Your privacy, your choice

We use essential cookies to make sure the site can function. We also use optional cookies for advertising, personalisation of content, usage analysis, and social media.

By accepting optional cookies, you consent to the processing of your personal data - including transfers to third parties. Some third parties are outside of the European Economic Area, with varying standards of data protection.

See our privacy policy for more information on the use of your personal data.

for further information and to change your choices.

Skip to main content

Functional regeneration strategies of hair follicles: advances and challenges

Abstract

Hair follicles are essential appendages of human skin that function in protection, sensation, thermoregulation and social interactions. The multicellular components, particularly the dermal papilla, matrix and bulge housing stem cells, enable cyclic hair growth postnatally. However, miniaturization and loss of hair follicles can occur in the context of ageing, trauma and various alopecia-related diseases. Conventional treatments involve the redistribution of existing follicles, which may not be viable in patients lacking follicular resources. Recent progress in the comprehension of morphogenesis and the development of biomaterials has significantly advanced follicle reconstruction, incorporating organ germ assembling, stem cell induction and bioprinting techniques. Despite these advancements, fully restoring hair follicles remains challenging due to the complexities of replicating embryonic signals and sustaining growth cycles. Identifying suitable cell sources for clinical applications also presents a hurdle. Here, we retrospect the progress made in the field of hair follicle regeneration, aiming to offer an exhaustive analysis on the benefits and limitations of these methods, and to foster the development of innovative solutions.

Introduction

Hair follicles (HFs) are self-sustaining mini-organs with diverse stem cell populations and intricate niches. Driven by the periodic activation of hair follicle stem cells (HFSCs), HFs undergo cyclical growth throughout the lifetime. Despite their ability to regenerate, HFs are susceptible to the effects of ageing, stress and environmental stimuli, resulting in reduced hair thickness and density. Currently, the main approach used to combat hair loss relies on the autograft of HF units; however, this method is inadequate for treating patients with insufficient HF reserves. Studies of HF regeneration are increasingly significant due to the prevalence of alopecia and the recognized contribution of HFs to wound healing [1].

Hair follicle morphogenesis

Understanding the mechanism that governs HF morphogenesis is essential for the reconstruction of functional HFs. The development of HF during embryogenesis shares the same driving force with the kidney, lung and tooth, known as epithelial-mesenchymal interactions (EMIs) [2, 3]. Owing to ethical constraints in human studies, knowledge of HF organogenesis is based mainly on mice. Around embryonic day 13, the unspecified epidermis receives “first dermal signals” from the mesenchyme and forms a thickened epithelial layer called the placode, marking the early stage of HF morphogenesis [4, 5]. Placode formation involves the activation of ectodysplasin A/ectodysplasin A receptor (EDA/EDAR) pathways within the epithelium, followed by Wnt and bone morphogenetic protein (BMP) signaling [6, 7]. EDA/EDAR and Wnt pathways collaboratively facilitate placode fate, while BMP represses placode development in adjacent areas [8]. The underlying mesenchymal cells subsequently receive “first epithelial signals” from the placode and aggregate to form the dermal condensate, which is controlled by fibroblast growth factor (FGF) signaling [9,10,11]. The dermal condensate communicates with the placode to boost the downgrowth of the hair germ, and further evolves into the dermal papilla (DP) enveloped by matrix cells via Wnt/SHH signaling [11, 12]. This signifies the emergence of the HF rudiment, where the DP and matrix collaborate to generate the hair shaft [13]. As HFs mature, the bulge emerges as a vital epithelial pool alongside the matrix, harbouring CD34/CD49f+ HFSCs for self-renewal [14, 15].

Hair cycle dynamics

A HF is a sustainable system in which cells are replenished through tightly regulated specification, differentiation and proliferation. Hair growth is fuelled by HFSCs in the bulge area under the regulation of DP, the signaling center [16,17,18]. Dependent on the dynamic epithelial-mesenchymal crosstalk, HFs exhibit cyclic growth (anagen), regression (catagen) and rest (telogen) after birth [16,17,18] (Fig. 1, adapted from [2] with permission). During telogen, DPs release inhibitory cues, especially BMPs, to maintain the quiescent state of HFSCs [19,20,21]. When transitioning to anagen, FGFs and BMP inhibitors, such as Noggin and transforming growth factor-β2 (TGFβ2), accumulate in the DP, and simultaneously, Wnt levels rise in the hair germ. These signals trigger the activation of HFSCs and the formation of new shafts [22, 23]. As catagen ensues, TGFβ1 and Wnt antagonists from the DP induce the apoptosis of basal epithelial cells [24, 25]. The dermal sheath contracts to retract DP upwards along the epithelial strand and relocate DP beneath the bulge, thereby posing appropriate spatial arrangements for cyclic switching [26, 27].

We can see that, EMIs play a pivotal role in HF morphogenesis and regeneration, although the identified signaling signatures are fragmentary. Recent breakthroughs in single-cell transcriptomics have propelled the exploration of a comprehensive regulatory network, but with so many factors involved, further research is needed to minimize redundancy. Benefitting from improved comprehension of underlying mechanisms and advanced bioengineering technologies, HF regeneration strategies have undergone a remarkable transformation, incorporating germ assembling, pluripotent stem cell (PSC) induction and bioprinting [28]. This review thoroughly traces the timeline of HF regeneration, providing novel insights into the optimization of HF bioengineering.

Fig. 1
figure 1

Signaling molecules involved in the hair cycle. (a) In telogen, bone morphogenetic proteins (BMPs), such as BMP4 from endothelial cells, BMP2 from adipocytes, and BMP4 from fibroblasts, maintain the quiescence of hair follicle stem cells (HFSCs). Oncostatin M (OSM) released by TREM2+ macrophages, and fibroblast growth factor 18 (FGF18) from the dermal papilla (DP) and bulge, also contribute to the dormancy of HFSCs. (b) During the transition from telogen to anagen, factors like FGF7, FGF10, Norrin, and BMP inhibitors (Noggin and transforming growth factor-β2 (TGFβ2)) promote the activation of HFSCs. Other contributors to this shift include endothelin 1 (EDN1) from endothelial cells, platelet-derived growth factor subunit A (PDGFA) from adipocyte precursor cells, and Wnts (Wnt7b and Wnt10a) from apoptotic CD11b+ macrophages. Additionally, cutaneous transient receptor potential cation channel subfamily V member 1 (TRPV1) innervations accelerate hair growth via Spp1 from CD9+CD26+ fibroblasts. (c) In anagen, sonic hedgehog (SHH) from transit-amplifying cells (TACs) further enhances the proliferation of HFSCs and the differentiation of adipocyte precursor cells, leading to the extension of hair follicles. (d) During the anagen-catagen transition, FGF5 from the matrix and outer root sheath (ORS), and Wnt antagonists (dickkopf WNT signaling pathway inhibitor 2 (DDK2) and Notum) from the DP induce the apoptosis of follicular cells. (e) In catagen, epithelial cells release EDN1 to stimulate calcium influx and dermal sheath retraction to relocate the DP. (f) In the subsequent telogen, the DP and the new bulge are positioned adjacent to the old bulge. Fig. 1 was adapted from [2] (Zhang B, Chen T. Local and systemic mechanisms that control the hair follicle stem cell niche. Nat Rev Mol Cell Biol. 2024;25(2):87-100. https://www.nature.com/nrm/), which is not part of the governing OA license, with reproduction permission.

