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
|
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
|
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
|
Bagheri AM, Mirzahashemi M, Salarpour S, Dehghnnoudeh Y, Banat IM, Ohadi M, Dehghannoudeh G. Potential anti-aging applications of microbial-derived surfactantsin cosmetic formulations. Crit Rev Biotechnol 2025; 45:766-787. [PMID: 39294002 DOI: 10.1080/07388551.2024.2393420] [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: 12/16/2023] [Revised: 07/14/2024] [Accepted: 07/30/2024] [Indexed: 09/20/2024]
Abstract
The skin aging process is a complex interaction of genetic, epigenetic, and environmental factors, such as chemical pollution and UV radiation. There is growing evidence that biosurfactants, especially those of microbial origin, have distinct age-supportive effects through different mechanisms, such as stimulation of fibroblast growth, high antioxidant capacities, and favorable anti-inflammatory properties. With a growing financial contribution of more than 15 m€per year, microbial surfactants (MSs) display unique biological effects on the skin including improved cell mobility, better nutrient access, and facilitated cellular growth under harsh conditions. Their biodegradable nature, unusual surface activity, good safety profile and tolerance to high temperature and pH variations widen their potential spectrum in biomedical and pharmaceutical applications. MSs typically have lower critical micelle concentration (CMC) levels than chemical surfactants enhancing their effectiveness. As natural surfactants, MSs are considered possible "green" alternatives to synthetic surfactants with better biodegradability, sustainability, and beneficial functional properties. This review therefore aims to explore the potential impacts of MSs as anti-aging ingredients.
Collapse
Affiliation(s)
- Amir Mohammad Bagheri
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Masoud Mirzahashemi
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Student Research Committee, Kerman University of Medical Sciences, Kerman, Iran
| | - Soodeh Salarpour
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Yasmin Dehghnnoudeh
- Departeman of Biology, Faculty of Science, York University, Toronto, Ontario, Canada
| | - Ibrahim M Banat
- School of Biomedical Sciences, Faculty of Life & Health Sciences, Ulster University, Coleraine, N. Ireland, UK
| | - Mandana Ohadi
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Gholamreza Dehghannoudeh
- Department of Pharmaceutics, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman, Iran
- Pharmaceutics Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
5
|
Torres M, Silberberg G, Vegvari A, Zubarev RA, Hunt M, Bansal R, Bachar-Wikstrom E, Wikstrom JD. The Temporal Dynamics of Proteins in Aged Skin Wound Healing and Comparison with Gene Expression. J Invest Dermatol 2025; 145:1534-1537.e9. [PMID: 39675661 DOI: 10.1016/j.jid.2024.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 07/28/2024] [Accepted: 09/12/2024] [Indexed: 12/17/2024]
Affiliation(s)
- Monica Torres
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Gilad Silberberg
- Champions Oncology, One University Plaza, Hackensack, New Jersey, USA
| | - Akos Vegvari
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Roman A Zubarev
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Matthew Hunt
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Ritu Bansal
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden; Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
6
|
Yang Z, Han X, Xing Z, He F, Qi T, Wang X, Fu R, Du C, Feng X, Wang Y, Yuan Q, Li F, Lan W, Xu Y. Combining transcriptomics and metabolomics to analyse the mechanism of allelopathy in Cyclachaena xanthiifolia. BMC PLANT BIOLOGY 2025; 25:660. [PMID: 40389813 PMCID: PMC12087043 DOI: 10.1186/s12870-025-06704-6] [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] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 05/12/2025] [Indexed: 05/21/2025]
Abstract
As a vicious invasive plant, Cyclachaena xanthiifolia has caused severe ecological disruption and significant reductions in crop yield, necessitating urgent control measures. However, the underlying mechanisms of its allelopathic invasion remain unclear, representing the primary bottleneck in current management strategies. In this study, we used metabolomic and transcriptomic analyses to evaluate the differences in allelopathy and related physiological and biochemical indices among different extract fractions of C.xanthiifolia, and to investigate how the allelopathy of C.xanthiifolia inhibits seed germination and seedling growth by altering metabolic pathways. GC-MS results identified several compounds with allelopathic potential, including fatty acids, terpenes, esters, alkanes, and aldehydes. Among them, n-butanol phase extract (NE) treatment significantly inhibited the germination and water absorption of mustard (Brassica juncea) seeds, changed the balance of the endogenous hormones abscisic acid (ABA) and gibberellins (GA) in seeds, destroyed the antioxidant enzyme system, and caused plasma membrane damage. Moreover, transcriptomic and broadly targeted metabolomic analyses showed that NE treatment interfered with primary metabolism, significantly enriched the carotenoid biosynthetic pathway, and led to a significant accumulation of ABA. The quantitative real-time PCR (qRT-PCR) results showed that the expression levels of 7 key genes involved in ABA biosynthesis and metabolic pathways were relatively high. The results showed that C.xanthiifolia may exert its allelopathic effects by disrupting the antioxidant enzyme system and interfering with primary metabolism and hormone signalling, and that the modulation of the ABA signalling pathway appears to play a key role.
Collapse
Affiliation(s)
- Zelin Yang
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Xiaoling Han
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Zhixiang Xing
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Fumeng He
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Tianshuai Qi
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Xue Wang
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Rao Fu
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Chong Du
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Xu Feng
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Yingnan Wang
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Qiang Yuan
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China
| | - Fenglan Li
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China.
- Heilongjiang Academy of Green Food Science, Harbin, 150023, China.
| | - Wei Lan
- School of Biology and Food Engineering, Fuyang Normal University, Fuyang, Anhui, 236037, P. R. China.
- Anhui Engineering Research Center for Functional Fruit Drink and Ecological Fermentation, Fuyang, Anhui, 236037, P. R. China.
| | - Yongqing Xu
- College of Life Sciences, Northeast Agricultural University, Harbin, 150030, China.
| |
Collapse
|
7
|
Zhang Y, Naaz A, Cheng TYN, Lin JJ, Gao M, Dorajoo R, Alfatah M. Systematic transcriptomics analysis of calorie restriction and rapamycin unveils their synergistic interaction in prolonging cellular lifespan. Commun Biol 2025; 8:753. [PMID: 40369174 PMCID: PMC12078523 DOI: 10.1038/s42003-025-08178-6] [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: 12/07/2023] [Accepted: 05/06/2025] [Indexed: 05/16/2025] Open
Abstract
Aging is a multifaceted biological process marked by the decline in both mitotic and postmitotic cellular function, often central to the development of age-related diseases. In the pursuit of slowing or even reversing the aging process, a prominent strategy of significant interest is calorie restriction (CR), also known as dietary restriction, and the potential influence of a drug called rapamycin (RM). Both CR and RM have demonstrated the capacity to extend healthspan and lifespan across a diverse array of species, including yeast, worms, flies, and mice. Nevertheless, their individual and combined effects on mitotic and postmitotic cells, as well as their comparative analysis, remain areas that demand a thorough investigation. In this study, we employ RNA-sequencing methodologies to comprehensively analyze the impact of CR, RM, and their combination (CR + RM) on gene expression in yeast cells. Our analysis uncovers distinctive, overlapping, and even contrasting patterns of gene regulation, illuminating the unique and shared effects of CR and RM. Furthermore, the transcriptional synergistic interaction of CR + RM is validated in extending the lifespan of both yeast and human cells.
Collapse
Affiliation(s)
- Yizhong Zhang
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Arshia Naaz
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Trishia Yi Ning Cheng
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jovian Jing Lin
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mingtong Gao
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Healthy Longevity, National University Health System, Singapore, Singapore
| | - Rajkumar Dorajoo
- Genome Institute of Singapore (GIS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Mohammad Alfatah
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Healthy Longevity Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Centre for Healthy Longevity, National University Health System, Singapore, Singapore.
| |
Collapse
|
8
|
Zhang X, Shi B, Zhao Z, Deng Y, Zhou X, Hu J. Deciphering the Transcriptomic Complexity of Yak Skin Across Different Ages and Body Sites. Int J Mol Sci 2025; 26:4601. [PMID: 40429746 PMCID: PMC12111109 DOI: 10.3390/ijms26104601] [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: 02/24/2025] [Revised: 05/08/2025] [Accepted: 05/09/2025] [Indexed: 05/29/2025] Open
Abstract
Differences in skin and hair phenotypes between the scapular and ventral regions of yaks (Bos grunniens) are obvious and become more prominent with age. However, the genetic mechanism that causes differences in yak skin at different ages has not been reported. In this study, we investigated the transcriptomic profile of yak skin across different ages (0.5 years, 2.5 years, and 4.5 years) and body sites (scapular and ventral regions). Differential gene expression analysis was initially conducted to explore the transcriptomic differences in skin at different ages and different body sites. Subsequently, weighted gene co-expression network analysis (WGCNA) was employed to analyze the transcriptomic data comprehensively. The results showed that, among all comparison groups, the Y2.5_S vs. Y2.5_V group (regional comparison) exhibited the highest number of DEGs, with 491 genes (179 upregulated and 312 downregulated), followed by the Y2.5_V vs. Y0.5_V group (age comparison), which had 370 DEGs (103 upregulated and 267 downregulated). DEGs such as IGF2BP3, ADCY8, FOSL1, and S100A8 were found in all comparison groups of different ages, and multiple members of the HOX gene family including HOXC10, HOXA9, HOXA6, HOXB9, and HOXB6 were differentially expressed in comparison groups at different sites. Functional enrichment analysis showed that there were more obvious differences in immune function between different ages of skin and more obvious differences in endocrine function between different parts of skin. WGCNA revealed that genes related with immunity such as GLYATL2, ACSL5, and SPDEF were the core genes of the co-expression module associated with the scapula region, and multiple genes related to hair follicle development such as FOXN1, OVOL1, DLX3, HOXC13, and TCHH were found to be the hub genes of the co-expression module associated with the ventral region. Overall, our study provides valuable insights into the transcriptomic complexity of yak skin across different ages and body sites. The differential gene expression patterns and co-expression network modules identified in this study lay the foundation for further research on skin biology and adaptation mechanisms in yaks.
Collapse
Affiliation(s)
| | | | | | | | | | - Jiang Hu
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou 730070, China; (X.Z.); (B.S.); (Z.Z.); (Y.D.); (X.Z.)
| |
Collapse
|
9
|
Böhm M, Stegemann A, Paus R, Kleszczyński K, Maity P, Wlaschek M, Scharffetter-Kochanek K. Endocrine Controls of Skin Aging. Endocr Rev 2025; 46:349-375. [PMID: 39998423 DOI: 10.1210/endrev/bnae034] [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: 04/04/2024] [Indexed: 02/26/2025]
Abstract
Skin is the largest organ of the human body and undergoes both intrinsic (chronological) and extrinsic aging. While intrinsic skin aging is driven by genetic and epigenetic factors, extrinsic aging is mediated by external threats such as UV irradiation or fine particular matters, the sum of which is referred to as exposome. The clinical manifestations and biochemical changes are different between intrinsic and extrinsic skin aging, albeit overlapping features exist, eg, increased generation of reactive oxygen species, extracellular matrix degradation, telomere shortening, increased lipid peroxidation, or DNA damage. As skin is a prominent target for many hormones, the molecular and biochemical processes underlying intrinsic and extrinsic skin aging are under tight control of classical neuroendocrine axes. However, skin is also an endocrine organ itself, including the hair follicle, a fully functional neuroendocrine "miniorgan." Here we review pivotal hormones controlling human skin aging focusing on IGF-1, a key fibroblast-derived orchestrator of skin aging, of GH, estrogens, retinoids, and melatonin. The emerging roles of additional endocrine players, ie, α-melanocyte-stimulating hormone, a central player of the hypothalamic-pituitary-adrenal axis; members of the hypothalamic-pituitary-thyroid axis; oxytocin, endocannabinoids, and peroxisome proliferator-activated receptor modulators, are also reviewed. Until now, only a limited number of these hormones, mainly topical retinoids and estrogens, have found their way into clinical practice as anti-skin aging compounds. Further research into the biological properties of endocrine players or its derivatives may offer the development of novel senotherapeutics for the treatment and prevention of skin aging.
