1
|
Arora M, Pavlíková Z, Kučera T, Kozlík P, Šopin T, Vacík T, Ľupták M, Duda M, Slanař O, Kutinová Canová N. Pharmacological effects of mTORC1/C2 inhibitor in a preclinical model of NASH progression. Biomed Pharmacother 2023; 167:115447. [PMID: 37683589 DOI: 10.1016/j.biopha.2023.115447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/29/2023] [Accepted: 09/04/2023] [Indexed: 09/10/2023] Open
Abstract
Knowledge of the benefits of mTOR inhibition concerning adipogenesis and inflammation has recently encouraged the investigation of a new generation of mTOR inhibitors for non-alcoholic steatohepatitis (NASH). We investigated whether treatment with a specific mTORC1/C2 inhibitor (Ku-0063794; KU) exerted any beneficial impacts on experimentally-induced NASH in vitro and in vivo. The results indicated that KU decreases palmitic acid-induced lipotoxicity in cultivated primary hepatocytes, thus emerging as a successful candidate for testing in an in vivo NASH dietary model, which adopted the intraperitoneal KU dosing route rather than oral application due to its significantly greater bioavailability in mice. The pharmacodynamics experiments commenced with the feeding of male C57BL/6 mice with a high-fat atherogenic western-type diet (WD) for differing intervals over several weeks aimed at inducing various phases of NASH. In addition to the WD, the mice were treated with KU for 3 weeks or 4 months. Acute and chronic KU treatments were observed to be safe at the given concentrations with no toxicity indications in the mice. KU was found to alleviate NASH-related hepatotoxicity, mitochondrial and oxidative stress, and decrease the liver triglyceride content and TNF-α mRNA in at least one set of in vivo experiments. The KU modulated liver expression of selected metabolic and oxidative stress-related genes depended upon the length and severity of the disease. Although KU failed to completely reverse the histological progression of NASH in the mice, we demonstrated the complexity of mTORC1/C2 signaling regulation and suggest a stratified therapeutic management approach throughout the disease course.
Collapse
Affiliation(s)
- Mahak Arora
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Zuzana Pavlíková
- Institute of Histology and Embryology, First Faculty of Medicine, Charles University, Prague, Czech Republic; Department of Anthropology and Human Genetics, Faculty of Science, Charles University, Prague, Czech Republic
| | - Tomáš Kučera
- Institute of Histology and Embryology, First Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Petr Kozlík
- Department of Analytical Chemistry, Faculty of Science, Charles University, Prague, Czech Republic
| | - Tijana Šopin
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Tomáš Vacík
- Institute of Biology and Medical Genetics of the First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Matej Ľupták
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Matthias Duda
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Ondřej Slanař
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic
| | - Nikolina Kutinová Canová
- Institute of Pharmacology, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czech Republic.
| |
Collapse
|
2
|
Gao X, Jin Y, Zhu W, Wu X, Wang J, Guo C. Regulation of Eukaryotic Translation Initiation Factor 4E as a Potential Anticancer Strategy. J Med Chem 2023; 66:12678-12696. [PMID: 37725577 DOI: 10.1021/acs.jmedchem.3c00636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2023]
Abstract
Eukaryotic translation initiation factors (eIFs) are highly expressed in cancer cells, especially eIF4E, the central regulatory node driving cancer cell growth and a potential target for anticancer drugs. eIF4E-targeting strategies primarily focus on inhibiting eIF4E synthesis, interfering with eIF4E/eIF4G interactions, and targeting eIF4E phosphorylation and peptide inhibitors. Although some small-molecule inhibitors are in clinical trials, no eIF4E inhibitors are available for clinical use. We provide an overview of the regulatory mechanisms of eIF4E and summarize the progress in developing and discovering eIF4E inhibitor strategies. We propose that interference with eIF4E/eIF4G interactions will provide a new perspective for the design of eIF4E inhibitors and may be a preferred strategy.
Collapse
Affiliation(s)
- Xintao Gao
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Yonglong Jin
- The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Wenyong Zhu
- Department of Thoracic Surgery, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, Qingdao, 266035, China
| | - Xiaochen Wu
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Jing Wang
- Department of Biology Science and Technology, Baotou Teacher's College, Baotou 014030, China
| | - Chuanlong Guo
- College of Chemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| |
Collapse
|
3
|
Tai Y, Zheng L, Liao J, Wang Z, Zhang L. Roles of the HIF-1α pathway in the development and progression of keloids. Heliyon 2023; 9:e18651. [PMID: 37636362 PMCID: PMC10448433 DOI: 10.1016/j.heliyon.2023.e18651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 07/17/2023] [Accepted: 07/24/2023] [Indexed: 08/29/2023] Open
Abstract
Keloids, a pathological scar that is induced by the consequence of aberrant wound healing, is still a major global health concern for its unsatisfactory treatment outcomes. HIF-1α, a main regulator of hypoxia, mainly acts through some proteins or signaling pathways and plays important roles in a variety of biological processes. Accumulating evidence has shown that HIF-1α played a crucial role in the process of keloid formation. In this review, we attempted to summarize the current knowledge on the association between HIF-1α expression and the development and progression of keloids. Through a comprehensive analysis, the molecular mechanisms underlying HIF-1α in keloids were shown to be correlated to the proliferation of fibroblasts, angiogenesis, and collagen deposits. The affected proteins and the signaling pathways were multiple. For instance, HIF-1α was reported to promote keloids formation by enhancing angiogenesis, fibroblast proliferation, and collagen deposition through the activation of periostin PI3K/Akt, TGF-β/Smad and TLR4/MyD88/NF-κB pathway. However, the specific effects of HIF-1α on keloids keloid illnesses in clinical practice is are entirely unclear, and further studies in clinical trials are still warranted. Therefore, an in-depth understanding of the biological mechanisms of HIF-1α in keloid formation is significant to develop promising therapeutic targets for the treatment of keloids in clinical practice.
Collapse
Affiliation(s)
- Yuncheng Tai
- Department of Burn Surgery, Taizhou Central Hospital (Taizhou University Hospital), Taizhou, 318000, Zhejiang, China
| | - Liying Zheng
- Postgraduate Department, First Affiliated Hospital of Gannan Medical College, Ganzhou, China
| | - Jiao Liao
- Department of Nephrology, Jiaxing Hospital of Traditional Chinese Medicine, Jiaxing, 314000, Zhejiang, China
| | - Zixiong Wang
- Department of Burn and Plastic Surgery, Xinjiang Military General Hospital, Urumqi, 830063, Xinjiang, China
| | - Lai Zhang
- Department of Orthopedics, Taizhou Municipal Hospital, Taizhou, 318000, Zhejiang, China
| |
Collapse
|
4
|
Guo C, Liang L, Zheng J, Xie Y, Qiu X, Tan G, Huang J, Wang L. UCHL1 aggravates skin fibrosis through an IGF-1-induced Akt/mTOR/HIF-1α pathway in keloid. FASEB J 2023; 37:e23015. [PMID: 37256780 DOI: 10.1096/fj.202300153rr] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 05/05/2023] [Accepted: 05/22/2023] [Indexed: 06/02/2023]
Abstract
Keloid is a heterogeneous disease featured by the excessive production of extracellular matrix. It is a great challenge for both clinicians and patients regarding the exaggerated and uncontrolled outgrowth and the therapeutic resistance of the disease. In this study, we verified that UCHL1 was drastically upregulated in keloid fibroblasts. UCHL1 had no effects on cell proliferation and migration, but instead promoted collagen I and α-SMA expression that was inhibited by silencing UCHL1 gene and by adding in LDN-57444, a pharmacological inhibitor for UCHL1 activity as well. The pathological process was mediated by IGF-1 promoted Akt/mTOR/HIF-1α signaling pathway because inhibition of any of them could reduce the expression of collagen I and α-SMA driven by UCHL1 in fibroblasts. Also, we found that UCHL1 expression in keloid fibroblasts was promoted by M2 macrophages via TGF-β1. These findings extend our understanding of the pathogenesis of keloid and provide potential therapeutic targets for the disease.