Follicle germ assembling

The pioneering work in HF regeneration dates back to 1970, established on the restoration of EMIs. Yuspa and colleagues grafted cultures derived from embryonic mouse skin onto lesions lacking epidermis and dermis and observed the formation of hair-bearing skin [29]. With the progress in cell isolation techniques, Worst and colleagues demonstrated that neogenesis of HFs occurred only when epidermal cells were transplanted alongside dermal cells [30]. Since then, HF restoration has become practicable through the codelivery of dissociated epithelial and mesenchymal progenitors via patch or chamber assays [31, 32]. While these engineered HFs largely recapitulated the natural anatomical features of HFs, their generative efficiency was limited, probably owing to insufficient EMIs.

The introduction of organ-germ culture revolutionized HF regeneration as an innovative delivery approach [33]. In this method, dissociated epithelial and mesenchymal cells self-organize into follicular primordia prior to transplantation (Fig. 2). Tsuji Lab reported that self-assembled hair follicle germs (HFGs) successfully produced pigmented HFs in vivo, with natural compartments and proper integration into the surrounding skin [34]. These HFs contained CD34/CD49f+ bulges and Sox2+ DPs, supporting cyclic growth (repeated at least 3 times). Nevertheless, protruding shafts occurred in < 1% of HFGs under Tsuji Lab’s protocol [35], necessitating improvements in trichogenicity. By adding 2% Matrigel before aggregation, Fukuda Team significantly enhanced the sprouting efficiency to nearly 100% [35]. This upswing was attributed to the morphological change in assemblages. Unlike the previous dumbbell-like conformation [34, 36], 2% Matrigel resulted in aggregates with a core‒shell structure, augmenting contacts between cells and reinforcing EMIs [35]. To enable clinical scalability, the high-throughput production of HFGs has been facilitated via culture on biomaterial substrates, such as polydimethylsiloxane and poly(ethylene-co-vinyl alcohol) [36, 37].

Fig. 2
figure 2

Methods for assembling hair follicle germs. (a) In initial attempts to regenerate HFs, mixed epithelial cells and mesenchymal cells from embryonic or neonatal mice were directly transplanted via patch or chamber assays. In the method of HFG formation, self-organized primordia are transplanted, with Matrigel supplement shown to enhance trichogenicity. (b) Strategies incorporating IGF2-VEGF, Wnt3a-Wnt10b and MMP14 or IFNγ/VEGF supplements effectively restore HF regeneration in adult cells. (c) Vibrissa DPCs and epithelial cells from adult mice serve as an alternative source for generating HFGs.

Appropriate cell sourcing

Studies on HFGs have focused on using progenitor cells extracted from embryonic or neonatal murine skin, with a few studies utilizing dermal papilla cells (DPCs) from adult HFs [34, 37] (Table 1). However, the use of embryonic or neonatal cells is not feasible clinically due to ethical concerns and immune rejection. Additionally, patient-derived DPCs tend to lose their intrinsic properties when cultured in vitro [38,39,40,41]. These limitations underscore the need to explore suitable cell sources, with reprogramming emerging as a potential strategy. Precursors induced through small molecule treatment and genetic engineering hold promise as viable sources [42,43,44,45]. It is noteworthy that, despite exhibiting phenotypes resembling natural progenitors, pluripotent stem cell (PSC)-derived precursors may fail to assemble into HFs due to inaccurate cell specification [46]. Therefore, the refinement of induction protocols from PSCs is crucial, offering the opportunity to generate abundant resources while ensuring safety.

Table 1 Cells and biomaterials used in follicle germ assembling

Rejuvenating adult cells

Environmental reprogramming presents a direct solution to address reproducibility challenges in mature cells. Lei and colleagues identified differential expression profiles between neonatal and adult cells and devised a corresponding system to restore morphogenetic competence [47]. Sequential supplements with IGF2-VEGF/Wnt3a-Wnt10b/MMP14 and continuous administration of PKC inhibitors reinitiated self-organization in adult mouse cells, prompting HF generation from 0 to approximately 40% of newborn-derived germs [47]. An alternative strategy involving PKC/PKR inhibition and IFNγ/VEGF supplement also enabled HF reconstruction from adult cells [48], indicating the potential to revitalize mature cells through ambient cues.

Preserving DPCs for inducibility

During the hair cycle, DPCs act as the primary trichogenic dermal cells [49]. While implanting dissected DPs from rodents and humans successfully stimulated HF growth [50], attempts to replicate this process using human DPCs or late-passage rodent DPCs failed owing to the diminished inducibility during culture [39, 50]. Studies have linked Wnt, BMP and FGF signaling to the hair-inducing potential of DPCs [51,52,53]. Accordingly, a specialized medium containing agonists of these pathways was formulated (Table 2). This medium was proven to partially recover the expression of DP signature genes, and restore the characteristic function of DPs to communicate with epithelial components [53].

Table 2 Approaches to inducible DPCs

Considering the tendency of DPCs to aggregate, three-dimensional culture has been proposed to revive the trichogenic phenotype and foster subsequent folliculogenesis [50, 54, 55]. Christiano Team reported that approximately 22% of transcripts perturbed by planar culture were regained in spherical culture [50]. Lef-1 overexpression further restored hair lineage signatures, leading to a 70% rate of HF formation in vitro [56]. Furthermore, Wu Team introduced immune regulation into the inducibility-preserving strategies of DPCs [57]. Calcium molybdate nanoparticles loaded with DPCs and macrophages not only supported the survival of DPCs, but also facilitated hair regrowth in vivo by creating an anti-inflammatory microenvironment [57].

Additionally, transforming dermal fibroblasts into DP-like cells represents an alternative approach (Table 2). Studies have shown that a cocktail composed of PDGF, FGFs and BIO [58], the small molecule TTNPB [59], and microencapsulated alginate-poly-L-lysine-alginate [60] can stimulate trichogenicity in dermal fibroblasts, as evidenced in patch assays.

The events subsequent to the transplantation of DPCs have not been fully elucidated, especially with respect to the origin of follicular epithelia — whether they stem from preexisting epidermal progenitors or if there is a redefinition of cell fate. Delving into the interactions of transplanted DPCs with native cells is essential for a comprehensive understanding of HF regeneration and degeneration. Enhanced knowledge of these processes will, in turn, contribute to the establishment of HFs that persist in the long term.

Skin organoid induction from pluripotent stem cells

Achievements in stem cell research have facilitated the generation of skin organoids from PSCs (Table 3). PSCs, including induced pluripotent stem cells (iPSCs) and embryonic stem cells (ESCs), have the potential to differentiate into distinct cells and tissue patterns under particular cues. The initial construction of skin equivalents involves two main steps: first, guiding PSCs to differentiate into keratinocytes and fibroblasts, the predominant cell types in the skin, and second, stacking these cells into a bilayer structure [61, 62]. These structures serve as basic frameworks lacking HFs or sebaceous glands. As our understanding on morphogenesis deepens, there is optimism about generating authentic skin substitutes through the recapitulation of key signals.

Table 3 Strategies for inducing skin organoids from PSCs

Murine skin induction from PSCs

In 2016, Tsuji Team successfully engineered an integumentary system containing HFs and sebaceous glands from murine iPSCs [63]. Embryoid bodies derived from iPSCs were transplanted as clusters, giving rise to integrated skin, HFs with properly arranged niches, and subcutaneous adipose tissue. Wnt10b supplement was proven to enhance the germination and maturation of HFs via EMIs [63]. This study represented a pioneering effort in regenerating skin with HFs using PSCs, albeit heavily dependent on the variables inherent in living organisms.

