iBet uBet web content aggregator. Adding the entire web to your favor.
iBet uBet web content aggregator. Adding the entire web to your favor.



Link to original content: https://www.mdpi.com/2079-7737/13/10/842
Dermal Papilla Cells: From Basic Research to Translational Applications
Next Article in Journal
MeHA: A Computational Framework in Revealing the Genetic Basis of Animal Mental Health Traits Under an Intensive Farming System—A Case Study in Pigs
Previous Article in Journal
The Impact of Eucalyptus and Pine Plantations on the Taxonomic and Functional Diversity of Dung Beetles (Coleoptera: Scarabaeidae) in the Southern Region of Ecuador
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Dermal Papilla Cells: From Basic Research to Translational Applications

1
School of Environmental and Chemical Engineering, Shanghai University, Shanghai 200444, China
2
School of Life Sciences, Shanghai University, Shanghai 200444, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biology 2024, 13(10), 842; https://doi.org/10.3390/biology13100842
Submission received: 26 August 2024 / Revised: 13 October 2024 / Accepted: 18 October 2024 / Published: 20 October 2024
(This article belongs to the Special Issue Stem Cells in Experimental Medicine)

Simple Summary

The dermal papilla (DP) is a specialized mesenchymal compartment that forms at the base of the hair follicle. It acts as a signaling center that regulates hair growth, shape, size, and color. Dermal papilla cells (DPCs) exhibit stem cell properties and show significant potential for use in cell therapy. They can be induced into other functional cells, regenerate skin tissues and new hair follicles when combined with epithelial stem cells, and promote hair growth and wound healing when administered on the skin. This review summarizes the functions of DPs in the hair follicle, shows how DPCs could be used in cell therapy, and discusses the challenges and recent advances in the field, from basic research to translational applications.

Abstract

As an appendage of the skin, hair protects against ultraviolet radiation and mechanical damage and regulates body temperature. It also reflects an individual’s health status and serves as an important method of expressing personality. Hair loss and graying are significant psychosocial burdens for many people. Hair is produced from hair follicles, which are exclusively controlled by the dermal papilla (DP) at their base. The dermal papilla cells (DPCs) comprise a cluster of specialized mesenchymal cells that induce the formation of hair follicles during early embryonic development through interaction with epithelial precursor cells. They continue to regulate the growth cycle, color, size, and type of hair after the hair follicle matures by secreting various factors. DPCs possess stem cell characteristics and can be cultured and expanded in vitro. DPCs express numerous stemness-related factors, enabling them to be reprogrammed into induced pluripotent stem cells (iPSCs) using only two, or even one, Yamanaka factor. DPCs are an important source of skin-derived precursors (SKPs). When combined with epithelial stem cells, they can reconstitute skin and hair follicles, participating in the regeneration of the dermis, including the DP and dermal sheath. When implanted between the epidermis and dermis, DPCs can induce the formation of new hair follicles on hairless skin. Subcutaneous injection of DPCs and their exosomes can promote hair growth. This review summarizes the in vivo functions of the DP; highlights the potential of DPCs in cell therapy, particularly for the treatment of hair loss; and discusses the challenges and recent advances in the field, from basic research to translational applications.

1. Introduction

Hair is generated by the skin appendage, the hair follicle, through interactions between epithelial and mesenchymal cells [1]. At the base of the hair follicle lies a structure called the dermal papilla (DP), which is formed by specialized mesenchymal cells during development and continues to release signaling molecules to the epithelial cells after hair follicle maturation, controlling hair growth, shape, size, and color [2,3,4,5]. Dermal papilla cells (DPCs) are a unique type of mesenchymal cell that can be cultured and expanded in vitro [6], and increasing evidence suggests their potential in cell therapy. This article reviews the functions of DPCs in hair follicle development and hair growth, provides a detailed description of their potential applications as specialized mesenchymal stem cells (MSCs), discusses recent research progress, and critically analyzes the challenges and limitations in translating these findings into clinical practice, highlighting the areas in which breakthroughs are needed to overcome these barriers.

2. Formation of DP and Its Function During Hair Follicle Morphogenesis

Basic research in mice reveals that hair follicle development during embryogenesis involves a series of reciprocal interactions between epithelial and mesenchymal cells [7]. Initially, dermal mesenchymal cells release signals to the epithelial cells, inducing epithelium thickening to form the placode. Conversely, the placode signals to the underlying dermal mesenchymal cells, inducing cell proliferation to form dermal condensates (DC) [8]. The reciprocal signaling interactions induce invagination and a downward extension of the basal cells of the placode, which ultimately envelop the DC. This process results in the maturation of the DP from the DC and ultimately leads to the maturation of the hair follicle [4]. During hair follicle formation, inner basal cells differentiate into the inner root sheath (IRS), which encapsulates the future hair shaft [9], while outer basal cells differentiate into the outer root sheath (ORS) [10]. The bulge region within the ORS contains hair follicle stem cells (HFSCs) and melanocyte stem cells [11]. The ORS cells differentiate into highly proliferative matrix cells at the bulb region, which surround the DP [12,13]. Melanocyte stem cells migrate and differentiate alongside HFSCs, and mature melanocytes reside within the matrix cells [14,15].
Key signaling pathways, such as the wingless/integrated (WNT), sonic hedgehog (SHH), and bone morphogenetic protein (BMP) pathways, are crucial in this process (Figure 1).
The WNT signal is recognized as the “first dermal signal” in HF morphogenesis [16], which precedes and is essential for the localized expression of regulatory genes and the initiation of HF placode formation from a uniform single layer of the basal epidermis [16,17]. The WNT activity in the placode epithelium promotes dermal cell proliferation and induces dermal condensate (DC) formation [18,19]. The β-catenin protein is uniformly activated in the upper dermis before its activation in the placode epithelium, followed by its activation in the underlying mesenchyme and DC [20,21]. The feedback from the dermal WNT then promotes the full induction of the placode signals [19,21].
SHH pathway, which acts downstream of the WNT signaling, is expressed in the epithelial cells and the DC, playing a crucial role in signaling from the epithelium to both the epithelial and mesenchymal cells [22,23]. SHH promotes the proliferation of the HF epithelium and its subsequent downward growth, as well as the formation of the DC [24]. While the induction of HFs is independent of SHH, its role in the ingrowth of the epidermis and the subsequent morphogenesis of the hair shaft is essential [23,25]. SHH signaling regulates specific DP signatures and maintains a reciprocal SHH–Noggin (BMP inhibitor) signaling loop to drive hair follicle morphogenesis [23,26].
BMP signaling acts in an inhibitory manner during HF morphogenesis. The ligands BMP2 and BMP4 are enriched in the placode and DC [27,28]. BMP signaling, along with its reporters and antagonists, exhibits a strict spatiotemporal expression pattern to properly regulate cell proliferation and differentiation in the epidermis and HFs [29,30]. The BMP receptor BMPR1A, expressed in HFs, is crucial for the differentiation of IRS and hair shaft progenitor cells [31,32]. Without BMPR1A activation, the terminal differentiation factor GATA-3 is downregulated, compromising its regulatory control over IRS differentiation [33]. BMP signaling is essential for HFSC quiescence and the promotion of TAC differentiation along different lineages during the hair cycle [34,35].
It is noteworthy that dermal fibroblasts and thus, DP, originate from different precursor cells in different parts of the body. Lineage tracing assays in mice show that dermal fibroblasts in the dorsal and ventral trunk originate from the somite, whereas dermal fibroblasts in the head and face originate from the neural crest [36,37]. The neural crest cells in birds and mice also migrate to the skin and give rise to melanocytes during embryogenesis [37,38]. Ablation of the WNT signaling in the mouse dermis leads to ectopic cartilage formation in the craniofacial and ventral trunk regions at the expense of the dermal and bone lineages [39]. Dermo-1/twist2 (twist basic helix-loop-helix transcription factor 2), a downstream transcription factor of WNT signaling, may mediate the function of WNT signaling in dermal cell development [39,40]. Dermo-1 knockout mice showed radically thin skin and sparse hair [41]. The forced expression of chicken Dermo-1 induces a dense dermis, feathers, and scales [42]. These findings highlight the significance of Dermo-1 in dermis maturation from different embryonic origins, which is crucial for hair follicle development and patterning.
In summary, DC/DP serves as a signaling center for hair follicle development, driving epithelial proliferation, placode formation, and downward growth of the hair follicle. This process involves diverse interactions between epithelial and mesenchymal cells, ultimately leading to the formation of different hair follicle lineage cells and the maturation of hair follicles [43,44].

3. DP Regulation in Mature Hair Follicles

3.1. DP Regulates Hair Growth/Cycle

After the maturation of the hair follicle, hair undergoes cyclical growth. The mature DP continues to serve as the signaling center for the hair follicles, finely regulating the quiescence, activation, proliferation, and differentiation of hair follicle epithelial cells, thereby controlling the hair cycle (Figure 1) [45,46]. During the growth phase (anagen), hair germ cells, which are derived directly from bulge HFSCs, proliferate to form transient amplifying matrix cells (TACs) [47]. These TACs surround the DPCs and receive signals from them, ultimately differentiating into hair shafts. In the regression phase (catagen), matrix cell proliferation ceases, and skin cell apoptosis occurs, leading to the degeneration of two-thirds of the lower follicle, while DPCs remain intact and migrate upwards along the epithelium [48]. In the resting phase (telogen), epithelial cells of the hair follicle remain in a quiescent state, with the DP located at the tip of the hair follicle, directly in contact with hair germ cells [49,50]. When quiescent hair germ cells receive activating signals from the DP, they rapidly proliferate into progenitor cells, which then differentiate into various mature cells, initiating a new hair growth cycle [50]. It should be noted that in mice, the growth cycle of all hairs on the back is synchronized until the second anagen, whereas in the adult human scalp, individual hairs maintain their own growth cycles. At any given time, approximately 90% of human scalp hair follicles are in the anagen phase, 1% are in the catagen phase, and 9% are in the telogen phase [51].
DPCs not only secrete factors such as WNTs [52], R-Spondins [53], transforming growth factor-β2 (TGF-β2) [54], hepatocyte growth factor (HGF) [55], and insulin-like growth factor 1 (IGF1) [56] to activate the WNT pathway and promote the proliferation of HFSCs [57], but also express the growth factors fibroblast growth factor 7 (FGF7) and FGF10 [58] to enhance the SHH signal in the hair follicle epithelial cells (Figure 1). The SHH signal acts downstream of WNT to stimulate the neighboring epithelial cells of the hair follicle [59], thereby accelerating hair growth [60].
Among these factors, DP-expressed R-Spondins potentiate WNT signaling to stimulate the proliferation of both HFSCs and epithelial progenitors during HF regeneration [61]. TGF-β2 activates the Smad2/3 pathway in HF stem cells, and antagonizes BMP signaling in HFSCs, thereby promoting the transition from the telogen to the anagen phase of the hair cycle [62]. HGF stimulated WNT/β-catenin activity in the hair matrix by upregulating WNT6 and WNT10B and inhibiting secreted frizzled-related protein 1 (SFRP1) in DP [55]. IGF-1 acts as an anti-apoptotic survival factor to inhibit cell death during the catagen phase of the hair cycle [63], as well as stimulates cell proliferation to promote hair growth [64]. The FGF and WNT signaling pathways interact in the DP to regulate the hair cycle by orchestrating the expression of WNT agonists (R-Spondins) and antagonists (Dickkopf-related protein 2 (DKK2) and Notum) [65]. Furthermore, DPCs express high levels of BMP pathway-related proteins such as BMP4, BMP6, BMP7, BMP receptor BMPR1A, and BMP inhibitor Noggin [66,67], which are involved in the complex mechanisms governing the transition of the hair follicle from the growth phase to the resting phase [68]. Noggin, with a 10–15 times higher affinity for BMP4 compared to that of BMPR-1A, likely prevents BMP4 interaction with BMPR-IA expressed in the secondary germ of the telogen hair follicles, thereby stimulating anagen initiation [69].
Previous studies have shown that the selective ablation of certain DPCs in mice delays the growth phase of the hair follicles [2]. Furthermore, the complete laser ablation of DPCs prevents hair follicles from entering the growth phase [70]. Under chronic stress, the hormone corticosterone, derived from the adrenal gland, inhibits growth arrest-specific gene 6 (GAS6) in DPCs, governing HFSCs’ quiescence and leading to hair loss [71]. Previous studies also suggest that the androgen receptor (AR), which is specifically expressed in DPCs in the skin, may be involved in androgenic alopecia [72].

