Abstract
Adult stem cells and their niches communicate intricately for tissue maintenance and regeneration. However, effectively coordinating these complex interactions is challenging. Here, we demonstrate that transient dedifferentiation of a fraction of epithelial stem cell progenies orchestrates beneficial changes within the entire skin’s cellular networks to favor repair. We achieved this by inducing a mosaic and reversible expression of reprogramming factors (Oct-4, Sox2, Klf4, and c-Myc) in the mouse epidermis. This in vivo partial epidermal reprogramming not only affected the partially reprogrammed cells, but also their microenvironment, including neighboring epithelial cells and T cells, conferring widespread healing characteristics even in the absence of injury. When a wound was introduced, these collective changes accelerated re-epithelialization in both wild-type and a hyperglycemic mouse disease model. Furthermore, the effects extended to dermal healing, leading to reduced scarring and altered angiogenesis. In conclusion, our work reveals that mosaic partial reprogramming of the epidermis influences various cell types within the skin during homeostasis and repair, leading to enhanced cutaneous wound healing.
Data availability
All scRNA-seq data are deposited in the SRA database and are publicly available under accession code PRJNA1139235. All other data supporting the findings of this study are provided in the Source data. Source data are provided with this paper.
Code availability
All original codes generated during scRNA-seq analysis are available at the following GitHub repository: https://github.com/CB-postech/scRNA-seq_mouse_Epi-iOSKM.
References
Arwert, E. N., Hoste, E. & Watt, F. M. Epithelial stem cells, wound healing and cancer. Nat. Rev. Cancer 12, 170–180 (2012).
Gurtner, G. C., Werner, S., Barrandon, Y. & Longaker, M. T. Wound repair and regeneration. Nature 453, 314–321 (2008).
Kumar, A., Godwin, J. W., Gates, P. B., Garza-Garcia, A. A. & Brockes, J. P. Molecular basis for the nerve dependence of limb regeneration in an adult vertebrate. Science 318, 772–777 (2007).
Martin, P. Wound healing-aiming for perfect skin regeneration. Science 276, 75–81 (1997).
Plikus, M. V. et al. Epithelial stem cells and implications for wound repair. Semin. Cell Dev. Biol. 23, 946–953 (2012).
Aragona, M. et al. Defining stem cell dynamics and migration during wound healing in mouse skin epidermis. Nat. Commun. 8, 14684 (2017).
Saxena, S., Vekaria, H., Sullivan, P. G. & Seifert, A. W. Connective tissue fibroblasts from highly regenerative mammals are refractory to ROS-induced cellular senescence. Nat. Commun. 10, 4400 (2019).
Mesa, K. R., Rompolas, P. & Greco, V. The Dynamic Duo: Niche/Stem Cell Interdependency. Stem Cell Rep. 4, 961–966 (2015).
Xin, T., Greco, V. & Myung, P. Hardwiring stem cell communication through tissue structure. Cell 164, 1212–1225 (2016).
Park, S. et al. Tissue-scale coordination of cellular behaviour promotes epidermal wound repair in live mice. Nat. Cell Biol. 19, 155–163 (2017).
Xue, M. & Jackson, C. J. Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv. Wound Care 4, 119–136 (2015).
Talbott, H. E., Mascharak, S., Griffin, M., Wan, D. C. & Longaker, M. T. Wound healing, fibroblast heterogeneity, and fibrosis. Cell Stem Cell 29, 1161–1180 (2022).
Mack, K. L. et al. Allele-specific expression reveals genetic drivers of tissue regeneration in mice. Cell Stem Cell 30, 1368–1381.e1366 (2023).
Ring, N. A. R. et al. The p-rpS6-zone delineates wounding responses and the healing process. Dev. Cell 58, 981–992 e986 (2023).
Doeser, M. C., Scholer, H. R. & Wu, G. Reduction of fibrosis and scar formation by partial reprogramming in vivo. Stem Cells 36, 1216–1225 (2018).
Reynolds, L. E. et al. Accelerated re-epithelialization in beta3-integrin-deficient- mice is associated with enhanced TGF-beta1 signaling. Nat. Med. 11, 167–174 (2005).
