Applied Materials Today
Volume 21, December 2020, 100805
Journal home page for Applied Materials Today

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

https://doi.org/10.1016/j.apmt.2020.100805Get rights and content

Highlights

  • The development of novel sources of human dermal papilla cells (hDPCs) is of great significance to hDPC-based hair follicle regeneration strategies. We have for the first time exploited the conversion of human dermal fibroblasts (hDFs) into hDPC-like cells in alginate-poly-L-lysine-alginate (APA) microcapsules. Our results show that the derived hDF-DPCs are comparable to primary DPCs on the molecular level, and they can induce the formation of hair follicle-like structures and hair pigmentation in nude mice. RNA-seq analyses indicate the activation of Wnt signaling pathway during the hDF-DPC conversion. Similarly, we demonstrate that mouse DFs differentiate into mouse DPC-like cells within the hydrogel microcapsules, indicating this approach is not limited to human cell type. The induction of dermal fibroblasts into DPC-like cells through biomimetic microencapsulation, devoid of chemical or gene reprogramming, would provide an alternative, abundant, and safe source of DPCs, which would help circumvent the bottleneck in DPC-based HF regeneration.

Abstract

Current approaches fail at obtaining adequate functional human dermal papilla cells (DPCs) to regenerate hair follicles (HFs). Here we induce human dermal fibroblasts (hDFs) into hDPC-like cells (hDF-DPCs) through alginate-poly-L-lysine-alginate (APA) microencapsulation. Our results show that the derived hDF-DPCs are comparable to primary DPCs on the molecular level, and they can induce the formation of hair follicle-like structures and hair pigmentation in nude mice. RNA-seq analyses indicate the activation of Wnt signaling pathway during the hDF-DPCs conversion. Likewise, we demonstrate that mouse DFs differentiate into mouse DPC-like cells within the hydrogel microcapsules. The induction of dermal fibroblasts into DPC-like cells through biomimetic microencapsulation, devoid of chemical or gene reprogramming, would provide an alternative, abundant, and safe source of DPCs, which would help circumvent the bottleneck in DPC-based HF regeneration.

Introduction

Hair loss is a common skin disorder associated with aging, diseases and medications, along with the full-thickness skin loss during severe wound injuries [1], [2], [3]. Current therapeutic treatments include medication and autologous hair transplantation [1,4]. Therapeutic drugs mainly prevent further hair loss rather than regenerate new hairs [5]. Hair transplant, a surgery that moves healthy hair follicles (HFs) from parts of the scalp to the bald area, is insufficient for patients with severe hair loss owing to the shortage of HFs [6]. With the advances of tissue engineering and regenerative medicine, cell therapy that aims at regenerating HFs emerges as a promising alternative to traditional HFs transplantation [7,8]. Dermal papilla (DP), a cluster of active specialized fibroblasts derived from dermal mesenchyme, is a spheroid structure located at the base of the HFs. DP cells (DPCs) are essential for HFs morphogenesis and regulation of hair cycling in both embryogenesis and postnatal regeneration [9,10]. However, the development of efficient expansion of DPCs with intrinsic features remains a great challenge, as DPCs proliferate slowly in vitro in two-dimensional (2D) cultures and quickly lose their hair-inducing activity during passaging [11,12]. 2D cultivation is unable to recapitulate the in vivo structures of densely packed DPCs within DPs [13]. Three-dimensional (3D) cultures could enable a microenvironment to facilitate HFs growth [14]. In light of such finding, we hypothesize that the 3D cultivation will allow us to obtain adequate DPCs for de novo folliculogenesis.

