1
|
Mekonnen TT, Ambekar YS, Zevallos-Delgado C, Nair A, Zvietcovich F, Zarkoob H, Singh M, Lim YW, Ferrer M, Aglyamov SR, Scarcelli G, Song MJ, Larin KV. Dual optical elastography detects TGF - β -induced alterations in the biomechanical properties of skin scaffolds. JOURNAL OF BIOMEDICAL OPTICS 2024; 29:095002. [PMID: 39295639 PMCID: PMC11409821 DOI: 10.1117/1.jbo.29.9.095002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 09/21/2024]
Abstract
Significance The skin's mechanical properties are tightly regulated. Various pathologies can affect skin stiffness, and understanding these changes is a focus in tissue engineering. Ex vivo skin scaffolds are a robust platform for evaluating the effects of various genetic and molecular interactions on the skin. Transforming growth factor-beta ( TGF - β ) is a critical signaling molecule in the skin that can regulate the amount of collagen and elastin in the skin and, consequently, its mechanical properties. Aim This study investigates the biomechanical properties of bio-engineered skin scaffolds, focusing on the influence of TGF - β , a signaling molecule with diverse cellular functions. Approach The TGF - β receptor I inhibitor, galunisertib, was employed to assess the mechanical changes resulting from dysregulation of TGF - β . Skin scaffold samples, grouped into three categories (control, TGF - β -treated, and TGF - β + galunisertib-treated), were prepared in two distinct culture media-one with aprotinin (AP) and another without. Two optical elastography techniques, namely wave-based optical coherence elastography (OCE) and Brillouin microscopy, were utilized to quantify the biomechanical properties of the tissues. Results Results showed significantly higher wave speed (with AP, p < 0.001 ; without AP, p < 0.001 ) and Brillouin frequency shift (with AP, p < 0.001 ; without AP, p = 0.01 ) in TGF - β -treated group compared with the control group. The difference in wave speed between the control and TGF - β + galunisertib with ( p = 0.10 ) and without AP ( p = 0.36 ) was not significant. Moreover, the TGF - β + galunisertib-treated group exhibited lower wave speed without and with AP and reduced Brillouin frequency shift than the TGF - β -treated group without AP, further strengthening the potential role of TGF - β in regulating the mechanical properties of the samples. Conclusions These findings offer valuable insights into TGF - β -induced biomechanical alterations in bio-engineered skin scaffolds, highlighting the potential of OCE and Brillouin microscopy in the development of targeted therapies in conditions involving abnormal tissue remodeling and fibrosis.
Collapse
Affiliation(s)
- Taye T. Mekonnen
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
- University of Sydney, Department of Mechanical Engineering, Sydney, New South Wales, Australia
| | - Yogeshwari S. Ambekar
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
- University of Maryland, Fischell Department of Bioengineering, College Park, Maryland, United States
| | | | - Achuth Nair
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Fernando Zvietcovich
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
- Pontificia Universidad Catolica del Peru, Department of Engineering, Lima, Peru
| | - Hoda Zarkoob
- National Institutes of Health, National Center for Advancing Translational Sciences, Rockville, Maryland, United States
| | - Manmohan Singh
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| | - Yi Wei Lim
- National Institutes of Health, National Center for Advancing Translational Sciences, Rockville, Maryland, United States
| | - Marc Ferrer
- National Institutes of Health, National Center for Advancing Translational Sciences, Rockville, Maryland, United States
| | - Salavat R. Aglyamov
- University of Houston, Department of Mechanical Engineering, Houston, Texas, United States
| | - Giuliano Scarcelli
- University of Maryland, Fischell Department of Bioengineering, College Park, Maryland, United States
| | - Min Jae Song
- National Institutes of Health, National Center for Advancing Translational Sciences, Rockville, Maryland, United States
| | - Kirill V. Larin
- University of Houston, Department of Biomedical Engineering, Houston, Texas, United States
| |
Collapse
|
2
|
Streutker EM, Devamoglu U, Vonk MC, Verdurmen WPR, Le Gac S. Fibrosis-on-Chip: A Guide to Recapitulate the Essential Features of Fibrotic Disease. Adv Healthc Mater 2024; 13:e2303991. [PMID: 38536053 DOI: 10.1002/adhm.202303991] [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: 11/14/2023] [Revised: 03/15/2024] [Indexed: 05/05/2024]
Abstract
Fibrosis, which is primarily marked by excessive extracellular matrix (ECM) deposition, is a pathophysiological process associated with many disorders, which ultimately leads to organ dysfunction and poor patient outcomes. Despite the high prevalence of fibrosis, currently there exist few therapeutic options, and importantly, there is a paucity of in vitro models to accurately study fibrosis. This review discusses the multifaceted nature of fibrosis from the viewpoint of developing organ-on-chip (OoC) disease models, focusing on five key features: the ECM component, inflammation, mechanical cues, hypoxia, and vascularization. The potential of OoC technology is explored for better modeling these features in the context of studying fibrotic diseases and the interplay between various key features is emphasized. This paper reviews how organ-specific fibrotic diseases are modeled in OoC platforms, which elements are included in these existing models, and the avenues for novel research directions are highlighted. Finally, this review concludes with a perspective on how to address the current gap with respect to the inclusion of multiple features to yield more sophisticated and relevant models of fibrotic diseases in an OoC format.
Collapse
Affiliation(s)
- Emma M Streutker
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Utku Devamoglu
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| | - Madelon C Vonk
- Department of Rheumatology, Radboud University Medical Center, Nijmegen, PO Box 9101, Nijmegen, 6500 HB, The Netherlands
| | - Wouter P R Verdurmen
- Department of Medical BioSciences, Radboud University Medical Center, Geert Grooteplein 28, Nijmegen, 6525 GA, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnoloygy and TechMed Centre, Organ-on-Chip Centre, University of Twente, Drienerlolaan 5, Enschede, 7522 NB, The Netherlands
| |
Collapse
|
3
|
Quizon MJ, Deppen JN, Barber GF, Kalelkar PP, Coronel MM, Levit RD, García AJ. VEGF-delivering PEG hydrogels promote vascularization in the porcine subcutaneous space. J Biomed Mater Res A 2024; 112:866-880. [PMID: 38189109 PMCID: PMC10984793 DOI: 10.1002/jbm.a.37666] [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: 11/16/2023] [Revised: 12/21/2023] [Accepted: 12/24/2023] [Indexed: 01/09/2024]
Abstract
For cell therapies, the subcutaneous space is an attractive transplant site due to its large surface area and accessibility for implantation, monitoring, biopsy, and retrieval. However, its poor vascularization has catalyzed research to induce blood vessel formation within the site to enhance cell revascularization and survival. Most studies focus on the subcutaneous space of rodents, which does not recapitulate important anatomical features and vascularization responses of humans. Herein, we evaluate biomaterial-driven vascularization in the porcine subcutaneous space. Additionally, we report the first use of cost-effective fluorescent microspheres to quantify perfusion in the porcine subcutaneous space. We investigate the vascularization-inducing efficacy of vascular endothelial growth factor (VEGF)-delivering synthetic hydrogels based on 4-arm poly(ethylene) glycol macromers with terminal maleimides (PEG-4MAL). We compare three groups: a non-degradable hydrogel with a VEGF-releasing PEG-4MAL gel coating (Core+VEGF gel); an uncoated, non-degradable hydrogel (Core-only); and naïve tissue. After 2 weeks, Core+VEGF gel has significantly higher tissue perfusion, blood vessel area, blood vessel density, and number of vessels compared to both Core-only and naïve tissue. Furthermore, healthy vital signs during surgery and post-procedure metrics demonstrate the safety of hydrogel delivery. We demonstrate that VEGF-delivering synthetic hydrogels induce robust vascularization and perfusion in the porcine subcutaneous space.