Collapse
Affiliation(s)
- Markus Böhm
- Department of Dermatology, University of Münster, Münster 48149, Germany
| | - Agatha Stegemann
- Department of Dermatology, University of Münster, Münster 48149, Germany
| | - Ralf Paus
- Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Musculoskeletal and Dermatological Sciences, The University of Manchester, Manchester M13 9PL, UK
- CUTANEON-Skin & Hair Innovations, 22335 Hamburgyi, Germany
- CUTANEON-Skin & Hair Innovations, 13125 Berlin, Germany
| | | | - Pallab Maity
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | - Meinhard Wlaschek
- Department of Dermatology and Allergic Diseases, Ulm University, 89081 Ulm, Germany
| | | |
Collapse
|
10
|
Li X, Wang Z, Li X, Fan X, Lu X, Li Y, Pan Y, Zhu Z, Zhu M, Li W, Chan L, Yu S, Pan Y, Wu Y. Transcriptional Study of Radiofrequency Device Using Experimental Mouse Model. Int J Mol Sci 2025; 26:4460. [PMID: 40362699 PMCID: PMC12072298 DOI: 10.3390/ijms26094460] [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: 03/26/2025] [Revised: 04/30/2025] [Accepted: 05/04/2025] [Indexed: 05/15/2025] Open
Abstract
Radiofrequencies have shown efficacy in addressing skin aging. Despite their effectiveness, few studies have explored how radiofrequencies affect the skin transcriptome. This study utilized mouse models divided into two age groups (four-month-old and one-year-old mice) to assess the impact of a radiofrequency device on skin collagen and elastin. A combination of histological analysis, Western blot analysis, real-time PCR and transcriptome sequencing was employed. Histological analysis revealed significant increases in dermis thickness and collagen fiber volume following radiofrequency treatment in both age groups. Quantitative PCR and Western blot analysis indicated that the levels of collagen-related genes and proteins were higher in the four-month-old group. Transcriptome sequencing identified 465 and 1867 differentially expressed genes (DEGs) in the skin of the 4-month-old mice and 1-year-old mice, respectively. GO and KEGG analyses elucidated the molecular mechanisms, revealing that the interleukin-17 and tumor necrosis factor signaling pathways may play crucial roles in collagen regeneration induced by radiofrequencies. Additionally, decreased expression of matrix metalloproteinase-9 and increased expression of the transcription factor Fos were identified as potential biomarkers of collagen regeneration. Immunofluorescence and immunohistochemistry staining demonstrated that radiofrequencies activate fibroblasts and inhibit macrophage alternative activation in the skin. This study identifies key genes and biological pathways involved in radiofrequency treatment and provides a foundation for a deeper understanding of the molecular mechanisms underlying collagen regeneration facilitated by radiofrequencies.
Collapse
Affiliation(s)
- Xiaofeng Li
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Zheng Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Xiaoman Li
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Xiaofeng Fan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Xinyu Lu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (S.Y.)
| | - Yanan Li
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Yehua Pan
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Ziyan Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Mingxi Zhu
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Wei Li
- School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China;
| | - Leo Chan
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| | - Suyun Yu
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (S.Y.)
| | - Yanhong Pan
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (S.Y.)
| | - Yuanyuan Wu
- Jiangsu Key Laboratory for Pharmacology and Safety Research of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, China; (X.L.); (Z.W.); (X.L.); (Y.L.); (Y.P.); (Z.Z.); (M.Z.); (L.C.)
| |
Collapse
|
11
|
Jia BB, Sun BK, Lee EY, Ren B. Emerging Techniques in Spatial Multiomics: Fundamental Principles and Applications to Dermatology. J Invest Dermatol 2025; 145:1017-1032. [PMID: 39503694 DOI: 10.1016/j.jid.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: 02/04/2024] [Revised: 09/09/2024] [Accepted: 09/09/2024] [Indexed: 04/25/2025]
Abstract
Molecular pathology, such as high-throughput genomic and proteomic profiling, identifies precise disease targets from biopsies but require tissue dissociation, losing valuable histologic and spatial context. Emerging spatial multi-omic technologies now enable multiplexed visualization of genomic, proteomic, and epigenomic targets within a single tissue slice, eliminating the need for labeling multiple adjacent slices. Although early work focused on RNA (spatial transcriptomics), spatial technologies can now concurrently capture DNA, genome accessibility, histone modifications, and proteins with spatially-resolved single-cell resolution. This review outlines the principles, advantages, limitations, and potential for spatial technologies to advance dermatologic research. By jointly profiling multiple molecular channels, spatial multiomics enables novel studies of copy number variations, clonal heterogeneity, and enhancer dysregulation, replete with spatial context, illuminating the skin's complex heterogeneity.
Collapse
Affiliation(s)
- Bojing B Jia
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, La Jolla, California, USA; Medical Scientist Training Program, University of California, San Diego, La Jolla, California, USA
| | - Bryan K Sun
- Department of Dermatology, University of California, Irvine, Irvine, California, USA
| | - Ernest Y Lee
- Department of Dermatology, University of California, San Francisco, San Francisco, California, USA
| | - Bing Ren
- Center for Epigenomics, Department of Cellular & Molecular Medicine, University of California, San Diego, La Jolla, California, USA; Institute of Genomic Medicine, Moores Cancer Center, School of Medicine, University of California, San Diego, La Jolla, California, USA.
| |
Collapse
|
12
|
Kou Y, Guo W, Wang Y, Kou C, Zhang B. Caviar extract inhibits skin photoaging by activating skin stem cells through NF-κB/MMPs/COL17A1 axis. Photochem Photobiol 2025; 101:683-696. [PMID: 39627162 DOI: 10.1111/php.14039] [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: 04/30/2024] [Revised: 09/29/2024] [Accepted: 09/29/2024] [Indexed: 05/17/2025]
Abstract
Ultraviolet radiations (UVR) produce harmful entities and reactive oxygen species (ROS) in skin cells, leading to skin photoaging. Caviar extract (CE) showed outstanding effects in delaying skin aging, but the underlying mechanism remains largely unknown. In this study, we prepared CE with acid protease and examined the anti-skin photoaging effects. The results showed that CE performed no cytotoxicity to HaCaT cells. For antioxidant properties, the EC50 values of DPPH and ABTS radical scavenging activity for CE were 1.27 and 5.20 mg/mL, respectively. It significantly reduced NF-κB, MMP-3 and MMP-9 protein expression levels, and increased IκB and TIMP-1 expression level in UVA-irradiated HaCaT cells. In the skin aging mice model, CE reduced the degree of UV-induced skin photoaging. Histological study confirmed that CE can ameliorate the adverse effects of UV exposure on the skin. Moreover, we found that CE could enhance the activities of Superoxide dismutase (SOD), and increased the contents of hydroxyproline (HYP) in photoaged mice skin. And CE elevated the protein expression level of COL17A1, KRT10, and KRT14 in mice skin. Taken together, our results bright systemic and new insights of CE into preventing UV-induced skin photoaging.
Collapse
Affiliation(s)
- Younan Kou
- College of Pharmacy, Nankai University, Tianjin, China
| | - Wuyan Guo
- Tianjin JieAngKang Bio&TCM-technology Development Co., Ltd., Tianjin, China
| | - Yun Wang
- Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou, Xuzhou, Jiangsu, China
| | - Changhua Kou
- Hepatobiliary Pancreatic Center, Xuzhou Central Hospital, Xuzhou, Xuzhou, Jiangsu, China
| | - Bo Zhang
- Institute for TCM-X, MOE Key Laboratory of Bioinformatics, Bioinformatics Division, BNRist, Department of Automation, Tsinghua University, Beijing, China
| |
Collapse
|
13
|
Fan Y, Zheng Y, Zhang Y, Xu G, Liu C, Hu J, Ji Q, Zhang S, Fang S, Lei J, Li LZ, Wang X, Xu X, Wang C, Wang S, Ma S, Song M, Jiang W, Zhu J, Feng Y, Wang J, Yang Y, Zhu G, Tian XL, Zhang H, Song W, Yang J, Yao Y, Liu GH, Qu J, Zhang W. ARID5A orchestrates cardiac aging and inflammation through MAVS mRNA stabilization. NATURE CARDIOVASCULAR RESEARCH 2025; 4:602-623. [PMID: 40301689 DOI: 10.1038/s44161-025-00635-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 03/10/2025] [Indexed: 05/01/2025]
Abstract
Elucidating the regulatory mechanisms of human cardiac aging remains a great challenge. Here, using human heart tissues from 74 individuals ranging from young (≤35 years) to old (≥65 years), we provide an overview of the histological, cellular and molecular alterations underpinning the aging of human hearts. We decoded aging-related gene expression changes at single-cell resolution and identified increased inflammation as the key event, driven by upregulation of ARID5A, an RNA-binding protein. ARID5A epi-transcriptionally regulated Mitochondrial Antiviral Signaling Protein (MAVS) mRNA stability, leading to NF-κB and TBK1 activation, amplifying aging and inflammation phenotypes. The application of gene therapy using lentiviral vectors encoding shRNA targeting ARID5A into the myocardium not only mitigated the inflammatory and aging phenotypes but also bolstered cardiac function in aged mice. Altogether, our study provides a valuable resource and advances our understanding of cardiac aging mechanisms by deciphering the ARID5A-MAVS axis in post-transcriptional regulation.
Collapse
Affiliation(s)
- Yanling Fan
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
| | - Yandong Zheng
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Yiyuan Zhang
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Gang Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Chun Liu
- Department of Physiology and Medicine, Cardiovascular Center, Cancer Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jianli Hu
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Qianzhao Ji
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuo Zhang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuaiqi Fang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jinghui Lei
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Lan-Zhu Li
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xing Wang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xi Xu
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Cui Wang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Si Wang
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Shuai Ma
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- Aging Biomarker Consortium, Beijing, China
| | - Moshi Song
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
| | - Wenjian Jiang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Junming Zhu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yijia Feng
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiangang Wang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Ying Yang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Guodong Zhu
- Institute of Gerontology, Guangzhou Geriatric Hospital, Guangzhou Medical University, Guangzhou, China
| | - Xiao-Li Tian
- Aging and Vascular Diseases, Human Aging Research Institute and School of Life Science, Nanchang University and Jiangxi Key Laboratory of Human Aging, Nanchang, China
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Weihong Song
- Oujiang Laboratory, Center for Geriatric Medicine and Institute of Aging, Key Laboratory of Alzheimer's Disease of Zhejiang Province, Zhejiang Provincial Clinical Research for Mental Disorders, The First-affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Jiayin Yang
- Liver Transplant Center, Organ Transplant Center, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Liver Transplantation, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital of Sichuan University, Chengdu, China
| | - Yan Yao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Guang-Hui Liu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| | - Jing Qu
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
- Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Weiqi Zhang
- China National Center for Bioinformation, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China.