In 2018, Koehler Lab detailed a stepwise formula to form murine folliculogenic skin from PSCs [64], based on an established protocol for inducing cranial surface ectoderm [65, 66]. In this method, the administration of a TGFβ inhibitor (SB431542) and BMP4 specified surface ectoderm at the outermost region, and the subsequent treatment with FGF and a BMP inhibitor (LDN-193189) committed the pre-placodal fate [64]. This skin organoid surprisingly mimicked the differentiation aligning to embryonic development. Derived HFs shared critical features with natural HFs in embryonic stages 7–8, containing αSMA+ dermal sheath, KRT5+ p63+ outer root sheath, GATA3+ inner root sheath, Ki67+ p63+ matrix, and AE13+ shaft. However, these HFs exhibited limitations in long-term culture, as the matrix deteriorated around day 32 and the shaft failed to shed physiologically, impeding the transition into the next cycle [64].

Human skin induction from PSCs

In 2020, Koehler Lab extended their strategy to human skin regeneration by refining the timing of LDN/FGF administration to day 3 of differentiation (Fig. 3) [67]. This adjustment consistently led to the development of epithelial cysts enveloped by cranial neural crest cells (CNCCs). Hair germs emerged on ~ day 70, aligning with the organogenesis process during gestation. After more than 100 days in culture, these organoids were comparable to 18-week foetal skin, featuring a stratified epidermis, a fat-rich dermis and pigmented HFs. On day 140, the cystic organoids were implanted into nude mice, and planar skin, an extended vasculature and pilosebaceous units were successfully established. These results signify the nearly complete reconstruction of human skin through self-assembly. Moreover, this methodology has shown considerable reproducibility in subsequent studies [68,69,70] and has come to the forefront in disease modeling [68, 69].

Fig. 3
figure 3

Strategies for inducing skin organoids from PSCs. (a) In the classical protocol, the use of a TGFβ inhibitor (SB431542) and BMP4 facilitates the differentiation of surface ectoderm, followed by the administration of FGF and a BMP inhibitor (LDN-193189) to prompt the formation of CNCCs. This process results in the development of enclosed cysts with an inside-out morphology, which matures into planar skin following transplantation. (b) A modified approach incorporates a Wnt activator (CHIR99021) and ALI culture, transforming the enclosed aggregates into larger, open skin organoids.

Transforming enclosed organoids into an open skin model

An apparent defect of these organoids is their inside-out morphology, in which epidermal cells are encircled by dermal components [67]. The aberrant structure imposes a maximum culture duration of 150 days; otherwise, squamous cells would accumulate in the core. To rectify the inverted conformation, Jung and colleagues introduced Wnt activation and air-liquid interface (ALI) culture into the SB/BMP-LDN/FGF protocol, resulting in enlarged open skin models (Fig. 3) [71]. Nevertheless, the lifespan of these organoids remained approximately 150 days. It is advisable to investigate methods for artificial ripening or prolonged culture so that fully developed structures can be harvested for restoration. On the other hand, determining the opportune stage for grafting may hold greater significance than adjusting the culture formula. While it took 4–5 months to coax PSCs into skin organoids with requisite appendages, single-cell RNA sequencing on day 48 revealed diverse cell populations resembling those in embryonic skin [67]. It is speculated that in situ incubation may operate more efficiently than various cocktails, as it covers biochemical messengers and subtle mechanical forces.

Scalp skin induction to be investigated

Notably, since CNCCs are responsible for the development of facial dermis, the organoids described above recapitulate only facial skin. Scalp HFs may be attained through the inclusion of mesodermal cells, or not. This represents an obstacle to holistic skin reconstruction from PSCs.

Skin substitute bioprinting

3D printing, evolving alongside fabrication technologies, brings a new dimension to HF engineering. Initially, 3D printing was utilized to produce noncellular scaffolds resembling HFs, after which the cells were manually seeded [56, 72]. This process was later simplified with the introduction of bioprinting (Table 4; Fig. 4). Bioprinting is a computer-aided technology allowing the precise deposition of bioinks — biomaterials loaded with cells [73]. Multiple factors, including cell types, biomaterial properties, fabrication modalities, and post-printing maturation, need to be considered when printing target tissues [74]. While primary cells and commercial cell lines are commonly used, the application of stem cells is restricted owing to their sensitivity to mechanical shear and viscosity [75, 76]. Biomaterials, as carriers and supporters of living cells, are expected to exhibit flexibility, biocompatibility, and controllable biodegradability [77, 78]. Polymers widely used in bioprinting include collagen, hyaluronic acid, alginate, agarose, chitosan and fibrin, depending on the desired pattern [79, 80]. An appropriate platform is then employed to accurately position bioinks according to the predetermined geometry. Finally, post-printing procedures, such as ALI and bioreactor processing, help to mature and functionalize these constructs [74, 81].

Table 4 Cells and biomaterials used in bioprinting
Fig. 4
figure 4

Application of bioprinting in hair-bearing skin regeneration. (a) Layer-by-layer deposition of fibroblasts, DPC spheres, and epithelial cells generates skin substitutes with trichogenicity. (b) Robot-assisted bioprinting utilizing precursors from neonatal mice enables on-site HF-inclusive skin repair.

Layer-by-layer bioprinting

Bioprinting involves four main modalities: droplet-based bioprinting (DBB), extrusion-based bioprinting (EBB), laser-assisted bioprinting (LAB), and stereolithography-based bioprinting (SBB) [82, 83]. In the realm of skin constructs, EBB is the preferred method due to its versatility [75, 77]. Miao Lab managed to engineer murine skin capable of HF regeneration through EBB [84]. During printing, cells encapsulated in gelatin-alginate hydrogels were deposited in stratified layers, with fibroblasts and human umbilical vein endothelial cells (HUVECs) forming the dermis, DPCs dot-printed into the middle stratum, and epithelial cells comprising the epidermis [84]. These scaffolds enabled the self-aggregation of DPCs and the initiation of EMIs in vitro. After 7 days of culture, DPCs and epithelial cells generated HF-like elongate structures, leading to substantial hair growth in an appropriate orientation following transplantation [84]. Notably, the proper inclusion of DPCs is decisive in HF formation, as direct deposition without pro-aggravating measures would result in the loss of trichogenic properties [85].

In a recent study by Karande Team, HFs were successfully incorporated into human skin constructs via EBB [86]. Spheroids consisting of DPCs and HUVECs were bioprinted within a pre-gelled dermal layer containing fibroblasts. Subsequently, type IV collagen was deposited to establish the dermal-epidermal junction, and keratinocytes were paved to generate the epidermis. Within 48 h of spheroid printing, HF-like columns extended up to the epidermal layer [86]. The resulting HFs resembled natural tissues, with a CK14+ outer root sheath and a CK10+ inner root sheath surrounding a dense core akin to the DP. Markers associated with DP inducibility, such as versican and alkaline phosphatase, were also detected. However, the model was not transplanted to evaluate HF sprouting or cycling capabilities in vivo [86].

In situ bioprinting

The studies outlined above are grounded on the transplantation of bioprinted architectures. Remarkably, a bioprinting robot has realized in situ regeneration of HF-equipped skin, utilizing skin-derived precursors from neonatal mice [87, 88]. This innovative approach allows one-step printing onto skin defects, in contrast to conventional layer-by-layer deposition and subsequent transplantation. Matrigel [87] and gelatin methacrylate [88] were identified as competent substrates supporting the survival and differentiation of these sensitive progenitors. In addition, intraoperative bioprinting of human adipose-derived extracellular matrix and stem cells led to the formation of HF-like extensions, suggesting the involvement of adipocytes in matrix shaping and downgrowth formation [89]. This discovery promisingly sparks interest in incorporating adipocytes for HF regeneration.