Collapse
Affiliation(s)
- Chipeng Guo
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lizhu Liang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingbin Zheng
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yang Xie
- Department of Dermatology, the Third Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou, China
| | - Xiaonan Qiu
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guozhen Tan
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingang Huang
- Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Liangchun Wang
- Department of Dermatology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
5
|
Roy T, Boateng ST, Uddin MB, Banang-Mbeumi S, Yadav RK, Bock CR, Folahan JT, Siwe-Noundou X, Walker AL, King JA, Buerger C, Huang S, Chamcheu JC. The PI3K-Akt-mTOR and Associated Signaling Pathways as Molecular Drivers of Immune-Mediated Inflammatory Skin Diseases: Update on Therapeutic Strategy Using Natural and Synthetic Compounds. Cells 2023; 12:1671. [PMID: 37371141 PMCID: PMC10297376 DOI: 10.3390/cells12121671] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 06/10/2023] [Accepted: 06/12/2023] [Indexed: 06/29/2023] Open
Abstract
The dysregulated phosphatidylinositol-3-kinase (PI3K)-Akt-mammalian target of rapamycin (mTOR) signaling pathway has been implicated in various immune-mediated inflammatory and hyperproliferative dermatoses such as acne, atopic dermatitis, alopecia, psoriasis, wounds, and vitiligo, and is associated with poor treatment outcomes. Improved comprehension of the consequences of the dysregulated PI3K/Akt/mTOR pathway in patients with inflammatory dermatoses has resulted in the development of novel therapeutic approaches. Nonetheless, more studies are necessary to validate the regulatory role of this pathway and to create more effective preventive and treatment methods for a wide range of inflammatory skin diseases. Several studies have revealed that certain natural products and synthetic compounds can obstruct the expression/activity of PI3K/Akt/mTOR, underscoring their potential in managing common and persistent skin inflammatory disorders. This review summarizes recent advances in understanding the role of the activated PI3K/Akt/mTOR pathway and associated components in immune-mediated inflammatory dermatoses and discusses the potential of bioactive natural products, synthetic scaffolds, and biologic agents in their prevention and treatment. However, further research is necessary to validate the regulatory role of this pathway and develop more effective therapies for inflammatory skin disorders.
Collapse
Affiliation(s)
- Tithi Roy
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Samuel T. Boateng
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Mohammad B. Uddin
- Department of Toxicology and Cancer Biology, Center for Research on Environmental Diseases, College of Medicine, University of Kentucky, Lexington, KY 40536, USA;
| | - Sergette Banang-Mbeumi
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
- Division for Research and Innovation, POHOFI Inc., Madison, WI 53744, USA
- School of Nursing and Allied Health Sciences, Louisiana Delta Community College, Monroe, LA 71203, USA
| | - Rajesh K. Yadav
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Chelsea R. Bock
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Joy T. Folahan
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Xavier Siwe-Noundou
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, P.O. Box 218, Pretoria 0208, South Africa;
| | - Anthony L. Walker
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
| | - Judy A. King
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA;
- College of Medicine, Belmont University, 900 Belmont Boulevard, Nashville, TN 37212, USA
| | - Claudia Buerger
- Department of Dermatology, Venerology and Allergology, Clinic of the Goethe University, 60590 Frankfurt am Main, Germany;
| | - Shile Huang
- Department of Biochemistry and Molecular Biology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA;
- Department of Hematology and Oncology, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130, USA
- Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Jean Christopher Chamcheu
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana at Monroe, Monroe, LA 71209, USA; (T.R.); (S.T.B.); (S.B.-M.); (R.K.Y.); (C.R.B.); (J.T.F.); (A.L.W.)
- Department of Pathology and Translational Pathobiology, LSU Health Shreveport, 1501 Kings Highway, Shreveport, LA 71103, USA;
| |
Collapse
|
6
|
Neves LMG, Wilgus TA, Bayat A. In Vitro, Ex Vivo, and In Vivo Approaches for Investigation of Skin Scarring: Human and Animal Models. Adv Wound Care (New Rochelle) 2023; 12:97-116. [PMID: 34915768 DOI: 10.1089/wound.2021.0139] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Significance: The cutaneous repair process naturally results in different types of scarring that are classified as normal or pathological. Affected individuals are often affected from an esthetic, physical (functional), and psychosocial perspective. The distinct nature of scarring in humans, particularly the formation of pathological scars, makes the study of skin scarring a challenge for researchers in this area. Several established experimental models exist for studying scar formation. However, the increasing development and validation of newly emerging models have made it possible to carry out studies focused on different variables that influence this unique process. Recent Advances: Experimental models such as in vitro, ex vivo, and in vivo models have obtained different degrees of success in the reproduction of the scar formation in its native milieu and true environment. These models also differ in their ability to elucidate the molecular, cellular, and structural mechanisms involved in scarring, as well as for testing new agents and approaches for therapies. The models reviewed here, including cells derived from human skin and in vivo animal models, have contributed to the advancement of skin scarring research. Critical Issues and Future Directions: The absence of experimental models that faithfully reproduce the typical characteristics of the different types of human skin scars makes the improvement of validated models and the establishment of new ones a critical unmet need. The fields of wound healing research combined with tissue engineering have offered newer alternatives for experimental studies with the potential to provide clinically useful knowledge about scar formation.
Collapse
Affiliation(s)
- Lia M G Neves
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom
| | - Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, Ohio, USA
| | - Ardeshir Bayat
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom.,Medical Research Council (MRC) Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
7
|
Huang S, Deng W, Dong Y, Hu Z, Zhang Y, Wang P, Cao X, Chen M, Cheng P, Xu H, Zhu W, Tang B, Zhu J. Melatonin influences the biological characteristics of keloid fibroblasts through the Erk and Smad signalling pathways. BURNS & TRAUMA 2023; 11:tkad005. [PMID: 36873285 PMCID: PMC9977354 DOI: 10.1093/burnst/tkad005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/31/2023] [Accepted: 01/17/2023] [Indexed: 03/06/2023]
Abstract
Background Keloids are abnormal fibrous hyperplasias that are difficult to treat. Melatonin can be used to inhibit the development of certain fibrotic diseases but has never been used to treat keloids. We aimed to discover the effects and mechanisms of melatonin in keloid fibroblasts (KFs). Methods Flow cytometry, CCK-8 assays, western blotting, wound-healing assays, transwell assays, collagen gel contraction assays and immunofluorescence assays were applied to demonstrate the effects and mechanisms of melatonin in fibroblasts derived from normal skin, hypertrophic scars and keloids. The therapeutic potential of the combination of melatonin and 5-fluorouracil (5-FU) was investigated in KFs. Results Melatonin significantly promoted cell apoptosis and inhibited cell proliferation, migration and invasion, contractile capability and collagen production in KFs. Further mechanistic studies demonstrated that melatonin could inhibit the cAMP/PKA/Erk and Smad pathways through the membrane receptor MT2 to alter the biological characteristics of KFs. Moreover, the combination of melatonin and 5-FU remarkably promoted cell apoptosis and inhibited cell migration and invasion, contractile capability and collagen production in KFs. Furthermore, 5-FU suppressed the phosphorylation of Akt, mTOR, Smad3 and Erk, and melatonin in combination with 5-FU markedly suppressed the activation of the Akt, Erk and Smad pathways. Conclusions Collectively, melatonin may inhibit the Erk and Smad pathways through the membrane receptor MT2 to alter the cell functions of KFs, while combination with 5-FU could exert even more inhibitory effects in KFs through simultaneous suppression of multiple signalling pathways.
Collapse
Affiliation(s)
- Shaobin Huang
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China.,Department of Cosmetic and Plastic Surgery, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou 510655, China.,Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Wuguo Deng
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Yunxian Dong
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Zhicheng Hu
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Yi Zhang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Nantong University, Nantong 226001, China
| | - Peng Wang
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Xiaoling Cao
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Miao Chen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China; Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| | - Pu Cheng
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Hailin Xu
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Wenkai Zhu
- Department of Obstetrics and Gynecology, School of Medicine, Stanford University, Stanford, CA 94305, United States
| | - Bing Tang
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| | - Jiayuan Zhu
- Department of Burn, First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, China
| |
Collapse
|
8
|
Yu Y, Wu H, Zhang Q, Ogawa R, Fu S. Emerging insights into the immunological aspects of keloids. J Dermatol 2021; 48:1817-1826. [PMID: 34549462 DOI: 10.1111/1346-8138.16149] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 08/18/2021] [Accepted: 08/25/2021] [Indexed: 12/21/2022]
Abstract
A special kind of scar, keloid, sometimes grows huge, disturbing patients in different ways. We discussed the pathogenesis of keloids and found researches about fibroblasts and collagen disorders, with little emphasis on immunity. Coupled with few effective treatments in keloid at present, we have focused on the immunological mechanisms of keloids with an aim to unravel some new therapeutic approaches in the future. In this review, the immunological processes are separately illustrated by the classification of different immune cells. In addition, we also discuss possible reasons for the repeated recurrence of keloids, the phenomenon of cell talks, and inflammation-related signal pathways involved in the pathogenesis of keloids.