- University of Chinese Academy of Sciences, Beijing, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
14
|
Arif M, Lehoczki A, Haskó G, Lohoff FW, Ungvari Z, Pacher P. Global and tissue-specific transcriptomic dysregulation in human aging: Pathways and predictive biomarkers. GeroScience 2025:10.1007/s11357-025-01672-z. [PMID: 40295347 DOI: 10.1007/s11357-025-01672-z] [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: 03/06/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Aging is a universal biological process that impacts all tissues, leading to functional decline and increased susceptibility to age-related diseases, particularly cardiometabolic disorders. While aging is characterized by hallmarks such as mitochondrial dysfunction, chronic inflammation, and dysregulated metabolism, the molecular mechanisms driving these processes remain incompletely understood, particularly in a tissue-specific context. To address this gap, we conducted a comprehensive transcriptomic analysis across 40 human tissues using data from the Genotype-Tissue Expression (GTEx) project, comparing individuals younger than 40 years with those older than 65 years. We identified over 17,000 differentially expressed genes (DEGs) across tissues, with distinct patterns of up- and down-regulation. Enrichment analyses revealed that up-regulated DEGs were associated with inflammation, immune responses, and apoptosis, while down-regulated DEGs were linked to mitochondrial function, oxidative phosphorylation, and metabolic processes. Using gene co-expression network (GCN) analyses, we identified 1,099 genes as dysregulated nodes (DNs) shared across tissues, reflecting global aging-associated transcriptional shifts. Integrating machine learning approaches, we pinpointed key aging biomarkers, including GDF15 and EDA2R, which demonstrated strong predictive power for aging and were particularly relevant in cardiometabolic tissues such as the heart, liver, skeletal muscle, and adipose tissue. These genes were also validated in plasma proteomics studies and exhibited significant correlations with clinical cardiometabolic health indicators. This study provides a multi-tissue, integrative perspective on aging, uncovering both systemic and tissue-specific molecular signatures. Our findings advance understanding of the molecular underpinnings of aging and identify novel biomarkers that may serve as therapeutic targets for promoting healthy aging and mitigating age-related diseases.
Collapse
Affiliation(s)
- Muhammad Arif
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
- Section On Fibrotic Disorders, National Institute and Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
- Department of Molecular and Clinical Medicine, SciLifeLab, Institute of Medicine, University of Gothenburg, Gothenburg, Sweden.
| | - Andrea Lehoczki
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - György Haskó
- Department of Anesthesiology, Columbia University, New York, NY, USA
| | - Falk W Lohoff
- Section On Clinical Genomics and Experimental Therapeutics, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Pal Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute On Alcohol Abuse and Alcoholism, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
15
|
Cheng D, Zhang Q, Shan W, Wang F. Facial Skin Aging: Effect of Aging in Different Layers of the Skin on Wrinkles and Sagging. JOURNAL OF BIOPHOTONICS 2025:e70028. [PMID: 40181530 DOI: 10.1002/jbio.70028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/26/2025] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
OBJECTIVE To investigate the impact of aging in different skin layers on the formation of wrinkles and sagging. METHODS In a single-center clinical study, the skin micromorphology, wrinkles, and sagging of 34 participants aged 42-60 years who use an antiaging product for 56 days are discussed retrospectively and observationally. The variation trend of each parameter and the correlation between micromorphology parameters and wrinkles and sagging are analyzed. RESULTS Parameters related to epidermal aging show a strong correlation with wrinkles and sagging, whereas parameters related to dermal aging show no significant correlation with wrinkles and sagging; in addition, parameters related to the dermal-epidermal junction (DEJ) show a moderate correlation with wrinkles and sagging. CONCLUSION For individuals in the rapid and stable aging phases, aging of the epidermis has a greater impact on wrinkles and sagging than aging of the dermis and DEJ.
Collapse
Affiliation(s)
- Dangdang Cheng
- Shanghai Jiyan Bio-Pharmaceutical Co. Ltd., Shanghai, China
- Yunnan Botanee Bio-Technology Group Co. Ltd., Yunnan, China
| | - Qin Zhang
- Shanghai Jiyan Bio-Pharmaceutical Co. Ltd., Shanghai, China
- Yunnan Botanee Bio-Technology Group Co. Ltd., Yunnan, China
| | - Wanwen Shan
- Shanghai Jiyan Bio-Pharmaceutical Co. Ltd., Shanghai, China
- Yunnan Botanee Bio-Technology Group Co. Ltd., Yunnan, China
| | - Feifei Wang
- Yunnan Botanee Bio-Technology Group Co. Ltd., Yunnan, China
- Yunnan Yunke Characteristic Plant Extraction Laboratory Co. Ltd., Yunnan, China
| |
Collapse
|
16
|
Cheung ST, Do Y, Kim E, Rella A, Goyarts E, Pernodet N, Wong YH. G Protein-Coupled Receptors in Skin Aging. J Invest Dermatol 2025; 145:749-765.e8. [PMID: 39186022 DOI: 10.1016/j.jid.2024.06.1288] [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: 06/17/2024] [Accepted: 06/24/2024] [Indexed: 08/27/2024]
Abstract
Skin aging is a complex biological process affected by a plethora of intrinsic and extrinsic factors that alter cutaneous functions through the modulations of signaling pathways and responses. Expressed in various cell types and skin tissue layers, G protein-coupled receptors (GPCRs) play a vital role in regulating skin aging. We have cataloged 156 GPCRs expressed in the skin and reviewed their roles in skin aging, such as pigmentation, loss of elasticity, wrinkles, rough texture, and aging-associated skin disorders. By exploring the GPCRs found in the skin, it may be possible to develop new treatment regimens for aging-associated skin conditions using GPCR ligands.
Collapse
Affiliation(s)
- Suet Ting Cheung
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Yelim Do
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Eunah Kim
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Antonella Rella
- Research and Development, The Estée Lauder Companies, New York, New York, USA
| | - Earl Goyarts
- Research and Development, The Estée Lauder Companies, New York, New York, USA
| | - Nadine Pernodet
- Research and Development, The Estée Lauder Companies, New York, New York, USA; Estée Lauder Research Laboratories, Melville, New York, USA
| | - Yung Hou Wong
- The Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China; The Biotechnology Research Institute, The Hong Kong University of Science and Technology, Hong Kong, China; Molecular Neuroscience Center, State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, China; Center for Aging Science, The Hong Kong University of Science and Technology, Hong Kong, China.
| |
Collapse
|
17
|
Zhao R, Zhang X, Geng Y, Lu D, Wang Y, Xie H, Zhang X, Xu S, Cao Y. SPRY1 regulates macrophage M1 polarization in skin aging and melanoma prognosis. Transl Oncol 2025; 54:102331. [PMID: 40023001 PMCID: PMC11915026 DOI: 10.1016/j.tranon.2025.102331] [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: 11/28/2024] [Revised: 01/28/2025] [Accepted: 02/10/2025] [Indexed: 03/04/2025] Open
Abstract
INTRODUCTION Skin aging is a complex, multifactorial process involving cellular damage, inflammation, and increased susceptibility to diseases. Despite its importance, the role of SPRY1 in skin aging remains poorly understood. This study aims to investigate the function of SPRY1 in skin aging, particularly its impact on macrophage M1 polarization, and explore its potential as a therapeutic target for mitigating skin aging and melanoma. METHODS Bioinformatics analyses were performed using datasets from the GTEx and GEO databases, alongside in vitro cellular experiments. These included Weighted Gene Co-expression Network Analysis (WGCNA), single-cell sequencing, and various cellular assays in RAW264.7 murine monocyte/macrophage leukemia cells and NIH/3T3 mouse skin fibroblasts. The assays comprised gene transfection, Cell Counting Kit-8 (CCK-8) assays, quantitative real-time PCR (qRT-PCR), and measurements of reactive oxygen species (ROS) and superoxide dismutase (SOD) activity. RESULTS SPRY1 was identified as a key gene within modules linked to skin aging. Single-cell sequencing revealed its enrichment in macrophages and keratinocytes. Knockdown of SPRY1 in RAW264.7 cells resulted in a shift from M1 to M2 macrophage polarization, reduced oxidative stress, and decreased expression of inflammatory markers. In NIH/3T3 cells, SPRY1 knockdown reduced cell viability and lowered the expression of inflammatory genes. Additionally, SPRY1 expression was downregulated in melanoma, and its reduced levels were associated with poorer survival outcomes. CONCLUSIONS SPRY1 accelerates skin aging by promoting macrophage M1 polarization and may serve as a promising therapeutic target. Future research should focus on in vivo validation and further exploration of its regulatory networks to develop novel treatments.
Collapse
Affiliation(s)
- Rongxin Zhao
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China
| | - Xun Zhang
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 XianXia Road, Shanghai 200336, China
| | - Yingnan Geng
- Department of Burns and Plastic Surgery, Second Affiliated Hospital of Naval Medical University, 415 Fengyang Road, Huangpu District, Shanghai 200003, China
| | - Dan Lu
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China
| | - Yuqing Wang
- Department of Dermatology, Xuzhou Huamei Cosmetology Hospital, Jiangsu, West Huaihai Road, Quanshan District, Xuzhou, Jiangsu, China
| | - Han Xie
- The Fifth People's Hospital of Shanghai, Fudan University, No. 128, Ruili Road, Minhang District, Shanghai, China
| | - Xiaofei Zhang
- Shanghai Xinmei Medical Beauty Outpatient Department, 202A, No.285, Jianguo West Road, Xuhui District, Shanghai, China.
| | - Shunming Xu
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China.
| | - Yanyun Cao
- Department of Dermatology, Pudong New Area People's Hospital, 490 Chuanhuang South Road, Pudong New Area, Shanghai, China.
| |
Collapse
|
18
|
Li Z, Xiao Y, Kang L, Li Y, Wang HC, Li Z, Yang Y, Huang J, Yu N, Long X. Comparative Study of Adipose-Derived Stem Cells from Localized Scleroderma Patients and Healthy Donors in Treating Skin Fibrosis. Plast Reconstr Surg 2025; 155:716e-726e. [PMID: 39348305 DOI: 10.1097/prs.0000000000011779] [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] [Indexed: 10/02/2024]
Abstract
BACKGROUND Localized scleroderma (LoS) is an autoimmune disease characterized by fibrosis of the skin and atrophy of the subcutaneous fat tissue. Use of adipose-derived mesenchymal stem cells (ASCs) is a promising treatment approach for LoS. However, ASCs from scleroderma patients (LoS ASCs) have been shown to exhibit altered characteristics compared with ASCs from healthy donors (healthy ASCs). This study aimed to compare the abilities of LoS ASCs and healthy ASCs in treating skin fibrosis. METHODS The paracrine ability of ASCs was tested with cytokine array. Bleomycin-challenged mouse models received subcutaneous injection of LoS ASCs and healthy ASCs. Pathologic staining and Western blotting of collagenase type I and α-smooth muscle actin was performed. Fibroblasts derived from LoS lesions (LoS FB) were co-cultured with ASCs, and subjected to RNA sequencing to further explore the similarities and differences in the treatment mechanism. RESULTS In vivo comparison revealed that healthy ASCs had a stronger proliferation ability and secreted higher levels of growth factors and cytokines, including vascular endothelial growth factor A, platelet-derived growth factor fibroblasts, and interleukin-10. Pathologic staining of the skin in mouse models treated with ASCs demonstrated that healthy ASCs were more effective in reducing dermal thickness and collagen deposition, and increasing microvessel density and the proportion of M2 macrophages. Co-culture with both healthy ASCs and LoS ASCs reduced the proliferation and migration abilities of LoS FB, and the protein expression of α-smooth muscle actin and collagenase type I. RNA sequencing and validation revealed potential difference in the canonical Wnt pathway. CONCLUSION Healthy ASCs exhibited stronger proliferation, paracrine, antifibrosis, proangiogenesis, and immunomodulation abilities in treating skin fibrosis in scleroderma mouse models. CLINICAL RELEVANCE STATEMENT Allogenic ASCs obtained from healthy donors are more efficient in treating skin fibrosis, and could serve as a potential alternative for patients who are not suitable candidates for liposuction surgery in the future.
