1
|
Wang JH, Li M, Xie PF, Si JY, Feng ZJ, Tang CF, Li JM. Procyanidin C1 ameliorates aging-related skin fibrosis through targeting EGFR to inhibit TGFβ/SMAD pathway. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156787. [PMID: 40315640 DOI: 10.1016/j.phymed.2025.156787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/03/2025] [Accepted: 04/17/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Aging-related skin fibrosis (SF) is a complex condition with limited treatment options. Procyanidin C1 (PCC1), a natural polyphenolic compound with demonstrated senolytic activity, has emerged as a potential therapeutic agent for fibrotic disorders through its selective elimination of senescent cells. However, its therapeutic efficacy and mechanisms in aging-related SF remain unclear. PURPOSE This study aimed to investigate the mechanisms of PCC1 in aging-related SF. RESULTS In D-galactose-induced L929 cells, PCC1 treatment significantly attenuated the expression of both senescence-associated markers (IL-1β, P16, P21 and LMNB1) and fibrosis-related markers (α-SMA, LOXL2 and COL1). Network pharmacology and experimental validation (molecular docking, DARTS, CETSA, MST) identified EGFR as a primary target, with PCC1 directly binding to and inhibiting EGFR phosphorylation. Furthermore, PCC1 treatment effectively down-regulated TGFβ1 expression and suppressed SMAD2/3 phosphorylation in D-galactose-induced L929 cells. Notably, PCC1 blocked NSC228155-induced EGFR phosphorylation and inhibited ERK/MAPK, AKT/mTOR and TGFβ/SMAD pathway activation. In bleomycin-induced SF mice, PCC1 significantly attenuated epidermal hyperplasia, improved collagen structure, restored the collagen I/III ratio, and reduced EGFR phosphorylation along with TGFβ1 expression and SMAD2/3 phosphorylation. CONCLUSION This study elucidates that PCC1 exerts its anti-fibrotic effects through dual mechanisms: resistance to cellular senescence and modulation of fibroblast heterogeneity. By directly binding to EGFR and inhibiting its phosphorylation, PCC1 subsequently suppresses multiple downstream signaling cascades, ultimately ameliorating TGFβ/SMAD-mediated SF. These findings establish PCC1 as a promising therapeutic candidate for aging-related skin fibrosis, offering a novel approach through targeted EGFR inhibition and comprehensive pathway modulation.
Collapse
Affiliation(s)
- Jun-Han Wang
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Min Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Peng-Fei Xie
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Jia-Yao Si
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Zhen-Jie Feng
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China
| | - Chuan-Feng Tang
- State Key Laboratory of Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Jian-Mei Li
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing 210023, China.
| |
Collapse
|
2
|
Fakhouri SC, Zhu H, Li Y, Ronicke M, Rigau AR, Dees C, Konstantinidis L, Schmid R, Matei A, Eckstein M, Geppert C, Ludolph I, Kreuter A, Sticherling M, Berking C, Horch RE, Schett G, Distler JHW, Bergmann C. Disturbed Spatial WNT Activation-A Potential Driver of the Reticularized Skin Phenotype in Systemic Sclerosis. Arthritis Rheumatol 2025; 77:740-749. [PMID: 39722197 PMCID: PMC12123256 DOI: 10.1002/art.43094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
OBJECTIVE Little is known on the mechanisms necessary to maintain the physiologic adult human skin integrity. This study aims to quantitatively describe anatomic changes in systemic sclerosis (SSc)-skin compared with controls and investigate the underlying mechanisms. METHODS Skin morphology was histologically assessed in 23 patients with SSc, 18 controls, and 15 patients with hypertrophic scars. Spatial WNT/β-catenin-activation was analyzed by RNAscope and immunofluorescence staining. Enrichment of reticular marker genes in predefined fibroblast subpopulations was done using Gene Ontology (GO) enrichment and gene set enrichment analysis. RESULTS SSc skin showed a decrease in number (P < 0.0001/P = 0.0004), area (P < 0.0001), and height (P < 0.0001) of papillae compared with controls and hypertrophic scars, respectively. The expression of papillary/reticular marker genes shifted toward a reticular expression profile in SSc. On the level of previously defined fibroblast populations, the increase of reticular marker genes was particularly pronounced in the PI16+ and SFRP4+ populations (P < 0.0001, respectively). Mechanistically, the expression of the WNT/β-catenin target AXIN2 and the number of fibroblasts with nuclear β-catenin-staining-pattern increased in the papillary compared with the reticular dermis in healthy skin. This polarization was lost in SSc with a two-fold increase in β-catenin-positive fibroblasts and AXIN2-expressing fibroblasts throughout the dermis (P = 0.0095). Enrichment of genes related to WNT/β-catenin-regulation was found in the PI16+ population that also relocates from the reticular to the papillary dermis in SSc. CONCLUSION We demonstrate an association of the "reticularized" skin phenotype in SSc with a profound loss of physiologic spatial WNT/β-catenin-activation. Rescuing the spatial WNT/β-catenin-activation might help restore the physiologic skin organization in future therapeutic approaches of fibrosing disorders.
Collapse
Affiliation(s)
- Sara Chenguiti Fakhouri
- Department of Internal Medicine 3 ‐ Rheumatology and Clinical Immunology, Friedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Honglin Zhu
- Department of Rheumatology and Immunology, Xiangya HospitalCentral South UniversityChangshaHunanPeople's Republic of China
| | - Yi‐Nan Li
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Hiller Research CenterUniversity Hospital Düsseldorf, Medical Faculty of Heinrich Heine UniversityDüsseldorfGermany
| | - Moritz Ronicke
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Department of DermatologyFriedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Aleix Rius Rigau
- Department of Internal Medicine 3 ‐ Rheumatology and Clinical Immunology, Friedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Deutsches Zentrum Immuntherapie (DZI)FAU Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Clara Dees
- Department of Internal Medicine 3 ‐ Rheumatology and Clinical Immunology, Friedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Laura Konstantinidis
- Department of Internal Medicine 3 ‐ Rheumatology and Clinical Immunology, Friedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Deutsches Zentrum Immuntherapie (DZI)FAU Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Ralf Schmid
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital ErlangenFriedrich Alexander University Erlangen‐Nürnberg FAUErlangenGermany
| | - Alexandru‐Emil Matei
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMDFrankfurt am MainGermany
| | - Markus Eckstein
- Department of PathologyFriedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Carol Geppert
- Department of PathologyFriedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Ingo Ludolph
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital ErlangenFriedrich Alexander University Erlangen‐Nürnberg FAUErlangenGermany
| | - Alexander Kreuter
- Department of Dermatology, Venereology, and AllergologyHELIOS St. Elisabeth Hospital OberhausenOberhausenGermany
| | - Michael Sticherling
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Department of DermatologyFriedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Carola Berking
- Deutsches Zentrum Immuntherapie (DZI), FAU Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Department of DermatologyFriedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Raymund E. Horch
- Department of Plastic and Hand Surgery, Laboratory for Tissue Engineering and Regenerative Medicine, University Hospital ErlangenFriedrich Alexander University Erlangen‐Nürnberg FAUErlangenGermany
| | - Georg Schett
- Department of Internal Medicine 3 ‐ Rheumatology and Clinical Immunology, Friedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum Erlangen, Erlangen, Germany, and Deutsches Zentrum Immuntherapie (DZI)FAU Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| | - Jörg H. W. Distler
- Department of Rheumatology, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany, and Hiller Research Center, University Hospital Düsseldorf, Medical Faculty of Heinrich Heine University, Düsseldorf, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, and Fraunhofer Cluster of Excellence for Immune Mediated Diseases CIMDFrankfurt am MainGermany
| | - Christina Bergmann
- Department of Internal Medicine 3 ‐ Rheumatology and Clinical Immunology, Friedrich‐Alexander‐University (FAU) Erlangen‐Nürnberg and Uniklinikum ErlangenErlangenGermany
| |
Collapse
|
3
|
Nakamizo S, Kabashima K, Morimoto N, Fujimori H, Yan X, Kabashima K. Single-Cell RNA Sequencing Reveals Age-Related Changes in Epidermal Cell Populations and Interactions. J Invest Dermatol 2025; 145:1502-1505.e5. [PMID: 39756533 DOI: 10.1016/j.jid.2024.12.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 01/07/2025]
Affiliation(s)
- Satoshi Nakamizo
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; Alliance Laboratory for Advanced Medical Research, Kyoto University Graduate School of Medicine, Kyoto, Japan.
| | | | - Naoki Morimoto
- Department of Plastic and Reconstructive Surgery, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | - Kenji Kabashima
- Department of Dermatology, Kyoto University Graduate School of Medicine, Kyoto, Japan; A∗STAR Skin Research Labs (A∗SRL) and Skin Research Institute of Singapore (SRIS), Agency for Science, Technology, and Research (A∗STAR), Singapore, Singapore.
| |
Collapse
|
4
|
Wlaschek M, Maity P, Koroma AK, Geiger H, Singh K, Scharffetter-Kochanek K. Imbalanced redox dynamics induce fibroblast senescence leading to impaired stem cell pools and skin aging. Free Radic Biol Med 2025; 233:292-301. [PMID: 40154755 DOI: 10.1016/j.freeradbiomed.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/05/2025] [Accepted: 03/17/2025] [Indexed: 04/01/2025]
Abstract
Skin function depends on a meticulously regulated dynamic interaction of distinct skin compartments such as the epidermis and dermis. Adaptive responses at the molecular and cellular level are essential for these interactions - and if dysregulated - drive skin aging and other pathologies. After defining the role of redox homeodynamics in physiology and aging pathology, we focus on the redox distress-dependent aging of dermal fibroblasts including their progenitors. We here discuss the prime role of senescent fibroblasts in the control of their own endogenous niche and stem cell niches for epidermal stem cells, hair follicle stem cells, adipocyte precursors and muscle stem cells. We here review that redox imbalance induced reduction in Insulin-like Growth Factor-1 drives skin aging by the depletion of stem cell pools. This IGF-1 reduction is mediated via the redox-sensitive transcription factor JunB and also by the redox-dependent changes in sphingolipid-metabolism, among others. In addition, we will discuss the changes in the extracellular matrix of the skin affecting cellular senescence and the skin integrity and function in aging. The aim is a deeper understanding of the two main redox-dependent hubs such as JunB-induced depletion of IGF-1, and the sphingolipid-mediated remodeling of the cell membrane with its impact on IGF-1, fibroblast heterogeneity, function, senescence and plasticity in skin aging.
Collapse
Affiliation(s)
- Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany; Aging Research Institute (arc), Ulm University, Ulm, Germany
| | - Pallab Maity
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany; Aging Research Institute (arc), Ulm University, Ulm, Germany
| | - Albert Kallon Koroma
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany; Aging Research Institute (arc), Ulm University, Ulm, Germany
| | - Hartmut Geiger
- Aging Research Institute (arc), Ulm University, Ulm, Germany; Institute for Molecular Medicine and Stem Cell Aging, Ulm University, Ulm, Germany
| | - Karmveer Singh
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany; Aging Research Institute (arc), Ulm University, Ulm, Germany
| | - Karin Scharffetter-Kochanek
- Department of Dermatology and Allergic Diseases, Ulm University, Ulm, Germany; Aging Research Institute (arc), Ulm University, Ulm, Germany.
| |
Collapse
|
5
|
Guo H, Chen J, Jiang L. The antioxidant stress effect of granulin precursor in vitiligo. Sci Rep 2025; 15:18189. [PMID: 40415096 DOI: 10.1038/s41598-025-03486-7] [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: 12/10/2024] [Accepted: 05/20/2025] [Indexed: 05/27/2025] Open
Abstract
The imbalance of the skin redox system is regarded as a crucial factor contributing to the loss of melanocytes in vitiligo. However, it remains unclear whether alterations in signal transmission between melanocytes and other cells impact the homeostasis of the skin microenvironment. Hence, leveraging single-cell sequencing and microarray data, we investigated the role of cell-cell and ligand receptor interactions in the pathogenesis of vitiligo. We discovered that the granulin-sortilin 1 ligand-receptor serves as an essential bridge for communication between melanocytes and other skin cells in normal skin, yet it is significantly downregulated in vitiligo lesions. Enrichment analysis indicates that the activation of granulin-sortilin 1 ligand-receptor is closely associated with the regulation of oxidative stress. In vitro experiments have verified that progranulin, the protein encoded by the granulin gene, enhances the ability of melanocytes to resist cell death induced by reactive oxygen species and markedly upregulates the expression of nuclear factor erythroid 2-related factor 2 and Heme Oxygenase-1. Notably, this process can be impeded by the interaction inhibitor. Moreover, the expression of nuclear factor erythroid 2-related factor 2 might be linked to the transcription of transcription factor EB activated by progranulin. In conclusion, the granulin-sortilin 1 ligand-receptor can activate the intracellular antioxidant system to counteract melanocyte death. The impairment of the granulin-sortilin 1 ligand-receptor may be implicated in melanocyte loss in vitiligo.