3.2. DP Regulates Hair Properties

In mammals, melanocytes determine the pigment deposition in the skin and hair [73]. The expression of melanin from melanocytes in the back skin of mice strictly follows the hair growth cycle, peaking in the late growth phase [74]. Previous studies have reported that DPCs regulate melanocytes via the WNT signaling pathway [75]. The plucking of hair from the backs of mice leads to the upregulation of the secretory factor endothelin 3 (EDN3) in the DPCs [76], and the deletion of EDN3 leads to coat white spotting in mice [76,77]. It has also been found that AGOUTI (also known as ASIP, agouti-signaling protein), specifically expressed in DPCs, competes with α-melanocyte-stimulating hormone (α-MSH) for binding to the melanocortin 1 receptor (MC1R), promoting the production of red/yellow pheomelanin instead of black/brown eumelanin [78,79,80]. Transient expression of AGOUTI in mice causes the production of a yellow-striped band near the tip of the black hair shaft. The serine protease CORIN is specifically expressed in DPCs during the growth phase and acts downstream of AGOUTI, inhibiting its pathway and balancing the production of pheomelanin and eumelanin [81]. The sex determining region Y-box 2 (SOX2) in DP regulates hair pigmentation through the upregulation of AGOUTI and the downregulation of CORIN [82,83].
The types of hair on the backs of mice include guard, awl, auchene, and zigzag, which form at different stages of development [84,85]. Guard hairs are the longest and smoothest, emerging the earliest during embryonic day 14.5; awl hairs are larger and blunter, appearing from embryonic day 16.5; auchene hairs, characterized by their bent middle, are the least abundant and also appear from embryonic day 16.5; zigzag hairs have 3–4 bends and are the most abundant, appearing from embryonic day 18.5 [84,85,86]. DPCs determine the different hair types [6,27]. SOX2 is expressed only in guard, awl, and auchene DP but not in zigzag DP. The removal of SOX2+ cells from DP results in the reconstitution of hair with only zigzag hairs, without the expression of awl and auchene hairs [87]. Sox18 knockout mice display a mild coat defect, with a reduced proportion of zigzag hairs and an increased proportion of auchene hairs [88,89]. Our recent study has demonstrated that the transcription factor early B cell factor-1 (EBF1), which is prominently expressed in DP, also plays a role in determining the type and length of hair. Specifically, the conditional knockout of EBF1 in DPCs leads to a reduction in hair length, a decrease in the amount of awl hair, and an increase in the amount of zigzag hair [90].
The number of DPCs within each hair follicle correlates with and determines the size and shape of the hair. Anatomical examination of hair follicles at postnatal day 11 reveals significant variations in the number of DPCs among the four types of follicles. As one type of hair transitions into larger and longer hairs, the number of DPCs significantly increases [2,91,92]. The selective ablation of DPCs in vivo using genetic mice models results in smaller hairs [2]. The genome sequencing of dogs with varying hair morphology has linked DP-expressed R-Spondin 2 (RSPO2) to properties such as the length and curliness of dog hair [93]. Scientists have discovered that the Hoxc gene family (HOXCs), expressed in mice DPCs, influences the activation of the downstream WNT signaling pathway, thereby affecting the length of hair across different parts of the mouse body [94].
In summary, the number and gene expression of DPCs correlate with or even determine hair properties such as hair color, size, type, and morphology, suggesting that they play an important role in hair formation.

4. Technological Advances in DP Basic Research and Establishment of Genetically Modified Mice as a Tool for Studying Functions of DP Genes

In the past, studies on the regulation of hair growth primarily focused on HFSCs, thanks to the creation of various genetically modified mice, such as K14-Cre, for specific gene knockout in skin epithelium [95]; K14-rtTA, for specific gene overexpression in skin epithelium [96]; and K19-CreERT, for specific gene knockout in HFSCs [97]. Due to the important role of DP in hair regulation, there has been increasing research on the functions of genes in DP. The mice reported as tools for gene knockout in DP include Tbx18-Cre [82,98], Corin-Cre [5], Sox2-CreER [99] and CD133-CreERT 2 [100,101], Hey2-CreERT2 [102], Lepr-Cre [90], and LeprB-Cre [103]. The agouti gene, which regulates hair coat color, is confined to expression in the DP after birth, but is widely expressed in dermal fibroblast precursors during embryonic development. Consequently, Agouti-Cre mice will induce gene knockout in both the DP and all dermal fibroblasts [104]. Tbx18-Cre targets precursor cells in DP [82,98], Corin-Cre targets DPCs after hair follicle formation [5], Sox2-CreER targets DPCs in guard, awl, and auchene hair follicles [99], and CD133-CreERT2 targets some DPCs [100,101].
These Cre mice help to reveal the regulatory functions of DP genes such as SOX2 [82,83,87,99,105], β-catenin [5,101], Fgfr1 and Fgfr2 [65], and EBF1 [90] in regards to hair properties. For example, the ablation or activation of β-Catenin, specifically in mice DP reveals, that β-Catenin in DP functions by switching the hair color from yellow to black, as well as by promoting postnatal hair growth [5,101]. Mice with Fgfr1/2 double knockout, specifically in DP, exhibit extremely long hairs [65].
One reason scientists continue to search for Cre mice is that existing mice lack specificity. Since DPCs originate from mesenchymal cells in the dermis [60,106], the Cre recombinase in these mice is more or less expressed in other dermal cells similar to DPCs, such as DS cells connected to DP and a small number of other dermal fibroblasts. Due to the similarity between DP and DS [91,107], it is difficult to achieve DP-specific expression using a single gene promoter to control Cre expression. However, using two promoters, such as those active in genes expressed in both DP and DS, along with those active specifically near the DP niche (e.g., Lef1), may eventually achieve DP-specific expression using both the Cre-LoxP and Dre-Rox systems [108]. It is worth mentioning that CreER mice can also be used to trace the lineage of DPCs in vivo [102], revealing whether they generate DS cells, whether they participate in dermal fibroblast generation during wound healing, and whether they participate in new DS and DP generation during hair follicle neogenesis after injury [109]. Obtaining definitive conclusions still requires the acquisition of dual-labeled, DP-specific CreER mice.
Another reason may be the significant cost required to transport these mice between different countries. In addition to Cre mice, establishing rTTA mice for gene overexpression specifically in DP will further facilitate the study of gene functions in DP [101].

5. Stemness and Potential Applications of DPCs in Cell Therapy

As the signaling centers for regulating hair follicle development and hair growth, DPCs express high levels of numerous growth factors. DPCs are particularly notable for their stem cell properties. This unique property makes them very promising for the development of cell products for cell therapy.

5.1. Reprogramming of DPCs into iPSCs

Compared with other somatic cells, DPCs express various stem cell factors and are more easily induced into pluripotent stem cells (iPSCs) [110]. Typically, the induction of mice embryonic or adult fibroblasts into iPSCs requires the addition of four transcription factors, i.e., octamer-binding transcription factor 4 (OCT4), SOX2, Krüppel-like factor 4 (KLF4), and and cellular myelocytomatosis oncogene (C-MYC) [111]. However, DPCs themselves express SOX2, C-MYC, and KLF4, so their induction into iPSCs only requires the addition of OCT4 and KLF4, or even just OCT4 [112,113]. Under the same conditions, the efficiency of reprogramming DPCs into iPSCs is about three times higher than that of fibroblasts [106].

5.2. DPCs Are a Main Source of SKPs

Skin-derived precursors (SKPs) are multipotent progenitor cells in the skin that are capable of self-renewal and display the characteristics of multipotent neural stem cells [114]. Freda D. Miller’s laboratory initially dissociated mice dermal tissues into single cells and cultured them in a medium containing epidermal growth factor (EGF) or FGF, where some cells formed floating spheres and proliferated during passage [115]. These cells differentiate along neuronal and glial cell lineages, while also expressing markers of MSCs, differentiating into smooth muscle cells, adipocytes, and other types of cells. Subsequently, the same laboratory found that SKPs derived from adult dermis possess characteristics similar to those of embryonic neural crest stem cells (NCSCs), with DP providing a niche for SKPs. Cell lineage tracing has shown that both mice hair and whisker follicle DP contain NCSCs, and whisker pad SKPs also originate from NCSCs, suggesting that DPCs are likely a main source of SKPs [116]. Further studies have shown that SOX2+ cells are distributed in DP and the lower parts of the dermal sheath (DS), and SKPs and SOX2+ cells cultured in vitro exhibit similar gene expression profiles and functions, i.e., both can reconstitute hair follicles when combined with epithelial cells from neonatal mice, differentiate into dermal fibroblasts, and regenerate DP and DS [117].
SKPs can also differentiate into adipocytes, hepatocytes, keratinocytes, melanocytes, and other cell types in vitro [118,119]. Steinbach et al. demonstrated that rat SKPs can be easily differentiated into functional vascular smooth muscle cells in vitro, laying the foundation for SKPs’ application in the treatment of ischemic diseases [120]. When transplanted into a normal or injured hippocampus, mice SKPs can survive and maintain their neural characteristics for at least five weeks [121]. When transplanted into the dysmyelinated brain, naive rodents or human SKPs can generate Schwann cells and myelinate central nervous system (CNS) axons [122,123]. SKPs can be used to treat spinal cord injuries by generating Schwann cells [123]. It has also been reported that SKPs transplanted into the aganglionic colon of pigs survive and express neuroglial differentiation markers [124,125]. SKPs differentiate into skeletogenic cell types in vivo to contribute to bone repair [126]. Human SKP-derived corneal endothelial cell-like cells transplanted into a monkey model of corneal endothelial dysfunction maintain functionality for two years [127,128].
DPCs can differentiate into adipocytes and osteoblasts using specific differentiation mediums, although whether DPCs have the same differentiation potential as SKPs remains to be verified [129]. Nevertheless, the fact that DPCs are a main source of SKPs indicates there is enormous potential for SKP-based cell therapies.
It is worth noting that Nestin-expressing hair follicle-accessible pluripotent stem cells (HAP) [130] are likely terminal Schwann cells that wrap around the hair follicle isthmus [131]. Human hair follicle-derived mesenchymal stem cells (HF-MSC) are derived from the DP and DS cells grown in a culture medium from isolated hair follicles [132]. HAP and HF-MSC both show characteristics of SKPs and are likely sources of SKPs, although their isolation methods are different.

5.3. DPCs and Their Derived Exosomes Promote Wound Healing

It has been reported that MSCs and their extracellular vesicles have significant potential to promote tissue repair and regeneration [133]. DPCs, as a special type of MSC, can differentiate into dermal fibroblasts during skin regeneration and wound healing; additionally, various growth factors and cytokines, either from DPCs or their derived exosomes, can promote angiogenesis, reduce inflammation, and facilitate wound healing [102,117,134,135,136,137].

5.4. DPCs Induce Hair Follicle Neogenesis In Vivo

Removal of the lower third of the whisker follicle leads to the loss of the ability to regenerate whiskers when the follicle is transplanted into ear skin. However, freshly isolated or culture-expanded DPCs implanted into the bases of rodent whisker follicles can restore the ability to grow whiskers [138,139]. Implanting DPCs between the epidermis and dermis of the hairless footpads in rats induces de novo hair follicle formation and hair growth [140]. Similarly, implanting DPCs between the epidermis and dermis of the human foreskin can induce hair growth on hairless skin [141]. Implanting DPCs into wounds on rat ears and backs has been found to aid in wound healing and induce new hair follicles [134,138]. Mixing DPCs with epithelial stem cells and transplanting them onto the backs of nude mice can reconstitute hairy skin [67,142,143,144].

