1
|
Morioka N, Ganier C, Watt FM. Fetal Fibroblast Heterogeneity Defines Dermal Architecture during Human Embryonic Skin Development. J Invest Dermatol 2025; 145:1081-1091.e7. [PMID: 39880186 DOI: 10.1016/j.jid.2024.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 12/23/2024] [Indexed: 01/31/2025]
Abstract
To investigate the heterogeneity of fibroblasts in human fetal skin, we analyzed published single-cell RNA-sequencing data (7 and 16 weeks after conception) and performed single-molecule FISH to map their spatial distribution and predicted dynamic interactions. Clustering revealed 8 fibroblast populations with changes in developmental stage-specific abundance. Proliferative cells (MKI67+) were present at all stages. The appearance of dermal papilla (PRDM1+) and hair follicle (SLC26A7+) fibroblasts coincided with hair follicle maturation, whereas fibroblasts (apolipoprotein E positive) specifically associated with blood vessels increased in abundance as the vessels developed. HOXC5 was a marker of the most abundant fibroblasts 7-8 weeks after conception; this cluster was diminished 9-13 weeks after conception and undetectable subsequently. A second population (PLAT+) decreased in abundance with the same kinetics. Fibroblasts corresponding to papillary dermis (GRP+) were predominant 9-13 weeks after conception, whereas reticular dermal fibroblasts (ASPN+) were the major cluster 14-16 weeks after conception. Partition-based graph abstraction and pseudotime analysis indicated that the HOXC5+ fibroblasts were closely connected with the papillary and hair follicle fibroblasts, whereas the PLAT+ fibroblasts were connected with reticular and vascular fibroblasts. Dermal papilla fibroblasts were the most highly differentiated. Integration of fetal and adult datasets distinguished the adult and fetal papillary clusters from the reticular clusters.
Collapse
Affiliation(s)
- Noriko Morioka
- Centre for Gene Therapy and Regenerative Medicine, Guy's Hospital, King's College London, London, United Kingdom; Frontier Research Center, Pola Chemical Industries, Yokohama, Japan
| | - Clarisse Ganier
- Centre for Gene Therapy and Regenerative Medicine, Guy's Hospital, King's College London, London, United Kingdom; Meta-organism Unit, Immunology Department, Institut Pasteur, Paris, France.
| | - Fiona M Watt
- Centre for Gene Therapy and Regenerative Medicine, Guy's Hospital, King's College London, London, United Kingdom; Directors' Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
2
|
Wen J, Jin S, Luo X, Chen C, Liu H, Li Y, Li J. Functionalized gelatin/poly(l-lactide-co-ε-caprolactone) fibrous membrane promotes scarless wound healing by modulating inflammation and reducing fibrosis. Int J Biol Macromol 2025; 306:141785. [PMID: 40054818 DOI: 10.1016/j.ijbiomac.2025.141785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/03/2025] [Accepted: 03/04/2025] [Indexed: 05/11/2025]
Abstract
In cases of deep skin defects, spontaneous tissue regeneration and excessive collagen deposition can result in the formation of proliferative scarring. Salvianolic acid B (SAB) demonstrates promising applications in the treatment of fibrotic diseases, including scarring. This study prepared biodegradable gelatin (Gel) and poly(lactic-co-ε-caprolactone) (PLCL) nanofibrous membranes using electrospinning technology and functionalized them with salvianolic acid B to create fibrous membrane dressings with anti-scarring properties. The resulting Gel/PLCL/SAB (GPS) fibrous membrane exhibited good mechanical properties and biodegradability. In vitro experiments demonstrated that the fibrous membranes of Gel/PLCL containing 0.5 wt% SAB (GPS0.5) exhibited good cytocompatibility, regulated macrophage function and polarization, and inhibited fibrosis-related genes, including CD36, α-SMA, and collagen I. In a mouse model of full-thickness skin defects, the GPS0.5 fibrous membrane effectively modulated the immune microenvironment and promoted vascular regeneration, thereby accelerating skin healing. Furthermore, the GPS0.5 fibrous membrane reduced the expression of CD36 and TGF-β1, downregulated collagen I and III, and promoted the regeneration of hair follicles, sebaceous glands, and other appendages, ultimately reducing scar formation. Thus, SAB-loaded Gel/PLCL fibrous membranes hold potential as bifunctional skin dressings that promote wound healing and inhibit fibrosis.
Collapse
Affiliation(s)
- Jing Wen
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Shue Jin
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Xue Luo
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Chunhong Chen
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Huan Liu
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Yubao Li
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China
| | - Jidong Li
- The Research Center for Nano-Biomaterials, Analytical and Testing Center, Sichuan University, Chengdu 610065, China.
| |
Collapse
|
3
|
Torregrossa M, Davies L, Hans-Günther M, Simon JC, Franz S, Rinkevich Y. Effects of embryonic origin, tissue cues and pathological signals on fibroblast diversity in humans. Nat Cell Biol 2025; 27:720-735. [PMID: 40263573 DOI: 10.1038/s41556-025-01638-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 02/18/2025] [Indexed: 04/24/2025]
Abstract
Fibroblasts, once perceived as a uniform cell type, are now recognized as a mosaic of distinct populations with specialized roles in tissue homeostasis and pathology. Here we provide a global overview of the expanding compendium of fibroblast cell types and states, their diverse lineage origins and multifaceted functions across various human organs. By integrating insights from developmental biology, lineage tracing and single-cell technologies, we highlight the complex nature of fibroblasts. We delve into their origination from embryonic mesenchyme and tissue-resident populations, elucidating lineage-specific behaviours in response to physiological cues. Furthermore, we highlight the pivotal role of fibroblasts in orchestrating tissue repair, connective tissue remodelling and immune modulation across diverse pathologies. This knowledge is essential to develop novel fibroblast-targeted therapies to restore steady-state fibroblast function and advance regenerative medicine strategies across multiple diseases.
Collapse
Affiliation(s)
- Marta Torregrossa
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Lindsay Davies
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm, Sweden
| | - Machens Hans-Günther
- Department for Plastic Surgery and Hand Surgery, Klinikum Rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jan C Simon
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany
| | - Sandra Franz
- Department of Dermatology, Venereology and Allergology, Leipzig University Medical Faculty, Leipzig, Germany.
| | - Yuval Rinkevich
- Chinese Institutes for Medical Research, Beijing, China.
- Capital Medical University, Beijing, China.
| |
Collapse
|
4
|
Scott RW. The Beauty of Fibroblasts Is Several Skin Layers Deep and Much Deeper. J Invest Dermatol 2025; 145:994-997. [PMID: 40072401 DOI: 10.1016/j.jid.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 04/25/2025]
Affiliation(s)
- R Wilder Scott
- Holland Bone and Joint Program, Sunnybrook Health Sciences Centre, Toronto, Canada; Biological Sciences Platform, Sunnybrook Research Institute, Toronto, Canada; Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada.
| |
Collapse
|
5
|
Xu J, Zhang H, Ye H. Research progress on the role of fascia in skin wound healing. BURNS & TRAUMA 2025; 13:tkaf002. [PMID: 40248160 PMCID: PMC12001785 DOI: 10.1093/burnst/tkaf002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 04/19/2025]
Abstract
The skin, the human body's largest organ, is perpetually exposed to environmental factors, rendering it vulnerable to potential injuries. Fascia, a vital connective tissue that is extensively distributed throughout the body, fulfils multiple functions, including support, compartmentalization, and force transmission. The role of fascia in skin wound healing has recently attracted considerable attention. In addition to providing mechanical support, fascia significantly contributes to intercellular signalling and tissue repair, establishing itself as a crucial participant in wound healing. This review synthesises the latest advancements in fascia research and its implications for skin wound healing.
Collapse
Affiliation(s)
- Jiamin Xu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital; School of Basic Medical Sciences; Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Hongyan Zhang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital; School of Basic Medical Sciences; Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| | - Haifeng Ye
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital; School of Basic Medical Sciences; Institute of Biomedical Innovation, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, China
| |
Collapse
|
6
|
Li DD, Lan N, Zhao P, Tang YY. Advances in Etiology and Prevention of Capsular Contracture After Breast Implantation. Aesthetic Plast Surg 2025; 49:1915-1926. [PMID: 39586860 PMCID: PMC12031949 DOI: 10.1007/s00266-024-04500-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/25/2024] [Indexed: 11/27/2024]
Abstract
Capsular contracture (CC) is one of the most common complications of breast implant usage in breast augmentation or reconstruction. The CC approach can cause breast hardening, pain, and varying degrees of deformity, affecting the quality of life of patients. Considerably, it has become one of the most common reasons for frequent surgeries. Nonetheless, the etiology and pathogenesis of CC remain unclear. Moreover, there exist still a lot of uncertainties regarding prevention and treatment measures. In this article, we present discussions on the research status of the etiology, pathogenesis, prevention, and treatment measures of CC. In summary, this study provides a reference for further research on CC and clinical use.Level of Evidence V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
Affiliation(s)
- Dan-Dan Li
- The Second Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 of Kunzhou Street, Xishan District, Kunming, 650000, China
| | - Nan Lan
- The Second Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 of Kunzhou Street, Xishan District, Kunming, 650000, China
| | - Ping Zhao
- The First Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, Xishan District, No. 519 of Kunzhou Street, Kunming, 650000, China.
| | - Yi-Yin Tang
- The Second Department of Breast Surgery, The Third Affiliated Hospital of Kunming Medical University, No. 519 of Kunzhou Street, Xishan District, Kunming, 650000, China.
| |
Collapse
|
7
|
Tiskratok W, Chuinsiri N, Limraksasin P, Kyawsoewin M, Jitprasertwong P. Extracellular Matrix Stiffness: Mechanotransduction and Mechanobiological Response-Driven Strategies for Biomedical Applications Targeting Fibroblast Inflammation. Polymers (Basel) 2025; 17:822. [PMID: 40292716 PMCID: PMC11946729 DOI: 10.3390/polym17060822] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
The extracellular matrix (ECM) is a dynamic network providing mechanical and biochemical cues that regulate cellular behavior. ECM stiffness critically influences fibroblasts, the primary ECM producers, particularly in inflammation and fibrosis. This review explores the role of ECM stiffness in fibroblast-driven inflammation and tissue remodeling, focusing on the physicochemical and biological mechanisms involved. Engineered materials, hydrogels, and polydimethylsiloxane (PDMS) are highlighted for replicating tissue-specific stiffness, enabling precise control over cell-matrix interactions. The surface functionalization of substrate materials, including collagen, polydopamine, and fibronectin, enhances bioactivity and fibroblast adhesion. Key mechanotransduction pathways, such as integrin signaling and YAP/TAZ activation, are related to regulating fibroblast behaviors and inflammatory responses. The role of fibroblasts in driving chronic inflammatory diseases emphasizes their therapeutic potentials. Advances in ECM-modifying strategies, including tunable biomaterials and hydrogel-based therapies, are explored for applications in tissue engineering, drug delivery, anti-inflammatory treatments, and diagnostic tools for the accurate diagnosis and prognosis of ECM stiffness-related inflammatory diseases. This review integrates mechanobiology with biomedical innovations, providing a comprehensive prognosis of fibroblast responses to ECM stiffness and outlining future directions for targeted therapies.
Collapse
Affiliation(s)
- Watcharaphol Tiskratok
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Nontawat Chuinsiri
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| | - Phoonsuk Limraksasin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Maythwe Kyawsoewin
- Center of Excellence for Dental Stem Cell Biology, Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (P.L.); (M.K.)
| | - Paiboon Jitprasertwong
- Institute of Dentistry, Suranaree University of Technology, Nakhon Ratchasima 30000, Thailand; (N.C.); (P.J.)
- Oral Health Centre, Suranaree University of Technology Hospital, Nakhon Ratchasima 30000, Thailand
| |
Collapse
|
8
|
Tang P, Wei F, Qiao W, Chen X, Ji C, Yang W, Zhang X, Chen S, Wu Y, Jiang M, Ma C, Shen W, Dong Q, Cao H, Xie M, Cai Z, Xu L, Shi J, Dong N, Chen J, Wang N. Engineering aortic valves via transdifferentiating fibroblasts into valvular endothelial cells without using viruses or iPS cells. Bioact Mater 2025; 45:181-200. [PMID: 39651397 PMCID: PMC11625219 DOI: 10.1016/j.bioactmat.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/30/2024] [Accepted: 11/14/2024] [Indexed: 12/11/2024] Open
Abstract
The technology of induced pluripotent stem cells (iPSCs) has enabled the conversion of somatic cells into primitive undifferentiated cells via reprogramming. This approach provides possibilities for cell replacement therapies and drug screening, but the potential risk of tumorigenesis hampers its further development and in vivo application. How to generate differentiated cells such as valvular endothelial cells (VECs) has remained a major challenge. Utilizing a combinatorial strategy of selective soluble chemicals, cytokines and substrate stiffness modulation, mouse embryonic fibroblasts are directly and efficiently transdifferentiated into induced aortic endothelial cell-like cells (iAECs), or human primary adult fibroblasts are transdifferentiated into induced valvular endothelial cell-like cells (hiVECs), without expressing pluripotency stem cell markers. These iAECs and hiVECs express VEC-associated genes and proteins and VEC-specific marker NFATC1 and are functional in culture and on decellularized porcine aortic valves, like mouse aortic endothelial cells or human primary aortic valvular endothelial cells. The iAECs and hiVECs seeded on decellularized porcine aortic valves stay intact and express VEC-associated proteins for 60 days after grafting into abdominal aorta of immune-compromised rats. In contrast, induced pluripotent stem cells (iPSCs) are less efficient in differentiating into VEC-like cells and pluripotency marker Nanog is expressed in a small subpopulation of iPSC-derived VEC-like cells that generate teratomas in SCID mice whereas hiVECs derived from transdifferentiation do not generate teratomas in vivo. Our findings highlight an approach to efficiently convert fibroblasts into iAECs and hiVECs and seed them onto decellularized aortic valves for safely generating autologous tissue-engineered aortic valves without using viruses or first reprogramming the cells into pluripotent stem cells.