Collapse
Affiliation(s)
- Michelle J. Quizon
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Juline N. Deppen
- Division of Cardiology, Emory University School of Medicine, 1440 Clifton Rd, Atlanta, GA 30322, USA
| | - Graham F. Barber
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Pranav P. Kalelkar
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - María M. Coronel
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| | - Rebecca D. Levit
- Division of Cardiology, Emory University School of Medicine, 1440 Clifton Rd, Atlanta, GA 30322, USA
| | - Andrés J. García
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr. NW, Atlanta, GA 30332, USA
| |
Collapse
|
4
|
Visone R, Paoletti C, Cordiale A, Nicoletti L, Divieto C, Rasponi M, Chiono V, Occhetta P. In Vitro Mechanical Stimulation to Reproduce the Pathological Hallmarks of Human Cardiac Fibrosis on a Beating Chip and Predict The Efficacy of Drugs and Advanced Therapies. Adv Healthc Mater 2024; 13:e2301481. [PMID: 37941521 PMCID: PMC11468947 DOI: 10.1002/adhm.202301481] [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: 05/08/2023] [Revised: 10/16/2023] [Indexed: 11/10/2023]
Abstract
Cardiac fibrosis is one of the main causes of heart failure, significantly contributing to mortality. The discovery and development of effective therapies able to heal fibrotic pathological symptoms thus remain of paramount importance. Micro-physiological systems (MPS) are recently introduced as promising platforms able to accelerate this finding. Here a 3D in vitro model of human cardiac fibrosis, named uScar, is developed by imposing a cyclic mechanical stimulation to human atrial cardiac fibroblasts (AHCFs) cultured in a 3D beating heart-on-chip and exploited to screen drugs and advanced therapeutics. The sole provision of a cyclic 10% uniaxial strain at 1 Hz to the microtissues is sufficient to trigger fibrotic traits, inducing a consistent fibroblast-to-myofibroblast transition and an enhanced expression and production of extracellular matrix (ECM) proteins. Standard of care anti-fibrotic drugs (i.e., Pirfenidone and Tranilast) are confirmed to be efficient in preventing the onset of fibrotic traits in uScar. Conversely, the mechanical stimulation applied to the microtissues limit the ability of a miRNA therapy to directly reprogram fibroblasts into cardiomyocytes (CMs), despite its proved efficacy in 2D models. Such results demonstrate the importance of incorporating in vivo-like stimulations to generate more representative 3D in vitro models able to predict the efficacy of therapies in patients.
Collapse
Affiliation(s)
- Roberta Visone
- BiomimX SrlMilan20157Italy
- Department of ElectronicsInformatics and BioengineeringPolitecnico di MilanoMilan20133Italy
| | - Camilla Paoletti
- Department of Mechanical and Aerospace EngineeringPolitecnico di TorinoTurin10129Italy
- Centro 3R (Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research)Pisa56122Italy
| | - Alessandro Cordiale
- Department of ElectronicsInformatics and BioengineeringPolitecnico di MilanoMilan20133Italy
| | - Letizia Nicoletti
- Department of Mechanical and Aerospace EngineeringPolitecnico di TorinoTurin10129Italy
- Centro 3R (Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research)Pisa56122Italy
| | - Carla Divieto
- Istituto Nazionale di Ricerca MetrologicaDivision of Advanced Materials and Life SciencesTurin10135Italy
| | - Marco Rasponi
- Department of ElectronicsInformatics and BioengineeringPolitecnico di MilanoMilan20133Italy
- Centro 3R (Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research)Pisa56122Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace EngineeringPolitecnico di TorinoTurin10129Italy
- Centro 3R (Interuniversity Center for the Promotion of 3Rs Principles in Teaching and Research)Pisa56122Italy
| | - Paola Occhetta
- BiomimX SrlMilan20157Italy
- Department of ElectronicsInformatics and BioengineeringPolitecnico di MilanoMilan20133Italy
| |
Collapse
|
5
|
Xie R, Pal V, Yu Y, Lu X, Gao M, Liang S, Huang M, Peng W, Ozbolat IT. A comprehensive review on 3D tissue models: Biofabrication technologies and preclinical applications. Biomaterials 2024; 304:122408. [PMID: 38041911 PMCID: PMC10843844 DOI: 10.1016/j.biomaterials.2023.122408] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/09/2023] [Accepted: 11/22/2023] [Indexed: 12/04/2023]
Abstract
The limitations of traditional two-dimensional (2D) cultures and animal testing, when it comes to precisely foreseeing the toxicity and clinical effectiveness of potential drug candidates, have resulted in a notable increase in the rate of failure during the process of drug discovery and development. Three-dimensional (3D) in-vitro models have arisen as substitute platforms with the capacity to accurately depict in-vivo conditions and increasing the predictivity of clinical effects and toxicity of drug candidates. It has been found that 3D models can accurately represent complex tissue structure of human body and can be used for a wide range of disease modeling purposes. Recently, substantial progress in biomedicine, materials and engineering have been made to fabricate various 3D in-vitro models, which have been exhibited better disease progression predictivity and drug effects than convention models, suggesting a promising direction in pharmaceutics. This comprehensive review highlights the recent developments in 3D in-vitro tissue models for preclinical applications including drug screening and disease modeling targeting multiple organs and tissues, like liver, bone, gastrointestinal tract, kidney, heart, brain, and cartilage. We discuss current strategies for fabricating 3D models for specific organs with their strengths and pitfalls. We expand future considerations for establishing a physiologically-relevant microenvironment for growing 3D models and also provide readers with a perspective on intellectual property, industry, and regulatory landscape.
