1
|
Chen W, Liang J, He S, Liang Q, Tian W, Lu A, Li D, Huang Z, Wu G. She-Chuang-Si-Wu-Tang Alleviates Inflammation and Itching Symptoms in a Psoriasis Mouse Model by Regulating the Th17/IL-17 Axis via the STAT3/MAPK Pathways. J Inflamm Res 2024; 17:5957-5975. [PMID: 39247836 PMCID: PMC11380483 DOI: 10.2147/jir.s472417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 08/28/2024] [Indexed: 09/10/2024] Open
Abstract
Purpose Psoriasis is an immune-related disorder characterized by silver scales, epidermis thickness, and itching. She-Chuang-Si-Wu-Tang (SSWT), a traditional Chinese medicine decoction, has been used clinically for 400 years. Although it benefits psoriasis treatment, the mechanism of action is still unclear. This study explores SSWT's molecular mechanism in treating psoriasis through network pharmacology analysis and experiments. Methods We identified relevant SSWT and psoriasis targets using network pharmacology and conducted SSWT quality control with high-performance liquid chromatography (HPLC). A mouse model of psoriasis was established using imiquimod (IMQ), with the drug administered continuously for seven days, spanning an eight-day period. During the experiment, we observed spontaneous scratching behaviors and assessed the Psoriasis Area and Severity Index (PASI) scores. At the conclusion of the experiment, we examined skin tissue pathology under an optical microscope and measured epidermal thickness. Additionally, we used enzyme-linked immunosorbent assay (ELISA) and quantitative reverse transcription polymerase chain reaction (qRT-PCR) to measure interleukin (IL)-23, IL-17A, IL-17F, and interferon (IFN)-γ levels in the mice's serum and their mRNA expression in the skin. Western blot analysis was conducted to assess protein levels related to signaling pathways. Results Results indicate that SSWT may target IL-17 signaling pathways and T helper (Th) 17 cell differentiation, as predicted by network pharmacology. SSWT significantly improved the PASI and Baker scores, reduced epidermal thickness, and decreased spontaneous scratching in IMQ-induced mice. Additionally, SSWT treatment significantly lowered the concentrations of inflammatory factors in the serum and skin lesions, as well as mRNA expression levels, compared to the IMQ group. Furthermore, SSWT significantly inhibited the phosphorylation of both the signal transducer and activator of transcription 3 (STAT3) and mitogen-activated protein kinase (MAPK) pathways. Conclusion In summary, this study unveiled the potential anti-psoriatic mechanism of SSWT, offering new evidence for its clinical application.
Collapse
Affiliation(s)
- Weixiong Chen
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People’s Republic of China
| | - Jianqiang Liang
- Department of Dermatology, the First People’s Hospital of Yulin, Yulin, Guangxi, 537000, People’s Republic of China
| | - Shuang He
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- Key Laboratory for Complementary and Alternative Medicine Experimental Animal Models of Guangxi, Nanning, Guangxi, 530299, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Qingsong Liang
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- Key Laboratory for Complementary and Alternative Medicine Experimental Animal Models of Guangxi, Nanning, Guangxi, 530299, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Wenting Tian
- College of Pharmacy, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Aobo Lu
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Demin Li
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Zhicheng Huang
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| | - Guanyi Wu
- College of Basic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, 530299, People’s Republic of China
- Key Laboratory for Complementary and Alternative Medicine Experimental Animal Models of Guangxi, Nanning, Guangxi, 530299, People’s Republic of China
- Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, Guangxi, 530299, People’s Republic of China
| |
Collapse
|
2
|
Liu B, Li A, Xu J, Cui Y. Single-Cell Transcriptional Analysis Deciphers the Inflammatory Response of Skin-Resident Stromal Cells. Front Surg 2022; 9:935107. [PMID: 35774389 PMCID: PMC9237500 DOI: 10.3389/fsurg.2022.935107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 05/23/2022] [Indexed: 11/24/2022] Open
Abstract
The skin is the outermost barrier of the body. It has developed a sophisticated system against the ever-changing environment. The application of single-cell technologies has revolutionized dermatology research and unraveled the changes and interactions across skin resident cells in the healthy and inflamed skin. Single-cell technologies have revealed the critical roles of stromal cells in an inflammatory response and explained a series of plausible previous findings concerning skin immunity. Here, we summarized the functional diversity of skin stromal cells defined by single-cell analyses and how these cells orchestrated events leading to inflammatory diseases, including atopic dermatitis, psoriasis, vitiligo, and systemic lupus erythematosus.
Collapse
Affiliation(s)
- Baoyi Liu
- Department of Dermatology, China–Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ang Li
- Department of Dermatology, China–Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingkai Xu
- Department of Dermatology, China–Japan Friendship Hospital, Beijing, China
| | - Yong Cui
- Department of Dermatology, China–Japan Friendship Hospital, Beijing, China
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Correspondence: Yong Cui
| |
Collapse
|
3
|
Vermeer MCSC, Andrei D, Kramer D, Nijenhuis AM, Hoedemaekers YM, Westers H, Jongbloed JDH, Pas HH, van den Berg MP, Silljé HHW, van der Meer P, Bolling MC. Functional investigation of two simultaneous or separately segregating DSP variants within a single family support the theory of a dose-dependent disease severity. Exp Dermatol 2022; 31:970-979. [PMID: 35325485 PMCID: PMC9322008 DOI: 10.1111/exd.14571] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 02/28/2022] [Accepted: 03/22/2022] [Indexed: 11/30/2022]
Abstract
Desmoplakin (DP) is an important component of desmosomes, essential in cell–cell connecting structures in stress‐bearing tissues. Over the years, many hundreds of pathogenic variants in DSP have been associated with different cutaneous and cardiac phenotypes or a combination, known as a cardiocutaneous syndrome. Of less than 5% of the reported DSP variants, the effect on the protein has been investigated. Here, we describe and have performed RNA, protein and tissue analysis in a large family where DSPc.273+5G>A/c.6687delA segregated with palmoplantar keratoderma (PPK), woolly hair and lethal cardiomyopathy, while DSPWT/c.6687delA segregated with PPK and milder cardiomyopathy. hiPSC‐derived cardiomyocytes and primary keratinocytes from carriers were obtained for analysis. Unlike the previously reported nonsense variants in the last exon of DSP that bypassed the nonsense‐mediated mRNA surveillance system leading to protein truncation, variant c.6687delA was shown to cause the loss of protein expression. Patients carrying both variants and having a considerably more severe phenotype were shown to have 70% DP protein reduction, while patients carrying only c.6687delA had 50% protein reduction and a milder phenotype. The analysis of RNA from patient cells did not show any splicing effect of the c.273+5G>A variant. However, a minigene splicing assay clearly showed alternative spliced transcripts originating from this variant. This study shows the importance of RNA and protein analyses to pinpoint the exact effect of DSP variants instead of solely relying on predictions. In addition, the particular pattern of inheritance, with simultaneous or separately segregating DSP variants within the same family, strongly supports the theory of a dose‐dependent disease severity.
