1
|
Mason W, Levin AM, Buhl K, Ouchi T, Parker B, Tan J, Ashammakhi N, Jones LR. Translational Research Techniques for the Facial Plastic Surgeon: An Overview. Facial Plast Surg 2023; 39:466-473. [PMID: 37339663 DOI: 10.1055/a-2113-5023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023] Open
Abstract
The field of facial plastic and reconstructive surgery (FPRS) is an incredibly diverse, multispecialty field that seeks innovative and novel solutions for the management of physical defects on the head and neck. To aid in the advancement of medical and surgical treatments for these defects, there has been a recent emphasis on the importance of translational research. With recent technological advancements, there are now a myriad of research techniques that are widely accessible for physician and scientist use in translational research. Such techniques include integrated multiomics, advanced cell culture and microfluidic tissue models, established animal models, and emerging computer models generated using bioinformatics. This study discusses these various research techniques and how they have and can be used for research in the context of various important diseases within the field of FPRS.
Collapse
Affiliation(s)
- William Mason
- Department of Otolaryngology, Henry Ford Hospital, Detroit, Michigan
| | - Albert M Levin
- Department of Public Health Science, Henry Ford Health, Detroit, Michigan
- Center for Bioinformatics, Henry Ford Health, Detroit, Michigan
| | - Katherine Buhl
- Department of Otolaryngology, Henry Ford Hospital, Detroit, Michigan
| | - Takahiro Ouchi
- Department of Otolaryngology, Henry Ford Hospital, Detroit, Michigan
| | - Bianca Parker
- Department of Otolaryngology, Henry Ford Hospital, Detroit, Michigan
| | - Jessica Tan
- Department of Otolaryngology, Henry Ford Hospital, Detroit, Michigan
| | - Nureddin Ashammakhi
- Institute for Quantitative Health Science and Engineering, Michigan State University, Michigan
- Department of Biomedical Engineering, College of Engineering, Michigan State University, Michigan
- College of Human Medicine, Michigan State University, Michigan
| | - Lamont R Jones
- Department of Otolaryngology, Henry Ford Hospital, Detroit, Michigan
| |
Collapse
|
2
|
Choi S, Ham S, Lee YI, Kim J, Lee WJ, Lee JH. Silibinin Downregulates Types I and III Collagen Expression via Suppression of the mTOR Signaling Pathway. Int J Mol Sci 2023; 24:14386. [PMID: 37762688 PMCID: PMC10531945 DOI: 10.3390/ijms241814386] [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: 07/04/2023] [Revised: 09/13/2023] [Accepted: 09/20/2023] [Indexed: 09/29/2023] Open
Abstract
Keloid scars are fibro-proliferative conditions characterized by abnormal fibroblast proliferation and excessive extracellular matrix deposition. The mammalian target of the rapamycin (mTOR) pathway has emerged as a potential therapeutic target in keloid disease. Silibinin, a natural flavonoid isolated from the seeds and fruits of the milk thistle, is known to inhibit the mTOR signaling pathway in human cervical and hepatoma cancer cells. However, the mechanisms underlying this inhibitory effect are not fully understood. This in vitro study investigated the effects of silibinin on collagen expression in normal human dermal and keloid-derived fibroblasts. We evaluated the effects of silibinin on the expressions of collagen types I and III and assessed its effects on the suppression of the mTOR signaling pathway. Our findings confirmed elevated mTOR phosphorylation levels in keloid scars compared to normal tissue specimens. Silibinin treatment significantly reduced collagen I and III expressions in normal human dermal and keloid-derived fibroblasts. These effects were accompanied by the suppression of the mTOR signaling pathway. Our findings suggest the potential of silibinin as a promising therapeutic agent for preventing and treating keloid scars. Further studies are warranted to explore the clinical application of silibinin in scar management.
Collapse
Affiliation(s)
- Sooyeon Choi
- Department of Dermatology & Cutaneous Biology, Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.C.); (S.H.); (Y.I.L.)
| | - Seoyoon Ham
- Department of Dermatology & Cutaneous Biology, Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.C.); (S.H.); (Y.I.L.)
| | - Young In Lee
- Department of Dermatology & Cutaneous Biology, Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.C.); (S.H.); (Y.I.L.)
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Republic of Korea; (J.K.); (W.J.L.)
| | - Jihee Kim
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Republic of Korea; (J.K.); (W.J.L.)
- Department of Dermatology, Yongin Severance Hospital, Yonsei University College of Medicine, Yongin 16995, Republic of Korea
| | - Won Jai Lee
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Republic of Korea; (J.K.); (W.J.L.)
- Department of Plastic Surgery, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ju Hee Lee
- Department of Dermatology & Cutaneous Biology, Research Institute, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; (S.C.); (S.H.); (Y.I.L.)
- Scar Laser and Plastic Surgery Center, Yonsei Cancer Hospital, Seoul 03722, Republic of Korea; (J.K.); (W.J.L.)
| |
Collapse
|
3
|
Fertala J, Wang ML, Rivlin M, Beredjiklian PK, Abboud J, Arnold WV, Fertala A. Extracellular Targets to Reduce Excessive Scarring in Response to Tissue Injury. Biomolecules 2023; 13:biom13050758. [PMID: 37238628 DOI: 10.3390/biom13050758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/24/2023] [Accepted: 04/25/2023] [Indexed: 05/28/2023] Open
Abstract
Excessive scar formation is a hallmark of localized and systemic fibrotic disorders. Despite extensive studies to define valid anti-fibrotic targets and develop effective therapeutics, progressive fibrosis remains a significant medical problem. Regardless of the injury type or location of wounded tissue, excessive production and accumulation of collagen-rich extracellular matrix is the common denominator of all fibrotic disorders. A long-standing dogma was that anti-fibrotic approaches should focus on overall intracellular processes that drive fibrotic scarring. Because of the poor outcomes of these approaches, scientific efforts now focus on regulating the extracellular components of fibrotic tissues. Crucial extracellular players include cellular receptors of matrix components, macromolecules that form the matrix architecture, auxiliary proteins that facilitate the formation of stiff scar tissue, matricellular proteins, and extracellular vesicles that modulate matrix homeostasis. This review summarizes studies targeting the extracellular aspects of fibrotic tissue synthesis, presents the rationale for these studies, and discusses the progress and limitations of current extracellular approaches to limit fibrotic healing.