Collapse
Affiliation(s)
- Peng Tang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Fuxiang Wei
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xing Chen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chenyang Ji
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Wanzhi Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Xinyu Zhang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Sihan Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yanyan Wu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Mingxing Jiang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Chenyu Ma
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Weiqiang Shen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Qi Dong
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Minghui Xie
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ziwen Cai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Junwei Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Bioinformatics and Molecular Imaging Key Laboratory, Laboratory for Cellular Biomechanics and Regenerative Medicine, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Ning Wang
- Institute for Mechanobiology, Department of Bioengineering, College of Engineering, Northeastern University, Boston, MA, 02115, USA
| |
Collapse
|
9
|
Yang MY, Quan HY, Li DL, Ruan J, Fan HY. Targeting TEAD would be a potential strategy for scarless wound repair: A preliminary study. Acta Histochem 2025; 127:152223. [PMID: 39667304 DOI: 10.1016/j.acthis.2024.152223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 11/10/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
Despite of decades of efforts, novel approaches are still limited to attenuate or prevent skin scarring. A previous report published in Science demonstrated that inhibition of YAP promotes scarless wound repair by regeneration. Due to the difficult drugability of targeting YAP, we speculated that inhibition of TEAD, a partner molecule of YAP, might exist similar therapeutic potential. Therefore, the aim of the study was to evaluate therapeutical effect of a novel inhibitor of TEAD auto-palmitoylation, VT107, on scar formation in a cutaneous wound healing model. Our findings confirmed VT107 exhibited favorable effect on preventing scarring, manifesting as reducing fibroblast proliferation and collagen denaturation, decreasing TGF-β1 and collagen deposition, as well as connective tissue growth factor (CTGF) expression. These findings provide a novel insight for the development of anti-scarring strategies. TEAD would become an ideal target for the treatment of scars.
Collapse
Affiliation(s)
- Ming-Yan Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, Shandong 264005, China
| | - Hong-Yuan Quan
- Guangxi University, No. 100 Daxue East Road, Nanning, Guangxi 530004, China
| | - Da-Lei Li
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, Shandong 264005, China
| | - Jian Ruan
- Yantai Center for Food and Drug Control, Yantai 264000, China.
| | - Hua-Ying Fan
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, No. 32 Qingquan Road, Laishan District, Yantai, Shandong 264005, China.
| |
Collapse
|
10
|
Shieh TM, Lin NC, Shen YW, Lan WC, Shih YH. Epithelium-derived exosomal dipeptidyl peptidase-4 involved in arecoline-induced oral submucous fibrosis. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167683. [PMID: 39837428 DOI: 10.1016/j.bbadis.2025.167683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 01/16/2025] [Accepted: 01/17/2025] [Indexed: 01/23/2025]
Abstract
INTRODUCTION Dipeptidyl peptidase-4 is known to be involved in the progression of several fibrogenic diseases, but its association with oral submucous fibrosis remains unclear. This study aims to ascertain whether dipeptidyl peptidase-4 plays a role in the pathogenesis of arecoline-induced oral submucous fibrosis. METHODS We assessed the expression of dipeptidyl peptidase-4 in arecoline-treated epithelial cells and the exosomes derived from cells. We cocultured the fibroblast and exosomes derived from epithelium cells and assessed fibrogenic activity by measuring collagen secretion, α-SMA expression, and gel contraction capability. An animal study was conducted to confirm the fibrogenic activity of exosomes derived from arecoline-treated epithelial cells. Additionally, we employed a dipeptidyl peptidase-4 inhibitor to assess its efficacy in mitigating fibrogenesis. RESULTS Following arecoline treatment, an increase dipeptidyl peptidase-4 expression was observed in exosomes from the treated epithelium cells. When these exosomes cocultured with fibroblast, fibrogenic gene α-SMA was upregulated, increased collagen secretion, and enhanced gel contraction capability. In a mouse model, the administration of arecoline-treated epithelium-derived exosomes induced oral submucous fibrosis phenotype, characterized by a reduction in incisal distance and epithelial atrophy. CONCLUSIONS These findings offer valuable insights into clinical strategies for combating oral fibrotic disease and contribute to the foundation of future research in this field.
Collapse
Affiliation(s)
- Tzong-Ming Shieh
- School of Dentistry, China Medical University, 404332 Taichung, Taiwan; Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei 112304, Taipei, Taiwan.
| | - Nan-Chin Lin
- Department of Oral and Maxillofacial Surgery, Show Chwan Memorial Hospital, 500 Changhua, Taiwan.
| | - Yen-Wen Shen
- Department of Dentistry, China Medical University Hospital, 404332 Taichung City, Taiwan.
| | - Wan-Chen Lan
- Department of Healthcare Administration, Asia University, 40454 Taichung, Taiwan.
| | - Yin-Hwa Shih
- Department of Healthcare Administration, Asia University, 40454 Taichung, Taiwan.
| |
Collapse
|
11
|
Tauch S, Hey J, Kast B, Gengenbacher N, Weiß L, Sator‐Schmitt M, Lohr S, Brobeil A, Schirmacher P, Utikal J, Augustin HG, Plass C, Angel P. A Unique Signature for Cancer-Associated Fibroblasts in Melanoma Metastases. Pigment Cell Melanoma Res 2025; 38:e70002. [PMID: 39924882 PMCID: PMC11808227 DOI: 10.1111/pcmr.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 11/27/2024] [Accepted: 01/16/2025] [Indexed: 02/11/2025]
Abstract
Cancer-associated fibroblasts (CAFs) represent a central cell population of the tumor microenvironment (TME). Recently, single-cell RNA-sequencing (scRNA-seq) analyses of primary tumors of different cancer entities yielded different classifications of CAF subsets underscoring the heterogeneity of CAFs within the TME. Here, we analyzed the transcriptional signatures of approximately 8400 CAFs and normal fibroblasts by scRNA-seq and compared genetic profiles of CAFs from murine melanoma primary tumors to CAFs from corresponding melanoma lung metastases. This revealed distinct subsets for primary tumor and metastasis-specific CAF populations, respectively. Combined with the spatial characterization of metastasis CAFs at the RNA and protein level, scRNA analyses indicate tumor-dependent crosstalk between neutrophils and CAFs, mediated via SAA3 and IL1b-related signaling pathways, which can be recapitulated in vitro. Analyzing tissue sections of human patient samples, this interaction was found to be present in human melanoma metastasis. Taken together, our data highlight unique characteristics of metastasis CAFs with potential therapeutic impact for melanoma metastasis.
Collapse
Affiliation(s)
- Saskia Tauch
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- Faculty of BiosciencesHeidelberg UniversityHeidelbergGermany
| | - Joschka Hey
- Division of Cancer EpigenomicsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Bettina Kast
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Nicolas Gengenbacher
- Division of Vascular Oncology and MetastasisGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- European Center for Angioscience (ECAS), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
| | - Lena Weiß
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Melanie Sator‐Schmitt
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Sabrina Lohr
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| | - Alexander Brobeil
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Peter Schirmacher
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Jochen Utikal
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
- Skin Cancer UnitGerman Cancer Research Center (DKFZ)HeidelbergGermany
- Department of Dermatology, Venereology and AllergologyUniversity Medical Center Mannheim, Ruprecht‐Karl University of HeidelbergMannheimGermany
| | - Hellmut G. Augustin
- Division of Vascular Oncology and MetastasisGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
- European Center for Angioscience (ECAS), Medical Faculty MannheimHeidelberg UniversityMannheimGermany
- DKFZ‐Hector Cancer Institute, University Medical Centre MannheimMannheimGermany
| | - Christoph Plass
- Division of Cancer EpigenomicsGerman Cancer Research Center (DKFZ)HeidelbergGermany
| | - Peter Angel
- Division Signal Transduction and Growth ControlGerman Cancer Research Center (DKFZ‐ZMBH Alliance)HeidelbergGermany
| |
Collapse
|
12
|
Mascharak S, Griffin M, Talbott HE, Guo JL, Parker J, Morgan AG, Valencia C, Kuhnert MM, Li DJ, Liang NE, Kratofil RM, Daccache JA, Sidhu I, Davitt MF, Guardino N, Lu JM, Abbas DB, Deleon NMD, Lavin CV, Adem S, Khan A, Chen K, Henn D, Spielman A, Cotterell A, Akras D, Downer M, Tevlin R, Lorenz HP, Gurtner GC, Januszyk M, Naik S, Wan DC, Longaker MT. Inhibiting mechanotransduction prevents scarring and yields regeneration in a large animal model. Sci Transl Med 2025; 17:eadt6387. [PMID: 39970235 DOI: 10.1126/scitranslmed.adt6387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/29/2025] [Indexed: 02/21/2025]
Abstract
Modulating mechanotransduction by inhibiting yes-associated protein (YAP) in mice yields wound regeneration without scarring. However, rodents are loose-skinned and fail to recapitulate key aspects of human wound repair. We sought to elucidate the effects of YAP inhibition in red Duroc pig wounds, the most human-like model of scarring. We show that one-time treatment with verteporfin, a YAP inhibitor, immediately after wounding is sufficient to prevent scarring and to drive wound regeneration in pigs. By performing single-cell RNA sequencing (scRNA-seq) on porcine wounds in conjunction with spatial proteomic analysis, we found perturbations in fibroblast dynamics with verteporfin treatment and the presence of putative pro-regenerative/profibrotic fibroblasts enriched in regenerating/scarring pig wounds, respectively. We also identified differences in enriched myeloid cell subpopulations after treatment and linked this observation to increased elaboration of interleukin-33 (IL-33) in regenerating wounds. Finally, we validated our findings in a xenograft wound model containing human neonatal foreskin engrafted onto nude mice and used scRNA-seq of human wound cells to draw parallels with fibroblast subpopulation dynamics in porcine wounds. Collectively, our findings provide support for the clinical translation of local mechanotransduction inhibitors to prevent human skin scarring, and they clarify a YAP/IL-33 signaling axis in large animal wound regeneration.
Collapse
Affiliation(s)
- Shamik Mascharak
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michelle Griffin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Heather E Talbott
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jason L Guo
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer Parker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Annah Grace Morgan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Caleb Valencia
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maxwell Michael Kuhnert
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dayan J Li
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Norah E Liang
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Rachel M Kratofil
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joseph A Daccache
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ikjot Sidhu
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Applied Bioinformatics Laboratories, NYU Langone Health, New York, NY 10016, USA
| | - Michael F Davitt
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nicholas Guardino
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - John M Lu
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Darren B Abbas
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nestor M D Deleon
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Christopher V Lavin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sandeep Adem
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Anum Khan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kellen Chen
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dominic Henn
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Amanda Spielman
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Asha Cotterell
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Deena Akras
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Mauricio Downer
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ruth Tevlin
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - H Peter Lorenz
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Geoffrey C Gurtner
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Shruti Naik
- Department of Pathology, NYU Langone Health, New York, NY 10016, USA
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Ronald O. Perelman Department of Dermatology, NYU Langone Health, New York, NY 10016, USA
- Perlmutter Cancer Center, NYU Langone Health, New York, NY 10016, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
13
|
Kang M, Ko UH, Oh EJ, Kim HM, Chung HY, Shin JH. Tension-sensitive HOX gene expression in fibroblasts for differential scar formation. J Transl Med 2025; 23:168. [PMID: 39930512 PMCID: PMC11808978 DOI: 10.1186/s12967-025-06191-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/31/2025] [Indexed: 02/14/2025] Open
Abstract
BACKGROUND Scar formation is a common end-point of the wound healing process, but its mechanisms, particularly in relation to abnormal scars such as hypertrophic scars and keloids, remain not fully understood. This study unveils a novel mechanistic insight into scar formation by examining the differential expression of Homeobox (HOX) genes in response to mechanical forces in fibroblasts derived from normal skin, hypertrophic scars, and keloids. METHODS We isolated fibroblasts from different scar types and conducted RNA sequencing (RNA-Seq) to identify differential gene expression patterns among the fibroblasts. Computational modeling provided insight into tension alterations following injury, and these findings were complemented by in vitro experiments where fibroblasts were subjected to exogenous tensile stress to investigate the link between mechanical tension and cellular behavior. RESULTS Our study revealed differential HOX gene expression among fibroblasts derived from normal skin, hypertrophic scars, and keloids. Computational simulations predicted injury-induced tension reduction in the skin, and in vitro experiments revealed a negative correlation between tension and fibroblast proliferation. Importantly, we discovered that applying mechanical tension to fibroblasts can modulate HOX gene expression, suggesting a pivotal role of mechanical cues in scar formation and wound healing. CONCLUSION This study proposes a model wherein successful wound healing and scar formation are critically dependent on maintaining tensional homeostasis in the skin, mediated by tension-sensitive HOX genes. Our findings highlight the potential of targeting mechanotransduction pathways and tension-sensitive HOX gene expression as therapeutic strategies for abnormal scar prevention and treatment, offering a new perspective on the complex process of scar formation.
