1
|
Romashin DD, Tolstova TV, Varshaver AM, Kozhin PM, Rusanov AL, Luzgina NG. Keratins 6, 16, and 17 in Health and Disease: A Summary of Recent Findings. Curr Issues Mol Biol 2024; 46:8627-8641. [PMID: 39194725 DOI: 10.3390/cimb46080508] [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: 06/05/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Keratins 6, 16, and 17 occupy unique positions within the keratin family. These proteins are not commonly found in the healthy, intact epidermis, but their expression increases in response to damage, inflammation, and hereditary skin conditions, as well as cancerous cell transformations and tumor growth. As a result, there is an active investigation into the potential use of these proteins as biomarkers for different pathologies. Recent studies have revealed the role of these keratins in regulating keratinocyte migration, proliferation, and growth, and more recently, their nuclear functions, including their role in maintaining nuclear structure and responding to DNA damage, have also been identified. This review aims to summarize the latest research on keratins 6, 16, and 17, their regulation in the epidermis, and their potential use as biomarkers in various skin conditions.
Collapse
Affiliation(s)
| | | | | | - Peter M Kozhin
- Institute of Biomedical Chemistry, Moscow 119121, Russia
| | | | | |
Collapse
|
2
|
Romashin D, Rusanov A, Tolstova T, Varshaver A, Netrusov A, Kozhin P, Luzgina N. Loss of mutant p53 in HaCaT keratinocytes promotes cadmium-induced keratin 17 expression and cell death. Biochem Biophys Res Commun 2024; 709:149834. [PMID: 38547608 DOI: 10.1016/j.bbrc.2024.149834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Cadmium exposure induces dermatotoxicity and epidermal barrier disruption and leads to the development of various pathologies. HaCaT cells are immortalized human keratinocytes that are widely used as alternatives to primary human keratinocytes, particularly for evaluating cadmium toxicity. HaCaT cells bear two gain-of-function (GOF) mutations in the TP53 gene, which strongly affect p53 function. Mutant forms of p53 are known to correlate with increased resistance to various stimuli, including exposure to cytotoxic substances. In addition, keratin 17 (KRT17) was recently shown to be highly expressed in HaCaT cells in response to genotoxic stress. Moreover, p53 is a direct transcriptional repressor of KRT17. However, the impact of TP53 mutations in HaCaT cells on the regulation of cell death and keratin 17 expression is unclear. In this study, we aimed to evaluate the impact of p53 on the response to Cd-induced cytotoxicity. METHODS AND RESULTS Employing the MTT assay and Annexin V/propidium iodide staining, we demonstrated that knockout of TP53 leads to a decrease in the sensitivity of HaCaT cells to the cytotoxic effects of cadmium. Specifically, HaCaT cells with TP53 knockout (TP53 KO HaCaT) exhibited cell death at a cadmium concentration of 10 μM or higher, whereas wild-type cells displayed cell death at a concentration of 30 μM. Furthermore, apoptotic cells were consistently detected in TP53 KO HaCaT cells upon exposure to low concentrations of cadmium (10 and 20 μM) but not in wild-type cells. Our findings also indicate that cadmium cytotoxicity is mediated by reactive oxygen species (ROS), which were significantly increased only in TP53 knockout cells treated with 30 μM cadmium. An examination of proteomic data revealed that TP53 knockout in HaCaT cells resulted in the upregulation of proteins involved in the regulation of apoptosis, redox systems, and DNA repair. Moreover, RT‒qPCR and immunoblotting showed that cadmium toxicity leads to dose-dependent induction of keratin 17 in p53-deficient cells but not in wild-type cells. CONCLUSIONS The connection between mutant p53 in HaCaT keratinocytes and increased resistance to cadmium toxicity was demonstrated for the first time. Proteomic profiling revealed that TP53 knockout in HaCaT cells led to the activation of apoptosis regulatory circuits, redox systems, and DNA repair. In addition, our data support the involvement of keratin 17 in the regulation of DNA repair and cell death. Apparently, the induction of keratin 17 is p53-independent but may be inhibited by mutant p53.
Collapse
Affiliation(s)
- Daniil Romashin
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexander Rusanov
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia.
| | - Tatiana Tolstova
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexandra Varshaver
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Alexander Netrusov
- Lomonosov Moscow State University, GSP-1, Leninskie Gory, Moscow, 119991, Russia
| | - Peter Kozhin
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| | - Nataliya Luzgina
- Institute of Biomedical Chemistry, 10 Pogodinskaya St., Moscow, 119121, Russia
| |
Collapse
|
3
|
Cohen E, Johnson CN, Wasikowski R, Billi AC, Tsoi LC, Kahlenberg JM, Gudjonsson JE, Coulombe PA. Significance of stress keratin expression in normal and diseased epithelia. iScience 2024; 27:108805. [PMID: 38299111 PMCID: PMC10828818 DOI: 10.1016/j.isci.2024.108805] [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: 09/06/2023] [Revised: 11/30/2023] [Accepted: 01/02/2024] [Indexed: 02/02/2024] Open
Abstract
A group of keratin intermediate filament genes, the type II KRT6A-C and type I KRT16 and KRT17, are deemed stress responsive as they are induced in keratinocytes of surface epithelia in response to environmental stressors, in skin disorders (e.g., psoriasis) and in carcinomas. Monitoring stress keratins is widely used to identify keratinocytes in an activated state. Here, we analyze single-cell transcriptomic data from healthy and diseased human skin to explore the properties of stress keratins. Relative to keratins occurring in healthy skin, stress-induced keratins are expressed at lower levels and show lesser type I-type II pairwise regulation. Stress keratins do not "replace" the keratins expressed during normal differentiation nor reflect cellular proliferation. Instead, stress keratins are consistently co-regulated with genes with roles in differentiation, inflammation, and/or activation of innate immunity at the single-cell level. These findings provide a roadmap toward explaining the broad diversity and contextual regulation of keratins.