Collapse
Affiliation(s)
- Haoran Guo
- Department of Dermatology, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Yuelu, Changsha, 410013, China
| | - Jing Chen
- Department of Dermatology, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Yuelu, Changsha, 410013, China
| | - Ling Jiang
- Department of Dermatology, Third Xiangya Hospital, Central South University, No.138 Tongzipo Road, Yuelu, Changsha, 410013, China.
| |
Collapse
|
6
|
Kraft M, Schoofs H, Petkova M, Andrade J, Grosso AR, Benedito R, De Roo AK, Boon LM, Vikkula M, Kapp FG, Hägerling R, Potente M, Mäkinen T. Angiopoietin-TIE2 feedforward circuit promotes PIK3CA-driven venous malformations. NATURE CARDIOVASCULAR RESEARCH 2025:10.1038/s44161-025-00655-9. [PMID: 40410415 DOI: 10.1038/s44161-025-00655-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/11/2025] [Indexed: 05/25/2025]
Abstract
Venous malformations (VMs) are vascular anomalies lacking curative treatments, often caused by somatic PIK3CA mutations that hyperactivate the PI3Kα-AKT-mTOR signaling pathway. Here, we identify a venous-specific signaling circuit driving disease progression, where excessive PI3Kα activity amplifies upstream TIE2 receptor signaling through autocrine and paracrine mechanisms. In Pik3caH1047R-driven VM mouse models, single-cell transcriptomics and lineage tracking revealed clonal expansion of mutant endothelial cells with a post-capillary venous phenotype, characterized by suppression of the AKT-inhibited FOXO1 and its target genes, including the TIE2 antagonist ANGPT2. An imbalance in TIE2 ligands, likely exacerbated by aberrant recruitment of smooth muscle cells producing the agonist ANGPT1, increased TIE2 activity in both mouse and human VMs. While mTOR blockade had limited effects on advanced VMs in mice, inhibiting TIE2 or ANGPT effectively suppressed their growth. These findings uncover a PI3K-FOXO1-ANGPT-TIE2 circuit as a core driver of PIK3CA-related VMs and highlight TIE2 as a promising therapeutic target.
Collapse
Affiliation(s)
- Marle Kraft
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Hans Schoofs
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Milena Petkova
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden
| | - Jorge Andrade
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ana Rita Grosso
- Associate Laboratory i4HB - Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Lisbon, Portugal
- UCIBIO - Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, Lisbon, Portugal
| | - Rui Benedito
- Molecular Genetics of Angiogenesis Group. Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - An-Katrien De Roo
- Center for Vascular Anomalies, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
- Department of Pathology, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Institute of Experimental and Clinical Research, UCLouvain, Brussels, Belgium
| | - Laurence M Boon
- Center for Vascular Anomalies, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
- Division of Plastic Surgery, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
- Laboratory of Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
| | - Miikka Vikkula
- Center for Vascular Anomalies, VASCERN VASCA European Reference Center, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
- Laboratory of Human Molecular Genetics, de Duve Institute, UCLouvain, Brussels, Belgium
- WELBIO Department, WEL Research Institute, Wavre, Belgium
| | - Friedrich G Kapp
- Department of Pediatric Hematology and Oncology, Children's Hospital, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, VASCERN VASCA European Reference Center, Freiburg, Germany
| | - René Hägerling
- Research Group 'Lymphovascular Medicine and Translational 3D-Histopathology', Institute of Medical and Human Genetics, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Center for Regenerative Therapies, Berlin, Germany
| | - Michael Potente
- Angiogenesis & Metabolism Laboratory, Center of Vascular Biomedicine, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Taija Mäkinen
- Uppsala University, Department of Immunology, Genetics and Pathology, Uppsala, Sweden.
- Translational Cancer Medicine Program and Department of Biochemistry and Developmental Biology, University of Helsinki, Helsinki, Finland.
- Wihuri Research Institute, Helsinki, Finland.
| |
Collapse
|
7
|
Hiratsuka T, Rognoni E. Protocol for detecting 3D dermal fibroblast cellular dynamics in mice using an intravital imaging technique. STAR Protoc 2025; 6:103801. [PMID: 40338747 DOI: 10.1016/j.xpro.2025.103801] [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: 12/23/2024] [Revised: 03/01/2025] [Accepted: 04/10/2025] [Indexed: 05/10/2025] Open
Abstract
Dermal fibroblasts play a central role in the production and remodeling of dermal extracellular matrix. Their spatially distinct subpopulations exhibit different functions during skin homeostasis, wound healing, and other diseases such as fibrosis and cancer. Here, we present a protocol for detecting 3D dermal fibroblast cellular dynamics in mice without fixing the tissue using an intravital imaging technique. We describe steps for preparing, labeling, and mounting mice. We then describe detailed procedures for intravital imaging, recovery, and quantification. For complete details on the use and execution of this protocol, please refer to Rognoni et al.1.
Collapse
Affiliation(s)
- Toru Hiratsuka
- Department of Oncogenesis and Growth Regulation, Research Center, Osaka International Cancer Institute, Chuoku, Osaka 541-8567, Japan; Department of Molcular Oncology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan.
| | - Emanuel Rognoni
- Centre for Cell Biology and Cutaneous Research, The Faculty of Medicine and Dentistry, Blizard Institute, Queen Mary University of London, London, UK
| |
Collapse
|
8
|
Ku YC, Lee YC, Hong YK, Lo YL, Kuo CH, Wang KC, Hsu CK, Yu CH, Lin SW, Wu HL. Deciphering the Dysregulating IGF-1-SP1-CD248 Pathway in Fibroblast Functionality during Diabetic Wound Healing. J Invest Dermatol 2025; 145:1180-1195. [PMID: 39293711 DOI: 10.1016/j.jid.2024.07.035] [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: 02/26/2024] [Revised: 07/08/2024] [Accepted: 07/31/2024] [Indexed: 09/20/2024]
Abstract
Reduced fibroblast activity is a critical factor in the progression of diabetic ulcers. CD248, a transmembrane glycoprotein prominently expressed in activated fibroblasts, plays a pivotal role in wound healing. However, the role of CD248 in diabetic wound healing and the CD248 regulatory pathway remains largely unexplored. Our study shows that CD248 expression is significantly reduced in skin wounds from both patients and mice with diabetes. Single-cell transcriptome data analyses reveal a marked reduction of CD248-enriched secretory-reticular fibroblasts in diabetic wounds. We identify IGF-1 as a key regulator of CD248 expression through the protein kinase B/mTOR signaling pathway and the SP1 transcription factor. Overexpression of CD248 enhances fibroblast motility, elucidating the under-representation of CD248-enriched fibroblasts in diabetic wounds. Immunohistochemical staining of diabetic wound samples further confirms low SP1 expression and fewer CD248-positive secretory-reticular fibroblasts. Further investigation reveals that elevated TNFα levels in diabetic environment promotes IGF-1 resistance, and inhibiting IGF-1 induced CD248 expression. In summary, our findings underscore the critical role of the IGF1-SP1-CD248 axis in activating reticular fibroblasts during wound-healing processes. Targeting this axis in fibroblasts could help develop a therapeutic regimen for diabetic ulcers.
Collapse
Affiliation(s)
- Ya-Chu Ku
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Yao-Chou Lee
- Division of Plastic and Reconstructive Surgery, Department of Surgery, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yi-Kai Hong
- Department of Dermatology, National Cheng Kung University Hospital, Tainan, Taiwan; Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Yung-Ling Lo
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Cheng-Hsiang Kuo
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuan-Chieh Wang
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| | - Chao-Kai Hsu
- Department of Dermatology, National Cheng Kung University Hospital, Tainan, Taiwan; International Center for Wound Repair and Regeneration, National Cheng Kung University, Tainan, Taiwan
| | - Chien-Hung Yu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shu-Wha Lin
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan.
| | - Hua-Lin Wu
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
9
|
Li D, Li Z, Liu S, Chen X, Che X, Deng G, Chen J, Li H, Wang R, Chen X, Su W, Su J. Single-cell RNA sequencing highlights the role of proinflammatory fibroblasts, vascular endothelial cells, and immune cells in the keloid immune microenvironment. Int J Dermatol 2025; 64:890-900. [PMID: 39450923 DOI: 10.1111/ijd.17516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 08/25/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024]
Abstract
BACKGROUND Keloids, characterized by an aberrant wound-healing process and a highly complex immune microenvironment, pose significant challenges for clinical management. Fibroblasts and vascular endothelial cells (VEC) were identified as the leading cells of keloid development. However, their roles in the keloid immune landscape have yet to be thoroughly elucidated. METHODS To explore the functional state of cells in the immune landscape of keloids, we conducted a single-cell RNA sequencing analysis on the tissue from three keloid lesions and two specimens of healthy skin. We simultaneously utilized available keloid data from the public database for external validation. RESULTS Specific subsets, such as proinflammatory fibroblasts (piF) and VEC, were markedly elevated in lesional skin compared to normal skin. Subsequent differential gene expression and Gene Ontology analyses indicated that these subsets may be involved in shaping the microenvironment. In keloids, there is an increased expression of immune-associated genes (P < 0.05), including TNFRSF6B, HGF, and TGFB3, alongside a decreased expression of inflammatory chemokines in the piF. Moreover, the significant upregulation of immune suppressive genes (P < 0.05), including CD39, CD73, and HIF1A, suggested the potential involvement of VEC as a conditional immune subpopulation in the keloid microenvironment. Immune cell communication analysis revealed preferential enrichment of macrophages and Tregs, highlighting intensified macrophage-centered interactions within the keloid microenvironment. CONCLUSION Our study highlighted the role of piF and VEC in the immune microenvironment of keloids for the first time, providing potential targets for therapeutic development.
Collapse
Affiliation(s)
- Daishi Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hu Nan Key Laboratory of Aging Biology, Changsha, China
| | - Zhaohuai Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Sitao Liu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hu Nan Key Laboratory of Aging Biology, Changsha, China
| | - Xiaozhen Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hu Nan Key Laboratory of Aging Biology, Changsha, China
| | - Xuanlin Che
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hu Nan Key Laboratory of Aging Biology, Changsha, China
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hu Nan Key Laboratory of Aging Biology, Changsha, China
| | - Jialing Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
- Department of Clinical Medicine, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - He Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Rong Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hu Nan Key Laboratory of Aging Biology, Changsha, China
| | - Wenru Su
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou, China
| | - Juan Su
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, China
- Hunan Engineering Research Center of Skin Health and Disease, Central South University, Changsha, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Central South University, Changsha, China
- National Clinical Research Center of Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Hu Nan Key Laboratory of Aging Biology, Changsha, China
| |
Collapse
|
10
|
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
|
11
|
Shima C, Ohashi A, Furukawa S, Yamamoto S, Kashimoto R, Satoh A. Collagen fiber and cellular dynamics of axolotl skin with aging. Dev Growth Differ 2025; 67:195-204. [PMID: 40127964 DOI: 10.1111/dgd.70005] [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: 11/21/2024] [Revised: 02/18/2025] [Accepted: 02/26/2025] [Indexed: 03/26/2025]
Abstract
As skin ages, its structure and function undergo significant transformations driven by complex cellular and molecular processes. In this study, we explore these changes using the axolotl, an amphibian model known for its transparent skin, allowing detailed observation of both epidermal and dermal layers. We found that axolotl skin, composed of an epidermis and a collagen-rich dermis with three distinct layers (stratum baladachinum, spongiosum, and compactum), shows clear age-related alterations. These changes include reduced fibroblast numbers, altered lattice-patterned cell morphology, disruption of the lattice patterned collagen fiber pattern, thickening the stratum spongiosum, and thinning of the stratum compactum. Notably, fibroblasts, which play a crucial role in collagen braiding, displayed diminished functionality in older axolotls. This study highlights how aging affects both the structural integrity of dermal collagen and cellular dynamics. Given the similarity between axolotl and mammalian skin, these findings may provide valuable insights into the mechanisms of skin aging and potential avenues for anti-aging therapies. This research offers a foundation for future studies aimed at understanding skin aging and regeneration.
Collapse
Affiliation(s)
- Chisaki Shima
- Graduate School of Environment, Life, natural Science and Technology, Okayama University, Okayama, Japan
| | - Ayaka Ohashi
- Graduate School of Environment, Life, natural Science and Technology, Okayama University, Okayama, Japan
| | - Saya Furukawa
- Graduate School of Environment, Life, natural Science and Technology, Okayama University, Okayama, Japan
| | - Sakiya Yamamoto
- Graduate School of Environment, Life, natural Science and Technology, Okayama University, Okayama, Japan
| | - Rena Kashimoto
- Division of Earth, Life, and Molecular Sciences, Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | - Akira Satoh
- Graduate School of Environment, Life, natural Science and Technology, Okayama University, Okayama, Japan
| |
Collapse
|
12
|
Li C, Liao J, Chen B, Wang Q. Heterogeneity of the tumor immune cell microenvironment revealed by single-cell sequencing in head and neck cancer. Crit Rev Oncol Hematol 2025; 209:104677. [PMID: 40023465 DOI: 10.1016/j.critrevonc.2025.104677] [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: 12/05/2024] [Revised: 02/16/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025] Open
Abstract
Head and neck cancer (HNC) is the sixth most common disease in the world. The recurrence rate of patients is relatively high, and the heterogeneity of tumor immune microenvironment (TIME) cells may be an important reason for this. Single-cell sequencing (SCS) is currently the most promising and mature application in cancer research. It can identify unique genes expressed in cells and study tumor heterogeneity. According to current research, the heterogeneity of immune cells has become an important factor affecting the occurrence and development of HNC. SCSs can provide effective therapeutic targets and prognostic factors for HNC patients through analyses of gene expression levels and cell heterogeneity. Therefore, this study analyzes the basic theory of HNC and the development of SCS technology, elaborating on the application of SCS technology in HNC and its potential value in identifying HNC therapeutic targets and biomarkers.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Jia Liao
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Bo Chen
- Department of Oncology, Suining Central Hospital, Suining, Sichuan 629000, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan 629000, China.
| |
Collapse
|
13
|
Xie R, Li C, Yun J, Zhang S, Zhong A, Cen Y, Li Z, Chen J. Identifying the Pattern Characteristics of Anoikis-Related Genes in Keloid. Adv Wound Care (New Rochelle) 2025; 14:223-237. [PMID: 38775414 DOI: 10.1089/wound.2024.0027] [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: 07/11/2024] Open
Abstract
Objective: Anoikis is a kind of programmed cell death that is triggered when cells lose contact with each other or with the matrix. However, the potential value of anoikis-related genes (ARGs) in keloid (KD) has not been investigated. Approach: We downloaded three keloid fibroblast (KF) RNA sequencing (RNA-seq) datasets from the Gene Expression Omnibus (GEO) and obtained 338 ARGs from a search of the GeneCards database and PubMed articles. Weighted correlation network analysis was used to construct the coexpression network and obtain the KF-related ARGs. The LASSO-Cox method was used to screen the hub ARGs and construct the best prediction model. Then, GEO single-cell sequencing datasets were used to verify the expression of hub genes. We used whole RNA-seq for gene-level validation and the correlation between KD immune infiltration and anoikis. Results: Our study comprehensively analyzed the role of ARGs in KD for the first time. The least absolute shrinkage and selection operator (LASSO) regression analysis identified six hub ARGs (HIF1A, SEMA7A, SESN1, CASP3, LAMA3, and SIK2). A large number of miRNAs participate in the regulation of hub ARGs. In addition, correlation analysis revealed that ARGs were significantly correlated with the infiltration levels of multiple immune cells in patients with KD. Innovation: We explored the expression characteristics of ARGs in KD, which is extremely important for determining the molecular pathways and mechanisms underlying KD. Conclusions: This study provides a useful reference for revealing the characteristics of ARGs in the pathogenesis of KD. The identified hub genes may provide potential therapeutic targets for patients. This study provides new ideas for individualized therapy and immunotherapy.
