51
|
Schneider M, Ziemer M, Lethaus B, Simon JC, Savkovic V. Generation of pigmented skin grafts from human hair follicles and dermal fibroblasts. Tissue Eng Part A 2021; 27:1333-1342. [PMID: 33573455 DOI: 10.1089/ten.tea.2020.0284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Skin equivalents are able to mimic key features of human skin and they can be used for a very broad range of applications, such as fundamental studies of skin biology, disease and toxicological models, as well as an alternative for animal testing. The high end of their use is in therapy of wound healing and repigmentation, disorders that massively affect individual health as well as quality of life and pose considerable burden to healthcare systems worldwide. Tissue-engineered skin grafts often originate from invasively obtained cell material (i.e. biopsy). Hereby, an unmet need for non-invasively gained autologous biological starting material has been created. The hair follicle, entirely non-invasively available by plucking, harbors a heterogeneous cell pool including stem cells with an immense differentiation capacity, hereby representing an attractive source of cells, especially for purposes of regenerative medicine. In this study, we engineered three-dimensional pigmented epidermal and dermoepidermal grafts using human keratinocytes and melanocytes from the outer root sheath of hair follicles combined with dermal fibroblasts. The grafts were generally anatomically correct and functional regarding stratification, formation of epidermal melanin units as well as extracellular matrix deposition, exhibiting moderate differences to the skin anatomy and function, typical for the in vitro culture.
Collapse
Affiliation(s)
- Marie Schneider
- Leipzig University, Saxon Incubator for Clinical Translation; Phillip-Rosenthal-Str. 55, Leipzig, Sachsen, Germany.,Leipzig University, Dept.of Hematology, Cell Therapy and Hemostaseology, University Hospital Leipzig, Liebigstr. 22 , 04103 Leipzig, Saxony, Germany;
| | - Mirjana Ziemer
- Leipzig University, Dept. of Dermatology, Venerology and Allergology, University Hospital Leipzig, Phillip-Rosenthal-Str. 23, Haus 10, Leipzig, Sachsen, Germany;
| | - Bernd Lethaus
- Leipzig University, Dept. of Cranio-Maxillofacial Surgery, University Hospital Leipzig, Liebigstr. 12, Leipzig, Sachsen, Germany;
| | - Jan Christoph Simon
- Leipzig University, 9180, Dept. of Dermatology, Venerology and Allergology, University Hospital Leipzig, Phillip-Rosenthal-Str. 23, Haus 10, Leipzig, Sachsen, Germany;
| | - Vuk Savkovic
- Leipzig University, Dept. of Cranio-Maxillofacial Surgery, Leipzig University Hospital, Liebigstr. 12, Leipzig, Sachsen, Germany;
| |
Collapse
|
52
|
De Leon SE, Cleuren L, Oo ZY, Stoddart PR, McArthur SL. Extending In-Plane Impedance Measurements from 2D to 3D Cultures: Design Considerations. Bioengineering (Basel) 2021; 8:11. [PMID: 33450860 PMCID: PMC7828367 DOI: 10.3390/bioengineering8010011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/21/2020] [Accepted: 12/29/2020] [Indexed: 11/22/2022] Open
Abstract
Three-dimensional (3D) cell cultures have recently emerged as tools for biologically modelling the human body. As 3D models make their way into laboratories there is a need to develop characterisation techniques that are sensitive enough to monitor the cells in real time and without the need for chemical labels. Impedance spectroscopy has been shown to address both of these challenges, but there has been little research into the full impedance spectrum and how the different components of the system affect the impedance signal. Here we investigate the impedance of human fibroblast cells in 2D and 3D collagen gel cultures across a broad range of frequencies (10 Hz to 5 MHz) using a commercial well with in-plane electrodes. At low frequencies in both 2D and 3D models it was observed that protein adsorption influences the magnitude of the impedance for the cell-free samples. This effect was eliminated once cells were introduced to the systems. Cell proliferation could be monitored in 2D at intermediate frequencies (30 kHz). However, the in-plane electrodes were unable to detect any changes in the impedance at any frequency when the cells were cultured in the 3D collagen gel. The results suggest that in designing impedance measurement devices, both the nature and distribution of the cells within the 3D culture as well as the architecture of the electrodes are key variables.