Collapse
Affiliation(s)
- Erez Cohen
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Craig N. Johnson
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Rachael Wasikowski
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Allison C. Billi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lam C. Tsoi
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - J. Michelle Kahlenberg
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Johann E. Gudjonsson
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Division of Rheumatology, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Pierre A. Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA
- Department of Dermatology, University of Michigan Medical School, Ann Arbor, MI, USA
- Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
4
|
Yue Z, Lin J, Lu X, Gao Q, Pan M, Zhang Y, Shen S, Zhu WG, Paus R. Keratin 17 Impacts Global Gene Expression and Controls G2/M Cell Cycle Transition in Ionizing Radiation-Induced Skin Damage. J Invest Dermatol 2023; 143:2436-2446.e13. [PMID: 37414246 DOI: 10.1016/j.jid.2023.02.043] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 01/30/2023] [Accepted: 02/08/2023] [Indexed: 07/08/2023]
Abstract
Keratin 17 (K17) is a cytoskeletal protein that is part of the intermediate filaments in epidermal keratinocytes. In K17-/- mice, ionizing radiation induced more severe hair follicle damage, whereas the epidermal inflammatory response was attenuated compared with that in wild-type mice. Both p53 and K17 have a major impact on global gene expression because over 70% of the differentially expressed genes in the skin of wild-type mice showed no expression change in p53-/- or K17-/- skin after ionizing radiation. K17 does not interfere with the dynamics of p53 activation; rather, global p53 binding in the genome is altered in K17-/- mice. The absence of K17 leads to aberrant cell cycle progression and mitotic catastrophe in epidermal keratinocytes, which is due to nuclear retention, thus reducing the degradation of B-Myb, a key regulator of the G2/M cell cycle transition. These results expand our understanding of the role of K17 in regulating global gene expression and ionizing radiation-induced skin damage.
Collapse
Affiliation(s)
- ZhiCao Yue
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China.
| | - JianQiong Lin
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - XiaoPeng Lu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China; Department of Biochemistry & Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - QingXiang Gao
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - MeiPing Pan
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - YaFei Zhang
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - SiTing Shen
- Department of Cell Biology & Medical Genetics, Shenzhen University Medical School, Shenzhen, China; International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China
| | - Wei-Guo Zhu
- International Cancer Center, Shenzhen University Medical School, Shenzhen, China; Guangdong Key Laboratory of Genome Instability and Human Disease Prevention, Shenzhen University Medical School, Shenzhen, China; Department of Biochemistry & Molecular Biology, Shenzhen University Medical School, Shenzhen, China
| | - Ralf Paus
- Dr. Philip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA; Center for Dermatology Research, School of Biological Sciences, The University of Manchester and NIHR Biomedical Research Center, Manchester, United Kingdom
| |
Collapse
|
5
|
Yang K, Song S, Zhang Y, Shen S, Xu X, Yue Z. Programmed gene expression change in mouse skin after ultraviolet radiation damage. Exp Dermatol 2021; 31:862-868. [PMID: 34951733 DOI: 10.1111/exd.14519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/24/2021] [Accepted: 12/21/2021] [Indexed: 11/29/2022]
Abstract
Ultraviolet (UV) radiation is a major cause of skin damage and carcinogenesis. Here, we systematically analyse the acute gene expression change in skin in vivo after UV exposure, aiming to establish the common C57BL/6 mouse strain as a convenient model for future pathological research and drug discovery. The back fur of C57BL/6 mice was depilated, and a mixed UV light source was used to irradiate the skin. Full-thickness skin samples were collected at 0, 0.5, 2, 6, 12 and 24 h. Total RNAs were extracted and subjected to RNA sequencing analysis. We found that the gene expression change in mouse skin is highly similar to previous reports in human skin. These include down-regulation of differentiation-related genes and extracellular matrix genes, and up-regulation of cytokine/chemokine genes. An early wave of activator protein 1 (AP-1) expression is induced, whereas activation of the p53 pathway is not significant. The impact of the AP-1 transcription factors and the antioxidant tea polyphenols is discussed. The analysis of acute gene expression change in skin after UV irradiation provides a starting point to investigate how the skin responds to genotoxic stress.