Collapse
Affiliation(s)
- Zhujun Li
- From the Department of Plastic Surgery
| | | | - Lin Kang
- Medical Research Center, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College
| | - Yunzhu Li
- From the Department of Plastic Surgery
| | - Hayson Chenyu Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine
| | - Ziming Li
- From the Department of Plastic Surgery
| | | | | | - Nanze Yu
- From the Department of Plastic Surgery
| | - Xiao Long
- From the Department of Plastic Surgery
| |
Collapse
|
19
|
Wang Y, Yang J, Luo Y, Zhao Z, Yuan Y, Li J, Liu Y, Yi Y, Xu X, Lan Y, Zou J, Li Q, Wang L, Pan Y, Yang Y, Xiong M, Wu M, Li J, Li W, Zhang Y, Cao Y, Zhu Y, Xiao ZXJ. Targeting IGF1-Induced Cellular Senescence to Rejuvenate Hair Follicle Aging. Aging Cell 2025:e70053. [PMID: 40159808 DOI: 10.1111/acel.70053] [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: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/13/2025] [Indexed: 04/02/2025] Open
Abstract
The insulin-like growth factor-1 (IGF-1) signaling pathway is known as a potent aging modifier, disruption of which consistently associates with lifespan extension across diverse species. Despite this established association, the mechanisms by which IGF-1 signaling modulates organ aging remain poorly understood. In this study, we assessed age-related changes in IGF-1 expression across multiple organs in mice and identified a more prominent increase in skin IGF-1 levels with aging-a phenomenon also observed in human skin. To explore the consequences of elevated IGF-1, we developed transgenic mice ectopically expressing human IGF-1 in the epidermis, driven by the bovine keratin 5 promoter (IGF-1 Tg). These mice exhibited premature aging of hair follicles, as evidenced by accelerated hair graying and loss. Single-cell RNA sequencing analyses of dorsal skin highlighted an upsurge in cellular senescence markers and the senescence-associated secretory phenotype (SASP) in hair follicle stem cells (HFSCs), alongside a decline in hair growth and HFSC exhaustion. Our findings indicate that excessive IGF-1 triggers HFSC senescence, thereby disrupting hair follicle homeostasis. Remarkably, interventions in IGF-1 signaling via downstream mechanisms-specifically blocking Ac-p53 activation via SIRT1 overexpression or senolytic treatment for senescent cell clearance, or reducing IGF-1 through dietary restriction-significantly reduced senescence markers, mitigated premature hair follicle aging phenotypes, and restored the stem cell pool. Our findings provide fundamental insights into the biological processes of hair aging and highlight the therapeutic promise of targeted interventions to rejuvenate aged HFSCs and promote hair follicle health.
Collapse
Affiliation(s)
- Yang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jian Yang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yue Luo
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Zhiqiang Zhao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yawen Yuan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Juan Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Liu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yong Yi
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Xiaoke Xu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuankunyu Lan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Juan Zou
- Department of Pathology, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, China
| | - Qintong Li
- Departments of Obstetrics & Gynecology and Pediatrics, West China Second University Hospital, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Sichuan University, Chengdu, Sichuan, China
| | - Liang Wang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Pan
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yuanhan Yang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Muzhao Xiong
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Min Wu
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Jinsong Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Weiyuxin Li
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yujun Zhang
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yang Cao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Yi Zhu
- Department of Physiology and Biomedical Engineering, Robert & Kogod Center on Aging, Mayo Clinic, Rochester, Minnesota, USA
| | - Zhi-Xiong Jim Xiao
- Center of Growth, Metabolism and Aging, Key Laboratory of Bio-Resource and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
20
|
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
|
21
|
Myeong J, Lee M, Lee B, Kim JH, Nam Y, Choi Y, Kim J, Jeon SY, Shim H, Jung DR, Shin Y, Jeong M, Oh B, Jung J, Kim CS, Han HS, Shin JH, Lee YH, Park NJY, Chong GO, Jeong Y. Microbial metabolites control self-renewal and precancerous progression of human cervical stem cells. Nat Commun 2025; 16:2327. [PMID: 40057497 PMCID: PMC11890575 DOI: 10.1038/s41467-025-57323-6] [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: 09/30/2024] [Accepted: 02/18/2025] [Indexed: 05/13/2025] Open
Abstract
Cervical cancer is the fourth most common female cancer, with the uterine ectocervix being the most commonly affected site. However, cervical stem cells, their differentiation, and their regulation remain poorly understood. Here, we report the isolation of a population enriched for human cervical stem cells and their regulatory mechanisms. Using single-cell RNA sequencing, we characterize the cellular heterogeneity of the human ectocervix and identify cluster-specific cell surface markers. By establishing normal and precancerous cervical organoids and an intralingual transplantation system, we show that ITGB4 and CD24 enable enrichment of human and murine ectocervical stem cells. We discover that Lactobacilli-derived lactic acid regulates cervical stem cells' self-renewal and early tumorigenesis through the PI3K-AKT pathway and YAP1. Finally, we show that D-lactic acid suppresses growth of normal and precancerous organoids, while L-lactic acid does not. Our findings reveal roles of human cervical stem cells and microbial metabolites in cervical health and diseases.
Collapse
Affiliation(s)
| | - Minho Lee
- Department of Life Science, Dongguk University, Gyeonggi-do, Korea
| | - Bawool Lee
- Department of New Biology, DGIST, Daegu, Korea
- New Biology Research Center, DGIST, Daegu, Korea
| | - Joon Hyung Kim
- Department of Life Science, Dongguk University, Gyeonggi-do, Korea
| | - Yeji Nam
- School of Undergraduate Studies, DGIST, Daegu, Korea
| | - Yeseul Choi
- Graduate Program, Department of Biomedical Science, Kyungpook National University School of Medicine, Daegu, Korea
- BK21 Four Program, Kyungpook National University School of Medicine, Daegu, Korea
| | | | - Se Young Jeon
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu, Korea
| | - Haewon Shim
- Department of Life Science, Dongguk University, Gyeonggi-do, Korea
| | - Da-Ryung Jung
- Department of Applied Biosciences, Kyungpook National University, Daegu, Korea
| | - Youngjin Shin
- Department of Life Science, Dongguk University, Gyeonggi-do, Korea
| | - Minsoo Jeong
- Department of Applied Biosciences, Kyungpook National University, Daegu, Korea
| | - Byungmoo Oh
- Department of New Biology, DGIST, Daegu, Korea
- New Biology Research Center, DGIST, Daegu, Korea
| | - Jaehun Jung
- Department of Life Science, Dongguk University, Gyeonggi-do, Korea
| | - Christine S Kim
- Department of New Biology, DGIST, Daegu, Korea
- New Biology Research Center, DGIST, Daegu, Korea
| | - Hyung Soo Han
- Graduate Program, Department of Biomedical Science, Kyungpook National University School of Medicine, Daegu, Korea
- BK21 Four Program, Kyungpook National University School of Medicine, Daegu, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu, Korea
- Department of Physiology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Jae-Ho Shin
- Department of Applied Biosciences, Kyungpook National University, Daegu, Korea
| | - Yoon Hee Lee
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu, Korea
- Clinical Omics Institute, Kyungpook National University, Daegu, Korea
- Department of Obstetrics and Gynecology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Nora Jee-Young Park
- Clinical Omics Institute, Kyungpook National University, Daegu, Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, Daegu, Korea
- Department of Pathology, Kyungpook National University School of Medicine, Daegu, Korea
| | - Gun Oh Chong
- Department of Obstetrics and Gynecology, Kyungpook National University Chilgok Hospital, Daegu, Korea.
- Clinical Omics Institute, Kyungpook National University, Daegu, Korea.
- Department of Obstetrics and Gynecology, Kyungpook National University School of Medicine, Daegu, Korea.
| | - Youngtae Jeong
- Department of New Biology, DGIST, Daegu, Korea.
- New Biology Research Center, DGIST, Daegu, Korea.
| |
Collapse
|
22
|
Li B, Ming H, Qin S, Nice EC, Dong J, Du Z, Huang C. Redox regulation: mechanisms, biology and therapeutic targets in diseases. Signal Transduct Target Ther 2025; 10:72. [PMID: 40050273 PMCID: PMC11885647 DOI: 10.1038/s41392-024-02095-6] [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: 06/20/2024] [Revised: 10/09/2024] [Accepted: 11/21/2024] [Indexed: 03/09/2025] Open
Abstract
Redox signaling acts as a critical mediator in the dynamic interactions between organisms and their external environment, profoundly influencing both the onset and progression of various diseases. Under physiological conditions, oxidative free radicals generated by the mitochondrial oxidative respiratory chain, endoplasmic reticulum, and NADPH oxidases can be effectively neutralized by NRF2-mediated antioxidant responses. These responses elevate the synthesis of superoxide dismutase (SOD), catalase, as well as key molecules like nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), thereby maintaining cellular redox homeostasis. Disruption of this finely tuned equilibrium is closely linked to the pathogenesis of a wide range of diseases. Recent advances have broadened our understanding of the molecular mechanisms underpinning this dysregulation, highlighting the pivotal roles of genomic instability, epigenetic modifications, protein degradation, and metabolic reprogramming. These findings provide a foundation for exploring redox regulation as a mechanistic basis for improving therapeutic strategies. While antioxidant-based therapies have shown early promise in conditions where oxidative stress plays a primary pathological role, their efficacy in diseases characterized by complex, multifactorial etiologies remains controversial. A deeper, context-specific understanding of redox signaling, particularly the roles of redox-sensitive proteins, is critical for designing targeted therapies aimed at re-establishing redox balance. Emerging small molecule inhibitors that target specific cysteine residues in redox-sensitive proteins have demonstrated promising preclinical outcomes, setting the stage for forthcoming clinical trials. In this review, we summarize our current understanding of the intricate relationship between oxidative stress and disease pathogenesis and also discuss how these insights can be leveraged to optimize therapeutic strategies in clinical practice.
Collapse
Affiliation(s)
- Bowen Li
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Hui Ming
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
| | - Siyuan Qin
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China
| | - Edouard C Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Jingsi Dong
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Zhongyan Du
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
- Key Laboratory of Blood-stasis-toxin Syndrome of Zhejiang Province, Hangzhou, China.
| | - Canhua Huang
- Department of Biotherapy, Institute of Oxidative Stress Medicine, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital and West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, PR China.
