Restoration of the intrinsic properties of human …...hypothesized that culture conditions...

12
Journal of Cell Science Restoration of the intrinsic properties of human dermal papilla in vitro Manabu Ohyama*, Tetsuro Kobayashi, Takashi Sasaki, Atsushi Shimizu and Masayuki Amagai Department of Dermatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan *Author for correspondence ([email protected]) Accepted 1 May 2012 Journal of Cell Science 125, 4114–4125 ß 2012. Published by The Company of Biologists Ltd doi: 10.1242/jcs.105700 Summary The dermal papilla (DP) plays pivotal roles in hair follicle morphogenesis and cycling. However, characterization and/or propagation of human DPs have been unsatisfactory because of the lack of efficient isolation methods and the loss of innate characteristics in vitro. We hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion of functional DP cells. To test this, we first characterized the global gene expression profile of microdissected, non-cultured human DPs. We performed a ‘two-step’ microarray analysis to exclude the influence of unwanted contaminants in isolated DPs and successfully identified 118 human DP signature genes, including 38 genes listed in the mouse DP signature. The bioinformatics analysis of the DP gene list revealed that WNT, BMP and FGF signaling pathways were upregulated in intact DPs and addition of 6-bromoindirubin-39- oxime, recombinant BMP2 and basic FGF to stimulate these respective signaling pathways resulted in maintained expression of in situ DP signature genes in primarily cultured human DP cells. More importantly, the exposure to these stimulants restored normally reduced DP biomarker expression in conventionally cultured DP cells. Cell growth was moderate in the newly developed culture medium. However, rapid DP cell expansion by conventional culture followed by the restoration by defined activators provided a sufficient number of DP cells that demonstrated characteristic DP activities in functional assays. The study reported here revealed previously unreported molecular mechanisms contributing to human DP properties and describes a useful technique for the investigation of human DP biology and hair follicle bioengineering. Key words: Human, Dermal papilla, Molecular signature, Cell culture Introduction The dermal papilla (DP) is a highly specialized mesenchymal component located at the base of hair follicles (Paus and Cotsarelis, 1999; Stenn and Paus, 2001; Driskell et al., 2011). Previous studies, including hair reconstitution experiments, have demonstrated the indispensable roles of DP in epithelial–mesenchymal interactions that enable hair follicle morphogenesis and cycling (for reviews see Oliver, 1991; Jahoda and Reynolds, 1996; Millar, 2002; Botchkarev and Kishimoto, 2003; Ohyama et al., 2010; Yang and Cotsarelis, 2010; Driskell et al., 2011). In mice, elegant transgenic approaches (Kishimoto et al., 1999; Rendl et al., 2005; Driskell et al., 2009) and newly identified cell surface markers such as CD133 (Ito et al., 2007) and a9-integrin (Rendl et al., 2005) enabled fluorescent labeling and targeted isolation of living DP cells. Sufficient numbers of mouse DP cells are now available for downstream applications such as microarray analysis and tissue regeneration assays (Rendl et al., 2005; Rendl et al., 2008; Driskell et al., 2009). However, isolation of human DP cells mostly depends on manual microdissection, which provides a limited number of DP cells (Messenger, 1984; Magerl et al., 2002; Ohyama et al., 2010). Accordingly, previous studies of human DP predominantly utilized cultured DP cells (Inui et al., 2003; Pflieger et al., 2006; Iino et al., 2007; Park et al., 2007; Shin et al., 2010). A potential disadvantage of this approach is the loss of some intrinsic properties of DP cells during culture (Ohyama et al., 2010; Yang and Cotsarelis, 2010). Loss of biological characteristics, especially hair inductive capacity, has been reported in rodent (Jahoda et al., 1984; Horne et al., 1986; Lichti et al., 1993; Weinberg et al., 1993) and canine (Kobayashi et al., 2011) DP cells, and is likely to occur in human DP cells. Thus, a clear demand exists for an approach that allows in vitro expansion of human DP cells without affecting their in situ characteristics. Previous studies reported various approaches to maintaining in vivo characteristics in cultured DP cells, including co-culture with keratinocytes (Reynolds and Jahoda, 1996; Inamatsu et al., 1998; Kobayashi et al., 2011), cultivation on extracellular matrices, and sphere formation (Osada et al., 2007; Young et al., 2009; Higgins et al., 2010; Kobayashi et al., 2011). The hair inductive capacity of rodent and canine DP cells was maintained in cultures treated with keratinocyte-derived factors (Inamatsu et al., 1998; Kobayashi et al., 2011) and ligands in signaling pathways involved in hair follicle morphogenesis, including Wnt3a and Bmp6 (Kishimoto et al., 2000; Shimizu and Morgan, 2004; Rendl et al., 2008). However, whether a similar approach is feasible with human DP cells has not been fully assessed. In the present study, we first elucidated the molecular signature of intact, non-cultured human DP. Subsequent bioinformatics analysis identified biological pathways that are activated in DP. By administering an optimized combination of activators of those pathways, a culture medium that maintained or even restored intrinsic DP characteristics was successfully developed. The current study uncovered previously unreported molecular characteristics of in vivo human DP and described an efficient methodology with which to propagate DP cells while maintaining intrinsic properties. 4114 Research Article

Transcript of Restoration of the intrinsic properties of human …...hypothesized that culture conditions...

Page 1: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

Restoration of the intrinsic properties of human dermalpapilla in vitro

Manabu Ohyama*, Tetsuro Kobayashi, Takashi Sasaki, Atsushi Shimizu and Masayuki AmagaiDepartment of Dermatology, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan

*Author for correspondence ([email protected])

Accepted 1 May 2012Journal of Cell Science 125, 4114–4125� 2012. Published by The Company of Biologists Ltddoi: 10.1242/jcs.105700

SummaryThe dermal papilla (DP) plays pivotal roles in hair follicle morphogenesis and cycling. However, characterization and/or propagation ofhuman DPs have been unsatisfactory because of the lack of efficient isolation methods and the loss of innate characteristics in vitro. We

hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion offunctional DP cells. To test this, we first characterized the global gene expression profile of microdissected, non-cultured human DPs.We performed a ‘two-step’ microarray analysis to exclude the influence of unwanted contaminants in isolated DPs and successfully

identified 118 human DP signature genes, including 38 genes listed in the mouse DP signature. The bioinformatics analysis of the DPgene list revealed that WNT, BMP and FGF signaling pathways were upregulated in intact DPs and addition of 6-bromoindirubin-39-oxime, recombinant BMP2 and basic FGF to stimulate these respective signaling pathways resulted in maintained expression of in situ

DP signature genes in primarily cultured human DP cells. More importantly, the exposure to these stimulants restored normally reducedDP biomarker expression in conventionally cultured DP cells. Cell growth was moderate in the newly developed culture medium.However, rapid DP cell expansion by conventional culture followed by the restoration by defined activators provided a sufficient

number of DP cells that demonstrated characteristic DP activities in functional assays. The study reported here revealed previouslyunreported molecular mechanisms contributing to human DP properties and describes a useful technique for the investigation of humanDP biology and hair follicle bioengineering.

Key words: Human, Dermal papilla, Molecular signature, Cell culture

IntroductionThe dermal papilla (DP) is a highly specialized mesenchymalcomponent located at the base of hair follicles (Paus and Cotsarelis,

1999; Stenn and Paus, 2001; Driskell et al., 2011). Previous studies,including hair reconstitution experiments, have demonstrated the

indispensable roles of DP in epithelial–mesenchymal interactionsthat enable hair follicle morphogenesis and cycling (for reviews see

Oliver, 1991; Jahoda and Reynolds, 1996; Millar, 2002; Botchkarev

and Kishimoto, 2003; Ohyama et al., 2010; Yang and Cotsarelis,2010; Driskell et al., 2011). In mice, elegant transgenic approaches

(Kishimoto et al., 1999; Rendl et al., 2005; Driskell et al., 2009) andnewly identified cell surface markers such as CD133 (Ito et al.,

2007) and a9-integrin (Rendl et al., 2005) enabled fluorescent

labeling and targeted isolation of living DP cells. Sufficient numbersof mouse DP cells are now available for downstream applications

such as microarray analysis and tissue regeneration assays (Rendlet al., 2005; Rendl et al., 2008; Driskell et al., 2009). However,

isolation of human DP cells mostly depends on manualmicrodissection, which provides a limited number of DP cells

(Messenger, 1984; Magerl et al., 2002; Ohyama et al., 2010).

Accordingly, previous studies of human DP predominantly utilizedcultured DP cells (Inui et al., 2003; Pflieger et al., 2006; Iino et al.,

2007; Park et al., 2007; Shin et al., 2010). A potential disadvantageof this approach is the loss of some intrinsic properties of DP cells

during culture (Ohyama et al., 2010; Yang and Cotsarelis, 2010).

Loss of biological characteristics, especially hair inductive capacity,has been reported in rodent (Jahoda et al., 1984; Horne et al., 1986;

Lichti et al., 1993; Weinberg et al., 1993) and canine (Kobayashi

et al., 2011) DP cells, and is likely to occur in human DP cells. Thus,a clear demand exists for an approach that allows in vitro expansionof human DP cells without affecting their in situ characteristics.