Collapse
Affiliation(s)
- Kaibin Yang
- Guangdong Key Laboratory for Genome Stability and Disease Prevention, Carson International Cancer Center, Department of Cell Biology and Medical Genetics, Shenzhen University School of Medicine, Shenzhen, China
| | - Shiting Song
- Guangdong Key Laboratory for Genome Stability and Disease Prevention, Carson International Cancer Center, Department of Cell Biology and Medical Genetics, Shenzhen University School of Medicine, Shenzhen, China
| | - Yafei Zhang
- Guangdong Key Laboratory for Genome Stability and Disease Prevention, Carson International Cancer Center, Department of Cell Biology and Medical Genetics, Shenzhen University School of Medicine, Shenzhen, China
| | - Siting Shen
- Guangdong Key Laboratory for Genome Stability and Disease Prevention, Carson International Cancer Center, Department of Cell Biology and Medical Genetics, Shenzhen University School of Medicine, Shenzhen, China
| | - Xingzhi Xu
- Guangdong Key Laboratory for Genome Stability and Disease Prevention, Carson International Cancer Center, Department of Cell Biology and Medical Genetics, Shenzhen University School of Medicine, Shenzhen, China
| | - Zhicao Yue
- Guangdong Key Laboratory for Genome Stability and Disease Prevention, Carson International Cancer Center, Department of Cell Biology and Medical Genetics, Shenzhen University School of Medicine, Shenzhen, China
| |
Collapse
|
6
|
Baraks G, Tseng R, Pan CH, Kasliwal S, Leiton CV, Shroyer KR, Escobar-Hoyos LF. Dissecting the Oncogenic Roles of Keratin 17 in the Hallmarks of Cancer. Cancer Res 2021; 82:1159-1166. [PMID: 34921015 PMCID: PMC9016724 DOI: 10.1158/0008-5472.can-21-2522] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 11/08/2021] [Accepted: 12/14/2021] [Indexed: 11/16/2022]
Affiliation(s)
- Gabriella Baraks
- Undergraduate Program in Biomedical Engineering, Stony Brook University, Stony Brook, New York
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Robert Tseng
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Chun-Hao Pan
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
- Molecular and Cellular Biology Graduate Program, Stony Brook University, New York
| | - Saumya Kasliwal
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Cindy V. Leiton
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
| | - Kenneth R. Shroyer
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
- Corresponding Authors: Kenneth R. Shroyer, Pathology, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794. Phone: 631-444-3000; E-mail: Kenneth.; and Luisa F. Escobar-Hoyos, 15 York Street PO Box 208040, New Haven, CT 06513. Phone: 203-737-2003; E-mail:
| | - Luisa F. Escobar-Hoyos
- Department of Pathology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York
- Department of Therapeutic Radiology and Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut
- Corresponding Authors: Kenneth R. Shroyer, Pathology, Stony Brook University, 101 Nicolls Rd, Stony Brook, NY 11794. Phone: 631-444-3000; E-mail: Kenneth.; and Luisa F. Escobar-Hoyos, 15 York Street PO Box 208040, New Haven, CT 06513. Phone: 203-737-2003; E-mail:
| |
Collapse
|
7
|
Piccini I, Brunken L, Chéret J, Ghatak S, Ramot Y, Alam M, Purba TS, Hardman J, Erdmann H, Jimenez F, Paus R, Bertolini M. PPARγ signaling protects hair follicle stem cells from chemotherapy-induced apoptosis and epithelial-mesenchymal transition. Br J Dermatol 2021; 186:129-141. [PMID: 34496034 DOI: 10.1111/bjd.20745] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Permanent chemotherapy-induced alopecia (pCIA), for which preventive interventions remain limited, can manifest with scarring. While the underlying pathomechanisms of pCIA are unclear, depletion of epithelial hair follicle (HF) stem cells (eHFSCs) is likely to play a role. OBJECTIVES To explore the hypothesis that eHFSCs undergo pathological epithelial-mesenchymal transition (EMT) besides apoptosis in pCIA, thus explaining the scarring phenotype. Furthermore, we tested whether a PPARγ modulator can prevent pCIA-associated pathomechanisms. METHODS Organ-cultured human scalp HFs were treated with the cyclophosphamide metabolite, 4-hydroperoxycyclophosphamide (4-HC). Additionally, HFs were pre-treated with the agnostic PPARγ modulator, N-Acetyl-GED-0507-34-Levo (NAGED), which we had previously shown to promote K15 expression and antagonize EMT in eHFSCs. RESULTS In accordance with anticipated hair bulb cytotoxicity, dystrophy and catagen induction, 4-HC promoted apoptosis along with increased p53 expression, DNA damage and pathological EMT in keratin 15+ (K15) bulge eHFSCs, as evidenced by decreased E-cadherin expression and the appearance of fibronectin- and vimentin-positive cells in the bulge. Pre-treatment with NAGED protected from 4-HC-induced hair bulb cytotoxicity/dystrophy, and halted apoptosis, p53 up-regulation, and EMT in the bulge, thereby significantly preventing the depletion of K15+ human eHFSCs ex vivo. CONCLUSIONS A cyclophosphamide metabolite alone suffices to damage and deplete human scalp eHFSCs by promoting apoptosis, DNA damage, and EMT ex vivo. Therefore, pCIA-therapeutic strategies need to target these pathological processes. Our data introduce the stimulation of PPARγ signaling as a novel intervention strategy for the prevention of pCIA, given the ability of NAGED to prevent chemotherapy-induced eHFSCs damage ex vivo.
Collapse
Affiliation(s)
- I Piccini
- Monasterium Laboratory, Münster, Germany
| | - L Brunken
- Monasterium Laboratory, Münster, Germany
| | - J Chéret
- Monasterium Laboratory, Münster, Germany.,Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - S Ghatak
- Monasterium Laboratory, Münster, Germany
| | - Y Ramot
- Department of Dermatology, Hadassah Medical Center, Hebrew University of Jerusalem, Jerusalem, Israel
| | - M Alam
- Monasterium Laboratory, Münster, Germany.,Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain.,Dept. of Dermatology & Venereology, Hamad Medical Corporation, Doha, Qatar.,Translational Research Institute, Academic Health System, Doha, Qatar
| | - T S Purba
- Centre for Dermatology Research, University of Manchester, NIHR Biomedical Research Centre, Manchester, UK
| | - J Hardman
- Centre for Dermatology Research, University of Manchester, NIHR Biomedical Research Centre, Manchester, UK.,St John's Institute of Dermatology, King's College London, London, United Kingdom
| | | | - F Jimenez
- Universidad Fernando Pessoa Canarias, Las Palmas de Gran Canaria, Spain.,Mediteknia Dermatology Clinic, Las Palmas de Gran Canaria, Spain
| | - R Paus
- Monasterium Laboratory, Münster, Germany.,Dr. Phillip Frost Department of Dermatology and Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, FL, USA.,Centre for Dermatology Research, University of Manchester, NIHR Biomedical Research Centre, Manchester, UK
| | | |
Collapse
|
8
|
Hiraganahalli Bhaskarmurthy D, Evan Prince S. Effect of Baricitinib on TPA-induced psoriasis like skin inflammation. Life Sci 2021; 279:119655. [PMID: 34043988 DOI: 10.1016/j.lfs.2021.119655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/10/2021] [Accepted: 05/14/2021] [Indexed: 10/21/2022]
Abstract
Psoriasis is a chronic inflammatory disorder of the skin and is characterized by hyper-dividing keratinocytes. This hyper-proliferation of keratinocytes is due to the high level of inflammatory cytokines. In this study, we evaluated the effect of topically applied Baricitinib, JAK1/2 inhibitor on chronic 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced psoriasis model in mice. To our knowledge, this is the first report evaluating the topical route of administration of Baricitinib in the context of psoriasis in vivo. TPA-induced inflammation was induced by the topical application of TPA in both ears. Thirty minutes before the application of TPA, the inner and outer surface of each ear was treated with Baricitinib for 6 days. Topical application of Baricitinib inhibited the expression of inflammation markers up-regulated by TPA. Besides, Baricitinib substantially reduced ear swelling, infiltration of leukocytes, the proliferation of epidermal cells, and angiogenesis of the dermal layer. The results suggest that Baricitinib significantly reduced phosphorylation of STAT3 and STAT1 levels in turn attenuating the downstream expression of inflammatory cytokines. Collectively, these results suggest that Baricitinib can be a potential therapeutic through topical route for psoriasis progresses.
