ch.n

atu

re.c

om

Page 5: Efficient generation of midbrain and hindbrain neurons from mouse embryonic stem cells

NATURE BIOTECHNOLOGY VOL 18 JUNE 2000 http://biotech.nature.com 679

when more than two groups were involved. If data were not normally distrib-uted, a nonparametric test (Mann–Whitney U-test) was used for the compar-isons of results. Data were expressed as mean ± s.e.m.

AcknowledgmentsThe R1 ES cell line was kindly provided by Dr. Heiner Westphal. The second EScell line used E14.1 was a gift of Dr. Tom Doetschman. We would like to thankDr. David Panchinsion for providing some of the PCR-primers and for criticaldiscussions of the manuscript.

1. Thomson, J.A. et al. Embryonic stem cell lines derived from human blastocysts.Science 282, 1145–1147 (1998).

2. Reubinoff, B. et al. Embryonic stem cell lines from human blastocysts: somaticdifferentiation in vitro. Nat. Biotechnol. 18, 399–404 (2000).

3. Shamblott, M.J. et al. Derivation of pluripotent stem cells from cultured humanprimordial germ cells. Proc. Natl. Acad. Sci. USA 95, 13726–13731 (1998).

4. Klug, M.G. et al., Genetically selected cardiomyocytes from differentiatingembryonic stem cells form stable intracardiac grafts. J. Clin. Invest. 98, 216–24(1996).

5. Keller, G. et al. Overexpression of HOX11 leads to the immortalization of embry-onic precursors with both primitive and definitive hematopoietic potential. Blood92, 877–87 (1998).

6. Wobus, A.M. & Boheler, K.R. Embryonic stem cells as developmental model invitro. Cells Tiss. Org. 165, 129–30 (1999).

7. Bain, G. et al. Embryonic stem cells express neuronal properties in vitro. Dev.Biol. 168, 342–357 (1995).

8. Deacon, T. et al. Blastula-stage stem cells can differentiate into dopaminergicand serotonergic neurons after transplantation. Exp. Neurol. 149, 28–41 (1998).

9. Brustle, O. et al. Embryonic stem cell-derived glial precursors: a source of myeli-nating transplants. Science 285, 754–756 (1999).

10. Okabe, S. et al. Development of neuronal precursor cells and functional potmi-totic neurons from embryonic stem cells in vitro. Mech. Dev. 59, 89–102 (1996).

11. Olanow, C.W., Kordower, J.H. & Freeman, T.B. Fetal nigral transplantation as atherapy for Parkinson’s disease. Trends Neurosci. 19, 102–109 (1996).

12. Breier, A., et al., Schizophrenia is associated with elevated amphetamine-induced synaptic dopamine concentrations: evidence from a novel positronemission tomography method. Proc. Natl. Acad. Sci. USA 94, 2569–2574 (1997).

13. Gramm, L.F. Drug therapy: fluoxetine. N. Engl. J. Med. 331, 1354 (1994).14. Volkow, N.D. et al. Decreased striatal dopaminergic responsiveness in detoxified

cocaine-dependent subjects. Nature 386, 830–833 (1997).15. Volkow, N.D. et al. Relationship between subjective effects of cocaine and

dopamine transporter occupancy. Nature 386, 827–830 (1997).16. Simeone, A. Otx1 and Otx2 in the development and evolution of the mammalian

brain. EMBO J. 17, 6790–6798 (1998).17. Acampora, D. & Simeone, A. Understanding the roles of Otx1 and Otx2 in the

control of brain morphogenesis. Trends Neurosci. 22, 116–122 (1999).18. Stoykova, A. & Gruss, P. Roles of Pax genes in developing and adult brain as

suggested by expression patterns. J. Neurosci. 14, 1395–1412 (1994).19. Rowitch, D.H. & McMahon, A. P., Pax-2 expression in the murine neural plate

precedes and encompasses the expression domains of Wnt-1 and En-1. MechDev. 52, 3–8 (1995).

20. Lendahl, U., Zimmerman, L.B. & McKay, R.D.G. CNS stem cells express a newclass of intermediate filament proteins. Cell 60, 585–595 (1990).

21. Frederiksen, K. & McKay, R.D.G. Proliferation and differentiation of rat neu-roephitelial precursor cells in vivo. J. Neurosci. 8, 1144–1151 (1988).

22. Johe, K. et al. Single factors direct the differentiation of stem cells from the fetaland adult nervous system. Genes Dev. 10, 3129–3140 (1996).

23. Vicario-Abejón, C. et al. Functions of basic-fibroblast growth factor and neu-rotrophins in the differentiation of hippocampal neurons. Neuron 15, 105–114(1995).

24. Studer, L., Tabar, V. & McKay, R. Transplantation of expanded mesencephalicprecursors leads to recovery in parkinsonian rats. Nat. Neurosci. 1, 290–295(1998).