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, PR China.
| |
Collapse
|
23
|
Liu Y, Wang P, Li J, Chen L, Shu B, Wang H, Liu H, Zhao S, Zhou J, Chen X, Xie J. Single-cell RNA sequencing reveals the impaired epidermal differentiation and pathological microenvironment in diabetic foot ulcer. BURNS & TRAUMA 2025; 13:tkae065. [PMID: 40040959 PMCID: PMC11879498 DOI: 10.1093/burnst/tkae065] [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: 04/10/2024] [Revised: 09/21/2024] [Accepted: 10/17/2024] [Indexed: 03/06/2025]
Abstract
Background Diabetic foot ulcer (DFU) is one of the most common and complex complications of diabetes, but the underlying pathophysiology remains unclear. Single-cell RNA sequencing (scRNA-seq) has been conducted to explore novel cell types or molecular profiles of DFU from various perspectives. This study aimed to comprehensively analyze the potential mechanisms underlying impaired re-epithelization of DFU in a single-cell perspective. Methods We conducted scRNA-seq on tissues from human normal skin, acute wound, and DFU to investigate the potential mechanisms underlying impaired epidermal differentiation and the pathological microenvironment. Pseudo-time and lineage inference analyses revealed the distinct states and transition trajectories of epidermal cells under different conditions. Transcription factor analysis revealed the potential regulatory mechanism of key subtypes of keratinocytes. Cell-cell interaction analysis revealed the regulatory network between the proinflammatory microenvironment and epidermal cells. Laser-capture microscopy coupled with RNA sequencing (LCM-seq) and multiplex immunohistochemistry were used to validate the expression and location of key subtypes of keratinocytes. Results Our research provided a comprehensive map of the phenotypic and dynamic changes that occur during epidermal differentiation, alongside the corresponding regulatory networks in DFU. Importantly, we identified two subtypes of keratinocytes: basal cells (BC-2) and diabetes-associated keratinocytes (DAK) that might play crucial roles in the impairment of epidermal homeostasis. BC-2 and DAK showed a marked increase in DFU, with an inactive state and insufficient motivation for epidermal differentiation. BC-2 was involved in the cellular response and apoptosis processes, with high expression of TXNIP, IFITM1, and IL1R2. Additionally, the pro-differentiation transcription factors were downregulated in BC-2 in DFU, indicating that the differentiation process might be inhibited in BC-2 in DFU. DAK was associated with cellular glucose homeostasis. Furthermore, increased CCL2 + CXCL2+ fibroblasts, VWA1+ vascular endothelial cells, and GZMA+CD8+ T cells were detected in DFU. These changes in the wound microenvironment could regulate the fate of epidermal cells through the TNFSF12-TNFRSF12A, IFNG-IFNGR1/2, and IL-1B-IL1R2 pathways, which might result in persistent inflammation and impaired epidermal differentiation in DFU. Conclusions Our findings offer novel insights into the pathophysiology of DFU and present potential therapeutic targets that could improve wound care and treatment outcomes for DFU patients.
Collapse
Affiliation(s)
- Yiling Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Peng Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Jingting Li
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Lei Chen
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Bin Shu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Hanwen Wang
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Hengdeng Liu
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Shixin Zhao
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| | - Junli Zhou
- Department of Burn and Plastic Surgery, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), No. 3 Wandao Road, Dongguan 523000, China
| | - Xiaodong Chen
- Department of Burn Surgery, The First People’s Hospital of Foshan, No. 3 Lingnan Road, Foshan 528000, China
| | - Julin Xie
- Department of Burn and Wound Repair Surgery, The First Affiliated Hospital of Sun Yat-sen University, No. 58 Zhongshan 2 Road, Guangzhou 510080, China
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, No. 58, Zhongshan 2 Road, Guangzhou 510080, China
| |
Collapse
|
24
|
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
|
25
|
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
|
26
|
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
|
27
|
Tong H, Guo X, Chen L, Wang H, Hu X, He A, Li C, Zhang T, Kang J, Fu Y. Quercetin prevents the loss of chondrogenic capacity in expansion cultured human auricular chondrocytes by alleviating mitochondrial dysfunction. Regen Ther 2025; 28:358-370. [PMID: 39896443 PMCID: PMC11783217 DOI: 10.1016/j.reth.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 12/25/2024] [Accepted: 01/04/2025] [Indexed: 02/04/2025] Open
Abstract
Objective To explore the characteristics of cellular senescence in human auricular chondrocytes during long-term in vitro culture and to evaluate the effects of anti-senescence treatments on enhancing their chondrogenic function. Methods Auricular chondrocytes exhibited senescence-related characteristics after prolonged expansion in culture. To identify senescence inducers, transcriptome sequencing was performed, with findings corroborated by transmission electron microscopy analyses. Quercetin was employed as an intervention to mitigate cellular senescence progression. The alterations in cellular senescence and mitochondrial function were evaluated. Regenerative cartilage tissue was developed through in vitro chondrogenic induction and in vivo implantation with GelMA hydrogel-loaded cells in nude mice. The impact of quercetin was substantiated through histological examinations. Results Mitochondrial dysfunction was a key characteristic of auricular chondrocytes after long-term expansion culture. Chondrocytes cultured with quercetin showed a lower proportion of senescent cells and reduced mitochondrial dysfunction. The chondrocytes cultured with continuous application of quercetin formed higher quality regenerative cartilage both in vitro and in vivo compared to the control group. Conclusion The results reveal that quercetin attenuates chondrocyte senescence by alleviating mitochondrial dysfunction, thereby preventing the loss of chondrogenic function in chondrocytes subjected to long-term expansion culture.
Collapse
Affiliation(s)
- Hua Tong
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Xudong Guo
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Lili Chen
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Honglei Wang
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Xuerui Hu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Aijuan He
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Chenlong Li
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| | - Tianyu Zhang
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- NHC Key Laboratory of Hearing Medicine, Fudan University, Shanghai 200031, China
| | - Jiuhong Kang
- School of Life Sciences and Technology, Tongji University, Shanghai 200092, China
| | - Yaoyao Fu
- Department of Facial Plastic and Reconstructive Surgery, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
- ENT Institute, Eye & ENT Hospital, Fudan University, Shanghai 200031, China
| |
Collapse
|
28
|
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
|
29
|
Sanborn MA, Wang X, Gao S, Dai Y, Rehman J. Unveiling the cell-type-specific landscape of cellular senescence through single-cell transcriptomics using SenePy. Nat Commun 2025; 16:1884. [PMID: 39987255 PMCID: PMC11846890 DOI: 10.1038/s41467-025-57047-7] [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: 04/29/2024] [Accepted: 02/06/2025] [Indexed: 02/24/2025] Open
Abstract
Senescent cells accumulate in most tissues with organismal aging, exposure to stressors, or disease progression. It is challenging to identify senescent cells because cellular senescence signatures and phenotypes vary widely across distinct cell types and tissues. Here we developed an analytical algorithm that defines cell-type-specific and universal signatures of cellular senescence across a wide range of cell types and tissues. We utilize 72 mouse and 64 human weighted single-cell transcriptomic signatures of cellular senescence to create the SenePy scoring platform. SenePy signatures better recapitulate in vivo cellular senescence than signatures derived from in vitro senescence studies. We use SenePy to map the kinetics of senescent cell accumulation in healthy aging as well as multiple disease contexts, including tumorigenesis, inflammation, and myocardial infarction. SenePy characterizes cell-type-specific in vivo cellular senescence and could lead to the identification of genes that serve as mediators of cellular senescence and disease progression.
Collapse
Grants
- R01-AG091545 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01HL160469 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01-HL152515 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL152515 NHLBI NIH HHS
- R01-HL163978 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- P01 HL160469 NHLBI NIH HHS
- F31-AG090005 U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
- T32- HL139439 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F31 AG090005 NIA NIH HHS
- R01 HL163978 NHLBI NIH HHS
- T32 HL139439 NHLBI NIH HHS
- R01 AG091545 NIA NIH HHS
- U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- U.S. Department of Health & Human Services | NIH | National Institute on Aging (U.S. National Institute on Aging)
Collapse
Affiliation(s)
- Mark A Sanborn
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA.
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA.
| | - Xinge Wang
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA
| | - Shang Gao
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA
| | - Yang Dai
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA
| | - Jalees Rehman
- Department of Biochemistry and Molecular Genetics, University of Illinois, College of Medicine, Chicago, Illinois, USA.
- Center for Bioinformatics and Quantitative Biology, University of Illinois Chicago, Chicago, Illinois, USA.
- Department of Biomedical Engineering, University of Illinois Chicago, College of Engineering and College of Medicine, Chicago, Illinois, USA.
- University of Illinois Cancer Center, Chicago, Illinois, USA.
| |
Collapse
|
30
|
Arakelyan NA, Kupriyanova DA, Vasilevska J, Rogaev EI. Sexual dimorphism in immunity and longevity among the oldest old. Front Immunol 2025; 16:1525948. [PMID: 40034689 PMCID: PMC11872714 DOI: 10.3389/fimmu.2025.1525948] [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: 11/10/2024] [Accepted: 01/28/2025] [Indexed: 03/05/2025] Open
Abstract
Human longevity is a sex-biased process in which sex chromosomes and sex-specific immunity may play a crucial role in the health and lifespan disparities between men and women. Generally, women have a higher life expectancy than men, exhibiting lower infection rates for a broad range of pathogens, which results in a higher prevalence of female centenarians compared to males. Investigation of the immunological changes that occur during the process of healthy aging, while taking into account the differences between sexes, can significantly enhance our understanding of the mechanisms that underlie longevity. In this review, we aim to summarize the current knowledge on sexual dimorphism in the human immune system and gut microbiome during aging, with a particular focus on centenarians, based exclusively on human data.
Collapse
Affiliation(s)
- Nelli A. Arakelyan
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Daria A. Kupriyanova
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Jelena Vasilevska
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
| | - Evgeny I. Rogaev
- Center for Genetics and Life Science, Sirius University of Science and Technology, Sochi, Russia
- Department of Psychiatry, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
31
|
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
|
32
|
Wu J, Shao T, Tang Z, Liu G, Li Z, Shi Y, Kang Y, Zuo J, Zhao B, Hu G, Liu J, Ji W, Zhang L, Niu Y. Highly efficient construction of monkey blastoid capsules from aged somatic cells. Nat Commun 2025; 16:1130. [PMID: 39875393 PMCID: PMC11775175 DOI: 10.1038/s41467-025-56447-z] [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: 06/01/2024] [Accepted: 01/16/2025] [Indexed: 01/30/2025] Open
Abstract
Blastoids-blastocyst-like structures created in vitro-emerge as a valuable model for early embryonic development research. Non-human primates stem cell-derived blastoids are an ethically viable alternative to human counterparts, yet the low formation efficiency of monkey blastoid cavities, typically below 30%, has limited their utility. Prior research has predominantly utilized embryonic stem cells. In this work, we demonstrate the efficient generation of blastoids from induced pluripotent stem cells and somatic cell nuclear transfer embryonic stem cells derived from aged monkeys, achieving an 80% formation efficiency. We also introduce a hydrogel-based microfluidics platform for the scalable and reproducible production of size-adjustable, biodegradable blastoid capsules, providing a stable 3D structure and mechanical protection. This advancement in the high-efficiency, scalable production of monkey blastoid capsules from reprogrammed aged somatic cells significantly enhances the study of embryonic development and holds promise for regenerative medicine.
Collapse
Affiliation(s)
- Junmo Wu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Tianao Shao
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Zengli Tang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Southwest United Graduate School, Kunming, Yunnan, China
| | - Gaojing Liu
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zhuoyao Li
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yuxi Shi
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Yu Kang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jiawei Zuo
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
- Southwest United Graduate School, Kunming, Yunnan, China
| | - Bo Zhao
- Key Laboratory of Genetic Evolution & Animal Models, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan, China
| | - Guangyu Hu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jiaqi Liu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Weizhi Ji
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
| | - Lei Zhang
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
| | - Yuyu Niu
- State Key Laboratory of Primate Biomedical Research; Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China.
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China.
- Southwest United Graduate School, Kunming, Yunnan, China.
| |
Collapse
|
33
|
Shi S, Ou X, Long J, Lu X, Xu S, Li G. The role of multiomics in revealing the mechanism of skin repair and regeneration. Front Pharmacol 2025; 16:1497988. [PMID: 39896077 PMCID: PMC11782119 DOI: 10.3389/fphar.2025.1497988] [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: 11/04/2024] [Accepted: 01/02/2025] [Indexed: 02/04/2025] Open
Abstract
Skin repair and regeneration are crucial processes in restoring the integrity of the skin after injury, with significant implications for medical treatments and plastic surgery. Multiomics, an integrated approach combining genomics, transcriptomics, proteomics, and metabolomics, offers unprecedented insights into the complex molecular and cellular mechanisms involved in skin healing. This review explores the transformative role of multiomics in elucidating the mechanisms of skin repair and regeneration. While genomic studies identify the genetic basis of wound healing, transcriptomics and proteomics uncover the dynamic changes in gene and protein expression, and metabolomics provides a snapshot of metabolic alterations associated with wound healing. Integrative multiomics studies can also identify novel biomarkers and therapeutic targets for skin regeneration. Despite the technical and biological challenges, the future of multiomics in skin research holds great promise for advancing personalized medicine and improving wound healing strategies. Through interdisciplinary collaboration, multiomics has the potential to revolutionize our understanding of skin repair, paving the way for innovative treatments in plastic surgery and beyond.