Challenges in tailoring bioinks

Throughout these studies, we can see that reestablishing HFs by bioprinting shares the same tenet with organ germs, that is the inducibility restoration of DPCs and the fate commitment of progenitors. While reprogramming strategies have been investigated to recover the trichogenic potential of DPCs, challenges persist in accessing DPCs and other primary cells owing to donor variations and massive clinical demand. Millions of cells are required to achieve a physiological density (greater than 107 cells/ml of bioink), making cell isolation and expansion an individualized and labor-intensive task [90]. Such inefficiency has prompted the exploration of alternative cell sources. Heartened by advancements in xenotransplantation, it is postulated that xenogeneic cells devoid of immunogenicity could serve as universal ingredients in bioinks [91,92,93]. The incorporation of genetically modified cells, for instance, porcine cells, holds promise for streamlining bioprinting processes and realizing immediate repair.

Another limitation of bioprinting is the restricted cell density permissible in bioinks to retain printability [94]. It is observed that bioprinted structures frequently undergo shrinkage while maintaining a relatively consistent conformation [75, 95]. The unexpected compaction necessitates adjustments to predefined parameters, but it also enables cell enrichment. Leveraging this idea, Fukuda Lab bioprinted paired collagen droplets containing mesenchymal and epithelial cells in close proximity, which allowed a spontaneous contraction through cell traction forces [96, 97]. This approach led to HFG-like constructs with cells enriched over tenfold and enhanced trichogenic potential in vivo. Consequently, the inherent shrinkage offers a promising solution to condense cells for future bioprinting projects.

Discussion

Organ loss and dysfunction caused by disease and trauma drives the development of regenerative therapies [98,99,100]. With the rising prevalence of hair loss, HF reestablishment has become one of the focal points in regenerative medicine [101, 102]. A genuine follicle is characterized by coordinative niches, including the distal papilla, bulged stem cell reservoir, and active transit-amplifying cells [18, 103]. Engineered HFs are expected to cycle stably, sprouting new hair shafts while extruding old ones, and stay synchronized with the native pattern. Current methods for HF regeneration are comprised of organ germs, PSC-derived organoids, and bioprinting, in which bioprinting is a facilitative instrument rather than a standalone approach [28, 104, 105]. Utilizing these strategies, the reconstruction of essential compartments and in vivo hair growth have been achieved, yet the cyclability throughout the lifespan remains a hurdle to fully functional HF substitutes [34, 64]. At the mechanism level, research efforts are focused primarily on the preparation of HF precursors or analogues, with a limited understanding on the interplay between implants and their surroundings. It is imperative to decipher the signaling crosstalk and cellular dynamics following transplantation. Clarifying whether the components of neogenetic HFs come solely from implants or are partially derived from existing or respecified progenitors in recipients will help find a way to sustainable regeneration.

Apart from unmet cycling goals, there are several intrinsic and technical challenges in HF reconstruction (Table 5). The organ-germ method, while appreciated as an early controllable approach, faces limitations due to its cut-and-paste nature. The progenitors extracted from embryonic or neonatal murine skin undergo transfer to generate hair in another individual [34,35,36], which is not suitable in clinical settings. Rather than utilizing allogeneic progenitors, revitalizing autologous cells holds potential for regenerating HFs in human. The restoration of trichogenicity in mature cells has been achieved through environmental signals like VEGF and PKC inhibition, although the durability of resulting HFs remains unexplored [47, 48]. Moreover, iPSCs are considered as a promising source for generating HF-equipped skin. iPSCs can be derived from various cell types, such as dermal fibroblasts, peripheral blood mononuclear cells, hepatocytes, pancreatic beta cells, and neural stem cells, offering benefits in terms of accessibility, expandability, and ethical compatibility [106]. Despite these advantages, several issues need to be addressed prior to their clinical application. Human iPSCs are “primed” and heterogeneous in differentiation potential, tumorigenicity and genome instability [107, 108]. These variations necessitate rigorous characterization and thorough selection during iPSC expansion and maturation, making customizing organoids extremely costly and time-consuming [109]. In order to maximize the utility of iPSCs, a practical strategy is to construct universal iPSC lines and to establish standardized differentiation protocols [110]. These iPSC lines, whether allogenic or xenogeneic, can be modified using CRISPR or other genome-editing tools to prevent immune rejection [111,112,113]. By producing standardized skin organoids, safety concerns can be minimized, and a timely clinical supply can be ensured. In addition, the accurate specification of iPSCs into epithelial and mesenchymal progenitors opens up the possibility of HFG transplantation as an alternative and convenient approach. All these prospects hinge on precise reprogramming and genetic editing techniques, requiring the collaborative efforts of researchers.

Table 5 Summary of advantages and drawbacks of present methods

Bioprinting technology emerges as an avenue to fabricating organ-mimicking structures with high resolution [114]. Current studies concerning HF bioprinting are guided by the principle of EMI restoration. Both DPC-based and progenitor-based bioinks have succeeded in integrating HFs into bioengineered skin [84, 86,87,88]. Recent employment of robotic systems has ushered in new opportunities for expeditious treatment using universal bioinks [87, 88], which comes down to optimizing cell sourcing and biomaterial selection. The biomaterials commonly used for printing iPSC-derived cells include Matrigel, alginate, agarose, gelatin methacryloyl, and nano-fibrillated cellulose [115]. Matrigel, derived from EHS mouse sarcoma cells, simulates the natural extracellular environment and is frequently utilized as a substrate for iPSC proliferation and differentiation [116]. Nevertheless, its heterologous nature and undefined chemical composition have raised safety concerns and experimental variations. Biomaterials such as hyaluronic acid and vitronectin also demonstrate the capacity to support iPSC culture, whereas further modifications are needed to align them with the bioactivity and formability requirements in bioprinting. Additionally, engineering improvements are necessary to alleviate thermal and mechanical stresses during printing, and thus to guarantee high viability and functionality of constructs. Apart from these technical constraints, a critical issue that tends to be overlooked in bioprinting and HFG studies is the cellular dynamics and niche reconstruction following transplantation. Although the comprehension of these cellular and molecular events is currently inadequate, novel technologies like live imaging and tracing present an accessible and efficient platform to visualize and illuminate these processes [117, 118].

Conclusions

In summary, although notable progress has been made in HF regeneration, it remains a work-in-progress. Future investigations should emphasize the regenerative nature and produce HFs from viable sources to propel organ reconstruction beyond basic science and ultimately benefit patients. Understanding the dynamics of regenerated HFs and their interplay with surroundings may offer solutions to the constrained lifespan of HFs. With deeper insights into the underlying mechanism, we will be able to define authentic HF reconstruction, normalize engineering procedures, and thus update therapeutics for alopecia and cutaneous defects. More importantly, achievements in HF neogenesis will encourage de novo organogenesis on a greater scale.

Data availability

Not applicable.

Abbreviations

ALI:

Air-liquid interface

BMP:

Bone morphogenetic protein

CNCC:

Cranial neural crest cell

DBB:

Droplet-based bioprinting

DDK2:

Dickkopf WNT signaling pathway inhibitor 2

DP:

Dermal papilla

DPC:

Dermal papilla cell

EBB:

Extrusion-based bioprinting

EDA:

Ectodysplasin A

EDAR:

Ectodysplasin A receptor

EDN1:

Endothelin 1

EMI:

Epithelial-mesenchymal interaction

ESC:

Embryonic stem cell

FGF:

Fibroblast growth factor

HF:

Hair follicle

HFG:

Hair follicle germ

HFSC:

Hair follicle stem cell

HUVEC:

Human umbilical vein endothelial cell

iPSC:

Induced pluripotent stem cell

LAB:

Laser-assisted bioprinting

ORS:

Outer root sheath

OSM:

Oncostatin M

PDGFA:

Platelet-derived growth factor subunit A

PSC:

Pluripotent stem cell

SBB:

Stereolithography-based bioprinting

SHH:

Sonic hedgehog signaling molecule

TAC:

Transit-amplifying cell

TGF:

Transforming growth factor

TRPV1:

Transient receptor potential cation channel subfamily V member 1

References

  1. Nuutila K. Hair follicle transplantation for Wound Repair. Adv Wound Care. 2021;10(3):153–63.

    Article  Google Scholar 

  2. Zhang B, Chen T. Local and systemic mechanisms that control the hair follicle stem cell niche. Nat Rev Mol Cell Biol. 2024;25(2):87–100.

