1
|
Peng Q, Liu Y, Huels A, Zhang C, Yu Y, Qiu W, Cai X, Zhao Y, Schikowski T, Merches K, Liu Y, Yang Y, Wang J, Zhao Y, Jin L, Zhang L, Krutmann J, Wang S. Genetic Variants in Telomerase Reverse Transcriptase Contribute to Solar Lentigines. J Invest Dermatol 2023; 143:1062-1072.e25. [PMID: 36572090 DOI: 10.1016/j.jid.2022.11.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 10/01/2022] [Accepted: 11/11/2022] [Indexed: 12/26/2022]
Abstract
Solar lentigines (SLs) are a hallmark of human skin aging. They result from chronic exposure to sunlight and other environmental stressors. Recent studies also imply genetic factors, but findings are partially conflicting and lack of replication. Through a multi-trait based analysis strategy, we discovered that genetic variants in telomerase reverse transcriptase were significantly associated with non-facial SL in two East Asian (Taizhou longitudinal cohort, n = 2,964 and National Survey of Physical Traits, n = 2,954) and one Caucasian population (SALIA, n = 462), top SNP rs2853672 (P-value for Taizhou longitudinal cohort = 1.32 × 10‒28 and P-value for National Survey of Physical Traits = 3.66 × 10‒17 and P-value for SALIA = 0.0007 and Pmeta = 4.93 × 10‒44). The same variants were nominally associated with facial SL but not with other skin aging or skin pigmentation traits. The SL-enhanced allele/haplotype upregulated the transcription of the telomerase reverse transcriptase gene. Of note, well-known telomerase reverse transcriptase‒related aging markers such as leukocyte telomere length and intrinsic epigenetic age acceleration were not associated with SL. Our results indicate a previously unrecognized role of telomerase reverse transcriptase in skin aging‒related lentigines formation.
Collapse
Affiliation(s)
- Qianqian Peng
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yu Liu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Anke Huels
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany; Faculty of Epidemiology, Rollins School of Public Health, Emory University, Atlanta, Georgia, USA
| | - Canfeng Zhang
- Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yao Yu
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China; CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Wenqing Qiu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xiyang Cai
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yuepu Zhao
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Tamara Schikowski
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Katja Merches
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Yun Liu
- MOE Key Laboratory of Metabolism and Molecular Medicine, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Yajun Yang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Human Phenome Institute, Fudan University, Shanghai, China; Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Human Phenome Institute, Fudan University, Shanghai, China; Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Yong Zhao
- Key Laboratory of Gene Engineering of the Ministry of Education, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Human Phenome Institute, Fudan University, Shanghai, China; Ministry of Education Key Laboratory of Contemporary Anthropology, Department of Anthropology and Human Genetics, School of Life Sciences, Fudan University, Shanghai, China
| | - Liang Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, China; Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jean Krutmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany; State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, Human Phenome Institute, Fudan University, Shanghai, China; Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Sijia Wang
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China; Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, China.
| |
Collapse
|
2
|
Mi Y, Zhong L, Lu S, Hu P, Pan Y, Ma X, Yan B, Wei Z, Yang G. Quercetin promotes cutaneous wound healing in mice through Wnt/β-catenin signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 290:115066. [PMID: 35122975 DOI: 10.1016/j.jep.2022.115066] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/17/2022] [Accepted: 01/30/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Oxytropis falcata Bunge is a legume distributed in Northwest China, which is mainly used to treat knife wounds and inflammation. Quercetin is a bioactive flavonoid in O. falcata and becomes a promising healing compound for its angiogenic and anti-inflammatory activities. However, the healing mechanism of quercetin in cutaneous wound remains elusive. AIM OF THE STUDY The purpose of this study was to evaluate the healing effect of quercetin on cutaneous wound models in vivo and in vitro, and to reveal the Wnt/β-catenin pathway and Telomerase reverse transcriptase (TERT) involved mechanisms. MATERIALS