5.5. DPCs and Their Derived Exosomes Promote Hair Growth

DPCs secrete various bioactive molecules such as growth factors and cytokines, which promote the proliferation and differentiation of HFSCs. The subcutaneous injection of mice DPCs can induce hair follicles from the resting phase into the growth phase and delay the onset of the regression phase, thereby promoting hair growth [145,146,147]. Recent studies have identified similar bioactive molecules in exosomes from cultured dermal papilla cells; injecting cell-free exosomes subcutaneously can also improve the scalp microenvironment, activate HFSCs, and promote hair growth [145,146,148,149,150]. The application of DPC-derived exosomes onto mouse full-thickness skin excisional wounds also promotes hair follicle neogenesis [137]. Therefore, the use of DPCs and their exosomes provides a potential therapeutic strategy for hair loss.

6. Strategies and Challenges of DPCs-Based Cell Therapy for Treating Alopecia

Human hair performs a wide range of functions, including thermoregulation, sensation of touch, and protection against ultraviolet radiation and mechanical damage [47,151,152]. Hair also reflects an individual’s health status, and hairstyles serve as an important method of self-expression. Loss of hair (alopecia) lowers an individual’s self-esteem and causes psychological distress [153].
Current treatments for alopecia do not meet the clinical needs of patients. For example, hair transplantation represents one of the most popular treatments for alopecia [154]. However, this method is constrained by the limited number of hair follicles available in the donor area and the cost of the treatment. Minoxidil and finasteride are the only two medications approved by the U.S. Food and Drug Administration for the treatment of alopecia, yet these treatments offer limited efficacy and cause significant side effects [155,156].
Based on their functions in inducing hair follicle neogenesis and promoting hair growth, DPCs can be applied in two potential strategies to treat alopecia: one involves reconstituting hair follicles, along with epithelial stem cells, and the other involves the subcutaneous injection of DPCs or their derived exosomes to promote hair growth (Figure 2). For the former strategy, the direct reconstitution of hairy skin on a patient’s scalp is impractical for the following reasons: (1) Current hair follicle reconstitution techniques in mice are based on hairy skin regeneration during wound healing, resulting in scar-like skin with impaired functionality [157]; (2) The density, size, and direction of hairs regenerated through spontaneous skin reconstitution from stem cells are uncontrollable [158]; (3) The reconstitution of colored hair also requires the inclusion of melanocyte stem cells in the reconstitution system [159]. An alternative approach involves reconstituting human follicles in immunodeficient large animals (e.g., pigs) and transplanting them to human scalps. However, hair follicles reconstituted in animals inevitably incorporate animal cells, such as vascular endothelial cells and dermal cells, raising questions about immune reactions and the survival of hair follicles after transplantation into human scalps. Furthermore, reconstituting hairy skin requires not only a large number of autologous DPCs but also melanocyte stem cells and HFSCs, which currently cannot be sufficiently maintained and expanded in vitro (Figure 2).
To implement the latter strategy, it is essential to isolate and expand DPCs on a large scale. In the case of autologous DPCs, a small number of healthy DPCs must be expanded in vitro through numerous passages while retaining their hair follicle induction activities. It is important to consider the potential for immune rejection from allogeneic DPCs and their exosomes. While primary DPCs can be obtained in large quantities from animals, there are significant concerns regarding immune rejection and ethical barriers (Figure 2). A common limitation of cell therapy is the poor survival of transplanted cells due to the immune rejection of allogeneic cells and failure to integrate the transplanted cells into the recipient tissues/organs [160]. To reduce immune rejection, the deletion of MHC (major histocompatibility complex) expression in transplanted cells and pretreatment with immunosuppressive drugs are common measures [161,162]. Strategies to reduce apoptotic signaling and increase cell adhesion, such as pretreatment with cytokines, growth factors, and anti-apoptotic molecules, have been developed to improve the long-term survival of transplanted cells [163]. These pretreatments can likely be applied topically to the scalp when using DPCs to treat alopecia. In addition to traditional drug therapies and hair transplantation, new therapies have been developed for the treatment of alopecia. For example, platelet-rich plasma (PRP) [164], MSCs [165], and MSCs-derived exosomes [166] or conditioned media [167] are injected into the scalp to stimulate HFSCs. A common mechanism behind these therapies is that the injected substances contain growth factors. DPCs are unique in that they accumulate high concentrations of growth factors that activate HFSCs, such as R-Spondins, VEGF, FGF, HGF, etc. [102,168]. DPCs-based cell therapy would offer significant advantages over other therapies for the treatment of alopecia.

7. Technological Advances in DP Translational Research—Obtaining DPCs on a Large Scale

Obtaining sufficient numbers of DPCs is the main obstacle to DPCs-based cell therapy. To solve this problem, scientists have made significant technological advances, including the large-scale isolation of DPCs using surface protein antibodies, the improvement of culture conditions to allow massive expansion of DPCs in vitro, and a technique for inducing iPSCs into DPCs, which provides an unlimited source of DPCs. These technologies not only advance translational research, including using DPCs as a source of SKPs that differentiate into other functional cells such as neurons, reprogramming DPCs into iPSCs, and using DPCs to promote wound healing and hair follicle regeneration or hair growth to treat hair loss, but also provide DPCs for basic research, including studying the functions of DP genes in vitro (Figure 3).

7.1. Large Scale Isolation of DPCs

To study the functions of DP in hair follicle development, hair cycle regulation, and hair regeneration, or to apply DPCs in disease treatment, the isolation and in vitro culture of DPCs are indispensable. Traditional isolation methods include microdissection, which involves the use of a scalpel under a stereomicroscope to physically separate dozens of DPCs from a single hair follicle [169,170]. this method is time-consuming and is only suitable for large hair follicles, such as mice whisker follicles and human scalp hair follicles. it has been reported that dpcs can be sorted using antibodies against the surface protein cd133/prom1, followed by flow cytometry [171]. however, cd133 is a general stem/progenitor cell marker [172], not highly specific to dpcs, and is expressed in other cells such as ors cells and matrix cells in the skin [173]. there are reports of using genetically labeled dpcs in mice via fluorescent proteins under the control of the dp-specific lef1 promoter [66,67,174]. however, this method relies on the construction of transgenic animals and has not been reported in species other than mice.
Our laboratory has identified a dp membrane protein, leptin receptor (lepr), and developed a more specific and efficient technology for functional dpc sorting on a large scale [168]. by analyzing the gene expression profile of dp cells using existing microarray and rna-seq databases, lepr was identified as the second most abundant surface protein in dp cells, exhibiting a higher level of specificity compared to that of the previously utilized cd133 marker [66,168,175]. the lepr antibody-based sorting method allows for the isolation of hundreds of thousands of dp cells in a few hours, a significant improvement in efficiency over that of traditional microdissection. Furthermore, LEPR+ cells sorted by this method retain their DP characteristics in vitro, as demonstrated by the expression of DP markers (such as alkaline phosphatase) and key secretory factors (such as RSPO1/2 and EDN3), as well as the successful reconstitution of hair follicles in nude mice when combined with epithelial stem cells [168].

7.2. Expansion of DPCs In Vitro

While undergoing culturing in a Petri dish, DPCs gradually lose their biological activity or stemness, especially the ability to induce hair follicle formation [176,177]. Generally, growth beyond the sixth passage of DPCs begins to slow down, accompanied by noticeable morphological changes such as the loss of aggregative growth [178,179]. In vivo, DPCs interact closely with epithelial cells during hair follicle development, with mature DPCs in direct contact with or even enveloped by epithelial cells [60]. The culturing of skin epithelial cells on feeder fibroblasts mimics the in vivo epithelial–mesenchymal cell interaction and has achieved long-term expansion [180]. Co-culturing DPCs with skin epithelial cells not only maintains the stemness of the epithelial cells [181], but also greatly extends the passage number of the DPCs. Similar effects are observed when DPCs are cultured using a skin epithelial cell-conditioned medium [147]. Under these conditions, DPCs can still maintain their hair follicle-inducing ability, even after 70 passages [182,183]. It has been reported that adding the growth factor FGF2 to DPCs can extend their passage number from 5 generations to over 30 generations [176].
Another successful approach is to culture DPCs into spheres using a three-dimensional suspension culture method, which facilitates cell–cell contact and enhances the ability of DPCs to induce hair follicle formation [141,176,184].
Additionally, immortalized DPCs have been established by introducing the SV40 large T antigen and the telomerase (TERT) gene [182,185,186]. These cell lines maintain the expression characteristics of DPCs, but whether or not they retain the ability to induce hair follicle formation is not mentioned.

7.3. Induction of iPSCs into DPCs

Another approach for obtaining a continuous supply of DPCs is through their induction from iPSCs. Scientists first induced iPSCs into neural crest stem cells (NCSCs) using recombinant Noggin and an inhibitor of activin/nodal and TGFβ signaling, and then further induced NCSCs into SKPs using a WNT signal activator in an SKP medium [187]. A similar trajectory was adopted to induce human iPSC differentiation into DP-like cells [188].
In another study, scientists differentiated iPSCs into induced mesenchymal cells (iMC) via a bone marrow stromal cell phenotype. Subsequently, using retinoic acid and a DP cell-activating culture medium, they reprogrammed a subset of LNGFR+ THY-1+ iMC to acquire DPC properties. When co-grafted with human keratinocytes in vivo, the induced DPCs produced fiber structures with a hair cuticle-like coat resembling the hair shaft [189]. Alternatively, scientists have directly reprogramed fibroblasts into DP-like cells using small molecules [190,191].

8. Conclusions and Perspectives

DPCs serve as the signaling centers for hair follicles, playing a crucial role in hair follicle development and the hair growth cycle. With recent technological advancements, DPCs have garnered increasing attention from scientists, and growing studies are exploring the functions of different genes in DPCs. It is noteworthy that the expression of different genes in DPCs undergoes dynamic changes during the hair cycle, likely involving secretory factors that promote and maintain various phases of hair growth. Hair growth is a finely tuned process in which the coordination of various secretory factors by DPCs determines the growth patterns, including initiation, termination, and pigmentation. Collecting DPCs at different time points and analyzing their expression profiles to reveal the sequential changes in DP genes and their interactions will greatly aid in understanding the hair growth signaling network. This knowledge will also establish a solid theoretical foundation for studying the mechanisms of pathological hair loss or graying.
As a unique type of MSC, DPCs can also serve as a cell source for cell therapy in various diseases, especially skin disorders like alopecia. Overcoming the barriers to DPC-based cell therapy involves maintaining stemness, while increasing passage numbers in 2D culturing, achieving cost-effective and simplified expansion in 3D culturing, and enhancing the long-term survival and functional performance of DPCs after subcutaneous injections. Continuously obtaining DPCs through iPSC induction represents a viable new approach, but improving the efficiency of iPSC induction and fully establishing this technological route will require further dedicated work from scientists.

Author Contributions

H.-L.Z.: materials collection; writing—original draft. X.-X.Q.: materials collection; writing—original draft. X.-H.L.: conceptualization, supervision, writing—review and editing, and funding acquisition. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the National Key Research and Development Program of China (funding number: 2020YFA01130002) and the National Natural Science Foundation of China (funding number: 81972563).

Conflicts of Interest

The authors declared no conflicts of interest.