Collapse
Affiliation(s)
- Jolanta Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mark L Wang
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Michael Rivlin
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Pedro K Beredjiklian
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Joseph Abboud
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - William V Arnold
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
- Rothman Institute of Orthopaedics, Thomas Jefferson University Hospital, Philadelphia, PA 19107, USA
| | - Andrzej Fertala
- Department of Orthopaedic Surgery, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| |
Collapse
|
4
|
Ma Y, Liu Z, Miao L, Jiang X, Ruan H, Xuan R, Xu S. Mechanisms underlying pathological scarring by fibroblasts during wound healing. Int Wound J 2023. [PMID: 36726192 DOI: 10.1111/iwj.14097] [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: 11/08/2022] [Accepted: 01/09/2023] [Indexed: 02/03/2023] Open
Abstract
Pathological scarring is an abnormal outcome of wound healing, which often manifests as excessive proliferation and transdifferentiation of fibroblasts (FBs), and excessive deposition of the extracellular matrix. FBs are the most important effector cells involved in wound healing and scar formation. The factors that promote pathological scar formation often act on the proliferation and function of FB. In this study, we describe the factors that lead to abnormal FB formation in pathological scarring in terms of the microenvironment, signalling pathways, epigenetics, and autophagy. These findings suggest that understanding the causes of abnormal FB formation may aid in the development of precise and effective preventive and treatment strategies for pathological scarring that are associated with improved quality of life of patients.
Collapse
Affiliation(s)
- Yizhao Ma
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Zhifang Liu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - LinLin Miao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Xinyu Jiang
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Hongyu Ruan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Rongrong Xuan
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| | - Suling Xu
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, China
| |
Collapse
|
5
|
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
|
6
|
Ahn KJ, Kim JS. TGF-β1 upregulates Sar1a expression and induces procollagen-I secretion in hypertrophic scarring fibroblasts. Open Med (Wars) 2022; 17:1473-1482. [PMID: 36188194 PMCID: PMC9483117 DOI: 10.1515/med-2022-0543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 07/11/2022] [Accepted: 07/20/2022] [Indexed: 11/15/2022] Open
Abstract
Hypertrophic scarring (HTS) is a common fibroproliferative disorder that typically follows thermal and other injuries involving the deep dermis. The underlying pathogenic mechanisms are regulated by transforming growth factor-β (TGF-β); however, the exact mechanisms in HTS have not been elucidated. We conducted this study to explore the cellular signaling mechanisms for expression of Sar1a, a coat protein complex II-associated small GTPase, in HTS fibroblasts (HTSF). We found that Sar1a was upregulated in HTSF as compared to that in normal fibroblasts. Furthermore, stimulation of TGF-β1 increased the expression of Sar1a in HTSF, and small interfering RNA for Sar1a suppressed procollagen-I (PC-I) secretion. Next we investigated the signaling mechanism from TGF-β1 to Sar1a expression and its association with PC-I secretion. In the presence of TGF-β-activated kinase 1 (TAK1), c-Jun N-terminal kinase, or p38 inhibitors, the effect of TGF-β1 on Sar1a expression and PC-I secretion significantly decreased; however, it had no effect on collagen-1A (Col-1A) expression. Further, the inhibitors of Smad3 or extracellular signal-regulated kinases inhibited TGF-β1-induced Col-1A expression but had no effect on PC-I secretion and Sar1a expression. Taken together, our results suggested that TGF-β1 induces Sar1a expression through TAK1 signaling and this signaling event regulates PC-I secretion in HTSF.
Collapse
Affiliation(s)
- Keun Jae Ahn
- Department of Science Education, Jeju National University, Jeju, 63063, Korea
| | - Jun-Sub Kim
- Department of Biotechnology, Korea National University of Transportation, Chungbuk, 27909, Korea
| |
Collapse
|
7
|
Pan X, Chen X, Ren Q, Yue L, Niu S, Li Z, Zhu R, Chen X, Jia Z, Zhen R, Ban J, Chen S. Single-cell transcriptome reveals effects of semaglutide on non-cardiomyocytes of obese mice. Biochem Biophys Res Commun 2022; 622:22-29. [PMID: 35843090 DOI: 10.1016/j.bbrc.2022.07.034] [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: 07/04/2022] [Revised: 07/07/2022] [Accepted: 07/08/2022] [Indexed: 11/02/2022]
Abstract
Non-cardiomyocytes (nonCMs) play an important part in cardiac fibrosis pathophysiology, but the underlying molecular pathways are unknown. Semaglutide has cardioprotective properties, but it is still unclear whether it helps with cardiac fibrosis and what the processes are. The goal of this study is to use single cell transcriptomics approaches to investigate the molecular mechanism of semaglutide's cardioprotective action in obese mice. We found 15 non-CMs, with fibroblasts making up the majority of them. We found eight DEGs that altered significantly following semaglutide treatment by screening for differentially expressed genes (DEGs). DEGs were shown to have biological activities primarily related to extracellular matrix and collagen synthesis and distribution, with Serpinh1 and Pcolce expression being the most dramatically altered. Serpinh1 and Pcolce were mostly found in fibroblasts, which play a key role in the fibrosis of the heart. Furthermore, we discovered that semaglutide lowered cardiac collagen content and alleviated obesity-induced ventricular wall hypertrophy. As a result, our findings show that Serpinh1 and Pcolce, which are expressed by fibroblasts, may play a role in the development of obese cardiac fibrosis. By reducing Serpinh1 and Pcolce expression and delaying cardiac fibrosis, semaglutide may have a cardioprotective effect.
Collapse
Affiliation(s)
- Xiaoyu Pan
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xing Chen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Nephrology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Qingjuan Ren
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Lin Yue
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Shu Niu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Zelin Li
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Ruiyi Zhu
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Xiaoyi Chen
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Zhuoya Jia
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Ruoxi Zhen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Jiangli Ban
- Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, China
| | - Shuchun Chen
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang, Hebei, China; Department of Endocrinology, Hebei General Hospital, Shijiazhuang, Hebei, 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: 4.7] [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
|
Lagoutte P, Bettler E, Vadon-Le Goff S, Moali C. Procollagen C-proteinase enhancer-1 (PCPE-1), a potential biomarker and therapeutic target for fibrosis. Matrix Biol Plus 2021; 11:100062. [PMID: 34435180 PMCID: PMC8377038 DOI: 10.1016/j.mbplus.2021.100062] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
The correct balance between collagen synthesis and degradation is essential for almost every aspect of life, from development to healthy aging, reproduction and wound healing. When this balance is compromised by external or internal stress signals, it very often leads to disease as is the case in fibrotic conditions. Fibrosis occurs in the context of defective tissue repair and is characterized by the excessive, aberrant and debilitating deposition of fibril-forming collagens. Therefore, the numerous proteins involved in the biosynthesis of fibrillar collagens represent a potential and still underexploited source of therapeutic targets to prevent fibrosis. One such target is procollagen C-proteinase enhancer-1 (PCPE-1) which has the unique ability to accelerate procollagen maturation by BMP-1/tolloid-like proteinases (BTPs) and contributes to trigger collagen fibrillogenesis, without interfering with other BTP functions or the activities of other extracellular metalloproteinases. This role is achieved through a fine-tuned mechanism of action that is close to being elucidated and offers promising perspectives for drug design. Finally, the in vivo data accumulated in recent years also confirm that PCPE-1 overexpression is a general feature and early marker of fibrosis. In this review, we describe the results which presently support the driving role of PCPE-1 in fibrosis and discuss the questions that remain to be solved to validate its use as a biomarker or therapeutic target.