Collapse
Affiliation(s)
- Deepak Hiraganahalli Bhaskarmurthy
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore-632014 India; Jubilant Biosys Ltd, Bangalore, Karnataka-560022 India
| | - Sabina Evan Prince
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore-632014 India.
| |
Collapse
|
9
|
Nair RR, Hsu J, Jacob JT, Pineda CM, Hobbs RP, Coulombe PA. A role for keratin 17 during DNA damage response and tumor initiation. Proc Natl Acad Sci U S A 2021; 118:e2020150118. [PMID: 33762306 PMCID: PMC8020757 DOI: 10.1073/pnas.2020150118] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
High levels of the intermediate filament protein keratin 17 (K17) are associated with poor prognoses for several human carcinomas. Studies in mouse models have shown that K17 expression is positively associated with growth, survival, and inflammation in skin and that lack of K17 delays onset of tumorigenesis. K17 occurs in the nucleus of human and mouse tumor keratinocytes where it impacts chromatin architecture, gene expression, and cell proliferation. We report here that K17 is induced following DNA damage and promotes keratinocyte survival. The presence of nuclear K17 is required at an early stage of the double-stranded break (DSB) arm of the DNA damage and repair (DDR) cascade, consistent with its ability to associate with key DDR effectors, including γ-H2A.X, 53BP1, and DNA-PKcs. Mice lacking K17 or with attenuated K17 nuclear import showed curtailed initiation in a two-step skin carcinogenesis paradigm. The impact of nuclear-localized K17 on DDR and cell survival provides a basis for the link between K17 induction and poor clinical outcomes for several human carcinomas.
Collapse
MESH Headings
- 9,10-Dimethyl-1,2-benzanthracene/administration & dosage
- 9,10-Dimethyl-1,2-benzanthracene/toxicity
- Active Transport, Cell Nucleus
- Animals
- Carcinogenesis/chemically induced
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Carcinoma/chemically induced
- Carcinoma/genetics
- Carcinoma/pathology
- Cell Nucleus/metabolism
- Cell Survival/genetics
- DNA Breaks, Double-Stranded/drug effects
- DNA Repair
- Female
- Gene Knockout Techniques
- HeLa Cells
- Humans
- Intravital Microscopy
- Keratin-17/genetics
- Keratin-17/metabolism
- Keratinocytes
- Keratins/genetics
- Keratins/metabolism
- Male
- Mice, Knockout
- Neoplasms, Experimental/chemically induced
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/pathology
- Time-Lapse Imaging
- Mice
Collapse
Affiliation(s)
- Raji R Nair
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Joshua Hsu
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| | - Justin T Jacob
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
| | - Christopher M Pineda
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109
| | - Ryan P Hobbs
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205;
| | - Pierre A Coulombe
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109;
- Department of Biochemistry and Molecular Biology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD 21205
- Department of Dermatology, University of Michigan, Ann Arbor, MI 48109
- Rogel Cancer Center, University of Michigan, Ann Arbor, MI 48109
| |
Collapse
|
10
|
Enaka M, Nakanishi M, Muragaki Y. The Gain-of-Function Mutation p53R248W Suppresses Cell Proliferation and Invasion of Oral Squamous Cell Carcinoma through the Down-Regulation of Keratin 17. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:555-566. [PMID: 33307039 DOI: 10.1016/j.ajpath.2020.11.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/20/2020] [Accepted: 11/30/2020] [Indexed: 11/28/2022]
Abstract
Keratin 17 (KRT17) expression promotes the proliferation and invasion of oral squamous cell carcinoma (OSCC), and mutations in TP53 have been reported in 65% to 85% of OSCC cases. We studied the correlation between KRT17 expression and TP53 mutants. Ca9-22 cells, which exhibit low KRT17 expression, carried mutant p53 (p53R248W) and p53R248W knockdown promoted KRT17 expression. p53R248W knockdown in Ca9-22 cells promoted migration and invasion activity. In contrast, in HSC3 cells, which have p53 nonsense mutations and exhibit high KRT17 expression, the overexpression of p53R248W decreased KRT17 expression, cell size, proliferation, and migration and invasion activities. In addition, p53R248W significantly suppressed MMP2 mRNA expression and enzyme activity. Moreover, s.c. and orthotopic xenografts were generated from p53R248W- or p53R248Q-expressing HSC3 cells. Tumors formed from p53R248W-expressing HSC3 cells grew more slowly and had a lower Ki-67 index than those derived from the control or p53R248Q-expressing HSC3 cells. Finally, the survival rate of the mice inoculated with p53R248W-expressing HSC3 cells was significantly higher than that of the control mice. These results indicate that the p53R248W mutant suppresses proliferation and invasion activity through the suppression of KRT17 expression. We propose that OSCC with p53R248W-expressing cells may be classified as a new OSCC type that has a good prognosis.