Collapse
Affiliation(s)
- Minwoo Kang
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Ung Hyun Ko
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Eun Jung Oh
- Department of Plastic & Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Hyun Mi Kim
- Department of Plastic & Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Ho Yun Chung
- Department of Plastic & Reconstructive Surgery, CMRI, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Jennifer H Shin
- Department of Mechanical Engineering, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.
| |
Collapse
|
14
|
Zhao S, Xie J, Zhang Q, Ni T, Lin J, Gao W, Zhao L, Yi M, Tu L, Zhang P, Wu D, Tang Q, Ma C, He Y, Li L, Wu G, Yan W. New Anti-Fibrotic Strategies for Keloids: Insights From Single-Cell Multi-Omics. Cell Prolif 2025:e13818. [PMID: 39902627 DOI: 10.1111/cpr.13818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 01/01/2025] [Accepted: 01/15/2025] [Indexed: 02/05/2025] Open
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.
Collapse
Affiliation(s)
- Songyun Zhao
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiaheng Xie
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
- Department of Plastic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Qian Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tianyi Ni
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jinde Lin
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Weicheng Gao
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Liping Zhao
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Min Yi
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Liying Tu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Pengpeng Zhang
- Department of Lung Cancer Surgery, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Dan Wu
- Department of Dermatology, Huashan Hospital of Fudan University, Shanghai, China
| | - Qikai Tang
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Chenfeng Ma
- Department of Neurosurgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yucang He
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liqun Li
- Department of Plastic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guoping Wu
- Department of Plastic Surgery, The Affiliated Friendship Plastic Surgery Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Yan
- Department of Burn and Plastic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
15
|
Allen RS, Seifert AW. Spiny mice (Acomys) have evolved cellular features to support regenerative healing. Ann N Y Acad Sci 2025; 1544:5-26. [PMID: 39805008 PMCID: PMC11830558 DOI: 10.1111/nyas.15281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Spiny mice (Acomys spp.) are warm-blooded (homeothermic) vertebrates whose ability to restore missing tissue through regenerative healing has coincided with the evolution of unique cellular and physiological adaptations across different tissue types. This review seeks to explore how these bizarre rodents deploy unique or altered injury response mechanisms to either enhance tissue repair or fully regenerate excised tissue compared to closely related, scar-forming mammals. First, we examine overall trends in healing Acomys tissues, including the cellular stress response, the ability to activate and maintain cell cycle progression, and the expression of certain features in reproductive adults that are normally associated with embryos. Second, we focus on specific cell types that exhibit precisely regulated proliferation to restore missing tissue. While Acomys utilize many of the same cell types involved in scar formation, these cells exhibit divergent activation profiles during regenerative healing. Considered together, current lines of evidence support sustained deployment of proregenerative pathways in conjunction with transient activation of fibrotic pathways to facilitate regeneration and improve tissue repair in Acomys.
Collapse
Affiliation(s)
- Robyn S. Allen
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
| | - Ashley W. Seifert
- Department of Biology, University of Kentucky, Lexington, Kentucky, USA
- The Spinal Cord and Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, Kentucky, USA
- Department of Veterinary Anatomy and Physiology, University of Nairobi, Nairobi, Kenya
| |
Collapse
|
16
|
Gomes MLNP, Krijnen PAJ, Middelkoop E, Niessen HWM, Boekema BKHL. Fetal Skin Wound Healing: Key Extracellular Matrix Components and Regulators in Scarless Healing. J Invest Dermatol 2025; 145:280-302. [PMID: 39152955 DOI: 10.1016/j.jid.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 08/19/2024]
Abstract
Fetal skin at early gestational stage is able to regenerate and heal rapidly after wounding. The exact mechanisms and molecular pathways involved in this process are however still largely unknown. The numerous differences in the skin of the early fetus versus skin in later developmental stages might provide clues for the mechanisms of scarless healing. This review summarizes the differences between mammalian fetal skin and the skin at later developmental phases in healthy and wounded conditions, focusing on extracellular matrix components, which are crucial factors in the microenvironment that direct cells and tissue functions and hence the wound healing process.
Collapse
Affiliation(s)
- Madalena Lopes Natário Pinto Gomes
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Tissue Function & Regeneration, Amsterdam Movement Sciences, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands
| | - Paul A J Krijnen
- Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, Amsterdam, The Netherlands
| | - Esther Middelkoop
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands; Tissue Function & Regeneration, Amsterdam Movement Sciences, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Burn Centre, Red Cross Hospital, Beverwijk, The Netherlands
| | - Hans W M Niessen
- Department of Pathology, Amsterdam UMC Location AMC, Amsterdam, The Netherlands; Amsterdam Cardiovascular Sciences Institute, Amsterdam UMC, Amsterdam, The Netherlands; Department of Cardio-thoracic Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands
| | - Bouke K H L Boekema
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC (Location VUmc), Amsterdam, The Netherlands; Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, The Netherlands.
| |
Collapse
|
17
|
Bashore AC, Coronel J, Xue C, Zhu LY, Reilly MP. Single-Cell Multimodal Profiling Reveals a Novel CD26 + Fibroblast Subpopulation in Atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.27.635067. [PMID: 39975362 PMCID: PMC11838260 DOI: 10.1101/2025.01.27.635067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Background Atherosclerosis involves complex interactions between lipids, immune cells, vascular smooth muscle cells (VSMCs), and fibroblasts within the arterial wall. While significant advances in single-cell technologies have shed light on the roles of immune cells and VSMCs in plaque development, fibroblasts remain underexplored, leaving critical gaps in understanding their contributions to disease progression and plaque stability. Comprehensive characterization of fibroblast phenotypes in atherosclerosis is essential to unravel their diverse functions and to distinguish between subsets that may play protective versus pathogenic roles in the disease process. Methods Here, we utilized CITE-seq (Cellular Indexing of Transcriptomes and Epitopes by Sequencing) to comprehensively profile fibroblast diversity in a mouse model of atherosclerosis. Mice were fed an atherogenic diet for 0, 8, 19, and 26 weeks, representing distinct stages of disease progression, enabling a detailed phenotypic characterization of fibroblasts throughout the course of atherosclerosis development. Results We identified four distinct fibroblast subpopulations, including a myofibroblast population closely resembling VSMC-derived chondromyocytes. The proportions of these fibroblast subsets exhibited a modest decline as atherosclerosis progressed. Through multimodal analysis, we identified CD26 as a highly expressed and specific marker for one of these fibroblast subpopulations, distinguishing it from other subsets. Using a combination of flow cytometry and immunohistochemistry, we demonstrated that CD26+ fibroblasts predominantly reside in the adventitia of healthy arteries. During atherosclerosis progression, these cells expand into the intima and primarily localize within the fibrous cap of the lesion. Conclusions Our multi-omic analysis highlights the phenotypic diversity and dynamic changes of fibroblasts during atherosclerosis progression. Among these, CD26+ fibroblasts emerge as a distinct subpopulation that expands within atherosclerotic lesions and may play a critical role in promoting plaque stability through their migration into the fibrous cap.
Collapse
Affiliation(s)
- Alexander C Bashore
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Johana Coronel
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Chenyi Xue
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Lucie Y Zhu
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
| | - Muredach P Reilly
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, USA
- Irving Institute for Clinical and Translational Research, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
18
|
Kudelka MR, Lavin Y, Sun S, Fuchs E. Molecular and cellular dynamics of squamous cell carcinomas across tissues. Genes Dev 2025; 39:18-35. [PMID: 39455281 PMCID: PMC11789493 DOI: 10.1101/gad.351990.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2024]
Abstract
Squamous cell carcinomas (SCCs), arising from the skin, head and neck, lungs, esophagus, and cervix, are collectively among the most common cancers and a frequent cause of cancer morbidity and mortality. Despite distinct stratified epithelial tissues of origin, converging evidence points toward shared biologic pathways across SCCs. With recent breakthroughs in molecular technologies have come novel SCC treatment paradigms, including immunotherapies and targeted therapy. This review compares commonalities and differences across SCCs from different anatomical sites, including risk factors and genetics, as well as cellular and molecular programs driving tumorigenesis. We review landmark discoveries of the "cancer stem cells" (CSCs) that initiate and propagate SCCs and their gene and translational regulation programs. This has led to an appreciation that interactions between CSCs and the immune system play key roles in invasion and therapeutic resistance. Here, we review the unifying principles of SCCs that have emerged from these exciting advances in our understanding of these epithelial cancers.
Collapse
Affiliation(s)
- Matthew R Kudelka
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Yonit Lavin
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10065, USA
| | - Siman Sun
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA
| | - Elaine Fuchs
- Howard Hughes Medical Institute, Robin Chemers Neustein Laboratory of Mammalian Cell Biology and Development, The Rockefeller University, New York, New York 10065, USA;
| |
Collapse
|
19
|
Lim CH, Kaminaka A, Lee SH, Moore S, Cronstein BN, Rabbani PS, Ito M. Dermal β-Catenin Is Required for Hedgehog-Driven Hair Follicle Neogenesis. J Invest Dermatol 2025; 145:42-49.e2. [PMID: 38810955 PMCID: PMC11599465 DOI: 10.1016/j.jid.2024.04.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/31/2024]
Abstract
Hair follicle neogenesis (HFN) occurs after large skin excisions in mice, serving as a rare regenerative model in mammalian wound healing. Wound healing typically results in fibrosis in mice and humans. We previously showed that small skin excisions in mice result in scarring devoid of HFN, displaying features of nonregenerative healing, and hedgehog (Hh) activation in the dermis of such wounds can induce HFN. In this study, we sought to verify the role of dermal Wnt/β-catenin signaling in HFN because this pathway is essential for hair follicle development but is also paradoxically well-characterized in fibrosis of adult wounds. By deletion of β-catenin in large wound myofibroblasts, we show that Wnt/β-catenin signaling is required for endogenous mechanisms of HFN. By utilizing a combined mouse model that simultaneously induces deletion of β-catenin and constitutive activation of Smoothened in myofibroblasts, we also found that β-catenin is required for Hh-driven dermal papilla formation. Transcriptome analysis confirms that Wnt/β-catenin and Hh pathways are activated in dermal papilla cells. Our results indicate that Wnt-active fibrotic status may also create a permissive state for the regenerative function of Hh, suggesting that activation of both Wnt and Hh pathways in skin wound fibroblasts must be ensured in future strategies to promote HFN.
Collapse
Affiliation(s)
- Chae Ho Lim
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA; Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, USA.
| | - Annette Kaminaka
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA; Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, USA
| | - Soung-Hoon Lee
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA; Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, USA
| | - Simone Moore
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA; Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, USA
| | - Bruce N Cronstein
- NYU-H+H Clinical and Translational Science Institute, NYU Grossman School of Medicine, New York, New York, USA
| | - Piul S Rabbani
- Hansjörg Wyss Department of Plastic Surgery, NYU Grossman School of Medicine, New York, New York, USA
| | - Mayumi Ito
- Ronald O. Perelman Department of Dermatology, NYU Grossman School of Medicine, New York, New York, USA; Department of Cell Biology, NYU Grossman School of Medicine, New York, New York, USA
| |
Collapse
|
20
|
Liu M, Liu X, Zhang J, Liang S, Gong Y, Shi S, Yuan X. Single-cell RNA sequencing reveals the heterogeneity of myofibroblasts in wound repair. Genomics 2025; 117:110982. [PMID: 39706310 DOI: 10.1016/j.ygeno.2024.110982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 12/09/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
Skin wound repair involves myofibroblasts crucial for tissue integrity. This study utilized single-cell RNA sequencing to explore myofibroblast diversity in various wound healing scenarios. Analysis of 89,148 cells from skin ulcers, keloids, and normal scars identified 13 cell clusters. Myofibroblast subcluster analysis unveiled 11 subsets, with subclusters 1 and 9 predominant in ulcers. Subcluster 1 exhibited heightened matrix metalloproteinase expression and involvement in bacterial response and angiogenesis, crucial in inflammation. Tissue validation confirmed subcluster 1 significance., while animal models supported upregulated CA12, TDO2, and IL-7R in chronic ulcers. These findings illuminate myofibroblast heterogeneity and their impact on wound healing, offering insights into potential therapeutic targets.