Collapse
Key Words
- ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs
- AS, aortic valve stenosis
- BMP, bone morphogenetic protein
- Biomarker
- CKD, chronic kidney disease
- CP, C-propeptide
- CUB, complement, Uegf, BMP-1
- CVD, cardiovascular disease
- Collagen
- DMD, Duchenne muscular dystrophy
- ECM, extracellular matrix
- EGF, epidermal growth factor
- ELISA, enzyme-linked immunosorbent assay
- Fibrillogenesis
- Fibrosis
- HDL, high-density lipoprotein
- HSC, hepatic stellate cell
- HTS, hypertrophic scar
- IPF, idiopathic pulmonary fibrosis
- LDL, low-density lipoprotein
- MI, myocardial infarction
- MMP, matrix metalloproteinase
- NASH, nonalcoholic steatohepatitis
- NTR, netrin
- OPMD, oculopharyngeal muscular dystrophy
- PABPN1, poly(A)-binding protein nuclear 1
- PCP, procollagen C-proteinase
- PCPE, procollagen C-proteinase enhancer
- PNP, procollagen N-proteinase
- Proteolysis
- SPC, subtilisin proprotein convertase
- TGF-β, transforming growth-factor β
- TIMP, tissue inhibitor of metalloproteinases
- TSPN, thrombospondin-like N-terminal
- Therapeutic target
- eGFR, estimated glomerular filtration rate
- mTLD, mammalian tolloid
- mTLL, mammalian tolloid-like
Collapse
Affiliation(s)
- Priscillia Lagoutte
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Emmanuel Bettler
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Sandrine Vadon-Le Goff
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| | - Catherine Moali
- University of Lyon, CNRS, Tissue Biology and Therapeutic Engineering Laboratory, LBTI, UMR5305, F-69367 Lyon, France
| |
Collapse
|
10
|
Signatures of Dermal Fibroblasts from RDEB Pediatric Patients. Int J Mol Sci 2021; 22:ijms22041792. [PMID: 33670258 PMCID: PMC7918539 DOI: 10.3390/ijms22041792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/05/2021] [Accepted: 02/08/2021] [Indexed: 12/13/2022] Open
Abstract
The recessive form of dystrophic epidermolysis bullosa (RDEB) is a debilitating disease caused by impairments in the junctions of the dermis and the basement membrane of the epidermis. Mutations in the COL7A1 gene induce multiple abnormalities, including chronic inflammation and profibrotic changes in the skin. However, the correlations between the specific mutations in COL7A1 and their phenotypic output remain largely unexplored. The mutations in the COL7A1 gene, described here, were found in the DEB register. Among them, two homozygous mutations and two cases of compound heterozygous mutations were identified. We created the panel of primary patient-specific RDEB fibroblast lines (FEB) and compared it with control fibroblasts from healthy donors (FHC). The set of morphological features and the contraction capacity of the cells distinguished FEB from FHC. We also report the relationships between the mutations and several phenotypic traits of the FEB. Based on the analysis of the available RNA-seq data of RDEB fibroblasts, we performed an RT-qPCR gene expression analysis of our cell lines, confirming the differential status of multiple genes while uncovering the new ones. We anticipate that our panels of cell lines will be useful not only for studying RDEB signatures but also for investigating the overall mechanisms involved in disease progression.
Collapse
|
11
|
Pan Y, Chen Z, Qi F, Liu J. Identification of drug compounds for keloids and hypertrophic scars: drug discovery based on text mining and DeepPurpose. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:347. [PMID: 33708974 PMCID: PMC7944324 DOI: 10.21037/atm-21-218] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Background Keloids (KL) and hypertrophic scars (HS) are forms of abnormal cutaneous scarring characterized by excessive deposition of extracellular matrix and fibroblast proliferation. Currently, the efficacy of drug therapies for KL and HS is limited. The present study aimed to investigate new drug therapies for KL and HS by using computational methods. Methods Text mining and GeneCodis were used to mine genes closely related to KL and HS. Protein-protein interaction analysis was performed using Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) and Cytoscape. The selection of drugs targeting the genes closely related to KL and HS was carried out using Pharmaprojects. Drug-target interaction prediction was performed using DeepPurpose, through which candidate drugs with the highest predicted binding affinity were finally obtained. Results Our analysis using text mining identified 69 KL- and HS-related genes. Gene enrichment analysis generated 25 genes, representing 7 pathways and 130 targeting drugs. DeepPurpose recommended 14 drugs as the final drug list, including 2 phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) inhibitors, 10 prostaglandin-endoperoxide synthase 2 (PTGS2) inhibitors and 2 vascular endothelial growth factor A (VEGFA) antagonists. Conclusions Drug discovery using in silico text mining and DeepPurpose may be a powerful and effective way to identify drugs targeting the genes related to KL and HS.
Collapse
Affiliation(s)
- Yuyan Pan
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhiwei Chen
- Big Data and Artificial Intelligence Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fazhi Qi
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiaqi Liu
- Department of Plastic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.,Artificial Intelligence Center for Plastic Surgery and Cutaneous Soft Tissue Cancers, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Xie F, Teng L, Xu J, Lu J, Zhang C, Yang L, Ma X, Zhao M. Adipose-derived mesenchymal stem cells inhibit cell proliferation and migration and suppress extracellular matrix synthesis in hypertrophic-scar and keloid fibroblasts. Exp Ther Med 2021; 21:139. [PMID: 33456506 PMCID: PMC7791925 DOI: 10.3892/etm.2020.9571] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 03/26/2020] [Indexed: 12/13/2022] Open
Abstract
Pathological scars occur during skin wound healing, and the use of adipose-derived stem cells (ADSCs) is one of the various treatments. The present study aimed to investigate the in vitro effects of ADSCs on the biological properties of hypertrophic scar fibroblasts (HSFs) and keloid fibroblasts (KFs), such as proliferation, migration, and the synthesis of extracellular matrix proteins. Transwell chambers were used to establish a co-culture system of ADSCs with normal skin fibroblasts (NFs), HSFs or KFs. The effect of ADSCs on the proliferation of fibroblasts was evaluated by CCK8 measurement, while the migration ability of fibroblasts was assessed using cell scratch assay. The expression of extracellular matrix proteins was measured by immunoblotting. Co-culture of NFs with ADSCs did not affect cell proliferation and migration, nor the expression of extracellular matrix proteins [collagen-I, collagen-III, fibronectin (FN) and α-smooth muscle actin (α-SMA)] in NFs. However, as with the inhibitor SB431542, ADSCs significantly inhibited cell proliferation and migration and the expression of extracellular matrix proteins (collagen-I, collagen-III, FN and α-SMA), but also suppressed the protein expression of transforming growth factor β1 (TGF-β1), phosphorylated (p-) mothers against decapentaplegic homolog (Smad) 2, p-Smad3 and Smad7 in HSFs and KFs. The results show that ADSCs inhibited cell proliferation and migration and the expression of extracellular matrix proteins in HSCs and KFs in vitro, possibly through inhibition of the TGF-β1/Smad pathway.