Previous studies reported various approaches to maintaining in

vivo characteristics in cultured DP cells, including co-culture withkeratinocytes (Reynolds and Jahoda, 1996; Inamatsu et al., 1998;Kobayashi et al., 2011), cultivation on extracellular matrices, and

sphere formation (Osada et al., 2007; Young et al., 2009; Higginset al., 2010; Kobayashi et al., 2011). The hair inductive capacity ofrodent and canine DP cells was maintained in cultures treated with

keratinocyte-derived factors (Inamatsu et al., 1998; Kobayashiet al., 2011) and ligands in signaling pathways involved in hairfollicle morphogenesis, including Wnt3a and Bmp6 (Kishimoto

et al., 2000; Shimizu and Morgan, 2004; Rendl et al., 2008).However, whether a similar approach is feasible with human DPcells has not been fully assessed.

In the present study, we first elucidated the molecular signature

of intact, non-cultured human DP. Subsequent bioinformaticsanalysis identified biological pathways that are activated in DP.By administering an optimized combination of activators of those

pathways, a culture medium that maintained or even restoredintrinsic DP characteristics was successfully developed. Thecurrent study uncovered previously unreported molecular

characteristics of in vivo human DP and described an efficientmethodology with which to propagate DP cells while maintainingintrinsic properties.

4114 Research Article

Page 2: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

ResultsMicroarray analysis of total RNA isolated from freshly

microdissected human DP

In our attempt to elucidate the global gene expression profile of

intact human DP (Fig. 1A), 15 DPs were microdissected from

anagen hair follicles obtained from four volunteers (Fig. 1A). As

controls, human DP cells and fibroblasts primarily cultured in

Dulbecco’s modified Eagle’s medium containing 10% fetal

bovine serum (10% FBS–DMEM) (de Almeida et al., 2005; Wu

et al., 2005; Iino et al., 2007) were prepared from the same

Fig. 1. Microarray comparisons between human DP and controls identified human DP signature genes. (A) Morphology and histology of the human DP in a

human anagen hair follicle. For microarray generation, total RNA was isolated from freshly microdissected DP (fDP) and cultured DP (cDP) cells and fibroblasts (Fibro).

Scale bars: 100 mm (fDP); 300 mm (cDP and Fibro). (B) Real-time PCR analysis detected higher expression of established DP signature genes in fDP compared with cDP

cells and Fibro, confirming successful RNA isolation (*P,0.05). (C) In approach 1 microarray analysis, the genes upregulated in cDP versus Fibro and not

downregulated in fDP versus cDP cells were selected. (D) Heat map of DP signature genes detected by approach 1. (E) Real-time PCR confirmation of approach 1 genes

(*P,0.05). (F) In approach 2, the genes increased in aggregated cDP (agDP) versus cDP cells and upregulated in fDP compared with cDP cells and Fibros were selected.

Scale bar: 200 mm (agDP). (G) Heat map of DP signature genes detected by approach 2. (H) Approach 2 genes confirmed by real-time PCR (*P,0.05).

Restoration of dermal papilla 4115

Page 3: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

individuals (Fig. 1A). Real-time PCR analysis of total RNA

isolated from freshly isolated human DP (fDP), cultured DP

(cDP) cells, and fibroblasts (Fibros) detected upregulation of

genes that have been used to confirm successful DP cell isolation

(Botchkarev and Kishimoto, 2003; Rendl et al., 2005; Ito et al.,

2007; Osada et al., 2007; Driskell et al., 2009; Ohyama et al.,

2010), such as alkaline phosphatase (ALPL), noggin (NOG),

WNT inhibitory factor 1 (WIF1), and versican (VCAN), in fDP as

compared with cDP cells and Fibros, demonstrating successful

RNA recovery (Fig. 1B). The same total RNA samples were

analyzed using four sets of Affymetrix HG-U133A 2.0 GeneChip

microarrays. Despite limitations in the starting materials, the

GeneChip data were of high quality (Khanna et al., 2004) and

comparable with one another (Ohyama et al., 2006), with

55.563.14% of transcripts found to be expressed and an

average correlation coefficient of 0.9560.03 for the

microarrays representing each population. The raw GeneChip

data files are accessible at NCBI’s Gene Expression Omnibus

through GEO Series accession number GSE31324 (http://www.

ncbi.nlm.nih.gov/geo/query/acc.cgi?acc5GSE31324).

Elucidation of gene expression profiles that distinguish

intact human DP from conventionally cultured human DPcells and fibroblasts

Global gene expression profiles for fDP, cDP and Fibro

GeneChips were subsequently compared to identify genes that

are overrepresented in fDP. It should be noted that fDP samples

contained microvasculature and possibly minor contaminants

such as matrix cells and melanocytes (Magerl et al., 2002;

Kobayashi et al., 2011), whereas cDP and Fibro samples

contained almost pure cell populations (Fig. 1A). To minimize

the influence of unwanted contaminants on gene expression

profiles, two respective microarray analyses were performed. In

each analysis, DP signature genes were selected by a ‘two-step’

filtering approach comprising primary gene screening by

comparing global gene expression profiles of two well-defined

subsets, and subsequent selection by assessing the expression

patterns of candidate genes among additional cell populations

(Ohyama et al., 2006).

In the first analysis (approach 1), cDP and Fibro GeneChip

data (n54) were compared to identify genes upregulated in cDP.

The expression levels of those genes in fDP and cDP cells were

then compared to identify genes that were equally or more highly

expressed in fDP (Fig. 1C). Among 105 genes detected as

upregulated in cDP compared with in Fibros, 70 genes

(supplementary material Table S1) were removed because

they were downregulated in fDP compared with in cDP.

Bioinformatics analysis using Database for Annotation,

Visualization and Integrated Discovery software (DAVID, ver.

6.7; http://david.abcc.ncifcrf.gov/) (Huang et al., 2009a; Huang

et al., 2009b) revealed that those genes were involved in

metabolic processes and responses to external stimulus,

suggesting that their upregulation was due to in vitro expansion.

No previously described DP gene (Botchkarev and Kishimoto,

2003; Rendl et al., 2005; Ito et al., 2007; Osada et al., 2007;

Driskell et al., 2009; Ohyama et al., 2010) was excluded in this

step. This analysis identified 35 genes upregulated in fDP

(Table 1; supplementary material Table S2). Differential expres-

sion of the selected genes was visualized by a heat map (Fig. 1D)

and validated by real-time PCR analyses of 11 randomly selected

genes (representative data in Fig. 1E). However, this approach

did not allow for the detection of genes whose expression was

rapidly downregulated in cDP cells to levels similar to those in

Fibros (Fig. 1F).

Previous studies reported that DP cell aggregation by

centrifugation in low cell binding plates, culturing on synthetic

membranes, or hanging drop culture recovered DP functions,

including DP biomarker expression. These results suggest that

genes upregulated by DP cell aggregation could contribute to the

biological distinctiveness of in situ DP (Osada et al., 2007; Young

et al., 2008; Higgins et al., 2010). Accordingly, in the second

analysis (approach 2), two additional sets of GeneChips were

generated from aggregated cDP (agDP) cells and cDP cells used

for agDP (cDP for agDP) and compared. Among the genes

detected as upregulated in agDP arrays, those more highly

expressed in fDP compared with both cDP and Fibros were

finally selected as DP signature genes (Fig. 1F). A total of 77

genes were identified in this analysis (Table 2; supplementary

Table 1. Top 15 human DP signature genes detected by approach 1

Affymetrix ID Gene name Gene symbolFold change cDP vs

FibroFold change fDP vs

cDP

204613_at Phospholipase C, gamma 2 PLCG2 10.2 2.1204653_at Transcription factor ap-2 alpha TFAP2A 8.3 5.2206115_at Early growth response 3 EGR3 6.0 3.4219743_at Hairy/enhancer-of-split related with YRPW motif 2 HEY2 5.9 7.7209541_at Insulin-like growth factor 1 IGF1 5.8 3.0203304_at Bmp and activin membrane-bound inhibitor homolog BAMBI 5.7 2.0221558_s_at Lymphoid enhancer-binding factor 1 LEF1 5.0 12.6204971_at Cystatin A (stefin A) CSTA 4.8 2.1201830_s_at Neuroepithelial cell transforming gene 1 NET1 4.8 1.7202411_at Interferon, alpha-inducible protein 27 IFI27 4.6 1.7221796_at Neurotrophic tyrosine kinase, receptor, type 2 NTRK2 4.5 6.4202626_s_ata

202625_atV-yes-1 Yamaguchi sarcoma viral related oncogene homolog LYN 4.1 3.0

201286_ata

201287_s_atSyndecan 1 SDC1 3.9 4.8

211368_s_at Caspase 1, apoptosis-related cysteine peptidase CASP1 3.8 3.9202388_at Regulator of G-protein signaling 2, 24kDa RGS2 3.7 10.4

aFor the genes supported by multiple probe sets, the probe set with the highest fold change between cDP and Fibro is indicated by bold letters.