Collapse
Affiliation(s)
- Sorel E. De Leon
- Bioengineering Research Group, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (S.E.D.L.); (Z.Y.O.); (P.R.S.)
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3168, Australia
| | - Lana Cleuren
- PXL University College, Hasselt University, 3500 Hasselt, Belgium;
| | - Zay Yar Oo
- Bioengineering Research Group, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (S.E.D.L.); (Z.Y.O.); (P.R.S.)
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3168, Australia
| | - Paul R. Stoddart
- Bioengineering Research Group, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (S.E.D.L.); (Z.Y.O.); (P.R.S.)
| | - Sally L. McArthur
- Bioengineering Research Group, Faculty of Science, Engineering and Technology, Swinburne University of Technology, Hawthorn, VIC 3122, Australia; (S.E.D.L.); (Z.Y.O.); (P.R.S.)
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Clayton, VIC 3168, Australia
| |
Collapse
|
53
|
Organoid culture to study epithelial cell differentiation and barrier formation in the colon: bridging the gap between monolayer cell culture and human subject research. In Vitro Cell Dev Biol Anim 2021; 57:174-190. [PMID: 33403624 DOI: 10.1007/s11626-020-00534-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022]
Abstract
Organoid culture provides a powerful technology that can bridge the gap between monolayer cell culture on the one hand and whole animal or human subject research on the other. Tissues from many different organs from multiple species, including human, have already been successfully adapted to organoid growth. While optimal culture conditions have not yet been established for all tissue types, it seems that most tissues will, ultimately, be amenable to this type of culture. The colon is one of the tissues in which organoid culture was first established as a technology and which has been most successfully employed. The ready availability of histologically normal tissue as well as both premalignant and malignant tissue (often from the same individual) makes this possible. While individual tumors are highly variable relative to one another in organoid culture, a high degree of genotypic consistency exists between the tumor tissue and the histologically normal counterpart from a given source. Further, source material and tumor tissue in organoid culture demonstrate a high degree of genotypic consistency. Even after 6-9 mo in continuous culture, drift in the mutational profile has been shown to be minimal. Colon tissue maintained in organoid culture, thus, provides a good surrogate for the tissue of origin-a surrogate, however, that is as amenable to intervention with molecular, pharmacological, and immunological approaches as are more-traditionally studied cell lines.
Collapse
|
54
|
Ferrà-Cañellas MDM, Munar-Bestard M, Garcia-Sureda L, Lejeune B, Ramis JM, Monjo M. BMP4 micro-immunotherapy increases collagen deposition and reduces PGE2 release in human gingival fibroblasts and increases tissue viability of engineered 3D gingiva under inflammatory conditions. J Periodontol 2021; 92:1448-1459. [PMID: 33393105 PMCID: PMC8724682 DOI: 10.1002/jper.20-0552] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 11/13/2020] [Accepted: 12/18/2020] [Indexed: 12/16/2022]
Abstract
Background We aimed to evaluate the effect of low doses (LD) bone morphogenetic protein‐2 (BMP2) and BMP4 micro‐immunotherapy (MI) in two in vitro models of periodontal wound healing/regeneration. Methods We first evaluated the effect of LD of BMP2 and BMP4 MI on a 2D cell culture using human gingival fibroblasts (hGF) under inflammatory conditions induced by IL1β. Biocompatibility, inflammatory response (Prostaglandin E2 (PGE2) release), collagen deposition and release of extracellular matrix (ECM) organization‐related enzymes (matrix metalloproteinase‐1 (MMP1) and metalloproteinase inhibitor 1 (TIMP1)) were evaluated after short (3 days) and long‐term (24 days) treatment with BMP2 or BMP4 MI. Then, given the results obtained in the 2D cell culture, LD BMP4 MI treatment was evaluated in a 3D cell culture model of human tissue equivalent of gingiva (GTE) under the same inflammatory stimulus, evaluating the biocompatibility, inflammatory response and effect on MMP1 and TIMP1 release. Results LD BMP4 was able to decrease the release of the inflammatory mediator PGE2 and completely re‐establish the impaired collagen metabolism induced by IL1β treatment. In the 3D model, LD BMP4 treatment improved tissue viability compared with the vehicle, with similar levels to 3D tissues without inflammation. No significant effects were observed on PGE2 levels nor MMP1/TIMP1 ratio after LD BMP4 treatment, although a tendency to decrease PGE2 levels was observed after 3 days. Conclusions LD BMP4 MI treatment shows anti‐inflammatory and regenerative properties on hGF, and improved viability of 3D gingiva under inflammatory conditions. LD BMP4 MI treatment could be used on primary prevention or maintenance care of periodontitis.
