A single-cell transcriptome atlas during Cashmere goat ...morphogenesis mainly used the mouse as a...

38
A single-cell transcriptome atlas during Cashmere goat hair follicle morphogenesis Wei Ge a , Wei-Dong Zhang a , Yue-Lang Zhang a , Yu-Jie Zheng a , Fang Li a , Shan-He Wang a , Jin-Wang Liu c , Shao-Jing Tan b , Zi-Hui Yan b , Lu Wang b , Wei Shen b , Lei Qu c , Xin Wang a a Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 712100, China; b College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China; c Life Science Research Center, Yulin University, Yulin, Shaanxi 719000, China ¶ Correspondence and reprint requests to: Prof. Xin Wang; E-mail: [email protected] . CC-BY-NC-ND 4.0 International license (which was not certified by peer review) is the author/funder. It is made available under a The copyright holder for this preprint this version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287 doi: bioRxiv preprint

Transcript of A single-cell transcriptome atlas during Cashmere goat ...morphogenesis mainly used the mouse as a...

  • A single-cell transcriptome atlas during Cashmere goat hair 1

    follicle morphogenesis 2

    Wei Ge a, Wei-Dong Zhang a, Yue-Lang Zhang a, Yu-Jie Zhenga, Fang Li a, Shan-He Wang a, 3

    Jin-Wang Liu c, Shao-Jing Tan b, Zi-Hui Yan b, Lu Wang b, Wei Shen b, Lei Qu c, Xin Wang a 4

    5

    a Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province,6

    College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi 7

    712100, China; 8

    b College of Life Sciences, Qingdao Agricultural University, Qingdao 266109, China; 9

    c Life Science Research Center, Yulin University, Yulin, Shaanxi 719000, China 10

    11

    12

    ¶ Correspondence and reprint requests to: 13

    Prof. Xin Wang; E-mail: [email protected] 14

    15

    16

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Abstract: 17

    Cashmere, also known as soft gold, is produced from the secondary hair follicles in 18

    Cashmere goats and it’s therefore of significance to investigate the molecular profiles 19

    during Cashmere goat hair follicle development. However, our current understanding of the 20

    machinery underlying Cashmere goat hair follicle remains largely unexplored and 21

    researches regarding hair follicle development mainly used the mouse as a research model. 22

    To provides comprehensively understanding on the cellular heterogeneity and cell lineage 23

    cell fate decisions, we performed single-cell RNA sequencing on 19,705 single cells from 24

    induction (embryonic day 60), organogenesis (embryonic day 90) and cytodifferentiation 25

    (embryonic day 120) stages of fetus Cashmere goat dorsal skin. Unsupervised clustering 26

    analysis identified 16 cell clusters and their corresponding cell types were also 27

    unprecedentedly characterized. Based on the lineage inference, we revealed detailed 28

    molecular landscape along the dermal and epidermal cell lineage developmental pathways. 29

    Notably, by cross-species comparasion of single cell data with murine model, we revelaed 30

    conserved programs during dermal condensate fate commitment and the heterochrony 31

    development of hair follicle development between mouse and Cashmere goat were also 32

    discussed here. Our work here delineate unparalleled molecular profiles of different cell 33

    populations during Cashmere goat hair follicle morphogenesis and provide a valuable 34

    resource for identifying biomarkers during Cashmere goat hair follicle development. 35

    36

    Key Words: Single-cell transcriptome; Developmental trajectories; Hair follicle 37

    morphogenesis 38

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Introduction 39

    Every year, more than 20,000 tons of Cashmere were generated in China and Cashmere 40

    goat has become an important source of income for the people lived in north China (Scott 41

    Waldron et al., 2014). Cashmere is the secondary hair follicles in Cashmere goats and forms 42

    as early as the fetus stage and due to the commercial value of Cashmere (Ansari-Renani et 43

    al., 2011, Geng et al., 2013), it’s therefore of great significance to reveal molecular 44

    pathways during early hair follicle development in Cashmere goats. Besides, by using 45

    mouse as a research model, research has demonstrated that molecular pathways during 46

    early hair follicle morphogenesis play important roles in regulating the hair characteristics, 47

    including hair fiber length, fineness and curvature (Duverger & Morasso, 2009), therefore, 48

    further enhancing the significance of revealing the molecular pathways driving hair follicle 49

    morphogenesis in Cashmere. However, due to the long-time duration of pregnancy (about 50

    145~159 days) in Cashmere goats (AJ Ritar et al., 1989), researches focused on hair follicle 51

    morphogenesis mainly used the mouse as a research model, while our current 52

    understanding on hair follicle morphogenesis in Cashmere goat remains largely unknown. 53

    Similar to murine hair follicle development, Cashmere goat hair follicle in uterus 54

    development can also be divided into three main stages: induction stage (about embryonic 55

    day 55 - 65), organogenesis (about embryonic day 85 - 95) and cytodifferentiation stages 56

    (around embryonic day 115) (Zhang Y et al., 2006). In mice, the molecular underpinnings 57

    underlying the induction stage has recently been comprehensively investigated by virtue of 58

    the development of single-cell RNA sequencing technology, while the late two stages 59

    remain not well-known (Saxena et al., 2019). According to what is known in mice, the 60

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • formation of placodes and dermal condensates (DC) are two marking events in the 61

    induction stage and requires a conserved crosstalk between dermal and epidermal cell 62

    populations, including Wnt/β-catenin signaling, Edar signaling and Fgf signaling (Chen et 63

    al., 2012, Huh et al., 2013, Zhang et al., 2009). More recently, Mok et al., demonstrated that 64

    murine DC formation can be further divided into three sub-stages using single-cell RNA 65

    sequencing (scRNA seq) technology and characterized detailed transcriptome signature 66

    genes during each substage, they also found that ECM/Adhesion signaling was vital for the 67

    early DC fate commitment (Mok et al., 2019). The organogenesis stage is characterized by 68

    the formation of dermal papilla (DP) cells, hair shaft and inner root sheath (IRS) and the 69

    molecular pathways involved includes PDGFα signaling and Shh signaling (Karlsson et al., 70

    1999, Ouspenskaia et al., 2016). For the cytodifferentiation stage, the differentiation of IRS, 71

    hair shaft and keratinocyte become obvious and Eda signaling is demonstrated to play a 72

    role (Duverger & Morasso, 2009, Millar, 2002). Noteworthy, the asynchronous 73

    development of different hair follicles, including guard hair follicles (starts from E13.5), 74

    awl, auchene hair follicles (starts from E15.5), and zigzag hair follicle (starts from E17.5) 75

    in mice, is also a marking event during the cytodifferentiation stage (Schlake, 2007). 76

    However, the molecular machinery underlying the asynchronous development of different 77

    hair follicles remains not well-known (Chi et al., 2013, Driskell et al., 2009). 78

    To preliminarily reveal molecular pathways involved during Cashmere goat hair 79

    follicle morphogenesis, several groups have collected skin samples from fetus goat and 80

    performed transcriptome sequencing analysis to reveal gene expression dynamics between 81

    different time points (Gao et al., 2016, Ren et al., 2016). However, due to the lack of 82

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • conserved markers to label particular cell types within the hair follicles, most studies used 83

    skin tissues to perform transcriptome sequencing analysis and generated the “equalized” 84

    expression matrices, which is sometimes, hard to reveal the real scenario. The paucity of 85

    information regarding the cell heterogeneity within the hair follicles has obviously become 86

    the main obstacle in dissecting the hair follicle morphogenesis. scRNA seq has recently 87

    became robust tool in dissecting cell heterogeneity and several groups have also 88

    successfully used scRNA seq technology to reveal the molecular machinery underlying 89

    murine hair follicle development (Ge et al., 2019, Gupta et al., 2019, Mok et al., 2019), 90

    further emphasized its application prospect in hair follicle development-related researches. 91

    Tackling the paucity of information regarding the cellular heterogeneity and molecular 92

    pathway underlying key cell fate decisions during Cashmere hair follicle development. 93

    Here, we reported a single-cell transcriptome landscape during Cashmere goat hair follicle 94

    morphogenesis based on 19,705 single-cell transcriptional profiles. We successfully 95

    identified different cell types during Cashmere goat hair follicle development and 96

    delineated their cell type-specific gene expression profiles, which provides valuable 97

    information for the identification of biomarkers and dissecting cellular heterogeneity during 98