Collapse
Affiliation(s)
- Miaonan Liu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoxuan Liu
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jingchi Zhang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shaocong Liang
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yan Gong
- Department of Burns and Wound Repairing, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shengjun Shi
- Department of Burns and Wound Repairing, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Xiaopeng Yuan
- Department of Laboratory Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China; Department of Laboratory Medicine, Shenzhen People's Hospital, The First Affiliated Hospital, Southern University of Science and Technology; The Second Clinical Medical College, Jinan University; Shenzhen 518020, Guangdong China..
| |
Collapse
|
21
|
Yue L, Chen F, Liu X, Wu C, Wang J, Lai J, Zhu H. miR-23b-3p Ameliorates LPS-Induced Pulmonary Fibrosis by Inhibiting EndMT via DPP4 Inhibition. Mol Biotechnol 2025; 67:175-186. [PMID: 38150089 PMCID: PMC11698795 DOI: 10.1007/s12033-023-00992-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 11/13/2023] [Indexed: 12/28/2023]
Abstract
Acute respiratory distress syndrome is a disease triggered by severe pulmonary and systemic inflammation that may lead to fibrosis and the decline of lung function. Lung capillary endothelial-to-mesenchymal transition (EndMT) is one of the primary sources of fibroblasts in pulmonary fibrosis. The role of miRNAs as molecular markers of pulmonary fibrosis, and miRNAs as nucleic acid drugs has attracted increasing attention. To mock EndMT process, Human pulmonary microvascular endothelial cells (HPMECs) were induced with lipopolysaccharide (LPS). Similarly, LPS treatment was used to generate a mouse model of LPS-induced EndMT and pulmonary fibrosis. LPS-induced EndMT in HPMECs resulted in a significant reduction of miR-23b-3p. miR-23b-3p inhibited the interstitial transition of HPMECs, and miR-23b-3p could mediate this process via inhibiting dipeptidyl peptidase-4 (DPP4). Dual-luciferase assays confirmed the regulatory mechanism of miR-23b-3p. In our mouse model of LPS-induced pulmonary fibrosis, miR-23b-3p and a DPP4 inhibitor (sitagliptin) individually alleviated LPS-induced EndMT progression and pulmonary fibrosis, and their combined use achieved the strongest remission effect. To sum up, miR-23b-3p alleviates EndMT in pulmonary fibrosis by inhibiting the expression of DPP4.
Collapse
Affiliation(s)
- Linlin Yue
- Department of Intensive Care Unit, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Feng Chen
- Department of Pediatric Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Xin Liu
- Department of Intensive Care Unit, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Chaoyu Wu
- Department of Intensive Care Unit, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Jie Wang
- Department of Intensive Care Unit, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Jiying Lai
- Department of Intensive Care Unit, The First Affiliated Hospital of Gannan Medical University, Ganzhou, 341000, Jiangxi, People's Republic of China
| | - Hongquan Zhu
- The First Affiliated Hospital of Gannan Medical University, 128 Jinling Avenue, Zhanggong District, Ganzhou, 341000, Jiangxi, People's Republic of China.
| |
Collapse
|
22
|
Kataria S, Rana I, Badarinath K, Zaarour RF, Kansagara G, Ahmed S, Rizvi A, Saha D, Dam B, Dutta A, Zirmire RK, Hajam EY, Kumar P, Gulyani A, Jamora C. Mindin regulates fibroblast subpopulations through distinct Src family kinases during fibrogenesis. JCI Insight 2024; 10:e173071. [PMID: 39739417 PMCID: PMC11948575 DOI: 10.1172/jci.insight.173071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 12/19/2024] [Indexed: 01/02/2025] Open
Abstract
Fibrosis results from excessive extracellular matrix (ECM) deposition, which causes tissue stiffening and organ dysfunction. Activated fibroblasts, central to fibrosis, exhibit increased migration, proliferation, contraction, and ECM production. However, it remains unclear if the same fibroblast performs all of the processes that fall under the umbrella term of "activation." Owing to fibroblast heterogeneity in connective tissues, subpopulations with specific functions may operate under distinct regulatory controls. Using a transgenic mouse model of skin fibrosis, we found that Mindin (also known as spondin-2), secreted by Snail-transgenic keratinocytes, differentially regulates fibroblast subpopulations. Mindin promotes migration and inflammatory gene expression in SCA1+ dermal fibroblasts via Fyn kinase. In contrast, it enhances contractility and collagen production in papillary CD26+ fibroblasts through c-Src signaling. Moreover, in the context of the fibrotic microenvironment of the tumor stroma, we found that differential responses of resident fibroblast subpopulations to Mindin extend to the generation of functionally heterogeneous cancer-associated fibroblasts. This study identifies Mindin as a key orchestrator of dermal fibroblast heterogeneity, reshaping cellular dynamics and signaling diversity in the complex landscapes of skin fibrosis and cancer.
Collapse
Affiliation(s)
- Sunny Kataria
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, India
- National Centre for Biological Sciences, Gandhi Krishi Vigyan Kendra Post, Bangalore, Karnataka, India
| | - Isha Rana
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Thanjavur, Tamil Nadu, India
| | - Krithika Badarinath
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- National Centre for Biological Sciences, Gandhi Krishi Vigyan Kendra Post, Bangalore, Karnataka, India
| | - Rania F. Zaarour
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Gaurav Kansagara
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, India
| | - Sultan Ahmed
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Abrar Rizvi
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Dyuti Saha
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, India
| | - Binita Dam
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Manipal Academy of Higher Education, Manipal, India
| | - Abhik Dutta
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Thanjavur, Tamil Nadu, India
| | - Ravindra K. Zirmire
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Thanjavur, Tamil Nadu, India
| | - Edries Yousaf Hajam
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Shanmugha Arts, Science, Technology and Research Academy (SASTRA) University, Thanjavur, Tamil Nadu, India
| | - Pankaj Kumar
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
| | - Akash Gulyani
- Integrative Chemical Biology, inStem, Bangalore, Karnataka, India
| | - Colin Jamora
- IFOM-inStem Joint Research Laboratory, Centre for Inflammation and Tissue Homeostasis, Institute for Stem Cell Science and Regenerative Medicine (inStem), Bangalore, Karnataka, India
- Department of Life Sciences, Shiv Nadar Institution of Eminence, Gautam Buddha Nagar, India
| |
Collapse
|
23
|
Huang Y, Wang N, Xing H, Tian J, Zhang D, Gao D, Hsia HC, Lu J, Raredon MSB, Kyriakides TR. Alteration of skin fibroblast steady state contributes to healing outcomes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.06.627278. [PMID: 39713414 PMCID: PMC11661132 DOI: 10.1101/2024.12.06.627278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Fibroblasts display complex functions associated with distinct gene expression profiles that influence matrix production and cell communications and the autonomy of tissue development and repair. Thrombospondin-2 (TSP-2), produced by fibroblasts, is a potent angiogenesis inhibitor and negatively associated with tissue repair. Single-cell (sc) sequencing analysis on WT and TSP2KO skin fibroblasts demonstrate distinct cell heterogeneity. Specifically, we found an enrichment of Sox10+ multipotent progenitor cells, identified as Schwann precursor cells, in TSP2KO fibroblasts, while fibrosis-related subpopulations decreased. Immunostaining of tissue and cells validated the increase of this Sox10+ population in KO fibroblasts. Furthermore, in silico analysis suggested enhanced pro-survival signaling, including WNT, TGF-β, and PDGF-β, alongside a reduced BMP4 response. Additionally, the creation of two TSP2KO NIH3T3 cell lines using the CRISPR/Cas9 technique allowed functional and signaling validation in a less complex system. Moreover, KO 3T3 cells exhibited enhanced migration and proliferation, with elevated levels of pro-regenerative molecules including TGF-β3 and Wnt4, and enrichment of nuclear β-catenin. These functional and molecular alterations likely contribute to improved healing and increased neurogenesis in TSP2-deficient wounds. Overall, our findings describe the heterogeneity of dermal fibroblasts and identify pro-regenerative features of TSP2KO fibroblasts.
Collapse
Affiliation(s)
- Yaqing Huang
- Department of Pathology, Yale University, New Haven, CT 06520, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA
| | - Nuoya Wang
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA
- Department of Anesthesiology, Yale University, New Haven, CT 06520, USA
| | - Hao Xing
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA
| | - Jingru Tian
- Department of Genetics, Yale University, New Haven, CT 06520, USA
| | - Dingyao Zhang
- Department of Genetics, Yale University, New Haven, CT 06520, USA
| | - Daqian Gao
- Plastic & Reconstructive Surgery, Yale University, New Haven, CT 06520, USA
| | - Henry C. Hsia
- Plastic & Reconstructive Surgery, Yale University, New Haven, CT 06520, USA
| | - Jun Lu
- Department of Genetics, Yale University, New Haven, CT 06520, USA
| | - Micha Sam Brickman Raredon
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA
- Department of Anesthesiology, Yale University, New Haven, CT 06520, USA
| | - Themis R. Kyriakides
- Department of Pathology, Yale University, New Haven, CT 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT 06520, USA
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
24
|
Fujiwara H. Dynamic duo: Cell-extracellular matrix interactions in hair follicle development and regeneration. Dev Biol 2024; 516:20-34. [PMID: 39059679 DOI: 10.1016/j.ydbio.2024.07.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 06/20/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
Ectodermal organs, such as hair follicles, originate from simple epithelial and mesenchymal sheets through a complex developmental process driven by interactions between these cell types. This process involves dermal condensation, placode formation, bud morphogenesis, and organogenesis, and all of these processes require intricate interactions among various tissues. Recent research has emphasized the crucial role of reciprocal and dynamic interactions between cells and the extracellular matrix (ECM), referred to as the "dynamic duo", in the development of ectodermal organs. These interactions provide spatially and temporally changing biophysical and biochemical cues within tissues. Using the hair follicle as an example, this review highlights two types of cell-ECM adhesion units-focal adhesion-type and hemidesmosome-type adhesion units-that facilitate communication between epithelial and mesenchymal cells. This review further explores how these adhesion units, along with other cell-ECM interactions, evolve during hair follicle development and regeneration, underscoring their importance in guiding both developmental and regenerative processes.
Collapse
|
25
|
Cao X, Wu X, Zhang Y, Qian X, Sun W, Zhao Y. Emerging biomedical technologies for scarless wound healing. Bioact Mater 2024; 42:449-477. [PMID: 39308549 PMCID: PMC11415838 DOI: 10.1016/j.bioactmat.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/16/2024] [Accepted: 09/01/2024] [Indexed: 09/25/2024] Open
Abstract
Complete wound healing without scar formation has attracted increasing attention, prompting the development of various strategies to address this challenge. In clinical settings, there is a growing preference for emerging biomedical technologies that effectively manage fibrosis following skin injury, as they provide high efficacy, cost-effectiveness, and minimal side effects compared to invasive and costly surgical techniques. This review gives an overview of the latest developments in advanced biomedical technologies for scarless wound management. We first introduce the wound healing process and key mechanisms involved in scar formation. Subsequently, we explore common strategies for wound treatment, including their fabrication methods, superior performance and the latest research developments in this field. We then shift our focus to emerging biomedical technologies for scarless wound healing, detailing the mechanism of action, unique properties, and advanced practical applications of various biomedical technology-based therapies, such as cell therapy, drug therapy, biomaterial therapy, and synergistic therapy. Finally, we critically assess the shortcomings and potential applications of these biomedical technologies and therapeutic methods in the realm of scar treatment.
Collapse
Affiliation(s)
- Xinyue Cao
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiangyi Wu
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Yuanyuan Zhang
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Xiaoyun Qian
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yuanjin Zhao
- Department of Otolaryngology Head and Neck Surgery, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518071, China
| |
Collapse
|
26
|
Wyetzner RH, Segal EX, Jussila AR, Atit RP. Topographical changes in extracellular matrix during skin fibrosis and recovery can be evaluated using automated image analysis algorithms. FEBS Lett 2024; 598:2995-3004. [PMID: 39054263 PMCID: PMC11665952 DOI: 10.1002/1873-3468.14987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 06/19/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
Skin fibrosis is characterized by fibroblast activation and intradermal fat loss, resulting in excess deposition and remodeling of dermal extracellular matrix (ECM). The topography of the dominant ECM proteins, such as collagens, can indicate skin stiffness and remains understudied in evaluating fibrotic skin. Here, we adapted two different unbiased image analysis algorithms to define collagen topography and alignment in a genetically inducible and reversible Wnt activation fibrosis model. We demonstrated that Wnt-activated fibrotic skin has altered collagen fiber characteristics and a loss of collagen alignment, which were restored in the reversible model. This study highlights how unbiased algorithms can be used to analyze ECM topography, providing novel avenues to evaluate fibrotic skin onset, recovery, and treatment.