References

  1. Chase, H.B. Growth of the hair. Physiol. Rev. 1954, 34, 113–126. [Google Scholar] [CrossRef] [PubMed]
  2. Chi, W.; Wu, E.; Morgan, B.A. Dermal papilla cell number specifies hair size, shape and cycling and its reduction causes follicular decline. Development 2013, 140, 1676–1683. [Google Scholar] [CrossRef] [PubMed]
  3. Elliott, K.; Messenger, A.G.; Stephenson, T.J. Differences in hair follicle dermal papilla volume are due to extracellular matrix volume and cell number: Implications for the control of hair follicle size and androgen responses. J. Investig. Dermatol. 1999, 113, 873–877. [Google Scholar] [CrossRef] [PubMed]
  4. Driskell, R.R.; Clavel, C.; Rendl, M.; Watt, F.M. Hair follicle dermal papilla cells at a glance. J. Cell Sci. 2011, 124, 1179–1182. [Google Scholar] [CrossRef]
  5. Enshell-Seijffers, D.; Lindon, C.; Wu, E.; Taketo, M.M.; Morgan, B.A. β-Catenin activity in the dermal papilla of the hair follicle regulates pigment-type switching. Proc. Natl. Acad. Sci. USA 2010, 107, 21564–21569. [Google Scholar] [CrossRef]
  6. Gan, Y.; Wang, H.; Du, L.; Li, K.; Qu, Q.; Liu, W.; Sun, P.; Fan, Z.; Wang, J.; Chen, R.; et al. Cellular Heterogeneity Facilitates the Functional Differences Between Hair Follicle Dermal Sheath Cells and Dermal Papilla Cells: A New Classification System for Mesenchymal Cells within the Hair Follicle Niche. Stem Cell Rev. Rep. 2022, 18, 2016–2027. [Google Scholar] [CrossRef]
  7. Hardy, M.H. The secret life of the hair follicle. Trends Genet. 1992, 8, 55–61. [Google Scholar] [CrossRef]
  8. Widelitz, R.B.; Chuong, C.-M. Early events in skin appendage formation: Induction of epithelial placodes and condensation of dermal mesenchyme. Investig. Dermatol. Symp. Proc. 1999, 4, 302–306. [Google Scholar] [CrossRef]
  9. Kaufman, C.K.; Zhou, P.; Pasolli, H.A.; Rendl, M.; Bolotin, D.; Lim, K.-C.; Dai, X.; Alegre, M.-L.; Fuchs, E. GATA-3: An unexpected regulator of cell lineage determination in skin. Genes Dev. 2003, 17, 2108–2122. [Google Scholar] [CrossRef]
  10. Okuyama, R.; Nguyen, B.-C.; Talora, C.; Ogawa, E.; di Vignano, A.T.; Lioumi, M.; Chiorino, G.; Tagami, H.; Woo, M.; Dotto, G. High commitment of embryonic keratinocytes to terminal differentiation through a Notch1-caspase 3 regulatory mechanism. Dev. Cell 2004, 6, 551–562. [Google Scholar] [CrossRef]
  11. Ohyama, M. Hair follicle bulge: A fascinating reservoir of epithelial stem cells. J. Dermatol. Sci. 2007, 46, 81–89. [Google Scholar] [CrossRef] [PubMed]
  12. Legué, E.; Nicolas, J.-F. Hair follicle renewal: Organization of stem cells in the matrix and the role of stereotyped lineages and behaviors. Development 2005, 132, 4143–4154. [Google Scholar] [CrossRef]
  13. Blanpain, C.; Lowry, W.E.; Geoghegan, A.; Polak, L.; Fuchs, E. Self-renewal, multipotency, and the existence of two cell populations within an epithelial stem cell niche. Cell 2004, 118, 635–648. [Google Scholar] [CrossRef]
  14. Lavker, R.M.; Sun, T.T. Epidermal stem cells: Properties, markers, and location. Proc. Natl. Acad. Sci. USA 2000, 97, 13473–13475. [Google Scholar] [CrossRef] [PubMed]
  15. Huang, L.; Zuo, Y.; Li, S.; Li, C. Melanocyte stem cells in the skin: Origin, biological characteristics, homeostatic maintenance and therapeutic potential. Clin. Transl. Med. 2024, 14, e1720. [Google Scholar] [CrossRef]
  16. Saxena, N.; Mok, K.-W.; Rendl, M. An updated classification of hair follicle morphogenesis. Exp. Dermatol. 2019, 28, 332–344. [Google Scholar] [CrossRef]
  17. Andl, T.; Reddy, S.T.; Gaddapara, T.; Millar, S.E. WNT Signals Are Required for the Initiation of Hair Follicle Development. Dev. Cell 2002, 2, 643–653. [Google Scholar] [CrossRef] [PubMed]
  18. Hu, X.-M.; Li, Z.-X.; Zhang, D.-Y.; Yang, Y.-C.; Fu, S.-A.; Zhang, Z.-Q.; Yang, R.-H.; Xiong, K. A systematic summary of survival and death signalling during the life of hair follicle stem cells. Stem Cell Res. Ther. 2021, 12, 453. [Google Scholar] [CrossRef]
  19. Chen, D.; Jarrell, A.; Guo, C.; Lang, R.; Atit, R. Dermal β-catenin activity in response to epidermal Wnt ligands is required for fibroblast proliferation and hair follicle initiation. Development 2012, 139, 1522–1533. [Google Scholar] [CrossRef]
  20. Zhu, N.; Yan, J.; Gu, W.; Yang, Q.; Lin, E.; Lu, S.; Cai, B.; Xia, B.; Liu, X.; Lin, C. Dermal papilla cell-secreted biglycan regulates hair follicle phase transit and regeneration by activating Wnt/β-catenin. Exp. Dermatol. 2024, 33, e14969. [Google Scholar] [CrossRef]
  21. Enshell-Seijffers, D.; Lindon, C.; Kashiwagi, M.; Morgan, B.A. beta-catenin activity in the dermal papilla regulates morphogenesis and regeneration of hair. Dev. Cell 2010, 18, 633–642. [Google Scholar] [CrossRef] [PubMed]
  22. Huelsken, J.; Vogel, R.; Erdmann, B.; Cotsarelis, G.; Birchmeier, W. beta-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin. Cell 2001, 105, 533–545. [Google Scholar] [CrossRef] [PubMed]
  23. Chiang, C.; Swan, R.Z.; Grachtchouk, M.; Bolinger, M.; Litingtung, Y.; Robertson, E.K.; Cooper, M.K.; Gaffield, W.; Westphal, H.; Beachy, P.A.; et al. Essential Role forSonic hedgehogduring Hair Follicle Morphogenesis. Dev. Biol. 1999, 205, 1–9. [Google Scholar] [CrossRef]
  24. Millar, S.E. Molecular Mechanisms Regulating Hair Follicle Development. J. Investig. Dermatol. 2002, 118, 216–225. [Google Scholar] [CrossRef]
  25. St-Jacques, B.; Dassule, H.R.; Karavanova, I.; Botchkarev, V.A.; Li, J.; Danielian, P.S.; McMahon, J.A.; Lewis, P.M.; Paus, R.; McMahon, A.P. Sonic hedgehog signaling is essential for hair development. Curr. Biol. 1998, 8, 1058–1069. [Google Scholar] [CrossRef]
  26. Woo, W.M.; Zhen, A.E.; Oro, A.E. Shh maintains dermal papilla identity and hair morphogenesis via a Noggin-Shh regulatory loop. Genes Dev. 2012, 26, 1235–1246. [Google Scholar] [CrossRef] [PubMed]
  27. Ge, W.; Tan, S.-J.; Wang, S.-H.; Li, L.; Sun, X.-F.; Shen, W.; Wang, X. Single-cell Transcriptome Profiling reveals Dermal and Epithelial cell fate decisions during Embryonic Hair Follicle Development. Theranostics 2020, 10, 7581–7598. [Google Scholar] [CrossRef]
  28. Li, X.; Xie, R.; Luo, Y.; Shi, R.; Ling, Y.; Zhao, X.; Xu, X.; Chu, W.; Wang, X. Cooperation of TGF-β and FGF signalling pathways in skin development. Cell Prolif. 2023, 56, e13489. [Google Scholar] [CrossRef]
  29. Plikus, M.V.; Mayer, J.A.; de la Cruz, D.; Baker, R.E.; Maini, P.K.; Maxson, R.; Chuong, C.-M. Cyclic dermal BMP signalling regulates stem cell activation during hair regeneration. Nature 2008, 451, 340–344. [Google Scholar] [CrossRef]
  30. Botchkarev, V.A. and A.A. Sharov, BMP signaling in the control of skin development and hair follicle growth. Differentiation 2004, 72, 512–526. [Google Scholar] [CrossRef]
  31. Zhang, G.; Chu, M.; Yang, H.; Li, H.; Shi, J.; Feng, P.; Wang, S.; Pan, Z. Expression, Polymorphism, and Potential Functional Sites of the BMPR1A Gene in the Sheep Horn. Genes 2024, 15, 376. [Google Scholar] [CrossRef] [PubMed]
  32. Sunkara, R.R.; Mehta, D.; Sarate, R.M.; Waghmare, S.K. BMP-AKT-GSK3β Signaling Restores Hair Follicle Stem Cells Decrease Associated with Loss of Sfrp1. Stem Cells 2022, 40, 802–817. [Google Scholar] [CrossRef]
  33. Kobielak, K.; Pasolli, H.A.; Alonso, L.; Polak, L.; Fuchs, E. Defining BMP functions in the hair follicle by conditional ablation of BMP receptor IA. J. Cell Biol. 2003, 163, 609–623. [Google Scholar] [CrossRef] [PubMed]
  34. Genander, M.; Cook, P.J.; Ramsköld, D.; Keyes, B.E.; Mertz, A.F.; Sandberg, R.; Fuchs, E. BMP Signaling and Its pSMAD1/5 Target Genes Differentially Regulate Hair Follicle Stem Cell Lineages. Cell Stem Cell 2014, 15, 619–633. [Google Scholar] [CrossRef]
  35. Guha, U.; Mecklenburg, L.; Cowin, P.; Kan, L.; O’Guin, W.M.; D’Vizio, D.; Pestell, R.G.; Paus, R.; Kessler, J.A. Bone morphogenetic protein signaling regulates postnatal hair follicle differentiation and cycling. Am. J. Pathol. 2004, 165, 729–740. [Google Scholar] [CrossRef]
  36. Jinno, H.; Morozova, O.; Jones, K.L.; Biernaskie, J.A.; Paris, M.; Hosokawa, R.; Rudnicki, M.A.; Chai, Y.; Rossi, F.; Marra, M.A.; et al. Convergent genesis of an adult neural crest-like dermal stem cell from distinct developmental origins. Stem Cells 2010, 28, 2027–2040. [Google Scholar] [CrossRef] [PubMed]
  37. Wong, C.E.; Paratore, C.; Dours-Zimmermann, M.T.; Rochat, A.; Pietri, T.; Suter, U.; Zimmermann, D.R.; Dufour, S.; Thiery, J.P.; Meijer, D.; et al. Neural crest-derived cells with stem cell features can be traced back to multiple lineages in the adult skin. J. Cell Biol. 2006, 175, 1005–1015. [Google Scholar] [CrossRef]
  38. Stocker, K.M.; Brown, A.M.; Ciment, G. Gene transfer of lacZ into avian neural tube and neural crest cells by retroviral infection of grafted embryonic tissues. J. Neurosci. Res. 1993, 34, 135–145. [Google Scholar] [CrossRef] [PubMed]
  39. Tran, T.H.; Jarrell, A.; Zentner, G.E.; Welsh, A.; Brownell, I.; Scacheri, P.C.; Atit, R. Role of canonical Wnt signaling/ß-catenin via Dermo1 in cranial dermal cell development. Development. 2010, 137, 3973–3984. [Google Scholar] [CrossRef]
  40. Li, L.; Cserjesi, P.; Olson, E.N. Dermo-1: A Novel Twist-Related bHLH Protein Expressed in the Developing Dermis. Dev. Biol. 1995, 172, 280–292. [Google Scholar]
  41. Šošić, D.; Richardson, J.A.; Yu, K.; Ornitz, D.M.; Olson, E.N. Twist Regulates Cytokine Gene Expression through a Negative Feedback Loop that Represses NF-κB Activity. Cell 2003, 112, 169–180. [Google Scholar] [CrossRef] [PubMed]