Collapse
Affiliation(s)
| | | | | | | | | | - Gang Li
- Department of Hand Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi an, China
| |
Collapse
|
34
|
Yu GT, Ganier C, Allison DB, Tchkonia T, Khosla S, Kirkland JL, Lynch MD, Wyles SP. Mapping epidermal and dermal cellular senescence in human skin aging. Aging Cell 2025; 24:e14358. [PMID: 39370688 PMCID: PMC11709101 DOI: 10.1111/acel.14358] [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: 05/12/2024] [Revised: 08/27/2024] [Accepted: 09/13/2024] [Indexed: 10/08/2024] Open
Abstract
Single-cell RNA sequencing and spatial transcriptomics enable unprecedented insight into cellular and molecular pathways implicated in human skin aging and regeneration. Senescent cells are individual cells that are irreversibly cell cycle arrested and can accumulate across the human lifespan due to cell-intrinsic and -extrinsic stressors. With an atlas of single-cell RNA-sequencing and spatial transcriptomics, epidermal and dermal senescence and its effects were investigated, with a focus on melanocytes and fibroblasts. Photoaging due to ultraviolet light exposure was associated with higher burdens of senescent cells, a sign of biological aging, compared to chronological aging. A skin-specific cellular senescence gene set, termed SenSkin™, was curated and confirmed to be elevated in the context of photoaging, chronological aging, and non-replicating CDKN1A+ (p21) cells. In the epidermis, senescent melanocytes were associated with elevated melanin synthesis, suggesting haphazard pigmentation, while in the dermis, senescent reticular dermal fibroblasts were associated with decreased collagen and elastic fiber synthesis. Spatial analysis revealed the tendency for senescent cells to cluster, particularly in photoaged skin. This work proposes a strategy for characterizing age-related skin dysfunction through the lens of cellular senescence and suggests a role for senescent epidermal cells (i.e., melanocytes) and senescent dermal cells (i.e., reticular dermal fibroblasts) in age-related skin sequelae.
Collapse
Affiliation(s)
- Grace T. Yu
- Mayo Clinic Medical Scientist Training ProgramMayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic Alix School of MedicineRochesterMinnesotaUSA
| | - Clarisse Ganier
- Centre for Gene Therapy and Regenerative MedicineKing's College London, Guy's HospitalLondonUK
| | | | - Tamara Tchkonia
- Division of Endocrinology and Metabolism, Department of MedicineCenter for Gerotherapeutics, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Sundeep Khosla
- Division of Endocrinology, Department of MedicineMayo ClinicRochesterMinnesotaUSA
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
| | - James L. Kirkland
- Division of Endocrinology and Metabolism, Department of MedicineCenter for Gerotherapeutics, Cedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Magnus D. Lynch
- Centre for Gene Therapy and Regenerative MedicineKing's College London, Guy's HospitalLondonUK
- St. John's Institute of DermatologyKing's College London, Guy's HospitalLondonUK
| | - Saranya P. Wyles
- Robert and Arlene Kogod Center on AgingMayo ClinicRochesterMinnesotaUSA
- Department of DermatologyMayo ClinicRochesterMinnesotaUSA
| |
Collapse
|
35
|
Mitra M, Batista SL, Coller HA. Transcription factor networks in cellular quiescence. Nat Cell Biol 2025; 27:14-27. [PMID: 39789221 DOI: 10.1038/s41556-024-01582-w] [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: 08/13/2024] [Accepted: 11/25/2024] [Indexed: 01/12/2025]
Abstract
Many of the cells in mammalian tissues are in a reversible quiescent state; they are not dividing, but retain the ability to proliferate in response to extracellular signals. Quiescence relies on the activities of transcription factors (TFs) that orchestrate the repression of genes that promote proliferation and establish a quiescence-specific gene expression program. Here we discuss how the coordinated activities of TFs in different quiescent stem cells and differentiated cells maintain reversible cell cycle arrest and establish cell-protective signalling pathways. We further cover the emerging mechanisms governing the dysregulation of quiescence TF networks with age. We explore how recent developments in single-cell technologies have enhanced our understanding of quiescence heterogeneity and gene regulatory networks. We further discuss how TFs and their activities are themselves regulated at the RNA, protein and chromatin levels. Finally, we summarize the challenges associated with defining TF networks in quiescent cells.
Collapse
Affiliation(s)
- Mithun Mitra
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Sandra L Batista
- Department of Computer Science, University of California, Los Angeles, Los Angeles, CA, USA
| | - Hilary A Coller
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA.
- Department of Biological Chemistry, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
36
|
Hu XM, Zheng S, Zhang Q, Wan X, Li J, Mao R, Yang R, Xiong K. PANoptosis signaling enables broad immune response in psoriasis: From pathogenesis to new therapeutic strategies. Comput Struct Biotechnol J 2024; 23:64-76. [PMID: 38125299 PMCID: PMC10730955 DOI: 10.1016/j.csbj.2023.11.049] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/24/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023] Open
Abstract
BACKGROUND Accumulating evidence suggests that regulated cell death, such as pyroptosis, apoptosis, and necroptosis, is deeply involved in the pathogenesis of psoriasis. As a newly recognized form of systematic cell death, PANoptosis is involved in a variety of inflammatory disorders through amplifying inflammatory and immune cascades, but its role in psoriasis remains elusive. OBJECTIVES To reveal the role of PANoptosis in psoriasis for a potential therapeutic strategy. METHODS Multitranscriptomic analysis and experimental validation were used to identify PANoptosis signaling in psoriasis. RNA-seq and scRNA-seq analyses were performed to establish a PANoptosis-mediated immune response in psoriasis, which revealed hub genes through WGCNA and predicted disulfiram as a potential drug. The effect and mechanism of disulfiram were verified in imiquimod (IMQ)-induced psoriasis. RESULTS Here, we found a highlighted PANoptosis signature in psoriasis patients through multitranscriptomic analysis and experimental validation. Based on this, two distinct PANoptosis patterns (non/high) were identified, which were the options for clinical classification. The high-PANoptosis-related group had a higher response rate to immune cell infiltration (such as M1 macrophages and keratinocytes). Subsequently, WGCNA showed the hub genes (e.g., S100A12, CYCS, NOD2, STAT1, HSPA4, AIM2, MAPK7), which were significantly associated with clinical phenotype, PANoptosis signature, and identified immune response in psoriasis. Finally, we explored disulfiram (DSF) as a candidate drug for psoriasis through network pharmacology, which ameliorated IMQ-mediated psoriatic symptoms through antipyroptosis-mediated inflammation and enhanced apoptotic progression. By analyzing the specific ligand-receptor interaction pairs within and between cell lineages, we speculated that DSF might exert its effects by targeting keratinocytes directly or targeting M1 macrophages to downregulate the proliferation of keratinocytes. CONCLUSIONS PANoptosis with its mediated immune cell infiltration provides a roadmap for research on the pathogenesis and therapeutic strategies of psoriasis.
Collapse
Affiliation(s)
- Xi-min Hu
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Shengyuan Zheng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qi Zhang
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
| | - Xinxing Wan
- Department of Endocrinology, Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Rui Mao
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou 510000, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, China
- Hunan Key Laboratory of Ophthalmology, Xiangya Hospital, Central South University, Changsha 410008, China
- Key Laboratory of Emergency and Trauma, Ministry of Education, College of Emergency and Trauma, Hainan Medical University, Haikou 571199, China
| |
Collapse
|
37
|
Xiao Y, Chen X, Chen Z, Dai W, Hu X, Zhang S, Zhong J, Chen J, Liu X, Liang L, Hu Y. Comparative single-cell transcriptomic analysis across tissues of aging primates reveals specific autologous activation of ZNF281 to mitigate oxidative stress in cornea. Aging Cell 2024; 23:e14319. [PMID: 39254179 PMCID: PMC11634732 DOI: 10.1111/acel.14319] [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: 03/23/2024] [Revised: 07/23/2024] [Accepted: 08/09/2024] [Indexed: 09/11/2024] Open
Abstract
Reactive oxygen species (ROS) and oxidative stress accelerate cellular aging, but their impact on different tissues varies. The cornea, known for its robust antioxidant defense systems, is relatively resistant to age-related diseases like cancer. However, the precise mechanisms by which the cornea maintains ROS homeostasis during aging remain unclear. Through comparative single-cell transcriptomic analysis of the cornea and other tissues in young and old nonhuman primates, we identified that a ZNF281 coding transcriptomic program is specifically activated in cornea during aging. Further investigation revealed that ZNF281 forms a positive feedback loop with FOXO3 to sense elevated levels of ROS and mitigate their effects potentially by regulating the mitochondrial respiratory chain and superoxide dismutase (SOD) expression. Importantly, we observed that overexpression of ZNF281 in MSCs prevented cellular senescence. In summary, these findings open up possibilities for understanding tissue-specific aging and developing new therapies targeting ROS damage.
Collapse
Affiliation(s)
- Yuhua Xiao
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Xu Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Zheyao Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Wangxuan Dai
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Xing Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Shuyao Zhang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Jiawei Zhong
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Jia Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Xu Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Lingyi Liang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| | - Youjin Hu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic CenterSun Yat‐Sen University, Guangdong Provincial Key Laboratory of Ophthalmology Visual ScienceGuangzhouChina
| |
Collapse
|
38
|
Alvarado-Vasquez N, Rangel-Escareño C, de Jesús Ramos-Abundis J, Becerril C, Negrete-García MC. The possible role of hypoxia-induced exosomes on the fibroblast metabolism in idiopathic pulmonary fibrosis. Biomed Pharmacother 2024; 181:117680. [PMID: 39549361 DOI: 10.1016/j.biopha.2024.117680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/15/2024] [Accepted: 11/07/2024] [Indexed: 11/18/2024] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) has a high incidence and prevalence among patients over 65 years old. While its exact etiology remains unknown, several risk factors have recently been identified. Hypoxia is associated with IPF due to the abnormal architecture of lung parenchyma and the accumulation of extracellular matrix produced by activated fibroblasts. Exosomes play a crucial role in intercellular communication during both physiological and pathological processes, including hypoxic diseases like IPF. Recent findings suggest that a hypoxic microenvironment influences the content of exosomes in various diseases, thereby altering cellular metabolism. Although the role of exosomes in IPF is an emerging area of research, the significance of hypoxic exosomes as inducers of metabolic reprogramming in fibroblasts is still underexplored. In this study, we analyze and discuss the relationship between hypoxia, exosomal cargo, and the metabolic reprogramming of fibroblasts in the progression of IPF.