    Cashmere goat hair follicle development. Besides, cell lineage inference analysis provides a 99

    comprehensively understanding of the molecular pathways underlying major cell lineage 100

    fate decisions. Our study here provides a valuable resource for understanding Cashmere 101

    goat hair follicle development, and will also have implications for future Cashmere goat 102

    breeding. 103

    104

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Results 105

    Single-cell sequencing and characterization of cellular heterogeneity during Cashmere 106

    goat hair follicle morphogenesis 107

    To provide in-depth insight into molecular profiles during Cashmere goat hair follicle 108

    development and main cell fates transitions, we collected skin samples from E60, E90 and 109

    E120 stage fetus Cashmere goat skin (Supplemental. Fig. S1a), which correspond to hair 110

    follicle induction, organogenesis and cytodifferentiation stage and performed single-cell 111

    RNA sequencing (Fig. 1a). We totally captured 7,000 single cells for each sample, and for 112

    each sample we detected at least 16,000 genes and the genome mapping rate was higher 113

    than 90% for all the samples (Supplemental. Fig. S1b). For quality control, we filtered cells 114

    according to the number of genes detected (Supplemental. Fig. S1c) and retained 115

    high-quality cells for downstream analysis. After quality control, we totally analyzed 116

    19,705 single-cell transcriptome expression profiles from E60 (6,825 single cells), E90 117

    (6,873 single cells) and E120 (6,007 single cells) stage fetus Cashmere goat back skin. 118

    To dissect cellular heterogeneity, we next performed t-distributed stochastic neighbor 119

    embedding (t-SNE) analysis and we totally identified 16 different cell clusters across three 120

    developmental times (Fig. 1b,c and Supplemental. Table 1). By analyzing cluster-specific 121

    expressed gene expression, we successfully identified the different cell types according to 122

    their marker gene expression (Supplemental. Fig. S1d). Briefly, we found that cluster 1, 4, 6, 123

    7 and 13 expressed high level of dermal cell lineage markers LUM and COL1A1 (Gupta et 124

    al., 2019), while cluster 0, 2, 3, 5, 8, 9 and 12 expressed high level of epithelial lineage 125

    markers KRT14 and KRT17 (Gu & Coulombe, 2007, Joost et al., 2016). Besides, we also 126

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • identified hair shaft clusters (LHX2 and MSX1, cluster 2, 5) (Yang et al., 2017), endothelial 127

    cluster (KDR and PECAM1, cluster 11) (Detmar et al., 1998), DP cluster (SOX2 and SOX18, 128

    cluster 13) (Driskell et al., 2009), pericyte cluster (ACTA2 and TPM2, cluster 15) 129

    (Paquet-Fifield et al., 2009), muscle cell cluster (CNMD and ARS1, cluster 17) and 130

    macrophage cell cluster (ALF1 and RGS1, cluster 16) (Lee et al., 2016). More importantly, 131

    we further delineated transcriptional characteristics for each cell types and identified a 132

    series of cell type-specific marker genes during Cashmere goat hair follicle development 133

    (Fig. 1d), and it was also worth noting that many cell type-specific expressed marker genes 134

    were also consistent with a murine scenario, such as dermal cell markers POSTN, DCN, 135

    APOE, epithelial cell markers KRT14, KRT15, and DP cell markers SOX2, SOX18. 136

    137

    Defining dermal cell lineage and epidermal cell lineage developmental trajectory 138

    along pseudoptime 139

    After the characterization of different cell clusters, we then want to investigate major cell 140

    fate transitions during hair follicle development. We, therefore, performed pseudotime 141

    trajectory construction analysis on dermal and epidermal cell clusters (Fig. 2). Since we 142

    have successfully characterized all cell clusters, we then selected dermal cell lineage cell 143

    clusters (Fig. 2a, cluster 1, 4, 6, 7 and 12) and epidermal cell lineage cell clusters (Fig. 2b, 144

    cluster 0, 2, 3, 5, 8, 9 and 11) to infer cell lineage developmental trajectory. For the dermal 145

    cell lineage, pseudotime trajectory displayed 2 branch points (Fig. 2c), while the epidermal 146

    cell lineage showed 3 branch points (Fig. 2d). Noteworthy, when the cells were color-coded 147

    with their corresponding developmental time, they also showed a time-ordered pattern 148

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • along the pseudotime. As for the branch point in the dermal and epidermal cell populations, 149

    based on the prior knowledge on cell lineage dynamics, during early hair follicle 150

    development, the dermal cell fate involves DC fate commitment and DP fate commitment 151

    (Fig. 2e) (Saxena et al., 2019), while epidermal cell fate involves matrix cell fate 152

    commitment, hair shaft/IRS fate commitment and keratinocyte fate commitment (Fig. 2f) 153

    (Forni et al., 2012, Millar, 2002, Schmidt-Ullrich & Paus, 2005), it’s therefore different 154

    branch points may represent the process of cell fate decisions. 155

    156

    Delineating developmental pathway during DC fate commitment 157

    After dermal cell lineage trajectory inference, we firstly focused on the first branch point on 158

    the dermal cell pseudotime trajectory to reveal the first dermal cell fate decision. By 159

    analyzing gene expression dynamics along pseudotime, we observed 2,679 differentially 160

    expressed genes at the end of cell fate commitment (Supplemental. Table 2) and gene 161

    functional enrichment analysis revealed that these genes enriched GO terms of “tissue 162

    morphogenesis, response to growth factor and cell morphogenesis involved in 163

    differentiation” (Fig. 3a). A comparison of overlapped GO terms between each gene set 164

    showed that they shared substantial GO terms (Supplemental. Fig. 2). Of particular notice, 165

    we observed a series of canonical murine dermal condensate cell markers such as APOD, 166

    LUM and APOE (Mok et al., 2019) in those differentially expressed genes, and the 167

    pseudotime expression pattern of DC markers APOD, SOX18, CTNNB1 and SOX2 was 168

    increased along pseudotime (Fig. 3b), therefore, we termed the first branch point as DC fate 169

    commitment. 170

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • To our knowledge, no reports yet have delineated cellular heterogeneity and 171

    transcriptional landscape during Cashmere goat hair follicle morphogenesis, and most 172

    researches regarding hair follicle development have been performed on the murine model. 173

    To gain in-depth insight into machinery driving DC fate commitment, we then compared 174

    DC fate signature genes with recently reported murine DC fate commitment signature 175

    genes (Fig. 3c) and observed 729 (about 14.5% of all) overlapped genes between Cashmere 176

    goat and murine DC signature genes (Supplemental. Table 3). Noteworthy, based on 177

    single-cell RNA sequencing on E15.0 dorsal skin, Mok et al. recently demonstrated that 178

    murine DC fate commitment can be divided into pre-DC, DC1 and DC2 stage at a more 179

    detailed level (Mok et al., 2019). By analyzing murine DC marker genes at different stages, 180

    we similarly found that DC signature genes of Cashmere goat also showed chronological 181

    expression patterns along pseudotime (Fig. 3d). Briefly, murine pre-DC markers DKK1 and 182

    LEF1 were elevated prior to DC1 and DC2 marker expressions, such as PRDM1, TRPS1, 183

    INHBA and RSPO3, it’s therefore plausible that DC fate commitment in Cashmere goat 184

    may also involve different stages. 185

    186

    Delineating DP cell heterogeneity along pseudotime 187

    After defining DC fate commitment, we then focused on the next branch point. We firstly 188

    compared differentially expressed gene expression between the two branches and observed 189

    that cell fate 1 elevated canonical DP marker genes, such as APOD, SOX18, and enriched 190

    GO terms of “tissue morphogenesis and epidermis development”, while cell fate 2 elevated 191

    genes such as CENPW, TOP2A and enriched GO terms of “mitotic cell cycle process and 192

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • DNA-dependent DNA replication” (Fig. 4a,b and Supplemental. Fig. 3a,b). To gain an 193

    in-depth understanding of the differences between the two branches, we further identified 194

    differentially expressed genes between the two branches using another differential analysis 195

    method (Wilcox, Wilcoxon Rank Sum test). Consistent with Monocle analysis, cell fate 1 196

    also showed higher expression of APOD, SOX18 and IGF1, while cell fate 2 showed 197

    elevated expression of CENPW, TOP2A and DCN (Fig. 4c). GO enrichment analysis 198

    similarly revealed that differentially expressed genes in cell fate 1 enriched GO terms of 199

    “tissue morphogenesis and epithelial cell differentiation”, while cell fate 2 enriched GO 200

    mainly related to the regulation of cell cycle (Supplemental. Fig. 3c). 201

    To dissect the heterogeneity within DP cells, we further performed 202

    immunofluorescence analysis on E120 Cashmere goat back skin tissues and observed 203

    different staining patterns between primary hair follicles (PF) and secondary hair follicles 204