Collapse
Affiliation(s)
- Maria Del Mar Ferrà-Cañellas
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain.,Preclinical Research Department, Labo'Life España, Consell, Spain
| | - Marta Munar-Bestard
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain.,Balearic Islands Health Research Institute (IdISBa), Palma de Mallorca, Spain
| | | | - Beatrice Lejeune
- Preclinical and Clinical Research, Regulatory Affairs Department, Labo'Life France, Nantes, France
| | - Joana Maria Ramis
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain.,Balearic Islands Health Research Institute (IdISBa), Palma de Mallorca, Spain
| | - Marta Monjo
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands, Palma de Mallorca, Spain.,Balearic Islands Health Research Institute (IdISBa), Palma de Mallorca, Spain
| |
Collapse
|
55
|
Przekora A. A Concise Review on Tissue Engineered Artificial Skin Grafts for Chronic Wound Treatment: Can We Reconstruct Functional Skin Tissue In Vitro? Cells 2020; 9:cells9071622. [PMID: 32640572 PMCID: PMC7407512 DOI: 10.3390/cells9071622] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
Chronic wounds occur as a consequence of a prolonged inflammatory phase during the healing process, which precludes skin regeneration. Typical treatment for chronic wounds includes application of autografts, allografts collected from cadaver, and topical delivery of antioxidant, anti-inflammatory, and antibacterial agents. Nevertheless, the mentioned therapies are not sufficient for extensive or deep wounds. Moreover, application of allogeneic skin grafts carries high risk of rejection and treatment failure. Advanced therapies for chronic wounds involve application of bioengineered artificial skin substitutes to overcome graft rejection as well as topical delivery of mesenchymal stem cells to reduce inflammation and accelerate the healing process. This review focuses on the concept of skin tissue engineering, which is a modern approach to chronic wound treatment. The aim of the article is to summarize common therapies for chronic wounds and recent achievements in the development of bioengineered artificial skin constructs, including analysis of biomaterials and cells widely used for skin graft production. This review also presents attempts to reconstruct nerves, pigmentation, and skin appendages (hair follicles, sweat glands) using artificial skin grafts as well as recent trends in the engineering of biomaterials, aiming to produce nanocomposite skin substitutes (nanofilled polymer composites) with controlled antibacterial activity. Finally, the article describes the composition, advantages, and limitations of both newly developed and commercially available bioengineered skin substitutes.
Collapse
Affiliation(s)
- Agata Przekora
- Department of Biochemistry and Biotechnology, Medical University of Lublin, Chodzki 1 Street, 20-093 Lublin, Poland
| |
Collapse
|
56
|
A Simple Method for the Production of Human Skin Equivalent in 3D, Multi-Cell Culture. Int J Mol Sci 2020; 21:ijms21134644. [PMID: 32629914 PMCID: PMC7369873 DOI: 10.3390/ijms21134644] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2020] [Revised: 06/26/2020] [Accepted: 06/27/2020] [Indexed: 01/01/2023] Open
Abstract
An important problem for researchers working in the field of dermatology is the preparation of the human skin equivalent (HSE). Here, we describe a simple and reliable protocol for preparing a skin model from the commercially available cell lines: keratinocytes, fibroblasts, and melanocytes. Importantly, in our 3D model, the keratinocytes are diverse that brings this model closer to the natural skin. For the production of HSE, we used available primary PCS-200-010, PCS-201-010, PCS-200-013, and immortalized CRL-4048 and CRL-4001 cell lines. We used genipin, which is necessary for collagen cross-linking and studied its cytotoxicity for keratinocytes and fibroblasts. The addition of 20 μM genipin reduced the shrinkage of the collagen in the constructs from 59% to 24% on day 12 of the culture of the construct. A higher concentration (80-200 µM) of genipin reduced shrinkage by 14% on average. Genipin in concentration 10 μM and below was not cytotoxic to the keratinocytes, and 150 μM and below to the fibroblasts. Hematoxylin and eosin staining showed that the morphology of HSEs was identical to that of native human skin. The immunohistochemical staining of the constructs showed the presence of vimentin-positive fibroblasts in the skin layer, while the melanocytes were in the epidermis and in the basal layer. We observed that the longer differentiation of constructs led to the higher secretion of GM-CSF, IL-10, IL-15, IL-1α, IL-6, IL-7, IL-8, and MCP-1. We also observed that the longer time of differentiation led to a more stable secretion of all analytes, which was reflected in the coefficient of variation. We described here a simple, reliable, and cost-effective production of the full-thickness human skin equivalents that can be used in the research and industry. With the global trend to decrease animal use for the research and testing, our HSE could be a useful testing tool and an alternative research model.