Collapse
Affiliation(s)
- Noé Alvarado-Vasquez
- Department of Molecular Biomedicine and Translational Research, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Mexico City 14080, Mexico
| | - Claudia Rangel-Escareño
- Computational Genomics, National Institute of Genomic Medicine, Mexico City 14610, Mexico; School of Engineering and Sciences, Tecnologico de Monterrey, NL 64700, Mexico
| | | | - Carina Becerril
- Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Mexico City 14080, Mexico
| | - María Cristina Negrete-García
- Molecular Biology Laboratory, Department of Research in Pulmonary Fibrosis, National Institute of Respiratory Diseases "Ismael Cosío Villegas", Mexico City 14080, Mexico.
| |
Collapse
|
39
|
Zhai R, Chang L, Jiang J, Wang B, Zhu W. Cellular and Molecular Basis of Environment-Induced Color Change in a Tree Frog. Animals (Basel) 2024; 14:3472. [PMID: 39682437 DOI: 10.3390/ani14233472] [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] [Revised: 11/19/2024] [Accepted: 11/28/2024] [Indexed: 12/18/2024] Open
Abstract
Background color matching is essential for camouflage and thermoregulation in ectothermic vertebrates, yet several key cellular-level questions remain unresolved. For instance, it is unclear whether the number of chromatophores or the activity of individual chromatophores plays a more critical role in this process. Using single-cell RNA sequencing (scRNA-seq), we investigated the cellular and molecular mechanisms underlying color change in Rhacophorus dugritei, which adapted to its background by displaying light-green skin on white and black skin on black within two days. We identified two types of chromatophores in their skin, both responsible for the observed color differences. Our findings reveal that morphological color change (MCC) is the dominant process, with the number of chromatophores being more influential in driving color change than the transcriptional activity of melanogenesis in individual cells. Additionally, melanophores from darker individuals exhibited increased activity in energy metabolism pathways, while those from lighter individuals showed stronger immune-related gene expression, suggesting that background adaptation involves more than just morphological changes. Overall, this study successfully applied single-cell sequencing technology to investigate skin pigmentation in a non-model organism. Our results suggest that MCC driven by chromatophore proliferation is a key mechanism of background adaptation, offering new insights into amphibian color adaptation and environmental adaptation in other vertebrates.
Collapse
Affiliation(s)
- Runliang Zhai
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Liming Chang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
| | - Jianping Jiang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Bin Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Wei Zhu
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu 610213, China
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
40
|
Xu X, Wen Q, Lan T, Zeng L, Zeng Y, Lin S, Qiu M, Na X, Yang C. Time-resolved single-cell transcriptomic sequencing. Chem Sci 2024; 15:19225-19246. [PMID: 39568874 PMCID: PMC11575584 DOI: 10.1039/d4sc05700g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Accepted: 10/19/2024] [Indexed: 11/22/2024] Open
Abstract
Cells experience continuous transformation under both physiological and pathological circumstances. Single-cell RNA sequencing (scRNA-seq) is competent in disclosing the disparities of cells; nevertheless, it poses challenges in linking the individual cell state at distinct time points. Although computational approaches based on scRNA-seq data have been put forward for trajectory analysis, the result is based on assumptions and fails to reflect the actual states. Consequently, it is necessary to incorporate a "time anchor" into the scRNA-seq library for the temporal documentation of the dynamic expression pattern. This review comprehensively overviews the time-resolved single-cell transcriptomic sequencing methodologies and applications. As scRNA-seq functions as the basis for profiling single-cell expression patterns, the review initially introduces various scRNA-seq approaches. Subsequently, the review focuses on the different experimental strategies for introducing a "time anchor" to scRNA-seq, highlighting their principles, strengths, weaknesses, and comparing their adaptation in various scenarios. Next, it provides a brief summary of applications in immunity response, cancer progression, and embryo development. Finally, the review concludes with a forward-looking perspective on future advancements in time-resolved single-cell transcriptomic sequencing.
Collapse
Affiliation(s)
- Xing Xu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
- Department of Laboratory Medicine, Key Laboratory of Clinical Laboratory Technology for Precision Medicine, School of Medical Technology and Engineering, Fujian Medical University Fuzhou 350122 China
| | - Qianxi Wen
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Tianchen Lan
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Liuqing Zeng
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Yonghao Zeng
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Shiyan Lin
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Minghao Qiu
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Xing Na
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis & Instrumentation, The Key Laboratory of Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Collaborative Innovation Center of Chemistry for Energy Materials, Department of Chemical Biology, Department of Chemical Engineering, College of Chemistry and Chemical Engineering, Xiamen University Xiamen 361005 China
- Institute of Molecular Medicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine Shanghai 200127 China
| |
Collapse
|
41
|
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
|
42
|
Cho CH, Sim WJ, Cho NC, Lim W, Lim TG. Structure-based virtual screening of natural compounds in preventing skin senescence: The role of epigallocatechin gallate in protein kinase C alpha-specific inhibition against UV-induced photoaging. Heliyon 2024; 10:e39933. [PMID: 39553571 PMCID: PMC11567019 DOI: 10.1016/j.heliyon.2024.e39933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/28/2024] [Accepted: 10/28/2024] [Indexed: 11/19/2024] Open
Abstract
This study combines high-throughput screening and virtual molecular docking to identify natural compounds targeting PKC in skin aging. Go 6983, a PKC inhibitor, showed potent suppression of MMP-1 transcription. EGCG was one of the candidates that showed it could significantly lower UVB-induced MMP-1 expression in HaCaT cells, and it had a strong affinity for PKCα. Interestingly, EGCG is exclusively bound to PKCα, not the δ and ζ isoforms. Blocking PKCα did not elevate UVB-induced MMP-1 expression in HaCaT cells. In a model of human skin, EGCG stopped collagen breakdown and changes in epidermal thickness that were caused by UV light from the sun. This suggests that EGCG could be useful in dermatology and drug development. These findings highlight the role of structure-based screening in identifying candidate compounds with applications in the cosmetic, dermatological, preventive health, and pharmaceutical fields.
Collapse
Affiliation(s)
- Cheol Hyeon Cho
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Woo-Jin Sim
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
| | - Nam-Chul Cho
- Korea Chemical Bank, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Wonchul Lim
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea
| | - Tae-Gyu Lim
- Department of Food Science & Biotechnology, Sejong University, Seoul 05006, Republic of Korea
- Department of Food Science & Biotechnology, and Carbohydrate Bioproduct Research Center, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
43
|
Avelino TM, Harb SV, Adamoski D, Oliveira LCM, Horinouchi CDS, Azevedo RJD, Azoubel RA, Thomaz VK, Batista FAH, d'Ávila MA, Granja PL, Figueira ACM. Unveiling the impact of hypodermis on gene expression for advancing bioprinted full-thickness 3D skin models. Commun Biol 2024; 7:1437. [PMID: 39528562 PMCID: PMC11555214 DOI: 10.1038/s42003-024-07106-4] [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: 12/11/2023] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
3D skin models have been explored as an alternative method to the use of animals in research and development. Usually, human skin equivalents comprise only epidermis or epidermis/dermis layers. Herein, we leverage 3D bioprinting technology to fabricate a full-thickness human skin equivalent with hypodermis (HSEH). The collagen hydrogel-based structure provides a mimetic environment for skin cells to adhere, proliferate and differentiate. The effective incorporation of the hypodermis layer is evidenced by scanning electron microscopy, immunofluorescence, and hematoxylin and eosin staining. The transcriptome results underscore the pivotal role of the hypodermis in orchestrating the genetic expression of a multitude of genes vital for skin functionality, including hydration, development and differentiation. Accordingly, we evidence the paramount significance of full-thickness human skin equivalents with hypodermis layer to provide an accurate in vitro platform for disease modeling and toxicology studies.
Collapse
Affiliation(s)
- Thayná M Avelino
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
| | - Samarah V Harb
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
| | - Douglas Adamoski
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
| | - Larissa C M Oliveira
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
| | - Cintia D S Horinouchi
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
| | - Rafael J de Azevedo
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
| | - Rafael A Azoubel
- Department of Manufacturing and Materials Engineering, School of Mechanical Engineering, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Vanessa K Thomaz
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
| | - Fernanda A H Batista
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil
- Molecular Research Laboratory in Cardiology, Dante Pazzanese Institute of Cardiology (IDPC), São Paulo, Brazil
| | - Marcos Akira d'Ávila
- Department of Manufacturing and Materials Engineering, School of Mechanical Engineering, Universidade Estadual de Campinas (UNICAMP), Campinas, Brazil
| | - Pedro L Granja
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto Nacional de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Ana Carolina M Figueira
- Brazilian Center for Research in Energy and Materials (CNPEM), OKNational Laboratory of Bioscience (LNBio), Campinas, Brazil.
| |
Collapse
|
44
|
Li C, Ren S, Yan C, Wang C, Jiang T, Kang Y, Chen J, Xiong H, Guo J, Jiang G, Liu S, Nie P, Chen Z. HES1 revitalizes the functionality of aged adipose-derived stem cells by inhibiting the transcription of STAT1. Stem Cell Res Ther 2024; 15:399. [PMID: 39501364 PMCID: PMC11539794 DOI: 10.1186/s13287-024-04002-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: 06/09/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
BACKGROUND The effectiveness of adipose-derived stem cells (ADSCs) in therapy diminishes with age. It has been reported that transcription factors (TFs) play a crucial role in the aging and functionality of stem cells. Nevertheless, there is limited understanding regarding the involvement of TFs in the aging mechanism of ADSCs. METHODS RNA sequencing (RNA-seq) was utilized to discern the differentially expressed genes in ADSCs obtained from donors of varying ages. TFs exhibiting significant variations across age groups were identified and subsequently validated. ADSCs were manipulated to exhibit either enhanced expression or reduced levels of HES1 and STAT1 via lentivirus transfection and small interfering RNA (siRNA) techniques. The impact of these genetic alterations on ADSCs' proliferation, migration, and cellular senescence was assessed using EdU, transwell, and senescence-activated β-galactosidase (SA-β-gal) staining assays. The DNA sequences bound by HES1 were investigated through the CUT & Tag assay. Lastly, the therapeutic efficacy of aged ADSCs with HES1 overexpression was evaluated in skin injury model of male Sprague-Dawley rats. RESULTS 678 genes showed differential expression between ADSCs obtained from young and old donors (Y-ADSCs and O-ADSCs), with 47 of these genes being TFs. Notably, the expression of the TF hairy and enhancer of split 1 (HES1) was notably reduced in ADSCs from old donors. Introducing HES1 overexpression in aged ADSCs resulted in improved cellular function and the suppression of cellular senescence, while reducing HES1 levels in young ADSCs had the opposite effect. Mechanistically, HES1 was found to interact with the promoter region of another TF, signal transducer and activator of transcription 1 (STAT1), to inhibit its transcription. Knocking down STAT1 could fully reverse the negative effects caused by decreased HES1 in ADSCs, leading to a reduction in the secretion of pro-inflammatory cytokines such as TNF-α, IL-6, and IL-8. Ultimately, restoring HES1 expression in aged ADSCs demonstrated enhanced therapeutic potential in promoting skin wound healing. CONCLUSION HES1 acts as an inhibitor of cellular senescence in the aging progression of ADSCs through the modulation of STAT1 expression, suggesting a promising avenue for rejuvenating senescent ADSCs and improving wound healing.
