New Anti-Fibrotic Strategies for Keloids: Insights From Single-Cell Multi-Omics

Songyun Zhao , Jiaheng Xie , Qian Zhang , Tianyi Ni , Jinde Lin , Weicheng Gao , Liping Zhao , Min Yi , Liying Tu , Pengpeng Zhang , Dan Wu , Qikai Tang , Chenfeng Ma , Yucang He , Liqun Li , Guoping Wu , Wei Yan

Cell Proliferation ›› 2025, Vol. 58 ›› Issue (6) : e13818

PDF
Cell Proliferation ›› 2025, Vol. 58 ›› Issue (6) : e13818 DOI: 10.1111/cpr.13818
ORIGINAL ARTICLE

New Anti-Fibrotic Strategies for Keloids: Insights From Single-Cell Multi-Omics

Author information +
History +
PDF

Abstract

Keloids are complex pathological skin scars characterised by excessive growth of fibrous tissue and abnormal accumulation of extracellular matrix (ECM). Despite various treatment options available, the treatment of keloids remains a major clinical challenge due to high recurrence rates and inconsistent therapeutic outcomes. By collecting three keloid tissues and three normal skin samples and utilising single-cell RNA sequencing (scRNA-seq), we delved into the cellular heterogeneity and molecular mechanisms of keloids. Our study identified multiple fibroblast subpopulations within keloid tissue. Enrichment and cell–cell communication analyses revealed that POSTN-positive mesenchymal fibroblasts (POSTN+ mesenchymal fibs) are more prevalent in keloids and exhibit higher transforming growth factor β (TGF-β) signalling activity, potentially playing a central role in excessive fibrosis. In contrast, IGFBP2-positive fibroblasts (IGFBP2+ fibs) are more abundant in normal skin, insensitive to TGF-β and Periostin signalling, and possess anti-fibrotic potential, possibly related to limited tissue repair and regenerative capacity. Trajectory analysis inferred the differentiation states and patterns of different fibroblast subpopulations. Additionally, we explored the heterogeneity of endothelial cells, finding an endothelial cell subpopulation (EC10) exhibiting mesenchymal activation characteristics, which may work with specific fibroblasts to promote abnormal angiogenesis and endothelial-to-mesenchymal transition processes. Spatial transcriptomics analysis has shown that the proportion of IGFBP2+ fibroblasts relatively increases in acne keloidalis after hormonal treatment, further demonstrating their value as potential therapeutic targets. Ultimately, we separated these two subpopulations using flow cytometry, highlighting their opposing roles in the secretion of the ECM. These findings provide new insights into the pathogenesis of keloids and lay the theoretical foundation for the development of innovative anti-fibrotic treatment strategies.

Keywords

anti-fibrotic therapy / fibroblasts / keloid / scRNA-seq / spatial transcriptomics / TGF-β

Cite this article

Download citation ▾
Songyun Zhao, Jiaheng Xie, Qian Zhang, Tianyi Ni, Jinde Lin, Weicheng Gao, Liping Zhao, Min Yi, Liying Tu, Pengpeng Zhang, Dan Wu, Qikai Tang, Chenfeng Ma, Yucang He, Liqun Li, Guoping Wu, Wei Yan. New Anti-Fibrotic Strategies for Keloids: Insights From Single-Cell Multi-Omics. Cell Proliferation, 2025, 58(6): e13818 DOI:10.1111/cpr.13818

登录浏览全文

4963

注册一个新账户 忘记密码

References

[1]

J. P. Andrews, J. Marttala, E. Macarak, J. Rosenbloom, and J. Uitto, “Keloids: The Paradigm of Skin Fibrosis—Pathomechanisms and Treatment,” Matrix Biology 51 (2016): 37-46, https://doi.org/10.1016/j.matbio.2016.01.013.

[2]

S. A. Davis, S. R. Feldman, and A. J. McMichael, “Management of Keloids in the United States, 1990-2009: An Analysis of the National Ambulatory Medical Care Survey,” Dermatologic Surgery 39, no. 7 (2013): 988-994, https://doi.org/10.1111/dsu.12182.

[3]

C. Y. Ung, A. Warwick, A. Onoufriadis, et al., “Comorbidities of Keloid and Hypertrophic Scars Among Participants in UK Biobank,” JAMA Dermatology 159, no. 2 (2023): 172-181, https://doi.org/10.1001/jamadermatol.2022.5607.