    (SF) (Fig. 4d). For the BMP2 staining, we found that BMP2 specifically expressed in the 205

    matrix cells surrounding DP cells, while in the secondary hair follicle, BMP2 positive cells 206

    could be found in both DP cells and surrounding matrix cells. Besides, we found that 207

    PCNA, a cell cycle-related marker, was expressed mainly in surrounding matrix cells in the 208

    primary hair follicle, while in the secondary hair follicles, we observed high expression of 209

    PCNA both in DP cells and surrounding matrix cells. Taken together, our analysis here 210

    demonstrated that different hair follicles showed different gene expression patterns in 211

    Cashmere goat, which may also plausible for the asynchronous development of different 212

    hair follicles. 213

    214

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Delineating developmental pathway during the first epidermal cell fate decision 215

    After revealing dermal cell fate decisions, we then focused on the epidermal cell clusters. 216

    Monocle pseudotime trajectory inference analysis revealed that epidermal cells showed 217

    three different branch points. We then firstly focused on the first branch point and analyzed 218

    differentially expressed gene dynamics along pseudotime. Based on k-means clustering, we 219

    observed four distinct gene clusters. As expected, we observed a series matrix cell markers 220

    at the end of pseodotime, including HOXC13, KRT25 and KRT71, thus deciphering a 221

    matrix cell fate commitment (Fig. 5a). Gene functional enrichment analysis showed that 222

    matrix cell expressed signature genes enriched GO terms of “keratinocyte differentiation, 223

    epidermis development, and skin epidermis development” (Fig. 5b), while comparison of 224

    GO terms between different gene set reveals that gene set 1,2 differs to that of gene set 3,4 225

    (Supplemental. Fig. 4b). 226

    To gain in-depth insight into molecular profiles during matrix cell fate commitment, we 227

    further analyzed the signature gene expression pattern along pseudotime. For the gene set 4, 228

    namely matrix cell fate, they enriched a series of keratin family genes, such as KRT25, 229

    KRT27, KRT84, and their expression was increased along pseudotime (Fig. 5c). For the 230

    gene set 3 and 2, we found that they transiently elevated expression of LEF1, SBSN, SOX18, 231

    SOX9, and enriched GO terms of “cardiac epithelial to mesenchymal transition, 232

    mesenchymal cell differentiation and skin development, morphogenesis of an epithelium, 233

    respectively. For the gene set 1, they enriched genes such as VIM, LUM and COL1A1 and 234

    both showed decreased expression along pseudotime. Immunohistochemistry staining 235

    analysis also confirmed that LEF1 and CTNNB1 were expressed in the upper epidermis, 236

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • which was also consistent with a murine scenario (Polakis, 2001, Tsai et al., 2014). 237

    238

    Delineating cell fate decisions during hair shaft and IRS cell fate commitment 239

    After deciphering matrix cell fate commitment in the first epidermal trajectory point, we 240

    next focused on the next branch point. By analyzing differentially expressed genes along 241

    pseudotime, we found that cell fate 1 enriched canonical hair shaft markers, including SHH, 242

    VDR, and HOXC13, while cell fate 2 enriched canonical IRS markers, such as SOX9, 243

    KRT14, SBSN and LEF1 (Fig. 6a) (Yang et al., 2017). Our immunofluorescence results also 244

    confirmed their expression in the Cashmere hair follicles (Supplemental. Fig. 5a). For the 245

    pre-branch, we observed genes such as LUM, COL1A, OGN, SOX18 and they all showed 246

    decreased expression along pseudotime (Fig. 6b). For the hair shaft fate (cell fate 1), we 247

    also observed elevated expression of DCN, TOP2A, CENPW and H2AFZ and enriched GO 248

    terms of “mitotic cell cycle process and DNA replication” (Supplemental. Fig. 5 a,b). For 249

    the IRS cell fate (cell fate 2), we observed elevated expression of PRDM1, KRT1, SOX9, 250

    KRT14 and enriched GO terms of “supramolecular fiber organization and tissues 251

    morphogenesis”. 252

    Besides, we also compared our identified hair shaft and IRS signature genes with our 253

    recently identified murine hair shaft and IRS signature genes (Ge et al., 2019). Interestingly, 254

    we only observed about ~8.8% overlapped IRS signature genes between mouse and goat 255

    (Fig. 6c and Supplemental. Table 4), and these overlapped genes mainly consisted of 256

    Keratin family genes, such as KRT14, KRT17, KRT79. As for overlapped hair shaft 257

    signature genes, we only observed ~4.6% genes (Fig. 6d), including MSX1, HOXC13, 258

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • APOD, LHX2, SHH, and VDR. To further validate our analysis, we compared the 259

    expression of PCNA and VDR between E90 Cashmere goat skin and E16.5 mouse skin 260

    tissues, and the immunohistochemistry showed that they showed similar expression 261

    patterns during hair follicle development (Fig. 6e). These data together demonstrate that 262

    hair shaft and IRS specification may require a conserved program during cell fate decisions. 263

    264

    Revealing the developmental pathway during keratinocyte cell fate commitment 265

    After delineating the first two epidermal cell lineage cell fate decisions, we then focused on 266

    the last branch point. Pseudotime trajectory analysis also revealed two different branches, 267

    and we then analyzed pseudotime gene expression dynamics between the two branches (Fig. 268

    7a). Analyzing differentially expressed genes along psusotime, we found that cell fate 1 269

    enriched genes such as VDR, BMP4, STAR, KRT85 and KRT14, while cell fate 2 enriched 270

    genes such as BMP2, SHH, CUX1 and ETV5. Noteworthy, KRT14 has been identified as a 271

    marker for keratinocyte both in humans and mice (Green et al., 2003, Joost et al., 2016), we, 272

    therefore, termed this fate as keratinocytes. To gain in-depth insight into their 273

    corresponding cell type, we performed gene functional enrichment analysis for each gene 274

    set and analyzed pseudotime GO enrichment dynamics (Fig. 7b and Supplemental. Fig. 6a). 275

    The result showed that the pre-branch mainly enriched genes related to “regulation of 276

    response process”, while for the cell fate 1, the signature genes mainly enriched GO terms 277

    of “development differentiation of epidermal”. Interestingly, for cell fate 2, these 278

    differentially expressed genes mainly enriched in GO terms of “regulation of cell cycle” 279

    and showed lower expression of KRT14 (Fig. 7c), it’s therefore plausible that keratinocyte 280

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • differentiation at this stage is not synchronized. To confirm such hypothesis, we performed 281

    immunofluorescence analysis of VDR, PCNA, BMP2, CTNNB1 and LEF1, and the results 282

    showed that the expression of VDR and BMP2 in the outer layer epidermis was not 283

    homogeneous, with some cell clusters showed higher expression while some cell 284

    populations showed lower expression (Fig. 7d). For the pre-branch enriched CTNNB1, it 285

    was uniformly expressed in the interfollicular epidermis. Besides, the expression pattern of 286

    PCNA and LEF1 was even more significant and they were partially expressed in the 287

    epidermis. Taken together, these results together emphasize that keratinocyte differentiation 288

    in Cashmere goat is asynchronous and requires different gene expression profiles. 289

    290

    Discussion 291

    The development of scRNA seq in recent years has been demonstrated as a robust tool for 292

    the developmental biologist to investigate organogenesis and has provided us with 293

    unparalleled insight into mammalian development. In the past decades, the number of 294

    papers using scRNA seq based technology has increased exponentially and scRNA has also 295

    been awarded Science's 2018 Breakthrough of the Year (Angerer et al., 2017, Pennisi, 296

    2018). Here, we successfully constructed single-cell atlas during Cashmere goat hair 297

    follicle development. Based on the downstream analysis, we provided unparalleled insight 298

    into the cellular heterogeneity and major cell fate decisions during the Cashmere hair 299

    follicle in uterus morphogenesis. As far as we have known, this is the first study to 300

    comprehensively delineating molecular profiles of various cell types and revealing major 301

    cell fate decisions during Cashmere hair follicle development. More importantly, by 302

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • analyzing cluster-specific gene expression profiles, our data here provides a valuable 303

    resource for identification of markers for future studies in Cashmere skin tissues and also 304

    provides an in-depth understanding of the Cashmere hair follicle development. 305

    In the current study, we totally analyzed 19,705 single-cell transcriptional profiles from 306

    three different points, which can represent major cell types during hair follicle development. 307