Collapse
Affiliation(s)
- Mayu Enaka
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Masako Nakanishi
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, Japan
| | - Yasuteru Muragaki
- Department of Pathology, Wakayama Medical University School of Medicine, Wakayama, Japan.
| |
Collapse
|
11
|
Guscetti F, Nassiri S, Beebe E, Rito Brandao I, Graf R, Markkanen E. Molecular homology between canine spontaneous oral squamous cell carcinomas and human head-and-neck squamous cell carcinomas reveals disease drivers and therapeutic vulnerabilities. Neoplasia 2020; 22:778-788. [PMID: 33142242 PMCID: PMC7642746 DOI: 10.1016/j.neo.2020.10.003] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/02/2020] [Accepted: 10/04/2020] [Indexed: 12/11/2022]
Abstract
Spontaneously occurring canine oral squamous cell carcinomas (COSCC) are viewed as a useful model for human head and neck squamous cell carcinomas (HNSCC). To date however, the molecular basis of COSCC remains poorly understood. To identify changes pertinent to cancer cells in COSCC, we specifically analyzed tumor cells and matched normal epithelium from clinical formalin-fixed paraffin-embedded specimens using laser-capture-microdissection coupled with RNA-sequencing (RNAseq). Our results identify strong contributions of epithelial-to-mesenchymal transition (EMT), classical tumor-promoting (such as E2F, KRAS, MYC, mTORC1, and TGFB1 signaling) and immune-related pathways in the tumor epithelium of COSCC. Comparative analyses of COSCC with 43 paired tumor/normal HNSCC from The Cancer Genome Atlas revealed a high homology in transcriptional reprogramming, and identified processes associated with cell cycle progression, immune processes, and loss of cellular differentiation as likely central drivers of the disease. Similar to HNSCC, our analyses suggested a ZEB2-driven partial EMT in COSCC and identified selective upregulation of KRT14 and KRT17 in COSCC. Beyond homology in transcriptional signatures, we also found therapeutic vulnerabilities strongly conserved between the species: these included increased expression of PD-L1 and CTLA-4, coinciding with EMT and revealing the potential for immune checkpoint therapies, and overexpression of CDK4/6 that sensitized COSCC to treatment with palbociclib. In summary, our data significantly extend the current knowledge of molecular aberrations in COSCC and underline the potential of spontaneous COSCC as a model for HNSCC to interrogate therapeutic vulnerabilities and support translation of novel therapies from bench to bedside.
Collapse
Affiliation(s)
- Franco Guscetti
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Sina Nassiri
- Bioinformatics Core Facility, SIB Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Erin Beebe
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Inês Rito Brandao
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Ramona Graf
- Institute of Veterinary Pathology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland
| | - Enni Markkanen
- Institute of Veterinary Pharmacology and Toxicology, Vetsuisse Faculty, University of Zürich, Zürich, Switzerland.
| |
Collapse
|
12
|
Yi L, Cui J, Hu N, Li L, Chen Y, Mu H, Yin J, Wei S, Gong Y, Wei Y, Liu B, Ding D. iTRAQ-Based Proteomic Profiling of Potential Biomarkers in Rat Serum for Uranium Tailing Suspension Intratracheal Instillation. J Proteome Res 2020; 20:995-1004. [PMID: 33151695 DOI: 10.1021/acs.jproteome.0c00766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Protection against low-dose ionizing radiation is of great significance. Uranium tailings are formed as a byproduct of uranium mining and a potential risk to organisms. In this study, we identified potential biomarkers associated with exposure to low-dose radiation from uranium tailings. We established a Wistar rat model of low dose rate irradiation by intratracheal instillation of a uranium tailing suspension. We observed pathological changes in the liver, lung, and kidney tissues of the rats. Using isobaric tags for relative and absolute quantification, we screened 17 common differentially expressed proteins in three dose groups. We chose alpha-1 antiproteinase (Serpina1), keratin 17 (Krt17), and aldehyde dehydrogenase (Aldh3a1) for further investigation. Our data showed that expression of Serpina1, Krt17, and Aldh3a1 had changed after the intratracheal instillation in rats, which may be potential biomarkers for uranium tailing low-dose irradiation. However, the underlying mechanisms require further investigation.
