Elsevier

Biomaterials

Volume 167, June 2018, Pages 121-131
Biomaterials

Inducing hair follicle neogenesis with secreted proteins enriched in embryonic skin

https://doi.org/10.1016/j.biomaterials.2018.03.003Get rights and content

Abstract

Organ development is a sophisticated process of self-organization. However, despite growing understanding of the developmental mechanisms, little is known about how to reactivate them postnatally for regeneration. We found that treatment of adult non-hair fibroblasts with cell-free extract from embryonic skin conferred upon them the competency to regenerate hair follicles. Proteomics analysis identified three secreted proteins enriched in the embryonic skin, apolipoprotein-A1, galectin-1 and lumican that together were essential and sufficient to induce new hair follicles. These 3 proteins show a stage-specific co-enrichment in the perifolliculogenetic embryonic dermis. Mechanistically, exposure to embryonic skin extract or to the combination of the 3 proteins altered the gene expression to an inductive hair follicle dermal papilla fibroblast-like profile and activated Igf and Wnt signaling, which are crucial for the regeneration process. Therefore, a cocktail of organ-specific extracellular proteins from the embryonic environment can render adult cells competent to re-engage in developmental interactions for organ neogenesis. Identification of factors that recreate the extracellular context of respective developing tissues can become an important strategy to promote regeneration in adult organs.

Introduction

Compared with the profound spontaneous restoration of lost tissues and organs in lower vertebrates [[1], [2], [3]], mammals show limited regeneration after injury and damaged tissues are often replaced by morphologically and functionally debilitating scars [4]. To promote regeneration, attempts have been made by transplantation of stem cells, organoids or bioartificial organs [[5], [6], [7]], yet in most cases functional incorporation and maintenance of transplanted cells remains challenging [8]. In skin, it usually heals by fibrosis rather than regeneration after injury, leading to scar formation and permanent loss of skin appendages, prominently hair follicles (HFs) [9]. Although promotion of skin healing has been attempted by transplanting cultured keratinocytes or skin equivalents [10,11], regeneration of HFs is still an unmet clinical need.

The HF is a mini-organ composed of an epithelial cylinder and specialized dermal papilla (DP) fibroblasts [12]. Similar to other ectodermal mini-organs, HF development depends on well-choreographed epithelial-mesenchymal interactions initiated by the crosstalk between epithelial and dermal embryonic skin progenitors [12]. During HF morphogenesis, epithelial progenitors gradually adopt a follicular fate, whereas dermal progenitors differentiate into DP fibroblasts. In normal adult skin, new HFs generally can not form [12]. As an exception, HFs can regenerate spontaneously in the center of very large wounds and with low efficiency in humans via incompletely understood mechanisms [[13], [14], [15]]. Such examples indicate that, in principle, the potential for HF neogenesis is preserved and can possibly be unleashed.

To induce new HFs postnatally, Oliver et al. demonstrated that microdissected DP could induce new HFs from epidermis when transplanted into the subepidermal space [16]. Following this seminal work, HF neogenesis in adults has largely relied on culture-expanded DP fibroblasts, which are capable of reinitiating developmental epithelial-mesenchymal interactions with competent keratinocytes [[17], [18], [19], [20]]. The HF-inducing ability of cultured DP fibroblasts is easily lost during culture and this limits their use for large-scale HF regeneration [17]. In addition to adult DP cells, freshly isolated newborn murine dermal fibroblasts have also been demonstrated to be capable of inducing HF neogenesis [7,19,[21], [22], [23]], but such HF inductivity of dermal fibroblasts are quickly lost in postnatal life.

An alternative strategy for inducing new HFs is through the use of embryonic skin [24]. In contrast to adult skin, wounded embryonic skin heals without scarring and with extensive HF neogenesis [25]. When embryonic dermal tissues are combined with postnatal epithelium, they are able to induce new HFs [24]. What are the key factors that define the HF-inducing ability of embryonic skin? It was demonstrated that isolated embryonic dermal cells are able to induce new HFs when combined with keratinocytes [26]. On the other hand, it has also been suggested that embryonic tissues create unique extracellular environments conducive to regeneration [25,27]. The cell-free extracellular matrix from adult tissue has been shown to enhance tissue regeneration [6,[27], [28], [29], [30]]. Isolation of defined factors present in the extracellular matrix that can elicit neogenesis of an organ can open novel, therapeutically amenable organ-specific regeneration protocols. However, induction of the neogenesis of a specific organ by defined extracellular factors has not been achieved. Here, we demonstrate that a cocktail of 3 secreted protein factors enriched in the developmental skin can induce neogenesis of the HF miniorgan through reactivating the epithelial-mesenchymal crosstalk between adult skin cells.