Collapse
|
57
|
Choi KYG, Wu BC, Lee AHY, Baquir B, Hancock REW. Utilizing Organoid and Air-Liquid Interface Models as a Screening Method in the Development of New Host Defense Peptides. Front Cell Infect Microbiol 2020; 10:228. [PMID: 32509598 PMCID: PMC7251080 DOI: 10.3389/fcimb.2020.00228] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 04/23/2020] [Indexed: 12/24/2022] Open
Abstract
Host defense peptides (HDPs), also known as antimicrobial peptides, are naturally occurring polypeptides (~12–50 residues) composed of cationic and hydrophobic amino acids that adopt an amphipathic conformation upon folding usually after contact with membranes. HDPs have a variety of biological activities including immunomodulatory, anti-inflammatory, anti-bacterial, and anti-biofilm functions. Although HDPs have the potential to address the global threat of antibiotic resistance and to treat immune and inflammatory disorders, they have yet to achieve this promise. Indeed, there are several challenges associated with bringing peptide-based drug candidates from the lab bench to clinical practice, including identifying appropriate indications, stability, toxicity, and cost. These challenges can be addressed in part by the development of innate defense regulator (IDR) peptides and peptidomimetics, which are synthetic derivatives of HDPs with similar or better efficacy, increased stability, and reduced toxicity and cost of the original HDP. However, one of the largest gaps between basic research and clinical application is the validity and translatability of conventional model systems, such as cell lines and animal models, for screening HDPs and their derivatives as potential drug therapies. Indeed, such translation has often relied on animal models, which have only limited validity. Here we discuss the recent development of human organoids for disease modeling and drug screening, assisted by the use of omics analyses. Organoids, developed from primary cells, cell lines, or human pluripotent stem cells, are three-dimensional, self-organizing structures that closely resemble their corresponding in vivo organs with regards to immune responses, tissue organization, and physiological properties; thus, organoids represent a reliable method for studying efficacy, formulation, toxicity and to some extent drug stability and pharmacodynamics. The use of patient-derived organoids enables the study of patient-specific efficacy, toxicogenomics and drug response predictions. We outline how organoids and omics data analysis can be leveraged to aid in the clinical translation of IDR peptides.