Collapse
Affiliation(s)
- Mathilde C S C Vermeer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Daniela Andrei
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Duco Kramer
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Albertine M Nijenhuis
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yvonne M Hoedemaekers
- Department of Genetics, Radboud University Nijmegen, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Helga Westers
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan D H Jongbloed
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hendri H Pas
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maarten P van den Berg
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Herman H W Silljé
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Maria C Bolling
- Department of Dermatology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
4
|
Maik-Rachline G, Wortzel I, Seger R. Alternative Splicing of MAPKs in the Regulation of Signaling Specificity. Cells 2021; 10:cells10123466. [PMID: 34943973 PMCID: PMC8699841 DOI: 10.3390/cells10123466] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) cascades transmit signals from extracellular stimuli to a variety of distinct cellular processes. The MAPKKs in each cascade specifically phosphorylate and activate their cognate MAPKs, indicating that this step funnels various signals into a seemingly linear pathway. Still, the effects of these cascades vary significantly, depending on the identity of the extracellular signals, which gives rise to proper outcomes. Therefore, it is clear that the specificity of the signals transmitted through the cascades is tightly regulated in order to secure the desired cell fate. Indeed, many regulatory components or processes that extend the specificity of the cascades have been identified. Here, we focus on a less discussed mechanism, that is, the role of distinct components in each tier of the cascade in extending the signaling specificity. We cover the role of distinct genes, and the alternatively spliced isoforms of MAPKKs and MAPKs, in the signaling specificity. The alternatively spliced MEK1b and ERK1c, which form an independent signaling route, are used as the main example. Unlike MEK1/2 and ERK1/2, this route’s functions are limited, including mainly the regulation of mitotic Golgi fragmentation. The unique roles of the alternatively spliced isoforms indicate that these components play an essential role in determining the proper cell fate in response to distinct stimulations.
Collapse
|
5
|
Rizaldy D, Toriyama M, Kato H, Fukui R, Fujita F, Nakamura M, Okada F, Morita A, Ishii KJ. Increase in primary cilia in the epidermis of patients with atopic dermatitis and psoriasis. Exp Dermatol 2021; 30:792-803. [PMID: 33455013 DOI: 10.1111/exd.14285] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 01/09/2021] [Accepted: 01/11/2021] [Indexed: 12/19/2022]
Abstract
Primary cilia influence cell activity, and thus have a unique role in maintaining cell proliferation and differentiation. In atopic dermatitis (AD) and psoriasis, areas of skin inflammation exhibit dysregulated keratinocyte homeostasis. The role of primary cilia in these conditions remains unclear. The objectives of this study is to elucidate the incidence of primary cilia in skin inflammation and the potential mechanism underlying the dysregulation of keratinocytes. Primary cilia were observed using immunofluorescence staining. Normal skin samples were compared with skin samples from patients with AD or psoriasis in terms of cilia numbers and length. The effect of cytokine stimulation on ciliogenesis in keratinocytes was analysed using a primary keratinocyte culture. IFT88, an important ciliary intraflagellar protein, was blocked in Th2 and Th17 cytokines-stimulated keratinocytes. These effects were analysed with quantitative polymerase chain reaction and Western blot. Significant increases in ciliated cells were observed in AD and psoriasis skin samples compared with normal skin samples. The stimulation of keratinocytes using Th2 and Th17 cytokines modulated the formation of primary cilia. The amount of IFT88 in the primary cilia associated with the phosphorylation of JNK, but not p38, in keratinocytes stimulated with interleukin-13, 17A and 22. An increase of ciliated cells in the epidermis may impair keratinocyte differentiation under stress conditions caused by inflammation in both AD and psoriasis patients.
Collapse
Affiliation(s)
- Defri Rizaldy
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Department of Pharmaceutical Biology, School of Pharmacy, Bandung Institute of Technology, Bandung, Indonesia
| | - Manami Toriyama
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Laboratory for Molecular Signal Transduction, Graduate School of Science and Technology, Nara Institute of Science and Technology, Nara, Japan
| | - Hiroko Kato
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Runa Fukui
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan
| | - Fumitaka Fujita
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Mandom Corporation, Osaka, Japan
| | - Motoki Nakamura
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Department of Geriatric and Environmental Dermatology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Fumihiro Okada
- Laboratory of Advanced Cosmetic Science, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan.,Mandom Corporation, Osaka, Japan
| | - Akimichi Morita
- Department of Geriatric and Environmental Dermatology, Graduate School of Medical Sciences, Nagoya City University, Nagoya, Japan
| | - Ken J Ishii
- Laboratory of Mock up Vaccine, Center for Vaccine and Adjuvant Research (CVAR), National Institutes of Biomedical Innovation, Health and Nutrition (NIBIOHN), Osaka, Japan.,Laboratory of Vaccine Science, WPI Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan.,Division of Vaccine Science, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
6
|
De Gregorio V, Urciuolo F, Netti PA, Imparato G. In Vitro Organotypic Systems to Model Tumor Microenvironment in Human Papillomavirus (HPV)-Related Cancers. Cancers (Basel) 2020; 12:E1150. [PMID: 32375253 PMCID: PMC7281263 DOI: 10.3390/cancers12051150] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Revised: 04/30/2020] [Accepted: 05/01/2020] [Indexed: 12/16/2022] Open
Abstract
Despite the well-known role of chronic human papillomavirus (HPV) infections in causing tumors (i.e., all cervical cancers and other human malignancies from the mucosal squamous epithelia, including anogenital and oropharyngeal cavity), its persistence is not sufficient for cancer development. Other co-factors contribute to the carcinogenesis process. Recently, the critical role of the underlying stroma during the HPV life cycle and HPV-induced disease have been investigated. The tumor stroma is a key component of the tumor microenvironment (TME), which is a specialized entity. The TME is dynamic, interactive, and constantly changing-able to trigger, support, and drive tumor initiation, progression, and metastasis. In previous years, in vitro organotypic raft cultures and in vivo genetically engineered mouse models have provided researchers with important information on the interactions between HPVs and the epithelium. Further development for an in-depth understanding of the interaction between HPV-infected tissue and the surrounding microenvironment is strongly required. In this review, we critically describe the HPV-related cancers modeled in vitro from the simplified 'raft culture' to complex three-dimensional (3D) organotypic models, focusing on HPV-associated cervical cancer disease platforms. In addition, we review the latest knowledge in the field of in vitro culture systems of HPV-associated malignancies of other mucosal squamous epithelia (anogenital and oropharynx), as well as rare cutaneous non-melanoma associated cancer.