Collapse
Affiliation(s)
- Ruxin Xie
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Chenyu Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jiao Yun
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shiwei Zhang
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ai Zhong
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ying Cen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junjie Chen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Le Maître M, Guerrier T, Sanges S, Chepy A, Collet A, Launay D. Beyond circulating B cells: Characteristics and role of tissue-infiltrating B cells in systemic sclerosis. Autoimmun Rev 2025; 24:103782. [PMID: 40010623 DOI: 10.1016/j.autrev.2025.103782] [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: 01/10/2025] [Revised: 02/21/2025] [Accepted: 02/22/2025] [Indexed: 02/28/2025]
Abstract
B cells play a key role in the pathophysiology of systemic sclerosis (SSc). While they are less characterized than their circulating counterparts, tissue-infiltrating B cells may have a more direct pathological role in tissues. In this review, we decipher the multiple evidence of B cells infiltration in the skin and lungs of SSc patients and animal models of SSc but also of other chronic fibrotic diseases with similar pathological mechanisms such as chronic graft versus host disease, idiopathic pulmonary fibrosis or morphea. We also recapitulate the current knowledge about mechanisms of B cells infiltration and their functions in tissues. Finally, we discuss B cell targeted therapies, and their specific impact on infiltrated B cells. Understanding the local consequences of infiltrating B cells is an important step for a better management of patients and the improvement of therapies in SSc.
Collapse
Affiliation(s)
- Mathilde Le Maître
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France.
| | - Thomas Guerrier
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France
| | - Sébastien Sanges
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| | - Aurélien Chepy
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| | - Aurore Collet
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Institut d'Immunologie, Pôle de Biologie Pathologie Génétique, Lille, France
| | - David Launay
- Univ. Lille, Inserm, CHU Lille, U1286 - INFINITE - Institute for Translational Research in Inflammation, Lille, France; CHU Lille, Département de Médecine Interne et Immunologie Clinique, F-59000 Lille, France; Centre National de Référence Maladies Auto-immunes Systémiques Rares du Nord, Nord-Ouest, Méditerranée et Guadeloupe (CeRAINOM), F-59000 Lille, France; Health Care Provider of the European Reference Network on Rare Connective Tissue and Musculoskeletal Diseases (ReCONNET), France
| |
Collapse
|
15
|
Kamat P, Macaluso N, Li Y, Agrawal A, Winston A, Pan L, Stewart T, Starich B, Milcik N, Min C, Wu PH, Walston J, Fan J, Phillip JM. Single-cell morphology encodes functional subtypes of senescence in aging human dermal fibroblasts. SCIENCE ADVANCES 2025; 11:eads1875. [PMID: 40279419 PMCID: PMC12024660 DOI: 10.1126/sciadv.ads1875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 03/21/2025] [Indexed: 04/27/2025]
Abstract
Cellular senescence, a hallmark of aging, reveals context-dependent phenotypes across multiple biological length scales. Despite its mechanistic importance, identifying and characterizing senescence across cell populations is challenging. Using primary dermal fibroblasts, we combined single-cell imaging, machine learning, several induced senescence conditions, and multiple protein biomarkers to define functional senescence subtypes. Single-cell morphology analysis revealed 11 distinct morphology clusters. Among these, we identified three as bona fide senescence subtypes (C7, C10, and C11), with C10 exhibiting the strongest age dependence within an aging cohort. In addition, we observed that a donor's senescence burden and subtype composition were indicative of susceptibility to doxorubicin-induced senescence. Functional analysis revealed subtype-dependent responses to senotherapies, with C7 being most responsive to the combination of dasatinib and quercetin. Our single-cell analysis framework, SenSCOUT, enables robust identification and classification of senescence subtypes, offering applications in next-generation senotherapy screens, with potential toward explaining heterogeneous senescence phenotypes based on the presence of senescence subtypes.
Collapse
Affiliation(s)
- Pratik Kamat
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Nico Macaluso
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Yukang Li
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Anshika Agrawal
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Aaron Winston
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Lauren Pan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Teasia Stewart
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Bartholomew Starich
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Nicholas Milcik
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Chanhong Min
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Pei-Hsun Wu
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
| | - Jeremy Walston
- Department of Geriatric Medicine and Gerontology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Jean Fan
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jude M. Phillip
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Translational Tissue Engineering Center (TTEC), Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
16
|
Thiagarajan L, Sanchez-Alvarez R, Kambara C, Rajasekar P, Wang Y, Halloy F, Hall J, Stark HJ, Martin I, Boukamp P, Kurinna S. miRNA-29 regulates epidermal and mesenchymal functions in skin repair. FEBS Lett 2025. [PMID: 40285401 DOI: 10.1002/1873-3468.70051] [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: 11/19/2024] [Revised: 03/06/2025] [Accepted: 03/15/2025] [Indexed: 04/29/2025]
Abstract
MicroRNAs (miRNAs) control organogenesis in mammals by inhibiting translation of mRNA. Skin is an excellent model to study the role of miRNAs in epidermis and the mesenchyme. Previous research demonstrated miRNA-29 family functions in skin; however, the mRNA targets and the downstream mechanisms of miRNA-29-mediated regulation are missing. We used the miRNA crosslinking and immunoprecipitation method to find direct targets of miRNA-29 in keratinocytes and fibroblasts from human skin. miRNA-29 inhibition using modified antisense oligonucleotides in 2D and 3D cultures of keratinocytes and fibroblasts enhanced cell-to-matrix adhesion through autocrine and paracrine mechanisms of miRNA-29-dependent tissue growth. We reveal a full transcriptome of human keratinocytes with enhanced adhesion to the matrix, which supports regeneration of the epidermis and is regulated by miRNA-29. Impact statement The functions of small, therapeutically targetable microRNA molecules identified in our study can provide a new approach to improve wound healing by restoring and enhancing the inner molecular mechanisms of a cell and its surrounding matrix. We also provide a plethora of new mRNA targets for follow-up studies of cell adhesion and extracellular matrix formation in humans.
Collapse
Affiliation(s)
- Lalitha Thiagarajan
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| | - Rosa Sanchez-Alvarez
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| | - Chiho Kambara
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| | | | - Yuluang Wang
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland
| | - François Halloy
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland
| | - Jonathan Hall
- Department of Chemistry and Applied Biosciences, Institute of Pharmaceutical Sciences, ETH Zurich, Switzerland
| | | | - Iris Martin
- German Cancer Research Center, Heidelberg, Germany
| | | | - Svitlana Kurinna
- Division of Cell Matrix Biology and Regenerative Medicine, School of Biological Sciences, University of Manchester, UK
| |
Collapse
|
17
|
Huang C, Shao Y, Bai J, Zhao Y, Ogawa R. Fibroproliferative conditions: the 3R approach bridging fibrosis and tumors. Trends Mol Med 2025:S1471-4914(25)00060-7. [PMID: 40268589 DOI: 10.1016/j.molmed.2025.03.009] [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: 08/02/2024] [Revised: 02/10/2025] [Accepted: 03/21/2025] [Indexed: 04/25/2025]
Abstract
Soft-tissue fibroproliferative conditions (FPCs) affect many organs. All demonstrate the accumulation of (myo)fibroblasts and extracellular matrix. Currently, FPCs are classified according to the affected body site/organ. To promote research into the etiological mechanisms that drive pathological FPCs, we propose a new, more clinically grounded, FPC classification that is based on the intent and severity of the fibroproliferation. There are three categories: responsive, replacement, and reconstructive FPCs. Reconstructive FPCs (e.g., keloids) have quasi-neoplastic behaviors, including local invasiveness, and serve as a bridge between fibrosis and cancers. Comparisons of reconstructive FPCs to both cancers and the other FPC categories may help elucidate their pathogenic cellular properties, microenvironmental components, and intracellular-signaling mechanisms. Thus, the new FPC classification may promote research in the fibrosis field.
Collapse
Affiliation(s)
- Chenyu Huang
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China.
| | - Yue Shao
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China.
| | - Jianbo Bai
- Institute of Biomechanics and Medical Engineering, Department of Engineering Mechanics, School of Aerospace Engineering, Tsinghua University, Beijing 100084, China
| | - Yi Zhao
- Department of Dermatology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo 113-8603, Japan
| |
Collapse
|
18
|
Kazmi A, Gill R, Restrepo P, Ji AL. The spatial and single-cell landscape of skin: Charting the multiscale regulation of skin immune function. Semin Immunol 2025; 78:101958. [PMID: 40267702 DOI: 10.1016/j.smim.2025.101958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/25/2025]
Abstract
Immune regulation is a key function of the skin, a barrier tissue that exhibits spatial compartmentalization of innate and adaptive immune cells. Recent advances in single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) have facilitated systems-based investigations into the molecular and cellular features of skin immunity at single-cell resolution, identifying cell types that maintain homeostasis in a coordinated manner, and those that exhibit dysfunctional cell-cell interactions in disease. Here, we review how technological innovation is uncovering the multiple scales of heterogeneity in the immune landscape of the skin. The microanatomic scale encompasses the skin's diverse cellular components and multicellular spatial organization, which govern the functional cell interactions and behaviors necessary to protect the host. On the macroanatomic scale, understanding heterogeneity in cutaneous tissue architecture across anatomical sites promises to unearth additional functional immune variation and resulting disease consequences. We focus on how single-cell and spatial dissection of the immune system in experimental models and in humans has led to a deeper understanding of how each cell type in the skin contributes to overall immune function in a context-dependent manner. Finally, we highlight translational opportunities for adopting these technologies, and insights gleaned from them, into the clinic.
Collapse
Affiliation(s)
- Abiha Kazmi
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Raman Gill
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Paula Restrepo
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Andrew L Ji
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Black Family Stem Cell Institute, Institute of Regenerative Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
19
|
Petrova B, Guler AT. Recent Developments in Single-Cell Metabolomics by Mass Spectrometry─A Perspective. J Proteome Res 2025; 24:1493-1518. [PMID: 39437423 PMCID: PMC11976873 DOI: 10.1021/acs.jproteome.4c00646] [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: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/15/2024] [Indexed: 10/25/2024]
Abstract
Recent advancements in single-cell (sc) resolution analyses, particularly in sc transcriptomics and sc proteomics, have revolutionized our ability to probe and understand cellular heterogeneity. The study of metabolism through small molecules, metabolomics, provides an additional level of information otherwise unattainable by transcriptomics or proteomics by shedding light on the metabolic pathways that translate gene expression into functional outcomes. Metabolic heterogeneity, critical in health and disease, impacts developmental outcomes, disease progression, and treatment responses. However, dedicated approaches probing the sc metabolome have not reached the maturity of other sc omics technologies. Over the past decade, innovations in sc metabolomics have addressed some of the practical limitations, including cell isolation, signal sensitivity, and throughput. To fully exploit their potential in biological research, however, remaining challenges must be thoroughly addressed. Additionally, integrating sc metabolomics with orthogonal sc techniques will be required to validate relevant results and gain systems-level understanding. This perspective offers a broad-stroke overview of recent mass spectrometry (MS)-based sc metabolomics advancements, focusing on ongoing challenges from a biologist's viewpoint, aimed at addressing pertinent and innovative biological questions. Additionally, we emphasize the use of orthogonal approaches and showcase biological systems that these sophisticated methodologies are apt to explore.