Collapse
Affiliation(s)
- Yangyiyi Yu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Haijing Wu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Qing Zhang
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| | - Rei Ogawa
- Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School, Tokyo, Japan
| | - Siqi Fu
- Hunan Key Laboratory of Medical Epigenomics, Department of Dermatology, Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Abstract
Scar is a common way of healing after tissue injury. The poor scar healing will not only cause dysfunction of tissues and organs but also affect the appearance of the patients’ body surface, which causes the pressure of life and spirit to the patients. However, the formation of scar tissue is an extremely complex process and its mechanism is not fully understood. At present, there is no treatment method to eliminate scars completely. Fibroblasts are the most abundant cells in the dermis, which have the ability to synthesize and remodel extracellular matrix (ECM). Myofibroblasts actively participate in the wound healing process and influence the outcome. Therefore, both of them play important roles in wound healing and scar formation. Adipose tissue-derived stem cells (ADSCs) are pluripotent stem cells that can act on target cells by paracrine. Adipose tissue stem cell-derived exosomes (ADSC-Exos) are important secretory substances of ADSCs. They are nanomembrane vesicles that can transport a variety of cellular components and fuse with target cells. In this review, we will discuss the effects of ADSCs and ADSC-Exos on the behavior of fibroblasts and myofibroblasts during wound healing and scarring stage in combination with recent studies.
Collapse
|
10
|
Jiang M, Bu WB, Chen YJ, Li L, Xiao T, Gu H. Mediation of Anti-Keloid Effects of mTOR Inhibitors by Autophagy-Independent Machinery. INTERNATIONAL JOURNAL OF DERMATOLOGY AND VENEREOLOGY 2021; 4:210-218. [DOI: 10.1097/jd9.0000000000000189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Accepted: 07/30/2021] [Indexed: 01/04/2025]
Abstract
Abstract
Objective:
Blocking mechanistic target of rapamycin (mTOR) activation with mTOR inhibitors has promising therapeutic potential for keloids. However, the precise mechanism of mTOR inhibitors remains unclear. This study was aimed to investigate the role of autophagy machinery in the anti-keloid effects of mTOR inhibitors.
Methods:
We first validated the biological effects induced by the mTOR inhibitors rapamycin (100 nmol/L) and KU-0063794 (5 μmol/L) on the proliferation, apoptosis, migration, and collagen synthesis of keloid fibroblasts (KFs) derived from Han Chinese persons through a Cell Counting Kit-8 assay, 5-Bromo-2’-deoxyuridine incorporation, Annexin V/propidium iodide staining, migration, and western blotting. To explore whether autophagy machinery is involved in the anti-keloid effects of mTOR inhibitors, we first blocked the autophagy activation induced by rapamycin and KU-0063794 with a pharmacological autophagy inhibitor (wortmannin) or by silencing the key autophagy gene (ATG5), and we then re-evaluated these biological effects on KFs.
Results:
Blocking mTOR activation with either rapamycin or KU-0063794 completely inhibited proliferation, migration, and collagen synthesis of primary KFs but did not affect apoptosis. Incubating KFs with the autophagy inhibitor wortmannin or performing ATG5 silencing abrogated the subsequent activation of autophagic activity induced by rapamycin (rapamycin + E-64d + pepstatin vs. rapamycin + wortmannin + E-64d + pepstatin: 1.88 ± 0.38 vs. 1.02 ± 0.35, F = 6.86, P = 0.013), (non-sense control + rapamycin vs. ATG5 small interfering RNA + rapamycin: 1.46 ± 0.18 vs. 0.75 ± 0.20, respectively; F = 7.68, P = 0.01) or KU-0063794 (KU-0063794 + E-64d + pepstatin vs. KU-0063794 + wortmannin + E-64d + pepstatin: 1.65 ± 0.35 vs. 0.76 ± 0.17, F = 10.01, P = 0.004), (NC + KU-0063794 vs. ATG5 small interfering RNA + KU-0063794: 1.59 ± 0.50 vs. 0.77 ± 0.09, F = 5.93, P = 0.02) as evidenced by decreased accumulation of LC3-II. However, blockage of autophagy induction in mTOR inhibitor-treated KFs with both methods did not disturb their anti-keloid effects, such as inhibition of cell viability, cell migration, and collagen synthesis (P > 0.05 each).
Conclusion:
Blocking mTOR activation with the mTOR inhibitors rapamycin and KU-0063794 showed anti-keloid effects in KFs. Restoration of autophagy inhibition by mTOR inhibitors does not contribute to their anti-keloid effects.
Collapse
Affiliation(s)
- Meng Jiang
- Department of Physiotherapy, Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical, Nanjing, Jiangsu 210042, China
| | - Wen-Bo Bu
- Department of Dermatologic Surgery, Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical, Nanjing, Jiangsu 210042, China
| | - Yu-Jie Chen
- Department of Physiotherapy, Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical, Nanjing, Jiangsu 210042, China
| | - Li Li
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu 210042, China
| | - Ta Xiao
- Department of Physiotherapy, Hospital for Skin Diseases (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical, Nanjing, Jiangsu 210042, China
| | - Heng Gu
- Jiangsu Key Laboratory of Molecular Biology for Skin Diseases and STIs, Hospital for Skin Disease (Institute of Dermatology), Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu 210042, China
| |
Collapse
|
11
|
Dual inhibition of HDAC and tyrosine kinase signaling pathways with CUDC-907 attenuates TGFβ1 induced lung and tumor fibrosis. Cell Death Dis 2020; 11:765. [PMID: 32943605 PMCID: PMC7499263 DOI: 10.1038/s41419-020-02916-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 12/17/2022]
Abstract
TGFβ1 signaling is a critical driver of collagen accumulation in pulmonary fibrotic diseases and a well-characterized regulator of cancer associated fibroblasts (CAF) activation in lung cancer. Myofibroblasts induced by TGFβ1 and other factors are key players in the pathogenesis of lung fibrosis and tumor. Tremendous attention has been gained to targeting myofibroblasts in order to inhibit the progression of fibrosis and myofibroblast-induced tumor progression and metastasis. Here we determined the therapeutic efficacy of simultaneously targeting PI3K and HDAC pathways in lung myofibroblasts and CAF with a single agent and to evaluate biomarkers of treatment response. CUDC-907 is a first-in-class compound, functioning as a dual inhibitor of HDACs and PI3K/AKT pathway. We investigated its effects in counteracting the activity of TGFβ1-induced myofibroblasts/CAF in regard to cell proliferation, migration, invasion, apoptosis in vitro antifibrosis efficiency in vivo. We found that CUDC-907 inhibited myofibroblasts/CAF cell proliferation, migration and apoptosis in a dose-dependent manner and caused cell cycle arrest at G1-S phase. CUDC-907 not only inhibited myofibroblasts markers expression, but also significantly inhibited the phosphorylation level of AKT, mTOR, Smad2/3, and promoted acetylation of histones. Furthermore, the observed inhibitory effect was also confirmed in bleomycin-induced mice lung fibrosis and nude mouse transplanted tumor model. Overall, these data suggest that dual inhibition of HDAC and the tyrosine kinase signaling pathways with CUDC-907 is a promising treatment strategy for TGFβ1-induced lung and tumor fibrosis.
Collapse
|
12
|
Zhou BY, Wang WB, Wu XL, Zhang WJ, Zhou GD, Gao Z, Liu W. Nintedanib inhibits keloid fibroblast functions by blocking the phosphorylation of multiple kinases and enhancing receptor internalization. Acta Pharmacol Sin 2020; 41:1234-1245. [PMID: 32327724 PMCID: PMC7608201 DOI: 10.1038/s41401-020-0381-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 02/13/2020] [Indexed: 01/22/2023]
Abstract
Keloid is a benign skin tumor characterized by its cell hyperproliferative activity, invasion into normal skin, uncontrolled growth, overproduction and deposition of extracellular matrices and high recurrence rate after various therapies. Nintedanib is a receptor tyrosine kinase inhibitor targeting VEGF, PDGF, FGF, and TGF-β receptors with proved efficacy in anti-angiogenesis and in treating various types of cancers. In this study, we investigated the effects of nintedanib on keloid fibroblasts in both in vitro and ex vivo models. Keloid fibroblasts were prepared from 54 keloid scar samples in active stages collected from 49 patients. We found that nintedanib (1−4 μM) dose-dependently suppressed cell proliferation, induced G0/G1 cell cycle arrest, and inhibited migration and invasion of keloid fibroblasts. The drug also significantly inhibited the gene and protein expression of collagen I (COL-1) and III (COL-3), fibronectin (FN), and connective growth factor (CTGF), as well as the gene expression of other pathological factors, such as alpha smooth muscle actin (α-SMA), plasminogen activator inhibitor-1 (PAI-1), FK506-binding protein 10 (FKBP10), and heat shock protein 47 (HSP47) in keloid fibroblasts. Furthermore, nintedanib treatment significantly suppressed the phosphorylation of p38, JNK, ERK, STAT3, and Smad, enhanced endocytosis of various growth factor receptors. Using an ex vivo tissue explant model, we showed that nintedanib significantly suppressed cell proliferation, migration, and collagen production. The drug also significantly disrupted microvessel structure ex vivo. In summary, our results demonstrate that nintedanib is likely to become a potential targeted drug for keloid systemic therapy.