Collapse
Affiliation(s)
- Renjian Xie
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Vaibhav Pal
- Department of Chemistry, Pennsylvania State University, University Park, PA, USA; The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA
| | - Yanrong Yu
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Xiaolu Lu
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Mengwei Gao
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Shijie Liang
- School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China
| | - Miao Huang
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China
| | - Weijie Peng
- Key Laboratory of Biomaterials and Biofabrication for Tissue Engineering in Jiangxi Province, Gannan Medical University, Ganzhou, JX, 341000, China; Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, JX, China; School of Pharmaceutics, Nanchang University, Nanchang, JX, 330006, China.
| | - Ibrahim T Ozbolat
- The Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA, USA; Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA; Department of Biomedical Engineering, Pennsylvania State University, University Park, PA, USA; Materials Research Institute, Pennsylvania State University, University Park, PA, USA; Department of Neurosurgery, Pennsylvania State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Penn State University, Hershey, PA, 17033, USA; Department of Medical Oncology, Cukurova University, Adana, 01130, Turkey; Biotechnology Research and Application Center, Cukurova University, Adana, 01130, Turkey.
| |
Collapse
|
6
|
Padmanabhan J, Chen K, Sivaraj D, Henn D, Kuehlmann BA, Kussie HC, Zhao ET, Kahn A, Bonham CA, Dohi T, Beck TC, Trotsyuk AA, Stern-Buchbinder ZA, Than PA, Hosseini HS, Barrera JA, Magbual NJ, Leeolou MC, Fischer KS, Tigchelaar SS, Lin JQ, Perrault DP, Borrelli MR, Kwon SH, Maan ZN, Dunn JCY, Nazerali R, Januszyk M, Prantl L, Gurtner GC. Allometrically scaling tissue forces drive pathological foreign-body responses to implants via Rac2-activated myeloid cells. Nat Biomed Eng 2023; 7:1419-1436. [PMID: 37749310 PMCID: PMC10651488 DOI: 10.1038/s41551-023-01091-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 08/02/2023] [Indexed: 09/27/2023]
Abstract
Small animals do not replicate the severity of the human foreign-body response (FBR) to implants. Here we show that the FBR can be driven by forces generated at the implant surface that, owing to allometric scaling, increase exponentially with body size. We found that the human FBR is mediated by immune-cell-specific RAC2 mechanotransduction signalling, independently of the chemistry and mechanical properties of the implant, and that a pathological FBR that is human-like at the molecular, cellular and tissue levels can be induced in mice via the application of human-tissue-scale forces through a vibrating silicone implant. FBRs to such elevated extrinsic forces in the mice were also mediated by the activation of Rac2 signalling in a subpopulation of mechanoresponsive myeloid cells, which could be substantially reduced via the pharmacological or genetic inhibition of Rac2. Our findings provide an explanation for the stark differences in FBRs observed in small animals and humans, and have implications for the design and safety of implantable devices.
Collapse
Affiliation(s)
- Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| | - Dharshan Sivaraj
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| | - Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Britta A Kuehlmann
- Department of Plastic and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Hudson C Kussie
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Eric T Zhao
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Anum Kahn
- Cell Sciences Imaging Facility (CSIF), Beckman Center, Stanford University, Stanford, CA, USA
| | - Clark A Bonham
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Teruyuki Dohi
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Thomas C Beck
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Artem A Trotsyuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA
| | - Zachary A Stern-Buchbinder
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter A Than
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Hadi S Hosseini
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Janos A Barrera
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Noah J Magbual
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Melissa C Leeolou
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Katharina S Fischer
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Seth S Tigchelaar
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - John Q Lin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - David P Perrault
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Mimi R Borrelli
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Sun Hyung Kwon
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Zeshaan N Maan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - James C Y Dunn
- Division of Pediatric Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Rahim Nazerali
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Lukas Prantl
- Department of Plastic and Reconstructive Surgery, University Hospital Regensburg, Regensburg, Germany
| | - Geoffrey C Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, USA.
| |
Collapse
|
7
|
Sapudom J, Karaman S, Quartey BC, Mohamed WKE, Mahtani N, Garcia-Sabaté A, Teo J. Collagen Fibril Orientation Instructs Fibroblast Differentiation Via Cell Contractility. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301353. [PMID: 37249413 PMCID: PMC10401101 DOI: 10.1002/advs.202301353] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 05/03/2023] [Indexed: 05/31/2023]
Abstract
Collagen alignment is one of the key microarchitectural signatures of many pathological conditions, including scarring and fibrosis. Investigating how collagen alignment modulates cellular functions will pave the way for understanding tissue scarring and regeneration and new therapeutic strategies. However, current approaches for the fabrication of three-dimensional (3D) aligned collagen matrices are low-throughput and require special devices. To overcome these limitations, a simple approach to reconstitute homogeneous 3D collagen matrices with adjustable degree of fibril alignment using 3D printed inclined surfaces is developed. By characterizing the mechanical properties of reconstituted matrices, it is found that the elastic modulus of collagen matrices is enhanced with an increase in the alignment degree. The reconstituted matrices are used to study fibroblast behavior to reveal the progression of scar formation where a gradual enhancement of collagen alignment can be observed. It is found that matrices with aligned fibrils trigger fibroblast differentiation into myofibroblasts via cell contractility, while collagen stiffening through a crosslinker does not. The results suggest the impact of collagen fibril organization on the regulation of fibroblast differentiation. Overall, this approach to reconstitute 3D collagen matrices with fibril alignment opens opportunities for biomimetic pathological-relevant tissue in vitro, which can be applied for other biomedical research.
Collapse
Affiliation(s)
- Jiranuwat Sapudom
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Shaza Karaman
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Brian Chesney Quartey
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Walaa Kamal Eldin Mohamed
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Nick Mahtani
- School of Engineering, Ecole Polytechnique Federale de Lausanne, Lausanne, 1015, Switzerland
| | - Anna Garcia-Sabaté
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
| | - Jeremy Teo
- Laboratory for Immuno Bioengineering Research and Applications, Division of Engineering, New York University Abu Dhabi, Abu Dhabi, 129188, UAE
- Department of Mechanical and Biomedical Engineering, Tandon School of Engineering, New York University, New York, 11201, USA
| |
Collapse
|
8
|
Luu RJ, Hoefler BC, Gard AL, Ritenour CR, Rogers MT, Kim ES, Coppeta JR, Cain BP, Isenberg BC, Azizgolshani H, Fajardo-Ramirez OR, García-Cardeña G, Lech MP, Tomlinson L, Charest JL, Williams C. Fibroblast activation in response to TGFβ1 is modulated by co-culture with endothelial cells in a vascular organ-on-chip platform. Front Mol Biosci 2023; 10:1160851. [PMID: 37577751 PMCID: PMC10421749 DOI: 10.3389/fmolb.2023.1160851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 06/06/2023] [Indexed: 08/15/2023] Open
Abstract
Background: Tissue fibrosis is a major healthcare burden that affects various organs in the body for which no effective treatments exist. An underlying, emerging theme across organs and tissue types at early stages of fibrosis is the activation of pericytes and/or fibroblasts in the perivascular space. In hepatic tissue, it is well known that liver sinusoidal endothelial cells (EC) help maintain the quiescence of stellate cells, but whether this phenomenon holds true for other endothelial and perivascular cell types is not well studied. Methods: The goal of this work was to develop an organ-on-chip microvascular model to study the effect of EC co-culture on the activation of perivascular cells perturbed by the pro-fibrotic factor TGFβ1. A high-throughput microfluidic platform, PREDICT96, that was capable of imparting physiologically relevant fluid shear stress on the cultured endothelium was utilized. Results: We first studied the activation response of several perivascular cell types and selected a cell source, human dermal fibroblasts, that exhibited medium-level activation in response to TGFβ1. We also demonstrated that the PREDICT96 high flow pump triggered changes in select shear-responsive factors in human EC. We then found that the activation response of fibroblasts was significantly blunted in co-culture with EC compared to fibroblast mono-cultures. Subsequent studies with conditioned media demonstrated that EC-secreted factors play at least a partial role in suppressing the activation response. A Luminex panel and single cell RNA-sequencing study provided additional insight into potential EC-derived factors that could influence fibroblast activation. Conclusion: Overall, our findings showed that EC can reduce myofibroblast activation of perivascular cells in response to TGFβ1. Further exploration of EC-derived factors as potential therapeutic targets in fibrosis is warranted.