Collapse
Affiliation(s)
- Vincenza De Gregorio
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| | - Francesco Urciuolo
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80125 Naples, Italy; (F.U.); (P.A.N.)
| | - Paolo Antonio Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, 80125 Naples, Italy; (F.U.); (P.A.N.)
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, 80125 Naples, Italy
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI) University of Naples Federico II, 80125 Naples, Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB, Istituto Italiano di Tecnologia, 80125 Naples, Italy
| |
Collapse
|
7
|
Solé-Boldo L, Raddatz G, Schütz S, Mallm JP, Rippe K, Lonsdorf AS, Rodríguez-Paredes M, Lyko F. Single-cell transcriptomes of the human skin reveal age-related loss of fibroblast priming. Commun Biol 2020; 3:188. [PMID: 32327715 PMCID: PMC7181753 DOI: 10.1038/s42003-020-0922-4] [Citation(s) in RCA: 231] [Impact Index Per Article: 57.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 03/31/2020] [Indexed: 12/21/2022] Open
Abstract
Fibroblasts are an essential cell population for human skin architecture and function. While fibroblast heterogeneity is well established, this phenomenon has not been analyzed systematically yet. We have used single-cell RNA sequencing to analyze the transcriptomes of more than 5,000 fibroblasts from a sun-protected area in healthy human donors. Our results define four main subpopulations that can be spatially localized and show differential secretory, mesenchymal and pro-inflammatory functional annotations. Importantly, we found that this fibroblast 'priming' becomes reduced with age. We also show that aging causes a substantial reduction in the predicted interactions between dermal fibroblasts and other skin cells, including undifferentiated keratinocytes at the dermal-epidermal junction. Our work thus provides evidence for a functional specialization of human dermal fibroblasts and identifies the partial loss of cellular identity as an important age-related change in the human dermis. These findings have important implications for understanding human skin aging and its associated phenotypes.
Collapse
Affiliation(s)
- Llorenç Solé-Boldo
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Günter Raddatz
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Sabrina Schütz
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany
| | - Jan-Philipp Mallm
- Division of Chromatin Networks, German Cancer Research Center and Bioquant, 69120, Heidelberg, Germany
| | - Karsten Rippe
- Division of Chromatin Networks, German Cancer Research Center and Bioquant, 69120, Heidelberg, Germany
| | - Anke S Lonsdorf
- Department of Dermatology, University Hospital, Ruprecht-Karls University of Heidelberg, 69120, Heidelberg, Germany
| | - Manuel Rodríguez-Paredes
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany.
| | - Frank Lyko
- Division of Epigenetics, DKFZ-ZMBH Alliance, German Cancer Research Center, 69120, Heidelberg, Germany.
| |
Collapse
|
8
|
Xu F, Xu J, Xiong X, Deng Y. Salidroside inhibits MAPK, NF-κB, and STAT3 pathways in psoriasis-associated oxidative stress via SIRT1 activation. Redox Rep 2020; 24:70-74. [PMID: 31495284 PMCID: PMC6748574 DOI: 10.1080/13510002.2019.1658377] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Objectives: To unveil the role of SIRT1 in limiting oxidative stress in psoriasis and to further discuss the therapeutic prospects of salidroside in psoriasis. Methods: Literature from 2002 to 2019 was searched with “psoriasis”, “oxidative stress”, “SIRT1”, “salidroside” as the key words. Then, Oxidative stress in psoriasis and the role of SIRT1 were summarized and the potential role of salidroside in the disease was speculated. Results: Oxidative stress might contribute to the pathogenesis of psoriasis. High levels of ROS produced during oxidative stress lead to the release of inflammatory mediators, that, in turn, induce angiogenesis and excessive proliferation of keratinocytes. SIRT1 is a member of the sirtuin family, of which the activation lead to the inhibition of such oxidative stress signaling pathways MAPK, NF-κB, and STAT3, down-regulation of inflammatory factors, suppression of inflammation and keratinocyte hyperproliferation, and inhibition of angiogenesis. Salidroside, the main ingredient of Rhodiola, is known to exert antioxidant roles, which has been attributed to SIRT1 activation. Conclusion: Salidroside might inhibit oxidative stress singling pathways via SIRT1 activation, and could be as an ideal candidate for management of psoriasis.
Collapse
Affiliation(s)
- Fengli Xu
- Department of Dermatology, The First Affiliated Hospital of Southwest Medical University , Luzhou , People's Republic of China
| | - Jixiang Xu
- Department of Dermatology, The First Affiliated Hospital of Southwest Medical University , Luzhou , People's Republic of China
| | - Xia Xiong
- Department of Dermatology, The First Affiliated Hospital of Southwest Medical University , Luzhou , People's Republic of China
| | - Yongqiong Deng
- Department of Dermatology, The First Affiliated Hospital of Southwest Medical University , Luzhou , People's Republic of China
| |
Collapse
|
9
|
Bal-Öztürk A, Miccoli B, Avci-Adali M, Mogtader F, Sharifi F, Çeçen B, Yaşayan G, Braeken D, Alarcin E. Current Strategies and Future Perspectives of Skin-on-a-Chip Platforms: Innovations, Technical Challenges and Commercial Outlook. Curr Pharm Des 2019; 24:5437-5457. [PMID: 30727878 DOI: 10.2174/1381612825666190206195304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 01/09/2023]
Abstract
The skin is the largest and most exposed organ in the human body. Not only it is involved in numerous biological processes essential for life but also it represents a significant endpoint for the application of pharmaceuticals. The area of in vitro skin tissue engineering has been progressing extensively in recent years. Advanced in vitro human skin models strongly impact the discovery of new drugs thanks to the enhanced screening efficiency and reliability. Nowadays, animal models are largely employed at the preclinical stage of new pharmaceutical compounds development for both risk assessment evaluation and pharmacokinetic studies. On the other hand, animal models often insufficiently foresee the human reaction due to the variations in skin immunity and physiology. Skin-on-chips devices offer innovative and state-of-the-art platforms essential to overcome these limitations. In the present review, we focus on the contribution of skin-on-chip platforms in fundamental research and applied medical research. In addition, we also highlighted the technical and practical difficulties that must be overcome to enhance skin-on-chip platforms, e.g. embedding electrical measurements, for improved modeling of human diseases as well as of new drug discovery and development.