Collapse
Affiliation(s)
- Boryana Petrova
- Medical
University of Vienna, Vienna 1090, Austria
- Department
of Pathology, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
| | - Arzu Tugce Guler
- Department
of Pathology, Boston Children’s Hospital, Boston, Massachusetts 02115, United States
- Institute
for Experiential AI, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
20
|
Xie R, Li C, Zhao T, Zhang S, Zhong A, Chen N, Li Z, Chen J. Integration of Flow Cytometry and Single-Cell RNA Sequencing Analysis to Explore the Fibroblast Subpopulations in Keloid that Correlate with Recurrence. Adv Wound Care (New Rochelle) 2025. [PMID: 40177712 DOI: 10.1089/wound.2024.0262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025] Open
Abstract
Objective: Fibroblasts (FBs) are the cytological basis of keloid (KD) formation. This study aimed to identify the key pathogenic target cell subpopulation involved in KD recurrence. Approach: Single-cell RNA sequencing data were retrieved from public databases, revealing distinct gene expression patterns in FB subpopulations. Flow cytometry (FCM) was used to identify the surface molecular phenotypes of FBs that affect KD recurrence. Simultaneously, logistic regression analysis was performed to assess the predictive value of changes in FB subpopulation percentages for clinical KD recurrence. Results: The percentage of keloid fibroblasts was significantly greater than that in normal tissues. Through further clustering analysis of the FB population, we obtained four subpopulations, FB1-FB4, in which the percentages of FB1 subpopulation were increased, and functional enrichment analysis suggested that the FB1 subpopulation may play a greater role in extracellular matrix collagen oversynthesis in KD. In addition, the gene expression of CD26 (DPP4), CD117 (c-KIT), and CD34 in the FB1 subpopulation was significantly higher than that in FB2-4 subpopulations. Moreover, the percentage of CD26+/CD117+/CD34+ cell subpopulations in the FCM data of patients with KD recurrence was significantly increased. Regression analysis confirmed that the CD26+/CD117+/CD34+ FB subpopulation was a risk factor for relapse. Innovation: We demonstrated that the molecular phenotypic and functional heterogeneity of FBs influences KD recurrence. Conclusion: We identified key pathogenic FB subpopulations that may affect KD development, which can be used as potential markers to predict recurrence and provide potential target cell populations for future clinical treatment.
Collapse
Affiliation(s)
- Ruxin Xie
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Chenyu Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Tian Zhao
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Shiwei Zhang
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ai Zhong
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Nengbin Chen
- Cosmetic Burn and Plastic Surgery, The People's Hospital of Leshan, Leshan, China
| | - Zhengyong Li
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Junjie Chen
- Department of Burn and Plastic Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
21
|
Liu M, Zheng S, Li H, Budowle B, Wang L, Lou Z, Ge J. High resolution tissue and cell type identification via single cell transcriptomic profiling. PLoS One 2025; 20:e0318151. [PMID: 40138334 PMCID: PMC11940611 DOI: 10.1371/journal.pone.0318151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 01/11/2025] [Indexed: 03/29/2025] Open
Abstract
Tissue identification can be instrumental in reconstructing a crime scene but remains a challenging task in forensic investigations. Conventionally, identifying the presence of certain tissue from tissue mixture by predefined cell type markers in bulk fashion is challenging due to limitations in sensitivity and accuracy. In contrast, single-cell RNA sequencing (scRNA-Seq) is a promising technology that has the potential to enhance or even revolutionize tissue and cell type identification. In this study, we developed a high sensitive general purpose single cell annotation pipeline, scTissueID, to accurately evaluate the single cell profile quality and precisely determine the cell and tissue types based on scRNA profiles. By incorporating a crucial and unique reference cell quality differentiation phase of targeting only high confident cells as reference, scTissueID achieved better and consistent performance in determining cell and tissue types compared to 8 state-of-art single cell annotation pipelines and 6 widely adopted machine learning algorithms, as demonstrated through a large-scale and comprehensive comparison study using both forensic-relevant and Human Cell Atlas (HCA) data. We highlighted the significance of cell quality differentiation, a previously undervalued factor. Thus, this study offers a tool capable of accurately and efficiently identifying cell and tissue types, with broad applicability to forensic investigations and other biomedical research endeavors.
Collapse
Affiliation(s)
- Muyi Liu
- Center for Human Identification, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
- Department of Cell Biology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Suilan Zheng
- Department of Chemistry, Purdue University, West Lafayette, Indiana, United States of America
| | - Hongmin Li
- Department of Computer Science, California State University, East Bay, Hayward, California, United States of America
| | - Bruce Budowle
- Department of Forensic Medicine, University of Helsinki, Finland
| | - Le Wang
- Department of Electronic and Information Engineering, North China University of Technology, Beijing, China
| | - Zhaohuan Lou
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jianye Ge
- Center for Human Identification, University of North Texas Health Science Center, Fort Worth, Texas, United States of America
| |
Collapse
|
22
|
Parisi L, Mansour F, Rihs S, Schnyder I, La Scala GC, Katsaros C, Degen M. The Skin-to-Mucosa Ratio Defines the Osteogenic Potential of Lip Fibroblasts. J Dent Res 2025:220345251321806. [PMID: 40108556 DOI: 10.1177/00220345251321806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
Fibroblasts isolated from discarded lip tissue obtained during cheiloplasty in patients with cleft lip/palate (CLP) show promising osteogenic potential and may be an appealing cell source for autologous bone regeneration. As the lip is a mucocutaneous junction, explant cultures from unseparated lip biopsies produce mesenchymal outgrowths composed of skin- and mucosa-derived fibroblasts. The proportions of the 2 fibroblast populations, however, differ among CLP patients and depend on the morphology of the excised sample, which is unique for each donor. Understanding the osteogenic activities of CLP fibroblast populations with varying skin-to-mucosa ratios is critical for their therapeutic application. We isolated CLP fibroblasts from 10 unseparated lip biopsies and comprehensively evaluated them for their bone differentiation capacities in vitro, demonstrating heterogeneous osteogenic potentials. Because there are no markers that can distinguish skin from mucosa fibroblasts, we used the respective and matching CLP keratinocytes to ascertain the skin-to-mucosa ratio of the 10 specimens. Thus, we found that CLP fibroblasts isolated from biopsies with high skin-to-mucosa ratios had a much higher osteogenic capacity than those derived from biopsies with low skin-to-mucosa ratios. To validate and solidify these findings, we carefully separated skin and mucosa tissues during corrective lip surgery to isolate pure skin and mucosa CLP lip fibroblasts. Indeed, skin had a higher osteogenic potential than their mucosal counterparts did. Furthermore, we discovered that the high osteogenic activity in skin was limited to specific subpopulations of yet unknown identities. Our findings indicate that skin fibroblasts perform better than their mucosal counterparts do, even though both types of fibroblasts can differentiate into bone-forming cells.
Collapse
Affiliation(s)
- L Parisi
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - F Mansour
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - S Rihs
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - I Schnyder
- University Clinic for Pediatric Surgery, Bern University Hospital, Bern, Switzerland
| | - G C La Scala
- Division of Pediatric Surgery, Department of Pediatrics, University Hospital of Geneva, Geneva, Switzerland
| | - C Katsaros
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| | - M Degen
- Laboratory for Oral Molecular Biology, Department of Orthodontics and Dentofacial Orthopedics, University of Bern, Bern, Switzerland
| |
Collapse
|
23
|
Liu S, Lu S, Pang Z, Li J, Zhou M, Ding Z, Feng Z. Inflammatory Transformation of Skin Basal Cells as a Key Driver of Cutaneous Aging. Int J Mol Sci 2025; 26:2617. [PMID: 40141258 PMCID: PMC11942461 DOI: 10.3390/ijms26062617] [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: 01/29/2025] [Revised: 03/09/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
This study comprehensively investigated keratinocyte subpopulation heterogeneity and developmental trajectories during skin aging using single-cell sequencing, transcriptomics, and facial aging-related genome-wide association studies (GWAS) data. We identified three major subpopulations: basal cells (BCs), spinous cells (SCs), and IFI27+ keratinocytes. Single-cell pseudotime analysis revealed that basal cells can differentiate along two distinct paths: toward spinous differentiation or the inflammatory state. With aging, the proportion of IFI27+ cells significantly increased, displaying more active inflammatory and immunomodulatory signals. Through cell-cell communication analysis, we found that the signaling pathways, including NOTCH, PTPR, and PERIOSTIN, exhibited distinct characteristics along different branches. Integration of the GWAS data revealed significant loci on chromosomes 2, 3, 6, and 9 that were spatially correlated with key biological pathways (including antigen processing, oxidative stress, and apoptosis). These findings reveal the complex cellular and molecular mechanisms underlying skin aging, offering potential targets for novel diagnostic approaches and therapeutic interventions.
Collapse
Affiliation(s)
| | | | | | | | | | - Zhenhua Ding
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (S.L.); (Z.P.); (J.L.); (M.Z.)
| | - Zhijun Feng
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, China; (S.L.); (S.L.); (Z.P.); (J.L.); (M.Z.)
| |
Collapse
|
24
|
Li W, Li X, Zhang Y, Zhu B, Xu X, Xiao H, Zhang S. Altered Arginine Metabolism Affects Proliferation and Radiosensitivity of Keloids. Exp Dermatol 2025; 34:e70077. [PMID: 40095415 DOI: 10.1111/exd.70077] [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: 11/12/2023] [Revised: 02/20/2025] [Accepted: 03/01/2025] [Indexed: 03/19/2025]
Abstract
Keloid is characterised by the reprogramming of cellular metabolism, wherein keloid cells adapt their metabolic pathways to meet the demands for energy and biosynthetic precursors. Investigating the intricate relationship between cellular metabolism and the biological behaviour of keloid holds the potential to yield novel therapeutic strategies for keloid. To elucidate the molecular alterations and potential underlying regulatory mechanisms in keloids, we created comprehensive metabolic profiling at the pathway level by analysing metabolomic, transcriptomic and single-cell RNA-sequencing data from keloids and adjacent skin. Viability assay and clonogenic assay were performed to validate the function of the metabolic pathway(s) in primary keloid fibroblast cells. Integrated analysis revealed an upregulation of arginine and proline metabolism in keloids. According to single-cell RNA-seq data, elevated expression of genes related to arginine and proline metabolism, such as P4HA3, P4HA2, P4HA1, PYCR1, OAT and ASS1, was predominately highly expressed in fibroblast-2. Fibroblast-2 displayed more obvious phenotypes of mesenchymal fibroblast. Critical genes from integrated analysis including P4HA3, P4HA2, P4HA1, PYCR1 and AZIN2, and metabolites including fumaric acid and 2-oxo-5-amino-pentanoic acid showed prognostic relevance with disease-free survival of keloid. Additionally, an In vitro study showed that arginine deprivation therapy (ADT) inhibited and radiosensitised the proliferation of keloid-derived fibroblasts. In conclusion, our thorough multiomics study deepens our understanding of the link between arginine and proline metabolism and keloid proliferation and radiosensitivity. Elevated activity of arginine and proline metabolism in mesenchymal fibroblasts may be a potential therapeutic pathway for keloid.
Collapse
Affiliation(s)
- Wei Li
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoqian Li
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, China
| | - Yange Zhang
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Baochen Zhu
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xuewen Xu
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Haitao Xiao
- Department of Plastic and Burns Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
- NHC Key Laboratory of Nuclear Technology Medical Transformation, Mianyang Central Hospital, Mianyang, China
- Second Affiliated Hospital of Chengdu Medical College, China National Nuclear Corporation 416 Hospital, Chengdu, China
| |
Collapse
|
25
|
Almet AA, Liu Y, Nie Q, Plikus MV. Integrated Single-Cell Analysis Reveals Spatially and Temporally Dynamic Heterogeneity in Fibroblast States during Wound Healing. J Invest Dermatol 2025; 145:645-659.e25. [PMID: 39019149 DOI: 10.1016/j.jid.2024.06.1281] [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: 01/31/2023] [Revised: 06/14/2024] [Accepted: 06/26/2024] [Indexed: 07/19/2024]
Abstract
Wound healing is a dynamic process over temporal and spatial scales. Key to repair outcomes are fibroblasts; yet, how they modulate healing across time and in different wound regions remains incompletely understood. By integrating single-cell RNA-sequencing datasets of mouse skin and wounds, we infer that fibroblasts are the most transcriptionally dynamic skin-resident cells, evolving during postnatal skin maturation and rapidly after injury toward distinct late scar states. We show that transcriptional dynamics in fibroblasts are largely driven by genes encoding extracellular matrix and signaling factors. Lineage trajectory inference and spatial gene mapping reveal that Prg4-expressing fibroblasts transiently emerge along early wound edges. Within days, they become replaced by long-lasting and likely noninterconverting fibroblast populations, including Col25a1-expressing and Pamr1-expressing fibroblasts that occupy subepidermal and deep scar regions, respectively, where they engage in reciprocal signaling with immune cells. Signaling inference shows that fibroblast-immune crosstalk repeatedly uses some signaling pathways across wound healing time, whereas use of other signaling pathways is time and space limited. Collectively, we uncovered high transcriptional plasticity by wound fibroblasts, with early states transiently forming distinct microniches along wound edges and in the fascia, followed by stable states that stratify scar tissue into molecularly dissimilar upper and lower layers.
Collapse
Affiliation(s)
- Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA
| | - Yingzi Liu
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA
| | - Qing Nie
- Department of Mathematics, University of California, Irvine, Irvine, California, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA.
| | - Maksim V Plikus
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, California, USA; Department of Developmental and Cell Biology, University of California, Irvine, Irvine, California, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, California, USA.
| |
Collapse
|
26
|
Kumaran G, Carroll L, Muirhead N, Bottomley MJ. How Can Spatial Transcriptomic Profiling Advance Our Understanding of Skin Diseases? J Invest Dermatol 2025; 145:522-535. [PMID: 39177547 DOI: 10.1016/j.jid.2024.07.006] [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: 03/01/2024] [Revised: 05/23/2024] [Accepted: 07/04/2024] [Indexed: 08/24/2024]
Abstract
Spatial transcriptomic (ST) profiling is the mapping of gene expression within cell populations with preservation of positional context and represents an exciting new approach to develop our understanding of local and regional influences upon skin biology in health and disease. With the ability to probe from a few hundred transcripts to the entire transcriptome, multiple ST approaches are now widely available. In this paper, we review the ST field and discuss its application to dermatology. Its potential to advance our understanding of skin biology in health and disease is highlighted through the illustrative examples of 3 research areas: cutaneous aging, tumorigenesis, and psoriasis.