Collapse
|
13
|
Limandjaja GC, Niessen FB, Scheper RJ, Gibbs S. The Keloid Disorder: Heterogeneity, Histopathology, Mechanisms and Models. Front Cell Dev Biol 2020; 8:360. [PMID: 32528951 PMCID: PMC7264387 DOI: 10.3389/fcell.2020.00360] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 04/22/2020] [Indexed: 12/22/2022] Open
Abstract
Keloids constitute an abnormal fibroproliferative wound healing response in which raised scar tissue grows excessively and invasively beyond the original wound borders. This review provides a comprehensive overview of several important themes in keloid research: namely keloid histopathology, heterogeneity, pathogenesis, and model systems. Although keloidal collagen versus nodules and α-SMA-immunoreactivity have been considered pathognomonic for keloids versus hypertrophic scars, conflicting results have been reported which will be discussed together with other histopathological keloid characteristics. Importantly, histopathological keloid abnormalities are also present in the keloid epidermis. Heterogeneity between and within keloids exists which is often not considered when interpreting results and may explain discrepancies between studies. At least two distinct keloid phenotypes exist, the superficial-spreading/flat keloids and the bulging/raised keloids. Within keloids, the periphery is often seen as the actively growing margin compared to the more quiescent center, although the opposite has also been reported. Interestingly, the normal skin directly surrounding keloids also shows partial keloid characteristics. Keloids are most likely to occur after an inciting stimulus such as (minor and disproportionate) dermal injury or an inflammatory process (environmental factors) at a keloid-prone anatomical site (topological factors) in a genetically predisposed individual (patient-related factors). The specific cellular abnormalities these various patient, topological and environmental factors generate to ultimately result in keloid scar formation are discussed. Existing keloid models can largely be divided into in vivo and in vitro systems including a number of subdivisions: human/animal, explant/culture, homotypic/heterotypic culture, direct/indirect co-culture, and 3D/monolayer culture. As skin physiology, immunology and wound healing is markedly different in animals and since keloids are exclusive to humans, there is a need for relevant human in vitro models. Of these, the direct co-culture systems that generate full thickness keloid equivalents appear the most promising and will be key to further advance keloid research on its pathogenesis and thereby ultimately advance keloid treatment. Finally, the recent change in keloid nomenclature will be discussed, which has moved away from identifying keloids solely as abnormal scars with a purely cosmetic association toward understanding keloids for the fibroproliferative disorder that they are.
Collapse
Affiliation(s)
- Grace C. Limandjaja
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Frank B. Niessen
- Department of Plastic Surgery, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Rik J. Scheper
- Department of Pathology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center (location VUmc), Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
14
|
Yan L, Wang LZ, Xiao R, Cao R, Pan B, Lv XY, Jiao H, Zhuang Q, Sun XJ, Liu YB. Inhibition of microRNA-21-5p reduces keloid fibroblast autophagy and migration by targeting PTEN after electron beam irradiation. J Transl Med 2020; 100:387-399. [PMID: 31558773 DOI: 10.1038/s41374-019-0323-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/14/2019] [Accepted: 08/22/2019] [Indexed: 01/17/2023] Open
Abstract
Electron beam (EB) irradiation is useful to reduce the recurrence of keloids; however, the underlying mechanism remains unknown. MicroRNA-21 (miR-21), which regulates autophagy during cancer radiation therapy, was identified as a potential therapeutic target for keloids. Here, we investigate the regulatory mechanism(s) of miR-21-5p on keloid fibroblast autophagy and migration after EB irradiation. The microRNA expression profile of the keloid dermis was examined by performing a microRNA microarray. Levels of LC3B and Beclin-1 were detected by immunohistochemical and western blot analysis in the keloid dermis and fibroblasts. Autophagy and apoptosis were tested in keloid fibroblasts after EB irradiation or transfection with an miR-21-5p inhibitor using electron microscopy, a Cyto-ID Green Autophagy Detection Kit, and an Annexin V PE Apoptosis Detection Kit. Migration was analyzed by an in vitro scratch-wound healing assay. Mechanistic tests were performed using small interfering RNAs to phosphatase and tensin homolog (siPTEN). Levels of miR-21-5p, PTEN, programmed cell death 4 (PDCD4), p-AKT, and apoptosis- and autophagy-associated genes were examined by qRT-PCR and western blotting. LC3B expression and migration ability were enhanced in fibroblasts and the keloid margin dermis compared with those in the adjacent normal skin. Both EB irradiation and an miR-21-5p inhibitor reduced keloid fibroblast autophagy, which was accompanied by decreased expression of miR-21-5p, p-AKT, and LC3B-II and increased expression of PTEN, PDCD4, and apoptosis-related genes. MiR-21-5p downregulation inhibited migration and suppressed LC3B expression and this was reversed by PTEN reduction. In conclusion, with increasing apoptosis, EB irradiation inhibits autophagy in keloid fibroblasts by reducing miR-21-5p, which regulates migration and LC3B expression via PTEN/AKT signaling. These data suggest a potential mechanism wherein miR-21-5p inhibition regulates autophagy and migration in EB-irradiated keloid fibroblasts, effectively preventing local invasion and recurrence. Therefore, miR-21-5p could be a new therapeutic target, to replace EB irradiation, and control keloid relapse.
Collapse
Affiliation(s)
- Li Yan
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Lian-Zhao Wang
- Comprehensive Treatment Center of Scar, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Rui Cao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Bo Pan
- Auricular Plastic and Reconstructive Surgery Center, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Xiao-Yan Lv
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Hu Jiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Qiang Zhuang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Xue-Jian Sun
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China
| | - Yuan-Bo Liu
- Department of Plastic and Reconstructive Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Ba-Da-Chu Road 33#, 100144, Beijing, P.R. China.
| |
Collapse
|
15
|
Tu T, Huang J, Lin M, Gao Z, Wu X, Zhang W, Zhou G, Wang W, Liu W. CUDC‑907 reverses pathological phenotype of keloid fibroblasts in vitro and in vivo via dual inhibition of PI3K/Akt/mTOR signaling and HDAC2. Int J Mol Med 2019; 44:1789-1800. [PMID: 31545402 PMCID: PMC6777681 DOI: 10.3892/ijmm.2019.4348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/02/2019] [Indexed: 02/06/2023] Open
Abstract
Keloids are benign skin tumors with a high recurrence rate following surgical excision. Abnormal intracellular signaling is one of the key mechanisms involved in its pathogenesis. Over-activated phosphoinositide 3-kinase/RAC-alpha serine/threonine-protein kinase/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway and overproduction of histone deacetylases 2 (HDAC2) have also been observed in keloid fibroblasts (KFs). The present study aimed to explore the possibility of reversing the KF pathological phenotype using CUDC-907, a dual inhibitor of PI3K/Akt/mTOR pathway and HDACs. KFs and keloid xenografts were treated with CUDC-907 to examine its inhibitory effects on the pathological activities of KFs in vitro and in vivo. CUDC-907 inhibited cell proliferation, migration, invasion and extracellular matrix deposition of in vitro cultured KFs and also suppressed collagen accumulation and disrupted the capillaries of keloid explants ex vivo and in vivo. A mechanistic study of CUDC-907 revealed the initiation of cell cycle arrest at G2/M phase along with the enhanced expression of cyclin-dependent kinase inhibitor 1 and decreased expression of cyclin B in cells treated with CUDC-907. CUDC-907 not only inhibited AKT and mTOR phosphorylation and promoted the acetylation of histone H3, but also significantly inhibited the phosphorylation levels of Smad2/3 and Erk. These preclinical data demonstrating its anti-keloid effects suggest that CUDC-907 may represent a candidate drug for systemic keloid therapy.
Collapse
Affiliation(s)
- Tian Tu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Jia Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Miaomiao Lin
- Department of Otolaryngology, Suzhou First People's Hospital, Suzhou, Anhui 234000, P.R. China
| | - Zhen Gao
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Xiaoli Wu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wenjie Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Guangdong Zhou
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wenbo Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| | - Wei Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, P.R. China
| |
Collapse
|
16
|
Ud-Din S, Foden P, Mazhari M, Al-Habba S, Baguneid M, Bulfone-Paus S, McGeorge D, Bayat A. A Double-Blind, Randomized Trial Shows the Role of Zonal Priming and Direct Topical Application of Epigallocatechin-3-Gallate in the Modulation of Cutaneous Scarring in Human Skin. J Invest Dermatol 2019; 139:1680-1690.e16. [DOI: 10.1016/j.jid.2019.01.030] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 01/13/2019] [Accepted: 01/14/2019] [Indexed: 01/19/2023]
|
17
|
Coentro JQ, Pugliese E, Hanley G, Raghunath M, Zeugolis DI. Current and upcoming therapies to modulate skin scarring and fibrosis. Adv Drug Deliv Rev 2019; 146:37-59. [PMID: 30172924 DOI: 10.1016/j.addr.2018.08.009] [Citation(s) in RCA: 129] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 07/08/2018] [Accepted: 08/26/2018] [Indexed: 12/12/2022]
Abstract
Skin is the largest organ of the human body. Being the interface between the body and the outer environment, makes it susceptible to physical injury. To maintain life, nature has endowed skin with a fast healing response that invariably ends in the formation of scar at the wounded dermal area. In many cases, skin remodelling may be impaired, leading to local hypertrophic scars or keloids. One should also consider that the scarring process is part of the wound healing response, which always starts with inflammation. Thus, scarring can also be induced in the dermis, in the absence of an actual wound, during chronic inflammatory processes. Considering the significant portion of the population that is subject to abnormal scarring, this review critically discusses the state-of-the-art and upcoming therapies in skin scarring and fibrosis.