Collapse
Affiliation(s)
| | - Ella X. Segal
- Department of BiologyCase Western Reserve UniversityClevelandOHUSA
| | - Anna R. Jussila
- Department of BiologyCase Western Reserve UniversityClevelandOHUSA
| | - Radhika P. Atit
- Department of BiologyCase Western Reserve UniversityClevelandOHUSA
- Department of Genetics and Genome SciencesCase Western Reserve UniversityClevelandOHUSA
- Department of DermatologyCase Western Reserve UniversityClevelandOHUSA
| |
Collapse
|
27
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Kleb SS, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Fibroblast-Mediated Macrophage Recruitment Supports Acute Wound Healing. J Invest Dermatol 2024:S0022-202X(24)02956-7. [PMID: 39581458 DOI: 10.1016/j.jid.2024.10.609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/26/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single-nuclei RNA sequencing, we defined changes in gene expression associated with inflammation 1 day after wounding in mouse skin. Compared with those in keratinocytes and myeloid cells, we detected enriched expression of proinflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL-33, compared with SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound-bed macrophages and monocytes during injury-induced inflammation, with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M Amuso
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - MaryEllen R Haas
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Paula O Cooper
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Sana Hafiz
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Shatha Salameh
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Miguel F Mazumder
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Violet Josephson
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Sarah S Kleb
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Khatereh Khorsandi
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Anelia Horvath
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, District of Columbia, USA
| | - Brett A Shook
- The Department of Biochemistry & Molecular Medicine, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA; The Department of Dermatology, School of Medicine & Health Sciences, The George Washington University, Washington, District of Columbia, USA.
| |
Collapse
|
28
|
Kim WS, Prasongyuenyong K, Ko A, Debnath R, Chen Z, Zhou JX, Shaaf E, Ko KI. ICAM1 + gingival fibroblasts modulate periodontal inflammation to mitigate bone loss. Front Immunol 2024; 15:1484483. [PMID: 39650645 PMCID: PMC11621011 DOI: 10.3389/fimmu.2024.1484483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 11/04/2024] [Indexed: 12/11/2024] Open
Abstract
Tissue-resident fibroblasts are heterogeneous and provide an endogenous source of cytokines that regulate immunologic events in many osteolytic diseases. Identifying distinct inflammatory fibroblast subsets and conducting mechanistic in vivo studies are critical for understanding disease pathogenesis and precision therapeutics, which is poorly explored in periodontitis. Here, we surveyed published single-cell datasets for fibroblast-specific analysis and show that Intercellular Adhesion Molecule-1 (ICAM1) expression selectively defines a fibroblast subset that exhibits an inflammatory transcriptional profile associated with nuclear factor-κB (NF-κB) pathway. ICAM1+ fibroblasts expand in both human periodontitis and murine ligature-induced periodontitis model, which have upregulated expression of CCL2 and CXCL1 compared to other fibroblast populations. Using a mouse model to selectively target gingival stromal cells, we further show that disruption of an inflammatory pathway by inhibiting transcriptional activity of NF-κB in these cells accelerated periodontal bone loss. Mechanistically, this was linked to a reduction of CCL2 expression by the ICAM1+ fibroblasts, leading to impaired macrophage recruitment and efferocytosis that was associated with persistent neutrophilic inflammation. These results may have a significant therapeutic implication as ICAM1+ gingival fibroblasts exert a protective response by regulating innate immune responses that are needed for the controlled inflammatory events in early stages of periodontitis.
Collapse
Affiliation(s)
- William S. Kim
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kawintip Prasongyuenyong
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hatyai, Songkhla, Thailand
| | - Annette Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Rahul Debnath
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Zhaoxu Chen
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Jonathan X. Zhou
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Emon Shaaf
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Kang I. Ko
- Department of Periodontics, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
29
|
Li X, Zhang KW, Zhang ZY, Wu JJ, Yuan ZD, Yuan FL, Chen J. Inhibiting dipeptidyl peptidase 4 positive fibroblasts using zinc sulfide cellulose nanofiber scaffolds to achieve scarless healing. Int J Biol Macromol 2024; 282:137525. [PMID: 39537047 DOI: 10.1016/j.ijbiomac.2024.137525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Wound regeneration with integral function and cutaneous appendages remains challenging in wound dressing applications. Cellulose nanofibers (CNF) exhibit remarkable characteristics in wound dressing applications; however, their utility in the wound healing process is limited by insufficient scar inhibition and regenerative healing. Herein, inspired by fibroblast heterogeneity mediating wound healing and skin regeneration, we developed a CNF scaffold designed to block Dipeptidyl Peptidase 4 positive (DPP4+) fibroblasts for regenerative healing. CNF encapsulated sitagliptin (SITA) and zinc sulfide nanoparticles (NZnS), namely CNF@SITA@NZnS, to fabricate a novel biomaterial for scar reduction and regenerative healing. The scaffold promoted scarless healing and hair follicle regeneration in rats. In vivo experiments, the wounds in the scaffold showed less skin fibrosis, a better collagen ratio and more new hair follicles. In vitro experiments showed that the scaffold material promoted scarless healing, possibly by inhibiting the secretion of extracellular matrix and fibroblast-to-myofibroblast conversion. The promotion of hair follicle regeneration by the scaffold material may be due to promotion of the migration and proliferation of human hair follicle papilla cells. RNA sequencing is performed to explore the underlying mechanisms, which can activate ECM-receptor interaction pathway in favor of the wound healing process. The inhibiting effect of CNF@SITA@NZnS scaffold on DPP4+ fibroblasts can be a potential target to reduce scarring and promote skin regeneration.
Collapse
Affiliation(s)
- Xia Li
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Kai-Wen Zhang
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Yu Zhang
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China
| | - Feng-Lai Yuan
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China.
| | - Jinghua Chen
- Institute of Integrated Chinese and Western Medicine, The Hospital Affiliated to Jiangnan University, Wuxi, Jiangsu 214041, China; Key Laboratory of Carbohydrate Chemistry and Biotechnology, Ministry of Education, School of Pharmaceutical Sciences, Jiangnan University, Wuxi 214122, China.
| |
Collapse
|
30
|
Forsthuber A, Aschenbrenner B, Korosec A, Jacob T, Annusver K, Krajic N, Kholodniuk D, Frech S, Zhu S, Purkhauser K, Lipp K, Werner F, Nguyen V, Griss J, Bauer W, Soler Cardona A, Weber B, Weninger W, Gesslbauer B, Staud C, Nedomansky J, Radtke C, Wagner SN, Petzelbauer P, Kasper M, Lichtenberger BM. Cancer-associated fibroblast subtypes modulate the tumor-immune microenvironment and are associated with skin cancer malignancy. Nat Commun 2024; 15:9678. [PMID: 39516494 PMCID: PMC11549091 DOI: 10.1038/s41467-024-53908-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) play a key role in cancer progression and treatment outcome. This study dissects the intra-tumoral diversity of CAFs in basal cell carcinoma, squamous cell carcinoma, and melanoma using molecular and spatial single-cell analysis. We identify three distinct CAF subtypes: myofibroblast-like RGS5+ CAFs, matrix CAFs (mCAFs), and immunomodulatory CAFs (iCAFs). Large-cohort tissue analysis reveals significant shifts in CAF subtype patterns with increasing malignancy. Two CAF subtypes exhibit immunomodulatory properties via different mechanisms. mCAFs sythesize extracellular matrix and may restrict T cell invasion in low-grade tumors via ensheathing tumor nests, while iCAFs are enriched in late-stage tumors, and express high levels of cytokines and chemokines to aid immune cell recruitment and activation. This is supported by the induction of an iCAF-like phenotype with immunomodulatory functions in primary healthy fibroblasts exposed to skin cancer cell secretomes. Thus, targeting CAF variants holds promise to enhance immunotherapy efficacy in skin cancers.
Collapse
Affiliation(s)
- Agnes Forsthuber
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bertram Aschenbrenner
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Ana Korosec
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Tina Jacob
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Karl Annusver
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Krajic
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Daria Kholodniuk
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sophie Frech
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Shaohua Zhu
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Kim Purkhauser
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Lipp
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Franziska Werner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Vy Nguyen
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Ana Soler Cardona
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Benedikt Weber
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Bernhard Gesslbauer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Clement Staud
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Jakob Nedomansky
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Christine Radtke
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan N Wagner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Maria Kasper
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden.
| | - Beate M Lichtenberger
- Skin and Endothelium Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
31
|
Wang HH, Korah M, Jing SL, Berry CE, Griffin MF, Longaker MT, Januszyk M. Characterizing Fibroblast Heterogeneity in Diabetic Wounds Through Single-Cell RNA-Sequencing. Biomedicines 2024; 12:2538. [PMID: 39595104 PMCID: PMC11592066 DOI: 10.3390/biomedicines12112538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024] Open
Abstract
Diabetes mellitus is an increasingly prevalent chronic metabolic disorder characterized by physiologic hyperglycemia that, when left uncontrolled, can lead to significant complications in multiple organs. Diabetic wounds are common in the general population, yet the underlying mechanism of impaired healing in such wounds remains unclear. Single-cell RNA-sequencing (scRNAseq) has recently emerged as a tool to study the gene expression of heterogeneous cell populations in skin wounds. Herein, we review the history of scRNAseq and its application to the study of diabetic wound healing, focusing on how innovations in single-cell sequencing have transformed strategies for fibroblast analysis. We summarize recent research on the role of fibroblasts in diabetic wound healing and describe the functional and cellular heterogeneity of skin fibroblasts. Moreover, we highlight future opportunities in diabetic wound fibroblast research, with a focus on characterizing distinct fibroblast subpopulations and their lineages. Leveraging single-cell technologies to explore fibroblast heterogeneity and the complex biology of diabetic wounds may reveal new therapeutic targets for improving wound healing and ultimately alleviate the clinical burden of chronic wounds.
Collapse
Affiliation(s)
- Helen H. Wang
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (H.H.W.); (M.K.); (S.L.J.); (C.E.B.); (M.F.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Maria Korah
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (H.H.W.); (M.K.); (S.L.J.); (C.E.B.); (M.F.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Serena L. Jing
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (H.H.W.); (M.K.); (S.L.J.); (C.E.B.); (M.F.G.)
| | - Charlotte E. Berry
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (H.H.W.); (M.K.); (S.L.J.); (C.E.B.); (M.F.G.)
| | - Michelle F. Griffin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (H.H.W.); (M.K.); (S.L.J.); (C.E.B.); (M.F.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (H.H.W.); (M.K.); (S.L.J.); (C.E.B.); (M.F.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA; (H.H.W.); (M.K.); (S.L.J.); (C.E.B.); (M.F.G.)
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
32
|
Ben-Shaanan TL, Knöpper K, Duan L, Liu R, Taglinao H, Xu Y, An J, Plikus MV, Cyster JG. Dermal TRPV1 innervations engage a macrophage- and fibroblast-containing pathway to activate hair growth in mice. Dev Cell 2024; 59:2818-2833.e7. [PMID: 38851191 PMCID: PMC11537826 DOI: 10.1016/j.devcel.2024.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 02/25/2024] [Accepted: 05/15/2024] [Indexed: 06/10/2024]
Abstract
Pain, detected by nociceptors, is an integral part of injury, yet whether and how it can impact tissue physiology and recovery remain understudied. Here, we applied chemogenetics in mice to locally activate dermal TRPV1 innervations in naive skin and found that it triggered new regenerative cycling by dormant hair follicles (HFs). This was preceded by rapid apoptosis of dermal macrophages, mediated by the neuropeptide calcitonin gene-related peptide (CGRP). TRPV1 activation also triggered a macrophage-dependent induction of osteopontin (Spp1)-expressing dermal fibroblasts. The neuropeptide CGRP and the extracellular matrix protein Spp1 were required for the nociceptor-triggered hair growth. Finally, we showed that epidermal abrasion injury induced Spp1-expressing dermal fibroblasts and hair growth via a TRPV1 neuron and CGRP-dependent mechanism. Collectively, these data demonstrated a role for TRPV1 nociceptors in orchestrating a macrophage and fibroblast-supported mechanism to promote hair growth and enabling the efficient restoration of this mechano- and thermo-protective barrier after wounding.