25. Studer, L., Tabar, V. & McKay, R.D.G. Survival of expanded dopaminergic precur-sors is critical for clinical trials. Nat. Neurosci. 1, 537 (1998).

26. Ye, W. et al. FGF and SHH signals control dopaminergic and serotonergic cellfate in the anterior neural plate. Cell 93, 755–766 (1998).

27. Branton, R.L., Love, R.M. & Clarke, D.J. cAMP included during cell suspensionpreparation improves survival of dopaminergic neurons in vitro . Neuroreport 9,3223–3227 (1998).

28. Kalir, H.H. & Mytilineou, C. Ascorbic acid in mesencephalic cultures: effects ondopaminergic neuron development. J. Neurochem. 57, 458–464 (1991).

29. Brustle, O. et al. In vitro-generated neural precursors participate in mammalianbrain development. Proc. Natl. Acad. Sci. USA 94, 14809–14814 (1997).

30. Spenger, C. et al. Fetal ventral mesencephalon of human and rat origin main-tained in vitro and transplanted to 6-hydroxydopamine-lesioned rats gives rise tografts rich in dopaminergic neurons. Exp. Brain Res. 112, 47–57 (1996).

31. Wagner, J. et al. Induction of a midbrain dopaminergic phenotype in Nurr1-over-expressing neural stem cells by type 1 astrocytes. Nat. Biotechnol. 17, 653–659(1999).

32. Joyner, A.L. Engrailed, Wnt and Pax genes regulate midbrain–hindbrain develop-ment. Trends Genet. 12, 15–20 (1996).

33. Studer, L. et al. Noninvasive dopamine determination by reversed phase HPLC inthe medium of free-floating roller tube cultures of rat fetal ventral mesen-cephalon: a tool to assess dopaminergic tissue prior to grafting . Brain Res. Bull.41, 143–150 (1996).

34. Auerbach, J.M., Eiden, M.V. & McKay, R.D.G. Tranplanted CNS stem cells formfunctional synapses in vivo. Eur. J. Neurosci. 12, 1696–1704 (2000).

Genome-directedprimers for selectivelabeling of bacterialtranscripts for DNAmicroarray analysisAdel M. Talaat, Preston Hunter, and Stephen AlbertJohnston*

Center for Biomedical Inventions and Department of Medicine, University ofTexas-Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX75390-8573. *Corresponding author ([email protected])

Received 9 March, 2000; accepted 25 April, 2000

DNA microarrays have the ability to analyze the expression of thou-sands of the same set of genes under at least two different experi-mental conditions1. However, DNA microarrays require substantialamounts of RNA to generate the probes, especially when bacterialRNA is used for hybridization (50 µg of bacterial total RNA con-tains approximately 2 µg of mRNA)2. We have developed a comput-er-based algorithm for prediction of the minimal number ofprimers to specifically anneal to all genes in a given genome. Thealgorithm predicts, for example, that 37 oligonucleotides shouldprime all genes in the Mycobacterium tuberculosis genome. We test-ed the usefulness of the genome-directed primers (GDPs) in com-parison to random primers for gene expression profiling usingDNA microarrays. Both types of primers were used to generate flu-orescent-labeled probes and to hybridize to an array of 960mycobacterial genes. Compared to random-primer probes, theGDP probes were more sensitive and more specific, especially whenmammalian RNA samples were spiked with mycobacterial RNA.The GDPs were used for gene expression profiling of mycobacterialcultures grown to early log or stationary growth phases. Thisapproach could be useful for accurate genome-wide expressionanalysis, especially for in vivo gene expression profiling, as well asdirected amplification of sequenced genomes.

We developed a computer algorithm to define the minimal num-ber of oligonucleotides of a given length capable of priming all geneswithin any genome. Using the genome sequence of Mycobacteriumtuberculosis, we applied the algorithm, setting the oligonucleotideslength at eight or seven bases, and requiring 100% coverage of the3,924 open reading frames (ORF) in the genome3 (Fig. 1A). Thesearch was limited to the first 500 bp of each complementarysequence of each ORF to generate long probes for efficienthybridization. The priming efficiency of the mycobacterial genome-directed primers (mtGDPs; 37 primers) was compared to the prim-ing efficiency of seven- or six-nucleotide random primers in a stan-dard reverse transcription reaction. Probes generated from the samemycobacterial RNA (log phase cultures) using mtGDPs or randomprimers were hybridized simultaneously to the same slides, and thesignal intensities of the 960 arrayed genes were calculated and com-pared4,5. There was a high correlation level between the signals ofboth probes for the whole array of genes (r = 0.97). Additionally, sig-nals generated by mtGDPs were significantly higher than randomprimer-generated signals (P < 0.05) (Fig. 1B, C). Signal intensities

TECHNICAL REPORTS

© 2000 Nature America Inc. • http://biotech.nature.com©

200

0 N

atu

re A

mer

ica

Inc.

• h

ttp

://b

iote

ch.n

atu

re.c

om