Collapse
Affiliation(s)
- Lan Yi
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Jian Cui
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Nan Hu
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China
| | - Linwei Li
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Yonglin Chen
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Hongxiang Mu
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Jie Yin
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Shuang Wei
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Yaqi Gong
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Yuanyun Wei
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Bang Liu
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China.,Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, Hunan 421001, P. R. China.,Hengyang Medical College, Institute of Cytology and Genetics, University of South China, Hengyang 421001, Hunan Province, P. R. China
| | - Dexin Ding
- The Hengyang Key Laboratory of Cellular Stress Biology, Key Discipline Laboratory for National Defense for Biotechnology in Uranium Mining and Hydrometallurgy, University of South China, Hengyang, Hunan 421001, P. R. China
| |
Collapse
|
13
|
CircRNA-1926 Promotes the Differentiation of Goat SHF Stem Cells into Hair Follicle Lineage by miR-148a/b-3p/ CDK19 Axis. Animals (Basel) 2020; 10:ani10091552. [PMID: 32887226 PMCID: PMC7552268 DOI: 10.3390/ani10091552] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 08/18/2020] [Accepted: 08/26/2020] [Indexed: 12/22/2022] Open
Abstract
Simple Summary Cashmere is the fiber derived from cashmere goats. Its textiles have been favored by consumers due to their typical features, like fine, light, softness, and comfort. Circular RNAs (circRNAs) are thought to play roles in cashmere growth of cashmere goats. CircRNA-1926 was previously identified in cashmere goats, but its functional roles are unclear. In this study, we firstly confirmed the expression of circRNA-1926 in secondary hair follicle bulge of cashmere goats with a significantly higher level at anagen than the counterpart of telogen. Next, we showed that circRNA-1926 promotes the differentiation of hair follicle stem cell into hair follicle lineage in cashmere goats. Mechanistically, we found that circRNA-1926 regulated the CDK19 expression via sponging miR-148a/b-3p. Our results have demonstrated that circRNA-1926 promotes the differentiation of secondary hair follicle stem cells into hair follicle lineages in cashmere goats through sponging miR-148a/b-3p to promote the expression of the CDK19 gene. The results from this study provided novel insight into the functional roles of circRNA-1926 in hair follicle regeneration and cashmere growth. Abstract Circular RNAs (CircRNAs) are a type of non-coding RNAs, which contain a covalently closed loop structure without 5′ to 3′ free ends. CircRNAs play essential roles in the regeneration of secondary hair follicle (SHF) and cashmere growth in goats. CircRNA-1926 was previously identified in SHF of cashmere goats, but its potential roles are unclear. In this study, we confirmed the expression of circRNA-1926 in SHF bulge of nine cashmere goats with a significantly higher level at anagen than that of telogen. Through the use of both overexpression and siRNA interference, we showed that circRNA-1926 promoted the differentiation of SHF stem cell into hair follicle lineage in cashmere goats which was evaluated via indictor genes Keratin 7 and Keratin 17. Using RNA pull-down, we found that circRNA-1926 bound with miR-148a/b-3p. Additionally, our data indicated that circRNA-1926 promoted the expression of the CDK19 gene. Using dual-luciferase reporter assays, it was revealed that circRNA-1926 positively regulated the CDK19 expression through miR-148a/b-3p. The results from this study demonstrated that circRNA-1926 contributes the differentiation of SHF stem cells into hair follicle lineages in cashmere goats via sponging miR-148a/b-3p to enhance CDK19 expression.
Collapse
|
14
|
Gao Q, Zhou G, Lin SJ, Paus R, Yue Z. How chemotherapy and radiotherapy damage the tissue: Comparative biology lessons from feather and hair models. Exp Dermatol 2018; 28:413-418. [PMID: 30457678 DOI: 10.1111/exd.13846] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 11/11/2018] [Accepted: 11/16/2018] [Indexed: 12/20/2022]
Abstract
Chemotherapy and radiotherapy are common modalities for cancer treatment. While targeting rapidly growing cancer cells, they also damage normal tissues and cause adverse effects. From the initial insult such as DNA double-strand break, production of reactive oxygen species (ROS) and a general stress response, there are complex regulatory mechanisms that control the actual tissue damage process. Besides apoptosis, a range of outcomes for the damaged cells are possible including cell cycle arrest, senescence, mitotic catastrophe, and inflammatory responses and fibrosis at the tissue level. Feather and hair are among the most actively proliferating (mini-)organs and are highly susceptible to both chemotherapy and radiotherapy damage, thus provide excellent, experimentally tractable model systems for dissecting how normal tissues respond to such injuries. Taking a comparative biology approach to investigate this has turned out to be particularly productive. Started in chicken feather and then extended to murine hair follicles, it was revealed that in addition to p53-mediated apoptosis, several other previously overlooked mechanisms are involved. Specifically, Shh, Wnt, mTOR, cytokine signalling and ROS-mediated degradation of adherens junctions have been implicated in the damage and/or reparative regeneration process. Moreover, we show here that inflammatory responses, which can be prominent upon histological examination of chemo- or radiotherapy-damaged hair follicle, may not be essential for the hair loss phenotype. These studies point to fundamental, evolutionarily conserved mechanisms in controlling tissue responses in vivo, and suggest novel strategies for the prevention and management of adverse effects that arise from chemo- or radiotherapy.
Collapse
Affiliation(s)
- QingXiang Gao
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
| | - GuiXuan Zhou
- Institute of Life Sciences, Fuzhou University, Fuzhou, Fujian, China
| | - Sung-Jan Lin
- Institute of Biomedical Engineering, National Taiwan University, Taipei, Taiwan.,Department of Dermatology, National Taiwan University Hospital and College of Medicine, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Ralf Paus
- Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida.,Centre for Dermatology Research, University of Manchester, Manchester, UK
| | - ZhiCao Yue
- Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida
| |
Collapse
|
15
|
Yang L, Zhang S, Wang G. Keratin 17 in disease pathogenesis: from cancer to dermatoses. J Pathol 2018; 247:158-165. [PMID: 30306595 DOI: 10.1002/path.5178] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 09/17/2018] [Accepted: 10/02/2018] [Indexed: 12/16/2022]
Abstract