Collapse
Affiliation(s)
- Chengcheng Li
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Sen Ren
- Department of Neurosurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Chengqi Yan
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Cheng Wang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Tao Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Yu Kang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Jing Chen
- Department of Dermatology, Wuhan No.1 Hospital, Wuhan, Hubei, 430000, China
| | - Hewei Xiong
- Department of Emergency Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahe Guo
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Guoyong Jiang
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Shuoyuan Liu
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Pengjuan Nie
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China
| | - Zhenbing Chen
- Department of Hand Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1277 Jiefang Avenue, Wuhan, 430022, China.
| |
Collapse
|
45
|
Li Y, Wang Q, Xuan Y, Zhao J, Li J, Tian Y, Chen G, Tan F. Investigation of human aging at the single-cell level. Ageing Res Rev 2024; 101:102530. [PMID: 39395577 DOI: 10.1016/j.arr.2024.102530] [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: 07/02/2024] [Revised: 08/18/2024] [Accepted: 09/30/2024] [Indexed: 10/14/2024]
Abstract
Human aging is characterized by a gradual decline in physiological functions and an increased susceptibility to various diseases. The complex mechanisms underlying human aging are still not fully elucidated. Single-cell sequencing (SCS) technologies have revolutionized aging research by providing unprecedented resolution and detailed insights into cellular diversity and dynamics. In this review, we discuss the application of various SCS technologies in human aging research, encompassing single-cell, genomics, transcriptomics, epigenomics, and proteomics. We also discuss the combination of multiple omics layers within single cells and the integration of SCS technologies with advanced methodologies like spatial transcriptomics and mass spectrometry. These approaches have been essential in identifying aging biomarkers, elucidating signaling pathways associated with aging, discovering novel aging cell subpopulations, uncovering tissue-specific aging characteristics, and investigating aging-related diseases. Furthermore, we provide an overview of aging-related databases that offer valuable resources for enhancing our understanding of the human aging process.
Collapse
Affiliation(s)
- Yunjin Li
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China
| | - Qixia Wang
- Department of General Practice, Xi'an Central Hospital, Xi'an, Shaanxi 710000, China
| | - Yuan Xuan
- Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, China
| | - Jian Zhao
- Department of Oncology-Pathology Karolinska Institutet, BioClinicum, Solna, Sweden
| | - Jin Li
- Shandong Zhifu Hospital, Yantai, Shandong 264000, China
| | - Yuncai Tian
- Shanghai AZ Science and Technology Co., Ltd, Shanghai 200000, China
| | - Geng Chen
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China.
| | - Fei Tan
- Shanghai Skin Disease Hospital, School of Medicine, Tongji University, Shanghai 200443, China; Shanghai Skin Disease Clinical College, The Fifth Clinical Medical College, Anhui Medical University, Shanghai Skin Disease Hospital, Shanghai 200443, China.
| |
Collapse
|
46
|
Gadre P, Markova P, Ebrahimkutty M, Jiang Y, Bouzada FM, Watt FM. Emergence and properties of adult mammalian epidermal stem cells. Dev Biol 2024; 515:129-138. [PMID: 39059680 DOI: 10.1016/j.ydbio.2024.07.014] [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: 12/10/2023] [Revised: 05/08/2024] [Accepted: 07/23/2024] [Indexed: 07/28/2024]
Abstract
In this review we discuss how the mammalian interfollicular epidermis forms during development, maintains homeostasis, and is repaired following wounding. Recent studies have provided new insights into the relationship between the stem cell compartment and the differentiating cell layers; the ability of differentiated cells to dedifferentiate into stem cells; and the epigenetic memory of epidermal cells following wounding.
Collapse
Affiliation(s)
- Purna Gadre
- Directors' Unit, EMBL-Heidelberg, Meyerhofstr. 1, 69117, Heidelberg, Germany
| | - Pavlina Markova
- Directors' Unit, EMBL-Heidelberg, Meyerhofstr. 1, 69117, Heidelberg, Germany
| | | | - Yidan Jiang
- Directors' Unit, EMBL-Heidelberg, Meyerhofstr. 1, 69117, Heidelberg, Germany
| | - Francisco M Bouzada
- Directors' Unit, EMBL-Heidelberg, Meyerhofstr. 1, 69117, Heidelberg, Germany
| | - Fiona M Watt
- Directors' Unit, EMBL-Heidelberg, Meyerhofstr. 1, 69117, Heidelberg, Germany.
| |
Collapse
|
47
|
Liu F, Liu J, Luo Y, Wu S, Liu X, Chen H, Luo Z, Yuan H, Shen F, Zhu F, Ye J. A Single-Cell Metabolic Profiling Characterizes Human Aging via SlipChip-SERS. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2406668. [PMID: 39231358 PMCID: PMC11538647 DOI: 10.1002/advs.202406668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/12/2024] [Indexed: 09/06/2024]
Abstract
Metabolic dysregulation is a key driver of cellular senescence, contributing to the progression of systemic aging. The heterogeneity of senescent cells and their metabolic shifts are complex and unexplored. A microfluidic SlipChip integrated with surface-enhanced Raman spectroscopy (SERS), termed SlipChip-SERS, is developed for single-cell metabolism analysis. This SlipChip-SERS enables compartmentalization of single cells, parallel delivery of saponin and nanoparticles to release intracellular metabolites and to realize SERS detection with simple slipping operations. Analysis of different cancer cell lines using SlipChip-SERS demonstrated its capability for sensitive and multiplexed metabolic profiling of individual cells. When applied to human primary fibroblasts of different ages, it identified 12 differential metabolites, with spermine validated as a potent inducer of cellular senescence. Prolonged exposure to spermine can induce a classic senescence phenotype, such as increased senescence-associated β-glactosidase activity, elevated expression of senescence-related genes and reduced LMNB1 levels. Additionally, the senescence-inducing capacity of spermine in HUVECs and WRL-68 cells is confirmed, and exogenous spermine treatment increased the accumulation and release of H2O2. Overall, a novel SlipChip-SERS system is developed for single-cell metabolic analysis, revealing spermine as a potential inducer of senescence across multiple cell types, which may offer new strategies for addressing ageing and ageing-related diseases.
Collapse
Affiliation(s)
- Fugang Liu
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Jiaqing Liu
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Yang Luo
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Siyi Wu
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Xu Liu
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Haoran Chen
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Zhewen Luo
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Haitao Yuan
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Feng Shen
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Fangfang Zhu
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
| | - Jian Ye
- School of Biomedical EngineeringShanghai Jiao Tong UniversityShanghai200030China
- State Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200032China
- Institute of Medical RoboticsShanghai Jiao Tong UniversityShanghai200240China
- Shanghai Key Laboratory of Gynecologic OncologyRen Ji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| |
Collapse
|
48
|
Zhou F, Sun Y, Chen X, Hou W, Shen J, Lai W, Han K, Zheng Y. Differences in cell subsets between sun-exposed and unexposed skin: preliminary single-cell sequencing and biological analysis from a single case. Front Med (Lausanne) 2024; 11:1453940. [PMID: 39540047 PMCID: PMC11558528 DOI: 10.3389/fmed.2024.1453940] [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: 06/24/2024] [Accepted: 09/23/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction The composition and subsets of skin cells continuously change in a dynamic manner. However, the specific microcosmic alterations of human photoaged skin, independent of chronologic aging, remain unclear and have been infrequently analyzed. This study aimed to evaluate the biological processes and mechanisms underlying cell-subgroup alterations in skin photoaging. Methods We utilized single-cell sequencing and biological analysis from a single case to investigate the effects of photoaging. Skin punch biopsies were taken from sun-exposed forearm skin and unexposed buttock skin from the same individual for comparative analysis. Results Our analysis identified 25 cell clusters and 12 skin cell types, revealing significant changes in unique gene expressions between the sun-exposed and unexposed skin samples. A comparison of cell numbers within each cluster revealed 9 dominant cell clusters in sun-exposed skin and 16 dominant cell clusters in unexposed skin. Enrichment analysis indicated that PD-L1 expression and the PD-1 checkpoint pathway were more prominent in sun-exposed skin, while MAPK, TNF-alpha, TGF-beta, and apoptosis pathways were more enriched in hair follicle cells of sun-exposed skin. Discussion This study reveals changes in cell components in photoaged skin from a single case and provides novel insights into cellular subpopulations and pathology during repeated UVA-induced skin damage. These findings enhance our understanding of the complex interplay between different cells in photoaged skin and offer potential targets for preventing human skin photoaging and UV-induced skin cancers.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yu Sun
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xinling Chen
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Wenyi Hou
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jing Shen
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Lai
- Department of Dermato-venereology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kai Han
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yue Zheng
- Department of Dermatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
49
|
Zhao M, He Z, Liu L, Wang Y, Gao L, Shang Y, Zhu M. Causal and mediating effects of lipid and facial aging: association study integrating GWAS, eQTL, mQTL, and pQTL data. Lipids Health Dis 2024; 23:342. [PMID: 39434152 PMCID: PMC11492622 DOI: 10.1186/s12944-024-02328-1] [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] [Accepted: 10/07/2024] [Indexed: 10/23/2024] Open
Abstract
BACKGROUND Increasing evidence suggests a potential causal association between lipid levels and facial aging. The aim of this study was to investigate the relationship between levels of specific lipids and facial aging via Mendelian randomization methods. Additionally, this study aimed to identify mediators and explore relevant genes and drug targets. METHODS In this study, genome-wide association data on plasma lipids from 7,174 Finnish individuals in the UK Biobank were used. Two-sample Mendelian randomization was applied to assess the causal effects of specific lipids on facial aging. Sensitivity and pleiotropy analyses were conducted to ensure the robustness and reliability of the results. Multivariate Mendelian randomization was conducted to account for the potential impact of confounding factors. Furthermore, summary-data-based Mendelian randomization was used to identify relevant genes, which were validated through multiomics data. Finally, drug‒gene interactions were explored via molecular docking techniques. RESULTS Two-sample Mendelian randomization analysis revealed a causal relationship between lipid levels and facial aging. According to the multivariate Mendelian randomization results, smoking was found to mediate this association, and these lipids remained significantly associated with facial aging, even after accounting for environmental confounders. Using summary-data-based Mendelian randomization, CYP21A2, CCND1, PSMA4, and MED1 were identified as potential gene targets, with MED1 further validated through pQTL and mQTL data. Additionally, the MED1 protein was found to bind spontaneously with astragalin, fenofibrate, and ginsenoside. CONCLUSIONS The results revealed a causal relationship between lipid levels and facial aging, revealing key gene targets that were still significantly associated with facial aging after controlling for environmental confounders. Additionally, the interactions between MED1 and certain drugs may indicate potential pathways for therapeutic interventions related to facial aging.
Collapse
Affiliation(s)
- Mingjian Zhao
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China
| | - Zhanchen He
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China
| | - Lukuan Liu
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yichen Wang
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China
| | - LinQi Gao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China
| | - Yuxuan Shang
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China.
| | - Mengru Zhu
- Department of Plastic Surgery, The First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian, 116011, China.
| |
Collapse
|
50
|
Chovatiya G, Wang AB, Versluis P, Bai CK, Huang SY, DeBerardine M, Ray J, Ozer A, Lis JT, Tumbar T. A lineage-specific nascent RNA assay unveils principles of gene regulation in tissue biology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.15.618417. [PMID: 39464031 PMCID: PMC11507779 DOI: 10.1101/2024.10.15.618417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
Gene regulatory mechanisms that modulate RNA Polymerase II activity are difficult to access in mammalian tissues composed of multiple cell lineages. Here, we develop a nascent RNA assay (PReCIS-seq) that measures lineage-specific transcriptionally-engaged Pol II on genes and DNA enhancer elements in intact mouse tissue. By employing keratinocytes as a prototype lineage, we unearth Pol II promoter-recruitment versus pause-release mechanisms operating in adult skin homeostasis. Moreover, we relate active enhancer proximity and transcription factor binding motifs on promoters to Pol II activity and promoter-proximal pausing level. Finally, we find Pol II firing rapidly into elongation on lineage identity genes and highly paused on cellular safeguarding genes in a context-dependent manner. Our work provides a basic platform to investigate mechanistic principles of gene regulation in individual lineages of complex mammalian tissues.
Collapse
Affiliation(s)
- Gopal Chovatiya
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Alex B Wang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Philip Versluis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Chris K Bai
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Sean Y Huang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Michael DeBerardine
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Judhajeet Ray
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Abdullah Ozer
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - John T Lis
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Tudorita Tumbar
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|