[4]

A. King, M. Guirguis, S. Satkunanathan, M. Saad, and R. Bose, “Intralesional 5-Fluorouracil for Keloids: A Systematic Review,” Journal of Cutaneous Medicine and Surgery 28, no. 4 (2024): 381-386, https://doi.org/10.1177/12034754241256346.

[5]

P. Martin and R. Nunan, “Cellular and Molecular Mechanisms of Repair in Acute and Chronic Wound Healing,” British Journal of Dermatology 173, no. 2 (2015): 370-378, https://doi.org/10.1111/bjd.13954.

[6]

Y. Bao, S. Xu, Z. Pan, et al., “Comparative Efficacy and Safety of Common Therapies in Keloids and Hypertrophic Scars: A Systematic Review and Meta-Analysis,” Aesthetic Plastic Surgery 44, no. 1 (2020): 207-218, https://doi.org/10.1007/s00266-019-01518-y.

[7]

M. Fernandez-Guarino, S. Bacci, L. A. Perez Gonzalez, M. Bermejo-Martinez, A. Cecilia-Matilla, and M. L. Hernandez-Bule, “The Role of Physical Therapies in Wound Healing and Assisted Scarring,” International Journal of Molecular Sciences 24, no. 8 (2023): 7487, https://doi.org/10.3390/ijms24087487.

[8]

C. C. Lee, C. H. Tsai, C. H. Chen, Y. C. Yeh, W. H. Chung, and C. B. Chen, “An Updated Review of the Immunological Mechanisms of Keloid Scars,” Frontiers in Immunology 14 (2023): 1117630, https://doi.org/10.3389/fimmu.2023.1117630.

[9]

M. V. Plikus, X. Wang, S. Sinha, et al., “Fibroblasts: Origins, Definitions, and Functions in Health and Disease,” Cell 184, no. 15 (2021): 3852-3872, https://doi.org/10.1016/j.cell.2021.06.024.

[10]

T. A. Wynn and T. R. Ramalingam, “Mechanisms of Fibrosis: Therapeutic Translation for Fibrotic Disease,” Nature Medicine 18, no. 7 (2012): 1028-1040, https://doi.org/10.1038/nm.2807.

[11]

M. Zhang, H. Chen, H. Qian, and C. Wang, “Characterization of the Skin Keloid Microenvironment,” Cell Communication and Signaling: CCS 21, no. 1 (2023): 207, https://doi.org/10.1186/s12964-023-01214-0.

[12]

Y. Li, M. Li, C. Qu, et al., “The Polygenic Map of Keloid Fibroblasts Reveals Fibrosis-Associated Gene Alterations in Inflammation and Immune Responses,” Frontiers in Immunology 12 (2021): 810290, https://doi.org/10.3389/fimmu.2021.810290.

[13]

N. N. Do and S. A. Eming, “Skin Fibrosis: Models and Mechanisms,” Current Research in Translational Medicine 64, no. 4 (2016): 185-193, https://doi.org/10.1016/j.retram.2016.06.003.

[14]

Y. Rinkevich, G. G. Walmsley, M. S. Hu, et al., “Skin Fibrosis. Identification and Isolation of a Dermal Lineage With Intrinsic Fibrogenic Potential,” Science 348, no. 6232 (2015): aaa2151, https://doi.org/10.1126/science.aaa2151.

[15]

Y. Xia, Y. Wang, Y. Hao, et al., “Deciphering the Single-Cell Transcriptome Network in Keloids With Intra-Lesional Injection of Triamcinolone Acetonide Combined With 5-Fluorouracil,” Frontiers in Immunology 14 (2023): 1106289, https://doi.org/10.3389/fimmu.2023.1106289.

[16]

C. Feng, M. Shan, Y. Xia, et al., “Single-Cell RNA Sequencing Reveals Distinct Immunology Profiles in Human Keloid,” Frontiers in Immunology 13 (2022): 940645, https://doi.org/10.3389/fimmu.2022.940645.

[17]

L. Sole-Boldo, G. Raddatz, S. Schutz, et al., “Single-Cell Transcriptomes of the Human Skin Reveal Age-Related Loss of Fibroblast Priming,” Communications Biology 3, no. 1 (2020): 188, https://doi.org/10.1038/s42003-020-0922-4.