Keratin 17 (K17) is a type I intermediate filament mainly expressed in the basal cells of epithelia. As a multifaceted cytoskeletal protein, K17 regulates a myriad of biological processes, including cell proliferation and growth, skin inflammation and hair follicle cycling. Aberrant overexpression of K17 is found in various diseases ranging from psoriasis to malignancies such as breast, cervical, oral squamous and gastric carcinomas. Moreover, genetic mutation in KRT17 is related to tissue-specific diseases, represented by steatocystoma multiplex and pachyonychia congenita. In this review, we summarize our findings concerning the regulatory mechanisms of K17 overexpression in psoriasis and compare them to the literature relating to other diseases. We discuss data that proinflammatory cytokines, including interleukin-17 (IL-17), IL-22, interferon-gamma (IFN-γ), transforming growth factor-beta (TGF-β) and transcription factors glioma-associated oncogene homolog 1/2 (Gli1/2), Nrf2 and p53 can regulate K17 by transcriptional and translational control. Moreover, post-translational modification, including phosphorylation and ubiquitination, is involved in the regulation of K17 stability and biological functions. We therefore review the current understanding of the K17 regulatory mechanism and its pathogenic role in diseases from dermatoses to cancer. Prospects for anti-K17 therapy in diagnosis, prognosis and disease treatment are also discussed. Copyright © 2018 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Luting Yang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| | - Shaolong Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, PR, China
| |
Collapse
|
16
|
Wang M, Gao Q, Teng X, Pan M, Lin T, Zhou G, Xu B, Yue Z. Ionizing radiation, but not ultraviolet radiation, induces mitotic catastrophe in mouse epidermal keratinocytes with aberrant cell cycle checkpoints. Exp Dermatol 2018; 27:791-794. [PMID: 29672918 DOI: 10.1111/exd.13665] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2018] [Indexed: 01/03/2023]
Abstract
Ultraviolet radiation (UVR) and ionizing radiation (IR) are common genotoxic stresses that damage human skin, although the specific damages to the genomic DNA are different. Here, we show that in the mouse glabrous skin, both UVR and IR induce DNA damage, cell cycle arrest, and condensed cell nuclei. However, only IR induces mitotic catastrophe (MC) in the epidermis. This is because UVR induces a complete blockage of pRB phosphorylation and cell cycle arrest in the G1 phase, whereas pRB phosphorylation remains positive in a significant portion of the epidermal keratinocytes following IR exposure. Furthermore, Cyclin B1 expression is significantly downregulated only by IR but not UVR. Finally, there are more MC cells in the epidermis of p53-/- mice after IR exposure as compared to wild-type mice. Our results suggest that although both IR and UVR are genotoxic, they show distinct impacts on the cell cycle machinery and thus damage the epidermal keratinocytes via different mechanisms.
Collapse
Affiliation(s)
- Ming Wang
- Institute of Life Sciences, Fuzhou University, Fuzhou, China.,College of Chemistry, Fuzhou University, Fuzhou, China
| | - QingXiang Gao
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - Xu Teng
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - MeiPing Pan
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - TianMiao Lin
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - GuiXuan Zhou
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| | - BenHua Xu
- Department of Radiation Oncology, Union Hospital Affiliated with Fujian Medical University, Fuzhou, China
| | - ZhiCao Yue
- Institute of Life Sciences, Fuzhou University, Fuzhou, China
| |
Collapse
|
17
|
Zhuang Y, Han C, Li B, Jin L, Dang E, Fang H, Qiao H, Wang G. NB-UVB irradiation downregulates keratin-17 expression in keratinocytes by inhibiting the ERK1/2 and STAT3 signaling pathways. Arch Dermatol Res 2018; 310:147-156. [PMID: 29349514 DOI: 10.1007/s00403-018-1812-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Revised: 01/03/2018] [Accepted: 01/12/2018] [Indexed: 12/21/2022]
Abstract
Keratin-17 (K17) is a cytoskeletal protein produced by keratinocytes (KCs), which is overexpressed in psoriasis and may play a pivotal role in its pathogenesis. Narrow-band ultraviolet B (NB-UVB) irradiation is used as a general treatment for psoriasis, although its impact on K17 expression has yet to be determined. In this study, we aimed to investigate the effect of NB-UVB irradiation on K17 expression and its signaling pathways. After exposure to NB-UVB irradiation, immortalized human keratinocytes (HaCaT cells) were analyzed by flow cytometry, CCK-8 assays and transmission electron microscopy to examine proliferation. Meanwhile, K17 expression in primary human epithelial keratinocytes was detected by quantitative real-time polymerase chain reaction (qRT-PCR), western blot analysis and immunofluorescence. HaCaT cells pre-incubated with PD-98059 and piceatannol were subjected to western blot analysis to examine ERK1/2 and STAT3 phosphorylation. The ears of mice treated with imiquimod (IMQ) and irradiated by NB-UVB were taken to examine K17 expression by qRT-PCR, western blot analysis, and immunofluorescence. Our results showed that 400 mJ/cm2 of NB-UVB irradiation was the maximum tolerable dose for HaCaT cells and could cause inhibited HaCaT cell proliferation and moderate increase of the early apoptosis. Furthermore, NB-UVB irradiation could downregulate K17 expression by inhibiting the ERK1/2 and STAT3 signaling pathways. In experiments conducted in vivo, NB-UVB irradiation with doses of MED or higher could eliminate the IMQ-induced psoriasis-like dermatitis and inhibit K17 expression. These results indicated that NB-UVB irradiation may eliminate chronic psoriatic plaques by suppressing K17 expression via the ERK1/2 and STAT3 signaling pathways.
Collapse
Affiliation(s)
- Yuchen Zhuang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Changxu Han
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Bing Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Liang Jin
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Erle Dang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hui Fang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Hongjiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
18
|
Jang H, Myung H, Lee J, Myung JK, Jang WS, Lee SJ, Bae CH, Kim H, Park S, Shim S. Impaired Skin Barrier Due to Sebaceous Gland Atrophy in the Latent Stage of Radiation-Induced Skin Injury: Application of Non-Invasive Diagnostic Methods. Int J Mol Sci 2018; 19:ijms19010185. [PMID: 29316698 PMCID: PMC5796134 DOI: 10.3390/ijms19010185] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 01/01/2018] [Accepted: 01/03/2018] [Indexed: 12/18/2022] Open
Abstract
Radiation-induced skin injury can take the form of serious cutaneous damage and have specific characteristics. Asymptomatic periods are classified as the latent stage. The skin barrier plays a critical role in the modulation of skin permeability and hydration and protects the body against a harsh external environment. However, an analysis on skin barrier dysfunction against radiation exposure in the latent stage has not been conducted. Thus, we investigated whether the skin barrier is impaired by irradiation in the latent stage and aimed to identify the molecules involved in skin barrier dysfunction. We analyzed skin barrier function and its components in SKH1 mice that received 20 and 40 Gy local irradiation. Increased transepidermal water loss and skin pH were observed in the latent stage of the irradiated skin. Skin barrier components, such as structural proteins and lipid synthesis enzymes in keratinocyte, increased in the irradiated group. Interestingly, we noted sebaceous gland atrophy and increased serine protease and inflammatory cytokines in the irradiated skin during the latent period. This finding indicates that the main factor of skin barrier dysfunction in the latent stage of radiation-induced skin injury is sebaceous gland deficiency, which could be an intervention target for skin barrier impairment.
Collapse
Affiliation(s)
- Hyosun Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Hyunwook Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Janet Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Jae Kyung Myung
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Won-Suk Jang
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Sun-Joo Lee
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Chang-Hwan Bae
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Hyewon Kim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Sunhoo Park
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
- Department of Pathology, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| | - Sehwan Shim
- Laboratory of Radiation Exposure & Therapeutics, National Radiation Emergency Medical Center, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Korea.
| |
Collapse
|
19
|
E-Cadherin–Mediated Cell Contact Controls the Epidermal Damage Response in Radiation Dermatitis. J Invest Dermatol 2017; 137:1731-1739. [DOI: 10.1016/j.jid.2017.03.036] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 03/23/2017] [Accepted: 03/30/2017] [Indexed: 11/23/2022]
|
20
|
Genome-wide analysis suggests a differential microRNA signature associated with normal and diabetic human corneal limbus. Sci Rep 2017; 7:3448. [PMID: 28615632 PMCID: PMC5471258 DOI: 10.1038/s41598-017-03449-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/27/2017] [Indexed: 12/19/2022] Open
Abstract
Small non-coding RNAs, in particular microRNAs (miRNAs), regulate fine-tuning of gene expression and can impact a wide range of biological processes. However, their roles in normal and diseased limbal epithelial stem cells (LESC) remain unknown. Using deep sequencing analysis, we investigated miRNA expression profiles in central and limbal regions of normal and diabetic human corneas. We identified differentially expressed miRNAs in limbus vs. central cornea in normal and diabetic (DM) corneas including both type 1 (T1DM/IDDM) and type 2 (T2DM/NIDDM) diabetes. Some miRNAs such as miR-10b that was upregulated in limbus vs. central cornea and in diabetic vs. normal limbus also showed significant increase in T1DM vs. T2DM limbus. Overexpression of miR-10b increased Ki-67 staining in human organ-cultured corneas and proliferation rate in cultured corneal epithelial cells. MiR-10b transfected human organ-cultured corneas showed downregulation of PAX6 and DKK1 and upregulation of keratin 17 protein expression levels. In summary, we report for the first time differential miRNA signatures of T1DM and T2DM corneal limbus harboring LESC and show that miR-10b could be involved in the LESC maintenance and/or their early differentiation. Furthermore, miR-10b upregulation may be an important mechanism of corneal diabetic alterations especially in the T1DM patients.
Collapse
|
21
|
Abstract
Chemo- and radiation therapy are the main modalities for cancer treatment. A major limiting factor is their toxicity to normal tissue, thus reducing the dose and duration of the therapy. The hair follicle, gastrointestinal tract, and hematopoietic system are among the target organs that often show side effects in cancer therapy . Although these organs are highly mitotic in common, the molecular mechanism of the damage remains unclear. The feather follicle is a fast-growing mini-organ, which allows observation and manipulation on each follicle individually. As a model system, the feather follicle is advantageous because of the following reasons: (1) its complex structure is regulated by a set of evolutionarily conserved molecular pathways, thus facilitating the effort to dissect the specific signaling events involved; (2) its morphology allows the continuity of normal-perturbed-normal structure in a single feather, thus "recording" the damaging effect of chemo- and radiation therapy; (3) further histological and molecular analysis of the damage response can be performed on each plucked feather; thus, it is not necessary to sacrifice the experimental animal. Here, we describe methods of applying the feather model to study the molecular mechanism of chemo- and radiation therapy-induced tissue damage.
Collapse
Affiliation(s)
- Zhicao Yue
- Institute of Life Sciences, Fuzhou University, #2 Xue Yuan Road, Fuzhou, Fujian, 350108, China.
| | - Benhua Xu
- Department of Radiation Oncology, The Union Hospital Affiliated with Fujian Medical University, Fuzhou, Fujian, 350000, China
| |
Collapse
|
22
|
Genetic susceptibility to cutaneous radiation injury. Arch Dermatol Res 2016; 309:1-10. [PMID: 27878387 DOI: 10.1007/s00403-016-1702-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 09/30/2016] [Accepted: 11/16/2016] [Indexed: 12/31/2022]
Abstract
The use of ionizing radiation is critical to cancer treatment and fluoroscopic procedures. However, despite efforts to minimize total radiation dose, many patients experience toxic cutaneous side-effects of ionizing radiation, ranging from mild erythema to subcutaneous fibrosis, telangiectasia formation, and ulceration. Extent of injury is highly variable among patients. Studying the genetic determinants of radiation injury can help develop protocols to reduce radiation toxicity, as well as drive research into effective modulators of the genes and gene products associated with radiation injury. Many studies in the past two decades have identified single-nucleotide polymorphisms that may be associated with susceptibility to cutaneous radiation injury, such as those in genes related to the following cellular responses to ionizing radiation: inflammation, DNA repair, oxidation and stress response, and cell-cycle and apoptosis. This review summarizes the current literature on potential major genes and polymorphisms, in the previously described damage response pathways, that are involved in susceptibility to cutaneous radiation injury. Potential pitfalls of current research and further avenues of discovery will be explored.
Collapse
|