Collapse
Affiliation(s)
- Ayça Bal-Öztürk
- Department of Analytical Chemistry, Faculty of Pharmacy, İstinye University, 34010, Zeytinburnu, Istanbul, Turkey,Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey
| | - Beatrice Miccoli
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium,Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Ferzaneh Mogtader
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey,NanoBMT, Cyberpark, Bilkent 06800, Ankara, Turkey
| | - Fatemeh Sharifi
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Berivan Çeçen
- Biomechanics Department, Institute of Health Science, Dokuz Eylul University, 35340, Inciraltı, Izmir, Turkey; Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Gökçen Yaşayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34668, Haydarpaşa, Istanbul, Turkey
| | - Dries Braeken
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium
| | - Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34668, Haydarpaşa, Istanbul, Turkey
| |
Collapse
|
10
|
Suppression of Stromal Interferon Signaling by Human Papillomavirus 16. J Virol 2019; 93:JVI.00458-19. [PMID: 31292244 DOI: 10.1128/jvi.00458-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 06/29/2019] [Indexed: 01/09/2023] Open
Abstract
Human papillomaviruses (HPVs) infect squamous epithelia and cause several important cancers. Immune evasion is critical for viral persistence. Fibroblasts in the stromal microenvironment provide growth signals and cytokines that are required for proper epithelial differentiation, maintenance, and immune responses and are critical in the development of many cancers. In this study, we examined the role of epithelial-stromal interactions in the HPV16 life cycle using organotypic (raft) cultures as a model. Rafts were created using uninfected human foreskin keratinocytes (HFKs) and HFKs containing either wild-type HPV16 or HPV16 with a stop mutation to prevent the expression of the viral oncogene E5. Microarray analysis revealed significant changes in gene expression patterns in the stroma in response to HPV16, some of which were E5 dependent. Interferon (IFN)-stimulated genes (ISGs) and extracellular matrix remodeling genes were suppressed, the most prominent pathways affected. STAT1, IFNAR1, IRF3, and IRF7 were knocked down in stromal fibroblasts using lentiviral short hairpin RNA (shRNA) transduction. HPV late gene expression and viral copy number in the epithelium were increased when the stromal IFN pathway was disrupted, indicating that the stroma helps control the late phase of the HPV life cycle in the epithelium. Increased late gene expression correlated with increased late keratinocyte differentiation but not decreased IFN signaling in the epithelium. These studies show HPV16 has a paracrine effect on stromal innate immunity, reveal a new role for E5 as a stromal innate immune suppressor, and suggest that stromal IFN signaling may influence keratinocyte differentiation.IMPORTANCE The persistence of high-risk human papillomavirus (HPV) infections is the key risk factor for developing HPV-associated cancers. The ability of HPV to evade host immunity is a critical component of its ability to persist. The environment surrounding a tumor is increasingly understood to be critical in cancer development, including immune evasion. Our studies show that HPV can suppress the expression of immune-related genes in neighboring fibroblasts in a three-dimensional (3D) model of human epithelium. This finding is significant, because it indicates that HPV can control innate immunity not only in the infected cell but also in the microenvironment. In addition, the ability of HPV to regulate stromal gene expression depends in part on the viral oncogene E5, revealing a new function for this protein as an immune evasion factor.
Collapse
|
11
|
Context-dependent effect of sPLA 2-IIA induced proliferation on murine hair follicle stem cells and human epithelial cancer. EBioMedicine 2019; 48:364-376. [PMID: 31521610 PMCID: PMC6838435 DOI: 10.1016/j.ebiom.2019.08.053] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Tissue stem cells (SCs) and cancer cells proliferation is regulated by many common signalling mechanisms. These mechanisms temporally balance proliferation and differentiation events during normal tissue homeostasis and repair. However, the effect of these aberrant signalling mechanisms on the ultimate fate of SCs and cancer cells remains obscure. METHODS To evaluate the functional effects of Secretory Phospholipase A2-IIA (sPLA2-IIA) induced abnormal signalling on normal SCs and cancer cells, we have used K14-sPLA2-IIA transgenic mice hair follicle stem cells (HFSCs), DMBA/TPA induced mouse skin tumour tissues, human oral squamous cell carcinoma (OSCC) and skin squamous cell carcinoma (SCC) derived cell lines. FINDINGS Our study demonstrates that sPLA2-IIA induces rapid proliferation of HFSCs, thereby altering the proliferation dynamics leading to a complete loss of the slow cycling H2BGFP positive HFSCs. Interestingly, in vivo reversion study by JNK inhibition exhibited a significant delay in post depilation hair growth, confirming that sPLA2-IIA promotes HFSCs proliferation through JNK/c-Jun signalling. In a different cellular context, we showed increased expression of sPLA2-IIA in human OSCC and mouse skin cancer tissues. Importantly, a xenograft of sPLA2-IIA knockdown cells of OSCC and SCC cell lines showed a concomitant reduction of tumour volume in NOD-SCID mice and decreased JNK/c-Jun signalling. INTERPRETATION This study unravels how an increased proliferation induced by a common proliferation inducer (sPLA2-IIA) alters the fate of normal SCs and cancer cells distinctively through common JNK/c-Jun signalling. Thus, sPLA2-IIA can be a potential target for various diseases including cancer. FUND: This work was partly supported by the Indian Council of Medical Research (ICMR-3097) and ACTREC (42) grants.
Collapse
|
12
|
Perez‐Aso M, Roca A, Bosch J, Martínez‐Teipel B. Striae reconstructed, a full thickness skin model that recapitulates the pathology behind stretch marks. Int J Cosmet Sci 2019; 41:311-319. [DOI: 10.1111/ics.12538] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 05/03/2019] [Indexed: 01/08/2023]
Affiliation(s)
- M. Perez‐Aso
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| | - A. Roca
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| | - J. Bosch
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| | - B. Martínez‐Teipel
- Provital, S.A. Gorgs Llado 200, 08210 Barbera del Valles Barcelona Spain
| |
Collapse
|
13
|
Arasa J, Terencio MC, Andrés RM, Marín-Castejón A, Valcuende-Cavero F, Payá M, Montesinos MC. Defective Induction of COX-2 Expression by Psoriatic Fibroblasts Promotes Pro-inflammatory Activation of Macrophages. Front Immunol 2019; 10:536. [PMID: 30984165 PMCID: PMC6448046 DOI: 10.3389/fimmu.2019.00536] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Accepted: 02/27/2019] [Indexed: 12/20/2022] Open
Abstract
Fibroblasts play an important role as members of the innate immune system through the secretion of COX-2-derived inflammatory mediators such as prostaglandin E2 (PGE2). However, it has been described that dermal fibroblasts behave like mesenchymal stem cells reducing lymphocyte recruitment and dendritic cell activation through PGE2 release. As the role of fibroblasts in psoriasis remains poorly characterized, in the present study we have evaluated the possible influence of PGE2 derived from dermal fibroblasts as modulator of the immune response in psoriatic skin. Our results indicate that under inflammatory conditions, psoriatic fibroblasts showed defective induction of COX-2, which resulted in diminished production of PGE2, in contrast to healthy fibroblasts. This phenotype correlated with deficient c-Jun N-terminal kinase (JNK) activation, in accordance with the hypothesis that alterations in members of the JNK pathway are associated with psoriasis. Furthermore, conditioned medium from psoriatic fibroblasts promoted the polarization of monocytic cells toward a pro-inflammatory profile, effect that was mimicked in healthy fibroblasts after pre-incubation with indomethacin. These results are consistent with a prominent role of dermal fibroblasts in the regulation of inflammatory response through the participation of COX-derived metabolites. This resolutive behavior seems to be defective in psoriatic fibroblasts, offering a possible explanation for the chronification of the disease and for the exacerbation triggered by nonsteroidal anti-inflammatory drugs (NSAIDS) such as indomethacin.