Collapse
Affiliation(s)
- Tamar L Ben-Shaanan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Konrad Knöpper
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lihui Duan
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ruiqi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Hanna Taglinao
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Ying Xu
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jinping An
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Jason G Cyster
- Howard Hughes Medical Institute and Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
33
|
Suzdaltseva Y, Selezneva A, Sergeev N, Kiselev SL. Initial WNT/β-Catenin or BMP Activation Modulates Inflammatory Response of Mesodermal Progenitors Derived from Human Induced Pluripotent Stem Cells. Cells 2024; 13:1820. [PMID: 39513926 PMCID: PMC11545028 DOI: 10.3390/cells13211820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 10/29/2024] [Accepted: 11/01/2024] [Indexed: 11/16/2024] Open
Abstract
Wound healing in adults largely depends on the functional state of multipotent mesenchymal stromal cells (MSCs). Human fetal tissues at the early stages of development are known to heal quickly with a full-quality restoration of the original structure. The differences in the molecular mechanisms that determine the functional activity of mesodermal cells in fetuses and adults remain virtually unknown. Using two independent human induced pluripotent stem cell (iPSC) lines, we examined the effects of the initial WNT and BMP activation on the differentiation of iPSCs via mesodermal progenitors into MSCs and highlighted the functions of these cells that are altered by the proinflammatory microenvironment. The WNT-induced mesoderm commitment of the iPSCs enhanced the expression of paraxial mesoderm (PM)-specific markers, while the BMP4-primed iPSCs exhibited increased levels of lateral mesoderm (LM)-specific genes. The inflammatory status and migration rate of the isogenic iPSC-derived mesoderm cells were assessed via gene expression analysis and scratch assay under the receptor-dependent activation of the proinflammatory IFN-γ or TNF-α signaling pathway. Reduced IDO1 and ICAM1 expression levels were detected in the WNT- and BMP-induced MSC progenitors compared to the isogenic MSCs in response to stimulation with IFN-γ and TNF-α. The WNT- and BMP-induced MSC progenitors exhibited a higher migration rate than isogenic MSCs upon IFN-γ exposure. The established isogenic cellular model will provide new opportunities to elucidate the mechanisms of regeneration and novel therapeutics for wound healing.
Collapse
Affiliation(s)
- Yulia Suzdaltseva
- Department of Epigenetics, Vavilov Institute of General Genetics of the Russian Academy of Sciences, 119333 Moscow, Russia
| | | | | | | |
Collapse
|
34
|
Zeyer KA, Bornert O, Nelea V, Bao X, Leytens A, Sharoyan S, Sengle G, Antonyan A, Bruckner-Tuderman L, Dengjel J, Reinhardt DP, Nyström A. Dipeptidyl Peptidase-4-Mediated Fibronectin Processing Evokes a Profibrotic Extracellular Matrix. J Invest Dermatol 2024; 144:2477-2487.e13. [PMID: 38570029 DOI: 10.1016/j.jid.2024.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/21/2024] [Accepted: 03/13/2024] [Indexed: 04/05/2024]
Abstract
Fibronectin serves as a platform to guide and facilitate deposition of collagen and fibrillin microfibrils. During development of fibrotic diseases, altered fibronectin deposition in the extracellular matrix (ECM) is generally an early event. After this, dysregulated organization of fibrillins and fibrillar collagens occurs. Because fibronectin is an essential orchestrator of healthy ECM, perturbation of its ECM-organizational capacity may be involved in development of fibrosis. To investigate this, we employed recessive dystrophic epidermolysis bullosa as a disease model with progressive, severe dermal fibrosis. Fibroblasts from donors with recessive dystrophic epidermolysis bullosa in 2-dimensional and 3-dimensional cultures displayed dysregulated fibronectin deposition. Our analyses revealed that increase of profibrotic dipeptidyl peptidase-4-positive fibroblasts coincides with altered fibronectin deposition. Dipeptidyl peptidase-4 inhibitors normalized deposition of fibronectin and subsequently of fibrillin microfibrils and collagen I. Intriguingly, proteomics and inhibitor and mutagenesis studies disclosed that dipeptidyl peptidase-4 modulates ECM deposition through the proteolysis of the fibronectin N-terminus. Our study provides mechanistic insights into the observed profibrotic activities of dipeptidyl peptidase-4 and extends the understanding of fibronectin-guided ECM assembly in health and disease.
Collapse
Affiliation(s)
- Karina A Zeyer
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Olivier Bornert
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Valentin Nelea
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Xinyi Bao
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany; Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Alexandre Leytens
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Svetlana Sharoyan
- H. Buniatian Institute of Biochemistry of Armenian NAS, Yerevan, Republic of Armenia
| | - Gerhard Sengle
- Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Medical Faculty, University of Cologne, Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics (CCMB), Cologne, Germany
| | - Alvard Antonyan
- H. Buniatian Institute of Biochemistry of Armenian NAS, Yerevan, Republic of Armenia
| | - Leena Bruckner-Tuderman
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany
| | - Jörn Dengjel
- Department of Biology, University of Fribourg, Fribourg, Switzerland
| | - Dieter P Reinhardt
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada; Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, Canada
| | - Alexander Nyström
- Department of Dermatology, Medical Faculty, Medical Center - University of Freiburg, Freiburg, Germany.
| |
Collapse
|
35
|
Olutoye OO, Eriksson E, Menchaca AD, Kirsner RS, Tanaka R, Schultz G, Weir D, Wagner TL, Fabia RB, Naik-Mathuria B, Liu PY, Ead JK, Adebayo T, Armstrong DG, McMullin N, Samora JB, Akingba AG. Management of Acute Wounds-Expert Panel Consensus Statement. Adv Wound Care (New Rochelle) 2024; 13:553-583. [PMID: 38618741 DOI: 10.1089/wound.2023.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2024] Open
Abstract
Significance: The Wound Healing Foundation recognized the need for consensus-based unbiased recommendations for the treatment of wounds. As a first step, a consensus on the treatment of chronic wounds was developed and published in 2022. The current publication on acute wounds represents the second step in this process. Acute wounds may result from any number of conditions, including burns, military and combat operations, and trauma to specific areas of the body. The management of acute wounds requires timely and evidence-driven intervention to achieve optimal clinical outcomes. This consensus statement provides the clinician with the necessary foundational approaches to the causes, diagnosis, and therapeutic management of acute wounds. Presented in a structured format, this is a useful guide for clinicians and learners in all patient care settings. Recent Advances: Recent advances in the management of acute wounds have centered on stabilization and treatment in the military and combat environment. Specifically, advancements in hemostasis, resuscitation, and the mitigation of infection risk through timely initiation of antibiotics and avoidance of high-pressure irrigation in contaminated soft tissue injury. Critical Issues: Critical issues include infection control, pain management, and the unique considerations for the management of acute wounds in pediatric patients. Future Directions: Future directions include new approaches to preventing the progression and conversion of burns through the use of specific gel formulations. Additionally, the use of three-dimensional bioprinting and photo-modulation for reconstruction is a promising area for continued discovery.
Collapse
Affiliation(s)
- Oluyinka O Olutoye
- Department of Surgery, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Elof Eriksson
- Harvard Medical School, Cambridge, Massachusetts, USA
| | - Alicia D Menchaca
- Department of General Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert S Kirsner
- University of Miami Hospital and Clinics Wound Center, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Rica Tanaka
- Juntendo University Graduate School of Medicine, Division of Regenerative Therapy, Department of Plastic & Reconstructive Surgery, Juntendo University Hospital Podiatry Center, Tokyo, Japan
| | - Greg Schultz
- University of Florida, Gainesville, Florida, USA
| | - Dot Weir
- Saratoga Hospital Center for Wound Healing and Hyperbaric Medicine, Saratoga Springs, New York, USA
| | - Tracey L Wagner
- Department of Pediatrics, Section of Emergency Medicine, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - Renata B Fabia
- Department of Surgery, Nationwide Children's Hospital, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | | | - Paul Y Liu
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - J Karim Ead
- Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Temitope Adebayo
- Temple University School of Podiatric Medicine, Philadelphia, Pennsylvania, USA
| | - David G Armstrong
- Warren Alpert Medical School of Brown University, Providence, Rhode Island, USA
| | - Neil McMullin
- Plastic Surgery Consultant to the Surgeon General of the Army, Evans Army Community Hospital, Fort Carson, Colorado, USA
| | - Julie Balch Samora
- Department of Orthopedics, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio, USA
| | - A George Akingba
- Department of Vascular Surgery, VA Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
36
|
Vorstandlechner V, Mildner M. Much More Than Just Diabetes: The Pivotal Role of Dipeptidyl-Peptidase 4 in Skin Fibrosis. J Invest Dermatol 2024; 144:2341-2343. [PMID: 38842990 DOI: 10.1016/j.jid.2024.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 04/18/2024] [Accepted: 04/26/2024] [Indexed: 10/25/2024]
Affiliation(s)
- Vera Vorstandlechner
- Department of Plastic, Reconstructive and Aesthetic Surgery, Medical University of Vienna, Vienna, Austria
| | - Michael Mildner
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|
37
|
Liu Z, Xian L, Li J, Zheng S, Xie H. Single-cell RNA sequencing analysis reveals the role of TXNDC5 in keloid formation. Cytojournal 2024; 21:40. [PMID: 39563670 PMCID: PMC11574684 DOI: 10.25259/cytojournal_58_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 06/05/2024] [Indexed: 11/21/2024] Open
Abstract
Objective Thioredoxin domain-containing protein 5 (TXNDC5) is associated with fibrosis in a variety of organs, but its mechanism of action in keloid is unclear. In this study, we aimed to investigate the mechanism of TXNDC5 in keloid. Material and Methods Single-cell RNA sequencing data of keloid and normal scar samples obtained from public databases were normalized and clustered using the Seurat package. Pathway enrich analysis was conducted using biological process enrichment analysis and Gene Set Enrichment Analysis (GSEA). In addition, TXNDC5 expression and its effects on migration and invasion of keloid fibroblasts (KFs) were validated based on cell function experiments. Results A total of five cell types were obtained. The KF clusters were further clustered into two fibroblast subtypes (Fibroblast cells 1 and Fibroblast cells 2). Biological process enrichment analysis showed that transforming growth factor beta (TGF-β) signaling pathway was enriched in the two fibroblast subtypes. GSEA analysis demonstrated that genes in TGF-β signaling pathway were mainly enriched in Fibroblast cells 1, and that genes involved in cell proliferation, migration, and the TGF-β signaling pathway were all high-expressed in fibroblast cells 1. TXNDC5 was positively correlated with fibroblast proliferation, migration and TGF-β signaling pathway, and AUCell score. The cellular experiment confirmed that the messenger RNA and protein levels of TXNDC5 and TGF-β1 were high-expressed in KFs cells (P<0.001), and that knockdown of TXNDC5 downregulated TGF-β1 expression and inhibited migration and invasion of KFs (P<0.0001). Conclusion Our study indicated that TGF-β signaling pathway was enriched in fibroblast cells, and TXNDC5 was positively correlated with proliferation, migration, and TGF-β signaling pathway. Cellular experiment demonstrated that knocking down TXNDC5 downregulated TGF-β1 expression, and suppressed migration and invasion of KFs. The current discoveries provided a new therapeutic strategy for the treatment of keloid.
Collapse
Affiliation(s)
- Zhikun Liu
- Department of Plastic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Lining Xian
- Department of Dermatology, The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianmin Li
- The First Clinical Medical School, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shudan Zheng
- Department of Plastic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Hongju Xie
- Department of Plastic Surgery, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|
38
|
Kohlhauser M, Mayrhofer M, Kamolz LP, Smolle C. An Update on Molecular Mechanisms of Scarring-A Narrative Review. Int J Mol Sci 2024; 25:11579. [PMID: 39519131 PMCID: PMC11546163 DOI: 10.3390/ijms252111579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Fibroblasts, the principal cellular mediators of connective tissue remodeling, play a crucial role in the formation of physiological and pathological scars. Understanding the intricate interplay between fibroblasts and other cellular and molecular components is essential for elucidating the underlying mechanisms driving scar formation. Hypertrophic scars, keloids and atrophic scars arise from dysregulated wound healing processes characterized by persistent inflammation, aberrant collagen deposition, and impaired extracellular matrix remodeling. Fibroblasts play a central role in the pathogenesis of such pathological scars, driving aberrant extracellular matrix remodeling, subsequently contributing to the formation of raised or depressed fibrotic lesions. The investigation of complex interactions between fibroblasts and the microenvironment is crucial for developing targeted therapeutic interventions aimed at modulating fibroblast activity and improving clinical outcomes in patients with pathological scars. Further research into the molecular pathways governing fibroblast behavior and their heterogeneity holds promise for advancing scar management strategies. This narrative review was performed to shed light on the mechanisms behind scar formation, with a special focus on the role of fibroblasts in the formation of different types of scars, providing insights into the pathophysiology of these conditions. Through the analysis of current knowledge, this review seeks to identify the key cellular and molecular mechanisms involved in fibroblast activation, collagen synthesis, and extracellular matrix remodeling in hypertrophic scar, keloid, or atrophic scar formation.