Journal of Cell Science 125 (17)4116

Page 4: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

material Table S3). The data are displayed in a heat map (Fig. 1G).

Real-time PCR analyses of 16 randomly selected genes

demonstrated expression patterns consistent with microarray

data, although statistical significance between agDP and cDP

was not sufficient for two genes (representative data in Fig. 1H).

The genes upregulated by DP cell aggregation accounted for

approximately 20% of all genes detected as increased in fDP

among fDP, cDP, and Fibro subsets. Accordingly, a large

proportion of fDP genes (n5320; supplementary material Table

S4) were filtered out in the first step. A total of 36 known hair

follicle keratinocyte-related genes, 12 melanocyte-related genes,

and 8 vasculature-related genes (supplementary material Table S4)

(Rendl et al., 2005; Huang et al., 2009a; Huang et al., 2009b) were

found in the removed genes, whereas such genes were not included

in those upregulated in agDP cells. These results suggest that the

genes expressed by the contaminants in microdissected DP were

indeed excluded in the two-step approach.

A total of 108 genes were found to be upregulated in intact

human DP cells by two microarray analyses. When the genes

detected as increased in fDP prior to microarray generation

(Fig. 1B) were combined, 118 human DP signature genes were

identified in this study (Fig. 2A).

Bioinformatics analysis revealing functional aspects of DPsignature genes, including the signaling pathwaysinvolved

To categorize human DP signature genes, a functional

classification analysis was performed using DAVID software

(Huang et al., 2009a; Huang et al., 2009b), in which DP signature

genes were divided into 14 groups according to enrichment

scores (Fig. 2A). The clusters with high enrichment scores

(.2.0) mainly comprised genes involved in WNT [frizzled-

related protein (FRZB), Norrie disease (NDP), WIF1, wingless-

related MMTV integration site 5A (WNT5A)], bone

morphogenetic proteins 2 and 4 (BMP2, BMP4), NOG, and

sclerostin domain containing 1 (SOSTDC1) and fibroblast growth

factor [FGF; FGF7 and sprouty homologs 1 and 4 (SPRY1,

SPRY4)] signaling pathways, suggesting the significance of these

pathways in the maintenance of intrinsic DP properties (Fig. 2A).

DAVID gene ontology analysis annotated 100 of 118 signature

genes as those with binding functions and 23 and 17 genes with

transcription regulator and enzyme regulator activity, respectively.

These results indicate that most DP signature genes were involved in

intercellular or intracellular signal transduction or regulation.

Previously unreported human DP genes encoding receptor binding

molecules [e.g. glia maturation factor gamma (GMFG), lipoprotein

lipase (LPL), NDP and metastasis suppressor 1 (MTSS1)], activators

or inhibitors of enzyme activity [e.g. apolipoprotein E (APOE); G-

protein signaling 2, 24 kDa (RGS2); cyctatin A (CSTA); and stratifin

(SFN)], and transcription factors [e.g. ets variant 1 (ETV1)],

forkhead box O1 (FOXO1), growth arrest-specific 7 (GAS7), and

myocyte enhancer factor 2C (MEF2C)] were listed, which could

imply novel mechanisms contributing to human DP function

(Fig. 2B). The genes of membrane proteins [e.g. endothelin

receptor type A (EDNRA), G-protein-coupled receptor 125

(GPR125), and low-density lipoprotein receptor-related protein 4

Table 2. Top 25 human DP signature genes detected by approach 2

Afffymetrix ID Gene name Gene SymbolFold change agDP

vs cDPFold change fDP

vs cDP

203382_s_ata Apolipoprotein E APOE 50.2 92.1203381_s_at

213994_s_ata

213993_at209436_at

Spondin 1 SPON150.2 247.3

209437_s_at

202291_s_at Matrix Gla protein MGP 27.9 260.5208399_s_at Endothelin 3 EDN3 26.0 251.6202388_at Regulator of G-protein signaling 2, 24kda RGS2 23.4 10.4203549_s_at Lipoprotein lipase LPL 11.3 43.0203423_at Retinol binding protein 1, cellular RBP1 10.2 4.4209047_at Aquaporin 1 AQP1 9.8 22.6218807_at Vav 3 oncogene VAV3 9.8 33.7204040_at Ring finger protein 144 RNF144A 9.5 5.9204463_s_at Endothelin receptor type A EDNRA 8.9 28.3205258_at Inhibin, beta B INHBB 8.9 25.1219511_s_at Synuclein, alpha-interacting protein SNCAIP 8.9 16.3206022_at Norrie disease NDP 8.9 5.4202191_s_at Growth arrest-specific 7 GAS7 8.0 16.3

205290_s_ata

205289_atBone morphogenetic protein 2 BMP2

7.7 41.5

212558_at Sprouty homolog 1 SPRY1 7.7 13.9

216604_s_ata

202752_x_atSolute carrier family 7, member 8 SLC7A8

7.2 17.1

219407_s_at Laminin, gamma 3 LAMC3 7.0 38.7200795_at Sparc-like 1 SPARCL1 7.0 355.8203037_s_at Metastasis suppressor 1 MTSS1 6.3 46.0221911_at Ets variant gene 1 ETV1 5.7 5.9206165_s_at Chloride channel, calcium activated, family member 2 CLCA2 5.3 97.0202724_s_at Forkhead box O1a FOXO1 5.3 12.6203632_s_at G protein-coupled receptor, family C, group 5, member B GPRC5B 5.3 32.6

aFor the genes supported by multiple probe sets, the probe set with the highest fold change between agDP and cDP is indicated by bold letters.

Restoration of dermal papilla 4117

Page 5: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

(LRP4)] and extracellular matrix proteins [e.g. spondin 1 (SPON1)

and matrix Gla protein (MGP)] were also identified (Fig. 2B).

Importantly, 38 of these genes are reportedly over-

represented in mouse DP cells (Fig. 2C) (Sleeman et al.,

2000; Botchkarev and Kishimoto, 2003; Panteleyev et al., 2003;

O’Shaughnessy et al., 2004; Rendl et al., 2005; Driskell et al.,

2009). Most of them were established DP signature genes

(Botchkarev and Kishimoto, 2003; Rendl et al., 2005; Ito et al.,

2007; Osada et al., 2007; Driskell et al., 2009; Ohyama et al.,

2010), including those encoding key molecules that enhance

[e.g. FGF7, insulin-like growth factor-1 (IGF1), and WNT5A]

or inhibit (e.g. FRZB and SOSTDC1) interactions between DP

and the hair matrix (Fig. 2B). These findings indicate that a

fundamental molecular mechanism is conserved between mouse

and human DP cells.

Development of DP activation culture (DPAC) medium: theoptimized culture conditions that maintain the intrinsic

properties of human DP cells

As an application of the results obtained above, we attempted to

develop a culture medium that allows human DP cell expansion

while sustaining intrinsic properties, including the molecular

signature. Because the bioinformatics analysis implied the

involvement of WNT, BMP, and FGF signaling pathways in

the maintenance of human DP properties, we anticipated that

stimulation of these pathways during culture would improve the

biological characteristics of cultured DP cells. To test this

hypothesis, 6-bromoindirubin-39-oxime (BIO), recombinant

BMP2 (rBMP), and basic FGF (bFGF) were individually added

to conventionally used 10% FBS–DMEM to stimulate the WNT,

BMP, and FGF pathways, respectively.

Fig. 2. Bioinformatics analyses of the molecular signature of human DP. (A) Human DP signature genes were subdivided into 14 groups by functional

classification. Clusters with high enrichment scores (.2) contained genes involved in WNT, BMP and FGF signaling. (B) Schematic illustration of the outcome of

gene ontology analysis. DP signature genes were enriched for those with binding functions, including signaling molecules, enzyme activity regulators and

transcription factors. Previously unreported genes encoding membrane and extracellular matrix molecules were also detected. (C) Thirty-eight DP signature genes

were conserved between mice and humans, suggesting that they have essential roles in DP biology.

Journal of Cell Science 125 (17)4118

Page 6: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

To assess the effect of each activator on DP properties,

expression of the classic DP genes ALPL, lymphoid enhancer-

binding factor 1 (LEF1), and NOG, as well as of RGS2, a DP

gene identified in this study, was monitored (Fig. 3A).