Collapse
Affiliation(s)
- Jorge Arasa
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain.,Departament of Pharmacology, Faculty of Pharmacy, Universitat de València, Valencia, Spain
| | - María Carmen Terencio
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain.,Departament of Pharmacology, Faculty of Pharmacy, Universitat de València, Valencia, Spain
| | - Rosa María Andrés
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain.,Departament of Pharmacology, Faculty of Pharmacy, Universitat de València, Valencia, Spain
| | - Asunción Marín-Castejón
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain.,Departament of Pharmacology, Faculty of Pharmacy, Universitat de València, Valencia, Spain
| | - Francisca Valcuende-Cavero
- Department of Dermatology, University Hospital La Plana, Vila-real, Spain.,Predepartamental Unit of Medicine, Universitat Jaume I, Castellón, Spain
| | - Miguel Payá
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain.,Departament of Pharmacology, Faculty of Pharmacy, Universitat de València, Valencia, Spain
| | - María Carmen Montesinos
- Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, Valencia, Spain.,Departament of Pharmacology, Faculty of Pharmacy, Universitat de València, Valencia, Spain
| |
Collapse
|
14
|
Therapeutic potential of endogenous stem cells and cellular factors for scar-free skin regeneration. Drug Discov Today 2019; 24:69-84. [DOI: 10.1016/j.drudis.2018.10.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Revised: 08/28/2018] [Accepted: 10/25/2018] [Indexed: 12/20/2022]
|
15
|
Abraham DM, Lee TE, Watson LJ, Mao L, Chandok G, Wang HG, Frangakis S, Pitt GS, Shah SH, Wolf MJ, Rockman HA. The two-pore domain potassium channel TREK-1 mediates cardiac fibrosis and diastolic dysfunction. J Clin Invest 2018; 128:4843-4855. [PMID: 30153110 PMCID: PMC6205385 DOI: 10.1172/jci95945] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Accepted: 08/23/2018] [Indexed: 01/08/2023] Open
Abstract
Cardiac two-pore domain potassium channels (K2P) exist in organisms from Drosophila to humans; however, their role in cardiac function is not known. We identified a K2P gene, CG8713 (sandman), in a Drosophila genetic screen and show that sandman is critical to cardiac function. Mice lacking an ortholog of sandman, TWIK-related potassium channel (TREK-1, also known Kcnk2), exhibit exaggerated pressure overload-induced concentric hypertrophy and alterations in fetal gene expression, yet retain preserved systolic and diastolic cardiac function. While cardiomyocyte-specific deletion of TREK-1 in response to in vivo pressure overload resulted in cardiac dysfunction, TREK-1 deletion in fibroblasts prevented deterioration in cardiac function. The absence of pressure overload-induced dysfunction in TREK-1-KO mice was associated with diminished cardiac fibrosis and reduced activation of JNK in cardiomyocytes and fibroblasts. These findings indicate a central role for cardiac fibroblast TREK-1 in the pathogenesis of pressure overload-induced cardiac dysfunction and serve as a conceptual basis for its inhibition as a potential therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Howard A Rockman
- Department of Medicine
- Department of Cell Biology, and
- Department of Molecular Genetics and Microbiology, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
16
|
Inhibition of apoptosis signal-regulating kinase 1 alters the wound epidermis and enhances auricular cartilage regeneration. PLoS One 2017; 12:e0185803. [PMID: 29045420 PMCID: PMC5646791 DOI: 10.1371/journal.pone.0185803] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 09/19/2017] [Indexed: 11/19/2022] Open
Abstract
Why regeneration does not occur in mammals remains elusive. In lower vertebrates, epimorphic regeneration of the limb is directed by the wound epidermis, which controls blastema formation to promote regrowth of the appendage. Herein, we report that knockout (KO) or inhibition of Apoptosis Signal-regulated Kinase-1 (ASK1), also known as mitogen-activated protein kinase kinase kinase 5 (MAP3K5), after full thickness ear punch in mice prolongs keratinocyte activation within the wound epidermis and promotes regeneration of auricular cartilage. Histological analysis showed the ASK1 KO ears displayed enhanced protein markers associated with blastema formation, hole closure and regeneration of auricular cartilage. At seven days after punch, the wound epidermis morphology was markedly different in the KO, showing a thickened stratum corneum with rounded cell morphology and a reduction of both the granular cell layer and decreased expression of filament aggregating protein. In addition, cytokeratin 6 was expressed in the stratum spinosum and granulosum. Topical application of inhibitors of ASK1 (NQDI-1), the upstream ASK1 activator, calcium activated mitogen kinase 2 (KN93), or the downstream target, c-Jun N-terminal kinase (SP600125) also resulted in enhanced regeneration; whereas inhibition of the other downstream target, the p38 α/β isoforms, (SB203580) had no effect. The results of this investigation indicate ASK1 inhibition prolongs keratinocyte and blastemal cell activation leading to ear regeneration.
Collapse
|
17
|
Genome-wide Target Enrichment-aided Chip Design: a 66 K SNP Chip for Cashmere Goat. Sci Rep 2017; 7:8621. [PMID: 28819310 PMCID: PMC5561203 DOI: 10.1038/s41598-017-09285-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Accepted: 07/14/2017] [Indexed: 01/24/2023] Open
Abstract
Compared with the commercially available single nucleotide polymorphism (SNP) chip based on the Bead Chip technology, the solution hybrid selection (SHS)-based target enrichment SNP chip is not only design-flexible, but also cost-effective for genotype sequencing. In this study, we propose to design an animal SNP chip using the SHS-based target enrichment strategy for the first time. As an update to the international collaboration on goat research, a 66 K SNP chip for cashmere goat was created from the whole-genome sequencing data of 73 individuals. Verification of this 66 K SNP chip with the whole-genome sequencing data of 436 cashmere goats showed that the SNP call rates was between 95.3% and 99.8%. The average sequencing depth for target SNPs were 40X. The capture regions were shown to be 200 bp that flank target SNPs. This chip was further tested in a genome-wide association analysis of cashmere fineness (fiber diameter). Several top hit loci were found marginally associated with signaling pathways involved in hair growth. These results demonstrate that the 66 K SNP chip is a useful tool in the genomic analyses of cashmere goats. The successful chip design shows that the SHS-based target enrichment strategy could be applied to SNP chip design in other species.