Collapse
Affiliation(s)
- Girishkumar Kumaran
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Liam Carroll
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | | | - Matthew J Bottomley
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom.
| |
Collapse
|
27
|
Thau H, Gerjol BP, Hahn K, von Gudenberg RW, Knoedler L, Stallcup K, Emmert MY, Buhl T, Wyles SP, Tchkonia T, Tullius SG, Iske J. Senescence as a molecular target in skin aging and disease. Ageing Res Rev 2025; 105:102686. [PMID: 39929368 DOI: 10.1016/j.arr.2025.102686] [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] [Revised: 01/27/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025]
Abstract
Skin aging represents a multifactorial process influenced by both intrinsic and extrinsic factors, collectively known as the skin exposome. Cellular senescence, characterized by stable cell cycle arrest and secretion of pro-inflammatory molecules, has been implicated as a key driver of physiological and pathological skin aging. Increasing evidence points towards the role of senescence in a variety of dermatological diseases, where the accumulation of senescent cells in the epidermis and dermis exacerbates disease progression. Emerging therapeutic strategies such as senolytics and senomorphics offer promising avenues to target senescent cells and mitigate their deleterious effects, providing potential treatments for both skin aging and senescence-associated skin diseases. This review explores the molecular mechanisms of cellular senescence and its role in promoting age-related skin changes and pathologies, while compiling the observed effects of senotherapeutics in the skin and discussing the translational relevance.
Collapse
Affiliation(s)
- Henriette Thau
- Van Cleve Cardiac Regenerative Medicine Program Mayo Clinic, Rochester, Minesota, USA; Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Bastian P Gerjol
- Department of Internal Medicine, Klinik Hirslanden, Zurich, Switzerland
| | - Katharina Hahn
- Department of Dermatology, Venereology and Allergology, Göttingen University Medical Center, Göttingen, Germany
| | - Rosalie Wolff von Gudenberg
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Leonard Knoedler
- Department of Oral and Maxillofacial Surgery, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health Berlin, Germany
| | - Kenneth Stallcup
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany
| | - Maximilian Y Emmert
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany; Institute for Regenerative Medicine (IREM), University of Zurich, Zurich, Switzerland
| | - Timo Buhl
- Department of Dermatology, Venereology and Allergology, Göttingen University Medical Center, Göttingen, Germany
| | | | - Tamar Tchkonia
- Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Stefan G Tullius
- Division of Transplant Surgery, Department of Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jasper Iske
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum der Charité (DHZC), Berlin, Germany; Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
| |
Collapse
|
28
|
Huang Y, Zhu S, Yao S, Zhai H, Liu C, Han JDJ. Unraveling aging from transcriptomics. Trends Genet 2025; 41:218-235. [PMID: 39424502 DOI: 10.1016/j.tig.2024.09.006] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 09/19/2024] [Indexed: 10/21/2024]
Abstract
Research into aging constitutes a pivotal endeavor aimed at elucidating the underlying biological mechanisms governing aging and age-associated diseases, as well as promoting healthy longevity. Recent advances in transcriptomic technologies, such as bulk RNA sequencing (RNA-seq), single-cell transcriptomics, and spatial transcriptomics, have revolutionized our ability to study aging at unprecedented resolution and scale. These technologies present novel opportunities for the discovery of biomarkers, elucidation of molecular pathways, and development of targeted therapeutic strategies for age-related disorders. This review surveys recent breakthroughs in different types of transcripts on aging, such as mRNA, long noncoding (lnc)RNA, tRNA, and miRNA, highlighting key findings and discussing their potential implications for future studies in this field.
Collapse
Affiliation(s)
- Yuanfang Huang
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shouxuan Zhu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Shuai Yao
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Haotian Zhai
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Chenyang Liu
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Center for Quantitative Biology (CQB), Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China; Peking University Chengdu Academy for Advanced Interdisciplinary Biotechnologies, Chengdu, China.
| |
Collapse
|
29
|
Ohlendieck CM, Matellan C, Manresa MC. Regulation of pathologic fibroblast functions in digestive diseases: a role for hypoxia? Am J Physiol Gastrointest Liver Physiol 2025; 328:G229-G242. [PMID: 39873349 DOI: 10.1152/ajpgi.00277.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/14/2024] [Accepted: 01/10/2025] [Indexed: 01/30/2025]
Abstract
The recent uncovering of fibroblast heterogeneity has given great insight into the versatility of the stroma. Among other cellular processes, fibroblasts are now thought to contribute to the coordination of immune responses in a range of chronic inflammatory diseases and cancer. Although the pathologic roles of myofibroblasts, inflammatory fibroblasts, and cancer-associated fibroblasts in disease are reasonably well understood, the mechanisms behind their activation remain to be uncovered. In the gastrointestinal (GI) tract, several interleukins and tumor necrosis factor superfamily members have been identified as possible mediators driving the acquisition of inflammatory and fibrotic properties in fibroblasts. In addition to cytokines, other microenvironmental factors such as nutrient and oxygen availability are likely contributors to this process. In this respect, the phenomenon of low cellular oxygen levels known as hypoxia is common in a plethora of GI diseases. Indeed, the cross talk between hypoxia and inflammation is well-documented, with an abundance of studies suggesting that oxygen-sensing enzymes may have regulatory effects on inflammatory signaling pathways such as NF-κB. However, the impact that this has in GI fibroblasts in the context of chronic diseases has not been fully uncovered. Here we discuss the role of fibroblasts in GI diseases, the mediators that have emerged as regulators of their functions and the potential impact of hypoxia in this process, highlighting areas that require further investigation.
Collapse
Affiliation(s)
- Cian M Ohlendieck
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| | - Carlos Matellan
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Mario C Manresa
- Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
- School of Biomolecular and Biomedical Science, University College Dublin, Dublin, Ireland
| |
Collapse
|
30
|
Wyles SP, Yu GT, Ganier C, Tchkonia T, Lynch MD, Kuchel GA, Kirkland JL. SenSkin™: a human skin-specific cellular senescence gene set. GeroScience 2025:10.1007/s11357-025-01568-y. [PMID: 39998731 DOI: 10.1007/s11357-025-01568-y] [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: 01/30/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Cellular senescence gene sets have been leveraged to overcome the inadequate sensitivity or specificity of single markers. However, growing evidence of heterogeneity among tissues in senescent cell phenotypes and gene expression profiles has highlighted the need for tissue-specific gene sets. SenSkin™ was curated by an expert review of literature on cellular senescence in the skin and characterized with pathway analysis. To validate SenSkin™, it was evaluated for enrichment with chronological aging in a bulk RNA-sequencing (RNA-seq) dataset and a pseudobulk RNA-seq dataset. Further, changes to SenSkin™ in different skin cell types with photoaging were evaluated in two single-cell RNA-seq datasets. SenSkin™ predominantly included genes related to the senescence-associated secretory phenotype (SASP), which were associated with metabolism and multiple aspects of immune responses. SenSkin™ was more enriched in chronologically aged skin than other commonly used cellular senescence and aging gene sets. In scRNA-seq, SenSkin™ displayed significant upregulation due to photoaging in ten skin cell types. In conclusion, SenSkin™ is a human skin-specific senescence gene set validated in chronological aging and photoaging, which may be more effective at detecting senescent cells in the skin than non-tissue-specific gene sets.
Collapse
Affiliation(s)
- Saranya P Wyles
- Department of Dermatology, Mayo Clinic, 200 First Street SW, Rochester, MN, USA.
| | - Grace T Yu
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN, USA
| | - Clarisse Ganier
- Immunology Department, Metaorganism Unit, Institut Pasteur, Paris, France
| | - Tamar Tchkonia
- Center for Gerotherapeutics, Division of Endocrinology and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magnus D Lynch
- Centre for Gene Therapy and Regenerative Medicine, King's College London, Guy's Hospital, London, UK
- St. John's Institute of Dermatology, King's College London, Guy's Hospital, London, UK
| | - George A Kuchel
- UConn Center on Aging, University of Connecticut, Farmington, CT, USA
| | - James L Kirkland
- Center for Gerotherapeutics, Division of Endocrinology and Metabolism, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
31
|
Wang M, Wang M, Jiang J, Li K, Liang H, Wang N, Zou Y, Wang D, Zhou S, Tang Y, Wu W, Qiu W, Li X, Wang X, Xie Q, Xiang X, Zhou W, Yang L, Chuong CM, Lei M. THSD4 promotes hair growth by facilitating dermal papilla and hair matrix interactions. Theranostics 2025; 15:3571-3588. [PMID: 40093891 PMCID: PMC11905124 DOI: 10.7150/thno.103221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 02/06/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction: Aging causes striking changes in the extracellular matrix (ECM) in hair follicles, which has a profound influence on hair growth. How the ECM of dermal papilla (DP), the master regulator of hair growth, changes during aging remains largely unknown. Methods: Herovici staining, Western Blotting and immunofluorescence were used to assess DP ECM and protein expression in hair follicles. Bulk and single cell RNA-sequencing were used to analyze gene expression and predict upstream and downstream regulators of target genes. Skin organoid and mouse models were used for functional validation of molecular mechanisms. Results: Aged follicle DP shows drastic depletion of ECM in which Thrombospondin Type 1 Domain Containing 4 (Thsd4) is highly downregulated. THSD4 is specifically expressed in the interface between DP and hair matrix (HM). It promotes hair growth by enhancing the interaction between dermal (DP) and epithelial cells (HM) through the SDC4-THSD4-CXCL1 signaling axis in both skin organoids and mouse models. Murine dorsal hair follicles show upregulated THSD4, enhanced DP-HM interaction, and hair growth following exposure to low temperature. Conclusions: THSD4 is a key micro- and macro-environmental mediator to promote hair growth by facilitating epidermal-mesenchymal interactions during aging. These findings demonstrate the therapeutic potential of low-temperature treatment for treating unwanted hair loss.
Collapse
Affiliation(s)
- Miaomiao Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Mengyue Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Jingwei Jiang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Ke Li
- Shenzhen Accompany Technology Cooperation, ltd, Shenzhen 518000, China
| | - Huan Liang
- Shenzhen Accompany Technology Cooperation, ltd, Shenzhen 518000, China
| | - Nian'ou Wang
- Shenzhen Accompany Technology Cooperation, ltd, Shenzhen 518000, China
| | - Yi Zou
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
- Department of Burns and Plastic Surgery, Wuhan General Hospital of Chinese People's Liberation Army, Wuhan 430000, China
| | - Dehuan Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Siyi Zhou
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Yuchun Tang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wang Wu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Weiming Qiu
- Department of Burns and Plastic Surgery, Wuhan General Hospital of Chinese People's Liberation Army, Wuhan 430000, China
| | - Xinxin Li
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China
| | - Xusheng Wang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-Sen University, Sun Yat-Sen University, Shenzhen 518107, China
| | - Qiaoli Xie
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Xiao Xiang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Wei Zhou
- Radiation Oncology Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Li Yang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Mingxing Lei
- Key Laboratory of Biorheological Science and Technology of Ministry of Education &111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China
| |
Collapse
|
32
|
Koczkowska M, Kostecka A, Zawrzykraj M, Myszczyński K, Skoniecka A, Deptuła M, Tymińska A, Czerwiec K, Jąkalski M, Zieliński J, Crossman DK, Crowley MR, Cichorek M, Skowron PM, Pikuła M, Piotrowski A. Identifying differentiation markers between dermal fibroblasts and adipose-derived mesenchymal stromal cells (AD-MSCs) in human visceral and subcutaneous tissues using single-cell transcriptomics. Stem Cell Res Ther 2025; 16:64. [PMID: 39934849 PMCID: PMC11818286 DOI: 10.1186/s13287-025-04185-w] [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: 05/16/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Adipose-derived mesenchymal stromal cells (AD-MSCs) and fibroblasts are both widely used in regenerative medicine, demonstrating significant potential for personalized cell therapy. A major challenge in their use lies in their high biological similarity, encompassing morphology, differentiation capabilities, and flow cytometric markers, making their distinction difficult. METHODS In our study, we aimed to compare AD-MSCs obtained from two types of adipose tissue, subcutaneous and visceral, alongside skin fibroblasts. Notably, all tissue samples were sourced from the same donors. We analyzed the cells for surface antigens via flow cytometry and conducted single-cell RNA sequencing, followed by verification with quantitative PCR (qPCR). RESULTS Our results revealed phenotypic similarities between the isolated AD-MSCs and dermal fibroblasts, particularly in the expression of markers characteristic of AD-MSCs. However, through in-depth analyses, we identified distinct differences between these cell types. Specifically, we pinpointed 30 genes exhibiting the most significant variations in expression between AD-MSCs and fibroblasts. These genes are associated with biological processes such as tissue remodeling, cell movement, and activation in response to external stimuli. Among them, MMP1, MMP3, S100A4, CXCL1, PI16, IGFBP5, COMP were further validated using qPCR, clearly demonstrating their potential to differentiate between AD-MSCs and fibroblasts. CONCLUSIONS Our scRNA-seq analysis elucidates the transcriptional landscape of AD-MSCs and fibroblasts with unprecedented resolution, highlighting both the population-specific markers and the intrapopulation heterogeneity. Our findings underscore the importance of employing high-resolution techniques for cell identification.