Takahashi, K. & Yamanaka, S. Induction of pluripotent stem cells from mouse embryonic and adult fibroblast cultures by defined factors. Cell 126, 663–676 (2006).
Rais, Y. et al. Deterministic direct reprogramming of somatic cells to pluripotency. Nature 502, 65–70 (2013).
Maza, I. et al. Transient acquisition of pluripotency during somatic cell transdifferentiation with iPSC reprogramming factors. Nat. Biotechnol. 33, 769–774 (2015).
Abad, M. et al. Reprogramming in vivo produces teratomas and iPS cells with totipotency features. Nature 502, 340–345 (2013).
Ohnishi, K. et al. Premature termination of reprogramming in vivo leads to cancer development through altered epigenetic regulation. Cell 156, 663–677 (2014).
Ocampo, A. et al. In vivo amelioration of age-associated hallmarks by partial reprogramming. Cell 167, 1719–1733.e1712 (2016).
Browder, K. C. et al. In vivo partial reprogramming alters age-associated molecular changes during physiological aging in mice. Nat. Aging 2, 243–253 (2022).
Ha, J. et al. Intermediate cells of in vitro cellular reprogramming and in vivo tissue regeneration require desmoplakin. Sci. Adv. 8, eabk1239 (2022).
Yang, J. H. et al. Loss of epigenetic information as a cause of mammalian aging. Cell 186, 305–326.e327 (2023).
Rodriguez-Matellan, A., Alcazar, N., Hernandez, F., Serrano, M. & Avila, J. In vivo reprogramming ameliorates aging features in dentate gyrus cells and improves memory in mice. Stem Cell Rep. 15, 1056–1066 (2020).
Chondronasiou, D. et al. Multi-omic rejuvenation of naturally aged tissues by a single cycle of transient reprogramming. Aging Cell 21, e13578 (2022).
Wang, C. et al. In vivo partial reprogramming of myofibers promotes muscle regeneration by remodeling the stem cell niche. Nat. Commun. 12, 3094 (2021).
Hishida, T. et al. In vivo partial cellular reprogramming enhances liver plasticity and regeneration. Cell Rep. 39, 110730 (2022).
Kim, J. et al. Partial in vivo reprogramming enables injury-free intestinal regeneration via autonomous Ptgs1 induction. Sci. Adv. 9, eadi8454 (2023).
Lu, Y. et al. Reprogramming to recover youthful epigenetic information and restore vision. Nature 588, 124–129 (2020).
Chen, Y. et al. Reversible reprogramming of cardiomyocytes to a fetal state drives heart regeneration in mice. Science 373, 1537–1540 (2021).
Elbaz, J. et al. Highly efficient reprogrammable mouse lines with integrated reporters to track the route to pluripotency. Proc. Natl. Acad. Sci. USA 119, e2207824119 (2022).
Ichijo, R. et al. Tbx3-dependent amplifying stem cell progeny drives interfollicular epidermal expansion during pregnancy and regeneration. Nat. Commun. 8, 508 (2017).
Zhang, Y. V., White, B. S., Shalloway, D. I. & Tumbar, T. Stem cell dynamics in mouse hair follicles: a story from cell division counting and single cell lineage tracing. Cell Cycle 9, 1504–1510 (2010).
Mascre, G. et al. Distinct contribution of stem and progenitor cells to epidermal maintenance. Nature 489, 257–262 (2012).
Wang, S. et al. Single cell transcriptomics of human epidermis identifies basal stem cell transition states. Nat. Commun. 11, 4239 (2020).
Cao, X. et al. RIPK4 promotes epidermal differentiation through phase separation and activation of LATS1/2. Dev. Cell 60, 2761–2776.e2711 (2025).
Haensel, D. et al. Defining epidermal basal cell states during skin homeostasis and wound healing using single-cell transcriptomics. Cell Rep. 30, 3932–3947 e3936 (2020).
Zhulyn, O. et al. Evolutionarily divergent mTOR remodels translatome for tissue regeneration. Nature 620, 163–171 (2023).
Greenhalgh, D. G. The role of apoptosis in wound healing. Int. J. Biochem. Cell Biol. 30, 1019–1030 (1998).