Collapse
|
18
|
Human Papillomavirus and the Stroma: Bidirectional Crosstalk during the Virus Life Cycle and Carcinogenesis. Viruses 2017; 9:v9080219. [PMID: 28792475 PMCID: PMC5580476 DOI: 10.3390/v9080219] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/03/2017] [Accepted: 08/04/2017] [Indexed: 12/22/2022] Open
Abstract
Human papillomaviruses (HPVs) are double-stranded DNA (dsDNA) tumor viruses that are causally associated with human cancers of the anogenital tract, skin, and oral cavity. Despite the availability of prophylactic vaccines, HPVs remain a major global health issue due to inadequate vaccine availability and vaccination coverage. The HPV life cycle is established and completed in the terminally differentiating stratified epithelia, and decades of research using in vitro organotypic raft cultures and in vivo genetically engineered mouse models have contributed to our understanding of the interactions between HPVs and the epithelium. More recently, important and emerging roles for the underlying stroma, or microenvironment, during the HPV life cycle and HPV-induced disease have become clear. This review discusses the current understanding of the bidirectional communication and relationship between HPV-infected epithelia and the surrounding microenvironment. As is the case with other human cancers, evidence suggests that the stroma functions as a significant partner in tumorigenesis and helps facilitate the oncogenic potential of HPVs in the stratified epithelium.
Collapse
|
19
|
Woodby B, Scott M, Bodily J. The Interaction Between Human Papillomaviruses and the Stromal Microenvironment. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2016; 144:169-238. [PMID: 27865458 PMCID: PMC5727914 DOI: 10.1016/bs.pmbts.2016.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human papillomaviruses (HPVs) are small, double-stranded DNA viruses that replicate in stratified squamous epithelia and cause a variety of malignancies. Current efforts in HPV biology are focused on understanding the virus-host interactions that enable HPV to persist for years or decades in the tissue. The importance of interactions between tumor cells and the stromal microenvironment has become increasingly apparent in recent years, but how stromal interactions impact the normal, benign life cycle of HPVs, or progression of lesions to cancer is less understood. Furthermore, how productively replicating HPV impacts cells in the stromal environment is also unclear. Here we bring together some of the relevant literature on keratinocyte-stromal interactions and their impacts on HPV biology, focusing on stromal fibroblasts, immune cells, and endothelial cells. We discuss how HPV oncogenes in infected cells manipulate other cells in their environment, and, conversely, how neighboring cells may impact the efficiency or course of HPV infection.
Collapse
Affiliation(s)
- B Woodby
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - M Scott
- Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - J Bodily
- Louisiana State University Health Sciences Center, Shreveport, LA, United States.
| |
Collapse
|
20
|
Ghanavati Z, Orazizadeh M, Bayati V, Abbaspour MR, Khorsandi L, Mansouri E, Neisi N. Characterization of A Three-Dimensional Organotypic Co-Culture Skin Model for Epidermal Differentiation of Rat Adipose-Derived Stem Cells. CELL JOURNAL 2016; 18:289-301. [PMID: 27602310 PMCID: PMC5011316 DOI: 10.22074/cellj.2016.4553] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Accepted: 11/28/2015] [Indexed: 12/17/2022]
Abstract
Objective The organotypic co-culture is a well-known technique to examine cellular
interactions and their roles in stem cell proliferation and differentiation. This study
aims to evaluate the effects of dermal fibroblasts (DFs) on epidermal differentiation
of adipose-derived stem cells (ASCs) using a three-dimensional (3D) organotypic co-
culture technique.
Materials and Methods In this experimental research study, rat DFs and ASCs were
isolated and cultured separately on electrospun polycaprolactone (PCL) matrices.
The PCL matrices seeded by ASCs were superimposed on to the matrices seeded
by DFs in order to create a 3D organotypic co-culture. In the control groups, PCL
matrices seeded by ASCs were placed on matrices devoid of DFs. After 10 days, we
assessed the expressions of keratinocyte-related genes by real-time reverse transcriptase-polymerase chain reaction (RT-PCR) and expression of pan-cytokeratin
protein by immunofluorescence in the differentiated keratinocyte-like cells from co-
culture and control groups. Keratinocyte-like cell morphologies were also observed
by scanning electron microscopy (SEM).
Results The early, intermediate, and terminal differentiation keratinocyte markers-Cytokeratin14, Filaggrin, and Involucrin significantly expressed in the co-culture groups com-
pared to the control ones (P<0.05). We observed pan-cytokeratin in keratinocyte-like cells
of both groups by immunofluorescence. SEM observation of the co-culture groups showed
that the differentiated keratinocyte-like cells developed a polygonal cobblestone shape,
considered characteristic of keratinocytes.
Conclusion The 3D organotypic co-culture bilayered construct that consisted of DFs and
ASCs was an effective technique for epidermal differentiation of ASCs. This co-culture
might be useful for epidermal differentiation of stem cells for future applications in skin
regeneration.
Collapse
Affiliation(s)
- Zeinab Ghanavati
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahmoud Orazizadeh
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Vahid Bayati
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad Reza Abbaspour
- Targeted Drug Delivery Research Center, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Layasadat Khorsandi
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Esrafil Mansouri
- Cellular and Molecular Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran; Department of Anatomical Sciences, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Niloofar Neisi
- Department of Medical Virology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
21
|
JNK Signaling: Regulation and Functions Based on Complex Protein-Protein Partnerships. Microbiol Mol Biol Rev 2016; 80:793-835. [PMID: 27466283 DOI: 10.1128/mmbr.00043-14] [Citation(s) in RCA: 321] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The c-Jun N-terminal kinases (JNKs), as members of the mitogen-activated protein kinase (MAPK) family, mediate eukaryotic cell responses to a wide range of abiotic and biotic stress insults. JNKs also regulate important physiological processes, including neuronal functions, immunological actions, and embryonic development, via their impact on gene expression, cytoskeletal protein dynamics, and cell death/survival pathways. Although the JNK pathway has been under study for >20 years, its complexity is still perplexing, with multiple protein partners of JNKs underlying the diversity of actions. Here we review the current knowledge of JNK structure and isoforms as well as the partnerships of JNKs with a range of intracellular proteins. Many of these proteins are direct substrates of the JNKs. We analyzed almost 100 of these target proteins in detail within a framework of their classification based on their regulation by JNKs. Examples of these JNK substrates include a diverse assortment of nuclear transcription factors (Jun, ATF2, Myc, Elk1), cytoplasmic proteins involved in cytoskeleton regulation (DCX, Tau, WDR62) or vesicular transport (JIP1, JIP3), cell membrane receptors (BMPR2), and mitochondrial proteins (Mcl1, Bim). In addition, because upstream signaling components impact JNK activity, we critically assessed the involvement of signaling scaffolds and the roles of feedback mechanisms in the JNK pathway. Despite a clarification of many regulatory events in JNK-dependent signaling during the past decade, many other structural and mechanistic insights are just beginning to be revealed. These advances open new opportunities to understand the role of JNK signaling in diverse physiological and pathophysiological states.