Collapse
Affiliation(s)
| | - Anna Kostecka
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland
| | - Małgorzata Zawrzykraj
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Kamil Myszczyński
- Centre of Biostatistics and Bioinformatics Analysis, Medical University of Gdansk, Gdansk, Poland
| | - Aneta Skoniecka
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Milena Deptuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Agata Tymińska
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Katarzyna Czerwiec
- Division of Clinical Anatomy, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Marcin Jąkalski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland
| | - Jacek Zieliński
- Department of Surgical Oncology, Transplant Surgery and General Surgery, Medical University of Gdansk, Gdansk, Poland
| | - David K Crossman
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael R Crowley
- Genomic Core Facility, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Mirosława Cichorek
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland
| | - Piotr M Skowron
- Department of Molecular Biotechnology, Faculty of Chemistry, University of Gdansk, Gdansk, Poland
| | - Michał Pikuła
- Division of Embryology, Department of Anatomy, Medical University of Gdansk, Gdansk, Poland.
| | - Arkadiusz Piotrowski
- 3P-Medicine Laboratory, Medical University of Gdansk, Gdansk, Poland.
- Department of Biology and Pharmaceutical Botany, Medical University of Gdansk, Gdansk, Poland.
| |
Collapse
|
33
|
Morozova V, Pellegata D, Charles RP, Gertsch J. Carboxylesterase 1-mediated endocannabinoid metabolism in skin: role in melanoma progression in BRaf V600E/Pten -/- mice. Cancer Metab 2025; 13:8. [PMID: 39934865 DOI: 10.1186/s40170-025-00378-2] [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: 10/22/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND Melanoma is a highly aggressive skin cancer with a poor prognosis. The endocannabinoids 2-arachidonoylgylcerol (2-AG) and anandamide have been linked to melanoma progression, though their roles remain unclear. We hypothesized that the 2-AG-arachidonate-prostaglandin axis could drive aggressive melanoma progression. METHODS The genetically engineered melanoma mouse model B6-Tyr::CreERT2; BRafCA; PtenloxP was characterized by targeted metabolomics. Functionally expressed serine hydrolases in the tumor tissue were identified by chemoproteomics. Pharmacological inhibition of carboxylesterase 1 (CES1) was achieved through chronic in vivo i.p. treatment with JZL184 (10 mg/kg daily), confirmed by activity-based protein profiling (ABPP) and targeted lipidomics. CES1-mediated 2-AG hydrolysis was further confirmed in radiotracer-based assays using CES1-transfected cell lines. RESULTS The diacylglycerol and protein kinase C activator 1-stearoyl-2-arachidonoyl-sn-glycerol (SAG) was significantly elevated in the nodular-like melanoma tissues, along with 2-AG and arachidonic acid (ARA), compared to normal skin. AEA and other N-acylethanolamines were decreased, while, notably, prostaglandin levels remained unchanged. Significant changes in the levels of neuromodulators and neurotransmitters, including serotonin and adenosine, were observed. Pronounced differences between serine hydrolase activity in normal skin and melanoma tissue were identified by ABPP. Intriguingly, CES1 was identified as the only 2-AG-hydrolyzing enzyme in this melanoma tissue, as MAGL and ABHD6/12 were not expressed. The MAGL inhibitor JZL184 also efficiently inhibited CES1 in vitro and in vivo, increasing glycerol esters and reducing tumor progression. Additionally, scRNA-seq data from previous studies revealed divergent MAGL/CES1 expression patterns across different human melanoma subtypes. CONCLUSIONS A role of CES1 expression in skin is demonstrated for the first time. Our study suggests that 2-AG degradation to arachidonate favors melanoma progression, either reflecting the carcinogenic role of ARA or that monoacylglycerols like 2-AG and/or other CES1 substrates may exert antitumor effects, indicating that CES1 could be a potential therapeutic target. CES1 expression and high SAG, 2-AG, and ARA levels may be a signature of specific BRAF-driven malignant melanoma subtypes which are associated with discrete metabolic adaptations.
Collapse
Affiliation(s)
- Veronika Morozova
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, CH-3012, Switzerland
| | - Daniele Pellegata
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, CH-3012, Switzerland
| | - Roch-Philippe Charles
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, CH-3012, Switzerland
| | - Jürg Gertsch
- Institute of Biochemistry and Molecular Medicine, University of Bern, Bern, CH-3012, Switzerland.
| |
Collapse
|
34
|
Sun S, Jiang M, Ma S, Ren J, Liu GH. Exploring the heterogeneous targets of metabolic aging at single-cell resolution. Trends Endocrinol Metab 2025; 36:133-146. [PMID: 39181730 DOI: 10.1016/j.tem.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 08/27/2024]
Abstract
Our limited understanding of metabolic aging poses major challenges to comprehending the diverse cellular alterations that contribute to age-related decline, and to devising targeted interventions. This review provides insights into the heterogeneous nature of cellular metabolism during aging and its response to interventions, with a specific focus on cellular heterogeneity and its implications. By synthesizing recent findings using single-cell approaches, we explored the vulnerabilities of distinct cell types and key metabolic pathways. Delving into the cell type-specific alterations underlying the efficacy of systemic interventions, we also discuss the complexity of integrating single-cell data and advocate for leveraging computational tools and artificial intelligence to harness the full potential of these data, develop effective strategies against metabolic aging, and promote healthy aging.
Collapse
Affiliation(s)
- Shuhui Sun
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing 100029, China.
| | - Mengmeng Jiang
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
| | - Shuai Ma
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium, Beijing 100101, China.
| | - Jie Ren
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium, Beijing 100101, China; Key Laboratory of RNA Innovation, Science and Engineering, China National Center for Bioinformation, Beijing 100101, China; Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing 100101, China; Sino-Danish College, University of Chinese Academy of Sciences, Beijing 101408, China; School of Future Technology, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Guang-Hui Liu
- Key Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China; Aging Biomarker Consortium, Beijing 100101, China; Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing 100053, China; Aging Translational Medicine Center, Xuanwu Hospital, Capital Medical University, Beijing 100053, China.
| |
Collapse
|
35
|
Li R, Wang H, Wang X, Yang Y, Zhong K, Zhang X, Li H. MSC-EVs and UCB-EVs promote skin wound healing and spatial transcriptome analysis. Sci Rep 2025; 15:4006. [PMID: 39893214 PMCID: PMC11787299 DOI: 10.1038/s41598-025-87592-6] [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: 11/13/2024] [Accepted: 01/20/2025] [Indexed: 02/04/2025] Open
Abstract
Extracellular vesicles (EVs) are important paracrine mediators derived from various cells and biological fluids, including plasma, that are capable of inducing regenerative effects by transferring bioactive molecules such as microRNAs (miRNAs). This study investigated the effect of mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) isolated from umbilical cord blood and human umbilical cord plasma-derived extracellular vesicles (UCB-EVs) on wound healing and scar formation reduction. Spatial transcriptomics (ST) was used to study the effects of MSC-EVs and UCB-EVs on the heterogeneity of major cell types and wound healing pathways in mouse skin tissue. MSC-EVs and UCB-EVs were isolated using ultracentrifugation and identified using transmission electron microscopy, nanoparticle tracking analysis, and western blot. The effects of MSC-EVs and UCB-EVs on human dermal fibroblast-adult cell (HDF-a) migration and proliferation were evaluated using cell scratch assays, cell migration assays, and cell proliferation assays. In vivo, MSC-EVs and UCB-EVs were injected around full-cut wounds to evaluate their efficacy of wound healing by measuring wound closure rates and scar width and performing histological analysis. ST was performed on skin tissue samples from mice in each group after wound healing to analyze the heterogeneity of major cell types compared with the control group and investigate potential mechanisms affecting wound healing and scar formation. In vitro experiments demonstrated that MSC-EVs and UCB-EVs promoted the proliferation and migration of HDF-a cells. Local injection of MSC-EVs and UCB-EVs into the periphery of a mouse skin wound accelerated re-epithelialization, promoted wound healing, and reduced scar width. ST analysis of skin tissue from each group after wound healing revealed that MSC-EVs and UCB-EVs reduced the relative expression of marker genes in myofibroblasts, regulated wound healing, and decreased scar formation by reducing the expression of the TGF-β signaling pathway and increasing the expression of the Wnt signaling pathway. The results suggest that MSC-EVs and UCB-EVs play a significant role in the activity of cord blood plasma-derived mesenchymal stem cells and cord blood plasma. They can be considered promising new agents for promoting skin wound healing.
Collapse
Affiliation(s)
- Ruonan Li
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Haotian Wang
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Xiaolong Wang
- HenanYinfeng Biological Engineering Technology Co., LTD, No. 11 Changchun Road, Zhengzhou High tech Industrial Development Zone, Zhengzhou, 450000, China
| | - Yanbin Yang
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China
| | - Kai Zhong
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China.
| | - Xuemei Zhang
- HenanYinfeng Biological Engineering Technology Co., LTD, No. 11 Changchun Road, Zhengzhou High tech Industrial Development Zone, Zhengzhou, 450000, China.
| | - Heping Li
- Key Laboratory of Animal Biochemistry and Nutrition of Ministry of Agriculture and Rural Affairs, College of Veterinary Medicine, Henan Agricultural University, No.15 Longzihu University Area, Zhengdong New District, Zhengzhou, 450046, China.
| |
Collapse
|
36
|
Alkon N, Chennareddy S, Cohenour ER, Ruggiero JR, Stingl G, Bangert C, Rindler K, Bauer WM, Weninger W, Griss J, Jonak C, Brunner PM. Single-cell sequencing delineates T-cell clonality and pathogenesis of the parapsoriasis disease group. J Allergy Clin Immunol 2025; 155:461-478. [PMID: 39278361 DOI: 10.1016/j.jaci.2024.09.004] [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: 03/27/2024] [Revised: 07/30/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
BACKGROUND Mycosis fungoides (MF), the most common cutaneous T-cell lymphoma, is often underdiagnosed in early stages because of similarities with benign dermatoses such as atopic dermatitis (AD). Furthermore, the delineation from what is called "parapsoriasis en plaque", a disease that can appear either in a small- or large-plaque form, is still controversial. OBJECTIVE We sought to characterize the parapsoriasis disease spectrum. METHODS We performed single-cell RNA sequencing of skin biopsies from patients within the parapsoriasis-to-early-stage MF spectrum, stratified for small and large plaques, and compared them to AD, psoriasis, and healthy control skin. RESULTS Six of 8 large-plaque lesions harbored either an expanded alpha/beta or gamma/delta T-cell clone with downregulation of CD7 expression, consistent with a diagnosis of early-stage MF. In contrast, 6 of 7 small-plaque lesions were polyclonal in nature, thereby lacking a lymphomatous phenotype, and also revealed a less inflammatory microenvironment than early-stage MF or AD. Of note, polyclonal small- and large-plaque lesions characteristically harbored a population of NPY+ innate lymphoid cells and displayed a stromal signature of complement upregulation and antimicrobial hyperresponsiveness in fibroblasts and sweat gland cells, respectively. These conditions were clearly distinct from AD or psoriasis, which uniquely harbored CD3+CRTH2+ IL-13 expressing "TH2A" cells, or strong type 17 inflammation, respectively. CONCLUSION These data position polyclonal small- and large-plaque parapsoriasis lesions as a separate disease entity that characteristically harbors a so far undescribed innate lymphoid cell population. We thus propose a new term, "polyclonal parapsoriasis en plaque", for this kind of lesion because they can be clearly differentiated from early- and advanced-stage MF, psoriasis, and AD on several cellular and molecular levels.
Collapse
Affiliation(s)
- Natalia Alkon
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Sumanth Chennareddy
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Emry R Cohenour
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - John R Ruggiero
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Georg Stingl
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Christine Bangert
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Katharina Rindler
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang M Bauer
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Weninger
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Johannes Griss
- Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Constanze Jonak
- Department of Dermatology, Medical University of Vienna, Vienna, Austria.
| | - Patrick M Brunner
- Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY.
| |
Collapse
|
37
|
Hughes BK, Davis A, Milligan D, Wallis R, Mossa F, Philpott MP, Wainwright LJ, Gunn DA, Bishop CL. SenPred: a single-cell RNA sequencing-based machine learning pipeline to classify deeply senescent dermal fibroblast cells for the detection of an in vivo senescent cell burden. Genome Med 2025; 17:2. [PMID: 39810225 PMCID: PMC11731430 DOI: 10.1186/s13073-024-01418-0] [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: 06/08/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Senescence classification is an acknowledged challenge within the field, as markers are cell-type and context dependent. Currently, multiple morphological and immunofluorescence markers are required. However, emerging scRNA-seq datasets have enabled an increased understanding of senescent cell heterogeneity. METHODS Here we present SenPred, a machine-learning pipeline which identifies fibroblast senescence based on single-cell transcriptomics from fibroblasts grown in 2D and 3D. RESULTS Using scRNA-seq of both 2D and 3D deeply senescent fibroblasts, the model predicts intra-experimental fibroblast senescence to a high degree of accuracy (> 99% true positives). Applying SenPred to in vivo whole skin scRNA-seq datasets reveals that cells grown in 2D cannot accurately detect fibroblast senescence in vivo. Importantly, utilising scRNA-seq from 3D deeply senescent fibroblasts refines our ML model leading to improved detection of senescent cells in vivo. This is context specific, with the SenPred pipeline proving effective when detecting senescent human dermal fibroblasts in vivo, but not the senescence of lung fibroblasts or whole skin. CONCLUSIONS We position this as a proof-of-concept study based on currently available scRNA-seq datasets, with the intention to build a holistic model to detect multiple senescent triggers using future emerging datasets. The development of SenPred has allowed for the detection of an in vivo senescent fibroblast burden in human skin, which could have broader implications for the treatment of age-related morbidities. All code for the SenPred pipeline is available at the following URL: https://github.com/bethk-h/SenPred_HDF .