Collapse
Affiliation(s)
- João Q Coentro
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Eugenia Pugliese
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Geoffrey Hanley
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland
| | - Michael Raghunath
- Center for Cell Biology and Tissue Engineering, Institute for Chemistry and Biotechnology (ICBT), Zurich University of Applied Sciences (ZHAW), Wädenswil, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland; Science Foundation Ireland (SFI), Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI, Galway), Galway, Ireland.
| |
Collapse
|
18
|
Chen J, Liu K, Liu Y, Wang X, Zhang Z. Targeting mTORC1/2 with OSI-027 inhibits proliferation and migration of keloid keratinocytes. Exp Dermatol 2019; 28:270-275. [PMID: 30650200 DOI: 10.1111/exd.13882] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 01/02/2019] [Accepted: 01/11/2019] [Indexed: 12/14/2022]
Abstract
Keloid is a dermal proliferative disorder characterized by the excessive proliferation and migration of keratinocytes and fibroblasts. Over-activation of the serine/threonine protein kinase, mammalian target of rapamycin (mTOR), plays a pivotal role in the process. Here, we show that both mTOR complex 1 (mTORC1) and mTOR complex 2 (mTORC2) were hyper-activated in keloid-derived primary keratinocytes. Further, OSI-027, an mTOR kinase inhibitor, potently inhibited proliferation and migration of keloid keratinocytes. At the molecular level, OSI-027 disrupted the assembly of mTORC1 (mTOR-Raptor) and mTORC2 (mTOR-Rictor-mLST8). Further, OSI-027 almost completely blocked the phosphorylation of the mTORC1 substrates, S6K1, S6 and 4EBP1, and the mTORC2 substrate, AKT, at Ser-473. The OSI-027 treatment of keloid keratinocytes showed more effectively inhibited cell proliferation and migration compared to the mTORC1 inhibitor, rapamycin. Moreover, restoring mTORC1 activation by the introduction of the constitutively active S6K1 only partly alleviated OSI-027-induced inhibition of keloid keratinocytes. Notably, mTOR2 inhibition by Rictor siRNAs also inhibited keloid keratinocyte proliferation and migration, but less efficiently than OSI-027. Together, our results imply that concurrent targeting of mTORC1/2 by OSI-027 potently inhibits the proliferation and the migration of keloid keratinocytes. Thus, OSI-027 may have translational value for the treatment of keloid.
Collapse
Affiliation(s)
- Jun Chen
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Center for Specialty Strategy Research of Shanghai, JiaoTong University China Hospital Development Institute, Shanghai, China
| | - Ke Liu
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Center for Specialty Strategy Research of Shanghai, JiaoTong University China Hospital Development Institute, Shanghai, China
| | - Yang Liu
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Center for Specialty Strategy Research of Shanghai, JiaoTong University China Hospital Development Institute, Shanghai, China
| | - Xue Wang
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Center for Specialty Strategy Research of Shanghai, JiaoTong University China Hospital Development Institute, Shanghai, China
| | - Zhen Zhang
- Department of Dermatology and Dermatologic Surgery, Shanghai Ninth People's Hospital, Affiliated to Shanghai Jiaotong University School of Medicine, Center for Specialty Strategy Research of Shanghai, JiaoTong University China Hospital Development Institute, Shanghai, China
| |
Collapse
|
19
|
Qin D, Lee WH, Gao Z, Zhang W, Peng M, Sun T, Gao Y. Protective effects of antioxidin-RL from Odorrana livida against ultraviolet B-irradiated skin photoaging. Peptides 2018; 101:124-134. [PMID: 29341894 DOI: 10.1016/j.peptides.2018.01.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 01/11/2018] [Accepted: 01/12/2018] [Indexed: 01/14/2023]
Abstract
The unavoidable daily exposure of the skin to ultraviolet (UV) B radiation is proven to have deleterious effects. The action mechanism of antioxidin-RL, an antioxidant peptide purified from skin secretions of frog Odorrana livida with amino acid sequence of AMRLTYNRPCIYAT, is well characterized by NMR titration and mutation based on ABTS+ scavenging activities. In order to explore the protective effects of antioxidin-RL against UVB-irradiated skin photoaging, cell uptake assay was used to detect the location of antioxidin-RL molecules serving various biological functions in the cells. The protective effects of antioxidin-RL on UVB-induced response were examined in vitro and in vivo. Results showed that antioxidin-RL successfully penetrated the cell membrane and exerted a positive effect on cell migration. It also effectively inhibited the UVB-induced excessive production of ROS and prevented oxidative damage to DNAs and proteins. Moreover, the mRNA expressions of MMP-1, VEGF, COX-2, and pro-inflammatory cytokines, such as IL-6 and TNF-α in antioxidin-RL-treated HaCaT and HSF cells were significantly down-regulated whereas those of FGF, procollagen type I and TGF-β1 up-regulated. Antioxidin-RL effectively prevented UVB-induced erythema on mouse skin, thereby inhibiting UVB-induced skin thickening and inflammation and increasing collagen deposition as demonstrated by in vivo experiments. Hence, the novel antioxidant peptide antioxidin-RL can effectively reduce UVB-induced skin reactions in vivo and in vitro, providing potential molecules against UVB-induced inflammation and photoaging.
Collapse
Affiliation(s)
- Di Qin
- Key Laboratory of Biological Medicine in Universities of Shandong Province, School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Wen-Hui Lee
- Key Laboratory of Biological Medicine in Universities of Shandong Province, School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, Shandong, China; Key Laboratory of Bioactive Peptides of Yunnan Province, Kunming Institute of Zoology, Kunming, 650223, Yunnan, China; Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese Academy of Sciences, China
| | - Zhiqin Gao
- Key Laboratory of Biological Medicine in Universities of Shandong Province, School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Weifen Zhang
- School of Pharmacy, Weifang Medical University, Baotong Road, Weifang, 261053, Shandong, China
| | - Meiyu Peng
- School of Clinical Medicine, Weifang Medical University, Weifang, 261053, Shandong, China
| | - Tongyi Sun
- Key Laboratory of Biological Medicine in Universities of Shandong Province, School of Bioscience and Technology, Weifang Medical University, Weifang, 261053, Shandong, China.
| | - Yuanyuan Gao
- School of Pharmacy, Weifang Medical University, Baotong Road, Weifang, 261053, Shandong, China.
| |
Collapse
|
20
|
Long non-coding RNA expression profiling in the lesional tissue and derived fibroblasts of keloid. Postepy Dermatol Alergol 2017; 34:587-600. [PMID: 29422825 PMCID: PMC5799763 DOI: 10.5114/ada.2017.72466] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Accepted: 01/12/2017] [Indexed: 12/21/2022] Open
Abstract
Introduction Long non-coding RNA (lncRNA) plays a key role in various disorders. However, its role in keloid is still unclear. Aim We explored differentially expressed (DE) lncRNAs and mRNAs between keloid tissue (KT)s and normal tissue (NT)s, as well as keloid fibroblast (KFB)s and normal fibroblast (NFB)s, respectively. Material and methods We use KTs and NTs from the chest of 5 patients, and 3 pairs of KFBs and NFBs, to perform microarray respectively. Gene ontology and pathway analyses were conducted by online software DAVID (Database for Annotation, Visualization and Integrated Discovery). The validation of targeted lncRNAs were conducted by qRT-PCR in enlarged samples (79 KTs and 21 NTs). Results We identified 3680 DE-lncRNAs in tissue essay, and 1231 DE-lncRNAs in cell essay. Furthermore, we found that many lncRNAs and their relative mRNAs were regulated simultaneously in keloid. We identified that ENST00000439703 and uc003jox.1 were up-regulated in both of the above essays through comparing the results of lncRNA screening between tissue essay and cell essay; the results were confirmed through qRT-PCR in enlarged samples. Conclusions Our study demonstrates that numerous lncRNAs are involved in the pathogenesis and development of the keloid.