Paladini, R. D., Takahashi, K., Bravo, N. S. & Coulombe, P. A. Onset of re-epithelialization after skin injury correlates with a reorganization of keratin filaments in wound edge keratinocytes: defining a potential role for keratin 16. J. Cell Biol. 132, 381–397 (1996).
Lim, C., Lim, J. & Choi, S. Wound-induced hair follicle neogenesis as a promising approach for hair regeneration. Mol. Cells 46, 573–578 (2023).
Hong, W. X. et al. The role of hypoxia-inducible factor in wound healing. Adv. Wound Care 3, 390–399 (2014).
Liu, S. et al. A tissue injury sensing and repair pathway distinct from host pathogen defense. Cell 186, 2127–2143.e2122 (2023).
Koh, T. J. & DiPietro, L. A. Inflammation and wound healing: the role of the macrophage. Expert Rev. Mol. Med. 13, e23 (2011).
Keyes, B. E. et al. Impaired epidermal to dendritic T cell signaling slows wound repair in aged skin. Cell 167, 1323–1338.e1314 (2016).
Lin, Y. et al. PIK3R1 negatively regulates the epithelial-mesenchymal transition and stem-like phenotype of renal cancer cells through the AKT/GSK3beta/CTNNB1 signaling pathway. Sci. Rep. 5, 8997 (2015).
Liu, Y. et al. Pan-cancer analysis on the role of PIK3R1 and PIK3R2 in human tumors. Sci. Rep. 12, 5924 (2022).
Welsh, N., Makeeva, N. & Welsh, M. Overexpression of the Shb SH2 domain-protein in insulin-producing cells leads to altered signaling through the IRS-1 and IRS-2 proteins. Mol. Med. 8, 695–704 (2002).
Holmqvist, K. et al. The adaptor protein shb binds to tyrosine 1175 in vascular endothelial growth factor (VEGF) receptor-2 and regulates VEGF-dependent cellular migration. J. Biol. Chem. 279, 22267–22275 (2004).
Schubert, M. et al. Perturbation-response genes reveal signaling footprints in cancer gene expression. Nat. Commun. 9, 20 (2018).
Ersahin, T., Tuncbag, N. & Cetin-Atalay, R. The PI3K/AKT/mTOR interactive pathway. Mol. Biosyst. 11, 1946–1954 (2015).
Zhang, X., Tang, N., Hadden, T. J. & Rishi, A. K. Akt, FoxO and regulation of apoptosis. Biochim Biophys. Acta 1813, 1978–1986 (2011).
Hannenhalli, S. & Kaestner, K. H. The evolution of Fox genes and their role in development and disease. Nat. Rev. Genet. 10, 233–240 (2009).
Gharbi, S. I. et al. Exploring the specificity of the PI3K family inhibitor LY294002. Biochem. J. 404, 15–21 (2007).
Agarwal, S., Mirzoeva, S., Readhead, B., Dudley, J. T. & Budunova, I. PI3K inhibitors protect against glucocorticoid-induced skin atrophy. EBioMedicine 41, 526–537 (2019).
Squarize, C. H., Castilho, R. M., Bugge, T. H. & Gutkind, J. S. Accelerated wound healing by mTOR activation in genetically defined mouse models. PLoS ONE 5, e10643 (2010).
van Lengerich, B., Agnew, C., Puchner, E. M., Huang, B. & Jura, N. EGF and NRG induce phosphorylation of HER3/ERBB3 by EGFR using distinct oligomeric mechanisms. Proc. Natl. Acad. Sci. USA 114, E2836–E2845 (2017).
El-Rayes, B. F. & LoRusso, P. M. Targeting the epidermal growth factor receptor. Br. J. Cancer 91, 418–424 (2004).
Barrientos, S., Stojadinovic, O., Golinko, M. S., Brem, H. & Tomic-Canic, M. Growth factors and cytokines in wound healing. Wound Repair Regen. 16, 585–601 (2008).
Gallini, S. et al. Injury prevents Ras mutant cell expansion in mosaic skin. Nature 619, 167–175 (2023).
Gibbs, S. et al. Epidermal growth factor and keratinocyte growth factor differentially regulate epidermal migration, growth, and differentiation. Wound Repair Regen. 8, 192–203 (2000).