Collapse
Affiliation(s)
- Bethany K Hughes
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Andrew Davis
- Unilever R&D, Merritt Blvd, Trumbull, CT, 06611, USA
| | - Deborah Milligan
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Ryan Wallis
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Federica Mossa
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Michael P Philpott
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Linda J Wainwright
- Unilever R&D, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - David A Gunn
- Unilever R&D, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Cleo L Bishop
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| |
Collapse
|
38
|
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
|
39
|
Wang Y, Wang X, Yuan Z, Liu F, Luo X, Yang J. Identifying Potential Drug Targets for Keloid: A Mendelian Randomization Study. J Invest Dermatol 2025; 145:77-84.e6. [PMID: 38797322 DOI: 10.1016/j.jid.2024.04.023] [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: 12/06/2023] [Revised: 04/03/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024]
Abstract
Keloids are a skin fibrosis disease characterized by troublesome symptoms, a varying degree of recurrence and inevitable side effects from treatments. Thus, identifying their drug targets is necessary. A 2-sample Mendelian randomization analysis was conducted using proteins from the intersection of the deCODE database and "The Druggable Genome and Support for Target Identification and Validation in Drug Development" as the exposure variable. The outcome variable was based on recently published GWAS of keloids. Summary data-based Mendelian randomization and colocalization analysis was employed to distinguish pleiotropy from linkage. Candidate targets underwent drug target analysis. The primary findings were validated through single-cell RNA-sequencing data, Western Blot and immunofluorescence staining on keloids. Seven proteins were identified as potential drug targets for keloids. Among these proteins, Hedgehog-interacting protein, neurotrimin [NTM], KLKB1, and CRIPTO showed positive correlations with keloids, while PLXNC1, SCG3 and PDGFD exhibited negative correlations. Combined with the single-cell RNA-sequencing data, NTM, PLXNC1, and PDGFD were found highly expressed in the fibroblasts. NTM showed a significant increase in keloids as compared to normal scars. In accordance with the analysis, higher levels of protein expression of NTM in keloids compared to normal skin was observed. The identified proteins may be appealing drug targets for keloids treatment with a special emphasis on NTM.
Collapse
Affiliation(s)
- Yinmin Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiuxia Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhaoqi Yuan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; The First People's Hospital of the Lancang Lahu Autonomous County, Yunnan, China
| | - Xusong Luo
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Jun Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
40
|
Smith NJ, Reddin I, Policelli P, Oh S, Zainal N, Howes E, Jenkins B, Tracy I, Edmond M, Sharpe B, Amendra D, Zheng K, Egawa N, Doorbar J, Rao A, Mahadevan S, Carpenter MA, Harris RS, Ali S, Hanley C, Buisson R, King E, Thomas GJ, Fenton TR. Differentiation signals induce APOBEC3A expression via GRHL3 in squamous epithelia and squamous cell carcinoma. EMBO J 2025; 44:1-29. [PMID: 39548236 PMCID: PMC11696371 DOI: 10.1038/s44318-024-00298-9] [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: 05/11/2024] [Revised: 10/21/2024] [Accepted: 10/24/2024] [Indexed: 11/17/2024] Open
Abstract
Two APOBEC DNA cytosine deaminase enzymes, APOBEC3A and APOBEC3B, generate somatic mutations in cancer, thereby driving tumour development and drug resistance. Here, we used single-cell RNA sequencing to study APOBEC3A and APOBEC3B expression in healthy and malignant mucosal epithelia, validating key observations with immunohistochemistry, spatial transcriptomics and functional experiments. Whereas APOBEC3B is expressed in keratinocytes entering mitosis, we show that APOBEC3A expression is confined largely to terminally differentiating cells and requires grainyhead-like transcription factor 3 (GRHL3). Thus, in normal tissue, neither deaminase appears to be expressed at high levels during DNA replication, the cell-cycle stage associated with APOBEC-mediated mutagenesis. In contrast, in squamous cell carcinoma we find that, there is expansion of GRHL3expression and activity to a subset of cells undergoing DNA replication and concomitant extension of APOBEC3A expression to proliferating cells. These findings suggest that APOBEC3A may play a functional role during keratinocyte differentiation, and offer a mechanism for acquisition of APOBEC3A mutagenic activity in tumours.
Collapse
Affiliation(s)
- Nicola J Smith
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- School of Biosciences, University of Kent, Canterbury, UK
| | - Ian Reddin
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Bio-R Bioinformatics Research Facility, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Paige Policelli
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Cell, Gene and RNA Therapies, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Sunwoo Oh
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Nur Zainal
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Emma Howes
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Benjamin Jenkins
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ian Tracy
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Mark Edmond
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Benjamin Sharpe
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Damian Amendra
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Ke Zheng
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Nagayasu Egawa
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - John Doorbar
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Anjali Rao
- Gilead Sciences, Research Department, 324 Lakeside Dr, Foster City, CA, 94404, USA
| | - Sangeetha Mahadevan
- Gilead Sciences, Research Department, 324 Lakeside Dr, Foster City, CA, 94404, USA
| | - Michael A Carpenter
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Reuben S Harris
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
- Howard Hughes Medical Institute, University of Texas Health San Antonio, San Antonio, TX, 78229, USA
| | - Simak Ali
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Christopher Hanley
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Rémi Buisson
- Department of Biological Chemistry, School of Medicine, University of California Irvine, Irvine, CA, USA
| | - Emma King
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
| | - Gareth J Thomas
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK
- Institute for Life Sciences, University of Southampton, Southampton, UK
| | - Tim R Fenton
- School of Cancer Sciences, Faculty of Medicine, University of Southampton, Southampton, UK.
- Institute for Life Sciences, University of Southampton, Southampton, UK.
| |
Collapse
|
41
|
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
|
42
|
Chen R, Zou J, Chen J, Wang L, Kang R, Tang D. Immune aging and infectious diseases. Chin Med J (Engl) 2024; 137:3010-3049. [PMID: 39679477 PMCID: PMC11706578 DOI: 10.1097/cm9.0000000000003410] [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: 09/20/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT The rise in global life expectancy has led to an increase in the older population, presenting significant challenges in managing infectious diseases. Aging affects the innate and adaptive immune systems, resulting in chronic low-grade inflammation (inflammaging) and immune function decline (immunosenescence). These changes would impair defense mechanisms, increase susceptibility to infections and reduce vaccine efficacy in older adults. Cellular senescence exacerbates these issues by releasing pro-inflammatory factors, further perpetuating chronic inflammation. Moreover, comorbidities, such as cardiovascular disease and diabetes, which are common in older adults, amplify immune dysfunction, while immunosuppressive medications further complicate responses to infections. This review explores the molecular and cellular mechanisms driving inflammaging and immunosenescence, focusing on genomic instability, telomere attrition, and mitochondrial dysfunction. Additionally, we discussed how aging-associated immune alterations influence responses to bacterial, viral, and parasitic infections and evaluated emerging antiaging strategies, aimed at mitigating these effects to improve health outcomes in the aging population.
Collapse
Affiliation(s)
- Ruochan Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ju Zou
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Jiawang Chen
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Ling Wang
- Department of Infectious Diseases, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- National Clinical Research Center for Geriatric Disorders (Xiangya), Changsha, Hunan 410008, China
| | - Rui Kang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75235, USA
| |
Collapse
|
43
|
Mao R, Zhang T, Yang Z, Li J. Unveiling Novel Protein Biomarkers for Psoriasis Through Integrated Analysis of Human Plasma Proteomics and Mendelian Randomization. PSORIASIS (AUCKLAND, N.Z.) 2024; 14:179-193. [PMID: 39669686 PMCID: PMC11635628 DOI: 10.2147/ptt.s492205] [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: 08/19/2024] [Accepted: 11/26/2024] [Indexed: 12/14/2024]
Abstract
Background Current pharmacological treatments for psoriasis are generally non-specific and have significant limitations, particularly in the realm of targeted biologic therapies. There is an urgent need to identify and develop new therapeutic targets to improve treatment options. Objective The aim of this study was to explore the proteome associated with psoriasis in large population cohorts to discover novel biomarkers that could guide therapy. Methods We analyzed data from 54,306 participants enrolled in the UK Biobank Pharmacological Proteomics Project (UKB-PPP). We investigated the relationship between 2923 serum proteins and the risk of psoriasis using multivariate Cox regression models initially. This was complemented by two-sample Mendelian randomization (TSMR), Summary-data-based Mendelian Randomization (SMR), and coloc colocalization studies to identify genetic correlations with protein targets linked to psoriasis. A protein scoring system was created using the Cox proportional hazards model, and cumulative risk curves were generated to analyze psoriasis incidence variations. Results Our study pinpointed 62 proteins significantly linked to the risk of developing psoriasis. Further analysis through TSMR narrowed these down to ten proteins with strong causal relationships to the disease. Additional deep-dive analyses such as SMR, colocalization, and differential expression studies highlighted four critical proteins (MMP12, PCSK9, PRSS8, and SCLY). We calculated a protein score based on the levels of these proteins, with higher scores correlating with increased risk of psoriasis. Conclusion This study's integration of proteomic and genetic data from a European adult cohort provides compelling evidence of several proteins as viable predictive biomarkers and potential therapeutic targets for psoriasis, facilitating the advancement of targeted treatment strategies.
Collapse
Affiliation(s)
- Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Tongtong Zhang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People’s Hospital of Chengdu, Affiliated Hospital of Southwest Jiaotong University & The Second Affiliated Hospital of Chengdu, Chongqing Medical University, Chengdu, 610031, People’s Republic of China
| | - Ziye Yang
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, People’s Republic of China
| |
Collapse
|
44
|
Schiavinato A, Marcous F, Zuk AV, Keene DR, Tufa SF, Mosquera LM, Zigrino P, Mauch C, Eckes B, Francois K, De Backer J, Hunzelmann N, Moinzadeh P, Krieg T, Callewaert B, Sengle G. New insights into the structural role of EMILINs within the human skin microenvironment. Sci Rep 2024; 14:30345. [PMID: 39639116 PMCID: PMC11621341 DOI: 10.1038/s41598-024-81509-5] [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: 04/23/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024] Open
Abstract
Supramolecular extracellular matrix (ECM) networks play an essential role in skin architecture and function. Elastin microfibril interface-located proteins (EMILINs) comprise a family of three extracellular glycoproteins that serve as essential structural components of the elastin/fibrillin microfibril network, and exert crucial functions in cellular signaling. Little is known about the structural nature of EMILIN networks in skin. We therefore investigated the spatiotemporal localization of EMILIN-1, -2, -3 in human skin induced by aging, UV-exposure, fibrosis, and connective tissue disorder. Confocal immunofluorescence and immunogold electron microscopy analysis identified all EMILINs as components of elastic fibers and elastin-free oxytalan fibers inserted into the basement membrane (BM). Further, our ultrastructural analysis demonstrates cellular contacts of dermally localized EMILIN-1 positive fibers across the BM with the surface of basal keratinocytes. Analysis of skin biopsies and fibroblast cultures from fibrillin-1 deficient Marfan patients revealed that EMILINs require intact fibrillin-1 as deposition scaffold. In patients with scleroderma and the bleomycin-induced murine fibrosis model EMILIN-2 was upregulated. EMILIN-3 localizes to the tips of candelabra-like oxytalan fibers, and to specialized BMs engulfing hair follicles and sebaceous glands. Our data identify EMILINs as important markers to monitor rearrangements of the dermal ECM architecture induced by aging and pathological conditions.
Collapse
Affiliation(s)
- Alvise Schiavinato
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, 50931, Cologne, Germany
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
| | - Fady Marcous
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, 50931, Cologne, Germany
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Alexandra V Zuk
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Douglas R Keene
- Micro-Imaging Center, Shriners Children's, Portland, OR, 97239, USA
| | - Sara F Tufa
- Micro-Imaging Center, Shriners Children's, Portland, OR, 97239, USA
| | - Laura M Mosquera
- Center for Medical Genetics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Pediatrics, Division of Pediatric Cardiology, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000, Ghent, Belgium
| | - Paola Zigrino
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Cornelia Mauch
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Beate Eckes
- Translational Matrix Biology, Faculty of Medicine, University Hospital Cologne, 50931, Cologne, Germany
| | - Katrien Francois
- Department of Cardiovascular Surgery, Ghent University Hospital, 9000, Ghent, Belgium
| | - Julie De Backer
- Center for Medical Genetics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Cardiology, Ghent University Hospital, 9000, Ghent, Belgium
| | - Nicolas Hunzelmann
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Pia Moinzadeh
- Department of Dermatology, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany
| | - Thomas Krieg
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany
- Translational Matrix Biology, Faculty of Medicine, University Hospital Cologne, 50931, Cologne, Germany
- Cologne Excellence Cluster On Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany
| | - Bert Callewaert
- Center for Medical Genetics, Ghent University Hospital, 9000, Ghent, Belgium
- Department of Biomolecular Medicine, Ghent University, 9000, Ghent, Belgium
| | - Gerhard Sengle
- Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Joseph-Stelzmann-Str. 52, 50931, Cologne, Germany.
- Center for Biochemistry, Faculty of Medicine and University Hospital Cologne, University of Cologne, 50931, Cologne, Germany.