Collapse
|
22
|
Arasa J, Terencio MC, Andrés RM, Valcuende-Cavero F, Montesinos MC. Decreased SAPK/JNK signalling affects cytokine release and STAT3 activation in psoriatic fibroblasts. Exp Dermatol 2015; 24:800-2. [PMID: 26120912 DOI: 10.1111/exd.12787] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2015] [Indexed: 01/15/2023]
Affiliation(s)
- Jorge Arasa
- Department of Pharmacology, Faculty of Pharmacy, University of Valencia, Valencia, Spain.,Center of Molecular Recognition and Technological Development (IDM), Valencia, Spain
| | - María Carmen Terencio
- Department of Pharmacology, Faculty of Pharmacy, University of Valencia, Valencia, Spain.,Center of Molecular Recognition and Technological Development (IDM), Valencia, Spain
| | - Rosa M Andrés
- Department of Pharmacology, Faculty of Pharmacy, University of Valencia, Valencia, Spain.,Center of Molecular Recognition and Technological Development (IDM), Valencia, Spain
| | - Francisca Valcuende-Cavero
- Department of Dermatology, University Hospital La Plana, Vila-real, Spain.,Department of Medicine and Surgery, CEU Cardenal Herrera University, Castellón de la Plana, Spain
| | - María Carmen Montesinos
- Department of Pharmacology, Faculty of Pharmacy, University of Valencia, Valencia, Spain.,Center of Molecular Recognition and Technological Development (IDM), Valencia, Spain
| |
Collapse
|
23
|
Vapniarsky N, Arzi B, Hu JC, Nolta JA, Athanasiou KA. Concise Review: Human Dermis as an Autologous Source of Stem Cells for Tissue Engineering and Regenerative Medicine. Stem Cells Transl Med 2015; 4:1187-98. [PMID: 26253713 DOI: 10.5966/sctm.2015-0084] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 07/08/2015] [Indexed: 12/16/2022] Open
Abstract
UNLABELLED The exciting potential for regenerating organs from autologous stem cells is on the near horizon, and adult dermis stem cells (DSCs) are particularly appealing because of the ease and relative minimal invasiveness of skin collection. A substantial number of reports have described DSCs and their potential for regenerating tissues from mesenchymal, ectodermal, and endodermal lineages; however, the exact niches of these stem cells in various skin types and their antigenic surface makeup are not yet clearly defined. The multilineage potential of DSCs appears to be similar, despite great variability in isolation and in vitro propagation methods. Despite this great potential, only limited amounts of tissues and clinical applications for organ regeneration have been developed from DSCs. This review summarizes the literature on DSCs regarding their niches and the specific markers they express. The concept of the niches and the differentiation capacity of cells residing in them along particular lineages is discussed. Furthermore, the advantages and disadvantages of widely used methods to demonstrate lineage differentiation are considered. In addition, safety considerations and the most recent advancements in the field of tissue engineering and regeneration using DSCs are discussed. This review concludes with thoughts on how to prospectively approach engineering of tissues and organ regeneration using DSCs. Our expectation is that implementation of the major points highlighted in this review will lead to major advancements in the fields of regenerative medicine and tissue engineering. SIGNIFICANCE Autologous dermis-derived stem cells are generating great excitement and efforts in the field of regenerative medicine and tissue engineering. The substantial impact of this review lies in its critical coverage of the available literature and in providing insight regarding niches, characteristics, and isolation methods of stem cells derived from the human dermis. Furthermore, it provides analysis of the current state-of-the-art regenerative approaches using human-derived dermal stem cells, with consideration of current guidelines, to assist translation toward therapeutic use.
Collapse
Affiliation(s)
- Natalia Vapniarsky
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Boaz Arzi
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Jerry C Hu
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Jan A Nolta
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| | - Kyriacos A Athanasiou
- Department of Biomedical Engineering, Department of Surgical and Radiological Sciences, School of Veterinary Medicine, Institute for Regenerative Cures and Department of Internal Medicine, School of Medicine, and Department of Orthopaedic Surgery, University of California, Davis, Davis, California, USA
| |
Collapse
|
24
|
Llames S, García-Pérez E, Meana Á, Larcher F, del Río M. Feeder Layer Cell Actions and Applications. TISSUE ENGINEERING PART B-REVIEWS 2015; 21:345-53. [PMID: 25659081 DOI: 10.1089/ten.teb.2014.0547] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Cultures of growth-arrested feeder cells have been used for years to promote cell proliferation, particularly with low-density inocula. Basically, feeder cells consist in a layer of cells unable to divide, which provides extracellular secretions to help another cell to proliferate. It differs from a coculture system because only one cell type is capable to proliferate. It is known that feeder cells support the growth of target cells by releasing growth factors to the culture media, but this is not the only way that feeder cells promote the growth of target cells. In this work, we discuss the different mechanisms of action of feeder cells, tackling questions as to why for some cell cultures the presence of feeder cell layers is mandatory, while in some other cases, the growth of target cells can be achieved with just a conditioned medium. Different treatments to avoid feeder cells to proliferate are revised, not only the classical treatments as mitomycin or γ-irradiation but also the not so common treatments as electric pulses or chemical fixation. Regenerative medicine has been gaining importance in recent years as a discipline that moves biomedical technology from the laboratory to the patients. In this context, human stem and pluripotent cells play an important role, but the presence of feeder cells is necessary for these progenitor cells to grow and differentiate. This review addresses recent specific applications, including those associated to the growth of embryonic and induced pluripotent stem cells. In addition, we have also dealt with safety issues, including feeder cell sources, as major factors of concern for clinical applications.