Zhong, H. et al. Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics. Cancer Res. 60, 1541–1545 (2000).
Peng, X. H. et al. Cross-talk between epidermal growth factor receptor and hypoxia-inducible factor-1alpha signal pathways increases resistance to apoptosis by up-regulating survivin gene expression. J. Biol. Chem. 281, 25903–25914 (2006).
Ma, S., Rao, L., Freedberg, I. M. & Blumenberg, M. Transcriptional control of K5, K6, K14, and K17 keratin genes by AP-1 and NF-kappaB family members. Gene Expr. 6, 361–370 (1997).
Chung, B. M., Murray, C. I., Van Eyk, J. E. & Coulombe, P. A. Identification of novel interaction between annexin A2 and keratin 17: evidence for reciprocal regulation. J. Biol. Chem. 287, 7573–7581 (2012).
Fuchs, E. & Raghavan, S. Getting under the skin of epidermal morphogenesis. Nat. Rev. Genet. 3, 199–209 (2002).
Chedid, M., Rubin, J. S., Csaky, K. G. & Aaronson, S. A. Regulation of keratinocyte growth factor gene expression by interleukin 1. J. Biol. Chem. 269, 10753–10757 (1994).
MacDonald, B. T., Tamai, K. & He, X. Wnt/beta-catenin signaling: components, mechanisms, and diseases. Dev. Cell 17, 9–26 (2009).
Sigal, M. et al. Stromal R-spondin orchestrates gastric epithelial stem cells and gland homeostasis. Nature 548, 451–455 (2017).
Sunkara, R. R. et al. SFRP1 in skin tumor initiation and cancer stem cell regulation with potential implications in epithelial cancers. Stem Cell Rep. 14, 271–284 (2020).
Tumbar, T. et al. Defining the epithelial stem cell niche in skin. Science 303, 359–363 (2004).
Liu, J. et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc. Natl. Acad. Sci. USA 110, 20224–20229 (2013).
Jameson, J. et al. A role for skin gammadelta T cells in wound repair. Science 296, 747–749 (2002).
Ramirez, K., Witherden, D. A. & Havran, W. L. All hands on DE(T)C: Epithelial-resident gammadelta T cells respond to tissue injury. Cell Immunol. 296, 57–61 (2015).
Boothby, I. C., Cohen, J. N. & Rosenblum, M. D. Regulatory T cells in skin injury: at the crossroads of tolerance and tissue repair. Sci. Immunol. 5, eaaz9631 (2020).
Sharp, L. L., Jameson, J. M., Cauvi, G. & Havran, W. L. Dendritic epidermal T cells regulate skin homeostasis through local production of insulin-like growth factor 1. Nat. Immunol. 6, 73–79 (2005).
Konieczny, P. et al. Interleukin-17 governs hypoxic adaptation of injured epithelium. Science 377, eabg9302 (2022).
Witherden, D. A. et al. The CD100 receptor interacts with its plexin B2 ligand to regulate epidermal gammadelta T cell function. Immunity 37, 314–325 (2012).
Cai, Y., Fleming, C. & Yan, J. New insights of T cells in the pathogenesis of psoriasis. Cell Mol. Immunol. 9, 302–309 (2012).
Sutton, C. E., Mielke, L. A. & Mills, K. H. IL-17-producing gammadelta T cells and innate lymphoid cells. Eur. J. Immunol. 42, 2221–2231 (2012).
Islam, S. A. et al. Mouse CCL8, a CCR8 agonist, promotes atopic dermatitis by recruiting IL-5+ T(H)2 cells. Nat. Immunol. 12, 167–177 (2011).
Hedrick, M. N. et al. CCR6 is required for IL-23-induced psoriasis-like inflammation in mice. J. Clin. Investig. 119, 2317–2329 (2009).
Mabuchi, T. et al. CCR6 is required for epidermal trafficking of gammadelta-T cells in an IL-23-induced model of psoriasiform dermatitis. J. Investig. Dermatol. 133, 164–171 (2013).
Richmond, J. M., Strassner, J. P., Essien, K. I. & Harris, J. E. T-cell positioning by chemokines in autoimmune skin diseases. Immunol. Rev. 289, 186–204 (2019).