Collapse
|
21
|
Abstract
BACKGROUND Keloid and hypertrophic scars represent an aberrant response to the wound healing process. These scars are characterized by dysregulated growth with excessive collagen formation, and can be cosmetically and functionally disruptive to patients. OBJECTIVE Objectives are to describe the pathophysiology of keloid and hypertrophic scar, and to compare differences with the normal wound healing process. The classification of keloids and hypertrophic scars are then discussed. Finally, various treatment options including prevention, conventional therapies, surgical therapies, and adjuvant therapies are described in detail. MATERIALS AND METHODS Literature review was performed identifying relevant publications pertaining to the pathophysiology, classification, and treatment of keloid and hypertrophic scars. RESULTS Though the pathophysiology of keloid and hypertrophic scars is not completely known, various cytokines have been implicated, including interleukin (IL)-6, IL-8, and IL-10, as well as various growth factors including transforming growth factor-beta and platelet-derived growth factor. Numerous treatments have been studied for keloid and hypertrophic scars,which include conventional therapies such as occlusive dressings, compression therapy, and steroids; surgical therapies such as excision and cryosurgery; and adjuvant and emerging therapies including radiation therapy, interferon, 5-fluorouracil, imiquimod, tacrolimus, sirolimus, bleomycin, doxorubicin, transforming growth factor-beta, epidermal growth factor, verapamil, retinoic acid, tamoxifen, botulinum toxin A, onion extract, silicone-based camouflage, hydrogel scaffold, and skin tension offloading device. CONCLUSION Keloid and hypertrophic scars remain a challenging condition, with potential cosmetic and functional consequences to patients. Several therapies exist which function through different mechanisms. Better understanding into the pathogenesis will allow for development of newer and more targeted therapies in the future.
Collapse
|
22
|
Mlitz V, Gendronneau G, Berlin I, Buchberger M, Eckhart L, Tschachler E. The Expression of the Endogenous mTORC1 Inhibitor Sestrin 2 Is Induced by UVB and Balanced with the Expression Level of Sestrin 1. PLoS One 2016; 11:e0166832. [PMID: 27861561 PMCID: PMC5115827 DOI: 10.1371/journal.pone.0166832] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Accepted: 11/05/2016] [Indexed: 01/28/2023] Open
Abstract
Sestrin 2 (SESN2) is an evolutionarily conserved regulator of mechanistic target of rapamycin complex 1 (mTORC1) which controls central cellular processes such as protein translation and autophagy. Previous studies have suggested that SESN2 itself is subjected to regulation at multiple levels. Here, we investigated the expression of SESN2 in the skin and in isolated skin cells. SESN2 was detected by immunofluorescence analysis in fibroblasts and keratinocytes of human skin. Differentiation of epidermal keratinocytes was not associated with altered SESN2 expression and siRNA-mediated knockdown of SESN2 did not impair stratum corneum formation in vitro. However, SESN2 was increased in both cell types when the expression of its paralog SESN1 was blocked by siRNA-mediated knock down, indicating a compensatory mechanism for the control of expression. Irradiation with UVB but not with UVA significantly increased SESN2 expression in both keratinocytes and fibroblasts. Upregulation of SESN2 expression could be completely blocked by suppression of p53. These results suggest that SESN2 is dispensable for normal epidermal keratinization but involved in the UVB stress response of skin cells.
Collapse
Affiliation(s)
- Veronika Mlitz
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | | | - Irina Berlin
- Department of Biology and Women Beauty, Chanel R&T, Pantin, France
| | - Maria Buchberger
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Leopold Eckhart
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
- * E-mail: (ET); (LE)
| | - Erwin Tschachler
- Research Division of Biology and Pathobiology of the Skin, Department of Dermatology, Medical University of Vienna, Vienna, Austria
- * E-mail: (ET); (LE)
| |
Collapse
|
23
|
Georgiadis C, Syed F, Petrova A, Abdul-Wahab A, Lwin SM, Farzaneh F, Chan L, Ghani S, Fleck RA, Glover L, McMillan JR, Chen M, Thrasher AJ, McGrath JA, Di WL, Qasim W. Lentiviral Engineered Fibroblasts Expressing Codon-Optimized COL7A1 Restore Anchoring Fibrils in RDEB. J Invest Dermatol 2016; 136:284-92. [PMID: 26763448 PMCID: PMC4759620 DOI: 10.1038/jid.2015.364] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Revised: 07/27/2015] [Accepted: 08/03/2015] [Indexed: 01/06/2023]
Abstract
Cells therapies, engineered to secrete replacement proteins, are being developed to ameliorate otherwise debilitating diseases. Recessive dystrophic epidermolysis bullosa (RDEB) is caused by defects of type VII collagen, a protein essential for anchoring fibril formation at the dermal-epidermal junction. Whereas allogeneic fibroblasts injected directly into the dermis can mediate transient disease modulation, autologous gene-modified fibroblasts should evade immunological rejection and support sustained delivery of type VII collagen at the dermal-epidermal junction. We demonstrate the feasibility of such an approach using a therapeutic grade, self-inactivating-lentiviral vector, encoding codon-optimized COL7A1, to transduce RDEB fibroblasts under conditions suitable for clinical application. Expression and secretion of type VII collagen was confirmed with transduced cells exhibiting supranormal levels of protein expression, and ex vivo migration of fibroblasts was restored in functional assays. Gene-modified RDEB fibroblasts also deposited type VII collagen at the dermal-epidermal junction of human RDEB skin xenografts placed on NOD-scid IL2Rgamma(null) recipients, with reconstruction of human epidermal structure and regeneration of anchoring fibrils at the dermal-epidermal junction. Fibroblast-mediated restoration of protein and structural defects in this RDEB model strongly supports proposed therapeutic applications in man.
Collapse
Affiliation(s)
- Christos Georgiadis
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Farhatullah Syed
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Anastasia Petrova
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Alya Abdul-Wahab
- St John's Institute of Dermatology, King's College London (Guy's campus), London, United Kingdom
| | - Su M Lwin
- St John's Institute of Dermatology, King's College London (Guy's campus), London, United Kingdom
| | - Farzin Farzaneh
- Department of Haematological Medicine, King's College London, The Rayne Institute, London, United Kingdom
| | - Lucas Chan
- Department of Haematological Medicine, King's College London, The Rayne Institute, London, United Kingdom
| | - Sumera Ghani
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Roland A Fleck
- Centre for Ultrastructural Imaging, King's College London, London, United Kingdom
| | - Leanne Glover
- Centre for Ultrastructural Imaging, King's College London, London, United Kingdom
| | - James R McMillan
- The Robin Eady National Diagnostic Epidermolysis Bullosa Laboratory, Viapath LLP, St Thomas' Hospital, London, United Kingdom
| | - Mei Chen
- Department of Dermatology, University of Southern California, Los Angeles, California, USA
| | - Adrian J Thrasher
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - John A McGrath
- St John's Institute of Dermatology, King's College London (Guy's campus), London, United Kingdom
| | - Wei-Li Di
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Waseem Qasim
- UCL Institute of Child Health, Molecular and Cellular Immunology Section & Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom.
| |
Collapse
|
24
|
Cordaro M, Paterniti I, Siracusa R, Impellizzeri D, Esposito E, Cuzzocrea S. KU0063794, a Dual mTORC1 and mTORC2 Inhibitor, Reduces Neural Tissue Damage and Locomotor Impairment After Spinal Cord Injury in Mice. Mol Neurobiol 2016; 54:2415-2427. [PMID: 26960330 DOI: 10.1007/s12035-016-9827-0] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Accepted: 03/03/2016] [Indexed: 10/22/2022]
Abstract
Autophagy is an intracellular catabolic mechanism for the degradation of cytoplasmic constituents in the autophagosomal-lysosomal pathway. This mechanism plays an important role in homeostasis and it is defective in certain diseases. Preceding studies have revealed that autophagy is developing as an important moderator of pathological responses associated to spinal cord injury (SCI) and plays a crucial role in secondary injury initiating a progressive degeneration of the spinal cord. Thus, based on this evidence in this study, we used two different selective inhibitors of mTOR activity to explore the functional role of autophagy in an in vivo model of SCI as well as to determine whether the autophagic process is involved in spinal cord tissue damage. We treated animals with a novel synthetic inhibitor temsirolimus and with a dual mTORC1 and mTORC2 inhibitor KU0063794 matched all with the well-known inhibitor of mTOR the rapamycin. Our results demonstrated that mTOR inhibitors could regulate the neuroinflammation associated to SCI and the results that we obtained evidently demonstrated that rapamycin and temsirolimus significantly diminished the expression of iNOS, COX2, GFAP, and re-established nNOS levels, but the administration of KU0063794 is able to blunt the neuroinflammation better than rapamycin and temsirolimus. In addition, neuronal loss and cell mortality in the spinal cord after injury were considerably reduced in the KU0063794-treated mice. Accordingly, taken together our results denote that the administration of KU0063794 produced a neuroprotective function at the lesion site following SCI, representing a novel therapeutic approach after SCI.