  42. Hornik, C.; Krishan, K.; Yusuf, F.; Scaal, M.; Brand-Saberi, B. cDermo-1 misexpression induces dense dermis, feathers, and scales. Dev. Biol. 2005, 277, 42–50. [Google Scholar] [CrossRef] [PubMed]
  43. Ramos, R.; Guerrero-Juarez, C.F.; Plikus, M.V. Hair Follicle Signaling Networks: A Dermal Papilla–Centric Approach. J. Investig. Dermatol. 2013, 133, 2306–2308. [Google Scholar] [CrossRef] [PubMed]
  44. Sennett, R.; Rendl, M. Mesenchymal–epithelial interactions during hair follicle morphogenesis and cycling. Semin. Cell Dev. Biol. 2012, 23, 917–927. [Google Scholar] [CrossRef]
  45. Alonso, L.; Fuchs, E. The hair cycle. J. Cell Sci. 2006, 119, 391–393. [Google Scholar] [CrossRef]
  46. Matsuzaki, T.; Yoshizato, K. Role of hair papilla cells on induction and regeneration processes of hair follicles. Wound Repair. Regen. 1998, 6, 524–530. [Google Scholar] [CrossRef]
  47. Schneider, M.R.; Schmidt-Ullrich, R.; Paus, R. The hair follicle as a dynamic miniorgan. Curr. Biol. 2009, 19, R132–R142. [Google Scholar] [CrossRef] [PubMed]
  48. Lindner, G.; A Botchkarev, V.; Botchkareva, N.V.; Ling, G.; Van Der Veen, C.; Paus, R. Analysis of apoptosis during hair follicle regression (catagen). Am. J. Pathol. 1997, 151, 1601. [Google Scholar]
  49. Wosicka, H.; Cal, K. Targeting to the hair follicles: Current status and potential. J. Dermatol. Sci. 2010, 57, 83–89. [Google Scholar] [CrossRef]
  50. Lin, X.; Zhu, L.; He, J. Morphogenesis, Growth Cycle and Molecular Regulation of Hair Follicles. Front. Cell Dev. Biol. 2022, 10, 899095. [Google Scholar] [CrossRef]
  51. Burg, D.; Yamamoto, M.; Namekata, M.; Haklani, J.; Koike, K.; Halasz, M. Promotion of anagen, increased hair density and reduction of hair fall in a clinical setting following identification of FGF5-inhibiting compounds via a novel 2-stage process. Clin. Cosmet. Investig. Dermatol. 2017, 10, 71–85. [Google Scholar] [CrossRef] [PubMed]
  52. Kandyba, E.; Kobielak, K. Wnt7b is an important intrinsic regulator of hair follicle stem cell homeostasis and hair follicle cycling. Stem Cells 2014, 32, 886–901. [Google Scholar] [CrossRef] [PubMed]
  53. Smith, A.A.; Li, J.; Liu, B.; Hunter, D.; Pyles, M.; Gillette, M.; Dhamdhere, G.R.; Abo, A.; Oro, A.; Helms, J.A. Activating Hair Follicle Stem Cells via R-spondin2 to Stimulate Hair Growth. J. Investig. Dermatol. 2016, 136, 1549–1558. [Google Scholar] [CrossRef] [PubMed]
  54. Soma, T.; Tsuji, Y.; Hibino, T. Involvement of transforming growth factor-beta2 in catagen induction during the human hair cycle. J. Investig. Dermatol. 2002, 118, 993–997. [Google Scholar] [CrossRef] [PubMed]
  55. Nicu, C.; O’sullivan, J.D.; Ramos, R.; Timperi, L.; Lai, T.; Farjo, N.; Farjo, B.; Pople, J.; Bhogal, R.; Hardman, J.A.; et al. Dermal Adipose Tissue Secretes HGF to Promote Human Hair Growth and Pigmentation. J. Investig. Dermatol. 2021, 141, 1633–1645.e13. [Google Scholar] [CrossRef]
  56. Tang, L.; Bernardo, O.; Bolduc, C.; Lui, H.; Madani, S.; Shapiro, J. The expression of insulin-like growth factor 1 in follicular dermal papillae correlates with therapeutic efficacy of finasteride in androgenetic alopecia. J. Am. Acad. Dermatol. 2003, 49, 229–233. [Google Scholar] [CrossRef]
  57. Morgan, B.A. The dermal papilla: An instructive niche for epithelial stem and progenitor cells in development and regeneration of the hair follicle. Cold Spring Harb. Perspect. Med. 2014, 4, a015180. [Google Scholar] [CrossRef]
  58. Ohuchi, H.; Tao, H.; Ohata, K.; Itoh, N.; Kato, S.; Noji, S.; Ono, K. Fibroblast growth factor 10 is required for proper development of the mouse whiskers. Biochem. Biophys. Res. Commun. 2003, 302, 562–567. [Google Scholar] [CrossRef]
  59. Greco, V.; Chen, T.; Rendl, M.; Schober, M.; Pasolli, H.A.; Stokes, N.; dela Cruz-Racelis, J.; Fuchs, E. A Two-Step Mechanism for Stem Cell Activation during Hair Regeneration. Cell Stem Cell 2009, 4, 155–169. [Google Scholar] [CrossRef]
  60. Blanpain, C.; Fuchs, E. Epidermal stem cells of the skin. Annu. Rev. Cell Dev. Biol. 2006, 22, 339–373. [Google Scholar] [CrossRef]
  61. Hagner, A.; Shin, W.; Sinha, S.; Alpaugh, W.; Workentine, M.; Abbasi, S.; Rahmani, W.; Agabalyan, N.; Sharma, N.; Sparks, H.; et al. Transcriptional Profiling of the Adult Hair Follicle Mesenchyme Reveals R-spondin as a Novel Regulator of Dermal Progenitor Function. iScience 2020, 23, 101019. [Google Scholar] [CrossRef] [PubMed]
  62. Oshimori, N.; Fuchs, E. Paracrine TGF-β Signaling Counterbalances BMP-Mediated Repression in Hair Follicle Stem Cell Activation. Cell Stem Cell 2012, 10, 63–75. [Google Scholar] [CrossRef] [PubMed]
  63. Stewart, C.E.; Rotwein, P. Growth, differentiation, and survival: Multiple physiological functions for insulin-like growth factors. Physiol. Rev. 1996, 76, 1005–1026. [Google Scholar] [CrossRef] [PubMed]
  64. Li, J.; Yang, Z.; Li, Z.; Gu, L.; Wang, Y.; Sung, C. Exogenous IGF-1 promotes hair growth by stimulating cell proliferation and down regulating TGF-β1 in C57BL/6 mice in vivo. Growth Horm. IGF Res. 2014, 24, 89–94. [Google Scholar] [CrossRef]
  65. Harshuk-Shabso, S.; Dressler, H.; Niehrs, C.; Aamar, E.; Enshell-Seijffers, D. Fgf and Wnt signaling interaction in the mesenchymal niche regulates the murine hair cycle clock. Nat. Commun. 2020, 11, 5114. [Google Scholar] [CrossRef] [PubMed]
  66. Rendl, M.; Lewis, L.; Fuchs, E. Molecular dissection of mesenchymal-epithelial interactions in the hair follicle. PLoS Biol. 2005, 3, e331. [Google Scholar] [CrossRef]
  67. Rendl, M.; Polak, L.; Fuchs, E. BMP signaling in dermal papilla cells is required for their hair follicle-inductive properties. Genes Dev. 2008, 22, 543–557. [Google Scholar] [CrossRef]
  68. Hsu, Y.-C.; Li, L.; Fuchs, E. Emerging interactions between skin stem cells and their niches. Nat. Med. 2014, 20, 847–856. [Google Scholar] [CrossRef]
  69. Botchkarev, V.A. Bone Morphogenetic Proteins and Their Antagonists in Skin and Hair Follicle Biology. J. Investig. Dermatol. 2003, 120, 36–47. [Google Scholar] [CrossRef]
  70. Rompolas, P.; Deschene, E.R.; Zito, G.; Gonzalez, D.G.; Saotome, I.; Haberman, A.M.; Greco, V. Live imaging of stem cell and progeny behaviour in physiological hair-follicle regeneration. Nature 2012, 487, 496–499. [Google Scholar] [CrossRef]
  71. Choi, S.; Zhang, B.; Ma, S.; Gonzalez-Celeiro, M.; Stein, D.; Jin, X.; Kim, S.T.; Kang, Y.-L.; Besnard, A.; Rezza, A.; et al. Corticosterone inhibits GAS6 to govern hair follicle stem-cell quiescence. Nature 2021, 592, 428–432. [Google Scholar] [CrossRef] [PubMed]
  72. Deng, Z.; Chen, M.; Liu, F.; Wang, Y.; Xu, S.; Sha, K.; Peng, Q.; Wu, Z.; Xiao, W.; Liu, T.; et al. Androgen Receptor-Mediated Paracrine Signaling Induces Regression of Blood Vessels in the Dermal Papilla in Androgenetic Alopecia. J. Investig. Dermatol. 2022, 142, 2088–2099.e9. [Google Scholar] [CrossRef] [PubMed]
  73. Rabbani, P.; Takeo, M.; Chou, W.; Myung, P.; Bosenberg, M.; Chin, L.; Taketo, M.M.; Ito, M. Coordinated Activation of Wnt in Epithelial and Melanocyte Stem Cells Initiates Pigmented Hair Regeneration. Cell 2011, 145, 941–955. [Google Scholar] [CrossRef] [PubMed]
  74. O’Sullivan, J.D.B.; Nicu, C.; Picard, M.; Chéret, J.; Bedogni, B.; Tobin, D.J.; Paus, R. The biology of human hair greying. Biol. Rev. Camb. Philos. Soc. 2021, 96, 107–128. [Google Scholar] [CrossRef]
  75. Chi, W.; Enshell-Seijffers, D.; Morgan, B.A. De novo production of dermal papilla cells during the anagen phase of the hair cycle. J. Investig. Dermatol. 2010, 130, 2664–2666. [Google Scholar] [CrossRef]
  76. Li, H.; Fan, L.; Zhu, S.; Shin, M.K.; Lu, F.; Qu, J. Epilation induces hair and skin pigmentation through an EDN3/EDNRB-dependent regenerative response of melanocyte stem cells. Sci. Rep. 2017, 7, 7272. [Google Scholar] [CrossRef]
  77. Baynash, A.G.; Hosoda, K.; Giaid, A.; Richardson, J.A.; Emoto, N.; Hammer, R.E.; Yanagisawa, M. Interaction of endothelin-3 with endothelin-B receptor is essential for development of epidermal melanocytes and enteric neurons. Cell 1994, 79, 1277–1285. [Google Scholar] [CrossRef]
  78. Lu, D.; Willard, D.; Patel, I.R.; Kadwell, S.; Overton, L.; Kost, T.; Luther, M.; Chen, W.; Woychik, R.P.; Wilkison, W.O.; et al. Agouti protein is an antagonist of the melanocyte-stimulating-hormone receptor. Nature 1994, 371, 799–802. [Google Scholar] [CrossRef]
  79. Leeb, T.; Deppe, A.; Kriegesmann, B.; Brenig, B. Genomic structure and nucleotide polymorphisms of the porcine agouti signalling protein gene (ASIP). Anim. Genet. 2000, 31, 335–336. [Google Scholar] [CrossRef]
  80. Barsh, G.; Gunn, T.; He, L.; Schlossman, S.; Duke-Cohan, J. Biochemical and genetic studies of pigment-type switching. Pigment Cell Res. 2000, 13, 48–53. [Google Scholar] [CrossRef]
  81. Enshell-Seijffers, D.; Lindon, C.; Morgan, B.A. The serine protease Corin is a novel modifier of the agouti pathway. Development 2008, 135, 217–225. [Google Scholar] [CrossRef] [PubMed]
  82. Clavel, C.; Grisanti, L.; Zemla, R.; Rezza, A.; Barros, R.; Sennett, R.; Mazloom, A.R.; Chung, C.-Y.; Cai, X.; Cai, C.-L.; et al. Sox2 in the Dermal Papilla Niche Controls Hair Growth by Fine-Tuning BMP Signaling in Differentiating Hair Shaft Progenitors. Dev. Cell 2012, 23, 981–994. [Google Scholar] [CrossRef] [PubMed]
  83. Ng, K.J.; Lim, J.; Tan, Y.N.; Quek, D.; Lim, Z.; Pantelireis, N.; Clavel, C. Sox2 in the dermal papilla regulates hair follicle pigmentation. Cell Rep. 2022, 40, 111100. [Google Scholar] [CrossRef] [PubMed]