Yates, C. C. et al. Delayed reepithelialization and basement membrane regeneration after wounding in mice lacking CXCR3. Wound Repair Regen. 17, 34–41 (2009).
Sutton, C. E. et al. Interleukin-1 and IL-23 induce innate IL-17 production from gammadelta T cells, amplifying Th17 responses and autoimmunity. Immunity 31, 331–341 (2009).
Alam, M. S. et al. TNF plays a crucial role in inflammation by signaling via T cell TNFR2. Proc. Natl. Acad. Sci. USA 118, e2109972118 (2021).
Kennedy-Crispin, M. et al. Human keratinocytes’ response to injury upregulates CCL20 and other genes linking innate and adaptive immunity. J. Investig. Dermatol. 132, 105–113 (2012).
Martin, B., Hirota, K., Cua, D. J., Stockinger, B. & Veldhoen, M. Interleukin-17-producing gammadelta T cells selectively expand in response to pathogen products and environmental signals. Immunity 31, 321–330 (2009).
Mitsui, K. et al. The homeoprotein Nanog is required for maintenance of pluripotency in mouse epiblast and ES cells. Cell 113, 631–642 (2003).
Yamashiro, Y. et al. Ectopic coexpression of keratin 8 and 18 promotes invasion of transformed keratinocytes and is induced in patients with cutaneous squamous cell carcinoma. Biochem. Biophys. Res. Commun. 399, 365–372 (2010).
Lowes, M. A., Suarez-Farinas, M. & Krueger, J. G. Immunology of psoriasis. Annu. Rev. Immunol. 32, 227–255 (2014).
Depianto, D., Kerns, M. L., Dlugosz, A. A. & Coulombe, P. A. Keratin 17 promotes epithelial proliferation and tumor growth by polarizing the immune response in skin. Nat. Genet. 42, 910–914 (2010).
Subudhi, I. et al. Metabolic coordination between skin epithelium and type 17 immunity sustains chronic skin inflammation. Immunity 57, 1665–1680.e7 (2024).
Krueger, J. et al. Tofacitinib attenuates pathologic immune pathways in patients with psoriasis: a randomized phase 2 study. J. Allergy Clin. Immunol. 137, 1079–1090 (2016).
Zomer, H. D. & Trentin, A. G. Skin wound healing in humans and mice: Challenges in translational research. J. Dermatol. Sci. 90, 3–12 (2018).
Sidgwick, G. P. & Bayat, A. Extracellular matrix molecules implicated in hypertrophic and keloid scarring. J. Eur. Acad. Dermatol. Venereol. 26, 141–152 (2012).
Xie, Z. et al. Dual growth factor releasing multi-functional nanofibers for wound healing. Acta Biomater. 9, 9351–9359 (2013).
Lintel, H. et al. Transdermal deferoxamine administration improves excisional wound healing in chronically irradiated murine skin. J. Transl. Med. 20, 274 (2022).
Holm Nielsen, S. et al. Exploring the role of extracellular matrix proteins to develop biomarkers of plaque vulnerability and outcome. J. Intern. Med. 287, 493–513 (2020).
Tang, V. W. Collagen, stiffness, and adhesion: the evolutionary basis of vertebrate mechanobiology. Mol. Biol. Cell 31, 1823–1834 (2020).
Rodrigues, M., Kosaric, N., Bonham, C. A. & Gurtner, G. C. Wound healing: a cellular perspective. Physiol. Rev. 99, 665–706 (2019).
Botusan, I. R. et al. Stabilization of HIF-1alpha is critical to improve wound healing in diabetic mice. Proc. Natl. Acad. Sci. USA 105, 19426–19431 (2008).
Xu, J., Liu, X., Zhao, F., Zhang, Y. & Wang, Z. HIF1alpha overexpression enhances diabetic wound closure in high glucose and low oxygen conditions by promoting adipose-derived stem cell paracrine function and survival. Stem Cell Res. Ther. 11, 148 (2020).
Li, G. et al. A small molecule HIF-1alpha stabilizer that accelerates diabetic wound healing. Nat. Commun. 12, 3363 (2021).
DiPietro, L. A. Angiogenesis and scar formation in healing wounds. Curr. Opin. Rheumatol. 25, 87–91 (2013).