    To dissect cellular heterogeneity during Cashmere goat hair follicle development after tSNE 308

    analysis, we analyzed cluster-specific expressed signature genes for each cluster to infer 309

    their corresponding cell types. It’s worth noting that all the cell markers used in the current 310

    study were referenced from the murine model due to the paucity of information regarding 311

    marker gene expression during Cashmere goat hair follicle development. However, our 312

    study here showed that substantial murine cell type-specific biomarkers are identical to the 313

    Cashmere goat. Besides, by comparison of cell type-specific signature genes between 314

    Cashmere goat and mouse (Fig. 3c and Fig. 6c,d), we found that the transcriptome 315

    similarity decreased along pseudotime (revealed by overlapped signature genes in this 316

    study). Briefly, about 729 DC signature genes overlapped between Cashmere goat and 317

    mouse, and murine DC signature genes at different stages (pre-DC, DC1 and DC2) showed 318

    similar expression pattern along pseudotime in Cashmere goat (Mok et al., 2019). While for 319

    the late-stage IRS and hair shaft cell population in the late stage of hair follicle 320

    development, the percentage of overlapped genes decreased (126 and 93 overlapped genes, 321

    respectively). Similarly, by comparing gene expression across 7 different species from brain, 322

    heart, ovary, kidney, testis and liver, researches have demonstrated that organ-specific 323

    molecular profiles are more similar in early development and become more distinct during 324

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • development (Cardoso-Moreira et al., 2019), it’s therefore plausible that hair follicle 325

    development in Cashmere goat and mouse may also consistent with such theory. 326

    Similar to the murine model, we also observed differential gene expression profiles in 327

    different DP cell populations from different hair follicles (primary hair follicles vs 328

    secondary hair follicles), which was also consistent with our recent findings on mice (Ge et 329

    al., 2019). Besides, an in-depth comparison of differentially expressed genes revealed that 330

    different DP cell populations require different transcriptional profiles, and it’s, therefore, 331

    plausible that the different induction signals may be involved during the asynchronous 332

    development of hair follicles. Consistent with such hypothesis, researches found that SOX2 333

    was specifically expressed in guard hair follicles, but not zigzag hair follicles, while SOX18 334

    was demonstrated to regulate zigzag hair follicle morphogenesis (Driskell et al., 2009, 335

    James et al., 2003, Pennisi et al., 2000). Although our study here also demonstrates the 336

    asynchronous development of different hair follicles in Cashmere goat, the detailed 337

    machinery underlying such phenomenon was not investigated here and future studies may 338

    focus on such topics, which will definitely provide us new insight into the hair follicle 339

    biology and hair follicle regeneration. 340

    Based on single-cell pseudotime trajectory inference, our study here also highlighted 341

    the underappreciated cell fate decisions during hair follicle development. Different from 342

    our previous studies performed in mice, our pseudotime lineage trajectory of epidermal cell 343

    lineage showed three different branch points while murine epidermal cell lineage trajectory 344

    showed two branch points. by analyzing the gene expression profiles of the first two branch 345

    points, they actually showed similar cell fate decisions, while the additional branch point in 346

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Cashmere goat mainly comes from keratinocyte differentiation. Such differences in 347

    pseudotime trajectory may be partially explained by the differences in the development of 348

    hair follicles across species, for example, E120 stage Cashmere goat showed obvious hair 349

    fiber on the surface of the skin, while it was not until 6-7 postnatal day that the hair follicles 350

    in the mouse skin surface become visible. The difference in the pseudotime trajectory 351

    reveals that hair follicle development was heterochrony between Cashmere goat and mouse, 352

    which was frequent when comparing the development of specific organs across species. 353

    Finally, our dataset here provides an important resource for understanding the cellular 354

    heterogeneity and major cell fate decisions during Cashmere goat hair follicle development. 355

    For the first time, the detailed transcriptional landscape of different cell populations was 356

    delineated here at single-cell resolution, which provided valuable resources for the 357

    identification of biomarkers. Besides, our trajectory inference analysis here successfully 358

    recapitulated major cell fate decisions during Cashmere goat hair follicle development, 359

    which enabled us to comprehensively study the detailed developmental pathways involved 360

    during Cashmere goat hair follicle morphogenesis, and will also have implications for the 361

    Cashmere goat breeding work in animal husbandry. 362

    363

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Materials and methods 364

    Experimental Animals 365

    All the experimental Shaanbei White Cashmere goats involved in this study were obtained 366

    from the Shaanbei Cashmere Goat Engineering Technology Research Center of Shaanxi 367

    Province and were fed with Cashmere goat feeding standard (DB61/T583-2013) of Shaanxi 368

    Province. All pregnant goats were prepared using artificial insemination and all the 369

    experimental procedures involved goats in this study were approved by the Experimental 370

    Animal Manage Committee of Northwest A&F University. 371

    Single-cell suspension preparation 372

    The goat fetus at desired dates was isolated using cesarean operation when the pregnant 373

    goats were anesthetic with the compound ketamine. The skin tissues (0.5 cm × 0.5 cm) 374

    were isolated from the fetus back skin and were immediately transferred to the ice-cold 375

    DMEM/F12 media (Gibco, Beijing, China) with 50 U/ml penicillin and 50 mg/ml 376

    streptomycin (HyClone, Beijing, China). After washing three times with DMEM/F12 to 377

    remove contaminative blood cells, the skin tissues were then dissociated into single cells 378

    prior to sequencing. For E60 and E90 skin tissues, the obtained skin tissues were firstly 379

    incubated with 2 mg/ml collagenase IV (Sigma, St Louis, MO, USA) for 30 min at 37°C, 380

    and then the skin tissues were mechanically dissociated into single-cell suspensions with a 381

    1ml pipette tip. For E120 skin tissues, the skin tissues were firstly cut into ~3 mm skin 382

    pieces and then incubated with 2 mg/ml collagenase IV for 30 min. After incubation, the 383

    hair follicles within the skin tissues were isolated with a pair of precise forceps and the 384

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • pooled hair follicles were further dissociated into single cells with TypLE Express (Gibco, 385

    Grand Island, NY, USA) for 30 min at 37°C. The obtained single-cell suspensions were 386

    then washed three times with PBS supplemented with 0.04% BSA (Sigma, St Louis, MO, 387

    USA) and were filtered with a 40 μm cell strainer (BD Falcon, BD Biosciences, San Jose, 388

    CA, USA) to remove debris and cell aggregations. For each stage, the samples were 389

    obtained from at least 2 different goat fetus and for each fetus goat, the single-cell 390

    suspension was prepared separately until finally pooled together prior to single-cell 391

    barcoding. 392

    Single-cell library construction and sequencing 393

    Single-cell library was constructed using 10x Genomics’ Chromium Single Cell 3’ V3 Gel 394

    Beads Kit (10x Genomics, Pleasanton, CA, USA) according to the manufacture’s 395

    instructions. After cell counting, the single-cell suspension was adjusted to 1000 cells/μl 396

    and about 7,000 cells were obtained for each stage. The single-cell barcoding procedure 397

    was performed using a 10x Genomics Chromium barcoding system (10x Genomics, 398

    Pleasanton, CA, USA) according to the manufacturer's guide. After single-cell library 399

    construction, the Illumina HiSeq X Ten sequencer (Illumina, San Diego, CA, USA) was 400

    used to sequence and 150 bp pair ended reads were generated. 401

    10x Genomics single-cell RNA sequencing data processing 402

    The obtained raw sequencing files were processed with standard CellRanger (v2.2.0) 403

    pipeline according to the 10x Genomics official guide (https://www.10xgenomics.com/cn/). 404

    The produced raw base call files were firstly transformed into fastq files by using 405

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Cellranger mkfastq function. The goat ARS1 reference genome downloaded from ensemble 406

    was used as a reference genome (https://asia.ensembl.org/Capra_hircus/Info/Index). 407

    Cellranger count function was used to perform mapping, filtering low-quality cells, 408

    barcoding counting and UMI counting. 409

    After the standard Cellranger pipeline, the generated gene expression matrice files were 410

    then analyzed with Seurat (V2.3.4) package according to the official user guide 411