The prerequisite of 3D expansion of DPCs is the isolation and collection of a large number of DPCs. As DPCs proliferate slowly, the DPC shortage in DPC-based HFs regeneration still remains a bottleneck. The development of alternative sources of DPCs from other cell lines is thus desirable. DPCs are a specialized cell population sharing a same origin as inter-follicular dermal fibroblasts (DFs) and distinguish from the latter cells by their unique characteristics, such as aggregative behavior, distinct gene expression and the ability to induce the formation of new HFs. Fibroblasts are widely present in skin tissue, and can be extracted and expanded following well-established protocols. These advantages make fibroblasts a valuable cell source for cell therapy studies [15,16]. Although human dermal fibroblasts (hDFs) could differentiate into osteoblasts, neurons, cardiomyocytes, and melanocytes [17], [18], [19], [20], little effort has been made to investigate the derivation of functional DPC-like cells from hDFs.

Here we report a 3D cultivation system of alginate-poly-L-lysine-alginate (APA) microcapsules that restore the trichogenecity of DPCs pre-cultured under 2D conditions. Moreover, we for the first time induced hDFs into functional DPC-like cells through APA microencapsulation (Fig. S1). The hDFs in APA microcapsules exhibited the hDPCs features on the molecular level. We further elucidated that the hDFs into hDF-DPCs in APA microcapsules was activated by Wnt signaling pathways. These two strategies, separately or in combination, can supply a large number of safe and functional DPCs and circumvent the bottleneck in DPC-based regenerative strategies for de novo folliculogenesis.

Section snippets

Human Cell sources and isolation

All clinical procedures and protocols were based on informed consent and approved by the Ethics Committee of Central South University Xiangya Hospital. Human hair follicles were manually isolated from the occipital scalp obtained during hair transplant surgery. Primary hDFs were isolated from the circumcised foreskins of healthy human donors. All procedures were approved by Institutional Review Board of Central South University Xiangya Hospital (IRB No. 201611609) in accordance with the

Trichogenecity of cDPCs within APA microcapsules

Hydrogels have found great success in regenerating skin and skin appendage [27,28]. Our prior studies demonstrated that hydrogels can establish robust interaction with endogenous cells to promote complete skin regeneration [29]. In order to create a microenvironment to retain the hair inducing capacity, we developed APA hydrogel microcapsules, and encapsulated cDPCs within APA to establish spheroids (APA-DPCs) (Fig. 1A). ALP activity, which indicates the hair-inducing capacity of DPCs [30], was

Discussion

Many efforts had been made to obtain DPC-like cells from various stem cell lines, such as embryonic stem cells (ESCs), mesenchymal stem cells (MSCs) and inducible pluripotent stem cells (iPSCs) [25,[42], [43], [44]. Traditional processes are time-consuming and quite expensive. The induction of iPSCs into DPC-like cells, for example, requires the differentiation of iPSCs into neural crest (NC) cells, with a yield of approximately 20%-30%. The NC cells then undergo a second induction and

Conclusion

In summary, we first demonstrated that APA-cultivation successfully reinstated the hair-inducing capacity of 2D-cultured DPCs. Moreover, we for the first time differentiated hDFs into functional DPC-like cells through APA microencapsulation. The hDFs in APA microcapsules exhibited the hDPC features on the molecular level. We further elucidated that the hDFs into hDF-DPCs in APA microcapsules was activated by Wnt signaling pathways. It is worth to note that this approach is not limited to hDFs,

Data availability

Data supporting the findings of this study are within this manuscript or available from the corresponding authors upon reasonable request.

CRediT authorship contribution statement

Bei Xie: Conceptualization, Investigation, Formal analysis, Writing - original draft. Mengting Chen: Investigation, Validation, Writing - original draft. Pinghui Ding: Investigation, Validation, Formal analysis. Lei Lei: Funding acquisition, Writing - review & editing. Xing Zhang: Writing - review & editing. Dan Zhu: Methodology. Yujian Zou: Methodology. Zhili Deng: . Guoming Sun: Writing - review & editing. Ji Li: Methodology, Funding acquisition, Project administration, Supervision. Hemin Nie:

Declaration of Competing Interest

The authors declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper.