Collapse
Affiliation(s)
- Rebeccah J. Luu
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - B. Christopher Hoefler
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ashley L. Gard
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | | | - Miles T. Rogers
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Ernest S. Kim
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Jonathan R. Coppeta
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brian P. Cain
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Brett C. Isenberg
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Hesham Azizgolshani
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Oscar R. Fajardo-Ramirez
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Guillermo García-Cardeña
- Laboratory for Systems Mechanobiology, Center for Excellence in Vascular Biology, Department of Pathology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | | | | | - Joseph L. Charest
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| | - Corin Williams
- Bioengineering Division, The Charles Stark Draper Laboratory Inc., Cambridge, MA, United States
| |
Collapse
|
9
|
Keshavan S, Bannuscher A, Drasler B, Barosova H, Petri-Fink A, Rothen-Rutishauser B. Comparing species-different responses in pulmonary fibrosis research: Current understanding of in vitro lung cell models and nanomaterials. Eur J Pharm Sci 2023; 183:106387. [PMID: 36652970 DOI: 10.1016/j.ejps.2023.106387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 12/16/2022] [Accepted: 01/14/2023] [Indexed: 01/16/2023]
Abstract
Pulmonary fibrosis (PF) is a chronic, irreversible lung disease that is typically fatal and characterized by an abnormal fibrotic response. As a result, vast areas of the lungs are gradually affected, and gas exchange is impaired, making it one of the world's leading causes of death. This can be attributed to a lack of understanding of the onset and progression of the disease, as well as a poor understanding of the mechanism of adverse responses to various factors, such as exposure to allergens, nanomaterials, environmental pollutants, etc. So far, the most frequently used preclinical evaluation paradigm for PF is still animal testing. Nonetheless, there is an urgent need to understand the factors that induce PF and find novel therapeutic targets for PF in humans. In this regard, robust and realistic in vitro fibrosis models are required to understand the mechanism of adverse responses. Over the years, several in vitro and ex vivo models have been developed with the goal of mimicking the biological barriers of the lung as closely as possible. This review summarizes recent progress towards the development of experimental models suitable for predicting fibrotic responses, with an emphasis on cell culture methods, nanomaterials, and a comparison of results from studies using cells from various species.
Collapse
Affiliation(s)
- Sandeep Keshavan
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Anne Bannuscher
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland
| | - Hana Barosova
- Institute of Experimental Medicine of the Czech Academy of Sciences, Videnska 1083, Prague 14220, Czech Republic
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, Fribourg CH-1700, Switzerland; Chemistry Department, University of Fribourg, Chemin du Musée 9, Fribourg 1700, Switzerland
| | | |
Collapse
|
10
|
Amici JM, Taieb C, Le Floc'h C, Demessant A, Seité S, Cogrel O. The impact of visible scars on well-being and quality of life: An international epidemiological survey in adults. J Eur Acad Dermatol Venereol 2023; 37 Suppl 3:3-6. [PMID: 36635614 DOI: 10.1111/jdv.18856] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 11/29/2022] [Indexed: 01/14/2023]
Abstract
INTRODUCTION Scars are visible marks from various causes, including surgery, skin injury, burning or dermatological disease, and may impact the quality of life. OBJECTIVE To assess the impact of scars on quality of life (QoL). MATERIAL AND METHODS Data about sociodemography, presence, origin, and symptoms of scars were collected using an Internet survey between April and May 2020. Overall, 11,100 individuals answered the survey. In total, 48.5% of the responders had at least one scar of less than 1 year of age. Scars were mainly reported on the abdomen and face. Globally, 28.9% of subjects with recent scars reported pain, 23.7% reported burning, 35.0% reported itching, and 44.1% reported redness. Subjects were most frequently bothered by the visibility of their scars and the presence of marks. Incidences were significantly higher than for those with older scars. The average DLQI score was 7.44; it decreased to 2.90 after 1 year. Subjects with scars aged less than 3 months had their QoL more frequently impacted (33.9%) than those with scars aged 12 months or more (10.2%). In subjects reporting skin discomfort, clinical symptoms significantly impacted body movement, choice of clothes, leisure activities, and sexual life more than in those reporting no skin discomfort. Moreover, subjects felt significantly more impacted in their daily lives due to their skin discomfort. When feeling bothered by the visibility of their scars, significantly more subjects were also impacted in their body movement, choice of clothes, leisure activities, and sexual life than those subjects who did not feel bothered. Moreover, significantly more subjects felt embarrassed. CONCLUSION Scars significantly impact the subjects' quality of life. This impact is even more important when caused by recent and visible scars, with a lower DLQI score in subjects with more aged than in those with recent scars.
Collapse
Affiliation(s)
- Jean Michel Amici
- Dermatology Department, CHU Bordeaux, Hôpital Saint-André, Bordeaux, France
| | - Charles Taieb
- European Market Maintenance Assessment, Fontenay-sous-Bois, France
| | | | | | - Sophie Seité
- La Roche-Posay Dermatological Laboratories, Levallois-Perret, France
| | - Olivier Cogrel
- Dermatology Department, CHU Bordeaux, Hôpital Saint-André, Bordeaux, France.,French Society of Dermatological Surgery, Paris, France
| |
Collapse
|
11
|
Neves LMG, Wilgus TA, Bayat A. In Vitro, Ex Vivo, and In Vivo Approaches for Investigation of Skin Scarring: Human and Animal Models. Adv Wound Care (New Rochelle) 2023; 12:97-116. [PMID: 34915768 DOI: 10.1089/wound.2021.0139] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Significance: The cutaneous repair process naturally results in different types of scarring that are classified as normal or pathological. Affected individuals are often affected from an esthetic, physical (functional), and psychosocial perspective. The distinct nature of scarring in humans, particularly the formation of pathological scars, makes the study of skin scarring a challenge for researchers in this area. Several established experimental models exist for studying scar formation. However, the increasing development and validation of newly emerging models have made it possible to carry out studies focused on different variables that influence this unique process. Recent Advances: Experimental models such as in vitro, ex vivo, and in vivo models have obtained different degrees of success in the reproduction of the scar formation in its native milieu and true environment. These models also differ in their ability to elucidate the molecular, cellular, and structural mechanisms involved in scarring, as well as for testing new agents and approaches for therapies. The models reviewed here, including cells derived from human skin and in vivo animal models, have contributed to the advancement of skin scarring research. Critical Issues and Future Directions: The absence of experimental models that faithfully reproduce the typical characteristics of the different types of human skin scars makes the improvement of validated models and the establishment of new ones a critical unmet need. The fields of wound healing research combined with tissue engineering have offered newer alternatives for experimental studies with the potential to provide clinically useful knowledge about scar formation.