    (https://satijalab.org/seurat/vignettes.html). Quality control was performed using FilterCells 412

    function and cells with detected genes less than 200 and genes expressed less than 3 cells 413

    were filtered. After normalization and data scaling, the different datasets were integrated by 414

    using RunMultiCCA function. tSNE was used to perform dimension reduction analysis and 415

    different cell clusters were identified by using FindClusters function. The cluster 416

    specifically expressed genes were analyzed with FindAllMarkers function and with the 417

    parameter “min.pct = 0.25, thresh.use = 0.25”. 418

    Single-cell pseudotime lineage trajectory reconstruction 419

    Single-cell lineage reconstruction analysis was performed using Monocle (V2) packages 420

    according to the online tutorial (http://cole-trapnell-lab.github.io/monocle-release/docs/). 421

    The monocle object was constructed from Seurat object with newCellDataSet function, and 422

    Seurat determined variable genes were used as ordering genes to order cells in pseudotime 423

    along a trajectory. Dimension reduction was performed using DDRTree methods. To 424

    analyze differential gene expression between different cell branches, BEAM function was 425

    used and differentially expressed genes were identified with q-val < 1e-4. Branch-specific 426

    gene expression heatmap was plotted with plot_genes_branched_heatmap function and 427

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • different gene set was calculated according to k-means clustering. 428

    Immunohistochemistry staining analysis 429

    The immunofluorescence or enzyme horseradish peroxidase (HRP)-based 430

    immunohistochemistry analysis procedure was performed as we previously described (Ge 431

    et al., 2017, Liu et al., 2017). For immunofluorescence analysis, the paraffin-embedded skin 432

    tissues were firstly deparaffinized in xylene and further rehydrated in ethanol solutions. 433

    Antigen retrieval was performed in 0.01 M sodium citrate buffer at 96°C. Following a 434

    permeabilization procedure in 0.5 M Tris-HCI buffer supplemented with 0.5% TritonX-100 435

    (Sorlabio, Beijing, China) for 10 min, the slides were then blocked with 3 % BSA and 10 % 436

    donkey serum (Boster, Wuhan, China) in 0.5 M Tris-HCI buffer for 30 min. The primary 437

    antibodies diluted in the blocking buffer were incubated with the slides at 4°C overnight. 438

    The next morning, the corresponding secondary antibodies were then added in the slides 439

    and incubated at 37°C for 1 h. DAPI was used to stain the nuclei and the pictures were 440

    taken using Nikon AR1 confocal system (Nikon, Tokyo, Japan). For HRP based 441

    immunohistochemistry analysis, following antigen retrieval, the samples were firstly 442

    incubated with 3% H2O2 for 10 min at room temperature to remove endogenous peroxidase 443

    activity. The primary antibodies were incubated 4 °C overnight, and the corresponding 444

    HRP-labeled secondary antibodies were added in the next morning for 40 min at room 445

    temperature. After that, peroxidase substrate DAB (Zsbio, Beijing, China) was used for 446

    chromogenic reaction with hematoxylin used to stain nuclei. The slides were finally 447

    mounted with neutral resins and pictures were captured with an Olympus BX51 microscope 448

    imaging system (Olympus, Tokyo, Japan). All the primary and secondary antibodies used in 449

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • this study were listed in Supplementary Table 5. 450

    451

    Data availability 452

    The single-cell RNA sequencing data used in this research is deposited in NCBI GEO 453

    databases under accession number: GSE144351. 454

    455

    Acknowledgments 456

    This work was supported by the National Natural Science Foundation of China (31972556 457

    and 31671554). 458

    459

    460

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • References 461

    AJ Ritar, S Salamon, PD Ball, O'may P (1989) Ovulation and fertility in goats after intravaginal device-PMSG 462

    treatment. Small Ruminant Research 2: 323-331 463

    Angerer P, Simon L, Tritschler S, Wolf FA, Fischer D, Theis FJ (2017) Single cells make big data: New challenges 464

    and opportunities in transcriptomics. Current Opinion in Systems Biology 4: 85-91 465

    Ansari-Renani HR, Ebadi Z, Moradi S, Baghershah HR, Ansari-Renani MY, Ameli SH (2011) Determination of 466

    hair follicle characteristics, density and activity of Iranian cashmere goat breeds. Small Ruminant Research 95: 467

    128-132 468

    Cardoso-Moreira M, Halbert J, Valloton D, Velten B, Chen C, Shao Y, Liechti A, Ascencao K, Rummel C, 469

    Ovchinnikova S, Mazin PV, Xenarios I, Harshman K, Mort M, Cooper DN, Sandi C, Soares MJ, Ferreira PG, 470

    Afonso S, Carneiro M et al. (2019) Gene expression across mammalian organ development. Nature 571: 471

    505-509 472

    Chen D, Jarrell A, Guo C, Lang R, Atit R (2012) Dermal beta-catenin activity in response to epidermal Wnt 473

    ligands is required for fibroblast proliferation and hair follicle initiation. Development 139: 1522-33 474

    Chi W, Wu E, Morgan BA (2013) Dermal papilla cell number specifies hair size, shape and cycling and its 475

    reduction causes follicular decline. Development 140: 1676-83 476

    Detmar M, Brown LF, Schon MP, Elicker BM, Velasco P, Richard L, Fukumura D, Monsky W, Claffey KP, Jain RK 477

    (1998) Increased microvascular density and enhanced leukocyte rolling and adhesion in the skin of VEGF 478

    transgenic mice. The Journal of investigative dermatology 111: 1-6 479

    Driskell RR, Giangreco A, Jensen KB, Mulder KW, Watt FM (2009) Sox2-positive dermal papilla cells specify 480

    hair follicle type in mammalian epidermis. Development 136: 2815-23 481

    Duverger O, Morasso MI (2009) Epidermal patterning and induction of different hair types during mouse 482

    embryonic development. Birth Defects Res C Embryo Today 87: 263-72 483

    Forni MF, Trombetta-Lima M, Sogayar MC (2012) Stem cells in embryonic skin development. Biol Res 45: 484

    215-22 485

    Gao Y, Wang X, Yan H, Zeng J, Ma S, Niu Y, Zhou G, Jiang Y, Chen Y (2016) Comparative Transcriptome Analysis 486

    of Fetal Skin Reveals Key Genes Related to Hair Follicle Morphogenesis in Cashmere Goats. PloS one 11: 487

    e0151118 488

    Ge W, Tan S-J, Wang S-H, Li L, Sun X-F, Shen W, Wang X (2019) Single-cell transcriptome profiling reveals 489

    dermal and epithelium cell fate decisions during embryonic hair follicle development. bioRxiv 490

    Ge W, Zhao Y, Lai FN, Liu JC, Sun YC, Wang JJ, Cheng SF, Zhang XF, Sun LL, Li L, Dyce PW, Shen W (2017) 491

    Cutaneous applied nano-ZnO reduce the ability of hair follicle stem cells to differentiate. Nanotoxicology: 492

    1-34 493

    Geng R, Yuan C, Chen Y (2013) Exploring differentially expressed genes by RNA-Seq in cashmere goat (Capra 494

    hircus) skin during hair follicle development and cycling. PloS one 8: e62704 495

    Green H, Easley K, Iuchi S (2003) Marker succession during the development of keratinocytes from cultured 496

    human embryonic stem cells. Proceedings of the National Academy of Sciences of the United States of 497

    America 100: 15625-30 498

    Gu LH, Coulombe PA (2007) Keratin function in skin epithelia: a broadening palette with surprising shades. 499

    Current opinion in cell biology 19: 13-23 500

    Gupta K, Levinsohn J, Linderman G, Chen D, Sun TY, Dong D, Taketo MM, Bosenberg M, Kluger Y, Choate K, 501

    Myung P (2019) Single-Cell Analysis Reveals a Hair Follicle Dermal Niche Molecular Differentiation Trajectory 502

    that Begins Prior to Morphogenesis. Developmental cell 48: 17-31 e6 503

    Huh SH, Narhi K, Lindfors PH, Haara O, Yang L, Ornitz DM, Mikkola ML (2013) Fgf20 governs formation of 504

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • primary and secondary dermal condensations in developing hair follicles. Genes & development 27: 450-8 505

    James K, Hosking B, Gardner J, Muscat GE, Koopman P (2003) Sox18 mutations in the ragged mouse alleles 506

    ragged-like and opossum. Genesis 36: 1-6 507

    Joost S, Zeisel A, Jacob T, Sun X, La Manno G, Lonnerberg P, Linnarsson S, Kasper M (2016) Single-Cell 508

    Transcriptomics Reveals that Differentiation and Spatial Signatures Shape Epidermal and Hair Follicle 509