AND METHODS The effects of quercetin on the proliferation and migration of 4 kinds of skin cells were determined by CCK-8 and scratch assay. The wound-healing capacity of quercetin was evaluated in cutaneous wound model of C57BL/6 mice and the wound healing degree was observed by histological staining. The expressions of inflammatory factors, growth factors and the related proteins were detected via Western blot and RT-qPCR analyses. The molecular docking was adopted to evaluate the binding ability of quercetin and TERT. RESULTS Quercetin could promote both proliferation and migration of fibroblasts, and enhance cutaneous wound healing capacity in mice. Compared to the control group, the wound healing rates in low (1.5 mg/mL), medium (3.0 mg/mL) and high dose (6.0 mg/mL) quercetin groups reached 94.67%, 97.31% and 98.42%, respectively. Moreover, the dermal structure in quercetin treated mice restored normal and the content of collagen fiber became abundant after administration. The levels of inflammatory factors, including tumor necrosis factor-α, interleukin-1β and interleukin-6 were significantly reduced after quercetin administration. Among which, the level of IL-1β in cutaneous wound was 0.007 times higher than that of the control group when treated with quercetin of high dose (6.0 mg/mL). The improved level of GSH in quercetin treated cutaneous wounds also indicated its higher antioxidant ability. In addition, dose-dependent positive associations were found in the expression levels of vascular endothelial growth factor, fibroblast growth factor and alpha smooth muscle actin in quercetin treated cutaneous wounds. The significantly upregulated protein levels of Wnt and β-catenin further indicated the important role of quercetin in promoting wound healing in mice. According to molecular docking analysis, the formed hydrogen bonds between quercetin and Ala195, Gln308, Asn369 and Lys372 residues of TERT also indicated the indispensable role of TERT in improving wound healing capacity. CONCLUSION Quercetin effectively promoted cutaneous wound healing by enhancing the proliferation and migration of fibroblasts, as well as inhibiting inflammation and increasing the expression of growth factors in mice via Wnt/β-catenin signaling pathway and TERT. It provides a basis for a more thorough understanding of mechanism of action of O. falcata Bunge in the treatment of knife wounds and burns.
Collapse
Affiliation(s)
- Yuhui Mi
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Lei Zhong
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Saijian Lu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Po Hu
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Yang Pan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| | - Xuelin Ma
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Binghui Yan
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Zhenhuan Wei
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Guangming Yang
- School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| |
Collapse
|
4
|
Moftah NH, El-Barbary R, Rashed L, El-Sammad N. Assessment of telomerase activity in nonsegmental vitiligo tissue: a pilot study. Clin Exp Dermatol 2019; 44:747-752. [PMID: 30706515 DOI: 10.1111/ced.13892] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2018] [Indexed: 01/15/2023]
Abstract
BACKGROUND Vitiligo is characterized by loss of melanocytes; therefore, an increased risk of photoageing and cancer are expected. However, a low incidence of cancer and sun damage in vitiliginous skin has been reported. Telomerase is a specialized cellular enzyme catalysing the synthesis of telomeres, and an increased level of the telomerase activity has been highlighted in most of human cancer cells and cancer cell lines. AIM To assess relative telomerase activity (RTA) among patients with nonsegmental vitiligo. METHODS In this case-control study, skin biopsy specimens were taken from 20 patients (one from lesional and another from nonlesional skin) and from sun-protected skin from 10 healthy age-, sex- and skin phototype-matched healthy controls. PCR ELISA was performed for assessment of RTA. RESULTS RTA in lesional skin biopsies from patients with nonsegmental vitiligo was significantly decreased compared with nonlesional skin and healthy control skin samples, with no significant difference between the latter two. RTA in lesional skin was negatively correlated with Vitiligo Area Scoring Index but not correlated with Vitiligo Disease Activity score or RTA of nonlesional skin. Neither lesional nor nonlesional RTA levels showed any correlation with patient sex, age, skin phototype or with disease duration. CONCLUSION Low levels of RTA in vitiliginous skin may help to explain the lower chance of developing skin cancer and decreased incidence of actinic damage in vitiliginous skin.