Collapse
Affiliation(s)
- Lia M G Neves
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom
| | - Traci A Wilgus
- Department of Pathology, Ohio State University, Columbus, Ohio, USA
| | - Ardeshir Bayat
- Plastic & Reconstructive Surgery Research, Centre for Dermatology Research, Wound Healing Theme, NIHR Manchester Biomedical Research Centre, University of Manchester, Manchester, England, United Kingdom.,Medical Research Council (MRC) Wound Healing Unit, Hair and Skin Research Laboratory, Division of Dermatology, Department of Medicine, Groote Schuur Hospital, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
12
|
Amici JM, Taieb C, LeFloc'h C, Demessant A, Seité S, Cogrel O. Prevalence of scars: an international epidemiological survey in adults. J Eur Acad Dermatol Venereol 2022; 36:e799-e800. [PMID: 35633084 PMCID: PMC9544440 DOI: 10.1111/jdv.18277] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/13/2022] [Indexed: 12/03/2022]
Affiliation(s)
- Jean Michel Amici
- Dermatology Department, CHU Bordeaux, Hôpital Saint-André, Bordeaux, France
| | - Charles Taieb
- European Market Maintenance Assessment, Fontenay-sous-Bois, France
| | | | | | | | - Olivier Cogrel
- Dermatology Department, CHU Bordeaux, Hôpital Saint-André, Bordeaux, France.,President of the French Society of Dermatological Surgery
| |
Collapse
|
13
|
Sivaraj D, Padmanabhan J, Chen K, Henn D, Noishiki C, Trotsyuk AA, Kussie HC, Leeolou MC, Magbual NJ, Andrikopoulos S, Perrault DP, Barrera JA, Januszyk M, Gurtner GC. IQGAP1-mediated mechanical signaling promotes the foreign body response to biomedical implants. FASEB J 2022; 36:e22007. [PMID: 35051300 DOI: 10.1096/fj.202101354] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022]
Abstract
The aim of this study was to further elucidate the molecular mechanisms that mediate pathologic foreign body response (FBR) to biomedical implants. The longevity of biomedical implants is limited by the FBR, which leads to implant failure and patient morbidity. Since the specific molecular mechanisms underlying fibrotic responses to biomedical implants have yet to be fully described, there are currently no targeted approaches to reduce pathologic FBR. We utilized proteomics analysis of human FBR samples to identify potential molecular targets for therapeutic inhibition of FBR. We then employed a murine model of FBR to further evaluate the role of this potential target. We performed histological and immunohistochemical analysis on the murine FBR capsule tissue, as well as single-cell RNA sequencing (scRNA-seq) on cells isolated from the capsules. We identified IQ motif containing GTPase activating protein 1 (IQGAP1) as the most promising of several targets, serving as a central molecular mediator in human and murine FBR compared to control subcutaneous tissue. IQGAP1-deficient mice displayed a significantly reduced FBR compared to wild-type mice as evidenced by lower levels of collagen deposition and maturity. Our scRNA-seq analysis revealed that decreasing IQGAP1 resulted in diminished transcription of mechanotransduction, inflammation, and fibrosis-related genes, which was confirmed on the protein level with immunofluorescent staining. The deficiency of IQGAP1 significantly attenuates FBR by deactivating downstream mechanotransduction signaling, inflammation, and fibrotic pathways. IQGAP1 may be a promising target for rational therapeutic design to mitigate pathologic FBR around biomedical implants.
Collapse
Affiliation(s)
- Dharshan Sivaraj
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Jagannath Padmanabhan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Kellen Chen
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Dominic Henn
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Chikage Noishiki
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Artem A Trotsyuk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Hudson C Kussie
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Melissa C Leeolou
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Noah J Magbual
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Sophia Andrikopoulos
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - David P Perrault
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Janos A Barrera
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Michael Januszyk
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| | - Geoffrey C Gurtner
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
14
|
Henn D, Chen K, Fehlmann T, Trotsyuk AA, Sivaraj D, Maan ZN, Bonham CA, Barrera JA, Mays CJ, Greco AH, Moortgat Illouz SE, Lin JQ, Steele SR, Foster DS, Padmanabhan J, Momeni A, Nguyen D, Wan DC, Kneser U, Januszyk M, Keller A, Longaker MT, Gurtner GC. Xenogeneic skin transplantation promotes angiogenesis and tissue regeneration through activated Trem2 + macrophages. SCIENCE ADVANCES 2021; 7:eabi4528. [PMID: 34851663 PMCID: PMC8635426 DOI: 10.1126/sciadv.abi4528] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 10/09/2021] [Indexed: 05/07/2023]
Abstract
Skin allo- and xenotransplantation are the standard treatment for major burns when donor sites for autografts are not available. The relationship between the immune response to foreign grafts and their impact on wound healing has not been fully elucidated. Here, we investigated changes in collagen architecture after xenogeneic implantation of human biologic scaffolds. We show that collagen deposition in response to the implantation of human split-thickness skin grafts (hSTSGs) containing live cells recapitulates normal skin architecture, whereas human acellular dermal matrix (ADM) grafts led to a fibrotic collagen deposition. We show that macrophage differentiation in response to hSTSG implantation is driven toward regenerative Trem2+ subpopulations and found that hydrogel delivery of these cells significantly accelerated wound closure. Our study identifies the preclinical therapeutic potential of Trem2+ macrophages to mitigate fibrosis and promote wound healing, providing a novel effective strategy to develop advanced cell therapies for complex wounds.