Interestingly, the expression of these genes was not always

upregulated in a dose-dependent manner (Fig. 3A). BIO

markedly increased the expression of all tested genes at a

concentrations of 1 mM and higher. However, cell proliferation

was markedly suppressed in 2 mM BIO. The ameliorative effect

of rBMP on DP gene expression did not differ significantly

Fig. 3. Development of dermal papilla activation culture (DPAC) medium. (A) Differential effects of BIO, rBMP and bFGF on human DP signature gene

expression. (B) The combination of rBMP and bFGF significantly enhanced DP cell proliferation, whereas the combination of BIO plus bFGF and a combination

of all three activators moderately suppressed DP cell growth. Scale bar: 300 mm. (C) Treatment with all three activators significantly increased the expression of

all DP signature genes (*P,0.05). As a result, 10% FBS–DMEM containing 1 mM BIO, 200 ng/ml rBMP and 20 ng/ml bFGF was named ‘dermal papilla

activation culture’ (DPAC) medium. (D) Human DP cells primarily cultured in DPAC (dpacDP) maintained the expression of some DP signature genes at a level

comparable to that in fDP, while suppressing expression of the fibroblast gene PTX3 (*P,0.05 for cDPs). (E) At lower passages, the morphology of DP cells in

DPAC was distinct from those cultured under control conditions (10% FBS–DMEM). Numbers of cells with a fibroblast-like morphology increased as the passage

number increased. Scale bar: 300 mm. (F) DPAC sustained DP signature gene expression across a number of passages. (G) Alkaline phosphatase activity (red) was

maintained for more than four passages in DPAC, while it was almost completely lost within two passages under control conditions. Scale bar: 100 mm.

Restoration of dermal papilla 4119

Page 7: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

between 200 or 400 ng/ml. bFGF dose-dependently enhanced DPcell proliferation. However, an increase in dead cells, probably

due to overproliferation, was observed at 40 ng/ml (data notshown). Although downregulation of ALPL was observed at ahigher concentration, upregulation of other genes was maintained

at 20 ng/ml. Accordingly, each activator differently maintainedDP signature gene expression at an individual optimalconcentration of 1 mM for BIO, 200 ng/ml for rBMP, and20 ng/ml for bFGF (Fig. 3A).

We next prepared 10% FBS–DMEM containing optimalconcentrations of two or all three factors and cultured fDP

cells. The combination of rBMP and bFGF enhanced DP cellproliferation, whereas the mixture of BIO and bFGF or all threefactors reduced DP cell growth to approximately 30% that ofcontrols (Fig. 3B). In addition, DP cells hardly proliferated when

exposed to the combination of BIO and rBMP (data not shown).Importantly, the combination of all three factors causedstatistically significant (P,0.05) upregulation of all tested DP

signature genes compared with controls (Fig. 3C). Thus, 10%FBS–DMEM containing 1 mM BIO, 200 ng/ml rBMP, and20 ng/ml bFGF provided the most favorable conditions for

maintaining proliferative capacity and intrinsic gene expressionin cultured human DP cells. We named this medium ‘dermalpapilla activation culture’ or DPAC.

Expression of other classic DP signature genes in DP cellsprimarily cultured in DPAC (dpacDP) was comparable with orhigher than that in fDP (Fig. 3D). In addition, pentraxin 3

(PTX3), a Fibro signature gene upregulated in cDP comparedwith fDP, was significantly underrepresented in dpacDP versuscDP, further confirming the effect of DPAC (Fig. 3D).

Both the conventional medium (10% FBS–DMEM) and DPACallowed DP cell expansion for more than five passages (Fig. 3E).Early-passage dpacDP cells predominantly demonstrated a small

polygonal appearance distinct from the fibroblast-like morphologyof cDP cells. However, numbers of spindle-shaped cells increasedin higher-passage dpacDP (Fig. 3E). Consistent with thisobservation, ALPL, NOG, and BMP4 gene expression and

alkaline phosphatase enzyme (ALP) activity, which correlateswith the hair inductive capacity of DP cells (McElwee et al., 2003;Rendl et al., 2008), were maintained in early-passage dpacDP at

levels comparable with those in fDP (Fig. 3F,G). Thus, the effectsof DPAC persisted for several passages.

DPAC sustained the intrinsic molecular signature of intacthuman DP

Human DP cells primarily cultured in DPAC sustained theexpression of previously undescribed human DP signature genes(Fig. 4). LRP4, which is reportedly a specific marker of murineDP (Lrp4/corin) (Enshell-Seijffers et al., 2008; Enshell-Seijffers

et al., 2010), was almost identically expressed in fDP and dpacDP(Fig. 4). Likewise, BMP and activin membrane-bound inhibitorhomolog (BAMBI) and SPRY4, antagonists of TGF-b and

FGF pathways (Furthauer et al., 2001; Sekiya et al., 2004),respectively, and NDP, an activator of WNT signaling (Seitzet al., 2010), were similarly expressed in fDP and dpacDP cells

(Fig. 4). Expression of those not directly related to the activatedpathways, including ETV1; GPR125; guanylate cyclase 1,soluble, alpha 3 (GUCY1A3); and semaphorin-4C (SEMA4C),

were also well maintained (Fig. 4). Trichorhinophalangealsyndrome I (TRPS1), a gene responsible for human congenitalhair anomalies (Momeni et al., 2000), and sex-determining region

Y-box 2 (SOX2), a mouse DP gene that defines a hair shaft

phenotype (Driskell et al., 2009), were highly expressed in

dpacDP compared with cDP cells (Fig. 4). These findings

indicate that DPAC provides a useful tool to probe the links

between potential DP signature genes and human DP biology.

Restoration of impaired DP properties in conventionally

cultured human DP cells by DPAC

When DP cells initially cultured in 10% FBS–DMEM were

passaged into DPAC (yielding dpacDP P1 cells; Fig. 5A), their

morphology changed from spindle-shaped to a polygonal form

resembling that of DP cells primarily cultured in DPAC

(Fig. 5B). In line with this observation, DP signature genes

such as ALPL, LEF1, and NOG were upregulated after passaging

into DPAC (Fig. 5C). In addition, ALP activity was much higher

in dpacDP P1 cells compared with passage one cDP cells (cDP

P1 cells; Fig. 5A,D). Thus, DPAC ameliorated DP biomarker

expression reduced in conventionally cultured DP cells.

To assess whether exposure to DPAC improved DP functions,

either dpacDP P1 or cDP P1 cells were co-cultured with normal

human hair follicle keratinocytes (Fig. 6A) (Inui et al., 2002). In the

bulb of the hair follicle, DP cells interact with hair matrix cells,

actively dividing keratinocytes that generate the hair shaft (Rendl

et al., 2005; Rendl et al., 2008; Ohyama et al., 2010). Intriguingly,

dpacDP P1 cells significantly increased the expression of hair matrix

signature genes LEF1 and msh homeobox 2 (MSX2) (Rendl et al.,

2005) in keratinocytes compared with cDP P1 cells, suggesting that

exposure to DPAC ameliorated the capacity of DP cells to provoke

epithelial–mesenchymal interactions in the hair follicle (Fig. 6B).

An in vivo hair induction assay was subsequently performed.

DP-like cell aggregates were generated from dpacDP P1 or cDP P1

Fig. 4. DPAC maintained novel human DP signature gene expression in

vitro. The expression of newly identified human signature genes was

increased in cells cultured in DPAC (*P,0.05 for cDPs). Increased

expression of previously unreported human DP signature genes under DPAC

could further confirm their roles in DP biology.

Journal of Cell Science 125 (17)4120

Page 8: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

cells and placed between enzymatically separated afollicular murine

sole epidermis and dermis. The ‘sandwich’ composites were then

inserted subcutaneously into immunodeficient mice (Fig. 6C).

Intriguingly, developments suggestive of hair morphogenesis,

including epidermal invagination or papillary mesenchymal body

formation (Osada and Kobayashi, 2000; Osada et al., 2009)

(Fig. 6D), were observed in 9 of 21 mice in the DPAC-treated

group, but only in 2 of 18 mice in the control group (P,0.05). These

developments were surrounded by dermal cells positive for ALP and

human-specific vimentin staining (Fig. 6D,E), suggesting that they

were induced by transplanted human DP cells.

These findings demonstrate that DPAC restores some aspects

of DP cell function in conventionally cultured DP cells.

DiscussionThe success of microarray analysis greatly depends on the purity

of the tested cell populations (Rendl et al., 2005). Microdissected

DP predominantly comprised DP cells, as demonstrated by high

expression of established DP signature genes. However, the

vascular structure within DP could not be removed, and complete

detachment of the surrounding hair matrix cells and melanocytes

was hardly achieved by the microdissection technique (Ohyama

Fig. 5. DPAC medium restores impaired DP properties in conventionally

cultured human DP cells. (A) Intact human DPs (fDP) were cultured under

conventional culture conditions (which allow rapid cell proliferation), and

passaged into either DPAC or the conventional culture medium. (B) DP cells

started to change their morphology when passaged into DPAC. Scale bar:

300 mm. (C) Restoration of human DP signature gene expression after culture

in DPAC (*P,0.05). (D) ALP activity was recovered in cDPs cultured in

DPAC, but was barely detectable in cDPs passaged into a conventional

culture medium. Scale bar: 100 mm.