[18]

C. Kuppe, M. M. Ibrahim, J. Kranz, et al., “Decoding Myofibroblast Origins in Human Kidney Fibrosis,” Nature 589, no. 7841 (2021): 281-286, https://doi.org/10.1038/s41586-020-2941-1.

[19]

J. Shim, S. J. Oh, E. Yeo, et al., “Integrated Analysis of Single-Cell and Spatial Transcriptomics in Keloids: Highlights on Fibrovascular Interactions in Keloid Pathogenesis,” Journal of Investigative Dermatology 142, no. 8 (2022): 2128-2139 e11, https://doi.org/10.1016/j.jid.2022.01.017.

[20]

C. C. Deng, Y. F. Hu, D. H. Zhu, et al., “Single-Cell Rna-Seq Reveals Fibroblast Heterogeneity and Increased Mesenchymal Fibroblasts in Human Fibrotic Skin Diseases,” Nature Communications 12, no. 1 (2021): 3709, https://doi.org/10.1038/s41467-021-24110-y.

[21]

Y. Hao, S. Hao, E. Andersen-Nissen, et al., “Integrated Analysis of Multimodal Single-Cell Data,” Cell 184, no. 13 (2021): 3573-3587 e29, https://doi.org/10.1016/j.cell.2021.04.048.

[22]

Y. K. Hong, D. Y. Hwang, C. C. Yang, et al., “Profibrotic Subsets of Spp1(+) Macrophages and Postn(+) Fibroblasts Contribute to Fibrotic Scarring in Acne Keloidalis,” Journal of Investigative Dermatology 144, no. 7 (2024): 1491-1504 e10, https://doi.org/10.1016/j.jid.2023.12.014.

[23]

D. M. Cable, E. Murray, L. S. Zou, et al., “Robust Decomposition of Cell Type Mixtures in Spatial Transcriptomics,” Nature Biotechnology 40, no. 4 (2022): 517-526, https://doi.org/10.1038/s41587-021-00830-w.

[24]

J. Cao, M. Spielmann, X. Qiu, et al., “The Single-Cell Transcriptional Landscape of Mammalian Organogenesis,” Nature 566, no. 7745 (2019): 496-502, https://doi.org/10.1038/s41586-019-0969-x.

[25]

G. S. Gulati, S. S. Sikandar, D. J. Wesche, et al., “Single-Cell Transcriptional Diversity Is a Hallmark of Developmental Potential,” Science 367, no. 6476 (2020): 405-411, https://doi.org/10.1126/science.aax0249.

[26]

T. Wu, E. Hu, S. Xu, et al., “Clusterprofiler 4.0: A Universal Enrichment Tool for Interpreting Omics Data,” Innovations 2, no. 3 (2021): 100141, https://doi.org/10.1016/j.xinn.2021.100141.

[27]

Y. Wu, S. Yang, J. Ma, et al., “Spatiotemporal Immune Landscape of Colorectal Cancer Liver Metastasis at Single-Cell Level,” Cancer Discovery 12, no. 1 (2022): 134-153, https://doi.org/10.1158/2159-8290.CD-21-0316.

[28]

S. Aibar, C. B. Gonzalez-Blas, T. Moerman, et al., “Scenic: Single-Cell Regulatory Network Inference and Clustering,” Nature Methods 14, no. 11 (2017): 1083-1086, https://doi.org/10.1038/nmeth.4463.

[29]

X. Qiu, Q. Mao, Y. Tang, et al., “Reversed Graph Embedding Resolves Complex Single-Cell Trajectories,” Nature Methods 14, no. 10 (2017): 979-982, https://doi.org/10.1038/nmeth.4402.

[30]

X. Liu, W. Chen, Q. Zeng, et al., “Single-Cell Rna-Sequencing Reveals Lineage-Specific Regulatory Changes of Fibroblasts and Vascular Endothelial Cells in Keloids,” Journal of Investigative Dermatology 142, no. 1 (2022): 124-135 e11, https://doi.org/10.1016/j.jid.2021.06.010.

[31]

Y. Gao, H. Wang, S. Chen, et al., “Single-Cell N(6)-Methyladenosine Regulator Patterns Guide Intercellular Communication of Tumor Microenvironment That Contribute to Colorectal Cancer Progression and Immunotherapy,” Journal of Translational Medicine 20, no. 1 (2022): 197, https://doi.org/10.1186/s12967-022-03395-7.

[32]

J. R. W. Conway, D. D. Dinc, G. Follain, et al., “Igfbp2 Secretion by Mammary Adipocytes Limits Breast Cancer Invasion,” Science Advances 9, no. 28 (2023): eadg1840, https://doi.org/10.1126/sciadv.adg1840.

[33]

Z. Pan, T. Xu, L. Bao, et al., “Creb3l1 Promotes Tumor Growth and Metastasis of Anaplastic Thyroid Carcinoma by Remodeling the Tumor Microenvironment,” Molecular Cancer 21, no. 1 (2022): 190, https://doi.org/10.1186/s12943-022-01658-x.

[34]

Z. Yu, J. Zhu, H. Wang, H. Li, and X. Jin, “Function of Bclaf1 in Human Disease,” Oncology Letters 23, no. 2 (2022): 58, https://doi.org/10.3892/ol.2021.13176.

[35]

Y. Xia, Y. Wang, M. Shan, Y. Hao, and Z. Liang, “Decoding the Molecular Landscape of Keloids: New Insights From Single-Cell Transcriptomics,” Burns Trauma 11 (2023): tkad017, https://doi.org/10.1093/burnst/tkad017.

[36]

H. He, H. Suryawanshi, P. Morozov, et al., “Single-Cell Transcriptome Analysis of Human Skin Identifies Novel Fibroblast Subpopulation and Enrichment of Immune Subsets in Atopic Dermatitis,” Journal of Allergy and Clinical Immunology 145, no. 6 (2020): 1615-1628, https://doi.org/10.1016/j.jaci.2020.01.042.

[37]

J. C. Schupp, T. S. Adams, C. Cosme, et al., “Integrated Single-Cell Atlas of Endothelial Cells of the Human Lung,” Circulation 144, no. 4 (2021): 286-302, https://doi.org/10.1161/CIRCULATIONAHA.120.052318.

[38]

T. Zhang, X. F. Wang, Z. C. Wang, et al., “Current Potential Therapeutic Strategies Targeting the Tgf-Beta/Smad Signaling Pathway to Attenuate Keloid and Hypertrophic Scar Formation,” Biomedicine & Pharmacotherapy 129 (2020): 110287, https://doi.org/10.1016/j.biopha.2020.110287.

[39]

Y. F. Zhang, S. Z. Zhou, X. Y. Cheng, et al., “Baicalein Attenuates Hypertrophic Scar Formation via Inhibition of the Transforming Growth Factor-Beta/Smad2/3 Signalling Pathway,” British Journal of Dermatology 174, no. 1 (2016): 120-130, https://doi.org/10.1111/bjd.14108.

[40]

G. Gabbiani, “The Myofibroblast in Wound Healing and Fibrocontractive Diseases,” Journal of Pathology 200, no. 4 (2003): 500-503, https://doi.org/10.1002/path.1427.

[41]

R. H. Fan, X. M. Zhu, Y. W. Sun, H. Z. Peng, H. L. Wu, and W. J. Gao, “Ctrp6 Inhibits Fibrogenesis in Tgf-Beta1-Stimulated Human Dermal Fibroblasts,” Biochemical and Biophysical Research Communications 475, no. 4 (2016): 356-360, https://doi.org/10.1016/j.bbrc.2016.05.013.

[42]

E. J. Macarak, P. J. Wermuth, J. Rosenbloom, and J. Uitto, “Keloid Disorder: Fibroblast Differentiation and Gene Expression Profile in Fibrotic Skin Diseases,” Experimental Dermatology 30, no. 1 (2021): 132-145, https://doi.org/10.1111/exd.14243.

[43]

Y. I. Lee, J. E. Shim, J. Kim, et al., “Wnt5a Drives Interleukin-6-Dependent Epithelial-Mesenchymal Transition via the Jak/Stat Pathway in Keloid Pathogenesis,” Burns Trauma 10 (2022): tkac023, https://doi.org/10.1093/burnst/tkac023.

[44]

X. Lin, Y. Wang, Y. Jiang, et al., “Sumoylation Enhances the Activity of the Tgf-Beta/Smad and Hif-1 Signaling Pathways in Keloids,” Life Sciences 255 (2020): 117859, https://doi.org/10.1016/j.lfs.2020.117859.

[45]

Y. Har-Shai, I. Mettanes, Y. Zilberstein, O. Genin, I. Spector, and M. Pines, “Keloid Histopathology After Intralesional Cryosurgery Treatment,” Journal of the European Academy of Dermatology and Venereology 25, no. 9 (2011): 1027-1036, https://doi.org/10.1111/j.1468-3083.2010.03911.x.

[46]

M. F. Griffin, H. E. desJardins-Park, S. Mascharak, M. R. Borrelli, and M. T. Longaker, “Understanding the Impact of Fibroblast Heterogeneity on Skin Fibrosis,” Disease Models & Mechanisms 13, no. 6 (2020): dmm044164, https://doi.org/10.1242/dmm.044164.

[47]

T. Dehne, C. Karlsson, J. Ringe, M. Sittinger, and A. Lindahl, “Chondrogenic Differentiation Potential of Osteoarthritic Chondrocytes and Their Possible Use in Matrix-Associated Autologous Chondrocyte Transplantation,” Arthritis Research & Therapy 11, no. 5 (2009): R133, https://doi.org/10.1186/ar2800.

[48]

E. El Agha, R. Kramann, R. K. Schneider, et al., “Mesenchymal Stem Cells in Fibrotic Disease,” Cell Stem Cell 21, no. 2 (2017): 166-177, https://doi.org/10.1016/j.stem.2017.07.011.

[49]

E. Funayama, T. Chodon, A. Oyama, and T. Sugihara, “Keratinocytes Promote Proliferation and Inhibit Apoptosis of the Underlying Fibroblasts: An Important Role in the Pathogenesis of Keloid,” Journal of Investigative Dermatology 121, no. 6 (2003): 1326-1331, https://doi.org/10.1111/j.1523-1747.2003.12572.x.

[50]

Q. Jiao, L. Zhi, B. You, G. Wang, N. Wu, and Y. Jia, “Skin Homeostasis: Mechanism and Influencing Factors,” Journal of Cosmetic Dermatology 23, no. 5 (2024): 1518-1526, https://doi.org/10.1111/jocd.16155.

[51]

S. Mohan and D. J. Baylink, “Igf-Binding Proteins Are Multifunctional and Act via Igf-Dependent and -Independent Mechanisms,” Journal of Endocrinology 175, no. 1 (2002): 19-31, https://doi.org/10.1677/joe.0.1750019.

[52]

J. I. Jones, A. Gockerman, W. H. Busby, C. Camacho-Hubner, and D. R. Clemmons, “Extracellular Matrix Contains Insulin-Like Growth Factor Binding Protein-5: Potentiation of the Effects of Igf-I,” Journal of Cell Biology 121, no. 3 (1993): 679-687, https://doi.org/10.1083/jcb.121.3.679.

[53]

T. Kawakita, E. M. Espana, H. He, et al., “Keratocan Expression of Murine Keratocytes Is Maintained on Amniotic Membrane by Down-Regulating Transforming Growth Factor-Beta Signaling,” Journal of Biological Chemistry 280, no. 29 (2005): 27085-27092, https://doi.org/10.1074/jbc.M409567200.

[54]

W. Li, H. He, Y. T. Chen, Y. Hayashida, and S. C. Tseng, “Reversal of Myofibroblasts by Amniotic Membrane Stromal Extract,” Journal of Cellular Physiology 215, no. 3 (2008): 657-664, https://doi.org/10.1002/jcp.21345.

[55]

S. H. Park, K. W. Kim, and J. C. Kim, “The Role of Insulin-Like Growth Factor Binding Protein 2 (Igfbp2) in the Regulation of Corneal Fibroblast Differentiation,” Investigative Ophthalmology & Visual Science 56, no. 12 (2015): 7293-7302, https://doi.org/10.1167/iovs.15-16616.

[56]

D. Medici and R. Kalluri, “Endothelial-Mesenchymal Transition and Its Contribution to the Emergence of Stem Cell Phenotype,” Seminars in Cancer Biology 22, no. 5-6 (2012): 379-384, https://doi.org/10.1016/j.semcancer.2012.04.004.

RIGHTS & PERMISSIONS

2025 The Author(s). Cell Proliferation published by Beijing Institute for Stem Cell and Regenerative Medicine and John Wiley & Sons Ltd.

AI Summary AI Mindmap
PDF

23

Accesses

0

Citation

Detail

Sections
Recommended

AI思维导图

/