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50931, Cologne, Germany.
- Cologne Center for Musculoskeletal Biomechanics (CCMB), 50931, Cologne, Germany.
| |
Collapse
|
45
|
Prakoso NM, Sundari AM, Fadhilah, Abinawanto, Pustimbara A, Dwiranti A, Bowolaksono A. Methylglyoxal impairs human dermal fibroblast survival and migration by altering RAGE-hTERT mRNA expression in vitro. Toxicol Rep 2024; 13:101835. [PMID: 39687678 PMCID: PMC11646750 DOI: 10.1016/j.toxrep.2024.101835] [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: 08/24/2024] [Revised: 11/14/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Fibroblasts are native residents in dermal layer of human skin which are important for dermal regeneration and essential during cutaneous wound healing by releasing inflammatory markers and actively migrate to close an open wound. Premature skin ageing due to methylglyoxal (MGO) has recently caught the attention considering its potential to accelerate the emergence of skin ageing signs, however previous studies were only focused in primary neonatal dermal fibroblast and NIH3t3 fibroblast cell line. Therefore, thorough investigation is required to study the impact of MGO on primary human dermal fibroblast isolated from adult subject (HDFa). In our experiments, short exposure of MGO was observed to induced significant reductions in cell viability at concentrations of 7.5, 10, 12.5, 15, and 17.5 mM (p < 0.005) after 3 hours of treatment. The cellular death of HDFa at 10, 12.5 and 15 mM of MGO were also marked by increased in intracellular ROS level, indicating the involvement of oxidative stress-induced death in these cells. We also observed enlarge scratch areas of cells exposed with 7.5 and 10 mM MGO compared to control after 26 hours, thereby suggesting a decline in cell migration and viability in this group. We propose the increased ROS as the consequence of AGE-RAGE activation which was marked by significant elevation of RAGE mRNA on cells exposed to 10 mM MGO. Our data also suggest the occurrence of DNA damage events via ROS-induced oxidation or mediated by decline in hTERT mRNA expression.
Collapse
Affiliation(s)
- Nurul Muhammad Prakoso
- Master Program in Biology, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Ayu Mulia Sundari
- Master Program in Biology, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Fadhilah
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Abinawanto
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Anantya Pustimbara
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama 2268501, Japan
- Center for Photodynamic Medicine, Kochi Medical School, Kohasu, Oko-cho, Nankoku-shi, Kochi 783-8505, Japan
| | - Astari Dwiranti
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| | - Anom Bowolaksono
- Cellular and Molecular Mechanisms in Biological System (CEMBIOS) Research Group, Department of Biology, Faculty of Mathematics and Natural Science, University of Indonesia, Depok, West Java 16424, Indonesia
| |
Collapse
|
46
|
Park SS, Lee YK, Kim YH, Park SH, Kang HY, Kim JC, Kim DJ, Lim SB, Yoon G, Kim JH, Choi YW, Park TJ. Distribution and impact of p16 INK4A+ senescent cells in elderly tissues: a focus on senescent immune cell and epithelial dysfunction. Exp Mol Med 2024; 56:2631-2641. [PMID: 39617789 DOI: 10.1038/s12276-024-01354-4] [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: 05/23/2024] [Revised: 08/26/2024] [Accepted: 09/11/2024] [Indexed: 12/28/2024] Open
Abstract
Cellular senescence, recognized as a key hallmark of aging, leads to the accumulation of senescent cells in various tissues over time. While the detrimental effects of these cells on age-related pathological conditions are well-documented, there is still limited information about how senescent cells are distributed in normal tissues of both young and aged organs. Our research indicates that fully senescent p16INK4A+ cells are rarely identified in the parenchyma of organic tissues and in the stromal cells crucial for structural maintenance, such as fibroblasts and smooth muscle cells. Instead, p16INK4A+ cells are more commonly found in immune cells, whether they reside in the organ or are infiltrating. Notably, p16INK4A+ senescent T cells have been observed to induce apoptosis and inflammation in colonic epithelial cells through Granzyme A-PARs signaling, compromising the integrity of the epithelial lining. This study showed that the senescence of immune cells could affect the phenotypical change of the parenchymal cells in the elderly and suggests that targeting immunosenescence might be a strategy to control functional decline in this population.
Collapse
Affiliation(s)
- Soon Sang Park
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea
| | - Young-Kyoung Lee
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea
| | - Young Hwa Kim
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea
| | - So Hyun Park
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Hee Young Kang
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea
- Department of Dermatology, Ajou University School of Medicine, Suwon, Korea
| | - Jin Cheol Kim
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea
- Department of Dermatology, Ajou University School of Medicine, Suwon, Korea
| | - Dong Jun Kim
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea
| | - Su Bin Lim
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Gyesoon Yoon
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea
| | - Jang-Hee Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
| | - Yong Won Choi
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea.
- Department of Hematology and Oncology, Ajou University School of Medicine, Suwon, Korea.
| | - Tae Jun Park
- Inflammaging Translational Research Center, Ajou University Medical Center, Suwon, Korea.
- Department of Biochemistry and Molecular Biology, Ajou University School of Medicine, Suwon, Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.
| |
Collapse
|
47
|
Li D, Li M, Gao H, Hu K, Xie R, Fan J, Huang M, Liao C, Han C, Guo Z, Chen X, Li M. Integrative multiomics analysis reveals association of gut microbiota and its metabolites with susceptibility to keloids. Front Microbiol 2024; 15:1475984. [PMID: 39669776 PMCID: PMC11636970 DOI: 10.3389/fmicb.2024.1475984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024] Open
Abstract
Keloid scarring is a fibroproliferative disease of the skin, which can significantly impact one's quality of life through cosmetic concerns, physical discomfort (itchy; painful), restricted movement, and psychological distress. Owing to the poorly understood pathogenesis of keloids and their high recurrence rate, the efficacy of keloid treatment remains unsatisfactory, particularly in patients susceptible to multiple keloids. We conducted fecal metagenomic analyzes and both untargeted and targeted plasma metabolomics in patients with multiple keloids (MK, n = 56) and controls with normal scars (NS, n = 60); tissue-untargeted metabolomics (MK, n = 35; NS, n = 32), tissue-targeted metabolomics (MK, n = 41; NS, n = 36), and single-cell sequencing analyzes (GSE163973). Differences in the gut microbiota composition, plasma metabolites, and tissue metabolites were observed between the MK and NS groups; the core gut microbiota, Oxalobacter formigenes, Bacteroides plebeius, and Parabacteroides distasonis, were identified via the gut microbiome co-occurrence network. Single-cell data helped clarify the specific cells affected by plasma metabolites. An area under the curve analysis using a random forest model based on fecal metagenomics, plasma metabolomics, and tissue metabolomics revealed that gut bacteria, plasma, and tissue metabolites were effective in distinguishing between MK and NS groups. Decreased Bacteroides plebeius could lower uracil levels, altering systemic lipid metabolism, which may change the metabolic phenotype of secretory reticular fibroblasts in wounds, potentially leading to MK. These findings may open new avenues for understanding the multifactorial nature of keloid formation from the gut-skin axis and highlight the potential for novel therapeutic strategies targeting keloid lesions and the underlying systemic imbalances affected by the gut microbiome.
Collapse
Affiliation(s)
- Dang Li
- Nursing Department of Fujian Medical University Union Hospital, Fuzhou, China
| | - Minghao Li
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Hangqi Gao
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Kailun Hu
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Rongrong Xie
- Department of Plastic Surgery, The Second Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jing Fan
- Department of Gynecology, Fuzhou Children’s Hospital of Fujian Medical University, Fuzhou, China
| | - Mingquan Huang
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Chengxin Liao
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Chang Han
- Shanghai Majorbio Bio-Pharm Technology Co., Ltd., Shanghai, China
| | - Zhihui Guo
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Xiaosong Chen
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| | - Ming Li
- Department of Plastic Surgery and Regenerative Medicine, Fujian Medical University Union Hospital, Fuzhou, China
- Department of Plastic Surgery and Regenerative Medicine Institute, Fujian Medical University, Fuzhou, China
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, China
| |
Collapse
|
48
|
Dao Nyesiga G, Haslund-Vinding JL, Budde J, Lange JF, Blum N, Dukstaite K, Ohlsson L, Mathiesen T, Woetmann A, Vilhardt F. Flow Cytometry Analyses of Meningioma Immune Cell Composition Using a Short, Optimized Digestion Protocol. Cancers (Basel) 2024; 16:3942. [PMID: 39682129 DOI: 10.3390/cancers16233942] [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: 10/12/2024] [Revised: 11/19/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND Current challenges in meningioma treatment, including post-surgical complications and cognitive impairments, highlight the need for new treatment alternatives. Immunological interventions have shown promise. However, there is a knowledge gap in characterizing infiltrating immune cells in meningioma and their interplay. Further studies on immune cells in single-cell suspensions from digested meningioma tissues could identify targetable mechanisms for non-surgical treatment options with fewer side effects. This study aimed to optimize a protocol for faster digestion of meningioma tissues into viable single-cell suspensions and to identify infiltrating immune cell populations. METHODS We modified a commercial kit intended for whole skin dissociation to digest resected meningioma tissues into viable single-cell suspensions. Tumor-infiltrating immune cell populations were characterized using flow cytometry. RESULTS Flow cytometry analyses revealed that the digested tissue was composed of viable immune cells, including predominantly CD14+ macrophages and CD3+ T cells, with minor populations of CD56+ NK cells and CD19+ B cells. In both of the two patient samples tested, half of the tumor-associated macrophages were TIM-3+, with a small proportion co-expressing CD83. Women were more likely to have a lower proportion of immune cells, B cells, and NK cells. Female patients with a high proportion of immune cells had a higher proportion of macrophages. CONCLUSION We successfully optimized a protocol for generating single-cell suspensions with viable immune cells from meningioma tissues, revealing infiltrating antigen-presenting cells with an immunosuppressive phenotype, and lymphocytes. This short protocol allows advanced analyses of tumor-infiltrating cells using techniques such as single-cell RNA sequencing and flow cytometry, which require live, dissociated cells.
Collapse
Affiliation(s)
- Gillian Dao Nyesiga
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, 205 06 Malmo, Sweden
- Department of Neurosurgery, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark
| | | | - Josephine Budde
- Department of Neurosurgery, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark
| | - Josefine Føns Lange
- Department of Neurosurgery, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark
| | - Nadja Blum
- Department of Neurosurgery, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Kotryna Dukstaite
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Lars Ohlsson
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, 205 06 Malmo, Sweden
| | - Tiit Mathiesen
- Department of Neurosurgery, Copenhagen University Hospital-Rigshospitalet, 2100 Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences SUND, University of Copenhagen, 2200 Copenhagen, Denmark
- Department of Clinical Neuroscience, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Anders Woetmann
- LEO Foundation Skin Immunology Research Center, Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Frederik Vilhardt
- Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen, Denmark
| |
Collapse
|
49
|
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
|
50
|
Liu Z, Lin H, Li X, Xue H, Lu Y, Xu F, Shuai J. The network structural entropy for single-cell RNA sequencing data during skin aging. Brief Bioinform 2024; 26:bbae698. [PMID: 39757115 DOI: 10.1093/bib/bbae698] [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: 10/19/2024] [Revised: 11/29/2024] [Accepted: 12/18/2024] [Indexed: 01/07/2025] Open
Abstract
Aging is a complex and heterogeneous biological process at cellular, tissue, and individual levels. Despite extensive effort in scientific research, a comprehensive understanding of aging mechanisms remains lacking. This study analyzed aging-related gene networks, using single-cell RNA sequencing data from >15 000 cells. We constructed a gene correlation network, integrating gene expressions into the weights of network edges, and ranked gene importance using a random walk model to generate a gene importance matrix. This unsupervised method improved the clustering performance of cell types. To further quantify the complexity of gene networks during aging, we introduced network structural entropy. The findings of our study reveal that the overall network structural entropy increases in the aged cells compared to the young cells. However, network entropy changes varied greatly within different cell subtypes. Specifically, the network structural entropy among various cell types may increase, remain unchanged, or decrease. This wide range of changes may be closely related to their individual functions, highlighting the cellular heterogeneity and potential key network reconfigurations. Analyzing gene network entropy provides insights into the molecular mechanisms behind aging. This study offers new scientific evidence and theoretical support for understanding the changes in cell functions during aging.
Collapse
Affiliation(s)
- Zhilong Liu
- Department of Physics, Xiamen University, No. 422, Siming South Road, Xiamen, Fujian, 361005, China
| | - Hai Lin
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), No. 999, Jinshi Road, Yongzhong Street, Longwan District, Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou, Zhejiang, 325000, China
| | - Xiang Li
- Department of Physics, Xiamen University, No. 422, Siming South Road, Xiamen, Fujian, 361005, China
| | - Hao Xue
- Department of Computational Biology, Cornell University, 110 Biotechnology Building, Ithaca, 14853 NY, United States
| | - Yuer Lu
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), No. 999, Jinshi Road, Yongzhong Street, Longwan District, Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou, Zhejiang, 325000, China
| | - Fei Xu
- Department of Physics, Anhui Normal University, No. 189 Jiuhua South Road, Wuhu, Anhui, 241002, China
| | - Jianwei Shuai
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), No. 999, Jinshi Road, Yongzhong Street, Longwan District, Wenzhou, Zhejiang, 325000, China; Wenzhou Institute, University of Chinese Academy of Sciences, No. 1, Jinlian Road, Longwan District, Wenzhou, Zhejiang, 325000, China
| |
Collapse
|