Collapse
Affiliation(s)
- Dominic Henn
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Kellen Chen
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Tobias Fehlmann
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
| | - Artem A. Trotsyuk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Dharshan Sivaraj
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Zeshaan N. Maan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Clark A. Bonham
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Janos A. Barrera
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Chyna J. Mays
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Autumn H. Greco
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Sylvia E. Moortgat Illouz
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - John Qian Lin
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Sydney R. Steele
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Deshka S. Foster
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Jagannath Padmanabhan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Arash Momeni
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Dung Nguyen
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Derrick C. Wan
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Ulrich Kneser
- Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Michael Januszyk
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Andreas Keller
- Chair for Clinical Bioinformatics, Saarland University, Saarbrücken, Germany
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA
| | - Michael T. Longaker
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | - Geoffrey C. Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| |
Collapse
|
15
|
Coentro JQ, May U, Prince S, Zwaagstra J, Ritvos O, Järvinen TAH, Zeugolis DI. Adapting the Scar-in-a-Jar to Skin Fibrosis and Screening Traditional and Contemporary Anti-Fibrotic Therapies. Front Bioeng Biotechnol 2021; 9:756399. [PMID: 34765594 PMCID: PMC8576412 DOI: 10.3389/fbioe.2021.756399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
Skin fibrosis still constitutes an unmet clinical need. Although pharmacological strategies are at the forefront of scientific and technological research and innovation, their clinical translation is hindered by the poor predictive capacity of the currently available in vitro fibrosis models. Indeed, customarily utilised in vitro scarring models are conducted in a low extracellular matrix milieu, which constitutes an oxymoron for the in-hand pathophysiology. Herein, we coupled macromolecular crowding (enhances and accelerates extracellular matrix deposition) with transforming growth factor β1 (TGFβ1; induces trans-differentiation of fibroblasts to myofibroblasts) in human dermal fibroblast cultures to develop a skin fibrosis in vitro model and to screen a range of anti-fibrotic families (corticosteroids, inhibitors of histone deacetylases, inhibitors of collagen crosslinking, inhibitors of TGFβ1 and pleiotropic inhibitors of fibrotic activation). Data obtained demonstrated that macromolecular crowding combined with TGFβ1 significantly enhanced collagen deposition and myofibroblast transformation. Among the anti-fibrotic compounds assessed, trichostatin A (inhibitors of histone deacetylases); serelaxin and pirfenidone (pleiotropic inhibitors of fibrotic activation); and soluble TGFβ receptor trap (inhibitor of TGFβ signalling) resulted in the highest decrease of collagen type I deposition (even higher than triamcinolone acetonide, the gold standard in clinical practice). This study further advocates the potential of macromolecular crowding in the development of in vitro pathophysiology models.
Collapse
Affiliation(s)
- João Q Coentro
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Ulrike May
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Stuart Prince
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - John Zwaagstra
- Human Health Therapeutics Research Centre, National Research Council Canada, Montreal, QC, Canada
| | | | - Tero A H Järvinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland.,Tampere University Hospital, Tampere, Finland
| | - Dimitrios I Zeugolis
- Regenerative, Modular and Developmental Engineering Laboratory (REMODEL) and Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), National University of Ireland Galway (NUI Galway), Galway, Ireland.,Regenerative, Modular and Developmental Engineering Laboratory (REMODEL), Charles Institute of Dermatology, Conway Institute of Biomolecular and Biomedical Research and School of Mechanical and Materials Engineering, University College Dublin (UCD), Dublin, Ireland
| |
Collapse
|
16
|
Tu L, Lin Z, Huang Q, Liu D. USP15 Enhances the Proliferation, Migration, and Collagen Deposition of Hypertrophic Scar-Derived Fibroblasts by Deubiquitinating TGF-βR1 In Vitro. Plast Reconstr Surg 2021; 148:1040-1051. [PMID: 34546211 PMCID: PMC8542080 DOI: 10.1097/prs.0000000000008488] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 04/13/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Hypertrophic scar is a fibroproliferative disorder caused by skin injury. The incidence of hypertrophic scar following trauma or burns is 40 to 70 percent or 70 percent, respectively. It has been shown that transforming growth factor (TGF) β1/Smad signaling plays a crucial role in hypertrophic scar, and that USP15 can regulate the activity of TGFβ1/Smad signaling to affect the progression of the disease. However, the underlying mechanism of USP15 in hypertrophic scar remains unclear. The authors hypothesized that USP15 was up-regulated and enhanced the proliferation, migration, invasion, and collagen deposition of hypertrophic scar-derived fibroblasts by deubiquitinating TGF-β receptor I (TβRI) in vitro. METHODS Fibroblasts were isolated from human hypertrophic scars in vitro. The knockdown and overexpression of USP15 in hypertrophic scar-derived fibroblasts were performed using lentivirus infection. The effect of USP15 on hypertrophic scar-derived fibroblast proliferation, migration, and invasion, and the expression of TβRI, Smad2, Smad3, α-SMA, COL1, and COL3, were detected by Cell Counting Kit-8, scratch, invasion, quantitative real-time polymerase chain reaction, and Western blot assays. The interaction between USP15 and TβRI was detected by co-immunoprecipitation and ubiquitination assays. RESULTS The authors demonstrated that USP15 knockdown significantly inhibited the proliferation, migration, and invasion of hypertrophic scar-derived fibroblasts in vitro and down-regulated the expression of TβRI, Smad2, Smad3, α-SMA, COL1, and COL3; in addition, USP15 overexpression showed the opposite trends (p < 0.05). Co-immunoprecipitation and ubiquitination assays revealed that USP15 interacted with TβRI and deubiquitinated TβRI. CONCLUSION USP15 enhances the proliferation, migration, invasion, and collagen deposition of hypertrophic scar-derived fibroblasts by deubiquitinating TβRI in vitro.
Collapse
Affiliation(s)
- Longxiang Tu
- From the Institute of Burn and Departments of Orthopedic Surgery and Nursing, The First Affiliated Hospital of Nanchang University
| | - Zunwen Lin
- From the Institute of Burn and Departments of Orthopedic Surgery and Nursing, The First Affiliated Hospital of Nanchang University
| | - Qin Huang
- From the Institute of Burn and Departments of Orthopedic Surgery and Nursing, The First Affiliated Hospital of Nanchang University
| | - Dewu Liu
- From the Institute of Burn and Departments of Orthopedic Surgery and Nursing, The First Affiliated Hospital of Nanchang University
| |
Collapse
|
17
|
Berry-Kilgour C, Cabral J, Wise L. Advancements in the Delivery of Growth Factors and Cytokines for the Treatment of Cutaneous Wound Indications. Adv Wound Care (New Rochelle) 2021; 10:596-622. [PMID: 33086946 PMCID: PMC8392095 DOI: 10.1089/wound.2020.1183] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 10/19/2020] [Indexed: 01/05/2023] Open
Abstract
Significance: Wound healing involves the phasic production of growth factors (GFs) and cytokines to progress an acute wound to a resolved scar. Dysregulation of these proteins contributes to both wound chronicity and excessive scarring. Direct supplementation of GFs and cytokines for treatment of healing and scarring complications has, however, been disappointing. Failings likely relate to an inability to deliver recombinant proteins at physiologically relevant levels to an environment conducive to healing. Recent Advances: Inspired by the extracellular matrix, natural biomaterials have been developed that resemble human skin, and are capable of delivering bioactives. Hybrid biomaterials made using multiple polymers, fabrication methods, and proteins are proving efficacious in animal models of acute and impaired wound healing. Critical Issues: For clinical translation, these delivery systems must be tailored for specific wound indications and the correct phase of healing. GFs and cytokines must be delivered in a controlled manner that will target specific healing or scarring impairments. Preclinical assessment in clinically relevant animal models of impaired or excessive healing is critical. Future Directions: Clinical success will likely depend on the GF or cytokine selected, their compatibility with the chosen biomaterial(s), degradation rate of the fabricated system, and the degree of control over release kinetics. Further testing is essential to assess which wound indications are most suited to specific delivery systems and to prove whether they provide superior efficacy over direct protein therapies.