    Article  CAS  PubMed  Google Scholar 

  3. Sennett R, Rendl M. Mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling. Semin Cell Dev Biol. 2012;23(8):917–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Xu Z, Wang W, Jiang K, Yu Z, Huang H, Wang F, et al. Embryonic attenuated Wnt/β-catenin signaling defines niche location and long-term stem cell fate in hair follicle. Elife. 2015;4:e10567.

    Article  PubMed  PubMed Central  Google Scholar 

  5. Saxena N, Mok KW, Rendl M. An updated classification of hair follicle morphogenesis. Exp Dermatol. 2019;28(4):332–44.

    Article  PubMed  PubMed Central  Google Scholar 

  6. 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(8):1522–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Schmidt-Ullrich R, Tobin DJ, Lenhard D, Schneider P, Paus R, Scheidereit C. NF-kappaB transmits Eda A1/EdaR signalling to activate shh and cyclin D1 expression, and controls post-initiation hair placode down growth. Development. 2006;133(6):1045–57.

    Article  CAS  PubMed  Google Scholar 

  8. Ouspenskaia T, Matos I, Mertz AF, Fiore VF, Fuchs E. WNT-SHH antagonism specifies and expands stem cells prior to niche formation. Cell. 2016;164(1–2):156–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Huh SH, Närhi K, Lindfors PH, Häärä O, Yang L, Ornitz DM, et al. Fgf20 governs formation of primary and secondary dermal condensations in developing hair follicles. Genes Dev. 2013;27(4):450–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Biggs LC, Mäkelä OJ, Myllymäki SM, Das Roy R, Närhi K, Pispa J et al. Hair follicle dermal condensation forms via Fgf20 primed cell cycle exit, cell motility, and aggregation. Elife. 2018;7.

  11. Rishikaysh P, Dev K, Diaz D, Qureshi WM, Filip S, Mokry J. Signaling involved in hair follicle morphogenesis and development. Int J Mol Sci. 2014;15(1):1647–70.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Millar SE. Molecular mechanisms regulating hair follicle development. J Invest Dermatol. 2002;118(2):216–25.

    Article  CAS  PubMed  Google Scholar 

  13. Ge W, Tan SJ, Wang SH, Li L, Sun XF, Shen W, et al. Single-cell transcriptome profiling reveals dermal and epithelial cell fate decisions during embryonic hair follicle development. Theranostics. 2020;10(17):7581–98.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Inoue K, Aoi N, Sato T, Yamauchi Y, Suga H, Eto H, et al. Differential expression of stem-cell-associated markers in human hair follicle epithelial cells. Lab Invest. 2009;89(8):844–56.

    Article  CAS  PubMed  Google Scholar 

  15. Ouji Y, Ishizaka S, Nakamura-Uchiyama F, Okuzaki D, Yoshikawa M. Partial maintenance and long-term expansion of murine skin epithelial stem cells by Wnt-3a in vitro. J Invest Dermatol. 2015;135(6):1598–608.

    Article  CAS  PubMed  Google Scholar 

  16. Adav SS, Ng KW. Recent omics advances in hair aging biology and hair biomarkers analysis. Ageing Res Rev. 2023;91:102041.

    Article  CAS  PubMed  Google Scholar 

  17. Fantauzzo KA, Christiano AM. There and back again: hair follicle stem cell dynamics. Cell Stem Cell. 2011;8(1):8–9.

    Article  CAS  PubMed  Google Scholar 

  18. Hsu YC, Pasolli HA, Fuchs E. Dynamics between stem cells, niche, and progeny in the hair follicle. Cell. 2011;144(1):92–105.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Kobielak K, Stokes N, de la Cruz J, Polak L, Fuchs E. Loss of a quiescent niche but not follicle stem cells in the absence of bone morphogenetic protein signaling. Proc Natl Acad Sci U S A. 2007;104(24):10063–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Plikus MV, Mayer JA, de la Cruz D, Baker RE, Maini PK, Maxson R, et al. Cyclic dermal BMP signalling regulates stem cell activation during hair regeneration. Nature. 2008;451(7176):340–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Wang X, Liu Y, He J, Wang J, Chen X, Yang R. Regulation of signaling pathways in hair follicle stem cells. Burns Trauma. 2022;10.

  22. Oshimori N, Fuchs E. Paracrine TGF-β signaling counterbalances BMP-mediated repression in hair follicle stem cell activation. Cell Stem Cell. 2012;10(1):63–75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Morgan BA. 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(7):a015180.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Foitzik K, Lindner G, Mueller-Roever S, Maurer M, Botchkareva N, Botchkarev V, et al. Control of murine hair follicle regression (catagen) by TGF-beta1 in vivo. Faseb j. 2000;14(5):752–60.

    Article  CAS  PubMed  Google Scholar 

  25. 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(1):5114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Heitman N, Sennett R, Mok KW, Saxena N, Srivastava D, Martino P, et al. Dermal sheath contraction powers stem cell niche relocation during hair cycle regression. Science. 2020;367(6474):161–6.

    Article  CAS  PubMed  Google Scholar 

  27. Martino PA, Heitman N, Rendl M. The dermal sheath: an emerging component of the hair follicle stem cell niche. Exp Dermatol. 2021;30(4):512–21.

    Article  PubMed  Google Scholar 

  28. Ohyama M. Use of human intra-tissue stem/progenitor cells and induced pluripotent stem cells for hair follicle regeneration. Inflamm Regen. 2019;39:4.

    Article  PubMed  PubMed Central  Google Scholar 

  29. Yuspa SH, Morgan DL, Walker RJ, Bates RR. The growth of fetal mouse skin in cell culture and transplantation to F1 mice. J Invest Dermatol. 1970;55(6):379–89.

    Article  CAS  PubMed  Google Scholar 

  30. Worst PK, Mackenzie IC, Fusenig NE. Reformation of organized epidermal structure by transplantation of suspensions and cultures of epidermal and dermal cells. Cell Tissue Res. 1982;225(1):65–77.

    Article  CAS  PubMed  Google Scholar 

  31. Weinberg WC, Goodman LV, George C, Morgan DL, Ledbetter S, Yuspa SH, et al. Reconstitution of hair follicle development in vivo: determination of follicle formation, hair growth, and hair quality by dermal cells. J Invest Dermatol. 1993;100(3):229–36.

    Article  CAS  PubMed  Google Scholar 

  32. Zheng Y, Du X, Wang W, Boucher M, Parimoo S, Stenn K. Organogenesis from dissociated cells: generation of mature cycling hair follicles from skin-derived cells. J Invest Dermatol. 2005;124(5):867–76.

    Article  CAS  PubMed  Google Scholar 

  33. Nakao K, Morita R, Saji Y, Ishida K, Tomita Y, Ogawa M, et al. The development of a bioengineered organ germ method. Nat Methods. 2007;4(3):227–30.