Collapse
Affiliation(s)
- Marika Cordaro
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Irene Paterniti
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Rosalba Siracusa
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Daniela Impellizzeri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Emanuela Esposito
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy
| | - Salvatore Cuzzocrea
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno D'Alcontres, 31, 98166, Messina, Italy. .,Department of Pharmacological and Physiological Science, Saint Louis University, Saint Louis, MO, USA.
| |
Collapse
|
25
|
Oza VS, Mamlouk MD, Hess CP, Mathes EF, Frieden IJ. Role of Sirolimus in Advanced Kaposiform Hemangioendothelioma. Pediatr Dermatol 2016; 33:e88-92. [PMID: 26864138 DOI: 10.1111/pde.12787] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Kaposiform hemangioendothelioma (KHE) is an infiltrative vascular tumor that classically presents in infancy. Management typically focuses on treating Kasabach-Merritt phenomenon (KMP), a disorder of severe and at times life-threatening platelet trapping. However, the morbidity of KHE extends beyond KMP. The infiltrative nature of the tumor can lead to long-term disability and often makes complete surgical resection impossible. We report the case of a 10-year-old boy with a KHE of his right distal thigh who was unable to walk without assistance due to fibrotic change and right knee contracture. He had no laboratory evidence of KMP at the time of representation. Rapamycin was started in hopes of reducing the tumor burden. Within 2 months of therapy, fibrotic areas softened, his contracture nearly resolved, and there was marked improvement in his mobility. Rapamycin has been previously reported to be effective in managing cases of KHE complicated by KMP. Our report emphasizes the role for rapamycin in the treatment of KHE in the absence of KMP through the inhibition of vasculogenesis and fibrotic pathways.
Collapse
Affiliation(s)
- Vikash S Oza
- Ronald O. Perelman Department of Dermatology, New York University School of Medicine, New York, New York
| | - Mark D Mamlouk
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Christopher P Hess
- Department of Radiology, University of California, San Francisco, San Francisco, California
| | - Erin F Mathes
- Department of Dermatology, University of California, San Francisco, San Francisco, California
| | - Ilona J Frieden
- Department of Dermatology, University of California, San Francisco, San Francisco, California
| |
Collapse
|
26
|
Yamane T, Muramatsu A, Yoshino S, Matsui S, Shimura M, Tsujii Y, Iwatsuki K, Kobayashi-Hattori K, Oishi Y. mTOR inhibition by rapamycin increases ceramide synthesis by promoting transforming growth factor-β1/Smad signaling in the skin. FEBS Open Bio 2016; 6:317-25. [PMID: 27239444 PMCID: PMC4821357 DOI: 10.1002/2211-5463.12039] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 01/27/2016] [Accepted: 01/27/2016] [Indexed: 01/10/2023] Open
Abstract
Although mammalian target of rapamycin (mTOR) mediates a wide variety of biological functions, little information is available on the effect of mTOR on the functions of skin cells. In this study, we investigated effects of mTOR inhibition by rapamycin on ceramide synthesis in the skin of rats and human keratinocytes and its regulatory mechanisms. The phosphorylation of p70 S6 kinase, which indicates mTOR activation, was induced in the skin of rats fed a high-fat diet, but this abnormality was reversed by supplementation with rapamycin. Ceramide levels and the mRNA levels of serine palmitoyltransferase (SPT) and transforming growth factor (TGF)-β1 were suppressed in the skin of rats fed high-fat diets, but this abnormality was reversed by supplementation with rapamycin. TGF-β1-induced SPT mRNA expression was blocked by SB525334, an inhibitor of TGF-β1-induced Smad2/3 nuclear localization, in human keratinocytes. Rapamycin-induced SPT mRNA expression was blocked by an anti-TGF-β1 antibody or SB525334 in human keratinocytes. These results show that mTOR inhibition by rapamycin increases ceramide synthesis by promoting TGF-β1/Smad signaling in the skin.
Collapse
Affiliation(s)
- Takumi Yamane
- Department of Nutritional Science and Food Safety Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Aimi Muramatsu
- Department of Nutritional Science Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Sawako Yoshino
- Department of Nutritional Science and Food Safety Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Sho Matsui
- Department of Nutritional Science Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Mari Shimura
- Department of Nutritional Science and Food Safety Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Yoshimasa Tsujii
- Department of Applied Biology and Chemistry Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Ken Iwatsuki
- Department of Nutritional Science and Food Safety Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Kazuo Kobayashi-Hattori
- Department of Nutritional Science Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| | - Yuichi Oishi
- Department of Nutritional Science and Food Safety Faculty of Applied Bioscience Tokyo University of Agriculture Setagaya-ku Japan
| |
Collapse
|
27
|
Increased c-Met phosphorylation is related to keloid pathogenesis: implications for the biological behaviour of keloid fibroblasts. Pathology 2015; 46:25-31. [PMID: 24300717 DOI: 10.1097/pat.0000000000000028] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Keloid is induced by a pathological wound healing response, and hepatocyte growth factor (HGF) is known to be involved in tissue repair via the activation of its primary receptor, c-Met. We aimed to investigate whether c-Met activation is implicated in keloid pathogenesis. HGF, c-Met, phosphorylated c-Met (p-Met), Ki-67, collagen I protein, and MET gene expression were detected in five normal skin and 30 keloid tissues by immunohistochemistry and quantitative real-time polymerase chain reaction analysis, respectively. The influence of p-Met expression on the biological behaviour of keloid fibroblasts was further investigated with regard to cell proliferation, motility, invasiveness, collagen I expression, and intracellular signaling in vitro. p-Met protein and MET gene expression but not HGF or c-Met protein expression showed significant increases in keloid tissues than dermal layer of normal skin tissues. In keloid tissues, p-Met expression was significantly associated with keloid size, Ki-67 and collagen I expression. Moreover, p-Met expression was also related to proliferation, migration, invasiveness, collagen I expression and activation of AKT and Erk in keloid fibroblasts in vitro. c-Met activation may have a strong influence on keloid pathogenesis, and it can be investigated further as a potential molecular target for keloid therapy.
Collapse
|
28
|
Perl A. mTOR activation is a biomarker and a central pathway to autoimmune disorders, cancer, obesity, and aging. Ann N Y Acad Sci 2015; 1346:33-44. [PMID: 25907074 DOI: 10.1111/nyas.12756] [Citation(s) in RCA: 169] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The mechanistic target of rapamycin (mTOR) is a ubiquitous serine/threonine kinase, which plays pivotal roles in integrating growth signals on a cellular level. To support proliferation and survival under stress, two interacting complexes that harbor mTOR, mTORC1 and mTORC2, promote the transcription of genes involved in carbohydrate metabolism and lipogenesis, enhance protein translation, and inhibit autophagy. Although rapamycin was originally developed as an inhibitor of T cell proliferation for preventing organ transplant rejection, its molecular target, mTOR, has been subsequently identified as a central regulator of metabolic cues that drive lineage specification in the immune system. Owing to oxidative stress, the activation of mTORC1 has emerged as a central pathway for the pathogenesis of systemic lupus erythematosus and other autoimmune diseases. Paradoxically, mTORC1 has also been identified as a mediator of the Warburg effect that allows cell survival under hypoxia. Rapamycin and new classes of mTOR inhibitors are being developed to block not only transplant rejection and autoimmunity but also to treat obesity and various forms of cancer. Through preventing these diseases, personalized mTOR blockade holds promise to extend life span.
Collapse
Affiliation(s)
- Andras Perl
- Division of Rheumatology, Department of Medicine State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York.,Division of Rheumatology, Department of Microbiology and Immunology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York
| |
Collapse
|
29
|
Andreoli A, Ruf M, Itin P, Pluschke G, Schmid P. Phosphorylation of the ribosomal protein S6, a marker of mTOR (mammalian target of rapamycin) pathway activation, is strongly increased in hypertrophic scars and keloids. Br J Dermatol 2015; 172:1415-7. [DOI: 10.1111/bjd.13523] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Affiliation(s)
- A. Andreoli
- Swiss Tropical and Public Health Institute; Socinstr. 57 Basel Switzerland
- University of Basel; Petersplatz 1 Basel Switzerland
| | - M.T. Ruf
- Swiss Tropical and Public Health Institute; Socinstr. 57 Basel Switzerland
- University of Basel; Petersplatz 1 Basel Switzerland
| | - P. Itin
- Department of Dermatology; University Hospital; Basel Switzerland
| | - G. Pluschke
- Swiss Tropical and Public Health Institute; Socinstr. 57 Basel Switzerland
- University of Basel; Petersplatz 1 Basel Switzerland
| | - P. Schmid
- Swiss Tropical and Public Health Institute; Socinstr. 57 Basel Switzerland
- University of Basel; Petersplatz 1 Basel Switzerland
| |
Collapse
|
30
|
Fogel AL, Hill S, Teng JMC. Advances in the therapeutic use of mammalian target of rapamycin (mTOR) inhibitors in dermatology. J Am Acad Dermatol 2015; 72:879-89. [PMID: 25769191 DOI: 10.1016/j.jaad.2015.01.014] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Revised: 01/07/2015] [Accepted: 01/09/2015] [Indexed: 12/31/2022]
Abstract
Significant developments in the use of mammalian target of rapamycin (mTOR) inhibitors (mTORIs) as immunosuppressant and antiproliferative agents have been made. Recent advances in the understanding of the mTOR signaling pathway and its downstream effects on tumorigenesis and vascular proliferation have broadened the clinical applications of mTORIs in many challenging disorders such as tuberous sclerosis complex, pachyonychia congenita, complex vascular anomalies, and inflammatory dermatoses. Systemic mTORI therapy has shown benefits in these areas, but is associated with significant side effects that sometimes necessitate drug holidays. To mitigate the side effects of systemic mTORIs for dermatologic applications, preliminary work to assess the potential of percutaneous therapy has been performed, and the evidence suggests that percutaneous delivery of mTORIs may allow for effective long-term therapy while avoiding systemic toxicities. Additional large placebo-controlled, double-blinded, randomized studies are needed to assess the efficacy, safety, duration, and tolerability of topical treatments. The objective of this review is to provide updated information on the novel use of mTORIs in the management of many cutaneous disorders.