DiPietro, L. A. Angiogenesis and wound repair: when enough is enough. J. Leukoc. Biol. 100, 979–984 (2016).
Wilgus, T. A., Ferreira, A. M., Oberyszyn, T. M., Bergdall, V. K. & Dipietro, L. A. Regulation of scar formation by vascular endothelial growth factor. Lab. Investig. 88, 579–590 (2008).
Diao, J. S., Xia, W. S. & Guo, S. Z. Bevacizumab: a potential agent for prevention and treatment of hypertrophic scar. Burns 36, 1136–1137 (2010).
Ma, J. et al. Single-cell RNA-Seq analysis of diabetic wound macrophages in STZ-induced mice. J. Cell Commun. Signal 17, 103–120 (2023).
Avitabile, S. et al. Interleukin-22 promotes wound repair in diabetes by improving keratinocyte pro-healing functions. J. Investig. Dermatol. 135, 2862–2870 (2015).
Sun, Y., Song, L., Zhang, Y., Wang, H. & Dong, X. Adipose stem cells from type 2 diabetic mice exhibit therapeutic potential in wound healing. Stem Cell Res. Ther. 11, 298 (2020).
Okizaki, S. et al. Vascular endothelial growth factor receptor type 1 signaling prevents delayed wound healing in diabetes by attenuating the production of IL-1beta by recruited macrophages. Am. J. Pathol. 186, 1481–1498 (2016).
Saidak, Z. et al. Wnt/beta-catenin signaling mediates osteoblast differentiation triggered by peptide-induced alpha5beta1 integrin priming in mesenchymal skeletal cells. J. Biol. Chem. 290, 6903–6912 (2015).
Mirotsou, M. et al. Secreted frizzled related protein 2 (Sfrp2) is the key Akt-mesenchymal stem cell-released paracrine factor mediating myocardial survival and repair. Proc. Natl. Acad. Sci. USA 104, 1643–1648 (2007).
Kasashima, H. et al. Stromal SOX2 upregulation promotes tumorigenesis through the generation of a SFRP1/2-expressing cancer-associated fibroblast population. Dev. Cell 56, 95–110.e110 (2021).
Sharma, M., Chuang, W. W. & Sun, Z. Phosphatidylinositol 3-kinase/Akt stimulates androgen pathway through GSK3beta inhibition and nuclear beta-catenin accumulation. J. Biol. Chem. 277, 30935–30941 (2002).
He, Y. et al. Targeting PI3K/Akt signal transduction for cancer therapy. Signal Transduct. Target Ther. 6, 425 (2021).
Pugh, C. W. & Ratcliffe, P. J. Regulation of angiogenesis by hypoxia: role of the HIF system. Nat. Med 9, 677–684 (2003).
Forsythe, J. A. et al. Activation of vascular endothelial growth factor gene transcription by hypoxia-inducible factor 1. Mol. Cell Biol. 16, 4604–4613 (1996).
Vasioukhin, V., Degenstein, L., Wise, B. & Fuchs, E. The magical touch: genome targeting in epidermal stem cells induced by tamoxifen application to mouse skin. Proc. Natl. Acad. Sci. USA 96, 8551–8556 (1999).
Lee, D., Lim, J., Woo, K. C. & Kim, K. T. Piperonylic acid stimulates keratinocyte growth and survival by activating epidermal growth factor receptor (EGFR). Sci. Rep. 8, 162 (2018).
Landini, G., Martinelli, G. & Piccinini, F. Colour deconvolution: stain unmixing in histological imaging. Bioinformatics 37, 1485–1487 (2021).
Mascharak, S. et al. Preventing engrailed-1 activation in fibroblasts yields wound regeneration without scarring. Science 372, eaba2374 (2021).
Mascharak, S. et al. Multi-omic analysis reveals divergent molecular events in scarring and regenerative wound healing. Cell Stem Cell 29, 315–327.e316 (2022).
Ye, L., Wang, Z., Li, Z., Lv, C. & Man, M. Q. Validation of GPSkin Barrier((R)) for assessing epidermal permeability barrier function and stratum corneum hydration in humans. Ski. Res. Technol. 25, 25–29 (2019).