Collapse
Affiliation(s)
- Fang Xie
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| | - Li Teng
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| | - Jiajie Xu
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| | - Jianjian Lu
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| | - Chao Zhang
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| | - Liya Yang
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| | - Xiaoyang Ma
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| | - Minghao Zhao
- Cranio-Maxillo-Facial Surgery Department 2, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100144, P.R. China
| |
Collapse
|
13
|
Mukhatayev Z, Ostapchuk YO, Fang D, Le Poole IC. Engineered antigen-specific regulatory T cells for autoimmune skin conditions. Autoimmun Rev 2021; 20:102761. [PMID: 33476816 DOI: 10.1016/j.autrev.2021.102761] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 11/28/2020] [Indexed: 12/15/2022]
Abstract
Regulatory T cells (Tregs) are a subset of T cells responsible for the regulation of immune responses, thereby maintaining immune homeostasis and providing immune tolerance to both self and non-self-antigens. An increasing number of studies revealed Treg numbers and functions in a variety of autoimmune diseases. Treg deficiency can cause the development of several autoimmune skin diseases including vitiligo, alopecia areata, pemphigoid and pemphigus, psoriasis, and systemic sclerosis. Many clinical trials have been performed for autoimmune conditions using polyclonal Tregs, but efficiency can be significantly improved using antigen-specific Tregs engineered using T cell receptor (TCR) or chimeric antigen receptor (CAR) constructs. In this review, we systematically reviewed altered frequencies, impaired functions, and phenotypic features of Tregs in autoimmune skin conditions. We also summarized new advances in TCR and CAR based antigen-specific Tregs tested both in animal models and in clinics. The advantages and limitations of each approach were carefully discussed emphasizing possible clinical relevance to patients with autoimmune skin diseases. Moreover, we have reviewed potential approaches for engineering antigen-specific Tregs, and strategies for overcoming possible hurdles in clinical applications. Thereby, antigen-specific Tregs can be infused using autologous adoptive cell transfer to restore Treg numbers and to provide local immune tolerance for autoimmune skin disorders.
Collapse
Affiliation(s)
- Zhussipbek Mukhatayev
- Department of Dermatology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA; Department of Biology and Biotechnology, Al-Farabi Kazakh National University, Almaty, Kazakhstan; M.A. Aitkhozhin's Institute of Molecular Biology and Biochemistry, Almaty, Kazakhstan
| | | | - Deyu Fang
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - I Caroline Le Poole
- Department of Dermatology, Northwestern University, Chicago, IL, USA; Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
14
|
Limandjaja GC, Niessen FB, Scheper RJ, Gibbs S. Hypertrophic scars and keloids: Overview of the evidence and practical guide for differentiating between these abnormal scars. Exp Dermatol 2021; 30:146-161. [PMID: 32479693 PMCID: PMC7818137 DOI: 10.1111/exd.14121] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 05/21/2020] [Accepted: 05/25/2020] [Indexed: 12/12/2022]
Abstract
Although hypertrophic scars and keloids both generate excessive scar tissue, keloids are characterized by their extensive growth beyond the borders of the original wound, which is not observed in hypertrophic scars. Whether or not hypertrophic scars and keloids are two sides of the same coin or in fact distinct entities remains a topic of much debate. However, proper comparison between the two ideally occurs within the same study, but this is the exception rather than the rule. For this reason, the goal of this review was to summarize and evaluate all publications in which both hypertrophic scars and keloids were studied and compared to one another within the same study. The presence of horizontal growth is the mainstay of the keloid diagnosis and remains the strongest argument in support of keloids and hypertrophic scars being distinct entities, and the histopathological distinction is less straightforward. Keloidal collagen remains the strongest keloid parameter, but dermal nodules and α-SMA immunoreactivity are not limited to hypertrophic scars alone. Ultimately, the current hypertrophic scars-keloid differences are mostly quantitative in nature rather than qualitative, and many similar abnormalities exist in both lesions. Nonetheless, the presence of similarities does not equate the absence of fundamental differences, some of which may not yet have been uncovered given how much we still have to learn about the processes involved in normal wound healing. It therefore seems pertinent to continue treating hypertrophic scars and keloids as separate entities, until such a time as new findings more decisively convinces us otherwise.
Collapse
Affiliation(s)
- Grace C. Limandjaja
- Department of Molecular Cell Biology and ImmunologyAmsterdam University Medical Centre (location VUmc)Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Frank B. Niessen
- Department of Plastic SurgeryAmsterdam University Medical Centre (location VUmc)Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Rik J. Scheper
- Department of PathologyAmsterdam University Medical Centre (location VUmc)Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Susan Gibbs
- Department of Molecular Cell Biology and ImmunologyAmsterdam University Medical Centre (location VUmc)Vrije Universiteit AmsterdamAmsterdamThe Netherlands
- Department of Oral Cell BiologyAcademic Centre for Dentistry (ACTA)University of Amsterdam and Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
15
|
Ekstein SF, Wyles SP, Moran SL, Meves A. Keloids: a review of therapeutic management. Int J Dermatol 2020; 60:661-671. [PMID: 32905614 DOI: 10.1111/ijd.15159] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/20/2022]
Abstract
Keloid scar formation arises from a disorganized fibroproliferative collagen response that extends beyond the original wound margins because of excessive production of extracellular matrix (ECM). Despite treatment options for keloid scars including medical and surgical therapies, such as intralesional steroid injection and surgical excision, the recurrence rate remains high. Herein we consolidate recently published narrative reviews, systematic reviews, and meta-analyses to provide an overview of updated treatment recommendations for keloidal scar formation. PubMed search engine was used to access the MEDLINE database to investigate updates regarding keloid incidence and treatment. More than 100 articles were reviewed. Keloid management remains a multimodal approach. There continues to be no gold standard of treatment that provides a consistently low recurrence rate; however, the increasing number of available treatments and synergistic combinations of these treatments (i.e., laser-based devices in combination with intralesional steroids, or 5-fluorouracil (5-FU) in combination with steroid therapy) is showing favorable results. Future studies could target the efficacy of novel treatment modalities (i.e., autologous fat grafting or stem cell-based therapies) for keloid management. This review article provides updated treatment guidelines for keloids and discusses insight into management to assist patient-focused, evidence-based clinical decision making.