Collapse
Affiliation(s)
| | | | - Joyce M C Teng
- Dermatology, Stanford University School of Medicine, Palo Alto, California; Pediatrics, Stanford University School of Medicine, Palo Alto, California; Pediatric Dermatology, Stanford University School of Medicine, Palo Alto, California; Stanford University School of Medicine, Palo Alto, California.
| |
Collapse
|
31
|
Yang F, Tanaka M, Wataya-Kaneda M, Yang L, Nakamura A, Matsumoto S, Attia M, Murota H, Katayama I. Topical application of rapamycin ointment ameliorates Dermatophagoides farina body extract-induced atopic dermatitis in NC/Nga mice. Exp Dermatol 2014; 23:568-72. [PMID: 24903639 DOI: 10.1111/exd.12463] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/03/2014] [Indexed: 12/24/2022]
Abstract
Atopic dermatitis (AD), a chronic inflammatory skin disease characterized by relapsing eczema and intense prurigo, requires effective and safe pharmacological therapy. Recently, rapamycin, an mTOR (mammalian target of rapamycin) inhibitor, has been reported to play a critical role in immune responses and has emerged as an effective immunosuppressive drug. In this study, we assessed whether inhibition of mTOR signalling could suppress dermatitis in mice. Rapamycin was topically applied to inflamed skin in a murine AD model that was developed by repeated topical application of Dermatophagoides farina body (Dfb) extract antigen twice weekly for 7 weeks in NC/Nga mice. The efficacy of topical rapamycin treatment was evaluated immunologically and serologically. Topical application of rapamycin reduced inflammatory cell infiltration in the dermis, alleviated the increase of serum IgE levels and resulted in a significant reduction in clinical skin condition score and marked improvement of histological findings. In addition, increased mTOR phosphorylation in the lesional skin was observed in our murine AD model. Topical application of rapamycin ointment inhibited Dfb antigen-induced dermatitis in NC/Nga mice, promising a new therapy for atopic dermatitis.
Collapse
Affiliation(s)
- Fei Yang
- Department of Dermatology, Course of Integrated Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Ashcroft KJ, Syed F, Bayat A. Site-specific keloid fibroblasts alter the behaviour of normal skin and normal scar fibroblasts through paracrine signalling. PLoS One 2013; 8:e75600. [PMID: 24348987 PMCID: PMC3857170 DOI: 10.1371/journal.pone.0075600] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2013] [Accepted: 08/19/2013] [Indexed: 01/31/2023] Open
Abstract
Keloid disease (KD) is an abnormal cutaneous fibroproliferative disorder of unknown aetiopathogenesis. Keloid fibroblasts (KF) are implicated as mediators of elevated extracellular matrix deposition. Aberrant secretory behaviour by KF relative to normal skin fibroblasts (NF) may influence the disease state. To date, no previous reports exist on the ability of site-specific KF to induce fibrotic-like phenotypic changes in NF or normal scar fibroblasts (NS) by paracrine mechanisms. Therefore, the aim of this study was to investigate the influence of conditioned media from site-specific KF on the cellular and molecular behaviour of both NF and NS enabled by paracrine mechanisms. Conditioned media was collected from cultured primary fibroblasts during a proliferative log phase of growth including: NF, NS, peri-lesional keloid fibroblasts (PKF) and intra-lesional keloid fibroblasts (IKF). Conditioned media was used to grow NF, NS, PKF and IKF cells over 240 hrs. Cellular behavior was monitored through real time cell analysis (RTCA), proliferation rates and migration in a scratch wound assay. Fibrosis-associated marker expression was determined at both protein and gene level. PKF conditioned media treatment of both NF and NS elicited enhanced cell proliferation, spreading and viability as measured in real time over 240 hrs versus control conditioned media. Following PKF and IKF media treatments up to 240 hrs, both NF and NS showed significantly elevated proliferation rates (p<0.03) and migration in a scratch wound assay (p<0.04). Concomitant up-regulation of collagen I, fibronectin, α-SMA, PAI-1, TGF-β and CTGF (p<0.03) protein expression were also observed. Corresponding qRT-PCR analysis supported these findings (P<0.03). In all cases, conditioned media from growing marginal PKF elicited the strongest effects. In conclusion, primary NF and NS cells treated with PKF or IKF conditioned media exhibit enhanced expression of fibrosis-associated molecular markers and increased cellular activity as a result of keloid fibroblast-derived paracrine factors.
Collapse
Affiliation(s)
- Kevin J. Ashcroft
- Plastic & Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, United Kingdom
- Institute of Inflammation & Repair, University of Manchester, Manchester, United Kingdom
| | - Farhatullah Syed
- Plastic & Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, United Kingdom
- Institute of Inflammation & Repair, University of Manchester, Manchester, United Kingdom
| | - Ardeshir Bayat
- Plastic & Reconstructive Surgery Research, Manchester Institute of Biotechnology, University of Manchester, United Kingdom
- Institute of Inflammation & Repair, University of Manchester, Manchester, United Kingdom
- Department of Plastic and Reconstructive Surgery, University Hospital South Manchester Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom
- The University of Manchester, Manchester Academic Health Science Centre, University Hospital South Manchester Foundation Trust, Wythenshawe Hospital, Manchester, United Kingdom
- * E-mail:
| |
Collapse
|
33
|
Montané MH, Menand B. ATP-competitive mTOR kinase inhibitors delay plant growth by triggering early differentiation of meristematic cells but no developmental patterning change. JOURNAL OF EXPERIMENTAL BOTANY 2013; 64:4361-74. [PMID: 23963679 PMCID: PMC3808319 DOI: 10.1093/jxb/ert242] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
The TOR (target of rapamycin) protein, a large phosphatidylinositol 3-kinase-like protein kinase (PIKK) that is conserved in eukaryotes and is a central regulator of growth and metabolism. The analysis of function of TOR in plant growth and development has been limited by the fact that plants are very poorly sensitive to rapamycin. As the kinase domain of TOR is highly conserved, this study analysed the dose-dependent effect of three sets of first- and second-generation ATP-competitive inhibitors (called asTORis for active-site TOR inhibitors) recently developed for the human TOR kinase on Arabidopsis thaliana growth. All six asTORis inhibited plant root growth in a dose-dependent manner, with 50% growth inhibitory doses (GI50) of <10 μM and <1 μM for the first- and second-generation inhibitors, respectively, similarly to the values in mammalian cells. A genetic approach further demonstrated that only asTORis inhibited root growth in an AtTOR gene-dosage-dependent manner. AsTORis decreased the length of: (i) the meristematic zone (MZ); (ii) the division zone in the MZ; (iii) epidermal cells in the elongation zone; and (iv) root hair cells. Whereas meristematic cells committed to early differentiation, the pattern of cell differentiation was not affected per se. AsTORis-induced root hair growth phenotype was shown to be specific by using other growth inhibitors blocking the cell cycle or translation. AsTORis dose-dependent inhibition of growth and root hairs was also observed in diverse groups of flowering plants, indicating that asTORis can be used to study the TOR pathway in other angiosperms, including crop plants.
Collapse
Affiliation(s)
- Marie-Hélène Montané
- Aix-Marseille Université, Laboratoire de Génétique et Biophysique des Plantes, Marseille, F-13009, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7265 Biologie Végétale et Microbiologie Environnementales, Marseille, F-13009, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut de Biologie Environnementale et Biotechnologie, Marseille, F-13009, France
| | - Benoît Menand
- Aix-Marseille Université, Laboratoire de Génétique et Biophysique des Plantes, Marseille, F-13009, France
- Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7265 Biologie Végétale et Microbiologie Environnementales, Marseille, F-13009, France
- Commissariat à l’Energie Atomique et aux Energies Alternatives, Institut de Biologie Environnementale et Biotechnologie, Marseille, F-13009, France
- * To whom correspondence should be addressed.
| |
Collapse
|