Collapse
Affiliation(s)
- Sara Llames
- 1 Tissue Engineering Unit, Centro Comunitario de Sangre y Tejidos del Principado de Asturias, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER U714) , Oviedo, Spain
| | - Eva García-Pérez
- 1 Tissue Engineering Unit, Centro Comunitario de Sangre y Tejidos del Principado de Asturias, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER U714) , Oviedo, Spain .,2 TERMEG, Department of Bioengineering, Universidad Carlos III de Madrid (UC3M) , Madrid, Spain
| | - Álvaro Meana
- 1 Tissue Engineering Unit, Centro Comunitario de Sangre y Tejidos del Principado de Asturias, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER U714) , Oviedo, Spain
| | - Fernando Larcher
- 2 TERMEG, Department of Bioengineering, Universidad Carlos III de Madrid (UC3M) , Madrid, Spain .,3 Epithelial Biomedicine Division, CIEMAT, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER U714) , Madrid, Spain
| | - Marcela del Río
- 2 TERMEG, Department of Bioengineering, Universidad Carlos III de Madrid (UC3M) , Madrid, Spain .,3 Epithelial Biomedicine Division, CIEMAT, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER U714) , Madrid, Spain .,4 Instituto de Investigaciones Fundación Jiménez Díaz , Madrid, Spain
| |
Collapse
|
25
|
Schulz JN, Zeltz C, Sørensen IW, Barczyk M, Carracedo S, Hallinger R, Niehoff A, Eckes B, Gullberg D. Reduced granulation tissue and wound strength in the absence of α11β1 integrin. J Invest Dermatol 2015; 135:1435-1444. [PMID: 25634355 PMCID: PMC4407012 DOI: 10.1038/jid.2015.24] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Revised: 01/14/2015] [Accepted: 01/16/2015] [Indexed: 12/24/2022]
Abstract
Previous wound healing studies have failed to define a role for either α1β1 or α2β1 integrin in fibroblast-mediated wound contraction, suggesting the involvement of another collagen receptor in this process. Our previous work demonstrated that the integrin subunit α11 is highly induced during wound healing both at the mRNA and protein level, prompting us to investigate and dissect the role of the integrin α11β1 during this process. Therefore, we used mice with a global ablation of either α2 or α11 or both integrin subunits and investigated the repair of excisional wounds. Analyses of wounds demonstrated that α11β1 deficiency results in reduced granulation tissue formation and impaired wound contraction, independently of the presence of α2β1. Our combined in vivo and in vitro data further demonstrate that dermal fibroblasts lacking α11β1 are unable to efficiently convert to myofibroblasts, resulting in scar tissue with compromised tensile strength. Moreover, we suggest that the reduced stability of the scar is a consequence of poor collagen remodeling in α11−/− wounds associated with defective transforming growth factor-β–dependent JNK signaling.
Collapse
Affiliation(s)
| | - Cédric Zeltz
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Ida W Sørensen
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Malgorzata Barczyk
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Sergio Carracedo
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway
| | - Ralf Hallinger
- Department of Dermatology, University of Cologne, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopedics, German Sport University, Cologne, Germany; Cologne Center for Musculoskeletal Biomechanics, University of Cologne, Cologne, Germany
| | - Beate Eckes
- Department of Dermatology, University of Cologne, Cologne, Germany.
| | - Donald Gullberg
- Department of Biomedicine, Centre for Cancer Biomarkers (CCBIO), Norwegian Centre of Excellence, University of Bergen, Bergen, Norway.
| |
Collapse
|
26
|
Rimann M, Bono E, Annaheim H, Bleisch M, Graf-Hausner U. Standardized 3D Bioprinting of Soft Tissue Models with Human Primary Cells. ACTA ACUST UNITED AC 2015; 21:496-509. [PMID: 25609254 DOI: 10.1177/2211068214567146] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Indexed: 12/22/2022]
Abstract
Cells grown in 3D are more physiologically relevant than cells cultured in 2D. To use 3D models in substance testing and regenerative medicine, reproducibility and standardization are important. Bioprinting offers not only automated standardizable processes but also the production of complex tissue-like structures in an additive manner. We developed an all-in-one bioprinting solution to produce soft tissue models. The holistic approach included (1) a bioprinter in a sterile environment, (2) a light-induced bioink polymerization unit, (3) a user-friendly software, (4) the capability to print in standard labware for high-throughput screening, (5) cell-compatible inkjet-based printheads, (6) a cell-compatible ready-to-use BioInk, and (7) standard operating procedures. In a proof-of-concept study, skin as a reference soft tissue model was printed. To produce dermal equivalents, primary human dermal fibroblasts were printed in alternating layers with BioInk and cultured for up to 7 weeks. During long-term cultures, the models were remodeled and fully populated with viable and spreaded fibroblasts. Primary human dermal keratinocytes were seeded on top of dermal equivalents, and epidermis-like structures were formed as verified with hematoxylin and eosin staining and immunostaining. However, a fully stratified epidermis was not achieved. Nevertheless, this is one of the first reports of an integrative bioprinting strategy for industrial routine application.
Collapse
Affiliation(s)
- Markus Rimann
- Institute of Chemistry and Biological Chemistry (ICBC), Zurich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
| | - Epifania Bono
- Institute of Chemistry and Biological Chemistry (ICBC), Zurich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
| | - Helene Annaheim
- Institute of Chemistry and Biological Chemistry (ICBC), Zurich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
| | - Matthias Bleisch
- Institute of Chemistry and Biological Chemistry (ICBC), Zurich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
| | - Ursula Graf-Hausner
- Institute of Chemistry and Biological Chemistry (ICBC), Zurich University of Applied Sciences (ZHAW), Waedenswil, Switzerland
| |
Collapse
|
27
|
Pendaries V, Malaisse J, Pellerin L, Le Lamer M, Nachat R, Kezic S, Schmitt AM, Paul C, Poumay Y, Serre G, Simon M. Knockdown of Filaggrin in a Three-Dimensional Reconstructed Human Epidermis Impairs Keratinocyte Differentiation. J Invest Dermatol 2014; 134:2938-2946. [DOI: 10.1038/jid.2014.259] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 05/14/2014] [Accepted: 05/20/2014] [Indexed: 01/10/2023]
|
28
|
Abstract
Mammalian skin research represents the convergence of three complementary disciplines: cell biology, mouse genetics, and dermatology. The skin provides a paradigm for current research in cell adhesion, inflammation, and tissue stem cells. Here, I discuss recent insights into the cell biology of skin. Single-cell analysis has revealed that human epidermal stem cells are heterogeneous and differentiate in response to multiple extrinsic signals. Live-cell imaging, optogenetics, and cell ablation experiments show skin cells to be remarkably dynamic. High-throughput, genome-wide approaches have yielded unprecedented insights into the circuitry that controls epidermal stem cell fate. Last, integrative biological analysis of human skin disorders has revealed unexpected functions for elements of the skin that were previously considered purely structural.
Collapse
Affiliation(s)
- Fiona M Watt
- King's College London Centre for Stem Cells and Regenerative Medicine, 28th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.
| |
Collapse
|