Collapse
Affiliation(s)
- N H Moftah
- Department of Dermatology and Venereology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - R El-Barbary
- Department of Dermatology and Venereology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| | - L Rashed
- Department of Biochemistry, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - N El-Sammad
- Department of Dermatology and Venereology, Faculty of Medicine for Girls, Al-Azhar University, Cairo, Egypt
| |
Collapse
|
6
|
Lotfi RA, El Zawahry KM, Kamar ZA, Hashem Z. Effects of smoking on human telomerase reverse transcriptase expression in the skin. Int J Dermatol 2014; 53:1205-12. [PMID: 24601896 DOI: 10.1111/ijd.12467] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Telomeres are DNA-protein complexes that cap chromosomal ends, promoting chromosome stability. Telomerase is a ribonucleoprotein complex with a direct telomere protective function. Telomere shortening represents lifetime exposure to oxidative stress and is negatively correlated with age, smoking, and mortality. Smoking increases oxidative DNA modification and thus may influence telomere dynamics and human telomerase reverse transcriptase (hTERT) activity. OBJECTIVES This study investigated the effect of smoking on hTERT expression in the skin of smokers and non-smokers. METHODS A cross-sectional study in 20 current smokers and 20 non-smokers was conducted. Three-mm punch skin biopsies were obtained. Biopsies were examined in routine hematoxylin and eosin staining and immunohistochemistry to investigate expression of hTERT. RESULTS All skin biopsies from smokers and non-smokers showed cytoplasmic staining in epidermal cells. Sections positive for hTERT expression showed nuclear and some nucleolar staining in cells of the basal and suprabasal layers. In the dermis, hTERT expression was present in some skin appendages. The epidermis of smokers showed positive hTERT in 55% and negative hTERT in 45% of biopsies. The epidermis of non-smokers showed positive hTERT in 70% and negative hTERT in 30% of biopsies (P > 0.05). Among smokers, 20% showed positive and 80% showed negative hTERT expression in the dermis. Among non-smokers, 30% showed positive and 70% showed negative hTERT expression in the dermis. A higher mean pack year value was found in subjects with negative rather than positive hTERT expression (P < 0.01). In addition, pack year was inversely correlated with hTERT expression in the epidermis (P < 0.05): as pack year increased, hTERT expression decreased. Mean pack year values were higher in subjects with negative rather than positive hTERT expression in the dermis (P < 0.01). CONCLUSIONS This research focused on smoking as a lifestyle factor that may alter telomere length and subsequently telomerase kinetics in the skin. Findings showed a higher percentage of negative hTERT in the epidermis and dermis among smokers compared with non-smokers and a higher percentage of positive hTERT expression among non-smokers (not significant). The results of this work showed a statistically significant higher mean pack year count among cases with negative rather than positive hTERT expression in the epidermis and dermis. Pack year count was inversely correlated to hTERT scoring in the epidermis (percentage of cells stained) and hTERT expression in the dermis. Thus, smoking may affect telomerase activity in the skin, thereby contributing to skin aging.
Collapse
Affiliation(s)
- Ranya A Lotfi
- Department of Dermatology and Venereology, Ain Shams University, Cairo, Egypt
| | | | | | | |
Collapse
|
8
|
Chen IP, Henning S, Faust A, Boukamp P, Volkmer B, Greinert R. UVA-induced epigenetic regulation of P16(INK4a) in human epidermal keratinocytes and skin tumor derived cells. Photochem Photobiol Sci 2011; 11:180-90. [PMID: 21986889 DOI: 10.1039/c1pp05197k] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
UVA-radiation (315-400 nm) has been demonstrated to be capable of inducing DNA damage and is regarded as a carcinogen. While chromosomal aberrations found in UVA-irradiated cells and skin tumors provided evidence of the genetic involvement in UVA-carcinogenesis, its epigenetic participation is still illusive. We thus analysed the epigenetic patterns of 5 specific genes that are involved in stem cell fate (KLF4, NANOG), telomere maintenance (hTERT) and tumor suppression in cell cycle control (P16(INK4a), P21(WAFI/CIPI)) in chronically UVA-irradiated HaCaT human keratinocytes. A striking reduction of the permissive histone mark H3K4me3 has been detected in the promoter of P16(INK4a) (4-fold and 9-fold reduction for 10 and 15 weeks UVA-irradiated cells, respectively), which has often been found deregulated in skin cancers. This alteration in histone modification together with a severe promoter hypermethylation strongly impaired the transcription of P16(INK4a) (20-fold and 40-fold for 10 weeks and 15 weeks UVA-irradiation, respectively). Analysis of the skin tumor-derived cells revealed the same severe impairment of the P16(INK4a) transcription attributed to promoter hypermethylation and enrichment of the heterochromatin histone mark H3K9me3 and the repressive mark H3K27me3. Less pronounced UVA-induced epigenetic alterations were also detected for the other genes, demonstrating for the first time that UVA is able to modify transcription of skin cancer associated genes by means of epigenetic DNA and histone alterations.
Collapse
Affiliation(s)
- I-Peng Chen
- Dept. Mol. Cell Biology, Center of Dermatology, Elbekliniken, Stade/Buxtehude, Germany
| | | | | | | | | | | |
Collapse
|