Section snippets

Animals

All animal experiments were approved by the Institutional Animal Care and Use Committee at National Taiwan University. Animals were housed in animal facilities of National Taiwan University. Wistar rats were purchased from BioLASCO Taiwan Co. and C57BL/6 mice and nude mice (BALB/cAnN-Foxn1nu/CrlNarl) were from Taiwan National Laboratory Animal Center. Z/AP mice expressing the lacZ reporter gene to overexpress β-galactosidase were from Jackson Laboratory [31]. For invasive experiments, animals

Induction of HF neogenesis by cell-free extract from E15.5-E17.5 embryonic rat skin

We first determined whether cell-free extracts from embryonic skin can induce HF neogenesis. In the HF neogenesis assay, the contamination of preformed HFs in keratinocytes can lead to HF formation without co-administration of inductive mesenchymal cells [21]. We carefully removed preformed HFs from epidermal keratinocytes to avoid this and the isolated keratinocytes alone did not grow any new HFs in patch assays for HF neogenesis (Fig. S1b, and S1b’). Formation of new HFs in the HF neogenesis

Discussion

We show that the three core proteins, Apoa1, Lgals1, and Lum are necessary and sufficient to induce HF neogenesis from adult cells, and that additional factors can further enhance the efficiency. ApoA1 is the major protein component of plasma high-density lipoprotein particles [55]. Due to their function in lipid metabolism and transport, apolipoproteins are actively involved in tissue healing and regeneration where newly formed lipid membranes are required [[56], [57], [58]]. In addition to

Conclusion

Regeneration of lost tissues in adults depends on the recapitulation of the developmental process of the respective organs. Using the HF regeneration as a model, we unveiled a novel method to re-elicit the developmental scheme for organ neogenesis. We demonstrated that defined extracellular proteins enriched in the embryonic developmental environment can reactivate organogenesis from adult cells. Three core proteins, Apoa1, Lgals1 and Lum, are necessary and sufficient for this regenerative

Acknowledgements

We thank the staff of the 8th Core Lab, Department of Medical Research, National Taiwan University Hospital for technical support and lab members in the S.J.L. lab for discussion. The mass spectrometry analyses using the Orbitrap Fusion mass spectrometers were performed by the Common Mass Spectrometry Facilities, Institute of Biological Chemistry, Academia Sinica. Analysis of RNA-sequencing data was performed by the Bioinformatics Core, Center of Genomic Medicine, National Taiwan University.

References (84)

  • K.K.L. Phua et al.

    Non-linear enhancement of mRNA delivery efficiencies by influenza A derived NS1 protein engendering host gene inhibition property

    Biomaterials

    (2017)
  • Y. Zheng et al.

    Organogenesis from dissociated cells: generation of mature cycling hair follicles from skin-derived cells

    J. Invest. Dermatol.

    (2005)
  • W.Y. Huang et al.

    Stress-induced premature senescence of dermal papilla cells compromises hair follicle epithelial-mesenchymal interaction

    J. Dermatol. Sci.

    (2017)
  • J. Huelsken et al.

    beta-Catenin controls hair follicle morphogenesis and stem cell differentiation in the skin

    Cell

    (2001)
  • C.C. Tsou et al.

    IDEAL-Q, an automated tool for label-free quantitation analysis using an efficient peptide alignment approach and spectral data validation

    Mol. Cell. Proteomics : MCP

    (2010)
  • T. Andl et al.

    WNT signals are required for the initiation of hair follicle development

    Dev. Cell

    (2002)
  • C.G. Brouillette et al.

    Structural models of human apolipoprotein A-I: a critical analysis and review

    Biochim. Biophys. Acta

    (2001)
  • H.N. van der Vliet et al.

    Apolipoprotein A-V: a novel apolipoprotein associated with an early phase of liver regeneration

    J. Biol. Chem.

    (2001)
  • A.C. Puche et al.

    Role of galectin-1 in the developing mouse olfactory system

    Dev. Biol.

    (1996)
  • S. Saika et al.

    Role of lumican in the corneal epithelium during wound healing

    J. Biol. Chem.

    (2000)
  • B.S. Kim et al.

    Direct 3D cell-printing of human skin with functional transwell system

    Biofabrication

    (2017)
  • R. Sennett et al.

    Mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling

    Semin. Cell Dev. Biol.

    (2012)
  • J.P. Liu et al.

    Mice carrying null mutations of the genes encoding insulin-like growth factor I (Igf-1) and type 1 IGF receptor (Igf1r)

    Cell

    (1993)
  • V.A. Botchkarev et al.

    Modulation of BMP signaling by noggin is required for induction of the secondary (nontylotrich) hair follicles

    J. Invest. Dermatol.

    (2002)
  • C.A. Collins et al.

    Polyclonal origin and hair induction ability of dermal papillae in neonatal and adult mouse back skin

    Dev. Biol.

    (2012)
  • J.P. Brockes

    Amphibian limb regeneration: rebuilding a complex structure

    Science

    (1997)
  • S.L. Johnson et al.

    Temperature-sensitive mutations that cause stage-specific defects in Zebrafish fin regeneration

    Genetics

    (1995)
  • R.O. Becker et al.