Fig. 6. Functional restoration of human DP cells by DPAC. (A) Co-culture

system to assess the capacity to provoke the epithelial–mesenchymal

interaction in the hair follicle bulb. (B) dpacDP P1 cells unregulated hair

matrix signature genes in co-cultured hair follicle keratinocytes compared to

cDP P1 cells. (C) A ‘sandwich’ assay was performed using dpacDP and cDP

cell aggregates. The aggregates were placed between mouse sole afollicular

epidermis and dermis and composites were transplanted subcutaneously into

nude mice. Scale bar: 100 mm. (D) A hair peg-like papillary mesenchymal

body formed after transplantation of dpacDP aggregates. Note the ALP-

positive dermal cells (arrowheads) surrounding the structure, and the loss of

ALP expression in the control aggregate. Scale bar: 50 mm. (E) An anti-

human vimentin antibody specifically stains human DP, but not mouse DP.

This antibody detected ALP-positive cells surrounding a hair like structure

(arrowheads), indicating that they were of human origin. Scale bar: 10 mm

(mouse DP); 50 mm (human DP); 100 mm (experimental sample).

Restoration of dermal papilla 4121

Page 9: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

et al., 2010; Kobayashi et al., 2011). Because the isolation of a

pure DP cell population is currently not feasible in humans(Ohyama et al., 2010), a methodology that allows for theexclusion of non DP-related genes from fDP upregulated genes

must be developed to obtain a bona fide intact DP geneexpression profile.

We previously successfully identified the markers of humanhair follicle putative stem cells by ‘two-step’ microarray analyses

(Ohyama et al., 2006). We adopted this approach to filter outunwanted genes. Because no signal from possible contaminantswas detected on cDP, Fibro, or agDP arrays, comparisons

between cDP and Fibro GeneChips and agDP and cDPGeneChips should detect the differential expression of thegenes expressed in DP cells. These preceding microarrayanalyses provided the criteria with which to link a gene to DP

fate in each subsequent step. In approach 1, the second step couldbe crucial because some DP signature genes might be detected asdownregulated in fDP compared with cDP cells and discarded

due to the lower occupancy of DP cells in fDP subsets. However,previously reported DP genes (Botchkarev and Kishimoto, 2003;Rendl et al., 2005; Ito et al., 2007; Osada et al., 2007; Driskell

et al., 2009; Ohyama et al., 2010) were not among the genesexcluded in this step, suggesting that this loss is unlikely.

Theoretically, approach 1 could not detect any DP genedownregulated in cDP cells to a level similar to that in Fibros. In

truth, cDP cells exhibited a global gene expression patternresembling that of Fibros (Fig. 1G), indicating the need foradditional analyses. Although approach 2 identified as many as 77

DP signature genes, excluding those expressed in contaminants inmicrodissected DP, some DP signature genes likely remainedundetected because of the limitation intrinsic to this approach that

not all DP signature genes were upregulated by cell aggregation.Indeed, 12 established DP genes (Botchkarev and Kishimoto,2003; Rendl et al., 2005; Ito et al., 2007; Osada et al., 2007;Driskell et al., 2009; Ohyama et al., 2010) were upregulated in

fDP compared with cDP cells and Fibros, but were not detectedby approach 2. Importantly, half of those genes were selected byanother analysis and finally listed as DP signature genes. This

result, together with the observation that only four genes werecommon to both analyses, suggests that the combination ofindividual approaches was beneficial.

The current study identified 118 human DP signature genes,

whereas global gene expression profiling of FACS-sorted puremouse DP cells returned 225 DP signature genes (Rendl et al.,2005), most of which overlapped with the genes recently

identified in CD133+ mouse DP cells (Driskell et al., 2009).Thus, a considerable number of human DP signature genes werenot detected in the present study. Our two-step criteria were

sufficiently stringent to identify selective markers, but may havebeen too restrictive to fully unveil the molecular signature oftested populations. Successful genetic fluorescent labeling of

living human hair follicle cells through specific promoterexpression has been reported (Tiede et al., 2009). A similarapproach utilizing a promoter for a DP signature gene identifiedin this study or the use of potential cell surface markers described

below might enable the isolation of more homogenous human DPcell samples and allow for the detection of unidentified DP genes.

In addition to the elucidation of the roles of WNT, BMP, and

FGF signaling in the maintenance of DP properties, thebioinformatics analysis discovered attractive gene groupsimplying future applications. For instance, the modulation of

enzyme activators, including APOE and RGS2, could augmentDP cell functions. Among transcription regulator genes, those

involved in cell differentiation, such as FOXO1 (Takano et al.,2007), GAS7 (Moorthy et al., 2005), and MEF2C (Lin et al.,1997), are of particular interest. The introduction of exogenoustranscription factors could reprogram the biological properties of

target cells (Takahashi and Yamanaka, 2006; Tsai et al., 2010;Tsai et al., 2011). Likewise, forced expression of these factorsmay facilitate the conversion of other dermal cells into cells with

DP-like properties. Membrane proteins, represented by GPR125

and LRP4, may provide useful cell surface markers for livinghuman DP cell isolation. Of note, Lrp4/corin is reportedly

specific for mouse DP cells (Enshell-Seijffers et al., 2008;Enshell-Seijffers et al., 2010). The development of antibodiesagainst these molecules for immunohistochemical- and/orfluorescence-activated cell sorting analyses could represent an

important next step. Interaction with extracellular matrix plays akey role in the maintenance of DP cell properties (Ohyama et al.,2010). Upregulation of DP biomarker was observed when DP

cells were cultured on laminin (Kobayashi et al., 2011). SPON1or MGP may provide a supportive environment for human DPcells in culture.

For the development of DPAC, we adopted BIO, rBMP, andbasic FGF to stimulate biological pathways downregulatedduring conventional culture. BIO is a small moleculecommonly used to activate canonical WNT signaling in

cultured cells (Sato et al., 2004; Yamauchi and Kurosaka,2009). Although BMP6 improved mouse DP cell properties moreefficiently than BMP2/BMP4 (Rendl et al., 2008), we did not

detect upregulation of BMP6 in human fDP. Based on theobservations that BMP2 was markedly suppressed duringconventional culture and upregulated during DP cell

aggregation, we selected this molecule as a BMP signalingactivator. bFGF upregulated some DP signature genes,suggesting that active FGF signaling contributes not only to

cell proliferation, but also to the maintenance of DP properties.As suggested by decreased expression of NOG, SOX2, andTRPS1 in dpacDP compared with that in fDP cells, dpacDP cellsprobably did not perfectly reproduce the molecular signature of

in situ DP. Once a pure human DP cell population is isolated,microarray comparison between intact DP and dpacDP cells mustbe performed to more precisely assess the ability of DPAC to

sustain DP properties. The findings obtained by this analysiscould enable further improvement of DPAC.

In previous studies, supplementation of exogenous Wnt or

Bmp molecules maintained mouse DP cell properties in culture,but did not affect their proliferative capacities (Rendl et al.,2008). For clinical application, the use of BIO may beadvantageous because Wnt is not widely available in large

quantities and may exhibit unwanted effects via non-canonicalpathways (Shimizu and Morgan, 2004). However, consideringthat DP growth suppression due to BIO (Yamauchi and

Kurosaka, 2009) represents a major drawback of DPAC, BIOmay be substituted by recombinant WNT3A (Kishimoto et al.,2000; Shimizu and Morgan, 2004) to maintain the intrinsic

properties and proliferative capacity of dpacDP cells. Becausepast reports suggested that activation of multiple biologicalpathways is necessary to fully maintain DP characteristics in

vitro (Kishimoto et al., 2000; Shimizu and Morgan, 2004; Rendlet al., 2008), DPAC could be more potentiated by the addition ofactivators of other signaling pathways, including the Sonic

Journal of Cell Science 125 (17)4122

Page 10: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

hedgehog, Notch, and Endothelin pathways (Gao et al., 2008;

Driskell et al., 2009; Hu et al., 2010). In contrast to DP cells,

fibroblasts cultured in DPAC reduced ALP expression (data not

shown). Thus, the conversion of other dermal cells into the DP

cell lineage using DPAC is not feasible.

A unique feature of DPAC is that it restored the DP signature

gene expression and functions normally reduced in conventionally

cultured DP cells. The gene expression recovery seems to be

imperfect by, for instance, moderate upregulation of RGS2. For

full assessment of functional restoration, additional examinations,

including the evaluation of the response to androgen (Inui et al.,

2002) or adenosine (Iino et al., 2007), may be performed.

However, the co-culture experiment and in vivo hair induction

assay demonstrated that the most characteristic DP property,

communication with the epithelial component (Ohyama et al.,

2010; Driskell et al., 2011), was at least partly restored by DPAC.

These observations imply a promising strategy for the efficient

propagation of functional human DP cells by primary expansion

under conventional culture conditions, followed by functional

restoration. In fact, the yield of DP cells after the second passage

was more than 10 times higher than that obtained through DPAC

expansion from primary cultures, and the time required was

approximately 50% less, indicating substantial advantages. The

phase of isolated DPs might have affected the efficiency of the

restoration. In the present study, DPs were mostly microdissected

from full-anagen hair follicles. The role of DP in such hair follicles

is to support hair shaft elongation rather than hair induction

(Oliver, 1967; Paus and Cotsarelis, 1999). Early-anagen DP cells

may possess more potent biological capacities, including

resilience, than late-anagen DP cells. Although the use of such

DP cells is technically very challenging, it could improve the

outcome of the restoration experiments.