  84. Duverger, O.; Morasso, M.I. Epidermal patterning and induction of different hair types during mouse embryonic development. Birth Defects Res. C Embryo Today 2009, 87, 263–272. [Google Scholar] [CrossRef] [PubMed]
  85. Sundberg, J.P.; Hogan, M.E. Hair types and subtypes in the laboratory mouse. In Handbook of Mouse Mutations with Skin and Hair Abnormalities; CRC Press: Boca Raton, FL, USA, 2020; pp. 57–68. [Google Scholar]
  86. Schlake, T. Determination of hair structure and shape. Semin. Cell Dev. Biol. 2007, 18, 267–273. [Google Scholar] [CrossRef]
  87. Driskell, R.R.; Giangreco, A.; Jensen, K.B.; Mulder, K.W.; Watt, F.M. Sox2-positive dermal papilla cells specify hair follicle type in mammalian epidermis. Development 2009, 136, 2815–2823. [Google Scholar] [CrossRef]
  88. Pennisi, D.; Bowles, J.; Nagy, A.; Muscat, G.; Koopman, P. Mice null for sox18 are viable and display a mild coat defect. Mol. Cell Biol. 2000, 20, 9331–9336. [Google Scholar] [CrossRef]
  89. James, K.; Hosking, B.; Gardner, J.; Muscat, G.E.O.; Koopman, P. Sox18 mutations in the ragged mouse alleles ragged-like and opossum. Genesis 2003, 36, 1–6. [Google Scholar] [CrossRef]
  90. Song, H.; Zhang, L.; Zhong, W.-Q.; Chen, E.Q.; Qiu, X.-X.; Tang, Z.-L.; Liao, X.-H. EBF1 expressed in the dermal papilla regulates hair type and length. Genes Dis. 2024, 12, 101261. [Google Scholar] [CrossRef]
  91. Rahmani, W.; Abbasi, S.; Hagner, A.; Raharjo, E.; Kumar, R.; Hotta, A.; Magness, S.; Metzger, D.; Biernaskie, J. Hair Follicle Dermal Stem Cells Regenerate the Dermal Sheath, Repopulate the Dermal Papilla, and Modulate Hair Type. Dev. Cell 2014, 31, 543–558. [Google Scholar] [CrossRef]
  92. Ibrahim, L.; Wright, E.A. A quantitative study of hair growth using mouse and rat vibrissal follicles. I. Dermal papilla volume determines hair volume. J. Embryol. Exp. Morphol. 1982, 72, 209–224. [Google Scholar] [PubMed]
  93. Cadieu, E.; Neff, M.W.; Quignon, P.; Walsh, K.; Chase, K.; Parker, H.G.; VonHoldt, B.M.; Rhue, A.; Boyko, A.; Byers, A.; et al. Coat variation in the domestic dog is governed by variants in three genes. Science 2009, 326, 150–153. [Google Scholar] [CrossRef] [PubMed]
  94. Yu, Z.; Jiang, K.; Xu, Z.; Huang, H.; Qian, N.; Lu, Z.; Chen, D.; Di, R.; Yuan, T.; Du, Z.; et al. Hoxc-Dependent Mesenchymal Niche Heterogeneity Drives Regional Hair Follicle Regeneration. Cell Stem Cell 2018, 23, 487–500.e6. [Google Scholar] [CrossRef] [PubMed]
  95. Vasioukhin, V.; Degenstein, L.; Wise, B.; Fuchs, E. The magical touch: Genome targeting in epidermal stem cells induced by tamoxifen application to mouse skin. Proc. Natl. Acad. Sci. USA 1999, 96, 8551–8556. [Google Scholar] [CrossRef] [PubMed]
  96. Nguyen, H.; Rendl, M.; Fuchs, E. Tcf3 governs stem cell features and represses cell fate determination in skin. Cell 2006, 127, 171–183. [Google Scholar] [CrossRef]
  97. Means, A.L.; Xu, Y.; Zhao, A.; Ray, K.C.; Gu, G. A CK19(CreERT) knockin mouse line allows for conditional DNA recombination in epithelial cells in multiple endodermal organs. Genesis 2008, 46, 318–323. [Google Scholar] [CrossRef]
  98. Grisanti, L.; Clavel, C.; Cai, X.; Rezza, A.; Tsai, S.-Y.; Sennett, R.; Mumau, M.; Cai, C.-L.; Rendl, M. Tbx18 Targets Dermal Condensates for Labeling, Isolation, and Gene Ablation during Embryonic Hair Follicle Formation. J. Investig. Dermatol. 2013, 133, 344–353. [Google Scholar] [CrossRef]
  99. Arnold, K.; Sarkar, A.; Yram, M.A.; Polo, J.M.; Bronson, R.; Sengupta, S.; Seandel, M.; Geijsen, N.; Hochedlinger, K. Sox2+ Adult Stem and Progenitor Cells Are Important for Tissue Regeneration and Survival of Mice. Cell Stem Cell 2011, 9, 317–329. [Google Scholar] [CrossRef]
  100. Zhou, L.; Yang, K.; Carpenter, A.; Lang, R.A.; Andl, T.; Zhang, Y. CD133-positive dermal papilla-derived Wnt ligands regulate postnatal hair growth. Biochem. J. 2016, 473, 3291–3305. [Google Scholar] [CrossRef]
  101. Zhou, L.; Xu, M.; Yang, Y.; Yang, K.; Wickett, R.R.; Andl, T.; Millar, S.E.; Zhang, Y. Activation of beta-Catenin Signaling in CD133-Positive Dermal Papilla Cells Drives Postnatal Hair Growth. PLoS ONE 2016, 11, e0160425. [Google Scholar]
  102. Wang, Y.G.; Yuan, V.L.; Liao, X.H. Genetic lineage tracing in skin reveals predominant expression of HEY2 in dermal papilla during telogen and that HEY2(+) cells contribute to the regeneration of dermal cells during wound healing. Exp. Dermatol. 2023, 32, 2176–2179. [Google Scholar] [CrossRef] [PubMed]
  103. Liu, Y.; Guerrero-Juarez, C.F.; Xiao, F.; Shettigar, N.U.; Ramos, R.; Kuan, C.-H.; Lin, Y.-C.; Lomeli, L.d.J.M.; Park, J.M.; Oh, J.W.; et al. Hedgehog signaling reprograms hair follicle niche fibroblasts to a hyper-activated state. Dev. Cell 2022, 57, 1758–1775.e7. [Google Scholar] [CrossRef] [PubMed]
  104. Shen, X.-R.; Zhang, H.-L.; Zhao, X.-B.; Wang, Y.-G.; Tan, X.-Y.; Gao, L.; Sun, R.; Liao, X.-H. A Cre knockin mouse reveals specific expression of Agouti gene in mesenchymal lineage cells in multiple organs and provides a unique tool for conditional gene targeting. Transgenic Res. 2023, 32, 143–152. [Google Scholar] [CrossRef]
  105. Lesko, M.H.; Driskell, R.R.; Kretzschmar, K.; Goldie, S.J.; Watt, F.M. Sox2 modulates the function of two distinct cell lineages in mouse skin. Dev. Biol. 2013, 382, 15–26. [Google Scholar] [CrossRef]
  106. Muchkaeva, I.A.; Dashinimaev, E.B.; Artyuhov, A.S.; Myagkova, E.P.; Vorotelyak, E.A.; Yegorov, Y.Y.; Vishnyakova, K.S.; Kravchenko, J.E.; Chumakov, S.P.; Terskikh, V.V.; et al. Generation of iPS Cells from Human Hair Follice Dermal Papilla Cells. Acta Naturae 2014, 6, 45–53. [Google Scholar] [CrossRef] [PubMed]
  107. Yamao, M.; Inamatsu, M.; Ogawa, Y.; Toki, H.; Okada, T.; Toyoshima, K.-E.; Yoshizato, K. Contact between dermal papilla cells and dermal sheath cells enhances the ability of DPCs to induce hair growth. J. Investig. Dermatol. 2010, 130, 2707–2718. [Google Scholar] [CrossRef] [PubMed]
  108. He, L.; Li, Y.; Li, Y.; Pu, W.; Huang, X.; Tian, X.; Wang, Y.; Zhang, H.; Liu, Q.; Zhang, L.; et al. Enhancing the precision of genetic lineage tracing using dual recombinases. Nat. Med. 2017, 23, 1488–1498. [Google Scholar] [CrossRef]
  109. Martino, P.; Sunkara, R.; Heitman, N.; Rangl, M.; Brown, A.; Saxena, N.; Grisanti, L.; Kohan, D.; Yanagisawa, M.; Rendl, M. Progenitor-derived endothelin controls dermal sheath contraction for hair follicle regression. Nat. Cell Biol. 2023, 25, 222–234. [Google Scholar] [CrossRef]
  110. Hibberts, N.A.; Messenger, A.G.; Randall, V.A. Dermal papilla cells derived from beard hair follicles secrete more stem cell factor (SCF) in culture than scalp cells or dermal fibroblasts. Biochem. Biophys. Res. Commun. 1996, 222, 401–405. [Google Scholar] [CrossRef]
  111. Takahashi, K.; Yamanaka, S.J.C. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 2006, 126, 663–676. [Google Scholar] [CrossRef]
  112. Tsai, S.-Y.; Clavel, C.; Kim, S.; Ang, Y.-S.; Grisanti, L.; Lee, D.-F.; Kelley, K.; Rendl, M. Oct4 and klf4 reprogram dermal papilla cells into induced pluripotent stem cells. Stem Cells 2010, 28, 221–228. [Google Scholar] [CrossRef] [PubMed]
  113. Tsai, S.-Y.; Bouwman, B.A.; Ang, Y.-S.; Kim, S.J.; Lee, D.-F.; Lemischka, I.R.; Rendl, M. Single Transcription Factor Reprogramming of Hair Follicle Dermal Papilla Cells to Induced Pluripotent Stem Cells. Stem Cells 2011, 29, 964–971. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, Z.; Pradhan, S.; Liu, C.; Le, L.Q. Skin-derived precursors as a source of progenitors for cutaneous nerve regeneration. Stem Cells 2012, 30, 2261–2270. [Google Scholar] [CrossRef]
  115. Toma, J.G.; Akhavan, M.; Fernandes, K.J.L.; Barnabé-Heider, F.; Sadikot, A.; Kaplan, D.R.; Miller, F.D. Isolation of multipotent adult stem cells from the dermis of mammalian skin. Nat. Cell Biol. 2001, 3, 778–784. [Google Scholar] [CrossRef]
  116. Fernandes, K.J.L.; McKenzie, I.A.; Mill, P.; Smith, K.M.; Akhavan, M.; Barnabé-Heider, F.; Biernaskie, J.; Junek, A.; Kobayashi, N.R.; Toma, J.G.; et al. A dermal niche for multipotent adult skin-derived precursor cells. Nat. Cell Biol. 2004, 6, 1082–1093. [Google Scholar] [CrossRef]
  117. Biernaskie, J.; Paris, M.; Morozova, O.; Fagan, B.M.; Marra, M.; Pevny, L.; Miller, F.D. SKPs derive from hair follicle precursors and exhibit properties of adult dermal stem cells. Cell Stem Cell 2009, 5, 610–623. [Google Scholar] [CrossRef] [PubMed]
  118. De Kock, J.; Snykers, S.; Ramboer, E.; Demeester, S.; Heymans, A.; Branson, S.; Vanhaecke, T.; Rogiers, V. Evaluation of the multipotent character of human foreskin-derived precursor cells. Toxicol. In Vitro 2011, 25, 1191–1202. [Google Scholar] [CrossRef]
  119. Li, L.; Fukunaga-Kalabis, M.; Yu, H.; Xu, X.; Kong, J.; Lee, J.T.; Herlyn, M. Human dermal stem cells differentiate into functional epidermal melanocytes. J. Cell Sci. 2010, 123 Pt 6, 853–860. [Google Scholar] [CrossRef]
  120. Steinbach, S.K.; El-Mounayri, O.; DaCosta, R.S.; Frontini, M.J.; Nong, Z.; Maeda, A.; Pickering, J.G.; Miller, F.D.; Husain, M. Directed differentiation of skin-derived precursors into functional vascular smooth muscle cells. Arter. Thromb. Vasc. Biol. 2011, 31, 2938–2948. [Google Scholar] [CrossRef]
  121. Fernandes, K.J.; Kobayashi, N.R.; Gallagher, C.J.; Barnabé-Heider, F.; Aumont, A.; Kaplan, D.R.; Miller, F.D. Analysis of the neurogenic potential of multipotent skin-derived precursors. Exp. Neurol. 2006, 201, 32–48. [Google Scholar] [CrossRef]