Collapse
Affiliation(s)
- Caitlin Berry-Kilgour
- Department of Pharmacology and Toxicology, School of Biomedical Sciences; Dunedin, New Zealand
| | - Jaydee Cabral
- Department of Chemistry, University of Otago, Dunedin, New Zealand
- Department of Food Sciences, University of Otago, Dunedin, New Zealand
| | - Lyn Wise
- Department of Pharmacology and Toxicology, School of Biomedical Sciences; Dunedin, New Zealand
| |
Collapse
|
18
|
Sawant M, Hinz B, Schönborn K, Zeinert I, Eckes B, Krieg T, Schuster R. A story of fibers and stress: Matrix-embedded signals for fibroblast activation in the skin. Wound Repair Regen 2021; 29:515-530. [PMID: 34081361 DOI: 10.1111/wrr.12950] [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: 03/25/2021] [Revised: 05/13/2021] [Accepted: 05/28/2021] [Indexed: 12/12/2022]
Abstract
Our skin is continuously exposed to mechanical challenge, including shear, stretch, and compression. The extracellular matrix of the dermis is perfectly suited to resist these challenges and maintain integrity of normal skin even upon large strains. Fibroblasts are the key cells that interpret mechanical and chemical cues in their environment to turnover matrix and maintain homeostasis in the skin of healthy adults. Upon tissue injury, fibroblasts and an exclusive selection of other cells become activated into myofibroblasts with the task to restore skin integrity by forming structurally imperfect but mechanically stable scar tissue. Failure of myofibroblasts to terminate their actions after successful repair or upon chronic inflammation results in dysregulated myofibroblast activities which can lead to hypertrophic scarring and/or skin fibrosis. After providing an overview on the major fibrillar matrix components in normal skin, we will interrogate the various origins of fibroblasts and myofibroblasts in the skin. We then examine the role of the matrix as signaling hub and how fibroblasts respond to mechanical matrix cues to restore order in the confusing environment of a healing wound.
Collapse
Affiliation(s)
- Mugdha Sawant
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Boris Hinz
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Canada
| | - Katrin Schönborn
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Isabel Zeinert
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Beate Eckes
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany
| | - Thomas Krieg
- Translational Matrix Biology, University of Cologne, Medical Faculty, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.,Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany
| | - Ronen Schuster
- Laboratory of Tissue Repair and Regeneration, Faculty of Dentistry, University of Toronto, Toronto, Canada.,PhenomicAI, MaRS Centre, 661 University Avenue, Toronto, Canada
| |
Collapse
|
19
|
Epigenetic Modulation of Radiation-Induced Diacylglycerol Kinase Alpha Expression Prevents Pro-Fibrotic Fibroblast Response. Cancers (Basel) 2021; 13:cancers13102455. [PMID: 34070078 PMCID: PMC8158145 DOI: 10.3390/cancers13102455] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/04/2021] [Accepted: 05/12/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary To reduce long-term fibrosis risk after radiotherapy, we demonstrated with different experimental approaches that modulation of the epigenetic pattern at the DGKA enhancer can attenuate pro-fibrotic reactions in human fibroblasts. We used (epi)genomic editing of the DGKA enhancer and administration of various epigenetic drugs and were able to modulate radiation-induced expression of DGKA and pro-fibrotic collagens. Based on our results, clinical application of bromodomain inhibitors will open promising ways to epigenetically modulate DGKA expression and might provide novel therapeutic options to prevent or even reverse radiotherapy-induced fibrotic reactions. Abstract Radiotherapy, a common component in cancer treatment, can induce adverse effects including fibrosis in co-irradiated tissues. We previously showed that differential DNA methylation at an enhancer of diacylglycerol kinase alpha (DGKA) in normal dermal fibroblasts is associated with radiation-induced fibrosis. After irradiation, the transcription factor EGR1 is induced and binds to the hypomethylated enhancer, leading to increased DGKA and pro-fibrotic marker expression. We now modulated this DGKA induction by targeted epigenomic and genomic editing of the DGKA enhancer and administering epigenetic drugs. Targeted DNA demethylation of the DGKA enhancer in HEK293T cells resulted in enrichment of enhancer-related histone activation marks and radiation-induced DGKA expression. Mutations of the EGR1-binding motifs decreased radiation-induced DGKA expression in BJ fibroblasts and caused dysregulation of multiple fibrosis-related pathways. EZH2 inhibitors (GSK126, EPZ6438) did not change radiation-induced DGKA increase. Bromodomain inhibitors (CBP30, JQ1) suppressed radiation-induced DGKA and pro-fibrotic marker expression. Similar drug effects were observed in donor-derived fibroblasts with low DNA methylation. Overall, epigenomic manipulation of DGKA expression may offer novel options for a personalized treatment to prevent or attenuate radiotherapy-induced fibrosis.
Collapse
|
20
|
Zhang Y, Zeng W, Xia Y. TWEAK/Fn14 axis is an important player in fibrosis. J Cell Physiol 2020; 236:3304-3316. [PMID: 33000480 DOI: 10.1002/jcp.30089] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/15/2022]
Abstract
Fibrosis is a common pathological condition associated with abnormal repair after tissue injury. However, the etiology and molecular mechanisms of fibrosis are still not well-understood. Tumor necrosis factor (TNF)-like weak inducer of apoptosis (TWEAK) belongs to the TNF superfamily and acts by binding to its receptor, fibroblast growth factor-inducible 14 (Fn14), thereby activating a variety of intracellular signal transduction pathways in various types of cells. Besides promoting the expression of growth factors, activation of TWEAK/Fn14 signaling after tissue injury can promote the expression of pro-inflammatory cytokines, which trigger the immune response, thereby exacerbating the injury. Severe or repetitive injury leads to a dysregulated tissue repair process, in which the TWEAK/Fn14 axis promotes the activation and proliferation of myofibroblasts, induces the secretion of the extracellular matrix, and regulates profibrotic mediators to further perpetuate and sustain the fibrotic process. In this review, we summarize the available experimental evidence on the underlying molecular mechanisms by which the TWEAK/Fn14 pathway mediates the development and progression of fibrosis. In addition, we discuss the therapeutic potential of the TWEAK/Fn14 pathway in fibrosis-associated diseases based on evidence derived from multiple models and cells from injured tissue and fibrotic tissue.