In summary, we have elucidated the molecular signature of

intact human anagen DP cells. This guided the development of

DPAC for the efficient in vitro expansion of human DP cells that

retain their intrinsic properties. The present study sheds new light

on the biological properties of human DP cells and implies a

novel strategy for supplying sufficient functional human DP cells

for the development of molecules acting on in vivo DPs or for

regenerative medicine-based approaches to treat intractable hair

loss disorders.

Materials and MethodsPreparation of human dermal papillae

Human hair follicles were manually isolated from intact scalp skin obtained duringsurgical removal of benign skin tumors. DPs were microdissected from the bulbarportion of hair follicles under a dissection microscope as previously described(Williams et al., 1994). All experimental procedures were officially approved bythe Institutional Review Board of Keio University. All human donors providedwritten informed consent in accordance with Declaration of Helsinki guidelines.

Cell culture

Five or six DPs were placed on the bottom of a 60-mm dish and cultured in DMEM(Invitrogen, Carlsbad, CA, USA) containing penicillin and streptomycin (Roche,Basel, Switzerland) and 10% FBS. For fibroblast collection, the dermis of scalpsamples was cut into small blocks, 2–3 mm square. The blocks were attached to thebottom of 60-mm culture dishes and cultured in 10% FBS–DMEM. All cell culturewas performed at 37 C in 5% CO2. When confluent, primary cultured cells wereharvested and re-seeded (20,000 cells/dish). After primary culture, all cells werepassaged on a weekly basis.

Dermal papilla cell aggregation

Cultured DP cells were incubated with 2% trypsin and then transferred into low-cell-binding 96-well plates and centrifuged for several seconds to spin down theDP cells, and then incubated overnight at 37 C in 5% CO2.

RNA isolation and real-time PCR analysis

Total RNA was isolated from microdissected DP, DP cell aggregates and cultured

DP cells using an RNeasy Micro Kit (Qiagen, Hilden, Germany). For real-time

PCR analyses, cDNA was synthesized using SuperScript III First-Strand Synthesis

SuperMix for qRT-PCR (Invitrogen, Carlsbad, CA) according to the

manufacturer’s protocol. Real-time PCR analysis was performed using Power

SYBR Green PCR Master Mix (Applied Biosystems, Santa Clara, CA, USA) and a

StepOne Real-Time PCR system (Applied Biosystems). All primers were designed

using Primer ExpressH software (Applied Biosystems) and reactions wereperformed under the following cycling conditions: 10 min at 95 C, followed by

40 cycles of 15 s at 95 C and 60 s at 60 C. The primer sets used are shown in

supplementary material Table S5. Target mRNA expression levels normalized to

the GAPDH mRNA using the DCt method were calculated as 22DCt. GAPDH was

used as the reference gene because of its similar expression in microdissected and

cultured DP cell populations, as shown by consistent Ct values. Three independent

experiments were performed in duplicate. The results are presented as the means 6

standard error of the mean (s.e.m.).

Microarray generation

Isolated total RNA was amplified and biotin-labeled using a T7 primer based

GeneChipH Two-Cycle cDNA Synthesis Kit and an IVT Labeling Kit (Affymetrix

Inc., Santa Clara, CA, USA), respectively. Then, 20 mg of each labeled cRNA

sample was fragmented and hybridized to an Affymetrix HG-U133A 2.0 GeneChipmicroarray and scanned using GeneChip Scanner 3000 7G (Affymetrix Inc.)

according to the manufacturer’s protocol.

Microarray data analysis

To identify genes that are differentially expressed in intact human DP, two data

analyses individually consisting of two-step microarray comparisons wereperformed using Gene Chip Operating Software ver. 1.4 (Affymetrix Inc.). For

all microarrays, chip files (CHIP files) were generated according to the following

parameters: target signal scaling, 500; Alpha1, 0.05; Alpha2, 0.065; and Tau,

0.015. Gene expression files were generated based on the following parameters:

Gamma1H, 0.0045; Gamma1L, 0.0045; Gamma2H, 0.006; Gamma2L, 0.006; and

Perturbation, 1.1. Only probe sets detected as being expressed (present call) in

these files were analyzed. In the first analysis, global gene expression for the cDP

and Fibro chips was compared (n54), and probe sets whose targets wereupregulated $2-fold in cDPs were selected. Next, the expression levels of the

targets for these probe sets in fDP and cDP GeneChips were compared in order to

identify genes whose expression was upregulated in fDP or equally expressed in

fDP and cDPs. In the second analysis, two sets of GeneChips were used to analyze

total RNA samples from agDPs and cDPs in order to identify probe sets whose

targets were .1.5-fold upregulated by DP cell aggregation. Among the probe sets

upregulated in agDPs, those found to be .3-fold more highly expressed in fDP, as

compared to both cDPs and Fibros, were selected. The Affymetrix IDs of these

probe sets were uploaded into the gene batch viewer of DAVID (http://david.abcc.ncifcrf.gov/) (Huang et al., 2009a; Huang et al., 2009b) to generate a list of genes

whose expression is upregulated in intact human DP.

Bioinformatics analysis

The list of DP human signature genes was uploaded in DAVID, and the geneclustering analysis was performed with the following settings: similarity term

overlap, 3; similarity threshold, 0.25; initial group membership, 2; final group

membership, 2; and multiple linkage threshold, 0.50. Ontological functional

annotation for each gene was obtained by DAVID. In addition, gene expression

profiles for approaches 1 and 2 were analyzed by GeneSpring GX 11.5 software

(Agilent Technologies, Santa Clara, CA, USA). The MAS without baseline

transformation was applied for normalization. The genes were analyzed by

hierarchical clustering and drawn as heat maps using the GeneSpring default

setting.

Development of DPAC medium

DP cells were cultured in 10% FBS–DMEM containing BIO (1, 2, or 4 mM;

Sigma, St. Louis, MO, USA) rBMP2 (100, 200, or 400 ng/ml; R&D,

Minneapolis, MN, USA), or bFGF (10, 20, or 40 ng/ml; PEPROTECH, RockyHill, NJ, USA). The growth rates of DP cells under these different culture

conditions were monitored every 4–5 days by assessing the ratio between the size

of a starting DP and an outgrowing colony. Four weeks later, DP cells were

collected and total RNA was extracted. Then, ALPL, NOG, LEF1, and RGS2

expression levels were measured by real-time PCR as described above.

Subsequently, DP cells were cultured in 10% FBS–DMEM containing two

activators (1 mM BIO and 200 ng/ml rBMP, 1 mM BIO and 20 ng/ml FGF, or

200 ng/ml rBMP and 20 ng/ml FGF) or all three activators (1 mM BIO, 200 ng/

ml rBMP, and 20 ng/ml FGF). DP cell growth and signature gene expressionwere then assessed as described above.

Restoration of dermal papilla 4123

Page 11: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

Alkaline phosphatase activity detection

Detection of ALP activity in DP cells was performed using an ALP detection kit

(Chemicon Millipore, Temecula, CA, USA), according to the manufacturer’s

protocol.

Keratinocyte and DP co-culture

DP cells primarily cultured in 10% FBS–DMEM were seeded on the upper

compartment of 6 well Transwell culture dishes (Corning, Lowell, MA, USA) and

cultured either in DPAC or 10% FBS–DMEM for 1 week. At the same time,human middle follicle outer root sheath keratinocytes were cultured on the

collagen type I coated six-well plates in defined keratinocyte serum free medium

(Invitrogen, Carlsbad, CA, USA). At day 7, Transwell dishes with DP cells were

inserted into the plates with keratinocytes and cultured for additional 2 days inDMEM without FBS. Then, total RNA was isolated from keratinocytes for gene

expression analyses.

In vivo hair induction assay

Freshly isolated human DP cells were conventionally cultured in 10% FBS–

DMEM and passaged either in DPAC or 10% FBS–DMEM. After 4 weeks in

culture, aggregates of DPAC-treated and control DP cells were generated as

described above. Afollicular footpads were surgically removed from 8-week-old

ICR mice under a dissection microscope and incubated with dispase I (GodoSyusei, Tokyo, Japan; 1500 U/ml) for 30 min at 37 C. The epidermis was

separated from the dermis and cell aggregates were placed between them. The

footpad skin composites were incubated on Millicell-CM culture plate inserts

(Millipore, Billerica MA, USA) for 2 h at 37 C in 10% FBS–DMEM and graftedsubcutaneously into BALB/cnu/nu nude mice. Skin samples were harvested 2 weeks

later for histological analysis.

Human vimentin detection

Frozen sections were fixed through incubation with acetone for 20 min at 220 C

and incubated overnight at 4 C with a monoclonal anti-human vimentin-Cy3

antibody (Sigma). Cell nuclei were counterstained with 49,6-diamidino-2-phenylindole.