Collapse
Affiliation(s)
- Samuel F Ekstein
- Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Steven L Moran
- Division of Plastic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | | |
Collapse
|
16
|
Janikowska G, Kurzeja E, Janikowski M, Strzałka-Mrozik B, Pyka-Pająk A, Janikowski T. The Effect of Cyclosporine A on Dermal Fibroblast Cell - Transcriptomic Analysis of Inflammatory Response Pathway. Curr Pharm Biotechnol 2020; 21:1213-1223. [PMID: 32297577 DOI: 10.2174/1389201021666200416103928] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 03/25/2020] [Accepted: 03/25/2020] [Indexed: 01/21/2023]
Abstract
BACKGROUND The first immunosuppressive drug - cyclosporine A (CsA) has many unquestioned merits in maintaining organ transplants in patients, as well as, in the treatment of many inflammatory diseases, also associated with cutaneous manifestations. The main task of this drug is to suppress the inflammatory response at the sites of action, which is not well known. OBJECTIVE The objective of this study was to evaluate the influence of CsA in therapeutic concentration on the expression of genes associated with the inflammatory response pathway in normal human dermal fibroblasts (NHDF; CC-2511), and this study attempted to determine the mechanism of its action. METHODS The cytotoxicity MTT test was performed. The expression of the inflammatory response pathway genes was determined using HG-U133A_2.0 oligonucleotide microarrays. Statistical analysis was performed by GeneSpring 13.0 software using the PL-Grid platform. RESULTS Among the 5,300 mRNA, only 573 were changed significantly in response to CsA compared to the control fibroblasts (P≤0.05). CsA inhibited the expression of most genes associated with the inflammatory response in NHDFs. There were only 19 genes with a fold change (FC) lower than -2.0, among which EGR1, FOS, PBK, CDK1 and TOP2A had the lowest expression, as did CXCL2 which can directly impact inflammation. Furthermore, ZNF451 was strongly induced, and COL1A1, COL3A1, IL33, TNFRSFs were weakly up-regulated (FC lower than 2.0). CONCLUSION The CsA in therapeutic concentration influences the genes linked to the inflammatory response (in the transcriptional level) in human dermal fibroblasts. The findings suggest that the potential mechanism of CsA action in this concentration and on these genes can be associated with a profibrotic and proapoptotic, and genotoxic effects.
Collapse
Affiliation(s)
- Grażyna Janikowska
- Department of Analytical Chemistry, Medical University of Silesia in Katowice, Katowice, Poland
| | - Ewa Kurzeja
- Department of Analytical Chemistry, Medical University of Silesia in Katowice, Katowice, Poland
| | - Marcin Janikowski
- Student Scientific Club at the Department of Molecular Biology, Medical University of Silesia in Katowice, Katowice, Poland
| | | | - Alina Pyka-Pająk
- Department of Analytical Chemistry, Medical University of Silesia in Katowice, Katowice, Poland
| | | |
Collapse
|
17
|
Easter Island and Plastic Surgery in the Light of a Historical Study. J Craniofac Surg 2020; 31:601-603. [PMID: 32049905 DOI: 10.1097/scs.0000000000006247] [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] Open
|
18
|
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: 2.2] [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
|
19
|
Dohi T, Padmanabhan J, Akaishi S, Than PA, Terashima M, Matsumoto NN, Ogawa R, Gurtner GC. The Interplay of Mechanical Stress, Strain, and Stiffness at the Keloid Periphery Correlates with Increased Caveolin-1/ROCK Signaling and Scar Progression. Plast Reconstr Surg 2019; 144:58e-67e. [PMID: 31246819 DOI: 10.1097/prs.0000000000005717] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
BACKGROUND Fibroproliferative disorders result in excessive scar formation, are associated with high morbidity, and cost billions of dollars every year. Of these, keloid disease presents a particularly challenging clinical problem because the cutaneous scars progress beyond the original site of injury. Altered mechanotransduction has been implicated in keloid development, but the mechanisms governing scar progression into the surrounding tissue remain unknown. The role of mechanotransduction in keloids is further complicated by the differential mechanical properties of keloids and the surrounding skin. METHODS The authors used human mechanical testing, finite element modeling, and immunohistologic analyses of human specimens to clarify the complex interplay of mechanical stress, strain, and stiffness in keloid scar progression. RESULTS Changes in human position (i.e., standing, sitting, and supine) are correlated to dynamic changes in local stress/strain distribution, particularly in regions with a predilection for keloids. Keloids are composed of stiff tissue, which displays a fibrotic phenotype with relatively low proliferation. In contrast, the soft skin surrounding keloids is exposed to high mechanical strain that correlates with increased expression of the caveolin-1/rho signaling via rho kinase mechanotransduction pathway and elevated inflammation and proliferation, which may lead to keloid progression. CONCLUSIONS The authors conclude that changes in human position are strongly correlated with mechanical loading of the predilection sites, which leads to increased mechanical strain in the peripheral tissue surrounding keloids. Furthermore, increased mechanical strain in the peripheral tissue, which is the site of keloid progression, was correlated with aberrant expression of caveolin-1/ROCK signaling pathway. These findings suggest a novel mechanism for keloid progression.
Collapse
Affiliation(s)
- Teruyuki Dohi
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| | - Jagannath Padmanabhan
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| | - Satoshi Akaishi
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| | - Peter A Than
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| | - Masao Terashima
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| | - Noriko N Matsumoto
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| | - Rei Ogawa
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| | - Geoffrey C Gurtner
- From the Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine; the Department of Plastic, Reconstructive and Aesthetic Surgery, Nippon Medical School; and the Department of Civil and Environmental Engineering, Stanford University School of Engineering
| |
Collapse
|
20
|
Gohar O, Weiss T, Wineman E, Kessler E. Ascorbic Acid Promotes Procollagen C‐Proteinase Enhancer 1 Expression, Secretion, and Cell Membrane Localization. Anat Rec (Hoboken) 2019; 303:1670-1679. [DOI: 10.1002/ar.24182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2018] [Revised: 12/31/2018] [Accepted: 01/18/2019] [Indexed: 01/03/2023]
Affiliation(s)
- Ofra Gohar
- Tel Aviv University Sackler Faculty of MedicineMaurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center Tel‐Hashomer Israel
| | - Tali Weiss
- Tel Aviv University Sackler Faculty of MedicineMaurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center Tel‐Hashomer Israel
| | - Eitan Wineman
- Tel Aviv University Sackler Faculty of MedicineMaurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center Tel‐Hashomer Israel
| | - Efrat Kessler
- Tel Aviv University Sackler Faculty of MedicineMaurice and Gabriela Goldschleger Eye Research Institute, Sheba Medical Center Tel‐Hashomer Israel
| |
Collapse
|
21
|
Analysis of Keloid Response to 5-Fluorouracil Treatment and Long-Term Prevention of Keloid Recurrence. Plast Reconstr Surg 2019; 143:490-494. [PMID: 30531622 DOI: 10.1097/prs.0000000000005257] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Keloids are benign fibroproliferative skin tumors that can cause disfigurement and disability. Although they frequently recur after excision or medical management and can affect 6 to 16 percent of African Americans, there is no gold standard therapy. Keloids are challenging to study because there are no animal or in vitro models of this disorder. This makes it very difficult to validate data from treated tissue samples or cells and develop targeted therapies for this disease. In this study, the authors demonstrate that intralesional 5-fluorouracil injection after keloid excision prevents recurrence for 2 years, with no reported adverse events. The authors analyze the expression of treated and untreated biopsy specimens of the same keloids in their native context to capture insights that may be missed by in vitro cell culture models and correct for intrakeloid variability. Random forest analysis of the microarray data dramatically increased the statistical power of the authors' results, permitting hypothesis-free creation of a gene expression profile of 5-fluorouracil-treated keloids. Through this analysis, the authors found a set of genes, including YAP1 and CCL-2, whose expression changes predict 5-fluorouracil therapy status and include genes that have not previously been associated with keloid biology and are of unknown function. The authors further describe keloid heterogeneity for the first time using multidimensional analysis of their microarray results. The methods and tools the authors developed in this research may overcome some of the challenges in studying keloids and developing effective treatments for this disease. CLINICAL QUESTION/LEVEL OF EVIDENCE:: Therapeutic, V.