    Regeneration of the ventricular myocardium in amphibians

    Nature

    (1974)
  • T.A. Wynn et al.

    Mechanisms of fibrosis: therapeutic translation for fibrotic disease

    Nat Med

    (2012)
  • H.C. Ott et al.

    Perfusion-decellularized matrix: using nature's platform to engineer a bioartificial heart

    Nat Med

    (2008)
  • M. Lei 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)
  • V.F. Segers et al.

    Stem-cell therapy for cardiac disease

    Nature

    (2008)
  • P. Martin

    Wound healing–aiming for perfect skin regeneration

    Science

    (1997)
  • A. Veves et al.

    Apligraf Diabetic Foot Ulcer S. Graftskin, a human skin equivalent, is effective in the management of noninfected neuropathic diabetic foot ulcers: a prospective randomized multicenter clinical trial

    Diabetes Care

    (2001)
  • O'Connor NE, Mulliken JB, Banks-Schlegel S, Kehinde O, Green H. Grafting of burns with cultured epithelium prepared...
  • M. Ito et al.

    Wnt-dependent de novo hair follicle regeneration in adult mouse skin after wounding

    Nature

    (2007)
  • A.W. Seifert et al.

    Skin shedding and tissue regeneration in African spiny mice (Acomys)

    Nature

    (2012)
  • R.F. Oliver

    The induction of hair follicle formation in the adult hooded rat by vibrissa dermal papillae

    J. Embryol. Exp. Morphol.

    (1970)
  • C.A. Higgins et al.

    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)
  • R.J. Morris et al.

    Capturing and profiling adult hair follicle stem cells

    Nat. Biotechnol.

    (2004)
  • C. Ferraris et al.

    Adult epidermal keratinocytes are endowed with pilosebaceous forming abilities

    Int. J. Dev. Biol.

    (1997)
  • M.T. Longaker et al.

    The biology of fetal wound healing: a review

    Plast. Reconstr. Surg.

    (1991)
  • Cited by (24)

    • Design of a biofluid-absorbing bioactive sandwich-structured Zn–Si bioceramic composite wound dressing for hair follicle regeneration and skin burn wound healing

      2021, Bioactive Materials
      Citation Excerpt :

      This proves that our material mainly stimulates cell proliferation. In addition, a clear synergistic effect of the Zn2+/SiO32− combination in stimulating expression of growth factors (Stem cell indicator markers: VEGF, BMP-6, KGF, HGF; Hair follicle anagen markers: PDGF-α, PDGF-β, and C-Myc) was observed [46–49]. For stem cell indicator markers, Zn2+ alone only revealed limited enhancement of the expression of VEGF and BMP-6, but not HGF and KGF, while SiO32− alone did not show any stimulation effect on the expression of growth factors of HHDPCs.

    • Curcumin/Fe-SiO<inf>2</inf> nano composites with multi-synergistic effects for scar inhibition and hair follicle regeneration during burn wound healing

      2021, Applied Materials Today
      Citation Excerpt :

      The migration rate was analyzed by Image J software and determined by the ratio of the initial scratch area and the final scratch area. Gene expression of VEGF, BMP-6 and KGF in HHDPCs were detected by Q‐ RT‐ PCR, which is related to the viability of hair follicles [30,31]. HHDPCs were plated in 6‐well plates at a density of 1 × 105 cells/well for 24h.

    • Induction of dermal fibroblasts into dermal papilla cell-like cells in hydrogel microcapsules for enhanced hair follicle regeneration

      2020, Applied Materials Today
      Citation Excerpt :

      Alternatively, DPC-like cells can be obtained from differentiated cells. For example, non-hair adult murine fibroblasts can be committed to a DP fate after being transfected with 3 mRNAs (Apoa/Lgals1/Lum) [42,45–47], but it remains unclear whether this protocol is applicable for human cells. Moreover, the gene induction strategies raise safety concerns.

    • MoS<inf>2</inf> Nanoclusters-based biomaterials for disease- impaired wound therapy

      2020, Applied Materials Today
      Citation Excerpt :

      It effectively alleviated the dysfunction of diabetes on angiogenesis and provided adequate nutrition for wound healing, although the mechanism is still unclear. Previous studies have shown that the growth factors (e.g. VEGF and HGF) have important effects on hair follicle regeneration [38,39]. Zinc ion, one of bioactive inorganic ions, has an important function in hair follicle to treat hair disorders [40].

    • Engineering biomimetic and instructive materials for wound healing and regeneration

      2019, Current Opinion in Biomedical Engineering
      Citation Excerpt :

      More recently, a study has reported that scaffolds fabricated from decellularized neonatal tissues significantly improved the fibrotic outcome in an excisional mouse model [12]. Similarly, proteins extracted from embryonic skin rendered skin fibroblasts competent to regenerate functional hair follicles [46]. In a bottom-up approach, we engineered fibronectin nanofiber scaffolds, thus emulating the unique microenvironment of early embryogenesis.

    View all citing articles on Scopus
    View full text