Collapse
Affiliation(s)
- Yitian Zhang
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Weihui Zeng
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
21
|
Marchesini A, De Francesco F, Mattioli-Belmonte M, Zingaretti N, Riccio V, Orlando F, Zavan B, Riccio M. A New Animal Model for Pathological Subcutaneous Fibrosis: Surgical Technique and in vitro Analysis. Front Cell Dev Biol 2020; 8:542. [PMID: 32850775 PMCID: PMC7409519 DOI: 10.3389/fcell.2020.00542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 06/09/2020] [Indexed: 12/02/2022] Open
Abstract
Fibrosis is a condition that affects the connective tissue in an organ or tissue in the restorative or responsive phase as a result of injury. The consequences of excessive fibrotic tissue growth may lead to various physiological complications of deformity and impairment due to hypertrophic scars, keloids, and tendon adhesion without understating the psychological impact on the patient. However, no method accurately quantifies the rate and pattern of subcutaneous induced hypertrophic fibrosis. We, therefore, devised a rodent excisional model to evaluate the extent of fibrosis with talc. Tissue specimens were set on formalin, and paraffin sections for histological, immunohistochemical, and molecular analysis talc was used to induce the fibroproliferative mechanism typical of hypertrophic scars. This pathway is relevant to the activation of inflammatory and fibrotic agents to stimulate human hypertrophic scarring. This model reproduces morpho-functional features of human hypertrophic scars to investigate scar formation and assess potential anti-scarring therapies.
Collapse
Affiliation(s)
- Andrea Marchesini
- Department of Reconstructive Surgery and Hand Surgery, AOU "Ospedali Riuniti", Ancona, Italy
| | - Francesco De Francesco
- Department of Reconstructive Surgery and Hand Surgery, AOU "Ospedali Riuniti", Ancona, Italy
| | - Monica Mattioli-Belmonte
- Department of Clinical and Molecular Sciences, Università Politecnica delle Marche, Ancona, Italy
| | - Nicola Zingaretti
- Clinic of Plastic and Reconstructive Surgery, Department of Medical Area (DAME), Academic Hospital of Udine, University of Udine, Udine, Italy
| | - Valentina Riccio
- Veterinary Medical School, University of Camerino, Camerino, Italy
| | - Fiorenza Orlando
- Experimental Animal Models for Aging Unit, Scientific Technological Area, IRCCS INRNCA, Ancona, Italy
| | - Barbara Zavan
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Michele Riccio
- Department of Reconstructive Surgery and Hand Surgery, AOU "Ospedali Riuniti", Ancona, Italy
| |
Collapse
|
22
|
Henn D, Chen K, Maan ZN, Greco AH, Moortgat Illouz SE, Bonham CA, Barrera JA, Trotsyuk AA, Padmanabhan J, Momeni A, Wan DC, Nguyen D, Januszyk M, Gurtner GC. Cryopreserved human skin allografts promote angiogenesis and dermal regeneration in a murine model. Int Wound J 2020; 17:925-936. [PMID: 32227459 DOI: 10.1111/iwj.13349] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 03/11/2020] [Accepted: 03/13/2020] [Indexed: 12/20/2022] Open
Abstract
Cryopreserved human skin allografts (CHSAs) are used for the coverage of major burns when donor sites for autografts are insufficiently available and have clinically shown beneficial effects on chronic non-healing wounds. However, the biologic mechanisms behind the regenerative properties of CHSA remain elusive. Furthermore, the impact of cryopreservation on the immunogenicity of CHSA has not been thoroughly investigated and raised concerns with regard to their clinical application. To investigate the importance and fate of living cells, we compared cryopreserved CHSA with human acellular dermal matrix (ADM) grafts in which living cells had been removed by chemical processing. Both grafts were subcutaneously implanted into C57BL/6 mice and explanted after 1, 3, 7, and 28 days (n = 5 per group). A sham surgery where no graft was implanted served as a control. Transmission electron microscopy (TEM) and flow cytometry were used to characterise the ultrastructure and cells within CHSA before implantation. Immunofluorescent staining of tissue sections was used to determine the immune reaction against the implanted grafts, the rate of apoptotic cells, and vascularisation as well as collagen content of the overlaying murine dermis. Digital quantification of collagen fibre alignment on tissue sections was used to quantify the degree of fibrosis within the murine dermis. A substantial population of live human cells with intact organelles was identified in CHSA prior to implantation. Subcutaneous pockets with implanted xenografts or ADMs healed without clinically apparent rejection and with a similar cellular immune response. CHSA implantation largely preserved the cellularity of the overlying murine dermis, whereas ADM was associated with a significantly higher rate of cellular apoptosis, identified by cleaved caspase-3 staining, and a stronger dendritic cell infiltration of the murine dermis. CHSA was found to induce a local angiogenic response, leading to significantly more vascularisation of the murine dermis compared with ADM and sham surgery on day 7. By day 28, aggregate collagen-1 content within the murine dermis was greater following CHSA implantation compared with ADM. Collagen fibre alignment of the murine dermis, correlating with the degree of fibrosis, was significantly greater in the ADM group, whereas CHSA maintained the characteristic basket weave pattern of the native murine dermis. Our data indicate that CHSAs promote angiogenesis and collagen-1 production without eliciting a significant fibrotic response in a xenograft model. These findings may provide insight into the beneficial effects clinically observed after treatment of chronic wounds and burns with CHSA.
Collapse
Affiliation(s)
- Dominic Henn
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA.,Department of Hand, Plastic, and Reconstructive Surgery, BG Trauma Center Ludwigshafen, Ruprecht-Karls-University of Heidelberg, Heidelberg, Germany
| | - Kellen Chen
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Zeshaan N Maan
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Autumn H Greco
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Sylvia E Moortgat Illouz
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Clark A Bonham
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Janos A Barrera
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Artem A Trotsyuk
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Jagannath Padmanabhan
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Arash Momeni
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Derrick C Wan
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Dung Nguyen
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Michael Januszyk
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric and Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, California, USA
| |
Collapse
|
23
|
Jiang D, Rinkevich Y. Scars or Regeneration?-Dermal Fibroblasts as Drivers of Diverse Skin Wound Responses. Int J Mol Sci 2020; 21:E617. [PMID: 31963533 PMCID: PMC7014275 DOI: 10.3390/ijms21020617] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 01/13/2020] [Accepted: 01/15/2020] [Indexed: 12/13/2022] Open
Abstract
Scarring and regeneration are two physiologically opposite endpoints to skin injuries, with mammals, including humans, typically healing wounds with fibrotic scars. We aim to provide an updated review on fibroblast heterogeneity as determinants of the scarring-regeneration continuum. We discuss fibroblast-centric mechanisms that dictate scarring-regeneration continua with a focus on intercellular and cell-matrix adhesion. Improved understanding of fibroblast lineage-specific mechanisms and how they determine scar severity will ultimately allow for the development of antiscarring therapies and the promotion of tissue regeneration.
Collapse
Affiliation(s)
| | - Yuval Rinkevich
- Comprehensive Pneumology Center, Institute of Lung Biology and Disease, Helmholtz Zentrum München, Max-Lebsche-Platz 31, 81377 Munich, Germany;
| |
Collapse
|