Collapse
|
22
|
Yeo DC, Wiraja C, Paller AS, Mirkin CA, Xu C. Abnormal scar identification with spherical-nucleic-acid technology. Nat Biomed Eng 2019. [PMID: 30936446 DOI: 10.1038/s41551-018-0218-x.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The accurate diagnosis of scar type and severity relies on histopathology of biopsied tissue, which is invasive and time-consuming, causes discomfort and may exacerbate scarring. Here, we show that imaging nanoprobes for the live-cell detection of intracellular messenger RNA (mRNA) (also known as NanoFlares) enable measurements of the expression of connective tissue growth factor (CTGF) as a visual indicator of hypertrophic scars and keloids. During cell culture, NanoFlares enabled the distinction of hypertrophic and keloidal fibroblasts from normal fibroblasts, and the detection of changes in CTGF expression resulting from the regulatory effects of transforming growth factor-β (TGF-β) agonists and TGF-β antagonists. We also applied the NanoFlares topically to the skin of live mice and rabbits, and to ex vivo human skin models. Transepidermal penetration of the NanoFlares enabled the visual and spectroscopic quantification of underlying abnormal fibroblasts on the basis of CTGF mRNA expression. Our proof-of-concept studies of topically applied NanoFlare technology as a means of biopsy-free scar diagnosis may eventually inform therapeutic decisions on the basis of the mRNA-expression patterns of skin disorders.
Collapse
Affiliation(s)
- David C Yeo
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Christian Wiraja
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Amy S Paller
- NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore, Singapore. .,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Chad A Mirkin
- NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore, Singapore. .,Department of Chemistry, Northwestern University, Evanston, IL, USA. .,International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore. .,NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
23
|
Abstract
Fibrosis is characterized by excessive deposition of collagen and additional extracellular matrix (ECM) components in response to chronic injuries. Liver fibrosis often results from chronic hepatitis C virus infection and alcohol abuse that can deteriorate to cirrhosis and liver failure. Current noninvasive diagnostic methods of liver fibrosis are limited in their ability to detect and differentiate between early and intermediate stages of fibrosis. New biomarkers of fibrosis that reflect ECM turnover are therefore badly needed. Procollagen C-proteinase enhancer 1 (PCPE-1), a connective tissue glycoprotein that functions as a positive regulator of C-terminal procollagen processing and subsequent collagen fibril assembly, is a promising candidate. Its tissue distribution and expression profile overlap those of collagen, and its expression in fibrosis is upregulated in parallel to the increase in collagen expression. The potential of PCPE-1 as a biomarker of liver fibrosis was recently established using a CCl4 mouse model of liver fibrosis by showing that the increase in collagen and PCPE-1 content in the fibrotic mouse liver was reflected by elevated plasma levels of PCPE-1. This was achieved using a newly developed highly sensitive, specific, accurate, and reproducible ELISA for mouse PCPE-1, which is based on commercially available antibodies and is offered as a new research tool in the field. A similar ELISA test was developed for human PCPE-1, and preliminary results with plasma from liver fibrosis patients revealed increased plasma concentrations of PCPE-1 in some patients. The protocols of both ELISA tests are outlined herein in great detail to permit their application by any laboratory with similar interests.
Collapse
Affiliation(s)
- Efrat Kessler
- Maurice and Gabriela Goldschleger Eye Research Institute, Tel Aviv University Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, Israel. .,Clinical Biochemistry and Pharmacology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel.
| | - Eyal Hassoun
- Clinical Biochemistry and Pharmacology Institute, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
24
|
Özkan G, Güzel S, Atar RV, Fidan Ç, Kara SP, Ulusoy Ş. Elevated serum levels of procollagen C-proteinase enhancer-1 in patients with chronic kidney disease is associated with a declining glomerular filtration rate. Nephrology (Carlton) 2018; 24:938-942. [PMID: 30393914 DOI: 10.1111/nep.13521] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/30/2018] [Indexed: 02/06/2023]
Abstract
BACKGROUND Procollagen C-proteinase enhancer-1 (PCPE-1) is a 55 kDa glycoprotein, which increases the activity of procollagen C-proteinases that break down C-terminal propeptides. Studies have shown that PCPE-1 is involved in the fibrotic process that occurs in various tissues and organs. Our review of the literature revealed no data concerning the relation between PCPE-1 and chronic kidney disease (CKD). The purpose of this study was to determine PCPE-1 levels in CKD. METHODS One hundred thirty-one CKD patients and 34 healthy controls were included in our study. Demographic data were recorded, and routine biochemical tests were performed. Blood specimens were collected for PCPE-1 investigation. Demographic data, biochemical test results and PCPE-1 levels were compared between the control and patient groups. Parameters affecting PCPE-1 levels in our patient group were assessed. RESULTS Procollagen C-proteinase enhancer-1 levels were significantly higher in our patient group compared to the control group. Parameters affecting PCPE-1 elevation in the patient group were identified as systolic blood pressure, blood urea nitrogen, phosphorus, haemoglobin, intact parathormone levels, glomerular filtration rate and body mass index. CONCLUSION We determined high PCPE-1 levels in CKD patients. PCPE-1 levels being negatively correlated with glomerular filtration rate suggests that PCPE-1 may be associated with progression in CKD patients.
Collapse
Affiliation(s)
- Gülsüm Özkan
- Department of Nephrology, Namık Kemal University, School of Medicine, Tekirdağ, Turkey
| | - Savaş Güzel
- Department of Biochemistry, Namık Kemal University, School of Medicine, Tekirdağ, Turkey
| | - Reşit V Atar
- Department of Internal Medicine, Namık Kemal University, School of Medicine, Tekirdağ, Turkey
| | - Çiğdem Fidan
- Department of Biochemistry, Namık Kemal University, School of Medicine, Tekirdağ, Turkey
| | - Sonat P Kara
- Department of Internal Medicine, Namık Kemal University, School of Medicine, Tekirdağ, Turkey
| | - Şükrü Ulusoy
- Department of Nephrology, Karadeniz Technical University, School of Medicine, Trabzon, Turkey
| |
Collapse
|
25
|
Abstract
In this issue of Structure, Pulido et al. (2018) determine the crystal structure of procollagen C-proteinase enhancer-1 (PCPE-1)/procollagen III complex and identify that PCPE-1 unwinds the stalk of the procollagen III trimer, liberating a single chain to facilitate binding and cleavage by BMP-1 proteinases for subsequent fibrillar collagen assembly.