    Heterogeneity. Cell Syst 3: 221-237 e9 510

    Karlsson L, Bondjers C, Betsholtz C (1999) Roles for PDGF-A and sonic hedgehog in development of 511

    mesenchymal components of the hair follicle. Development 126: 2611-21 512

    Lee SB, Shim S, Kim MJ, Shin HY, Jang WS, Lee SJ, Jin YW, Lee SS, Park S (2016) Identification of a distinct 513

    subpopulation of fibroblasts from murine dermis: CD73(-) CD105(+) as potential marker of dermal fibroblasts 514

    subset with multipotency. Cell Biol Int 40: 1008-16 515

    Liu JC, Lai FN, Li L, Sun XF, Cheng SF, Ge W, Wang YF, Zhang XF, De Felici M, Dyce PW, Shen W (2017) Di 516

    (2-ethylhexyl) phthalate exposure impairs meiotic progression and DNA damage repair in fetal mouse 517

    oocytes in vitro. Cell death & disease 8: e2966 518

    Millar SE (2002) Molecular mechanisms regulating hair follicle development. The Journal of investigative 519

    dermatology 118: 216-25 520

    Mok KW, Saxena N, Heitman N, Grisanti L, Srivastava D, Muraro MJ, Jacob T, Sennett R, Wang Z, Su Y, Yang LM, 521

    Ma'ayan A, Ornitz DM, Kasper M, Rendl M (2019) Dermal Condensate Niche Fate Specification Occurs Prior 522

    to Formation and Is Placode Progenitor Dependent. Developmental cell 48: 32-48 e5 523

    Ouspenskaia T, Matos I, Mertz AF, Fiore VF, Fuchs E (2016) WNT-SHH Antagonism Specifies and Expands Stem 524

    Cells prior to Niche Formation. Cell 164: 156-169 525

    Paquet-Fifield S, Schluter H, Li A, Aitken T, Gangatirkar P, Blashki D, Koelmeyer R, Pouliot N, Palatsides M, Ellis 526

    S, Brouard N, Zannettino A, Saunders N, Thompson N, Li J, Kaur P (2009) A role for pericytes as 527

    microenvironmental regulators of human skin tissue regeneration. The Journal of clinical investigation 119: 528

    2795-806 529

    Pennisi D, Gardner J, Chambers D, Hosking B, Peters J, Muscat G, Abbott C, Koopman P (2000) Mutations in 530

    Sox18 underlie cardiovascular and hair follicle defects in ragged mice. Nature genetics 24: 434-7 531

    Pennisi E (2018) Chronicling embryos, cell by cell, gene by gene. Science 360: 367 532

    Polakis P (2001) More than one way to skin a catenin. Cell 105: 563-6 533

    Ren H, Wang G, Chen L, Jiang J, Liu L, Li N, Zhao J, Sun X, Zhou P (2016) Genome-wide analysis of long 534

    non-coding RNAs at early stage of skin pigmentation in goats (Capra hircus). BMC genomics 17: 67 535

    Saxena N, Mok KW, Rendl M (2019) An Updated Classification of Hair Follicle Morphogenesis. Experimental 536

    dermatology 537

    Schlake T (2007) Determination of hair structure and shape. Seminars in cell & developmental biology 18: 538

    267-73 539

    Schmidt-Ullrich R, Paus R (2005) Molecular principles of hair follicle induction and morphogenesis. Bioessays 540

    27: 247-61 541

    Scott Waldron, Colin Brown, Komarek. AM (2014) The Chinese Cashmere Industry A Global Value Chain 542

    Analysis. Development Policy Review 32: 589-610 543

    Tsai SY, Sennett R, Rezza A, Clavel C, Grisanti L, Zemla R, Najam S, Rendl M (2014) Wnt/beta-catenin signaling 544

    in dermal condensates is required for hair follicle formation. Developmental biology 385: 179-88 545

    Yang H, Adam RC, Ge Y, Hua ZL, Fuchs E (2017) Epithelial-Mesenchymal Micro-niches Govern Stem Cell 546

    Lineage Choices. Cell 169: 483-496 e13 547

    Zhang Y, Yin J, C L (2006) Study on development of skin and hair follicle from fetal inner mongolian Arbas 548

    cashmere goats. ACTA VETERINARIA ET ZOOTECHNICA SINICA, 37: 761 549

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Zhang Y, Tomann P, Andl T, Gallant NM, Huelsken J, Jerchow B, Birchmeier W, Paus R, Piccolo S, Mikkola ML, 550

    Morrisey EE, Overbeek PA, Scheidereit C, Millar SE, Schmidt-Ullrich R (2009) Reciprocal requirements for 551

    EDA/EDAR/NF-kappaB and Wnt/beta-catenin signaling pathways in hair follicle induction. Developmental cell 552

    17: 49-61 553

    554 555 556

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Figure Legends 557

    Fig. 1 Dissecting cellular heterogeneity during Cashmere goat hair follicle 558

    development. (a) Overall experimental design. (b) tSNE plot of all single cells labeled with 559

    developmental time. Cells from the different developmental points were color-coded with 560

    different colors. (c) tSNE plot of all single cells labeled with cell types according to their 561

    marker gene expression. Different colors represent different cell clusters and the cell 562

    number for each cluster was listed in the bracket. (d) Dot plot of representative marker 563

    genes for different cell clusters. The color intensity represents its expression level, and the 564

    dot size represents the positive cell percentage. 565

    566

    Fig. 2 Delineating dermal and epidermal cell lineage pseudotime developmental 567

    trajectory. (a) Dermal cell lineage highlighted in the tSNE plot. (b) Developmental 568

    trajectory of dermal cell lineage along pseudotime. Cells were color-coded with cell types 569

    identified by Seurat (left panel) and developmental time (right panel), respectively. (c) 570

    Epidermal cell lineage highlighted in the tSNE plot. (d) Developmental trajectory of 571

    epidermal cell lineage along pseudotime. Cells were also color-coded with cell types (left 572

    panel) and developmental time (right panel), respectively. (e) Main dermal cell lineage 573

    decisions during hair follicle morphogenesis. (f) Main epidermal cell lineage fate decisions 574

    during hair follicle morphogenesis. 575

    576

    Fig. 3 Revealing molecular profiles during DC fate commitment. (a) Pseudotime 577

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • expression heatmap during DC fate commitment. The four gene sets were determined by 578

    k-means clustering according to their expression pattern and GO terms for each gene set 579

    were listed in the right panel. (b) Immunofluorescence analysis of BMP2, CTNNB1, LEF1 580

    and K15 in E60 Cashmere goat skin tissues. Scale bars = 50 μm. (c) Veen diagram 581

    illustrating overlapped signature genes between E60 Cashmere goat DC cells and murine 582

    E13.5 DC signature genes. (d) Visualizing murine DC signature genes of different stages 583

    along pseudotime in Cashmere goat. The murine signature DC markers were listed in the 584

    top panel and their corresponding expression pattern in Cashmere goat was listed in the 585

    lower panel. Cells were color-coded according to their cluster identify. 586

    587

    Fig 4. DP cells from primary hair follicles and secondary hair follicles in Cashmere 588

    goat showed distinct gene expression profiles. (a) Heatmap illustrating dynamic gene 589

    expression profiles during DP cell fate commitment. The gene expression pattern for each 590

    gene set was listed in the middle panel, and the top 5 enriched GO terms for each gene set 591

    were listed in the right panel. (b) Pseudotime expression of cell fate 1 enriched genes 592

    APOD, SOX18 and cell fate 2 enriched genes CENPW, TOP2A. Cells were colored-coded 593

    with their corresponding cluster identity. (c) Volcano plot illustrating cell fate 1 and cell fate 594

    significantly enriched differentially expressed genes. (d) Immunofluorescence analysis of 595

    BMP2, K15, VDR and PCNA in the primary hair follicles and secondary hair follicles from 596

    E120 Cashmere skin sections. Scale bars = 50 μm. 597

    598

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Fig. 5 Delineating matrix cell fate decision along pseudotime. (a) Heatmap 599

    demonstrating dynamic gene expression patterns during matrix cell fate commitment. The 600

    differentially expressed genes were divided into 4 different gene sets based on k-means 601

    clustering. (b) Gene functional enrichment analysis of signature genes in the 4 different 602

    gene sets identified in Fig. 5a. (c) The pseudotime expression pattern of representative 603

    signature genes from each gene set. Cells were color-coded according to their cluster 604

    identity. (d) Immunofluorescence analysis of LEF1 and CTNNB1 in E60 Cashmere skin 605

    tissues. Scale bars = 50 μm. 606

    607

    Fig. 6 Revealing pseudotime gene expression dynamics during IRS and hair shaft cell 608

    fate commitment. (a) Heatmap illustrating pseudotime gene expression pattern of 609

    differentially expressed genes during IRS and hair shaft cell development. Cell fate 1 610

    depicts IRS fate, while cell fate 2 depicts hair shaft fates. (b) Pseudotime expression pattern 611

    of representative marker genes during IRS and hair shaft cell fate commitment. Cells were 612

    color-coded with their corresponding cluster identity and the solid line depicts cell fate 1, 613

    while the dashed line depicts cell fate 2. (c) Veen diagram demonstrating overlapped 614

    murine and Cashmere goat IRS signature genes. The representative overlapped genes were 615

    listed in the corresponding boxes. (d) Comparison of murine hair shaft signature genes with 616