    Article  CAS  PubMed  Google Scholar 

  34. Toyoshima KE, Asakawa K, Ishibashi N, Toki H, Ogawa M, Hasegawa T, et al. Fully functional hair follicle regeneration through the rearrangement of stem cells and their niches. Nat Commun. 2012;3:784.

    Article  PubMed  Google Scholar 

  35. Kageyama T, Shimizu A, Anakama R, Nakajima R, Suzuki K, Okubo Y, et al. Reprogramming of three-dimensional microenvironments for in vitro hair follicle induction. Sci Adv. 2022;8(42):eadd4603.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kageyama T, Yoshimura C, Myasnikova D, Kataoka K, Nittami T, Maruo S, et al. Spontaneous hair follicle germ (HFG) formation in vitro, enabling the large-scale production of HFGs for regenerative medicine. Biomaterials. 2018;154:291–300.

    Article  CAS  PubMed  Google Scholar 

  37. Yen CM, Chan CC, Lin SJ. High-throughput reconstitution of epithelial-mesenchymal interaction in folliculoid microtissues by biomaterial-facilitated self-assembly of dissociated heterotypic adult cells. Biomaterials. 2010;31(15):4341–52.

    Article  CAS  PubMed  Google Scholar 

  38. Jahoda CA, Horne KA, Oliver RF. Induction of hair growth by implantation of cultured dermal papilla cells. Nature. 1984;311(5986):560–2.

    Article  CAS  PubMed  Google Scholar 

  39. Jahoda CA, Reynolds AJ, Oliver RF. Induction of hair growth in ear wounds by cultured dermal papilla cells. J Invest Dermatol. 1993;101(4):584–90.

    Article  CAS  PubMed  Google Scholar 

  40. Thangapazham RL, Klover P, Wang JA, Zheng Y, Devine A, Li S, et al. Dissociated human dermal papilla cells induce hair follicle neogenesis in grafted dermal-epidermal composites. J Invest Dermatol. 2014;134(2):538–40.

    Article  CAS  PubMed  Google Scholar 

  41. Ohyama M, Veraitch O. Strategies to enhance epithelial-mesenchymal interactions for human hair follicle bioengineering. J Dermatol Sci. 2013;70(2):78–87.

    Article  CAS  PubMed  Google Scholar 

  42. Yang R, Zheng Y, Burrows M, Liu S, Wei Z, Nace A, et al. Generation of folliculogenic human epithelial stem cells from induced pluripotent stem cells. Nat Commun. 2014;5:3071.

    Article  PubMed  Google Scholar 

  43. Gnedeva K, Vorotelyak E, Cimadamore F, Cattarossi G, Giusto E, Terskikh VV, et al. Derivation of hair-inducing cell from human pluripotent stem cells. PLoS ONE. 2015;10(1):e0116892.

    Article  PubMed  PubMed Central  Google Scholar 

  44. Sugiyama-Nakagiri Y, Fujimura T, Moriwaki S. Induction of skin-derived Precursor cells from Human Induced Pluripotent Stem cells. PLoS ONE. 2016;11(12):e0168451.

    Article  PubMed  PubMed Central  Google Scholar 

  45. Veraitch O, Mabuchi Y, Matsuzaki Y, Sasaki T, Okuno H, Tsukashima A, et al. 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.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ebner-Peking P, Krisch L, Wolf M, Hochmann S, Hoog A, Vári B, et al. Self-assembly of differentiated progenitor cells facilitates spheroid human skin organoid formation and planar skin regeneration. Theranostics. 2021;11(17):8430–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lei M, Schumacher LJ, Lai YC, Juan WT, Yeh CY, Wu P, et al. Self-organization process in newborn skin organoid formation inspires strategy to restore hair regeneration of adult cells. Proc Natl Acad Sci U S A. 2017;114(34):E7101–10.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Lei M, Jiang J, Wang M, Wu W, Zhang J, Liu W, et al. Epidermal-dermal coupled spheroids are important for tissue pattern regeneration in reconstituted skin explant cultures. NPJ Regen Med. 2023;8(1):65.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Ohyama M, Zheng Y, Paus R, Stenn KS. The mesenchymal component of hair follicle neogenesis: background, methods and molecular characterization. Exp Dermatol. 2010;19(2):89–99.

    Article  PubMed  Google Scholar 

  50. Higgins CA, Chen JC, Cerise JE, Jahoda CA, Christiano AM. Microenvironmental reprogramming by three-dimensional culture enables dermal papilla cells to induce de novo human hair-follicle growth. Proc Natl Acad Sci U S A. 2013;110(49):19679–88.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Rendl M, Polak L, Fuchs E. BMP signaling in dermal papilla cells is required for their hair follicle-inductive properties. Genes Dev. 2008;22(4):543–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Soma T, Fujiwara S, Shirakata Y, Hashimoto K, Kishimoto J. Hair-inducing ability of human dermal papilla cells cultured under Wnt/β-catenin signalling activation. Exp Dermatol. 2012;21(4):307–9.

    Article  CAS  PubMed  Google Scholar 

  53. Ohyama M, Kobayashi T, Sasaki T, Shimizu A, Amagai M. Restoration of the intrinsic properties of human dermal papilla in vitro. J Cell Sci. 2012;125(Pt 17):4114–25.

    CAS  PubMed  Google Scholar 

  54. Kang BM, Kwack MH, Kim MK, Kim JC, Sung YK. Sphere formation increases the ability of cultured human dermal papilla cells to induce hair follicles from mouse epidermal cells in a reconstitution assay. J Invest Dermatol. 2012;132(1):237–9.

    Article  CAS  PubMed  Google Scholar 

  55. Osada A, Iwabuchi T, Kishimoto J, Hamazaki TS, Okochi H. Long-term culture of mouse vibrissal dermal papilla cells and de novo hair follicle induction. Tissue Eng. 2007;13(5):975–82.

    Article  CAS  PubMed  Google Scholar 

  56. Abaci HE, Coffman A, Doucet Y, Chen J, Jacków J, Wang E, et al. Tissue engineering of human hair follicles using a biomimetic developmental approach. Nat Commun. 2018;9(1):5301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Wu J, Ma J, Zhuang H, Ma H, Wu C. 3D bioprinting of calcium molybdate nanoparticles-containing immunomodulatory bioinks for hair regrowth. Nano Today. 2023;51:101917.

    Article  CAS  Google Scholar 

  58. Zhao Q, Li N, Zhang H, Lei X, Cao Y, Xia G, et al. Chemically induced transformation of human dermal fibroblasts to hair-inducing dermal papilla-like cells. Cell Prolif. 2019;52(5):e12652.

    Article  PubMed  PubMed Central  Google Scholar 

  59. 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(8).

  60. Xie B, Chen M, Ding P, Lei L, Zhang X, Zhu D, et al. Induction of dermal fibroblasts into dermal papilla cell-like cells in hydrogel microcapsules for enhanced hair follicle regeneration. Appl Mater Today. 2020;21:100805.

    Article  Google Scholar 

  61. Itoh M, Kiuru M, Cairo MS, Christiano AM. Generation of keratinocytes from normal and recessive dystrophic epidermolysis bullosa-induced pluripotent stem cells. Proc Natl Acad Sci U S A. 2011;108(21):8797–802.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Itoh M, Umegaki-Arao N, Guo Z, Liu L, Higgins CA, Christiano AM. Generation of 3D skin equivalents fully reconstituted from human induced pluripotent stem cells (iPSCs). PLoS ONE. 2013;8(10):e77673.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Takagi R, Ishimaru J, Sugawara A, Toyoshima KE, Ishida K, Ogawa M, et al. Bioengineering a 3D integumentary organ system from iPS cells using an in vivo transplantation model. Sci Adv. 2016;2(4):e1500887.