Collapse
Affiliation(s)
- Michael P Lockhart-Cairns
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Clair Baldock
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK.
| |
Collapse
|
26
|
Evaluation of serum procollagen C-proteinase enhancer 1 level as a fibrosis marker in patients with chronic hepatitis B. Eur J Gastroenterol Hepatol 2018; 30:918-924. [PMID: 29683980 DOI: 10.1097/meg.0000000000001123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIM Although liver biopsy has long been considered the gold standard for staging fibrosis, because of the disadvantages and risks of biopsy, several noninvasive processes such as serum biomarkers have been introduced for the assessment of liver fibrosis. The aim of this study was to assess the diagnostic value of serum procollagen C-proteinase enhancer 1 (PCPE-1) as a noninvasive fibrosis marker in treatment-naive chronic hepatitis B patients. PATIENTS AND METHODS This study included 126 patients with biopsy-proven hepatitis B and 50 healthy controls. Fibrosis stage was determined using the Ishak scoring system. The PCPE-1 level was measured using the enzyme-linked immunosorbent assay assay, and the aspartate aminotransferase to platelet ratio index and the FIB-4 index were calculated using the formulas described in Appendix 1 (Supplemental digital content 1, http://links.lww.com/EJGH/A277). RESULTS Serum PCPE-1 levels of chronic hepatitis B patients were found to be significantly lower than those of the healthy control group (4.49±2.74 vs. 42.9±59.6 pg/ml, respectively, P<0.001). There was a statistically significant negative correlation between serum PCPE-1 level and fibrosis stage (P=0.011; r=-0.226). A statistically significant negative correlation was found between serum PCPE-1 level and necroinflammatory activity (P=0.030; r=-0.194). PCPE-1 levels of patients with liver fibrosis scores of F1-2 were statistically significantly lower than those of the healthy control group (P<0.001) (area under the receiver operating characteristic: 0.955). The area under the receiver operating characteristic of the PCPE-1 level was 0.615 for the prediction of fibrosis (F0 vs. F1-6) (P=0.039). CONCLUSION Serum PCPE-1 might be used as a noninvasive marker of liver fibrosis. Further animal and human studies are needed to assess the utility of this marker.
Collapse
|
27
|
Le Poole IC, Mehrotra S. Replenishing Regulatory T Cells to Halt Depigmentation in Vitiligo. J Investig Dermatol Symp Proc 2018; 18:S38-S45. [PMID: 28941492 DOI: 10.1016/j.jisp.2016.10.023] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 09/19/2016] [Indexed: 02/07/2023]
Abstract
Vitiligo is a cutaneous autoimmune disease, especially devastating to patients with darker skin tones because of the contrast between unaffected and lesional skin. We studied immune cells infiltrating vitiligo skin and found very few regulatory T cells (Tregs). Vitiligo was not associated with a reduced frequency or function of circulating Tregs. To manipulate Treg function, we used mouse models expressing melanocyte-reactive TCRs, following changes in pelage color. We also isolated splenocytes to measure Treg function and evaluated cutaneous Treg abundance. Even small numbers of Tregs transferred into depigmenting mice could effectively interfere with depigmentation. The same holds true for treatment with rapamycin, readily translatable for use in human patients; such treatment may be well tolerated. Because vitiligo skin is relatively devoid of cells that produce the chemokine CCL22, whereas circulating Tregs express normal levels of its receptor CCR4, we overexpressed Ccl22 in the skin of vitiligo-prone mice to assess the resulting levels of depigmentation. Markedly reduced depigmentation was accompanied by Treg infiltration to the skin. With several options available to support a healthy balance between Tregs and effector T cells, the next challenge will be to render such treatment antigen specific and avoid general immunosuppression.
Collapse
Affiliation(s)
- I Caroline Le Poole
- Department of Pathology and Microbiology/Immunology, Oncology Research Institute, Loyola University, Chicago, Illinois, USA.
| | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
28
|
Yeo DC, Wiraja C, Paller AS, Mirkin CA, Xu C. Abnormal scar identification with spherical-nucleic-acid technology. Nat Biomed Eng 2018; 2:227-238. [PMID: 30936446 DOI: 10.1038/s41551-018-0218-x] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 03/12/2018] [Indexed: 12/11/2022]
Abstract
The accurate diagnosis of scar type and severity relies on histopathology of biopsied tissue, which is invasive and time-consuming, causes discomfort and may exacerbate scarring. Here, we show that imaging nanoprobes for the live-cell detection of intracellular messenger RNA (mRNA) (also known as NanoFlares) enable measurements of the expression of connective tissue growth factor (CTGF) as a visual indicator of hypertrophic scars and keloids. During cell culture, NanoFlares enabled the distinction of hypertrophic and keloidal fibroblasts from normal fibroblasts, and the detection of changes in CTGF expression resulting from the regulatory effects of transforming growth factor-β (TGF-β) agonists and TGF-β antagonists. We also applied the NanoFlares topically to the skin of live mice and rabbits, and to ex vivo human skin models. Transepidermal penetration of the NanoFlares enabled the visual and spectroscopic quantification of underlying abnormal fibroblasts on the basis of CTGF mRNA expression. Our proof-of-concept studies of topically applied NanoFlare technology as a means of biopsy-free scar diagnosis may eventually inform therapeutic decisions on the basis of the mRNA-expression patterns of skin disorders.
Collapse
Affiliation(s)
- David C Yeo
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Christian Wiraja
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Amy S Paller
- NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore, Singapore. .,Department of Dermatology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA.
| | - Chad A Mirkin
- NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore, Singapore. .,Department of Chemistry, Northwestern University, Evanston, IL, USA. .,International Institute for Nanotechnology, Northwestern University, Evanston, IL, USA.