    Cashmere goat hair shaft signature genes. The representative overlapped genes were listed 617

    in the rectangular boxes. (e) Comparison of PCNA and VDR expression in E90 Cashmere 618

    goat and E16.5 mouse skin tissues. Scale bars = 50 μm. 619

    620

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Fig. 7 Dissecting the asynchronous development of keratinocyte. (a) Heatmap 621

    illustrating gene expression dynamics during keratinocyte differentiation. (b) GO terms 622

    corresponding to the gene set in Figure 7a. The arrow indicates the elongation of 623

    pseudotime. (c) Expression of cell fate 1 signature genes KRT14, KRT17 and cell fate 2 624

    signature genes TOP2A, PCNA along the pseudotime trajectory. (d) Immunofluorescent 625

    staining analysis of VDR, PCNA, CTNNB1, BMP2, LEF1 and K15 in E90 Cashmere goat 626

    skin tissues. Scale bars = 50 μm. 627

    628

    Supplementary Figure Legends 629

    Supplementary Fig. 1 Single-cell dataset quality control and representative marker 630

    expression across all single cells. (a) Morphology of fetus Cashmere goat at E60, E90 and 631

    E120 and its corresponding skin structure revealed by HE staining. (b) Quality matrices 632

    revealed by CellRanger of all datasets used in this study. (c) Comparison of the number of 633

    genes detected and the number of UMI detected in each cell. (d) Evaluating key cell type 634

    markers across all single cells in the tSNE plot. 635

    636

    Supplementary Fig. 2 Gene function enrichment of differentially expressed genes 637

    during DC fate commitment. (a) The network of enriched GO terms for different gene 638

    sets during DC fate commitment. Each dot represents one GO terms and different colors 639

    represent different gene sets. (b) Circos plot demonstrating the number of overlapped GO 640

    terms between different gene sets. 641

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Supplementary Fig. 3 Gene function enrichment of differentially expressed genes 642

    during DP cell fate commitment. (a) The network of enriched GO terms for different gene 643

    sets during DP fate commitment. Each dot represents one GO terms and different colors 644

    represent different gene sets. (b) Circos plot demonstrating the number of overlapped GO 645

    terms between different gene sets. (c) Comparison of enriched GO terms between two 646

    different cell fate. 647

    648

    Supplementary Fig. 4 Comparison of enriched GO terms during matrix cell fate 649

    commitment. (a) Heatmap illustrating gene set specific enriched GO terms. (b) Circos plot 650

    demonstrating the number of overlapped GO terms between different gene sets. 651

    652

    Supplementary Fig. 5 Validation of hair shaft/IRS marker expression and comparison 653

    of enriched GO terms. (a) Immunohistochemistry analysis of hair shaft marker HOXC13 654

    and IRS marker SOX9 expression in Cashmere goat skin. Scale bars = 50 μm. (b) The 655

    network of enriched GO terms for different gene sets during hair shaft and IRS fate 656

    commitment. Each dot represents one GO terms and different colors represent different 657

    gene sets. (c) Heatmap illustrating gene set specific enriched GO terms. 658

    659

    Supplementary Fig. 6 Gene functional enrichment and representative gene expression 660

    along pseudtotime. (a) The network of enriched GO terms for different gene sets during 661

    keratinocyte differentiation. Each dot represents one GO terms and different colors 662

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • represent different gene sets. (b) Psedotime expression pattern of representative cell fate 1 663

    and cell fate 2 signature genes. 664

    665

    Supplementary Tables 666

    Supplementary table 1: All cluster specific differentially expressed genes. 667

    Supplementary table 2: DC signature genes for different gene sets. 668

    Supplementary table 3: DC signature genes comparasion between goat and mice. 669

    Supplementary table 4: IRS and hair shaft signature genes comparasion between goat and 670

    mice. 671

    Supplementary table 5: All antibodies used in this study. 672

    673

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • E60E90E120

    Figure 1

    (a)

    E60

    E90

    E120

    Fetus Cashmere goat skin biospy Single cell barcoding

    Beads Oil

    Sequencing & Dissecting heterogeneity

    (b) (c) Cell type (cells)

    (d)

    CD

    H19

    JSR

    P1

    ARSI

    CN

    MD

    L1C

    AM

    −1 2

    e

    0 25 50 750

    1

    23

    4

    5

    67

    89

    11

    1

    111

    LUM

    COL1

    A1

    POST

    N

    DCN

    APO

    E

    KRT1

    5

    KRT1

    7

    SOX9

    KRT1

    4

    KLF5

    RSP

    O2

    APO

    D

    SOX1

    8

    SOX2

    PCO

    LCE2

    PLVA

    P

    VWF

    PEC

    AM1

    KDR

    DEP

    P1

    KRT1

    0

    KRT1

    SBSN

    KRTD

    AP

    LRAT

    TBX2

    ACTA

    2

    TPM

    2

    EBF2

    ABC

    C9

    LCP1

    FCER

    1A

    RG

    S1

    RG

    S10

    LTC

    4S

    tSNE 1

    tSN

    E 2

    Clu

    ster

    ID

    dermal epithelial DP endothelial keratinocyte pericyte macrophagy muscle

    Developmental time

    tSNE 1

    tSN

    E 2

    0

    1

    2

    3

    4

    5

    6

    7

    8

    9

    1

    1

    1

    1

    1

    1

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 1 2

    Component 1

    Com

    pone

    nt 2

    Figure 2

    (a)1

    4

    1 2

    Component 1

    Com

    pone

    nt 2

    Cell type (cells)Developmental trajectory Developmental time

    ( )

    ( )

    ( )

    Component 1

    Com

    pone

    nt 2

    1

    2

    3

    Cell type (cells)Developmental trajectory

    ( )

    ( )

    ( )

    Dermal lineage

    Epidermal lineage

    tSNE 1

    tSN

    E 2

    tSNE 1

    tSN

    E 2

    Component 1

    Com

    pone

    nt 2

    1

    2

    3

    Developmental time

    (c)

    (b) (d)

    (e)

    Undiff. DC

    Fibro.

    DP

    (f)

    Undiff.

    Matrix Hair shaft Keratinocyte

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Figure 3

    (a)

    Pseudotime

    FbAPODHES1LUMDCNVIMSOX18SOX9APOESOX2LHX2COL1A1HOXC13JUNPDGFADKK1TPX2NOTCH3

    PTNLCKCORINCENPCCCDC66SFPQ

    SYNRGBDKRB2AUTS2MRPL16ACADLHUNKWDR36CAB39LANGPT1PRXL2APURBKLHL24UBE2J2NDUFS5

    0 10 20 30 40

    0.0 2.5 5.0 7.5

    0 2 4 6

    0 2 4 6

    tissue morphogenesisresponse to growth factor

    cell morphogenesis involved in differentiationepidermis development

    GO (BP) -Log(P)

    negative regulation of cell cycle

    positive regulation of organelle organization

    negative regulation of protein modification

    DNA-dependent DNA replication

    Wnt signaling pathway

    regulation of type I interferon production

    viral translational termination-reinitiation

    multicellular organismal homeostasis

    proteasomal protein catabolic process

    nucleobase-containing compound catabolic

    intracellular receptor signaling pathway

    anatomical structure homeostasis

    (b)

    1951(38.8%)

    2345(46.7%)

    729(14.5%)

    Cashmere goat DC signature genes

    E13.5 murine DCsignature genes

    TRPS1

    PRDM1

    0 25 50 75 100

    13

    10

    13

    10

    Pseudotime

    Expr

    essi

    on

    RSPO3

    INHBA

    0 25 50 75 100

    13

    1030

    13

    1030

    Pseudotime

    Expr

    essi

    on 1

    13

    467

    (c)

    LEF1

    DKK1

    0 25 50 75 100

    13

    10

    0.51.0

    3.05.0

    Pseudotime

    Expr

    essi

    on

    Pre-DC DC1 DC2

    Lef1, Dkk1, Twist, Fst Prdm1, Trps1, Sox18, Foxd1 Inhba, Rspo3, Foxp1, Hey1

    Pseudotime

    (d)

    Dc

    -3

    3Exp.

    1

    2

    3

    4

    SOX18

    APOD

    0 25 50 75 100

    13

    1030

    1

    10

    100

    Pseudotime

    Expr

    essi

    on

    SOX2

    CTNNB1

    0 25 50 75 100

    0.51.0

    3.05.0

    13

    1030

    Pseudotime

    Expr

    essi

    on

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Cell Fate 1 vs Cell Fate 2

    Figure 4

    (a)

    -3

    3Exp. 0 10 20

    0 20 40 60

    0 5 10 15 20

    0 1 2 3

    tissue morphogenesisepidermis development

    epithelial cell differentiationneuron projection morphogenesis

    GO (BP)

    mitotic cell cycle process

    DNA-dependent DNA replication

    cellular response to DNA damage stimulus

    microtubule cytoskeleton organization

    regulation of cell morphogenesis

    citrulline metabolic process

    protein-containing complex disassembly

    protein localization to membrane

    extracellular matrix organization

    skeletal system development

    blood vessel development

    cellular response to growth factor stimulus

    Pre-Branch Cell Fate 1 Cell Fate 2

    Pseudotime

    -Log(P)Signature gene expression

    1,53

    91.

    077

    559

    201

    BMP2

    /K15

    /DAP

    I

    Primary

    BMP2

    /K15

    /DAP

    I

    Secondary

    VDR

    /PC

    NA/

    DAP

    I

    VDR

    /PC

    NA/

    DAP

    I

    (b) (c) (d)

    SOX18

    APOD

    0 25 50 75 100

    13

    1030

    1

    10

    100

    Pseudotime

    Expr

    essi

    on

    1

    13

    467

    TOP2A

    CENPW

    0 25 50 75 100

    13

    1030

    13

    1030

    Pseudotime

    Expr

    essi

    on

    SOX18 SOX2

    FGF7KLF2

    H2AFZ

    CENPWLUM

    KIF20B TOP2A

    VIM

    DLK1

    0

    50

    100

    150

    200

    −2 20

    avg_logFC

    log1

    0(p_

    val_

    adj)

    Significant None

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Figure 5

    (a)KRT25KRT71KRT85KRT27KRT35KRT84AHCYFGF21HBP1HPRT1CLTBGANTIMP2KCNK7CTSVTXN

    POSTNVIMCOL1A1LUMACTC1COL1A2OGNTOP2A

    KRT28PADI3SBSNKRT14KRT19LRATKRT23KRT73BMP4LEF1KRT4KRT1KRT17KRT10PDGFASOX9HES1SOX18

    keratinocyte differentiationepidermis development

    skin epidermis developmentregulation of keratinocyte differentiation

    GO (BP) -Log(P)

    cardiac epithelial to mesenchymal transition

    mesenchymal cell differentiation

    epithelial to mesenchymal transition

    BMP signaling pathway

    skin development

    morphogenesis of an epithelium

    negative regulation of cell proliferation

    keratinocyte proliferation

    cell division

    skeletal system development

    ossification

    regulation of endothelial cell proliferation

    0.0 2.5 5.0 7.5

    0 2 4

    0 5 10 15

    0 5 10 15

    Pseudotime

    Undiff. Matrix

    KRT27

    KRT25

    0 25 50 75 100

    1e+01

    1e+03

    1e+05

    1e+021e+051e+08

    Expr

    essi

    on

    SBSN

    LEF1

    0 25 50 75 100

    1

    3

    10

    1

    10

    100Exp

    ress

    ion

    POSTN

    LUM

    0 25 50 75 100

    1

    10

    100

    110

    1001000

    Exp

    ress

    ion

    SOX9

    SOX18

    0 25 50 75 100

    13

    1030

    13

    1030

    Exp

    ress

    ion

    gene

    set 1

    gene

    set 2

    gene

    set 3

    gene

    set 4

    Pseudotime

    Cell clusters:0 2 3 5 8 9 12

    (b) (c)

    -3

    3Exp.

    4 3 2 1

    LEF1/DAPILEF1 DAPI

    E60

    skin

    (d)

    CTNNB1/DAPICTNNB1 DAPI

    E60

    skin

    epidermal

    dermal

    epidermal

    dermal

    epidermal

    dermal

    epidermal

    dermal

    epidermal

    dermal

    epidermal

    dermal

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • Figure 6

    (a)

    Pseudotime

    Cell fate 2

    -3

    3Exp.

    ALCAMCOL1A2LUMOGNVIMCOL1A1SOX18VCANFSTAPODDKK1DLK1NR3C1CDH11MICU3ACADSLRCH3

    H2AFZDCNTOP2APCNASHHVDREGFRLCKDCTETV5DCKSLF1MT3SNRPB

    SBSNKRT4KRT1KRT14SOX9HES1PRDM1ELF1

    MSX1KRT35ID3ID1AHCYHOXC13LEF1APOOUSP16RPS9MARS

    Cell fate 1

    OGN

    LUM

    0 25 50 75 100

    1

    10

    100

    13

    1030

    Expr

    essi

    on

    TOP2A

    DCN

    0 25 50 75 100

    1

    10

    100

    13

    1030

    Expr

    essi

    on

    SOX18

    COL1A1

    0 25 50 75 100

    1

    10

    100

    13

    1030

    Expr

    essi

    on

    H2AFZ

    CENPF

    0 25 50 75 100

    13

    1030

    13

    1030

    Expr

    essi

    on

    Cell clusters:0 2 3 5 8 9 12

    Pseudotime

    Cell fates:fate 1fate 2

    Pre-branch(b)

    581 (40.3%)

    306 (21.3%)

    57 (4%)

    427 (29.7%)

    69 (4.8%)

    Cashmere goat IRSsignature genes

    Murine IRS-1signature genes

    Murine IRS-2signature genes

    Cell fate 1

    KRT79, SBSN, KRT17,KRT7, KRT79, DUSP1

    KRT14, KRT5, KRT1,KRT10, KRT77, KLF5,KLF4, KLF3 ...

    (c)

    Murine hair shaftsignature genes

    491(23.9%)

    389(18.9%)

    1,085(52.7%)

    26(1.3%)

    67(3.3%)

    Goat hair shaft HS-1

    Goat hair shaftHS-2

    MSX1,KRT35, HOXC13APOO, BMP4 ...

    VDR, CUX1, SHH,LHX2, PCNA, DCN...

    (d)

    1

    2

    3

    4

    (e) E90 goat E16.5 mouse

    PCN

    A

    VDR

    E90 goat E16.5 mouse

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/

  • 1

    2

    3

    KRT14

    Figure 7

    (a) Cell fate 2

    -3

    3Exp.

    KRT85KRT35KRT14KRT84KRT28VDRBMP4STAR

    Cell fate 1 Pre-branch

    BMP2SHHCUX1ETV5IGSF8NARSURI1DNLZ

    SBSNKLK10KRT17SOX9FSTPARM1LAMA3

    CTNNB1POSTNH2AFZTIMP2KIF23TUBB2ABGNDLK1APOEVCANVIMASPHFMR1

    regulation of cell cycledevelopment differentiationepidermal cell population proliferation

    intermediate filament organization

    GO enrichment dynamics along pseudotime

    endocytosis virus host

    lipid metabolic process

    process metabolic cellular

    folding 'de novo'

    start

    cell fate 1 cell fate 2

    regulation response process

    1

    2

    3

    PCNA

    log10( + 0.1)

    1

    2

    3

    TOP2A

    component 1

    com

    pone

    nt 2

    Cel

    l fat

    e 1

    Cel

    l fat

    e 2

    1

    2

    3

    KRT17

    log10( + 0.1)

    BMP2/K15/DAPI LEF1/PCNA/DAPI

    Hi Hi

    low low

    low low

    Hi Hi

    VDR/PCNA/DAPI CTNNB1/DAPI

    (b)

    (c) (d)

    1

    2

    3

    4

    .CC-BY-NC-ND 4.0 International license(which was not certified by peer review) is the author/funder. It is made available under aThe copyright holder for this preprintthis version posted January 31, 2020. . https://doi.org/10.1101/2020.01.30.926287doi: bioRxiv preprint

    https://doi.org/10.1101/2020.01.30.926287http://creativecommons.org/licenses/by-nc-nd/4.0/