Statistical analysis

Statistically significant differences were identified by two-sided Student’s t-test orMann–Whitney U-test, depending on the size of the sample population. A P-value

of less than 0.05 was considered significant.

AcknowledgementsWe thank the late Jonathan C. Vogel (Dermatology Branch, CCR,National Cancer Institute, NIH, Bethesda, USA) for his technicaladvice and discussion.

FundingThis work was supported by a Grant-in-Aid for Scientific Researchfrom the Ministry of Education, Culture, Sports, Science andTechnology of Japan [grant number 18390315 to M.O.]; a JapaneseSociety for Investigative Dermatology Fellowship Shiseido Award2008 [to M.O.]; Keio University Research Grants for Life Sciencesand Medicine [to M.O.]; and Keio Gakuji Academic DevelopmentFunds [to M.O.].

Supplementary material available online at

http://jcs.biologists.org/lookup/suppl/doi:10.1242/jcs.105700/-/DC1

ReferencesBotchkarev, V. A. and Kishimoto, J. (2003). Molecular control of epithelial-

mesenchymal interactions during hair follicle cycling. J. Investig. Dermatol. Symp.

Proc. 8, 46-55.

de Almeida, H., Jr, Zigrino, P., Muller, F., Krieg, T., Korge, B. and Mauch, C.

(2005). Human scalp dermal papilla and fibrous sheath cells have a different

expression profile of matrix metalloproteinases in vitro when compared to scalp

dermal fibroblasts. Arch. Dermatol. Res. 297, 121-126.

Driskell, R. R., Giangreco, A., Jensen, K. B., Mulder, K. W. and Watt, F. M. (2009).

Sox2-positive dermal papilla cells specify hair follicle type in mammalian epidermis.

Development 136, 2815-2823.

Driskell, R. R., Clavel, C., Rendl, M. and Watt, F. M. (2011). Hair follicle dermal

papilla cells at a glance. J. Cell Sci. 124, 1179-1182.

Enshell-Seijffers, D., Lindon, C. and Morgan, B. A. (2008). The serine protease Corin

is a novel modifier of the Agouti pathway. Development 135, 217-225.

Enshell-Seijffers, D., Lindon, C., Kashiwagi, M. and Morgan, B. A. (2010). beta-catenin activity in the dermal papilla regulates morphogenesis and regeneration ofhair. Dev. Cell 18, 633-642.

Furthauer, M., Reifers, F., Brand, M., Thisse, B. and Thisse, C. (2001). sprouty4 actsin vivo as a feedback-induced antagonist of FGF signaling in zebrafish. Development

128, 2175-2186.

Gao, J., DeRouen, M. C., Chen, C. H., Nguyen, M., Nguyen, N. T., Ido, H., Harada,

K., Sekiguchi, K., Morgan, B. A., Miner, J. H. et al. (2008). Laminin-511 is anepithelial message promoting dermal papilla development and function during earlyhair morphogenesis. Genes Dev. 22, 2111-2124.

Higgins, C. A., Richardson, G. D., Ferdinando, D., Westgate, G. E. and Jahoda,

C. A. (2010). Modelling the hair follicle dermal papilla using spheroid cell cultures.Exp. Dermatol. 19, 546-548.

Horne, K. A., Jahoda, C. A. and Oliver, R. F. (1986). Whisker growth induced byimplantation of cultured vibrissa dermal papilla cells in the adult rat. J. Embryol. Exp.

Morphol. 97, 111-124.

Hu, B., Lefort, K., Qiu, W., Nguyen, B. C., Rajaram, R. D., Castillo, E., He, F.,

Chen, Y., Angel, P., Brisken, C. et al. (2010). Control of hair follicle cell fate byunderlying mesenchyme through a CSL-Wnt5a-FoxN1 regulatory axis. Genes Dev.

24, 1519-1532.

Huang, da. W., Sherman, B. T. and Lempicki, R. A. (2009a). Bioinformaticsenrichment tools: paths toward the comprehensive functional analysis of large genelists. Nucleic Acids Res. 37, 1-13.

Huang, da. W., Sherman, B. T. and Lempicki, R. A. (2009b). Systematic andintegrative analysis of large gene lists using DAVID bioinformatics resources. Nat.

Protoc. 4, 44-57.

Iino, M., Ehama, R., Nakazawa, Y., Iwabuchi, T., Ogo, M., Tajima, M. and Arase,

S. (2007). Adenosine stimulates fibroblast growth factor-7 gene expression viaadenosine A2b receptor signaling in dermal papilla cells. J. Invest. Dermatol. 127,1318-1325.

Inamatsu, M., Matsuzaki, T., Iwanari, H. and Yoshizato, K. (1998). Establishment ofrat dermal papilla cell lines that sustain the potency to induce hair follicles fromafollicular skin. J. Invest. Dermatol. 111, 767-775.

Inui, S., Fukuzato, Y., Nakajima, T., Yoshikawa, K. and Itami, S. (2002). Androgen-inducible TGF-beta1 from balding dermal papilla cells inhibits epithelial cell growth:a clue to understand paradoxical effects of androgen on human hair growth. FASEB J.

16, 1967-1969.

Inui, S., Fukuzato, Y., Nakajima, T., Yoshikawa, K. and Itami, S. (2003).Identification of androgen-inducible TGF-beta1 derived from dermal papilla cellsas a key mediator in androgenetic alopecia. J. Investig. Dermatol. Symp. Proc. 8, 69-71.

Ito, Y., Hamazaki, T. S., Ohnuma, K., Tamaki, K., Asashima, M. and Okochi, H.

(2007). Isolation of murine hair-inducing cells using the cell surface markerprominin-1/CD133. J. Invest. Dermatol. 127, 1052-1060.

Jahoda, C. A. and Reynolds, A. J. (1996). Dermal-epidermal interactions. Adultfollicle-derived cell populations and hair growth. Dermatol. Clin. 14, 573-583.

Jahoda, C. A., Horne, K. A. and Oliver, R. F. (1984). Induction of hair growth byimplantation of cultured dermal papilla cells. Nature 311, 560-562.

Khanna, S., Cheng, G., Gong, B., Mustari, M. J. and Porter, J. D. (2004). Genome-wide transcriptional profiles are consistent with functional specialization of theextraocular muscle layers. Invest. Ophthalmol. Vis. Sci. 45, 3055-3066.

Kishimoto, J., Ehama, R., Wu, L., Jiang, S., Jiang, N. and Burgeson, R. E. (1999).Selective activation of the versican promoter by epithelial- mesenchymal interactionsduring hair follicle development. Proc. Natl. Acad. Sci. USA 96, 7336-7341.

Kishimoto, J., Burgeson, R. E. and Morgan, B. A. (2000). Wnt signaling maintains thehair-inducing activity of the dermal papilla. Genes Dev. 14, 1181-1185.

Kobayashi, T., Fujisawa, A., Amagai, M., Iwasaki, T. and Ohyama, M. (2011).Molecular biological and immunohistological characterization of canine dermalpapilla cells and the evaluation of culture conditions. Vet. Dermatol. 22, 414-422.

Lichti, U., Weinberg, W. C., Goodman, L., Ledbetter, S., Dooley, T., Morgan, D.and Yuspa, S. H. (1993). In vivo regulation of murine hair growth: insights fromgrafting defined cell populations onto nude mice. J. Invest. Dermatol. 101, 124S-129S.

Lin, Q., Schwarz, J., Bucana, C. and Olson, E. N. (1997). Control of mouse cardiacmorphogenesis and myogenesis by transcription factor MEF2C. Science 276, 1404-1407.

Magerl, M., Kauser, S., Paus, R. and Tobin, D. J. (2002). Simple and rapid method toisolate and culture follicular papillae from human scalp hair follicles. Exp. Dermatol.

11, 381-385.

McElwee, K. J., Kissling, S., Wenzel, E., Huth, A. and Hoffmann, R. (2003).Cultured peribulbar dermal sheath cells can induce hair follicle development andcontribute to the dermal sheath and dermal papilla. J. Invest. Dermatol. 121, 1267-1275.

Messenger, A. G. (1984). The culture of dermal papilla cells from human hair follicles.Br. J. Dermatol. 110, 685-689.

Millar, S. E. (2002). Molecular mechanisms regulating hair follicle development. J.

Invest. Dermatol. 118, 216-225.

Momeni, P., Glockner, G., Schmidt, O., von Holtum, D., Albrecht, B., Gillessen-Kaesbach, G., Hennekam, R., Meinecke, P., Zabel, B., Rosenthal, A. et al. (2000).Mutations in a new gene, encoding a zinc-finger protein, cause tricho-rhino-phalangeal syndrome type I. Nat. Genet. 24, 71-74.

Moorthy, P. P., Kumar, A. A. and Devaraj, H. (2005). Expression of the Gas7 geneand Oct4 in embryonic stem cells of mice. Stem Cells Dev. 14, 664-670.