Acknowledgments

This work was supported by the National Natural Science Foundation of China (Nos. 31670997, 81773351, 81900982), Hunan Provincial Science and Technology Department (Nos. 2018SK2106, 2017JJ3421), and Changsha Municipal Bureau of Science and Technology (No. kq1901024). The authors declare no competing interest associated with this publication.

References (60)

  • V.A. Botchkarev et al.

    Molecular control of epithelial-mesenchymal interactions during hair follicle cycling

    J. Invest. Derm. Symp. P.

    (2003)
  • S. Reddy et al.

    Characterization of Wnt gene expression in developing and postnatal hair follicles and identification of Wnt5a as a target of Sonic hedgehog in hair follicle morphogenesis

    Mech. Develop.

    (2001)
  • C.H. Chang et al.

    Distinct Wnt members regulate the hierarchical morphogenesis of skin regions (spinal tract) and individual feathers

    Mech. Develop.

    (2004)
  • S.E. Millar

    Molecular mechanisms regulating hair follicle development

    J. Invest. Dermatol.

    (2002)
  • R. Sennett et al.

    Mesenchymal-epithelial interactions during hair follicle morphogenesis and cycling

    Semin. Cell Dev. Biol.

    (2012)
  • Y. Ito et al.

    Isolation of murine hair-inducing cells using the cell surface marker prominin-1/CD133

    J. Invest. Dermatol.

    (2007)
  • P. Rompolas et al.

    Stem cell dynamics in the hair follicle niche

    Semin. Cell Dev. Biol.

    (2014)
  • Y.C. Huang et al.

    Scalable production of controllable dermal papilla spheroids on PVA surfaces and the effects of spheroid size on hair follicle regeneration

    Biomaterials

    (2013)
  • S.M. Fan et al.

    Inducing hair follicle neogenesis with secreted proteins enriched in embryonic skin

    Biomaterials

    (2018)
  • T.H. Young et al.

    Self-assembly of dermal papilla cells into inductive spheroidal microtissues on poly(ethylene-co-vinyl alcohol) membranes for hair follicle regeneration

    Biomaterials

    (2008)
  • G. Su et al.

    Direct conversion of fibroblasts into neural progenitor-like cells by forced growth into 3D spheres on low attachment surfaces

    Biomaterials

    (2013)
  • H.Y. Yeh et al.

    The calcium-dependent regulation of spheroid formation and cardiomyogenic differentiation for MSCs on chitosan membranes

    Biomaterials

    (2012)
  • T. Andl et al.

    WNT signals are required for the initiation of hair follicle development

    Dev. Cell

    (2002)
  • Y. Yang et al.

    Versican gene: regulation by the β-catenin signaling pathway plays a significant role in dermal papilla cell aggregative growth

    J. Dermatol. Sci.

    (2012)
  • S.C. Chueh et al.

    Therapeutic strategy for hair regeneration: hair cycle activation, niche environment modulation, wound-induced follicle neogenesis and stem cell engineering

    Expert Opin. Biol. Th.

    (2013)
  • C.A. Higgins et al.

    Regenerative medicine and hair loss: how hair follicle culture has advanced our understanding of treatment options for androgenetic alopecia

    Regen. Med.

    (2014)
  • T.F. Cash

    The psychosocial consequences of androgenetic alopecia: a review of the research literature

    Brit. J. Dermatol.

    (1999)
  • J. Shapiro

    Current treatment of alopecia areata

    J. Invest. Derm. Symp. P.

    (2013)
  • D. Nanba et al.

    Establishment of cadherin-based intercellular junctions in the dermal papilla of the developing hair follicle

    Anat. Rec.

    (2003)
  • M. Ohyama et al.

    The mesenchymal component of hair follicle neogenesis: background, methods and molecular characterization

    Exp. Dermatol.

    (2010)
  • 1

    These authors contributed equally to this work.

    View full text