    Article  PubMed  PubMed Central  Google Scholar 

  64. Lee J, Bӧscke R, Tang PC, Hartman BH, Heller S, Koehler KR. Hair follicle development in mouse pluripotent stem cell-derived skin organoids. Cell Rep. 2018;22(1):242–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Koehler KR, Mikosz AM, Molosh AI, Patel D, Hashino E. Generation of inner ear sensory epithelia from pluripotent stem cells in 3D culture. Nature. 2013;500(7461):217–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Koehler KR, Hashino E. 3D mouse embryonic stem cell culture for generating inner ear organoids. Nat Protoc. 2014;9(6):1229–44.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Lee J, Rabbani CC, Gao H, Steinhart MR, Woodruff BM, Pflum ZE, et al. Hair-bearing human skin generated entirely from pluripotent stem cells. Nature. 2020;582(7812):399–404.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Ma J, Li W, Cao R, Gao D, Zhang Q, Li X, et al. Application of an iPSC-Derived Organoid Model for Localized Scleroderma Therapy. Adv Sci (Weinh). 2022;9(16):e2106075.

    Article  PubMed  Google Scholar 

  69. Ma J, Liu J, Gao D, Li X, Zhang Q, Lv L, et al. Establishment of human pluripotent stem cell-derived skin Organoids enabled Pathophysiological Model of SARS-CoV-2 infection. Adv Sci (Weinh). 2022;9(7):e2104192.

    Article  PubMed  Google Scholar 

  70. Ramovs V, Janssen H, Fuentes I, Pitaval A, Rachidi W, Chuva de Sousa Lopes SM, et al. Characterization of the epidermal-dermal junction in hiPSC-derived skin organoids. Stem Cell Rep. 2022;17(6):1279–88.

    Article  CAS  Google Scholar 

  71. Jung SY, You HJ, Kim MJ, Ko G, Lee S, Kang KS. Wnt-activating human skin organoid model of atopic dermatitis induced by Staphylococcus aureus and its protective effects by Cutibacterium acnes. iScience. 2022;25(10):105150.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Pan J, Yung Chan S, Common JE, Amini S, Miserez A, Birgitte Lane E, et al. Fabrication of a 3D hair follicle-like hydrogel by soft lithography. J Biomed Mater Res A. 2013;101(11):3159–69.

    Article  PubMed  Google Scholar 

  73. Ansaf RB, Ziebart R, Gudapati H, Simoes Torigoe RM, Victorelli S, Passos J, et al. 3D bioprinting-a model for skin aging. Regen Biomater. 2023;10:rbad060.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Datta P, Barui A, Wu Y, Ozbolat V, Moncal KK, Ozbolat IT. Essential steps in bioprinting: from pre- to post-bioprinting. Biotechnol Adv. 2018;36(5):1481–504.

    Article  CAS  PubMed  Google Scholar 

  75. Banerjee D, Singh YP, Datta P, Ozbolat V, O’Donnell A, Yeo M, et al. Strategies for 3D bioprinting of spheroids: a comprehensive review. Biomaterials. 2022;291:121881.

    Article  CAS  PubMed  Google Scholar 

  76. Xu H-Q, Liu J-C, Zhang Z-Y, Xu C-X. A review on cell damage, viability, and functionality during 3D bioprinting. Military Med Res. 2022;9(1).

  77. Weng T, Zhang W, Xia Y, Wu P, Yang M, Jin R, et al. 3D bioprinting for skin tissue engineering: current status and perspectives. J Tissue Eng. 2021;12:20417314211028574.

    Article  PubMed  PubMed Central  Google Scholar 

  78. Parak A, Pradeep P, du Toit LC, Kumar P, Choonara YE, Pillay V. Functionalizing bioinks for 3D bioprinting applications. Drug Discov Today. 2019;24(1):198–205.

    Article  CAS  PubMed  Google Scholar 

  79. Gopinathan J, Noh I. Recent trends in bioinks for 3D printing. Biomater Res. 2018;22:11.

    Article  PubMed  PubMed Central  Google Scholar 

  80. Masri S, Fauzi MB. Current insight of Printability Quality improvement strategies in natural-based Bioinks for skin regeneration and Wound Healing. Polym (Basel). 2021;13(7).

  81. Ríos-Galacho M, Martínez-Moreno D, López-Ruiz E, Gálvez-Martín P, Marchal JA. An overview on the Manufacturing of functional and mature Cellular skin substitutes. Tissue Eng Part B Rev. 2022;28(5):1035–52.

    Article  PubMed  Google Scholar 

  82. Datta P, Cabrera LY, Ozbolat IT. Ethical challenges with 3D bioprinted tissues and organs. Trends Biotechnol. 2023;41(1):6–9.

    Article  CAS  PubMed  Google Scholar 

  83. Wang Y, Yuan X, Yao B, Zhu S, Zhu P, Huang S. Tailoring bioinks of extrusion-based bioprinting for cutaneous wound healing. Bioact Mater. 2022;17:178–94.

    CAS  PubMed  PubMed Central  Google Scholar 

  84. Kang D, Liu Z, Qian C, Huang J, Zhou Y, Mao X, et al. 3D bioprinting of a gelatin-alginate hydrogel for tissue-engineered hair follicle regeneration. Acta Biomater. 2023;165:19–30.

    Article  CAS  PubMed  Google Scholar 

  85. Jorgensen AM, Gorkun A, Mahajan N, Willson K, Clouse C, Jeong CG, et al. Multicellular bioprinted skin facilitates human-like skin architecture in vivo. Sci Transl Med. 2023;15(716):eadf7547.

    Article  CAS  PubMed  Google Scholar 

  86. Motter Catarino C, Cigaran Schuck D, Dechiario L, Karande P. Incorporation of hair follicles in 3D bioprinted models of human skin. Sci Adv. 2023;9(41):eadg0297.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Zhao W, Chen H, Zhang Y, Zhou D, Liang L, Liu B, et al. Adaptive multi-degree-of-freedom in situ bioprinting robot for hair-follicle-inclusive skin repair: a preliminary study conducted in mice. Bioeng Transl Med. 2022;7(3):e10303.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Chen H, Ma X, Gao T, Zhao W, Xu T, Liu Z. Robot-assisted in situ bioprinting of gelatin methacrylate hydrogels with stem cells induces hair follicle-inclusive skin regeneration. Biomed Pharmacother. 2023;158:114140.

    Article  CAS  PubMed  Google Scholar 

  89. Kang Y, Yeo M, Derman ID, Ravnic DJ, Singh YP, Alioglu MA et al. Intraoperative Bioprinting of Human Adipose-derived Stem cells and Extra-cellular Matrix Induces Hair Follicle-Like Downgrowths and Adipose Tissue Formation during Full-thickness Craniomaxillofacial Skin Reconstruction. bioRxiv. 2023.

  90. Murphy SV, De Coppi P, Atala A. Opportunities and challenges of translational 3D bioprinting. Nat Biomed Eng. 2020;4(4):370–80.

    Article  PubMed  Google Scholar 

  91. Yang L, Güell M, Niu D, George H, Lesha E, Grishin D, et al. Genome-wide inactivation of porcine endogenous retroviruses (PERVs). Science. 2015;350(6264):1101–4.

    Article  CAS  PubMed  Google Scholar 

  92. Niu D, Wei HJ, Lin L, George H, Wang T, Lee IH, et al. Inactivation of porcine endogenous retrovirus in pigs using CRISPR-Cas9. Science. 2017;357(6357):1303–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Niu D, Ma X, Yuan T, Niu Y, Xu Y, Sun Z, et al. Porcine genome engineering for xenotransplantation. Adv Drug Deliv Rev. 2021;168:229–45.