Collapse
Affiliation(s)
- Michael Kohlhauser
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Marcel Mayrhofer
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- COREMED—Centre for Regenerative Medicine and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
| | - Christian Smolle
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
39
|
Cheng XC, Tong WZ, Rui W, Feng Z, Shuai H, Zhe W. Single-cell sequencing technology in skin wound healing. BURNS & TRAUMA 2024; 12:tkae043. [PMID: 39445224 PMCID: PMC11497848 DOI: 10.1093/burnst/tkae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 01/24/2024] [Accepted: 01/29/2024] [Indexed: 10/25/2024]
Abstract
Skin wound healing is a complicated biological process that mainly occurs in response to injury, burns, or diabetic ulcers. It can also be triggered by other conditions such as dermatitis and melanoma-induced skin cancer. Delayed healing or non-healing after skin injury presents an important clinical issue; therefore, further explorations into the occurrence and development of wound healing at the cellular and molecular levels are necessary. Single-cell sequencing (SCS) is used to sequence and analyze the genetic messages of a single cell. Furthermore, SCS can accurately detect cell expression and gene sequences. The use of SCS technology has resulted in the emergence of new concepts pertaining to wound healing, making it an important tool for studying the relevant mechanisms and developing treatment strategies. This article discusses the application value of SCS technology, the effects of the latest research on skin wound healing, and the value of SCS technology in clinical applications. Using SCS to determine potential biomarkers for wound repair will serve to accelerate wound healing, reduce scar formation, optimize drug delivery, and facilitate personalized treatments.
Collapse
Affiliation(s)
- Xu Cheng Cheng
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Wang Zi Tong
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Wang Rui
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Zhao Feng
- Department of Stem Cells and Regenerative Medicine, China Medical University, No. 77 Puhe Road, Shenyang 110013, China
| | - Hou Shuai
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| | - Wang Zhe
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang, No. 36 Sanhao Street, Shenyang 110004, China
| |
Collapse
|
40
|
Li Y, Liu A, Wang J, Yang C, Lv K, He W, Wu J, Chen W. Suture-anchored cutaneous tension induces persistent hypertrophic scarring in a novel murine model. BURNS & TRAUMA 2024; 12:tkae051. [PMID: 39429643 PMCID: PMC11491161 DOI: 10.1093/burnst/tkae051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 12/31/2023] [Indexed: 10/22/2024]
Abstract
Background Hypertrophic scars cause impaired skin appearance and function, seriously affecting physical and mental health. Due to medical ethics and clinical accessibility, the collection of human scar specimens is frequently restricted, and the establishment of scar experimental animal models for scientific research is urgently needed. The four most commonly used animal models of hypertrophic scars have the following drawbacks: the rabbit ear model takes a long time to construct; the immunodeficient mouse hypertrophic scar model necessitates careful feeding and experimental operations; female Duroc pigs are expensive to purchase and maintain, and their large size makes it difficult to produce a significant number of models; and mouse scar models that rely on tension require special skin stretch devices, which are often damaged and shed, resulting in unstable model establishment. Our group overcame the shortcomings of previous scar animal models and created a new mouse model of hypertrophic scarring induced by suture anchoring at the wound edge. Methods We utilized suture anchoring of incisional wounds to impose directional tension throughout the healing process, restrain wound contraction, and generate granulation tissue, thus inducing scar formation. Dorsal paired incisions were generated in mice, with wound edges on the upper back sutured to the rib cage and the wound edges on the lower back relaxed as a control. Macroscopic manifestation, microscopic histological analysis, mRNA sequencing, bioinformatics, and in vitro cell assays were also conducted to verify the reliability of this method. Results Compared with those in relaxed controls, the fibrotic changes in stretched wounds were more profound. Histologically, the stretched scars were hypercellular, hypervascular, and hyperproliferative with disorganized extracellular matrix deposition, and displayed molecular hallmarks of hypertrophic fibrosis. In addition, the stretched scars exhibited transcriptional overlap with mechanically stretched scars, and human hypertrophic and keloid scars. Phosphatidylinositol 3-kinase-serine/threonine-protein kinase B signaling was implicated as a profibrotic mediator of apoptosis resistance under suture-induced tension. Conclusions This straightforward murine model successfully induces cardinal molecular and histological features of pathological hypertrophic scarring through localized suture tension to inhibit wound contraction. The model enables us to interrogate the mechanisms of tension-induced fibrosis and evaluate anti-scarring therapies.
Collapse
Affiliation(s)
- Yashu Li
- Department of Plastic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kangjiang Road, Yangpu District, Shanghai 200092, People's Republic of China
| | - Anqi Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan 410008, People's Republic of China
- Key Laboratory for the Genetics of Developmental and Neuropsychiatric Disorders (Ministry of Education), Bio-X Institutes, Shanghai Jiao Tong University, Shanghai 200240, People's Republic of China
| | - Jingyan Wang
- Department of Plastic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kangjiang Road, Yangpu District, Shanghai 200092, People's Republic of China
| | - Changsheng Yang
- Department of Plastic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kangjiang Road, Yangpu District, Shanghai 200092, People's Republic of China
| | - Kaiyang Lv
- Department of Plastic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kangjiang Road, Yangpu District, Shanghai 200092, People's Republic of China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Medical University), No. 30 Gaotanyan main Street, Shapingba District, Chongqing 400038, People's Republic of China
| | - Jun Wu
- Department of Burn and Plastic Surgery, Shenzhen Institute of Translational Medicine, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, No. 3002 Sungang Road, Futian District, Shenzhen 518035, People's Republic of China
| | - Wenbin Chen
- Department of Plastic Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, No. 1665 Kangjiang Road, Yangpu District, Shanghai 200092, People's Republic of China
| |
Collapse
|
41
|
Mangoura SA, Ahmed MA, Zaka AZ. New Insights into the Pleiotropic Actions of Dipeptidyl Peptidase-4 Inhibitors Beyond Glycaemic Control. TOUCHREVIEWS IN ENDOCRINOLOGY 2024; 20:19-29. [PMID: 39526061 PMCID: PMC11548370 DOI: 10.17925/ee.2024.20.2.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 05/23/2024] [Indexed: 11/16/2024]
Abstract
Dipeptidyl peptidase-4 (DPP-4) is a multifunctional serine ectopeptidase that cleaves and modifies a plethora of substrates, including regulatory peptides, cytokines and chemokines. DPP-4 is implicated in the regulation of immune response, viral entry, cellular adhesion, metastasis and chemotaxis. Regarding its numerous substrates and extensive expression inside the body, multitasking DPP-4 has been assumed to participate in different pathophysiological mechanisms. DPP-4 inhibitors or gliptins are increasingly used for the treatment of type 2 diabetes mellitus. Several reports from experimental and clinical studies have clarified that DPP-4 inhibitors exert many beneficial pleiotropic effects beyond glycaemic control, which are mediated by anti-inflammatory, anti-oxidant, anti-fibrotic and anti-apoptotic actions. The present review will highlight the most recent findings in the literature about these pleiotropic effects and the potential mechanisms underlying these benefits, with a specific focus on the potential effectiveness of DPP-4 inhibitors in coronavirus disease-19 and diabetic kidney disease.
Collapse
Affiliation(s)
- Safwat A Mangoura
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr, Cairo, Egypt
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Marwa A Ahmed
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| | - Andrew Z Zaka
- Department of Medical Pharmacology, Faculty of Medicine, Assiut University, Assiut, Egypt
| |
Collapse
|
42
|
Li DJ, Berry CE, Wan DC, Longaker MT. Clinical, mechanistic, and therapeutic landscape of cutaneous fibrosis. Sci Transl Med 2024; 16:eadn7871. [PMID: 39321265 DOI: 10.1126/scitranslmed.adn7871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 09/03/2024] [Indexed: 09/27/2024]
Abstract
When dysregulated, skin fibrosis can lead to a multitude of pathologies. We provide a framework for understanding the wide clinical spectrum, mechanisms, and management of cutaneous fibrosis encompassing a variety of matrix disorders, fibrohistiocytic neoplasms, injury-induced scarring, and autoimmune scleroses. Underlying such entities are common mechanistic pathways that leverage morphogenic signaling, immune activation, and mechanotransduction to modulate fibroblast function. In light of the limited array of available treatments for cutaneous fibrosis, scientific insights have opened new therapeutic and investigative avenues for conditions that still lack effective interventions.
Collapse
Affiliation(s)
- Dayan J Li
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Department of Dermatology, Stanford University School of Medicine, Redwood City, CA 94063, USA
| | - Charlotte E Berry
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Derrick C Wan
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Michael T Longaker
- Department of Surgery, Division of Plastic and Reconstructive Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| |
Collapse
|
43
|
Gawronska-Kozak B, Machcinska-Zielinska S, Walendzik K, Kopcewicz M, Pääkkönen M, Wisniewska J. Hypoxia and Foxn1 alter the proteomic signature of dermal fibroblasts to redirect scarless wound healing to scar-forming skin wound healing in Foxn1 -/- mice. BMC Biol 2024; 22:193. [PMID: 39256768 PMCID: PMC11389453 DOI: 10.1186/s12915-024-01990-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 08/22/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Foxn1-/- deficient mice are a rare model of regenerative skin wound healing among mammals. In wounded skin, the transcription factor Foxn1 interacting with hypoxia-regulated factors affects re-epithelialization, epithelial-mesenchymal transition (EMT) and dermal white adipose tissue (dWAT) reestablishment and is thus a factor regulating scar-forming/reparative healing. Here, we hypothesized that transcriptional crosstalk between Foxn1 and Hif-1α controls the switch from scarless (regenerative) to scar-present (reparative) skin wound healing. To verify this hypothesis, we examined (i) the effect of hypoxia/normoxia and Foxn1 signalling on the proteomic signature of Foxn1-/- (regenerative) dermal fibroblasts (DFs) and then (ii) explored the effect of Hif-1α or Foxn1/Hif-1α introduced by a lentiviral (LV) delivery vector to injured skin of regenerative Foxn1-/- mice with particular attention to the remodelling phase of healing. RESULTS We showed that hypoxic conditions and Foxn1 stimulation modified the proteome of Foxn1-/- DFs. Hypoxic conditions upregulated DF protein profiles, particularly those related to extracellular matrix (ECM) composition: plasminogen activator inhibitor-1 (Pai-1), Sdc4, Plod2, Plod1, Lox, Loxl2, Itga2, Vldlr, Ftl1, Vegfa, Hmox1, Fth1, and F3. We found that Pai-1 was stimulated by hypoxic conditions in regenerative Foxn1-/- DFs but was released by DFs to the culture media exclusively upon hypoxia and Foxn1 stimulation. We also found higher levels of Pai-1 protein in DFs isolated from Foxn1+/+ mice (reparative/scar-forming) than in DFs isolated from Foxn1-/- (regenerative/scarless) mice and triggered by injury increase in Foxn1 and Pai-1 protein in the skin of mice with active Foxn1 (Foxn1+/+ mice). Then, we demonstrated that the introduction of Foxn1 and Hif-1α via lentiviral injection into the wounded skin of regenerative Foxn1-/- mice activates reparative/scar-forming healing by increasing the wounded skin area and decreasing hyaluronic acid deposition and the collagen type III to I ratio. We also identified a stimulatory effect of LV-Foxn1 + LV-Hif-1α injection in the wounded skin of Foxn1-/- mice on Pai-1 protein levels. CONCLUSIONS The present data highlight the effect of hypoxia and Foxn1 on the protein profile and functionality of regenerative Foxn1-/- DFs and demonstrate that the introduction of Foxn1 and Hif-1α into the wounded skin of regenerative Foxn1-/- mice activates reparative/scar-forming healing.
Collapse
Affiliation(s)
- Barbara Gawronska-Kozak
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland.
| | - Sylwia Machcinska-Zielinska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| | - Katarzyna Walendzik
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| | - Marta Kopcewicz
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| | - Mirva Pääkkönen
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Tykistökatu 6, Biocity 5 Floor, 20520, Turku, Finland
| | - Joanna Wisniewska
- Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Ul. Tuwima 10, 10-748, Olsztyn, Poland
| |
Collapse
|
44
|
Poss KD, Tanaka EM. Hallmarks of regeneration. Cell Stem Cell 2024; 31:1244-1261. [PMID: 39163854 PMCID: PMC11410156 DOI: 10.1016/j.stem.2024.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/12/2024] [Accepted: 07/24/2024] [Indexed: 08/22/2024]
Abstract
Regeneration is a heroic biological process that restores tissue architecture and function in the face of day-to-day cell loss or the aftershock of injury. Capacities and mechanisms for regeneration can vary widely among species, organs, and injury contexts. Here, we describe "hallmarks" of regeneration found in diverse settings of the animal kingdom, including activation of a cell source, initiation of regenerative programs in the source, interplay with supporting cell types, and control of tissue size and function. We discuss these hallmarks with an eye toward major challenges and applications of regenerative biology.