  122. McKenzie, I.A.; Biernaskie, J.; Toma, J.G.; Midha, R.; Miller, F.D. Skin-derived precursors generate myelinating Schwann cells for the injured and dysmyelinated nervous system. J. Neurosci. 2006, 26, 6651–6660. [Google Scholar] [CrossRef] [PubMed]
  123. Biernaskie, J.; Sparling, J.S.; Liu, J.; Shannon, C.P.; Plemel, J.R.; Xie, Y.; Miller, F.D.; Tetzlaff, W. Skin-derived precursors generate myelinating Schwann cells that promote remyelination and functional recovery after contusion spinal cord injury. J. Neurosci. 2007, 27, 9545–9559. [Google Scholar] [CrossRef] [PubMed]
  124. Thomas, A.-L.; Taylor, J.S.; Dunn, J.C.Y. Human skin-derived precursor cells xenografted in aganglionic bowel. J. Pediatr. Surg. 2020, 55, 2791–2796. [Google Scholar] [CrossRef]
  125. Thomas, A.-L.; Taylor, J.S.; Huynh, N.; Dubrovsky, G.; Chadarevian, J.-P.; Chen, A.; Baker, S.; Dunn, J.C. Autologous Transplantation of Skin-Derived Precursor Cells in a Porcine Model. J. Pediatr. Surg. 2020, 55, 194–200. [Google Scholar] [CrossRef]
  126. Lavoie, J.-F.; Biernaskie, J.A.; Chen, Y.; Bagli, D.; Alman, B.; Kaplan, D.R.; Miller, F.D. Skin-derived precursors differentiate into skeletogenic cell types and contribute to bone repair. Stem Cells Dev. 2009, 18, 893–906. [Google Scholar] [CrossRef]
  127. Shen, L.; Sun, P.; Du, L.; Zhu, J.; Ju, C.; Guo, H.; Wu, X. Long-Term Observation and Sequencing Analysis of SKPs-Derived Corneal Endothelial Cell-Like Cells for Treating Corneal Endothelial Dysfunction. Cell Transpl. 2021, 30, 9636897211017830. [Google Scholar] [CrossRef]
  128. Shen, L.; Sun, P.; Zhang, C.; Yang, L.; Du, L.; Wu, X. Therapy of corneal endothelial dysfunction with corneal endothelial cell-like cells derived from skin-derived precursors. Sci. Rep. 2017, 7, 13400. [Google Scholar] [CrossRef] [PubMed]
  129. Luo, X.; Liu, J.; Zhang, P.; Yu, Y.; Wu, B.; Jia, Q.; Liu, Y.; Xiao, C.; Cao, Y.; Jin, H.; et al. Isolation, characterization and differentiation of dermal papilla cells from Small-tail Han sheep. Anim. Biotechnol. 2023, 34, 3475–3482. [Google Scholar] [CrossRef]
  130. Hoffman, R.M.; Amoh, Y. Hair Follicle-Associated Pluripotent(HAP) Stem Cells. Prog. Mol. Biol. Transl. Sci. 2018, 160, 23–28. [Google Scholar]
  131. Song, H.; Zhao, X.; Chu, Q.; Zhang, J.; Gao, L.; Liao, X. Expression dynamics of lymphoid enhancer-binding factor 1 in terminal Schwann cells, dermal papilla, and interfollicular epidermis. Dev. Dyn. 2023, 252, 527–535. [Google Scholar] [CrossRef]
  132. Wang, B.; Liu, X.-M.; Liu, Z.-N.; Wang, Y.; Han, X.; Lian, A.-B.; Mu, Y.; Jin, M.-H.; Liu, J.-Y. Human hair follicle-derived mesenchymal stem cells: Isolation, expansion, and differentiation. World J. Stem Cells 2020, 12, 462–470. [Google Scholar] [CrossRef] [PubMed]
  133. Wu, P.; Zhang, B.; Shi, H.; Qian, H.; Xu, W. MSC-exosome: A novel cell-free therapy for cutaneous regeneration. Cytotherapy 2018, 20, 291–301. [Google Scholar] [CrossRef] [PubMed]
  134. Gharzi, A.; Reynolds, A.J.; Jahoda, C.A. Plasticity of hair follicle dermal cells in wound healing and induction. Exp. Dermatol. 2003, 12, 126–136. [Google Scholar] [CrossRef]
  135. Leirós, G.J.; Kusinsky, A.G.; Drago, H.; Bossi, S.; Sturla, F.; Castellanos, M.L.; Stella, I.Y.; Balañá, M.E. Dermal papilla cells improve the wound healing process and generate hair bud-like structures in grafted skin substitutes using hair follicle stem cells. Stem Cells Transl. Med. 2014, 3, 1209–1219. [Google Scholar] [CrossRef] [PubMed]
  136. Wang, Y.; Shen, K.; Sun, Y.; Cao, P.; Zhang, J.; Zhang, W.; Liu, Y.; Zhang, H.; Chen, Y.; Li, S.; et al. Extracellular vesicles from 3D cultured dermal papilla cells improve wound healing via Kruppel-like factor 4/vascular endothelial growth factor A -driven angiogenesis. Burns Trauma 2023, 11, tkad034. [Google Scholar] [CrossRef]
  137. Shang, Y.; Li, M.; Zhang, L.; Han, C.; Shen, K.; Wang, K.; Li, Y.; Zhang, Y.; Luo, L.; Jia, Y.; et al. Exosomes derived from mouse vibrissa dermal papilla cells promote hair follicle regeneration during wound healing by activating Wnt/beta-catenin signaling pathway. J. Nanobiotechnol. 2024, 22, 425. [Google Scholar] [CrossRef]
  138. Jahoda, C.A.; Horne, K.A.; Oliver, R.F. Induction of hair growth by implantation of cultured dermal papilla cells. Nature 1984, 311, 560–562. [Google Scholar] [CrossRef]
  139. Oliver, R.F. The experimental induction of whisker growth in the hooded rat by implantation of dermal papillae. J. Embryol. Exp. Morphol. 1967, 18, 43–51. [Google Scholar] [CrossRef]
  140. Reynolds, A.J.; Jahoda, C.A. Cultured dermal papilla cells induce follicle formation and hair growth by transdifferentiation of an adult epidermis. Development 1992, 115, 587–593. [Google Scholar] [CrossRef]
  141. Higgins, C.A.; Chen, J.C.; Cerise, J.E.; Jahoda, C.A.B.; Christiano, A.M. Microenvironmental reprogramming by three-dimensional culture enables dermal papilla cells to induce de novo human hair-follicle growth. Proc. Natl. Acad. Sci. USA 2013, 110, 19679–19688. [Google Scholar] [CrossRef]
  142. Kishimoto, J.; Burgeson, R.E.; Morgan, B.A. Wnt signaling maintains the hair-inducing activity of the dermal papilla. Genes Dev. 2000, 14, 1181–1185. [Google Scholar] [CrossRef] [PubMed]
  143. Lichti, U.; JAnders; Yuspa, S.H. Isolation and short-term culture of primary keratinocytes, hair follicle populations and dermal cells from newborn mice and keratinocytes from adult mice for in vitro analysis and for grafting to immunodeficient mice. Nat. Protoc. 2008, 3, 799–810. [Google Scholar] [CrossRef]
  144. Wu, J.-J.; Zhu, T.-Y.; Lu, Y.-G.; Liu, R.-Q.; Mai, Y.; Cheng, B.; Lu, Z.-F.; Zhong, B.-Y.; Tang, S.-Q. Hair follicle reformation induced by dermal papilla cells from human scalp skin. Arch. Dermatol. Res. 2006, 298, 183–190. [Google Scholar] [CrossRef] [PubMed]
  145. Zhou, L.; Wang, H.; Jing, J.; Yu, L.; Wu, X.; Lu, Z. Regulation of hair follicle development by exosomes derived from dermal papilla cells. Biochem. Biophys. Res. Commun. 2018, 500, 325–332. [Google Scholar] [CrossRef]
  146. Hu, S.; Li, Z.; Lutz, H.; Huang, K.; Su, T.; Cores, J.; Dinh, P.-U.C.; Cheng, K. Dermal exosomes containing miR-218-5p promote hair regeneration by regulating beta-catenin signaling. Sci. Adv. 2020, 6, eaba1685. [Google Scholar] [CrossRef]
  147. Hwang, J.; Zheng, M.; Le, T.N.H.; Kim, H.; Sung, J. Hair growth promoting effects of human dermal papilla cells in pig. Exp. Dermatol. 2023, 32, 1156–1158. [Google Scholar] [CrossRef] [PubMed]
  148. Li, J.; Zhao, B.; Dai, Y.; Zhang, X.; Chen, Y.; Wu, X. Exosomes Derived from Dermal Papilla Cells Mediate Hair Follicle Stem Cell Proliferation through the Wnt3a/beta-Catenin Signaling Pathway. Oxid. Med. Cell Longev. 2022, 2022, 9042345. [Google Scholar] [CrossRef]
  149. Chen, Y.; Huang, J.; Chen, R.; Yang, L.; Wang, J.; Liu, B.; Du, L.; Yi, Y.; Jia, J.; Xu, Y.; et al. Sustained release of dermal papilla-derived extracellular vesicles from injectable microgel promotes hair growth. Theranostics 2020, 10, 1454–1478. [Google Scholar] [CrossRef]
  150. Kwack, M.H.; Seo, C.H.; Gangadaran, P.; Ahn, B.; Kim, M.K.; Kim, J.C.; Sung, Y.K. Exosomes derived from human dermal papilla cells promote hair growth in cultured human hair follicles and augment the hair-inductive capacity of cultured dermal papilla spheres. Exp. Dermatol. 2019, 28, 854–857. [Google Scholar] [CrossRef]
  151. Geyfman, M.; Plikus, M.V.; Treffeisen, E.; Andersen, B.; Paus, R. Resting no more: Re-defining telogen, the maintenance stage of the hair growth cycle. Biol. Rev. Camb. Philos. Soc. 2015, 90, 1179–1196. [Google Scholar] [CrossRef]
  152. Matts, P.; Grosick, T.; Rust, R. Human body skin: Structure, function, and correlation with perceived personal care needs and treatment choice. J. Am. Acad. Dermatol. 2005, 52, P91. [Google Scholar]
  153. Kuty-Pachecka, M. Psychological and psychopathological factors in alopecia areata. Psychiatr. Pol. 2015, 49, 955–964. [Google Scholar] [CrossRef] [PubMed]
  154. Gupta, A.K.; Venkataraman, M.; Quinlan, E.M. Artificial hair implantation for hair restoration. J. Dermatol. Treat. 2022, 33, 1312–1318. [Google Scholar] [CrossRef]
  155. Nestor, M.S.; Ablon, G.; Gade, A.; Han, H.; Fischer, D.L. Treatment options for androgenetic alopecia: Efficacy, side effects, compliance, financial considerations, and ethics. J. Cosmet. Dermatol. 2021, 20, 3759–3781. [Google Scholar] [CrossRef]
  156. Rossi, A.; Cantisani, C.; Melis, L.; Iorio, A.; Scali, E.; Calvieri, S. Minoxidil use in dermatology, side effects and recent patents. Recent. Pat. Inflamm. Allergy Drug Discov. 2012, 6, 130–136. [Google Scholar] [CrossRef]
  157. Abreu, C.M.; Marques, A.P. Recreation of a hair follicle regenerative microenvironment: Successes and pitfalls. Bioeng. Transl. Med. 2022, 7, e10235. [Google Scholar] [CrossRef]
  158. Shen, Z.; Sun, L.; Liu, Z.; Li, M.; Cao, Y.; Han, L.; Wang, J.; Wu, X.; Sang, S. Rete ridges: Morphogenesis, function, regulation, and reconstruction. Acta Biomater. 2023, 155, 19–34. [Google Scholar] [CrossRef]
  159. Li, F.; Chen, W.; Huang, F.; Zhang, X.; Zhou, Y.; Liu, Z.; Wang, D. Role of different melanocyte populations in the reconstitution of pigmented hair follicles. Chin. J. Dermatol. 2023, 56, 118–124. [Google Scholar]
  160. Rusu, E.; Necula, L.G.; Neagu, A.I.; Alecu, M.; Stan, C.; Albulescu, R.; Tanase, C.P. Current status of stem cell therapy: Opportunities and limitations. Turk. J. Biol. 2016, 40, 955–967. [Google Scholar] [CrossRef]