    Article  CAS  PubMed  Google Scholar 

  94. Sun W, Starly B, Daly AC, Burdick JA, Groll J, Skeldon G, et al. The bioprinting roadmap. Biofabrication. 2020;12(2):022002.

    Article  CAS  PubMed  Google Scholar 

  95. Pleguezuelos-Beltrán P, Gálvez-Martín P, Nieto-García D, Marchal JA, López-Ruiz E. Advances in spray products for skin regeneration. Bioact Mater. 2022;16:187–203.

    PubMed  PubMed Central  Google Scholar 

  96. Nanmo A, Yan L, Asaba T, Wan L, Kageyama T, Fukuda J. Bioprinting of hair follicle germs for hair regenerative medicine. Acta Biomater. 2023;165:50–9.

    Article  CAS  PubMed  Google Scholar 

  97. Kageyama T, Yan L, Shimizu A, Maruo S, Fukuda J. Preparation of hair beads and hair follicle germs for regenerative medicine. Biomaterials. 2019;212:55–63.

    Article  CAS  PubMed  Google Scholar 

  98. Hoang DM, Pham PT, Bach TQ, Ngo ATL, Nguyen QT, Phan TTK, et al. Stem cell-based therapy for human diseases. Signal Transduct Target Ther. 2022;7(1):272.

    Article  PubMed  PubMed Central  Google Scholar 

  99. Jafarzadeh A, PourMohammad A, Goodarzi A. A systematic review of the efficacy, safety and satisfaction of regenerative medicine treatments, including platelet-rich plasma, stromal vascular fraction and stem cell-conditioned medium for hypertrophic scars and keloids. Int Wound J. 2024;21(4):e14557.

    Article  PubMed  Google Scholar 

  100. Jafarzadeh A, Mohammad AP, Goodarzi A. A systematic review of Case Series and clinical trials investigating Regenerative Medicine for the Treatment of Vitiligo. J Cosmet Dermatol. 2024.

  101. Lee JH, Choi S. Deciphering the molecular mechanisms of stem cell dynamics in hair follicle regeneration. Exp Mol Med. 2024;56(1):110–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Jafarzadeh A, Pour Mohammad A, Keramati H, Zeinali R, Khosravi M, Goodarzi A. Regenerative medicine in the treatment of specific dermatologic disorders: a systematic review of randomized controlled clinical trials. Stem Cell Res Ther. 2024;15(1):176.

    Article  PubMed  PubMed Central  Google Scholar 

  103. Buffoli B, Rinaldi F, Labanca M, Sorbellini E, Trink A, Guanziroli E, et al. The human hair: from anatomy to physiology. Int J Dermatol. 2014;53(3):331–41.

    Article  PubMed  Google Scholar 

  104. Nilforoushzadeh MA, Zare M, Zarrintaj P, Alizadeh E, Taghiabadi E, Heidari-Kharaji M, et al. Engineering the niche for hair regeneration - A critical review. Nanomedicine. 2019;15(1):70–85.

    Article  CAS  PubMed  Google Scholar 

  105. Correia M, Lopes J, Lopes D, Melero A, Makvandi P, Veiga F, et al. Nanotechnology-based techniques for hair follicle regeneration. Biomaterials. 2023;302:122348.

    Article  CAS  PubMed  Google Scholar 

  106. Mohite P, Puri A, Dave R, Budar A, Munde S, Ghosh SB, et al. Unlocking the therapeutic potential: odyssey of induced pluripotent stem cells in precision cell therapies. Int J Surg. 2024;110(10):6432–55.

    Article  PubMed  PubMed Central  Google Scholar 

  107. Shi Y, Inoue H, Wu JC, Yamanaka S. Induced pluripotent stem cell technology: a decade of progress. Nat Rev Drug Discov. 2017;16(2):115–30.

    Article  CAS  PubMed  Google Scholar 

  108. Yin X, Li Q, Shu Y, Wang H, Thomas B, Maxwell JT, et al. Exploiting urine-derived induced pluripotent stem cells for advancing precision medicine in cell therapy, disease modeling, and drug testing. J Biomed Sci. 2024;31(1):47.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Huang CY, Liu CL, Ting CY, Chiu YT, Cheng YC, Nicholson MW, et al. Human iPSC banking: barriers and opportunities. J Biomed Sci. 2019;26(1):87.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Cerneckis J, Cai HX, Shi YH. Induced pluripotent stem cells (iPSCs): molecular mechanisms of induction and applications. Signal Transduct Target Therapy. 2024;9(1).

  111. Ekser B, Li P, Cooper DKC. Xenotransplantation: past, present, and future. Curr Opin Organ Transpl. 2017;22(6):513–21.

    Article  Google Scholar 

  112. Cooper DKC, Gaston R, Eckhoff D, Ladowski J, Yamamoto T, Wang L, et al. Xenotransplantation-the current status and prospects. Br Med Bull. 2018;125(1):5–14.

    Article  CAS  PubMed  Google Scholar 

  113. Aschheim K, DeFrancesco L. Xenotransplantation: how close are we? Nat Biotechnol. 2023;41(4):452–60.

    Article  CAS  PubMed  Google Scholar 

  114. Wu Y, Qin M, Yang X. Organ bioprinting: progress, challenges and outlook. J Mater Chem B. 2023;11(43):10263–87.

    Article  CAS  PubMed  Google Scholar 

  115. Soman SS, Vijayavenkataraman S. Applications of 3D Bioprinted-Induced pluripotent stem cells in Healthcare. Int J Bioprint. 2020;6(4):280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Li Y, Li L, Chen ZN, Gao G, Yao R, Sun W. Engineering-derived approaches for iPSC preparation, expansion, differentiation and applications. Biofabrication. 2017;9(3):032001.

    Article  PubMed  Google Scholar 

  117. Pineda CM, Park S, Mesa KR, Wolfel M, Gonzalez DG, Haberman AM, et al. Intravital imaging of hair follicle regeneration in the mouse. Nat Protoc. 2015;10(7):1116–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Sun Q, Lee W, Hu H, Ogawa T, De Leon S, Katehis I, et al. Dedifferentiation maintains melanocyte stem cells in a dynamic niche. Nature. 2023;616(7958):774–82.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors acknowledge the assistance provided by Duanqing Pei, Bo Wang, Yue Qin and Junyang Li from School of Life Sciences, Westlake University in coordinating the article content. The authors appreciate the reproduction permission of Figure 1 from Springer Nature (Zhang B, Chen T. Local and systemic mechanisms that control the hair follicle stem cell niche. Nat Rev Mol Cell Biol. 2024;25(2):87-100. https://www.nature.com/articles/s41580-023-00662-3). Figures 2, 3 and 4 were drawn by Figdraw. The authors declare that they have not used AI-generated work in this manuscript.

Funding

This work was supported by the Key Discipline Project of Hangzhou (grant number 0020200044), and Medicine and Health Research Project of Zhejiang Province (grant number 2025KY1037).

Author information

Authors and Affiliations

Authors

Contributions

XC and ZZ conceptualized and wrote the manuscript. XQ performed the literature search. XC, HS and HC revised the manuscript. JZ supervised the writing process and reviewed the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Jufang Zhang.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Chu, X., Zhou, Z., Qian, X. et al. Functional regeneration strategies of hair follicles: advances and challenges. Stem Cell Res Ther 16, 77 (2025). https://doi.org/10.1186/s13287-025-04210-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13287-025-04210-y

Keywords