Collapse
Affiliation(s)
- Kenneth D Poss
- Duke Regeneration Center and Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA.
| | - Elly M Tanaka
- Institute of Molecular Biotechnology (IMBA), Austrian Academy of Sciences, Vienna Biocenter (VBC), 1030 Vienna, Austria.
| |
Collapse
|
45
|
Reiter L, Niehoff N, Weiland D, Helbig D, Eming SA, Krieg T, Etich J, Brachvogel B, Wiesner RJ, Knuever J. Mitochondrial DNA mutations attenuate Bleomycin-induced dermal fibrosis by inhibiting differentiation into myofibroblasts. Matrix Biol 2024; 132:72-86. [PMID: 39009171 DOI: 10.1016/j.matbio.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/17/2024]
Abstract
Post-mitotic, non-proliferative dermal fibroblasts have crucial functions in maintenance and restoration of tissue homeostasis. They are involved in essential processes such as wound healing, pigmentation and hair growth, but also tumor development and aging-associated diseases. These processes are energetically highly demanding and error prone when mitochondrial damage occurs. However, mitochondrial function in fibroblasts and the influence of mitochondrial dysfunction on fibroblast-specific demands are still unclear. To address these questions, we created a mouse model in which accelerated cell-specific mitochondrial DNA (mtDNA) damage accumulates. We crossed mice carrying a dominant-negative mutant of the mitochondrial replicative helicase Twinkle (RosaSTOP system) with mice that express fibroblast-specific Cre Recombinase (Collagen1A2 CreERT) which can be activated by Tamoxifen (TwinkleFIBRO). Thus, we are able to induce mtDNA deletions and duplications in specific cells, a process which resembles the physiological aging process in humans, where this damage accumulates in all tissues. Upon proliferation in vitro, Tamoxifen induced Twinkle fibroblasts deplete most of their mitochondrial DNA which, although not disturbing the stoichiometry of the respiratory chain complexes, leads to reduced ROS production and mitochondrial membrane potential as well as an anti-inflammatory and anti-fibrotic profile of the cells. In Sodium Azide treated wildtype fibroblasts, without a functioning respiratory chain, we observe the opposite, a rather pro-inflammatory and pro-fibrotic signature. Upon accumulation of mitochondrial DNA mutations in vivo the TwinkleFIBRO mice are protected from fibrosis development induced by intradermal Bleomycin injections. This is due to dampened differentiation of the dermal fibroblasts into α-smooth-muscle-actin positive myofibroblasts in TwinkleFIBRO mice. We thus provide evidence for striking differences of the impact that mtDNA mutations have in contrast to blunted mitochondrial function in dermal fibroblasts and skin homeostasis. These data contribute to improved understanding of mitochondrial function and dysfunction in skin and provide mechanistic insight into potential targets to treat skin fibrosis in the future.
Collapse
Affiliation(s)
- Lena Reiter
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Nadine Niehoff
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany
| | - Daniela Weiland
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany; Laboratory of Experimental Immunology, Institute of Virology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Doris Helbig
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany
| | - Sabine A Eming
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Institute of Zoology, Developmental Biology Unit, University of Cologne, 50674 Cologne, Germany
| | - Thomas Krieg
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Translational Matrix Biology, University of Cologne, Medical Faculty, 50931 Cologne, Germany
| | - Julia Etich
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Bent Brachvogel
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931 Cologne, Germany
| | - Rudolf J Wiesner
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, University of Cologne, 50931 Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Jana Knuever
- Department of Dermatology and Venereology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50937 Cologne, Germany.
| |
Collapse
|
46
|
Zhu B, Liang L, Hui L, Lu Y. Exploring the role of dermal sheath cells in wound healing and fibrosis. Wound Repair Regen 2024; 32:735-745. [PMID: 39129718 DOI: 10.1111/wrr.13206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/13/2024]
Abstract
Wound healing is a complex, dynamic process involving the coordinated interaction of diverse cell types, growth factors, cytokines, and extracellular matrix components. Despite emerging evidence highlighting their importance, dermal sheath cells remain a largely overlooked aspect of wound healing research. This review explores the multifunctional roles of dermal sheath cells in various phases of wound healing, including modulating inflammation, aiding in proliferation, and contributing to extracellular matrix remodelling. Special attention is devoted to the paracrine effects of dermal sheath cells and their role in fibrosis, highlighting their potential in improving healing outcomes, especially in differentiating between hairy and non-hairy skin sites. By drawing connections between dermal sheath cells activity and wound healing outcomes, this work proposes new insights into the mechanisms of tissue regeneration and repair, marking a step forward in our understanding of wound healing processes.
Collapse
Affiliation(s)
- Bing Zhu
- Translational Medicine Engineering Research Center of Inner Mongolia Autonomous Region, affiliated with Baotou Central Hospital, Baotou, China
| | - Lu Liang
- Translational Medicine Engineering Research Center of Inner Mongolia Autonomous Region, affiliated with Baotou Central Hospital, Baotou, China
| | - Lihua Hui
- Burn Research Institute of Inner Mongolia Autonomous Region, affiliated with Inner Mongolia Baogang Hospital, Baotou, China
| | - Yaojun Lu
- Translational Medicine Engineering Research Center of Inner Mongolia Autonomous Region, affiliated with Baotou Central Hospital, Baotou, China
| |
Collapse
|
47
|
Amuso VM, Haas MR, Cooper PO, Chatterjee R, Hafiz S, Salameh S, Gohel C, Mazumder MF, Josephson V, Khorsandi K, Horvath A, Rahnavard A, Shook BA. Deep skin fibroblast-mediated macrophage recruitment supports acute wound healing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.09.607357. [PMID: 39149286 PMCID: PMC11326280 DOI: 10.1101/2024.08.09.607357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Epithelial and immune cells have long been appreciated for their contribution to the early immune response after injury; however, much less is known about the role of mesenchymal cells. Using single nuclei RNA-sequencing, we defined changes in gene expression associated with inflammation at 1-day post-wounding (dpw) in mouse skin. Compared to keratinocytes and myeloid cells, we detected enriched expression of pro-inflammatory genes in fibroblasts associated with deeper layers of the skin. In particular, SCA1+ fibroblasts were enriched for numerous chemokines, including CCL2, CCL7, and IL33 compared to SCA1- fibroblasts. Genetic deletion of Ccl2 in fibroblasts resulted in fewer wound bed macrophages and monocytes during injury-induced inflammation with reduced revascularization and re-epithelialization during the proliferation phase of healing. These findings highlight the important contribution of deep skin fibroblast-derived factors to injury-induced inflammation and the impact of immune cell dysregulation on subsequent tissue repair.
Collapse
Affiliation(s)
- Veronica M. Amuso
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - MaryEllen R. Haas
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Paula O. Cooper
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ranojoy Chatterjee
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Sana Hafiz
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Shatha Salameh
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Chiraag Gohel
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Miguel F. Mazumder
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Violet Josephson
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Khatereh Khorsandi
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Anelia Horvath
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, The George Washington University, Washington, DC 20052, USA
| | - Brett A. Shook
- Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
- Department of Dermatology, School of Medicine and Health Sciences, The George Washington University, Washington, DC 20052, USA
| |
Collapse
|
48
|
Rahman MA, Amirkhani A, Mempin M, Ahn SB, Deva AK, Baker MS, Vickery K, Hu H. The Low-Abundance Plasma Proteome Reveals Differentially Abundant Proteins Associated with Breast Implant Capsular Contracture: A Pilot Study. Proteomes 2024; 12:22. [PMID: 39189262 PMCID: PMC11348101 DOI: 10.3390/proteomes12030022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/28/2024] Open
Abstract
Capsular contracture (CC) is one of the most common postoperative complications associated with breast implant-associated infections. The mechanisms that lead to CC remain poorly understood. Plasma is an ideal biospecimen for early proteomics biomarker discovery. However, as high-abundance proteins mask signals from low-abundance proteins, identifying novel or specific proteins as biomarkers for a particular disease has been hampered. Here, we employed depletion of high-abundance plasma proteins followed by Tandem Mass Tag (TMT)-based quantitative proteomics to compare 10 healthy control patients against 10 breast implant CC patients. A total of 450 proteins were identified from these samples. Among them, 16 proteins were significantly differentially expressed in which 5 proteins were upregulated and 11 downregulated in breast implant CC patients compared to healthy controls. Gene Ontology enrichment analysis revealed that proteins related to cell, cellular processes and catalytic activity were highest in the cellular component, biological process, and molecular function categories, respectively. Further, pathway analysis revealed that inflammatory responses, focal adhesion, platelet activation, and complement and coagulation cascades were enriched pathways. The differentially abundant proteins from TMT-based quantitative proteomics have the potential to provide important information for future mechanistic studies and in the development of breast implant CC biomarkers.
Collapse
Affiliation(s)
- Md. Arifur Rahman
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | | | - Maria Mempin
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Seong Beom Ahn
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Anand K. Deva
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Mark S. Baker
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Karen Vickery
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
| | - Honghua Hu
- Macquarie Medical School, Macquarie University, Sydney, NSW 2109, Australia (S.B.A.); (A.K.D.); (M.S.B.); (K.V.)
- Jinhua Institute of Zhejiang University, Jinhua 321016, China
| |
Collapse
|
49
|
Peña OA, Martin P. Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol 2024; 25:599-616. [PMID: 38528155 DOI: 10.1038/s41580-024-00715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/27/2024]
Abstract
Wound healing is a complex process that involves the coordinated actions of many different tissues and cell lineages. It requires tight orchestration of cell migration, proliferation, matrix deposition and remodelling, alongside inflammation and angiogenesis. Whereas small skin wounds heal in days, larger injuries resulting from trauma, acute illness or major surgery can take several weeks to heal, generally leaving behind a fibrotic scar that can impact tissue function. Development of therapeutics to prevent scarring and successfully repair chronic wounds requires a fuller knowledge of the cellular and molecular mechanisms driving wound healing. In this Review, we discuss the current understanding of the different phases of wound healing, from clot formation through re-epithelialization, angiogenesis and subsequent scar deposition. We highlight the contribution of different cell types to skin repair, with emphasis on how both innate and adaptive immune cells in the wound inflammatory response influence classically studied wound cell lineages, including keratinocytes, fibroblasts and endothelial cells, but also some of the less-studied cell lineages such as adipocytes, melanocytes and cutaneous nerves. Finally, we discuss newer approaches and research directions that have the potential to further our understanding of the mechanisms underpinning tissue repair.
Collapse
Affiliation(s)
- Oscar A Peña
- School of Biochemistry, University of Bristol, Bristol, UK.
| | - Paul Martin
- School of Biochemistry, University of Bristol, Bristol, UK.
| |
Collapse
|
50
|
Gao J, Liu H, Li L, Guo C, Wang Z, Cheng M, Tan S, Chen L, Shi J, Wu H, Feng C, Yu G, Ding C. Comprehensive proteomic characterization of urethral stricture disease in the Chinese population. Front Mol Biosci 2024; 11:1401970. [PMID: 39130371 PMCID: PMC11310122 DOI: 10.3389/fmolb.2024.1401970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 06/26/2024] [Indexed: 08/13/2024] Open
Abstract
Background Male urethral stricture disease (USD) is predominantly characterized by scar formation. There are few effective therapeutic drugs, and comprehensive molecular characterizations of USD formation remain undefined. Methods The proteomic profiling of twelve scar tissues and five matched normal adjacent tissues (NATs). Proteomic analysis methods were applied to explore the molecular characterizations of USD formation, including uncovering mechanistic pathways and providing novel biomarkers for scar formation. Results Comparative proteomic analysis showed that the extracellular matrix (ECM) and complement cascade signaling were predominant in scar tissues. COL11A1 and CD248 significantly contributed to the accumulation of ECM components. Our study presented diverse molecular mechanisms of scar formation across different ages and suggested the potential effects of PXK in Age 1 (<45) patients. Furthermore, immune infiltration studies indicated the therapeutic potential of inhibiting the complement system (C4A, C4B) in Age 2 (≥45) patients, providing a potential clinical strategy for USD. Conclusion This study illustrated the pathogenesis of USD formation and the diverse characteristics of USD patients with different ages, enhancing our understanding of the disease's pathogenesis and providing a valuable resource for USD treatment.
Collapse
Affiliation(s)
- Jiangtao Gao
- Department of Urology, The First People’s Hospital of Zhengzhou, Henan, China
| | - Hui Liu
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- State Key Laboratory Cell Differentiation and Regulation, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis, (111 Project), College of Life Science, Henan Normal University, Xinxiang, China
| | - Lingling Li
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Chunmei Guo
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Zhiyong Wang
- Key Medical Laboratory of Stem Cell Transformation and Application, Department of Pathology, The First People’s Hospital of Zhengzhou, Henan, China
| | - Mengya Cheng
- Department of Urology, The First People’s Hospital of Zhengzhou, Henan, China
| | - Subei Tan
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Lu Chen
- Department of Urology, The First People’s Hospital of Zhengzhou, Henan, China
| | - Jijing Shi
- Key Medical Laboratory of Stem Cell Transformation and Application, Department of Pathology, The First People’s Hospital of Zhengzhou, Henan, China
| | - Hui Wu
- Department of Urology, The First People’s Hospital of Zhengzhou, Henan, China
| | - Chao Feng
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai, China
| | - Guoying Yu
- State Key Laboratory Cell Differentiation and Regulation, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis, (111 Project), College of Life Science, Henan Normal University, Xinxiang, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Human Phenome Institute, Fudan University, Shanghai, China
- State Key Laboratory Cell Differentiation and Regulation, Overseas Expertise Introduction Center for Discipline Innovation of Pulmonary Fibrosis, (111 Project), College of Life Science, Henan Normal University, Xinxiang, China
- Institute of Cancer Research, Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, China
| |
Collapse
|