  161. Haworth, R.; Sharpe, M. Accept or Reject: The Role of Immune Tolerance in the Development of Stem Cell Therapies and Possible Future Approaches. Toxicol. Pathol. 2021, 49, 1308–1316. [Google Scholar] [CrossRef]
  162. Wood, K.J.; Goto, R. Mechanisms of rejection: Current perspectives. Transplantation 2012, 93, 1–10. [Google Scholar] [CrossRef]
  163. Lee, S.; Choi, E.; Cha, M.-J.; Hwang, K.-C. Cell adhesion and long-term survival of transplanted mesenchymal stem cells: A prerequisite for cell therapy. Oxid. Med. Cell Longev. 2015, 2015, 632902. [Google Scholar] [CrossRef] [PubMed]
  164. Stevens, J.; Khetarpal, S. Platelet-rich plasma for androgenetic alopecia: A review of the literature and proposed treatment protocol. Int. J. Womens Dermatol. 2019, 5, 46–51. [Google Scholar] [CrossRef] [PubMed]
  165. Krefft-Trzciniecka, K.; Piętowska, Z.; Nowicka, D.; Szepietowski, J.C. Human Stem Cell Use in Androgenetic Alopecia: A Systematic Review. Cells 2023, 12, 951. [Google Scholar] [CrossRef] [PubMed]
  166. Ajit, A.; Nair, M.D.; Venugopal, B. Exploring the Potential of Mesenchymal Stem Cell–Derived Exosomes for the Treatment of Alopecia. Regen. Eng. Transl. Med. 2021, 7, 119–128. [Google Scholar] [CrossRef]
  167. Chien, W.-Y.; Huang, H.-M.; Kang, Y.-N.; Chen, K.-H.; Chen, C. Stem cell-derived conditioned medium for alopecia: A systematic review and meta-analysis. J. Plast. Reconstr. Aesthet. Surg. 2024, 88, 182–192. [Google Scholar] [CrossRef]
  168. Gao, L.; Chen, E.Q.; Zhong, H.; Xie, J.; Song, H.; Zhao, X.; Lin, J.; Liu, Q.; Wang, S.; Wu, W.; et al. Large-scale isolation of functional dermal papilla cells using novel surface marker LEPTIN Receptor. Cytom. A 2022, 101, 675–681. [Google Scholar] [CrossRef]
  169. Topouzi, H.; Logan, N.J.; Williams, G.; Higgins, C.A. Methods for the isolation and 3D culture of dermal papilla cells from human hair follicles. Exp. Dermatol. 2017, 26, 491–496. [Google Scholar] [CrossRef]
  170. Nilforoushzadeh, M.; Jameh, E.R.; Jaffary, F.; Abolhasani, E.; Keshtmand, G.; Zarkob, H.; Mohammadi, P.; Aghdami, N. Hair follicle generation by injections of adult human follicular epithelial and dermal papilla cells into nude mice. Cell J. 2017, 19, 259. [Google Scholar]
  171. Ito, Y.; Hamazaki, T.S.; Ohnuma, K.; Tamaki, K.; Asashima, M.; Okochi, H. Isolation of Murine Hair-Inducing Cells Using the Cell Surface Marker Prominin-1/CD133. J. Investig. Dermatol. 2007, 127, 1052–1060. [Google Scholar] [CrossRef]
  172. Richardson, G.D.; Robson, C.N.; Lang, S.H.; Neal, D.E.; Maitland, N.J.; Collins, A.T. CD133, a novel marker for human prostatic epithelial stem cells. J. Cell Sci. 2004, 117 Pt 16, 3539–3545. [Google Scholar] [CrossRef] [PubMed]
  173. Sennett, R.; Wang, Z.; Rezza, A.; Grisanti, L.; Roitershtein, N.; Sicchio, C.; Mok, K.W.; Heitman, N.J.; Clavel, C.; Ma’ayan, A.; et al. An Integrated Transcriptome Atlas of Embryonic Hair Follicle Progenitors, Their Niche, and the Developing Skin. Dev. Cell 2015, 34, 577–591. [Google Scholar] [CrossRef] [PubMed]
  174. Kishimoto, J.; Ehama, R.; Wu, L.; Jiang, S.; Jiang, N.; Burgeson, R.E. Selective activation of the versican promoter by epithelial- mesenchymal interactions during hair follicle development. Proc. Natl. Acad. Sci. USA 1999, 96, 7336–7341. [Google Scholar] [CrossRef] [PubMed]
  175. Rezza, A.; Wang, Z.; Sennett, R.; Qiao, W.; Wang, D.; Heitman, N.; Mok, K.W.; Clavel, C.; Yi, R.; Zandstra, P.; et al. Signaling Networks among Stem Cell Precursors, Transit-Amplifying Progenitors, and their Niche in Developing Hair Follicles. Cell Rep. 2016, 14, 3001–3018. [Google Scholar] [CrossRef] [PubMed]
  176. Osada, A.; Iwabuchi, T.; Kishimoto, J.; Hamazaki, T.S.; Okochi, H. Long-term culture of mouse vibrissal dermal papilla cells and de novo hair follicle induction. Tissue Eng. 2007, 13, 975–982. [Google Scholar] [CrossRef]
  177. Reynolds, A.J.; Jahoda, C.A. Hair matrix germinative epidermal cells confer follicle-inducing capabilities on dermal sheath and high passage papilla cells. Development 1996, 122, 3085–3094. [Google Scholar] [CrossRef]
  178. Bratka-Robia, C.B.; Mitteregger, G.; Aichinger, A.; Egerbacher, M.; Helmreich, M.; Bamberg, E. Primary cell culture and morphological characterization of canine dermal papilla cells and dermal fibroblasts. Vet. Dermatol. 2002, 13, 1–6. [Google Scholar] [CrossRef]
  179. Abreu, C.M.; Cerqueira, M.T.; Pirraco, R.P.; Gasperini, L.; Reis, R.L.; Marques, A.P. Rescuing key native traits in cultured dermal papilla cells for human hair regeneration. J. Adv. Res. 2021, 30, 103–112. [Google Scholar] [CrossRef]
  180. Rheinwald, J.G.; Green, H. Serial cultivation of strains of human epidermal keratinocytes: The formation of keratinizing colonies from single cells. Cell 1975, 6, 331–343. [Google Scholar] [CrossRef]
  181. Bak, S.S.; Kwack, M.H.; Shin, H.S.; Kim, J.C.; Kim, M.K.; Sung, Y.K. Restoration of hair-inductive activity of cultured human follicular keratinocytes by co-culturing with dermal papilla cells. Biochem. Biophys. Res. Commun. 2018, 505, 360–364. [Google Scholar] [CrossRef]
  182. Inamatsu, M.; Matsuzaki, T.; Iwanari, H.; Yoshizato, K. Establishment of rat dermal papilla cell lines that sustain the potency to induce hair follicles from afollicular skin. J. Investig. Dermatol. 1998, 111, 767–775. [Google Scholar] [CrossRef] [PubMed]
  183. Shin, S.-H.; Park, S.-Y.; Kim, M.-K.; Kim, J.-C.; Sung, Y.-K. Establishment and characterization of an immortalized human dermal papilla cell line. BMB Rep. 2011, 44, 512–516. [Google Scholar] [CrossRef] [PubMed]
  184. Kang, B.M.; Kwack, M.H.; Kim, M.K.; Kim, J.C.; Sung, Y.K. Sphere formation increases the ability of cultured human dermal papilla cells to induce hair follicles from mouse epidermal cells in a reconstitution assay. J. Investig. Dermatol. 2012, 132, 237–239. [Google Scholar] [CrossRef]
  185. Kwack, M.H.; Yang, J.M.; Won, G.H.; Kim, M.K.; Kim, J.C.; Sung, Y.K. Establishment and characterization of five immortalized human scalp dermal papilla cell lines. Biochem. Biophys. Res. Commun. 2018, 496, 346–351. [Google Scholar] [CrossRef]
  186. Kwack, M.H.; Ben Hamida, O.; Kim, M.K.; Kim, M.K.; Sung, Y.K. Establishment and characterization of matched immortalized human frontal and occipital scalp dermal papilla cell lines from androgenetic alopecia. Sci. Rep. 2023, 13, 21421. [Google Scholar] [CrossRef] [PubMed]
  187. Sugiyama-Nakagiri, Y.; Fujimura, T.; Moriwaki, S. Induction of Skin-Derived Precursor Cells from Human Induced Pluripotent Stem Cells. PLoS ONE 2016, 11, e0168451. [Google Scholar] [CrossRef]
  188. Riabinin, A.; Kalabusheva, E.; Khrustaleva, A.; Akulinin, M.; Tyakht, A.; Osidak, E.; Chermnykh, E.; Vasiliev, A.; Vorotelyak, E. Trajectory of hiPSCs derived neural progenitor cells differentiation into dermal papilla-like cells and their characteristics. Sci. Rep. 2023, 13, 14213. [Google Scholar] [CrossRef]
  189. Veraitch, O.; Mabuchi, Y.; Matsuzaki, Y.; Sasaki, T.; Okuno, H.; Tsukashima, A.; Amagai, M.; Okano, H.; Ohyama, M. Induction of hair follicle dermal papilla cell properties in human induced pluripotent stem cell-derived multipotent LNGFR(+)THY-1(+) mesenchymal cells. Sci. Rep. 2017, 7, 42777. [Google Scholar] [CrossRef] [PubMed]
  190. Ma, Y.; Lin, Y.; Huang, W.; Wang, X. Direct Reprograming of Mouse Fibroblasts into Dermal Papilla Cells via Small Molecules. Int. J. Mol. Sci. 2022, 23, 4213. [Google Scholar] [CrossRef]
  191. Zhao, Q.; Li, N.; Zhang, H.; Lei, X.; Cao, Y.; Xia, G.; Duan, E.; Liu, S. Chemically induced transformation of human dermal fibroblasts to hair-inducing dermal papilla-like cells. Cell Prolif. 2019, 52, e12652. [Google Scholar] [CrossRef]
Figure 1. Key signaling molecules/pathways involved in the regulation of hair follicle morphogenesis and the hair cycle.
Figure 1. Key signaling molecules/pathways involved in the regulation of hair follicle morphogenesis and the hair cycle.
Biology 13 00842 g001
Figure 2. Technical and ethical challenges of two potential approaches of DPC-based cell therapy for treating alopecia. HF, hair follicle; iMC, induced mesenchymal cells. Created with BioRender.com.
Figure 2. Technical and ethical challenges of two potential approaches of DPC-based cell therapy for treating alopecia. HF, hair follicle; iMC, induced mesenchymal cells. Created with BioRender.com.
Biology 13 00842 g002
Figure 3. New technological advancements in basic and translational research of DPCs. HF, hair follicle; HS, hair shaft; DF, dermal fibroblasts. Created with BioRender.com (accessed on 6 August 2024).
Figure 3. New technological advancements in basic and translational research of DPCs. HF, hair follicle; HS, hair shaft; DF, dermal fibroblasts. Created with BioRender.com (accessed on 6 August 2024).
Biology 13 00842 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhang, H.-L.; Qiu, X.-X.; Liao, X.-H. Dermal Papilla Cells: From Basic Research to Translational Applications. Biology 2024, 13, 842. https://doi.org/10.3390/biology13100842

AMA Style

Zhang H-L, Qiu X-X, Liao X-H. Dermal Papilla Cells: From Basic Research to Translational Applications. Biology. 2024; 13(10):842. https://doi.org/10.3390/biology13100842

Chicago/Turabian Style

Zhang, He-Li, Xi-Xi Qiu, and Xin-Hua Liao. 2024. "Dermal Papilla Cells: From Basic Research to Translational Applications" Biology 13, no. 10: 842. https://doi.org/10.3390/biology13100842

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop