1
|
Zhang M, Chen H, Qian H, Wang C. Characterization of the skin keloid microenvironment. Cell Commun Signal 2023; 21:207. [PMID: 37587491 PMCID: PMC10428592 DOI: 10.1186/s12964-023-01214-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 07/02/2023] [Indexed: 08/18/2023] Open
Abstract
Keloids are a fibroproliferative skin disorder that develops in people of all ages. Keloids exhibit some cancer-like behaviors, with similar genetic and epigenetic modifications in the keloid microenvironment. The keloid microenvironment is composed of keratinocytes, fibroblasts, myofibroblasts, vascular endothelial cells, immune cells, stem cells and collagen fibers. Recent advances in the study of keloids have led to novel insights into cellular communication among components of the keloid microenvironment as well as potential therapeutic targets for treating keloids. In this review, we summarized the nature of genetic and epigenetic regulation in keloid-derived fibroblasts, epithelial-to-mesenchymal transition of keratinocytes, immune cell infiltration into keloids, the differentiation of keloid-derived stem cells, endothelial-to-mesenchymal transition of vascular endothelial cells, extracellular matrix synthesis and remodeling, and uncontrolled angiogenesis in keloids with the aim of identifying new targets for therapeutic benefit. Video Abstract.
Collapse
Affiliation(s)
- Mengwen Zhang
- The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Hailong Chen
- The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Huan Qian
- The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China
| | - Chen Wang
- The Second Affiliated Hospital of Zhejiang University School of Medicine, 88 Jiefang Road, Hangzhou, 310009, China.
| |
Collapse
|
2
|
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
|
3
|
Qiu ZK, Zhang MZ, Zhang WC, Li ZJ, Si LB, Long X, Yu NZ, Wang XJ. Role of HIF-1α in pathogenic mechanisms of keloids. J Cosmet Dermatol 2023; 22:1436-1448. [PMID: 36718786 DOI: 10.1111/jocd.15601] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 11/08/2022] [Accepted: 12/12/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUDS AND OBJECTIVE Keloids are defined as overrepairing products that develop after skin lesions. Keloids are characterized by the proliferation of fibroblasts and the overaccumulation of extracellular matrix components (mainly collagen), leading to a locally hypoxic microenvironment. Hence, this article was aimed to review hypoxia in pathogenesis of keloids. METHODS We reviewed and summarized the relevant published studies. RESULTS Hypoxia results in the accumulation of hypoxia-inducible factor 1α (HIF-1α) in keloids, contributing to overactivation of the fibrotic signaling pathway, epithelial-mesenchymal transition, and changes in metabolism, eventually leading to aggravated fibrosis, infiltrative growth, and radiotherapy resistance. CONCLUSION It is, therefore, essential to understand the role of HIF-1α in the pathogenic mechanisms of keloids in order to develop new therapeutic approaches.
Collapse
Affiliation(s)
- Zi-Kai Qiu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ming-Zi Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen-Chao Zhang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Jin Li
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lou-Bin Si
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Nan-Ze Yu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao-Jun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical college Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
4
|
Zhang QL, Li P, Hong L, Li RZ, Wang JQ, Cui X. The protein tyrosine kinase inhibitor genistein suppresses hypoxia-induced atrial natriuretic peptide secretion mediated by the PI3K/Akt-HIF-1α pathway in isolated beating rat atria. Can J Physiol Pharmacol 2021; 99:1184-1190. [PMID: 34612711 DOI: 10.1139/cjpp-2020-0503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Genistein, an isoflavonoid that can inhibit protein tyrosine kinase (PTK) phosphorylation, has been shown to play pivotal roles in the signal transduction pathways of hypoxic disorders. In this study, we established a rat model of isolated beating atrium and investigated the regulator role of genistein and its downstream signaling pathways in acute hypoxia-induced atrial natriuretic peptide (ANP) secretion. Radioimmunoassay was used to detect the ANP content in the atrial perfusates. Western blot analysis was used to determine the protein level of hypoxia-inducible factor 1α (HIF-1α), and GATA4 in the atrial tissue. The results showed that acute hypoxia substantially promoted ANP secretion, whereas this effect was partly attenuated by the PTKs inhibitor genistein (3 μM). By Western blotting analysis, we found that hypoxia-induced increase in phosphorylation of Akt and transcriptional factors, including HIF-1α, were also reversed by genistein. The perfused HIF-1α inhibitors rotenone (0.5 μM) or CAY10585 (10 μM) plus genistein significantly abolished the enhanced ANP section induced by hypoxia. Additionally, the perfused PI3K/Akt agonist insulin-like growth factor 1 (30 μM) also abolished ANP secretion induced by genistein and inhibited expression of HIF-1α. In summary, our data suggested that acute hypoxia markedly increased ANP secretion by PTKs through the phosphoinositide-3 kinase (PI3K)/HIF-1α dependent pathway.
Collapse
Affiliation(s)
- Qiu-Li Zhang
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Ping Li
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Lan Hong
- Department of Physiology, School of Medical Sciences, Yanbian University, Yanji, China
| | - Rui-Zhuang Li
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong, China
| | - Jia-Qi Wang
- Affiliated Hospital of Inner Mongolia University for the Nationalities, Tongliao, Neimenggu, China
| | - Xun Cui
- Department of Physiology, School of Medical Sciences, Yanbian University, Yanji, China
- Key Laboratory of Organism Functional Factors of the Changbai Mountain, Ministry of Education, Yanbian University, Yanji, China
- Cellular Function Research Center, Yanbian University, Yanji, China
| |
Collapse
|
5
|
Dorris ER, Russell J, Murphy M. Post-intubation subglottic stenosis: aetiology at the cellular and molecular level. Eur Respir Rev 2021; 30:30/159/200218. [PMID: 33472959 PMCID: PMC9489001 DOI: 10.1183/16000617.0218-2020] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/20/2020] [Indexed: 02/07/2023] Open
Abstract
Subglottic stenosis (SGS) is a narrowing of the airway just below the vocal cords. This narrowing typically consists of fibrotic scar tissue, which may be due to a variety of diseases. This review focuses on post-intubation (PI) SGS. SGS can result in partial or complete narrowing of the airway. This narrowing is caused by fibrosis and can cause serious breathing difficulties. It can occur in both adults and children. The pathogenesis of post-intubation SGS is not well understood; however, it is considered to be the product of an abnormal healing process. This review discusses how intubation can change the local micro-environment, leading to dysregulated tissue repair. We discuss how mucosal inflammation, local hypoxia and biomechanical stress associated with intubation can promote excess tissue deposition that occurs during the pathological process of SGS. COVID-19 may cause an increased incidence of subglottic stenosis (SGS). In this review, the cellular and molecular aetiology of post-intubation SGS is outlined and we discuss how better knowledge of the underlying biology can inform SGS management.https://bit.ly/2RSliRK
Collapse
Affiliation(s)
- Emma R Dorris
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland .,School of Medicine, University College Dublin, Dublin, Ireland
| | - John Russell
- Children's Hospital Ireland Crumlin, Dublin, Ireland
| | - Madeline Murphy
- National Children's Research Centre, Our Lady's Children's Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
6
|
DeFrates KG, Franco D, Heber-Katz E, Messersmith PB. Unlocking mammalian regeneration through hypoxia inducible factor one alpha signaling. Biomaterials 2021; 269:120646. [PMID: 33493769 PMCID: PMC8279430 DOI: 10.1016/j.biomaterials.2020.120646] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 12/19/2020] [Accepted: 12/29/2020] [Indexed: 02/08/2023]
Abstract
Historically, the field of regenerative medicine has aimed to heal damaged tissue through the use of biomaterials scaffolds or delivery of foreign progenitor cells. Despite 30 years of research, however, translation and commercialization of these techniques has been limited. To enable mammalian regeneration, a more practical approach may instead be to develop therapies that evoke endogenous processes reminiscent of those seen in innate regenerators. Recently, investigations into tadpole tail regrowth, zebrafish limb restoration, and the super-healing Murphy Roths Large (MRL) mouse strain, have identified ancient oxygen-sensing pathways as a possible target to achieve this goal. Specifically, upregulation of the transcription factor, hypoxia-inducible factor one alpha (HIF-1α) has been shown to modulate cell metabolism and plasticity, as well as inflammation and tissue remodeling, possibly priming injuries for regeneration. Since HIF-1α signaling is conserved across species, environmental or pharmacological manipulation of oxygen-dependent pathways may elicit a regenerative response in non-healing mammals. In this review, we will explore the emerging role of HIF-1α in mammalian healing and regeneration, as well as attempts to modulate protein stability through hyperbaric oxygen treatment, intermittent hypoxia therapy, and pharmacological targeting. We believe that these therapies could breathe new life into the field of regenerative medicine.
Collapse
Affiliation(s)
- Kelsey G DeFrates
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA.
| | - Daniela Franco
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA.
| | - Ellen Heber-Katz
- Laboratory of Regenerative Medicine, Lankenau Institute for Medical Research, Wynnewood, PA, USA.
| | - Phillip B Messersmith
- Department of Bioengineering and Materials Science and Engineering, University of California, Berkeley, CA, USA; Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| |
Collapse
|
7
|
Kim J, Kim B, Kim SM, Yang CE, Song SY, Lee WJ, Lee JH. Hypoxia-Induced Epithelial-To-Mesenchymal Transition Mediates Fibroblast Abnormalities via ERK Activation in Cutaneous Wound Healing. Int J Mol Sci 2019; 20:ijms20102546. [PMID: 31137604 PMCID: PMC6566997 DOI: 10.3390/ijms20102546] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/21/2022] Open
Abstract
Previous studies described the involvement of extracellular signal-related kinase (ERK) in systemic fibrotic diseases, but the role of ERK in cutaneous scarring is unknown. Although hypoxia drives tissue fibrosis by activating hypoxia-inducible factor-1α (HIF-1α), the specific roles of hypoxia and associated ERK phosphorylation in abnormal fibroblast activity during cutaneous scarring are unclear. Here, we investigated whether pathologic myofibroblast-like keloid fibroblast activity is promoted by hypoxia-induced epithelial-mesenchymal transition mediated by ERK activation. ERK phosphorylation was significantly increased in keloid tissue and fibroblasts. Human dermal fibroblasts cultured under hypoxia (1% O2) expressed phosphorylated ERK and exhibited activation of p38 mitogen-activated protein kinase signaling. Hypoxic human dermal fibroblasts showed increased protein and mRNA levels of epithelial-mesenchymal transition markers. Furthermore, administration of an ERK inhibitor (SCH772984) reduced the hypoxia-induced elevation of collagen type I levels in human dermal fibroblasts. Therefore, ERK may be a promising therapeutic target in profibrogenic diseases.
Collapse
Affiliation(s)
- Jihee Kim
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea.
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Korea.
| | - Bomi Kim
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Soo Min Kim
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Chae Eun Yang
- Department of Plastic and Reconstructive Surgery, Yonsei University Wonju College of Medicine, Wonju 26426, Korea.
| | - Seung Yong Song
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Korea.
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Won Jai Lee
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Korea.
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Yonsei University College of Medicine, Seoul 03722, Korea.
| | - Ju Hee Lee
- Department of Dermatology, Cutaneous Biology Research Institute, Yonsei University College of Medicine, Seoul 03722, Korea.
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Korea.
| |
Collapse
|
8
|
Zhang M, Liu S, Guan E, Liu H, Dong X, Hao Y, Zhang X, Zhao P, Liu X, Pan S, Wang Y, Wang X, Liu Y. Hyperbaric oxygen therapy can ameliorate the EMT phenomenon in keloid tissue. Medicine (Baltimore) 2018; 97:e11529. [PMID: 30024539 PMCID: PMC6086457 DOI: 10.1097/md.0000000000011529] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/21/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Hyperbaric oxygen therapy (HBOT) has been widely used in the clinical setting. In this study, HBOT therapy was evaluated for its ability to ameliorate the epithelial-to-mesenchymal transition (EMT) phenomenon in keloid tissue. METHODS Keloid patients were randomly divided into two groups: keloid patients (K group, 9 patients) and keloid patients receiving HBOT (O group, 9 patients). A third group with normal skin (S group, 9 patients) was established for control. Before HBOT and surgery, a laser Doppler flowmeter was used to measure the keloid blood supply of patients in the O group. Hematoxylin and eosin (H&E) staining was used to observe morphology. E-cadherin, ZO-1, vimentin, fibronectin, vascular endothelial growth factor (VEGF), and hypoxia inducible factor (HIF)-1α were measured by immunofluorescence staining and Western blot analysis. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to evaluate the mRNA expression level of these factors as well. RESULTS In the O group, keloid blood perfusion was significantly reduced after patients received HBOT. Compared with the K group, lower expression levels of vimentin, vibronectin, VEGF, and HIF-1α were observed in the O group, whereas the expression of E-cadherin and ZO-1 was significantly higher. The mRNA expression of E-cadherin and ZO-1 was also increased after HBOT. CONCLUSIONS The expression levels of factors related to the EMT phenomenon were significantly reversed in keloid patients after they received HBOT, indicating that HBOT may be an effective therapy against the EMT phenomenon in keloid patients.
Collapse
Affiliation(s)
- Mingzi Zhang
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Shu Liu
- Department of Plastic Surgery, China Meitan General Hospital Affiliated to North China University of Science and Technology, Beijing
| | - Enling Guan
- Department of Ear-Nose-Throat, Qingdao Huangdao District Hospital of Traditional Chinese Medicine, Qingdao, Shandong
| | - Hao Liu
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Xinhang Dong
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Yan Hao
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Xin Zhang
- College of Life Science and Bioengineering, Beijing University of Technology
| | - Pengxiang Zhao
- College of Life Science and Bioengineering, Beijing University of Technology
| | - Xuehua Liu
- Department of Hyperbaric Oxygen, Beijing Chao-Yang Hospital
| | - Shuyi Pan
- Department of Hyperbaric Oxygen, Navy General Hospital
| | - Youbin Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Xiaojun Wang
- Department of Plastic Surgery, Peking Union Medical College Hospital
| | - Yifang Liu
- International education college, Beijing Vocational College of Agriculture, Beijing, China
| |
Collapse
|
9
|
Rabieian R, Boshtam M, Zareei M, Kouhpayeh S, Masoudifar A, Mirzaei H. Plasminogen Activator Inhibitor Type-1 as a Regulator of Fibrosis. J Cell Biochem 2017; 119:17-27. [PMID: 28520219 DOI: 10.1002/jcb.26146] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 05/17/2017] [Indexed: 12/17/2022]
Abstract
Fibrosis is known as a frequent and irreversible pathological condition which is associated with organ failure. Tissue fibrosis is a central process in a variety of chronic progressive diseases such as diabetes, hypertension, and persistent inflammation. This state could contribute to chronic injury and the initiation of tissue repair. Fibrotic disorders represent abnormal wound healing with defective matrix turnover and clearance that lead to excessive accumulation of extracellular matrix components. A variety of identified growth factors, cytokines, and persistently activated myofibroblasts have critical roles in the pathogenesis of fibrosis. Irrespective of etiology, the transforming growth factor-β pathway is the major driver of fibrotic response. Plasminogen activator inhibitor-1 (PAI-1) is a crucial downstream target of this pathway. Transforming growth factor-β positively regulates PAI-1 gene expression via two main pathways including Smad-mediated canonical and non-canonical pathways. Overexpression of PAI-1 reduces extracellular matrix degradation via perturbing the plasminogen activation system. Indeed, elevated PAI-1 levels inhibit proteolytic activity of tissue plasminogen activator and urokinase plasminogen activator which could contribute to a variety of inflammatory elements in the injury site and to excessive matrix deposition. This review summarizes the current knowledge of critical pathways that regulate PAI-1 gene expression and suggests effective approaches for the treatment of fibrotic disease. J. Cell. Biochem. 119: 17-27, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Reyhaneh Rabieian
- Department of Genetics and Molecular Biology, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahshid Zareei
- Department of Biology, School of Sciences, University of Isfahan, Isfahan, Iran
| | - Shirin Kouhpayeh
- Department of Immunology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Aria Masoudifar
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Hamed Mirzaei
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
10
|
Xiong A, Liu Y. Targeting Hypoxia Inducible Factors-1α As a Novel Therapy in Fibrosis. Front Pharmacol 2017; 8:326. [PMID: 28611671 PMCID: PMC5447768 DOI: 10.3389/fphar.2017.00326] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/16/2017] [Indexed: 02/05/2023] Open
Abstract
Fibrosis, characterized by increased extracellular matrix (ECM) deposition, and widespread vasculopathy, has the prominent trait of chronic hypoxia. Hypoxia inducible factors-1α (HIF-1α), a key transcriptional factor in response to this chronic hypoxia, is involved in fibrotic disease, such as Systemic sclerosis (SSc). The implicated function of HIF-1α in fibrosis include stimulation of excessive ECM, vascular remodeling, and futile angiogenesis with further exacerbation of chronic hypoxia and deteriorate pathofibrogenesis. This review will focus on the molecular biological behavior of HIF-1α in regulating progressive fibrosis. Better understanding of the role for HIF-1α-regulated pathways in fibrotic disease will accelerate development of novel therapeutic strategies that target HIF-1α. Such new therapeutic strategies may be particularly effective for treatment of the prototypic, multisystem fibrotic, autoimmune disease SSc.
Collapse
Affiliation(s)
| | - Yi Liu
- Department of Rheumatology and Immunology, West China Hospital, Sichuan UniversityChengdu, China
| |
Collapse
|
11
|
Pamuk F, Lütfioğlu M, Aydoğdu A, Koyuncuoglu CZ, Cifcibasi E, Badur OS. The effect of low-level laser therapy as an adjunct to non-surgical periodontal treatment on gingival crevicular fluid levels of transforming growth factor-beta 1, tissue plasminogen activator and plasminogen activator inhibitor 1 in smoking and non-smoki. J Periodontal Res 2017; 52:872-882. [DOI: 10.1111/jre.12457] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/04/2017] [Indexed: 01/24/2023]
Affiliation(s)
- F. Pamuk
- Department of Periodontology; Faculty of Dentistry; Yeditepe Univesity; Istanbul Turkey
- Department of Periodontology; Faculty of Dentistry; Istanbul Aydin University; Istanbul Turkey
| | - M. Lütfioğlu
- Department of Periodontology; Faculty of Dentistry; Ondokuzmayis University; Samsun Turkey
| | - A. Aydoğdu
- Department of Periodontology; Faculty of Dentistry; Istanbul Research and Application Center; Baskent University; Istanbul Turkey
| | - C. Z. Koyuncuoglu
- Department of Periodontology; Faculty of Dentistry; Istanbul Aydin University; Istanbul Turkey
| | - E. Cifcibasi
- Department of Periodontology; Faculty of Dentistry; Istanbul University; Istanbul Turkey
| | - O. S. Badur
- Division of Virology and Immunology; Department of Microbiology; Faculty of Medicine; Istanbul University; Istanbul Turkey
| |
Collapse
|
12
|
Sharma M, Radhakrishnan R. Limited mouth opening in oral submucous fibrosis: reasons, ramifications, and remedies. J Oral Pathol Med 2016; 46:424-430. [PMID: 27743497 DOI: 10.1111/jop.12513] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/10/2016] [Indexed: 12/14/2022]
Abstract
Limited mouth opening (LMO) in oral submucous fibrosis (OSF) has been attributed to both the submucosal and muscle fibrosis (MF). While reflectory trismus was proposed before as an auxiliary mechanism by another group, the stretch-mediated muscle damage (MSD), histopathological changes in blood vessels (such as endothelial dysfunction, endothelial hypertrophy, and endarteritis obliterans), and upregulated anaerobic isoforms of lactate dehydrogenase (LDH) have been proposed by us as complementary events leading to MF. Additionally, the amount of hypoxia-mediated upregulation of anaerobic isoforms of LDH determines the extent of MF. Radiotherapy (RT)-mediated release of reactive oxygen species causes vascular damage thereby worsening hypoxia. While the alteration in LDH levels secondary to hypoxia enhances fibrosis, RT worsens it. Oral squamous cell carcinoma occurring in the background of OSF is an absolute contraindication for RT as it augurs unfavorable prognosis. An algorithm to demonstrate this with evidence is clearly depicted. The role of HIF-1α in the progression of OSF and its malignant transformation, and the consideration of hyperbaric oxygen therapy as a therapeutic remedy in OSF are underscored.
Collapse
Affiliation(s)
- Mohit Sharma
- Department Of Oral Pathology, ITS Dental College, Hospital and Research Center, Greater Noida, India
| | - Raghu Radhakrishnan
- Department of Oral Pathology, Manipal College of Dental Sciences, Manipal University, Manipal, India
| |
Collapse
|
13
|
Li Q, Zhang C, Fu X. Will stem cells bring hope to pathological skin scar treatment? Cytotherapy 2016; 18:943-956. [PMID: 27293205 DOI: 10.1016/j.jcyt.2016.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 04/19/2016] [Accepted: 05/10/2016] [Indexed: 12/12/2022]
Abstract
Pathological skin scars, such as keloids, aesthetically and psychosocially affect patients. The quest for scar reduction and the increasing recognition of patient satisfaction has led to the continued exploration of scar treatment. Stem cells are a promising source for tissue repair and regeneration. The multi-potency and secretory functions of these cells could offer possible treatments for pathological scars and have been examined in recent studies. Here, we analyze the factors that influence the formation of pathological skin scars, summarize recent research on pathological scar treatment with stem cells and elaborate on the possible mechanisms of this treatment. Additionally, other effects of stem cell treatments are also presented while evaluating potential side effects of stem cell-based pathological scar treatments. Thus, this review may provide meaningful guidance in the clinic for scar treatments with stem cells.
Collapse
Affiliation(s)
- Qiankun Li
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China
| | - Cuiping Zhang
- Stem Cell and Tissue Regeneration Laboratory, The First Affiliated Hospital, General Hospital of PLA, Beijing, China.
| | - Xiaobing Fu
- Wound Healing and Cell Biology Laboratory, Institute of Basic Medical Science, Chinese PLA General Hospital, Beijing, China; Stem Cell and Tissue Regeneration Laboratory, The First Affiliated Hospital, General Hospital of PLA, Beijing, China.
| |
Collapse
|
14
|
Ma X, Chen J, Xu B, Long X, Qin H, Zhao RC, Wang X. Keloid-derived keratinocytes acquire a fibroblast-like appearance and an enhanced invasive capacity in a hypoxic microenvironment in vitro. Int J Mol Med 2015; 35:1246-56. [PMID: 25777304 PMCID: PMC4380122 DOI: 10.3892/ijmm.2015.2135] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 03/05/2015] [Indexed: 11/05/2022] Open
Abstract
A keloid scar is an overgrowth of dense fibrous tissue that develops around a wound. These scars are raised scars that spread beyong the margins of the orinigal wound to normal skin by invasion. Keloid tissue consists of both an epithelium and dermal fibroblasts. Recent studies have primarily focused on keloid fibroblasts; however, the precise role of keratinocytes in the invasion process of keloids remains to be identified. Hypoxia is a typical characteristic of keloid scars, as well as other solid tumors. The expression of the transcription factor, hypoxia-inducible factor-1α (HIF-1α), is mainly induced by hypoxia and is known for its ability to induce proliferative and transformative changes in cells; its expression has been shown to correlate with tumor invasion and metastasis. In the present study, we used immunohistochemistry, fluorescence staining and western blot analysis and demonstrated that HIF-1α was highly expressed in both the epithelial layer of keloid tissue specimens and in hypoxia-exposed keratinocytes, which suggested that the keloid keratinocytes underwent epithelial-to-mesenchymal transition (EMT) in vitro. The high expression of mesenchymal markers, such as as vimentin and fibronectin was confirmed, as well as the reduced expression of E-cadherin and zonula occludens-1 (ZO-1) during this process by detection at the protein and mRNA level. Moreover, siRNA targeting HIF-1α reversed the changes which had occurred in the morphology of the keratinocytes (cells had acquired a fibroblast-like appearance) and suppressed the invasive ability of the keratinocytes. In conclusion, the present findings demonstrate that the hypoxia/HIF-1α microenvironment provides a favorable environment for keloid-derived keratinocytes to adopt a fibroblast-like appearance through EMT. This transition may be responsible for the enhanced capacity of keloid keratinocytes to invade, allowing the keloids to extend beyond the wound margin.
Collapse
Affiliation(s)
- Xiaoyang Ma
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Jia Chen
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Bei Xu
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Xiao Long
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| | - Han Qin
- Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China
| | - Robert Chunhua Zhao
- Institute of Basic Medical Sciences and School of Basic Medicine, Center of Excellence in Tissue Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100005, P.R. China
| | - Xiaojun Wang
- Department of Plastic and Reconstructive Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, P.R. China
| |
Collapse
|
15
|
ZHANG ZHE, NIE FANGFEI, KANG CHUNFU, CHEN BIN, QIN ZELIAN, MA JIANXUN, MA YONGGUANG, ZHAO XIA. Increased periostin expression affects the proliferation, collagen synthesis, migration and invasion of keloid fibroblasts under hypoxic conditions. Int J Mol Med 2014; 34:253-61. [DOI: 10.3892/ijmm.2014.1760] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 04/10/2014] [Indexed: 11/05/2022] Open
|
16
|
Arno AI, Amini-Nik S, Blit PH, Al-Shehab M, Belo C, Herer E, Jeschke MG. Effect of human Wharton's jelly mesenchymal stem cell paracrine signaling on keloid fibroblasts. Stem Cells Transl Med 2014; 3:299-307. [PMID: 24436441 DOI: 10.5966/sctm.2013-0120] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Keloid scars are abnormal benign fibroproliferative tumors with high recurrence rates and no current efficacious treatment. Accumulating evidence suggests that human umbilical cord Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) have antifibrotic properties. Paracrine signaling is considered one of the main underlying mechanisms behind the therapeutic effects of mesenchymal stem cells. However, the paracrine signaling effects of WJ-MSCs on keloids have not yet been reported. The aim of this study is to investigate paracrine signaling effects of human WJ-MSCs on keloid fibroblasts in vitro. Human umbilical cords and keloid skin samples were obtained, and WJ-MSCs and keloid fibroblasts were isolated and cultured. One-way and two-way paracrine culture systems between both cell types were investigated. Plasminogen activator inhibitor-I and transforming growth factor-β2 (TGF-β2) transcripts were upregulated in keloid fibroblasts cultured with WJ-MSC-conditioned medium (WJ-MSC-CM) and cocultured with inserts, while showing lower TGF-β3 gene expression. Interleukin (IL)-6, IL-8, TGF-β1, and TGF-β2 protein expression was also enhanced. The WJ-MSC-CM-treated keloid fibroblasts showed higher proliferation rates than their control keloid fibroblasts with no significant change in apoptosis rate or migration ability. In our culture conditions, the indirect application of WJ-MSCs on keloid fibroblasts may enhance their profibrotic phenotype.
Collapse
Affiliation(s)
- Anna I Arno
- Plastic Surgery Department and Burn Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain; Ross Tilley Burn Centre and Sunnybrook Research Institute and Gynecology and Obstetrics Department, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
17
|
Proteomic profiling reveals upregulated protein expression of hsp70 in keloids. BIOMED RESEARCH INTERNATIONAL 2013; 2013:621538. [PMID: 24260741 PMCID: PMC3821890 DOI: 10.1155/2013/621538] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 08/20/2013] [Accepted: 08/30/2013] [Indexed: 11/29/2022]
Abstract
Background. The biochemical characteristics of keloid-derived fibroblasts differ from those of adjacent normal fibroblasts, and these differences are thought to be the cause of abnormal fibrosis. Therefore, we investigated the characteristic proteins that are differentially expressed in keloid-derived fibroblasts using proteomics tools. Objective. We attempted to investigate the novel proteins that play important roles in the pathophysiology of keloids. Methods. Proteomics analysis was performed to identify differentially expressed proteins in keloid-derived fibroblasts. Keloid-derived fibroblasts and adjacent normal fibroblasts were analyzed with 2-DAGE. We validated these proteins with immunoblot analysis, real-time RT-PCR, and immunohistochemistry. Results. Sixteen differentially expressed protein spots were identified in keloid-derived fibroblasts. Among them, heat shock protein 70 (Hsp70) was specifically upregulated in keloid-derived fibroblasts. Also, immunohistochemistry and western blot analysis revealed increased Hsp70, TGF-β, and PCNA expressions in keloids compared to normal tissue. Conclusion. Hsp70 is overexpressed in keloid fibroblasts and tissue. The overexpression of Hsp70 may be involved in the pathogenesis of keloids, and the inhibition of Hsp70 could be a new therapeutic tool for the treatment of keloids.
Collapse
|
18
|
Lokmic Z, Musyoka J, Hewitson TD, Darby IA. Hypoxia and hypoxia signaling in tissue repair and fibrosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2012; 296:139-85. [PMID: 22559939 DOI: 10.1016/b978-0-12-394307-1.00003-5] [Citation(s) in RCA: 141] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Following injury, vascular damage results in the loss of perfusion and consequent low oxygen tension (hypoxia) which may be exacerbated by a rapid influx of inflammatory and mesenchymal cells with high metabolic demands for oxygen. Changes in systemic and cellular oxygen concentrations induce tightly regulated response pathways that attempt to restore oxygen supply to cells and modulate cell function in hypoxic conditions. Most of these responses occur through the induction of the transcription factor hypoxia-inducible factor-1 (HIF-1) which regulates many processes needed for tissue repair during ischemia in the damaged tissue. HIF-1 transcriptionally upregulates expression of metabolic proteins (GLUT-1), adhesion proteins (integrins), soluble growth factors (TGF-β and VEGF), and extracellular matrix components (type I collagen and fibronectin), which enhance the repair process. For these reasons, HIF-1 is viewed as a positive regulator of wound healing and a potential regulator of organ repair and tissue fibrosis. Understanding the complex role of hypoxia in the loss of function in scarring tissues and biology of chronic wound, and organ repair will aid in the development of pharmaceutical agents that can redress the detrimental outcomes often seen in repair and scarring.
Collapse
Affiliation(s)
- Zerina Lokmic
- Department of Surgery, St. Vincent's Hospital, University of Melbourne, Fitzroy, Victoria, Australia
| | | | | | | |
Collapse
|
19
|
Intraabdominal adhesion formation is associated with differential mRNA expression of metabolic genes PDHb and SDHa. Arch Gynecol Obstet 2012; 286:683-6. [DOI: 10.1007/s00404-012-2364-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Accepted: 04/23/2012] [Indexed: 10/28/2022]
|
20
|
Complex Regulation of the Pericellular Proteolytic Microenvironment during Tumor Progression and Wound Repair: Functional Interactions between the Serine Protease and Matrix Metalloproteinase Cascades. Biochem Res Int 2012; 2012:454368. [PMID: 22454771 PMCID: PMC3290807 DOI: 10.1155/2012/454368] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 11/21/2011] [Indexed: 01/08/2023] Open
Abstract
Spatial and temporal regulation of the pericellular proteolytic environment by local growth factors, such as EGF and TGF-β, initiates a wide repertoire of cellular responses coupled to a plasmin/matrix metalloproteinase (MMP) dependent stromal-remodeling axis. Cell motility and invasion, tumor metastasis, wound healing, and organ fibrosis, for example, represent diverse events controlled by expression of a subset of genes that encode various classes of tissue remodeling proteins. These include members of the serine protease and MMP families that functionally constitute a complex system of interacting protease cascades and titrated by their respective inhibitors. Several structural components of the extracellular matrix are upregulated by TGF-β as are matrix-active proteases (e.g., urokinase (uPA), plasmin, MMP-1, -3, -9, -10, -11, -13, -14). Stringent controls on serine protease/MMP expression and their topographic activity are essential for maintaining tissue homeostasis. Targeting individual elements in this highly interactive network may lead to novel therapeutic approaches for the treatment of cancer, fibrotic diseases, and chronic wounds.
Collapse
|
21
|
Abstract
Fibrosis is defined as a fibroproliferative or abnormal fibroblast activation-related disease. Deregulation of wound healing leads to hyperactivation of fibroblasts and excessive accumulation of extracellular matrix (ECM) proteins in the wound area, the pathological manifestation of fibrosis. The accumulation of excessive levels of collagen in the ECM depends on two factors: an increased rate of collagen synthesis and or decreased rate of collagen degradation by cellular proteolytic activities. The urokinase/tissue type plasminogen activator (uPA/tPA) and plasmin play significant roles in the cellular proteolytic degradation of ECM proteins and the maintenance of tissue homeostasis. The activities of uPA/tPA/plasmin and plasmin-dependent MMPs rely mostly on the activity of a potent inhibitor of uPA/tPA, plasminogen activator inhibitor-1 (PAI-1). Under normal physiologic conditions, PAI-1 controls the activities of uPA/tPA/plasmin/MMP proteolytic activities and thus maintains the tissue homeostasis. During wound healing, elevated levels of PAI-1 inhibit uPA/tPA/plasmin and plasmin-dependent MMP activities, and, thus, help expedite wound healing. In contrast to this scenario, under pathologic conditions, excessive PAI-1 contributes to excessive accumulation of collagen and other ECM protein in the wound area, and thus preserves scarring. While the level of PAI-1 is significantly elevated in fibrotic tissues, lack of PAI-1 protects different organs from fibrosis in response to injury-related profibrotic signals. Thus, PAI-1 is implicated in the pathology of fibrosis in different organs including the heart, lung, kidney, liver, and skin. Paradoxically, PAI-1 deficiency promotes spontaneous cardiac-selective fibrosis. In this review, we discuss the significance of PAI-1 in the pathogenesis of fibrosis in multiple organs.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA.
| | | |
Collapse
|
22
|
Sebastian A, Allan E, Allan D, Colthurst J, Bayat A. Addition of novel degenerate electrical waveform stimulation with photodynamic therapy significantly enhances its cytotoxic effect in keloid fibroblasts: First report of a potential combination therapy. J Dermatol Sci 2011; 64:174-84. [DOI: 10.1016/j.jdermsci.2011.08.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2011] [Revised: 08/15/2011] [Accepted: 08/29/2011] [Indexed: 12/17/2022]
|
23
|
Deschene K, Céleste C, Boerboom D, Theoret CL. Hypoxia regulates the expression of extracellular matrix associated proteins in equine dermal fibroblasts via HIF1. J Dermatol Sci 2011; 65:12-8. [PMID: 21999945 DOI: 10.1016/j.jdermsci.2011.09.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 09/05/2011] [Accepted: 09/15/2011] [Indexed: 01/20/2023]
Abstract
BACKGROUND Exuberant granulation tissue (EGT), a fibrotic healing disorder resembling the human keloid, occurs almost exclusively in limb wounds of horses and may be caused in part by a relative state of hypoxia within the wound. OBJECTIVE The objectives of this study were therefore to (1) assess the effects of hypoxia on equine dermal fibroblast (EDF) proliferation and apoptosis, (2) study the effects of hypoxia on the expression of key extracellular matrix (ECM) associated proteins and determine if such effects are dependent on hypoxia-inducible factor (HIF), and (3) determine if EDFs from the body or limb respond differently to hypoxia. METHODS EDFs were isolated and cultured from skin from body or limb under normoxic or hypoxic conditions for up to 7days. RESULTS Hypoxia significantly stimulated EDF proliferation, but had no effect on cell survival. The hypoxia-mimetic agent CoCl(2) up-regulated COL1A1 expression and down-regulated MMP2 expression, suggesting an increase in ECM synthesis and a decrease in turnover. Both regulatory effects were inhibited by the addition of echinomycin, indicating that they are mediated by the transcriptional regulatory activity of HIF. No differences were observed between EDFs originating from body or limb for any effect of hypoxia or CoCl(2), suggesting that EGT development does not depend on intrinsic properties of limb fibroblasts. CONCLUSIONS We conclude that hypoxia regulates ECM remodeling via HIF1 in EDFs, and that this may be an important determinant in the pathogenesis of equine EGT.
Collapse
Affiliation(s)
- Karine Deschene
- Université de Montréal, Département de biomédecine vétérinaire, Canada
| | | | | | | |
Collapse
|
24
|
PAI-1: An Integrator of Cell Signaling and Migration. Int J Cell Biol 2011; 2011:562481. [PMID: 21837240 PMCID: PMC3151495 DOI: 10.1155/2011/562481] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2011] [Revised: 05/09/2011] [Accepted: 05/17/2011] [Indexed: 12/23/2022] Open
Abstract
Cellular migration, over simple surfaces or through complex stromal barriers, requires coordination between detachment/re-adhesion cycles, involving structural components of the extracellular matrix and their surface-binding elements (integrins), and the precise regulation of the pericellular proteolytic microenvironment. It is now apparent that several proteases and protease inhibitors, most notably urokinase plasminogen activator (uPA) and plasminogen activator inhibitor type-1 (PAI-1), also interact with several cell surface receptors transducing intracellular signals that significantly affect both motile and proliferative programs. These events appear distinct from the original function of uPA/PAI-1 as modulators of the plasmin-based proteolytic cascade. The multifaceted interactions of PAI-1 with specific matrix components (i.e., vitronectin), the low-density lipoprotein receptor-related protein-1 (LRP1), and the uPA/uPA receptor complex have dramatic consequences on the migratory phenotype and may underlie the pathophysiologic sequalae of PAI-1 deficiency and overexpression. This paper focuses on the increasingly intricate role of PAI-1 as a major mechanistic determinant of the cellular migratory phenotype.
Collapse
|
25
|
Matsui T, Ito C, Oda M, Itoigawa M, Yokoo K, Okada T, Furukawa H. Lapachol suppresses cell proliferation and secretion of interleukin-6 and plasminogen activator inhibitor-1 of fibroblasts derived from hypertrophic scars. J Pharm Pharmacol 2011; 63:960-6. [DOI: 10.1111/j.2042-7158.2011.01292.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Abstract
Objectives
The pathogenesis and therapy of hypertrophic scar have not yet been established. Our aim was to investigate the antiproliferative and antisecretory effects of lapachol, isolated from the stem bark of Avicennia rumphiana Hall. f., on hypertrophic scar fibroblasts.
Methods
The effects of lapachol on hypertrophic scar fibroblast proliferation were measured using the MTT assay, cell-cycle analyses and lactate dehydrogenase assays. The type I collagen α-chain (COL1A1), interleukin-6 (IL-6) and plasminogen activator inhibitor-1 (PAI-1) mRNA and/or protein levels of hypertrophic scar-fibroblasts were quantitated by real-time PCR and ELISA.
Key findings
Lapachol at 25 and 50 µm significantly inhibited the in vitro proliferation of hypertrophic scar fibroblasts, but not fibroblasts from non-lesional skin sites. In addition, lapachol had no apparent effect on cell cycle and lactate dehydrogenase activity in conditioned medium from lapachol-treated hypertrophic scar fibroblasts was nearly equal to that in medium from vehicle-treated cells. Lapachol treatment also inhibited COL1A1 and PAI-1 mRNA levels in hypertrophic scar fibroblasts, but did not affect IL-6 mRNA levels. The protein levels of IL-6 and PAI-1 in conditioned medium from hypertrophic scar fibroblasts treated with 50 µm lapachol were lower than those from vehicle-treated hypertrophic scar fibroblasts.
Conclusions
Lapachol decreased the proliferation rate of hypertrophic scar fibroblasts. As IL-6 and PAI-1 secretion was also lowered in lapachol-treated hypertrophic scar fibroblasts, our findings suggested that lapachol may have suppressed extracellular matrix hyperplasia in wound healing and possibly alleviated the formation of hypertrophic scar.
Collapse
Affiliation(s)
- Takuya Matsui
- Faculty of Pharmacy, Meijo University, Tempaku-ku, Nagoya, Japan
- Department of Physiology, Aichi Medical University, Nagakute-cho, Aichi-gun, Japan
| | - Chihiro Ito
- Faculty of Pharmacy, Meijo University, Tempaku-ku, Nagoya, Japan
| | - Makiko Oda
- Department of Plastic Surgery, Aichi Medical University, Nagakute-cho, Aichi-gun, Japan
| | - Masataka Itoigawa
- Faculty of Human Wellness, Tokai Gakuen University, Tempaku, Nagoya, Aichi, Japan
| | - Kazuhisa Yokoo
- Department of Plastic Surgery, Aichi Medical University, Nagakute-cho, Aichi-gun, Japan
| | - Tadashi Okada
- Department of Physiology, Aichi Medical University, Nagakute-cho, Aichi-gun, Japan
| | - Hiroshi Furukawa
- Faculty of Pharmacy, Meijo University, Tempaku-ku, Nagoya, Japan
| |
Collapse
|
26
|
Rezvani HR, Ali N, Nissen LJ, Harfouche G, de Verneuil H, Taïeb A, Mazurier F. HIF-1α in epidermis: oxygen sensing, cutaneous angiogenesis, cancer, and non-cancer disorders. J Invest Dermatol 2011; 131:1793-805. [PMID: 21633368 DOI: 10.1038/jid.2011.141] [Citation(s) in RCA: 95] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Besides lung, postnatal human epidermis is the only epithelium in direct contact with atmospheric oxygen. Skin epidermal oxygenation occurs mostly through atmospheric oxygen rather than tissue vasculature, resulting in a mildly hypoxic microenvironment that favors increased expression of hypoxia-inducible factor-1α (HIF-1α). Considering the wide spectrum of biological processes, such as angiogenesis, inflammation, bioenergetics, proliferation, motility, and apoptosis, that are regulated by this transcription factor, its high expression level in the epidermis might be important to HIF-1α in skin physiology and pathophysiology. Here, we review the role of HIF-1α in cutaneous angiogenesis, skin tumorigenesis, and several skin disorders.
Collapse
|
27
|
Carrera S, de Verdier PJ, Khan Z, Zhao B, Mahale A, Bowman KJ, Zainol M, Jones GDD, Lee SW, Aaronson SA, Macip S. Protection of cells in physiological oxygen tensions against DNA damage-induced apoptosis. J Biol Chem 2010; 285:13658-65. [PMID: 20228054 DOI: 10.1074/jbc.m109.062562] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Oxygen availability has important effects on cell physiology. Although hyperoxic and hypoxic stresses have been well characterized, little is known about cellular functions in the oxygen levels commonly found in vivo. Here, we show that p53-dependent apoptosis in response to different DNA-damaging agents was reduced when normal and cancer cells were cultured at physiological oxygen tensions instead of the usual atmospheric levels. Different from what has been described in hypoxia, this was neither determined by decreases in p53 induction or its transactivation activity, nor by differences in the intracellular accumulation of reactive oxygen species. At these physiological oxygen levels, we found a constitutive activation of the ERK1/2 MAPK in all the models studied. Inhibition of this signaling pathway reversed the protective effect in some but not all cell lines. We conclude that a stress-independent constitutive activation of prosurvival pathways, including but probably not limited to MAPK, can protect cells in physiological oxygen tensions against genotoxic stress. Our results underscore the need of considering the impact of oxygen levels present in the tissue microenvironment when studying cell sensitivity to treatments such as chemotherapy and radiotherapy.
Collapse
Affiliation(s)
- Samantha Carrera
- Department of Biochemistry, University of Leicester, Lancaster Road, Leicester LE1 9HN, United Kingdom
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Genetics of keloid scarring. Arch Dermatol Res 2010; 302:319-39. [PMID: 20130896 DOI: 10.1007/s00403-009-1014-y] [Citation(s) in RCA: 147] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2009] [Revised: 11/30/2009] [Accepted: 12/07/2009] [Indexed: 12/15/2022]
Abstract
Keloid scarring, also known as keloid disease (KD), is a common, abnormally raised fibroproliferative cutaneous lesion that can occur following even minor skin trauma. The aetiopathogenesis of KD has remained an enigma todate compounded by an ill-defined clinical management. There is strong evidence suggesting a genetic susceptibility in individuals affected by KD, including familial heritability, common occurrence in twins and high prevalence in certain ethnic populations. This review aims to address the genetic aspects of KD that have been described in present literature that include inheritance patterns, linkage studies, case-control association studies, whole genome gene expression microarray studies and gene pathways that were significant in KD. In addition to our clinical and scientific background in KD, we used search engines, Scopus, Scirus and PubMed, which searched for key terms covering various genetic aspects of KD. Additionally, genes reported in seven whole genome gene expression microarray studies were separately compared in detail. Our findings indicate a varied inheritance pattern in KD (predominantly autosomal dominant), linkage loci (chromosomes 2q23 and 7p11), several human leukocyte antigen (HLA) alleles (HLA-DRB1*15, HLA-DQA1*0104, DQ-B1*0501 and DQB1*0503), negative candidate gene case-control association studies and at least 25 dysregulated genes reported in multiple microarray studies. The major pathways reportedly proposed to be involved in KD include apoptosis, mitogen-activated protein kinase, transforming growth factor-beta, interleukin-6 and plasminogen activator inhibitor-1. In summary, involvement of more than one gene is likely to be responsible for susceptibility to KD. A better understanding of the genes involved in KD may potentially lead to the development of more effective diagnostic, therapeutic and prognostic measures.
Collapse
|
29
|
He S, Liu X, Yang Y, Huang W, Xu S, Yang S, Zhang X, Roberts M. Mechanisms of transforming growth factor β1
/Smad signalling mediated by mitogen-activated protein kinase pathways in keloid fibroblasts. Br J Dermatol 2009; 162:538-46. [DOI: 10.1111/j.1365-2133.2009.09511.x] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
30
|
Park G, Yoon BS, Moon JH, Kim B, Jun EK, Oh S, Kim H, Song HJ, Noh JY, Oh C, You S. Green tea polyphenol epigallocatechin-3-gallate suppresses collagen production and proliferation in keloid fibroblasts via inhibition of the STAT3-signaling pathway. J Invest Dermatol 2008; 128:2429-41. [PMID: 18463684 DOI: 10.1038/jid.2008.103] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Keloids are benign skin tumors characterized by collagen accumulation and hyperproliferation of fibroblasts. To find an effective therapy for keloids, we explored the pharmacological potential of (-)-epigallocatechin-3-gallate (EGCG), a widely investigated tumor-preventive agent. When applied to normal and keloid fibroblasts (KFs) in vitro, proliferation and migration of KFs were more strongly suppressed by EGCG than normal fibroblast proliferation and migration (IC(50): 54.4 microM (keloid fibroblast (KF)) versus 63.0 microM (NF)). The level of Smad2/3, signal transducer and activator of transcription-3 (STAT3), and p38 phosphorylation is more enhanced in KFs, and EGCG inhibited phosphorylation of phosphatidylinositol-3-kinase (PI3K), extracellular signal-regulated protein kinase 1/2 (ERK1/2), and STAT3 (Tyr705 and Ser727). To evaluate the contribution of these pathways to keloid pathology, we treated KFs with specific inhibitors for PI3K, ERK1/2, or STAT3. Although a PI3K inhibitor significantly suppressed proliferation, PI3K and MEK/ERK inhibitors had a minor effect on migration and collagen production. However, a JAK2/STAT3 inhibitor and a STAT3 siRNA strongly suppressed proliferation, migration, and collagen production by KFs. We also found that treatment with EGCG suppressed growth and collagen production in the in vivo keloid model. This study demonstrates that EGCG suppresses the pathological characteristics of keloids through inhibition of the STAT3-signaling pathway. We propose that EGCG has potential in the treatment and prevention of keloids.
Collapse
Affiliation(s)
- Gyuman Park
- Department of Dermatology, School of Medicine, Korea University, Seoul, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
KÖSE OSMAN, WASEEM AHMAD. Keloids and Hypertrophic Scars: Are They Two Different Sides of the Same Coin? Dermatol Surg 2008; 34:336-46. [DOI: 10.1111/j.1524-4725.2007.34067.x] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
|
33
|
Zhang GY, Yi CG, Li X, Zheng Y, Niu ZG, Xia W, Meng Z, Meng CY, Guo SZ. Inhibition of vascular endothelial growth factor expression in keloid fibroblasts by vector-mediated vascular endothelial growth factor shRNA: a therapeutic potential strategy for keloid. Arch Dermatol Res 2008; 300:177-84. [DOI: 10.1007/s00403-007-0825-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2007] [Revised: 11/22/2007] [Accepted: 12/18/2007] [Indexed: 01/08/2023]
|
34
|
Palumbo C, Ferretti M, Bonucci P, Sena P, Bertoni L, Cavani F, Celli A, Rovesta C. Two peculiar conditions following a coma: A clinical case of heterotopic ossification concomitant with keloid formation. Clin Anat 2008; 21:348-54. [DOI: 10.1002/ca.20616] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
35
|
Segura T, Schmokel H, Hubbell JA. RNA interference targeting hypoxia inducible factor 1alpha reduces post-operative adhesions in rats. J Surg Res 2007; 141:162-70. [PMID: 17561118 DOI: 10.1016/j.jss.2006.07.045] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2006] [Revised: 07/24/2006] [Accepted: 07/31/2006] [Indexed: 01/17/2023]
Abstract
BACKGROUND To investigate the use of RNA interference mediated gene down-regulation targeting hypoxia inducible factor 1alpha (HIF-1alpha) and plasminogen activator inhibitor 1 (PAI-1) in an effort to prevent abdominal adhesion formation. MATERIALS AND METHODS Real time PCR and a PAI-1 protein activity assay were used in vitro to determine the efficacy of small interfering RNAs (siRNAs). For in vivo experiments, 57 white female rats were operated to generate ischemic and serosal injury to the uterine horns, and treated with saline, siRNA(Lamin A/C) (negative control), siRNA(HIF-1alpha), siRNA(PAI-1), or siRNA(HIF-1alpha) plus siRNA(PAI-1). The cationic polyer poly(ethylenimine) (PEI) was used as the delivery vehicle for all siRNAs delivered in vivo. Adhesions were analyzed by a blinded surgeon 8 days post-surgery. RESULTS After in vitro transfection with siRNA, at least 69% gene down-regulation was obtained for all siRNAs tested. In vitro siRNA-mediated down-regulation of HIF-1alpha, PAI-1 or their simultaneous delivery resulted in a significant decrease of PAI-1 protein activity (at least P < 0.05). Administration of 4 nmol siRNA(HIF-1alpha)/PEI complexes after injury to the uterine horns achieved a statistical reduction of post-operative adhesion formation with a reduction by 52% (P < 0.05). Delivery of 4 nmol siRNA(PAI-1)/PEI complexes and the simultaneous delivery of 2 nmol siRNA(HIF-1alpha) plus 2 nmol siRNA(PAI-1), resulted in a reduction of abdominal adhesion by 36% and 42%, respectively, with the reduction being statistically significant when compared directly to the saline control (P < 0.01). CONCLUSION These data show that administration of siRNA/PEI complexes within the peritoneal cavity can be used to prevent post-operative abdominopelvic adhesions.
Collapse
Affiliation(s)
- Tatiana Segura
- Ecole Polytechnique Fédérale de Lausanne (EPFL), Institute of Bioengineering and Institute of Chemical Sciences and Engineering, Lausanne, Switzerland
| | | | | |
Collapse
|
36
|
Hong KH, Yoo SA, Kang SS, Choi JJ, Kim WU, Cho CS. Hypoxia induces expression of connective tissue growth factor in scleroderma skin fibroblasts. Clin Exp Immunol 2007; 146:362-70. [PMID: 17034590 PMCID: PMC1942060 DOI: 10.1111/j.1365-2249.2006.03199.x] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Connective tissue growth factor (CTGF) plays a role in the fibrotic process of systemic sclerosis (SSc). Because hypoxia is associated with fibrosis in several profibrogenic conditions, we investigated whether CTGF expression in SSc fibroblasts is regulated by hypoxia. Dermal fibroblasts from patients with SSc and healthy controls were cultured in the presence of hypoxia or cobalt chloride (CoCl(2)), a chemical inducer of hypoxia-inducible factor (HIF)-1alpha. Expression of CTGF was evaluated by Northern and Western blot analyses. Dermal fibroblasts exposed to hypoxia (1% O(2)) or CoCl(2) (1-100 microM) enhanced expression of CTGF mRNA. Skin fibroblasts transfected with HIF-1alpha showed the increased levels of CTGF protein and mRNA, as well as nuclear staining of HIF-1alpha, which was enhanced further by treatment of CoCl(2). Simultaneous treatment of CoCl(2) and transforming growth factor (TGF)-beta additively increased CTGF mRNA in dermal fibroblasts. Interferon-gamma inhibited the TGF-beta-induced CTGF mRNA expression dose-dependently in dermal fibroblasts, but they failed to hamper the CoCl(2)-induced CTGF mRNA expression. In addition, CoCl(2) treatment increased nuclear factor (NF)-kappaB binding activity for CTGF mRNA, while decreasing IkappaBalpha expression in dermal fibroblasts. Our data suggest that hypoxia, caused possibly by microvascular alterations, up-regulates CTGF expression through the activation of HIF-1alpha in dermal fibroblasts of SSc patients, and thereby contributes to the progression of skin fibrosis.
Collapse
Affiliation(s)
- K-H Hong
- Department of Internal Medicine, Division of Rheumatology, School of Medicine, Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
37
|
Liao H, Hyman MC, Lawrence DA, Pinsky DJ. Molecular regulation of the PAI-1 gene by hypoxia: contributions of Egr-1, HIF-1alpha, and C/EBPalpha. FASEB J 2006; 21:935-49. [PMID: 17197388 DOI: 10.1096/fj.06-6285com] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hypoxia, as occurs during tissue ischemia, tips the natural anticoagulant/procoagulant balance of the endovascular wall to favor activation of coagulation. Plasminogen activator inhibitor-1 (PAI-1) is an important factor suppressing fibrinolysis under conditions of low oxygen tension. We previously reported that hypoxia induced PAI-1 mRNA and antigen expression in murine macrophages secondary to increased de novo transcription as well as increased mRNA stability. We now show in RAW264.7 murine macrophages that the transcription factors early growth response gene-1 (Egr-1), hypoxia-inducible factor-1alpha (HIF-1alpha), and CCAAT/enhancer binding protein alpha (C/EBPalpha) are quickly activated in hypoxia and are responsible for transcription and expression of PAI-1. Murine PAI-1 promoter constructs, including Egr, HIF-1alpha, and/or C/EBPalpha binding sites, were transfected into RAW 264.7 murine macrophages. To identify the relative importance of each of these putative hypoxia-responsive elements, cells were exposed to normobaric hypoxia, and transcriptional activity was recorded. At 16 h of hypoxic exposure, murine PAI-1 promoter deletion constructs that included Egr, HIF-1alpha, and/or C/EBPalpha binding sites demonstrated increased transcriptional activity. Mutation of each of these three murine PAI-1 promoter sites (or a combination of them) resulted in a marked reduction in hypoxia sensitivity as detected by transcriptional analysis. Functional data obtained using 32P-labeled Egr, HIF-1alpha response element (HRE), and C/EBPalpha oligonucleotides revealed induction of DNA binding activity in nuclear extracts from hypoxic RAW cells, with supershift analysis confirming activation of Egr-1, HIF-1alpha, or C/EBPalpha. ChIP analysis confirmed the authenticity of these interactions as each of these transcription factors binds to chromatin under hypoxic conditions. Further, the induction of PAI-1 by Egr-1, HIF-1alpha, or C/EBPalpha was replicated in primary peritoneal macrophages. These data suggest that although HIF-1alpha appears to dominate the PAI-1 transcriptional response in hypoxia, Egr-1 and C/EBPalpha greatly augment this response and can do so independent of HIF-1alpha or each other. These studies are relevant to ischemic up-regulation of the PAI-1 gene and consequent accrual of microvascular thrombus under ischemic conditions.
Collapse
Affiliation(s)
- Hui Liao
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | | | | | | |
Collapse
|
38
|
Martin-García RF, Busquets AC. Postsurgical Use of Imiquimod 5% Cream in the Prevention of Earlobe Keloid Recurrences: Results of an Open-Label, Pilot Study. Dermatol Surg 2006; 31:1394-8. [PMID: 16416606 DOI: 10.2310/6350.2005.31203] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
BACKGROUND Traditional surgical modalities for the management of earlobe keloids are often associated with high recurrence rates. A recent report suggests that imiquimod 5% cream can be effective in the prevention of keloid recurrences after surgical excision. OBJECTIVES To establish the safety and efficacy of imiquimod 5% cream in the prevention of recurrences of excised earlobe keloids. METHODS Patients who attended a dermatologic surgery clinic for the treatment of earlobe keloids were recruited into the study. Earlobe keloids underwent parallel shave excision. Imiquimod 5% cream was applied daily for 8 weeks followed by an observation period of 16 weeks. In patients who presented with bilateral earlobe keloids, paired comparisons of imiquimod versus intralesional steroid injections were performed. RESULTS Eight earlobes were treated with imiquimod 5% cream after parallel keloid removal. Twenty-four weeks after surgery, six (75%) remained recurrence free. Four patients underwent bilateral paired comparisons. At the end of the observation period, two patients (50%) remained recurrence free in the imiquimod-treated areas while experiencing recurrences in the intralesional steroid-treated areas. Local irritation secondary to imiquimod application required rest periods in three cases. In all cases, patients were able to resume therapy and completed the study without further complications. CONCLUSION Although small and uncontrolled, the results of this open-label, pilot study suggest that imiquimod 5% cream may prove to be a reasonably effective adjuvant therapeutic alternative for the prevention of recurrences in excised earlobe keloids.
Collapse
Affiliation(s)
- Rafael F Martin-García
- Department of Dermatology, University of Puerto Rico Medical Sciences Campus, San Juan, Puerto Rico.
| | | |
Collapse
|
39
|
Tso CL, Freije WA, Day A, Chen Z, Merriman B, Perlina A, Lee Y, Dia EQ, Yoshimoto K, Mischel PS, Liau LM, Cloughesy TF, Nelson SF. Distinct transcription profiles of primary and secondary glioblastoma subgroups. Cancer Res 2006; 66:159-67. [PMID: 16397228 DOI: 10.1158/0008-5472.can-05-0077] [Citation(s) in RCA: 161] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Glioblastomas are invasive and aggressive tumors of the brain, generally considered to arise from glial cells. A subset of these cancers develops from lower-grade gliomas and can thus be clinically classified as "secondary," whereas some glioblastomas occur with no prior evidence of a lower-grade tumor and can be clinically classified as "primary." Substantial genetic differences between these groups of glioblastomas have been identified previously. We used large-scale expression analyses to identify glioblastoma-associated genes (GAG) that are associated with a more malignant phenotype via comparison with lower-grade astrocytomas. We have further defined gene expression differences that distinguish primary and secondary glioblastomas. GAGs distinct to primary or secondary tumors provided information on the heterogeneous properties and apparently distinct oncogenic mechanisms of these tumors. Secondary GAGs primarily include mitotic cell cycle components, suggesting the loss of function in prominent cell cycle regulators, whereas primary GAGs highlight genes typical of a stromal response, suggesting the importance of extracellular signaling. Immunohistochemical staining of glioblastoma tissue arrays confirmed expression differences. These data highlight that the development of gene pathway-targeted therapies may need to be specifically tailored to each subtype of glioblastoma.
Collapse
Affiliation(s)
- Cho-Lea Tso
- Department of Human Genetics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Schultze-Mosgau S, Kopp J, Thorwarth M, Rödel F, Melnychenko I, Grabenbauer GG, Amann K, Wehrhan F. Plasminogen activator inhibitor-I–related regulation of procollagen I (α1 and α2) by antitransforming growth factor-β1 treatment during radiation-impaired wound healing. Int J Radiat Oncol Biol Phys 2006; 64:280-8. [PMID: 16377416 DOI: 10.1016/j.ijrobp.2005.09.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2005] [Revised: 08/28/2005] [Accepted: 09/06/2005] [Indexed: 01/18/2023]
Abstract
PURPOSE Plasminogen activator inhibitor (PAI)-1 mediates transforming growth factor-beta1 (TGF-beta1)-related signaling by stimulating collagen Type I synthesis in radiation-impaired wound healing. The regulation of alpha(I)-procollagen is contradictory in fibroblasts of different fibrotic lesions. It is not known whether anti-TGF-beta1 treatment specifically inhibits alpha(I)-procollagen synthesis. We used an experimental wound healing study to address anti-TGF-beta1-associated influence on alpha(I)-procollagen synthesis. METHODS AND MATERIALS A free flap was transplanted into the preirradiated (40 Gy) or nonirradiated neck region of Wistar rats: Group 1 (n = 8) surgery alone; Group 2 (n = 14) irradiation and surgery; Group 3 (n = 8) irradiation and surgery and anti-TGF-beta1 treatment. On the 14th postoperative day, skin samples were processed for fibroblast culture, in situ hybridization for TGF-beta1, immunohistochemistry, and immunoblotting for PAI-1, alpha1/alpha2(I)-procollagen. RESULTS Anti-TGF-beta1 significantly reduced TGF-beta1 mRNA (p < 0.05) and PAI-1 expression (p < 0.05). Anti-TGF-beta1 treatment in vivo significantly reduced alpha1(I)-procollagen protein (p < 0.05) and the number of expressing cells (p < 0.05) in contrast to significantly increased (p < 0.05) alpha2(I)-procollagen expression. CONCLUSION These results emphasize anti-TGF-beta1 treatment to reduce radiation-induced fibrosis by decreasing alpha1(I)-procollagen synthesis in vivo. alpha1(I)-procollagen and alpha2(I)-procollagen might be differentially regulated by anti-TGF-beta1 treatment. Increased TGF-beta signaling in irradiated skin fibroblasts seemed to be reversible, as shown by a reduction in PAI-1 expression after anti-TGF-beta1 treatment.
Collapse
Affiliation(s)
- Stefan Schultze-Mosgau
- Department of Oral and Maxillofacial Surgery/Plastic Surgery, Freidrich-Schiller-University of Jena, Jena, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
LEARNING OBJECTIVES After studying this article, the participant should be able to: 1. Have a greater appreciation of the extent of differences and similarities between keloid and hypertrophic scarring. 2. Have a greater appreciation of the significance of the stage of maturation of a keloid or hypertrophic scar with regard to its morphologic, biochemical, and molecular profile. 3. More critically review basic science research that is based on poorly characterized scar tissue. 4. More critically review clinical studies that are based on poorly characterized scar tissue. BACKGROUND Hypertrophic and keloid scars remain extremely challenging, particularly in their variable response to treatment. The understanding of hypertrophic and keloid scarring is evolving from a position where they were regarded as different stages of the same process to the contemporary perspective of two separate entities. This article reviews the differences in the two forms of scarring and discusses the implications for future research. METHODS The authors conducted a MEDLINE search of all English language reviews linking key words "hypertrophic," "keloid," and "scarring." RESULTS Over the past four decades, there has been considerable clinical and experimental research looking at the biological nature and therapeutic response of keloid and hypertrophic scarring. As more differences are emerging regarding the fundamental biology of the scars, investigators are giving more detailed characterization of their source material. It is evident that even within the broad categories of hypertrophic and keloid scarring there is a heterogenous distribution of pathologic connective tissue matrix biology. CONCLUSION Considerable advances have been made in our understanding of the fundamental biology of scarring. As research methodology becomes even more sophisticated, it will be even more crucial to extensively characterize source material, recognizing major differences not only between keloid and hypertrophic scar but also between scars of varying stages of maturation and histomorphological, biochemical, and molecular variations within individual scars.
Collapse
Affiliation(s)
- Andrew Burd
- Division of Plastic and Reconstructive Surgery, Department of Surgery, The Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong.
| | | |
Collapse
|
42
|
Postsurgical Use of Imiquimod 5% Cream in the Prevention of Earlobe Keloid Recurrences. Dermatol Surg 2005. [DOI: 10.1097/00042728-200511000-00003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Kaur B, Khwaja FW, Severson EA, Matheny SL, Brat DJ, Van Meir EG. Hypoxia and the hypoxia-inducible-factor pathway in glioma growth and angiogenesis. Neuro Oncol 2005; 7:134-53. [PMID: 15831232 PMCID: PMC1871894 DOI: 10.1215/s1152851704001115] [Citation(s) in RCA: 484] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Glioblastomas, like other solid tumors, have extensive areas of hypoxia and necrosis. The importance of hypoxia in driving tumor growth is receiving increased attention. Hypoxia-inducible factor 1 (HIF-1) is one of the master regulators that orchestrate the cellular responses to hypoxia. It is a heterodimeric transcription factor composed of alpha and beta subunits. The alpha subunit is stable in hypoxic conditions but is rapidly degraded in normoxia. The function of HIF-1 is also modulated by several molecular mechanisms that regulate its synthesis, degradation, and transcriptional activity. Upon stabilization or activation, HIF-1 translocates to the nucleus and induces transcription of its downstream target genes. Most important to gliomagenesis, HIF-1 is a potent activator of angiogenesis and invasion through its upregulation of target genes critical for these functions. Activation of the HIF-1 pathway is a common feature of gliomas and may explain the intense vascular hyperplasia often seen in glioblastoma multiforme. Activation of HIF results in the activation of vascular endothelial growth factors, vascular endothelial growth factor receptors, matrix metalloproteinases, plasminogen activator inhibitor, transforming growth factors alpha and beta, angiopoietin and Tie receptors, endothelin-1, inducible nitric oxide synthase, adrenomedullin, and erythropoietin, which all affect glioma angiogenesis. In conclusion, HIF is a critical regulatory factor in the tumor microenvironment because of its central role in promoting proangiogenic and invasive properties. While HIF activation strongly promotes angiogenesis, the emerging vasculature is often abnormal, leading to a vicious cycle that causes further hypoxia and HIF upregulation.
Collapse
Affiliation(s)
| | | | | | | | | | - Erwin G. Van Meir
- Address correspondence to Erwin G. Van Meir, Winship Cancer Institute, Emory University School of Medicine, 1365C Clifton Road, NE, Room C5078, Atlanta, GA 30322, USA (
)
| |
Collapse
|
44
|
Davila HH, Magee TR, Zuniga FI, Rajfer J, Gonzalez-Cadavid NF. Peyronie’s disease associated with increase in plasminogen activator inhibitor in fibrotic plaque. Urology 2005; 65:645-8. [PMID: 15833499 DOI: 10.1016/j.urology.2005.01.010] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2004] [Revised: 01/05/2005] [Accepted: 01/07/2005] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To investigate whether tissue expression of plasminogen activator inhibitor type 1 (PAI-1) is increased in the fibrotic plaque of human Peyronie's disease (PD). Increased tissue levels of PAI-1, an inhibitor of both fibrinolysis and collagenolysis, have been found in a variety of fibrotic conditions. Recently, it was reported that PAI-1 expression was also increased in the fibrotic plaque of an animal model of PD induced by the injection of fibrin into the tunica albuginea (TA) of the penis. METHODS Tissue (n = 10/group) and cells (n = 4/group) obtained from the penile TA plaque of patients with PD or from normal TA were subjected to RNA extraction and real-time reverse transcriptase-polymerase chain reaction. Tissues were also analyzed by immunohistochemistry (n = 8/group) for the detection of PAI-1 expression at the transcription and protein levels. RESULTS A significant 3.5-fold to 16-fold increase was found in both PAI-1 mRNA and protein levels in the human PD plaque and the respective fibroblast cultures compared with the normal non-PD TA. CONCLUSIONS The observed increase in PAI-1 in the human PD plaque agrees with what has been observed in the rat and suggests that PAI-1 may be a key pro-fibrotic factor in the development of human PD.
Collapse
Affiliation(s)
- Hugo H Davila
- Department of Urology, University of California, Los Angeles, School of Medicine, Los Angeles, California, USA
| | | | | | | | | |
Collapse
|
45
|
Chau CH, Clavijo CA, Deng HT, Zhang Q, Kim KJ, Qiu Y, Le AD, Ann DK. Etk/Bmx mediates expression of stress-induced adaptive genes VEGF, PAI-1, and iNOS via multiple signaling cascades in different cell systems. Am J Physiol Cell Physiol 2005; 289:C444-54. [PMID: 15788485 DOI: 10.1152/ajpcell.00410.2004] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We recently showed that Etk/Bmx, a member of the Tec family of nonreceptor protein tyrosine kinases, promotes tight junction formation during chronic hypoxic exposure and augments normoxic VEGF expression via a feedforward mechanism. Here we further characterized Etk's role in potentiating hypoxia-induced gene expression in salivary epithelial Pa-4 cells. Using transient transfection in conditionally activated Etk (DeltaEtk:ER) cells, we demonstrated that Etk enhances hypoxia-response element-dependent reporter activation in normoxia and hypoxia. This Etk-driven reporter activation is ameliorated by treatment with wortmannin or LFM-A13. Using lentivirus-mediated gene delivery and small interfering RNA, we provided direct evidence that hypoxia leads to transient Etk and Akt activation and hypoxia-mediated Akt activation is Etk dependent. Northern blot analyses confirmed that Etk activation led to induction of steady-state mRNA levels of endogenous VEGF and plasminogen activator inhibitor (PAI)-1, a hallmark of hypoxia-mediated gene regulation. We also demonstrated that Etk utilizes a phosphatidylinositol 3-kinase/Akt pathway to promote reporter activation driven by NF-kappaB, another oxygen-sensitive transcription factor, and to augment cytokine-induced inducible nitric oxide synthase expression in endothelial cells. To establish the clinical relevance of Etk-induced, hypoxia-mediated gene regulation, we examined Etk expression in keloid, which has elevated VEGF and PAI-1. We found that Etk is overexpressed in keloid (but not normal skin) tissues. The differential steady-state Etk protein levels were further confirmed in primary fibroblast cultures derived from these tissues, suggesting an Etk role in tissue fibrosis. Our results provide further understanding of Etk function within multiple signaling cascades to govern adaptive cytoprotection against extracellular stress in different cell systems, salivary epithelial cells, brain endothelial cells, and dermal fibroblasts.
Collapse
Affiliation(s)
- Cindy H Chau
- Department of Molecular Pharmacology and Toxicology, School of Medicine, University of Southern California, Los Angeles, California 90033-1049, USA
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Wu Y, Zhang Q, Ann DK, Akhondzadeh A, Duong HS, Messadi DV, Le AD. Increased vascular endothelial growth factor may account for elevated level of plasminogen activator inhibitor-1 via activating ERK1/2 in keloid fibroblasts. Am J Physiol Cell Physiol 2003; 286:C905-12. [PMID: 14644771 DOI: 10.1152/ajpcell.00200.2003] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Keloids are characterized as an "overexuberant" healing response in which disequilibrium between production and catabolism of extracellular matrix (ECM) occurs. Previous studies from our laboratory and others demonstrate an intrinsically higher level of plasminogen activator inhibitor-1 (PAI-1) expression in keloid tissues and cultured fibroblasts compared with normal bordering skin. These findings support the concept that an altered balance of activator and inhibitor activities in the plasminogen system, in particular, an overexpression of PAI-1, may partly contribute to keloid formation and tissue fibrosis. Vascular endothelial growth factor (VEGF) has been implicated as a critical factor in regulating angiogenesis and inflammation under both physiological and pathological conditions. This study was designed to assess whether VEGF plays a role in keloid fibrosis. We report that VEGF was expressed at higher levels in keloid tissues and their derived fibroblasts compared with their associated normal skin. We have further demonstrated that VEGF stimulated the expression of PAI-1, but not urokinase plasminogen activator (uPA), in keloid fibroblasts at both mRNA and protein levels, in a dose- and time-dependent manner. However, treatment of normal skin fibroblasts with VEGF exerted little effects on PAI-1 gene expression. Additionally, we have characterized for the first time that the extracellular signal-regulated kinase (ERK)1/2 signaling pathway is mainly involved in VEGF-induced PAI-1 expression and have demonstrated its potential as a target molecule for modulation of scar fibrosis. These findings suggest that VEGF may play an important role in keloid formation by altering ECM homeostasis toward a state of impaired degradation and excessive accumulation.
Collapse
Affiliation(s)
- Yidi Wu
- Department of Oral and Maxillofacial Surgery, Charles R Drew University of Medicine and Science, Los Angeles, CA 90059, USA
| | | | | | | | | | | | | |
Collapse
|