Journal of Cell Science 125 (17)4124

Page 12: Restoration of the intrinsic properties of human …...hypothesized that culture conditions sustaining the intrinsic molecular signature of the human DP could facilitate expansion

Journ

alof

Cell

Scie

nce

O’Shaughnessy, R. F., Yeo, W., Gautier, J., Jahoda, C. A. and Christiano, A. M.

(2004). The WNT signalling modulator, Wise, is expressed in an interaction-dependent manner during hair-follicle cycling. J. Invest. Dermatol. 123, 613-621.

Ohyama, M., Terunuma, A., Tock, C. L., Radonovich, M. F., Pise-Masison, C. A.,

Hopping, S. B., Brady, J. N., Udey, M. C. and Vogel, J. C. (2006). Characterizationand isolation of stem cell-enriched human hair follicle bulge cells. J. Clin. Invest. 116,249-260.

Ohyama, M., Zheng, Y., Paus, R. and Stenn, K. S. (2010). The mesenchymalcomponent of hair follicle neogenesis: background, methods and molecularcharacterization. Exp. Dermatol. 19, 89-99.

Oliver, R. F. (1967). The experimental induction of whisker growth in the hooded rat byimplantation of dermal papillae. J. Embryol. Exp. Morphol. 18, 43-51.

Oliver, R. F. (1991). Dermal-epidermal interactions and hair growth. J. Invest.

Dermatol. 96, 76S.

Osada, A. and Kobayashi, K. (2000). Appearance of hair follicle-induciblemesenchymal cells in the rat embryo. Dev. Growth Differ. 42, 19-27.

Osada, A., Iwabuchi, T., Kishimoto, J., Hamazaki, T. S. and Okochi, H. (2007).Long-term culture of mouse vibrissal dermal papilla cells and de novo hair follicleinduction. Tissue Eng. 13, 975-982.

Osada, A., Kobayashi, K., Masui, S., Hamazaki, T. S., Yasuda, K. and Okochi, H.

(2009). Cloned cells from the murine dermal papilla have hair-inducing ability. J.

Dermatol. Sci. 54, 129-131.

Panteleyev, A. A., Mitchell, P. J., Paus, R. and Christiano, A. M. (2003). Expressionpatterns of the transcription factor AP-2alpha during hair follicle morphogenesis andcycling. J. Invest. Dermatol. 121, 13-19.

Park, S. Y., Kwack, M. H., Chung, E. J., Im, S. U., Han, I. S., Kim, M. K., Kim, J. C.

and Sung, Y. K. (2007). Establishment of SV40T-transformed human dermal papillacells and identification of dihydrotestosterone-regulated genes by cDNA microarray.J. Dermatol. Sci. 47, 201-208.

Paus, R. and Cotsarelis, G. (1999). The biology of hair follicles. N. Engl. J. Med. 341,491-497.

Pflieger, D., Chabane, S., Gaillard, O., Bernard, B. A., Ducoroy, P., Rossier, J. and

Vinh, J. (2006). Comparative proteomic analysis of extracellular matrix proteinssecreted by two types of skin fibroblasts. Proteomics 6, 5868-5879.

Rendl, M., Lewis, L. and Fuchs, E. (2005). Molecular dissection of mesenchymal-epithelial interactions in the hair follicle. PLoS Biol. 3, e331.

Rendl, M., Polak, L. and Fuchs, E. (2008). BMP signaling in dermal papilla cells isrequired for their hair follicle-inductive properties. Genes Dev. 22, 543-557.

Reynolds, A. J. and Jahoda, C. A. (1996). Hair matrix germinative epidermal cellsconfer follicle-inducing capabilities on dermal sheath and high passage papilla cells.Development 122, 3085-3094.

Sato, N., Meijer, L., Skaltsounis, L., Greengard, P. and Brivanlou, A. H. (2004).Maintenance of pluripotency in human and mouse embryonic stem cells throughactivation of Wnt signaling by a pharmacological GSK-3-specific inhibitor. Nat. Med.

10, 55-63.

Seitz, R., Hackl, S., Seibuchner, T., Tamm, E. R. and Ohlmann, A. (2010). Norrinmediates neuroprotective effects on retinal ganglion cells via activation of the Wnt/beta-catenin signaling pathway and the induction of neuroprotective growth factors inMuller cells. J. Neurosci. 30, 5998-6010.

Sekiya, T., Adachi, S., Kohu, K., Yamada, T., Higuchi, O., Furukawa, Y.,

Nakamura, Y., Nakamura, T., Tashiro, K., Kuhara, S. et al. (2004). Identificationof BMP and activin membrane-bound inhibitor (BAMBI), an inhibitor of

transforming growth factor-beta signaling, as a target of the beta-catenin pathwayin colorectal tumor cells. J. Biol. Chem. 279, 6840-6846.

Shimizu, H. and Morgan, B. A. (2004). Wnt signaling through the beta-cateninpathway is sufficient to maintain, but not restore, anagen-phase characteristics ofdermal papilla cells. J. Invest. Dermatol. 122, 239-245.

Shin, H., Kwack, M. H., Shin, S. H., Oh, J. W., Kang, B. M., Kim, A. A., Kim, J.,Kim, M. K., Kim, J. C. and Sung, Y. K. (2010). Identification of transcriptionaltargets of Wnt/beta-catenin signaling in dermal papilla cells of human scalp hairfollicles: EP2 is a novel transcriptional target of Wnt3a. J. Dermatol. Sci. 58, 91-96.

Sleeman, M. A., Murison, J. G., Strachan, L., Kumble, K., Glenn, M. P., McGrath,

A., Grierson, A., Havukkala, I., Tan, P. L. and Watson, J. D. (2000). Geneexpression in rat dermal papilla cells: analysis of 2529 ESTs. Genomics 69, 214-224.

Stenn, K. S. and Paus, R. (2001). Controls of hair follicle cycling. Physiol. Rev. 81,449-494.

Takahashi, K. and Yamanaka, S. (2006). Induction of pluripotent stem cells frommouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663-676.

Takano, M., Lu, Z., Goto, T., Fusi, L., Higham, J., Francis, J., Withey, A., Hardt, J.,

Cloke, B., Stavropoulou, A. V. et al. (2007). Transcriptional cross talk between theforkhead transcription factor forkhead box O1A and the progesterone receptorcoordinates cell cycle regulation and differentiation in human endometrial stromalcells. Mol. Endocrinol. 21, 2334-2349.

Tiede, S., Koop, N., Kloepper, J. E., Fassler, R. and Paus, R. (2009). Nonviral in situgreen fluorescent protein labeling and culture of primary, adult human hair follicleepithelial progenitor cells. Stem Cells 27, 2793-2803.

Tsai, S. Y., Clavel, C., Kim, S., Ang, Y. S., Grisanti, L., Lee, D. F., Kelley, K. andRendl, M. (2010). Oct4 and klf4 reprogram dermal papilla cells into inducedpluripotent stem cells. Stem Cells 28, 221-228.

Tsai, S. Y., Bouwman, B. A., Ang, Y. S., Kim, S. J., Lee, D. F., Lemischka, I. R. and

Rendl, M. (2011). Single transcription factor reprogramming of hair follicle dermalpapilla cells to induced pluripotent stem cells. Stem Cells 29, 964-971.

Weinberg, W. C., Goodman, L. V., George, C., Morgan, D. L., Ledbetter, S., Yuspa,

S. H. and Lichti, U. (1993). Reconstitution of hair follicle development in vivo:determination of follicle formation, hair growth, and hair quality by dermal cells.J. Invest. Dermatol. 100, 229-236.

Williams, D., Profeta, K. and Stenn, K. S. (1994). Isolation and culture of follicularpapillae from murine vibrissae: an introductory approach. Br. J. Dermatol. 130, 290-297.

Wu, J. J., Liu, R. Q., Lu, Y. G., Zhu, T. Y., Cheng, B. and Men, X. (2005). Enzymedigestion to isolate and culture human scalp dermal papilla cells: a more efficientmethod. Arch. Dermatol. Res. 297, 60-67.

Yamauchi, K. and Kurosaka, A. (2009). Inhibition of glycogen synthase kinase-3enhances the expression of alkaline phosphatase and insulin-like growth factor-1 inhuman primary dermal papilla cell culture and maintains mouse hair bulbs in organculture. Arch. Dermatol. Res. 301, 357-365.

Yang, C. C. and Cotsarelis, G. (2010). Review of hair follicle dermal cells. J.

Dermatol. Sci. 57, 2-11.Young, T. H., Lee, C. Y., Chiu, H. C., Hsu, C. J. and Lin, S. J. (2008). Self-assembly

of dermal papilla cells into inductive spheroidal microtissues on poly(ethylene-co-vinyl alcohol) membranes for hair follicle regeneration. Biomaterials 29, 3521-3530.

Young, T. H., Tu, H. R., Chan, C. C., Huang, Y. C., Yen, M. H., Cheng, N. C., Chiu,

H. C. and Lin, S. J. (2009). The enhancement of dermal papilla cell aggregation byextracellular matrix proteins through effects on cell-substratum adhesivity and cellmotility. Biomaterials 30, 5031-5040.

Restoration of dermal papilla 4125