| | - Chenjie Xu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore. .,NTU-Northwestern Institute for Nanomedicine, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
29
|
Li Q, Qin Z, Nie F, Bi H, Zhao R, Pan B, Ma J, Xie X. Metabolic reprogramming in keloid fibroblasts: Aerobic glycolysis and a novel therapeutic strategy. Biochem Biophys Res Commun 2018; 496:641-647. [DOI: 10.1016/j.bbrc.2018.01.068] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 01/10/2018] [Indexed: 10/18/2022]
|
30
|
Massoudi D, Germer CJ, Glisch JM, Greenspan DS. Procollagen C-proteinase enhancer 1 (PCPE-1) functions as an anti-angiogenic factor and enhances epithelial recovery in injured cornea. Cell Tissue Res 2017; 370:461-476. [PMID: 28936615 DOI: 10.1007/s00441-017-2689-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Accepted: 08/28/2017] [Indexed: 12/19/2022]
Abstract
Procollagen C-proteinase enhancer 1 (PCPE-1) has been characterized as a protein capable of enhancing the activity of bone morphogenetic protein 1/tolloid-like proteinases in the biosynthetic processing of C-propeptides from procollagens I-III. This processing step is thought necessary to the formation of collagen I-III monomers capable of forming fibrils. Thus, PCPE-1 is predicted to play an important role in scarring, as scar tissue is predominantly composed of fibrillar collagen. Corneal scarring is of great clinical importance, as it leads to loss of visual acuity and, in severe cases, blindness. Here, we investigate a possible role for PCPE-1 in corneal scarring. Although differences in corneal opacity associated with scarring following injury of Pcolce -/- and wild-type (WT) mice using full-thickness excision or alkali burn models of corneal injury were not grossly apparent, differences in procollagen I processing levels between Pcolce -/- and WT primary corneal keratocytes were consistent with a role for PCPE-1 in corneal collagen deposition. An unexpected finding was that neoangiogenesis, which follows alkali burn cornea injury, was strikingly increased in Pcolce -/- cornea, compared to WT. A series of aortic ring assays confirmed the anti-angiogenic effects of PCPE-1. Another unexpected finding was of abnormalities of epithelial basement membrane and of re-epithelialization following Pcolce -/- corneal injury. Thus, PCPE-1 appears to be of importance as an anti-angiogenic factor and in re-epithelialization following injury in cornea and perhaps in other tissues as well.
Collapse
Affiliation(s)
- Dawiyat Massoudi
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, 4503 WIMRII, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Colin J Germer
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, 4503 WIMRII, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Jeffrey M Glisch
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, 4503 WIMRII, 1111 Highland Ave., Madison, WI, 53705, USA
| | - Daniel S Greenspan
- Department of Cell and Regenerative Biology, University of Wisconsin School of Medicine and Public Health, 4503 WIMRII, 1111 Highland Ave., Madison, WI, 53705, USA.
| |
Collapse
|
31
|
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
|
32
|
Hassoun E, Safrin M, Ziv H, Pri-Chen S, Kessler E. Procollagen C-Proteinase Enhancer 1 (PCPE-1) as a Plasma Marker of Muscle and Liver Fibrosis in Mice. PLoS One 2016; 11:e0159606. [PMID: 27458976 PMCID: PMC4961444 DOI: 10.1371/journal.pone.0159606] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 07/05/2016] [Indexed: 01/06/2023] Open
Abstract
Current non-invasive diagnostic methods of fibrosis are limited in their ability to identify early and intermediate stages of fibrosis and assess the efficacy of therapy. New biomarkers of fibrosis are therefore constantly sought for, leading us to evaluate procollagen C-proteinase enhancer 1 (PCPE-1), a fibrosis-related extracellular matrix glycoprotein, as a plasma marker of fibrosis. A sandwich ELISA that permitted accurate measurements of PCPE-1 concentrations in mouse plasma was established. Tissue fibrosis was assessed using histochemical, immunofluorescence, and immunoblotting analyses for type I collagen and PCPE-1. The normal plasma concentration of PCPE-1 in 6 weeks to 4 months old mice was ~200 ng/ml (189.5 ± 11.3 to 206.8 ± 13.8 ng/ml). PCPE-1 plasma concentrations in four and 8.5 months old mdx mice displaying fibrotic diaphragms increased 27 and 40% respectively relatively to age-matched control mice, an increase comparable to that of the N-propeptide of procollagen type III (PIIINP), a known blood marker of fibrosis. PCPE-1 plasma levels in mice with CCl4-induced liver fibrosis increased 34 to 50% relatively to respective controls and reflected the severity of the disease, namely increased gradually during the progression of fibrosis and went down to basal levels during recovery, in parallel to changes in the liver content of collagen I and PCPE-1. The results favor PCPE-1 as a potential new clinically valuable fibrosis biomarker.
Collapse
Affiliation(s)
- Eyal Hassoun
- Maurice and Gabriela Goldschleger Eye Research Institute, Tel-Aviv University Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| | - Mary Safrin
- Maurice and Gabriela Goldschleger Eye Research Institute, Tel-Aviv University Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| | - Hana Ziv
- Maurice and Gabriela Goldschleger Eye Research Institute, Tel-Aviv University Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| | - Sarah Pri-Chen
- Maurice and Gabriela Goldschleger Eye Research Institute, Tel-Aviv University Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| | - Efrat Kessler
- Maurice and Gabriela Goldschleger Eye Research Institute, Tel-Aviv University Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 52621, Israel
| |
Collapse
|
33
|
BMP-1/tolloid-like proteinases synchronize matrix assembly with growth factor activation to promote morphogenesis and tissue remodeling. Matrix Biol 2015; 44-46:14-23. [DOI: 10.1016/j.matbio.2015.02.006] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 11/20/2022]
|
34
|
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: 0.7] [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
|
35
|
Luo L, Zhang C, Zhao J, Wei Q, Li X. Effects of rapamycin on reduction of peridural fibrosis: an experimental study. Med Sci Monit 2015; 21:482-8. [PMID: 25677111 PMCID: PMC4335565 DOI: 10.12659/msm.893165] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background Peridural fibrosis (PF) is a normal complication after lumbar surgery. It is a challenge for both surgeons and patients. Rapamycin (RPM), a novel antibiotic with anti-proliferative and immunosuppressive properties, has been shown to be effective in preventing uncontrolled scar proliferation diseases. The object of the present research was to investigate the effects of RPM on inhibiting PF in vitro and in vivo. Material/Methods In vitro, the fibroblasts collected and isolated from the rat tail skin were cultured with/without RPM and cell counting was performed. In vivo, the double-blinded study was conducted in 60 healthy Wistar rats divided randomly into 3 groups: 1) RPM treatment group; 2) Vehicle treatment group; 3) Control group. Rats underwent a L1-L2 level laminectomy with a satisfactory anesthetization. Four weeks post-operatively, the Rydell score, histological analysis, hydroxyproline content, vimentin expressional level, and inflammatory cytokines expressional levels were assessed. Results In vitro, RPM showed ability to prevent fibroblast proliferation. In vivo, the laminectomy was well tolerated by all rats, which were killed 4 weeks post-operatively. The Rydell score, histological evaluation, hydroxyproline content, vimentin expression level, and inflammatory activity showed the positive effect of RPM in preventing peridural adhesion, inhibiting fibrotic formation and collagen synthesis, and down-regulating inflammation. Conclusions In the present primary study, RPM showed good efficacy in preventing the proliferation of fibroblasts. RPM can prevent rat peridural adhesion through inhibiting collagen synthesis, fibroblasts proliferation, and inflammatory activity.
Collapse
Affiliation(s)
- Like Luo
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Chifei Zhang
- Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Jinmin Zhao
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Qingjun Wei
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| | - Xiaofeng Li
- Department of Orthopedic Trauma and Hand Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China (mainland)
| |
Collapse
|