Deeds, M. C. et al. Single dose streptozotocin-induced diabetes: considerations for study design in islet transplantation models. Lab. Anim. 45, 131–140 (2011).
Le May, C. et al. Estrogens protect pancreatic beta-cells from apoptosis and prevent insulin-deficient diabetes mellitus in mice. Proc. Natl. Acad. Sci. USA 103, 9232–9237 (2006).
Graham, M. L., Janecek, J. L., Kittredge, J. A., Hering, B. J. & Schuurman, H. J. The streptozotocin-induced diabetic nude mouse model: differences between animals from different sources. Comp. Med. 61, 356–360 (2011).
Lee, D. et al. Diabetic sensory neuropathy and insulin resistance are induced by loss of UCHL1 in Drosophila. Nat. Commun. 15, 468 (2024).
McCarthy, D. J., Campbell, K. R., Lun, A. T. & Wills, Q. F. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 33, 1179–1186 (2017).
Lun, A. T., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Res 5, 2122 (2016).
Hao, Y. et al. Dictionary learning for integrative, multimodal and scalable single-cell analysis. Nat. Biotechnol. 42, 293–304 (2024).
Liberzon, A. et al. Molecular signatures database (MSigDB) 3.0. Bioinformatics 27, 1739–1740 (2011).
Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).
La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).
Setty, M. et al. Characterization of cell fate probabilities in single-cell data with Palantir. Nat. Biotechnol. 37, 451–460 (2019).
Kang, M. et al. Improved reconstruction of single-cell developmental potential with CytoTRACE 2. Nat. Methods 22, 2258–2263 (2025).
Jin, S. et al. Inference and analysis of cell-cell communication using CellChat. Nat. Commun. 12, 1088 (2021).
Kolberg, L. et al. g:Profiler-interoperable web service for functional enrichment analysis and gene identifier mapping (2023 update). Nucleic Acids Res. 51, W207–W212 (2023).
Szklarczyk, D. et al. The STRING database in 2023: protein-protein association networks and functional enrichment analyses for any sequenced genome of interest. Nucleic Acids Res. 51, D638–D646 (2023).
Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e1821 (2019).
Acknowledgements
We thank the members of Choi Laboratory for their helpful discussions and comments on this manuscript. This work was supported by National Research Foundation of Korea (NRF) grants funded by the Korean government (MSIT) (NRF-RS-2025-00560319, NRF-RS-2022-NR072134, NRF-RS-2022-NR067309, NRF-RS-2023-00223298, and NRF-RS-2024-00433755 to S.C.; NRF-RS-2024-00426031 to J.K.K.; NRF-RS-2023-00221112 to J.K.K. and S.C.). S.C. has been supported by a POSCO Science Fellowship from the POSCO TJ Park Foundation. M.K., E.C., Y.J., B.K., C.L., J.L., J.K.K. and S.C. were supported by a BK21 FOUR Research Fellowship funded by the Ministry of Education, Republic of Korea.
Author information
Authors and Affiliations
Contributions
S.C., M.K., and Y.J. conceived the project. M.K., Y.J., and B.K. performed most of the experiments and quantified and analyzed the data. E.C. and J.K.K. performed the bioinformatic analysis. Y.H.C. generated RNA-seq libraries. C.L. performed flow cytometry. J.L. performed splinted full-thickness cutaneous wounding. J.H.L, K.-P.K., and B.-K.K. provided intellectual input. J.K.K. and S.C. acquired funding. M.K., Y.J., J.K.K., and S.C. wrote the manuscript, with discussion and feedback from all co-authors.
Corresponding authors
Ethics declarations
Competing interests
The authors declare no competing interests.
Peer review
Peer review information
Nature Communications thanks the anonymous reviewer(s) for their contribution to the peer review of this work. A peer review file is available.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Source data
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, 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 changes were made. 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/4.0/.
About this article
Cite this article
Kwak, M., Choi, E., Jo, Y. et al. Mosaic partial epidermal reprogramming remodels neighbors and niches to refine skin homeostasis and repair. Nat Commun (2026). https://doi.org/10.1038/s41467-026-69047-2
Received:
Accepted:
Published:
DOI: https://doi.org/10.1038/s41467-026-69047-2