Collapse
Affiliation(s)
- Ka-Yee Grace Choi
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Bing Catherine Wu
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Amy Huei-Yi Lee
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada.,Department of Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, BC, Canada
| | - Beverlie Baquir
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Robert E W Hancock
- Department of Microbiology and Immunology, Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
58
|
Kwak BS, Jin SP, Kim SJ, Kim EJ, Chung JH, Sung JH. Microfluidic skin chip with vasculature for recapitulating the immune response of the skin tissue. Biotechnol Bioeng 2020; 117:1853-1863. [PMID: 32100875 DOI: 10.1002/bit.27320] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 02/16/2020] [Accepted: 02/24/2020] [Indexed: 12/19/2022]
Abstract
There is a considerable need for cell-based in vitro skin models for studying dermatological diseases and testing cosmetic products, but current in vitro skin models lack physiological relevance compared to human skin tissue. For example, many dermatological disorders involve complex immune responses, but current skin models are not capable of recapitulating the phenomena. Previously, we reported development of a microfluidic skin chip with a vessel structure and vascular endothelial cells. In this study, we cocultured dermal fibroblasts and keratinocytes with vascular endothelial cells, human umbilical vascular endothelial cells. We verified the formation of a vascular endothelium in the presence of the dermis and epidermis layers by examining the expression of tissue-specific markers. As the vascular endothelium plays a critical role in the migration of leukocytes to inflammation sites, we incorporated leukocytes in the circulating media and attempted to mimic the migration of neutrophils in response to external stimuli. Increased secretion of cytokines and migration of neutrophils was observed when the skin chip was exposed to ultraviolet irradiation, showing that the microfluidic skin chip may be useful for studying the immune response of the human tissue.
Collapse
Affiliation(s)
- Bong Shin Kwak
- Department of Chemical Engineering, Hongik University, Republic of Korea
| | - Seon-Pil Jin
- Department of Dermatology, Seoul National University Hospital, Republic of Korea.,Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University College of Medicine, Republic of Korea
| | - Su Jung Kim
- DYNEBIO INC., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Eun Joo Kim
- DYNEBIO INC., Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Jin Ho Chung
- Department of Dermatology, Seoul National University Hospital, Republic of Korea.,Institute of Human-Environmental Interface Biology, Medical Research Center, Seoul National University College of Medicine, Republic of Korea
| | - Jong Hwan Sung
- Department of Chemical Engineering, Hongik University, Republic of Korea
| |
Collapse
|
59
|
Establishment and characterization of an immortalized human chondrocyte cell line. Biotechnol Lett 2020; 42:707-716. [PMID: 32040675 DOI: 10.1007/s10529-020-02827-y] [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: 11/02/2019] [Accepted: 01/26/2020] [Indexed: 10/25/2022]
Abstract
OBJECTIVES Following a specific number of mitotic divisions, primary chondrocytes undergo proliferative senescence, thwarting efforts to expand sufficient populations in vitro suitable to meet the needs of scientific research or medical therapies. Therefore, the human telomerase reverse transcriptase (TERT) was used to immortalize human chondrocyte and establish a cell line that escape from cellular senescence. RESULTS The human chondrocytes were successfully immortalized by ectopic stable expression of TERT. The established TERT-Chondrocyte cell line showed robust proliferation capacity, even in late passages up to P20, and displayed little cellular senescence. Moreover, TERT-Chondrocyte cells at 20th passage showed similar chondrocyte properties to normal chondrocytes at early passages. CONCLUSIONS Ectopic stable expression of TERT is an effective way to immortalized human chondrocyte. The immortalized chondrocytes displayed little cellular senescence, showed promise as an in vitro model to investigate osteoarthritis, and may be a promising resource for cell-based therapy for damaged cartilage.
Collapse
|
60
|
Kallepalli A, McCall B, James DB, Junaid S, Halls J, Richardson MA. Optical investigation of three-dimensional human skin equivalents: A pilot study. JOURNAL OF BIOPHOTONICS 2020; 13:e201960053. [PMID: 31593618 DOI: 10.1002/jbio.201960053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/23/2019] [Accepted: 09/24/2019] [Indexed: 06/10/2023]
Abstract
Human skin equivalents (HSEs) are three-dimensional living models of human skin that are prepared in vitro by seeding cells onto an appropriate scaffold. They recreate the structure and biological behaviour of real skin, allowing the investigation of processes such as keratinocyte differentiation and interactions between the dermal and epidermal layers. However, for wider applications, their optical and mechanical properties should also replicate those of real skin. We therefore conducted a pilot study to investigate the optical properties of HSEs. We compared Monte Carlo simulations of (a) real human skin and (b) two-layer optical models of HSEs with (c) experimental measurements of transmittance through HSE samples. The skin layers were described using a hybrid collection of optical attenuation coefficients. A linear relationship was observed between the simulations and experiments. For samples thinner than 0.5 mm, an exponential increase in detected power was observed due to fewer instances of absorption and scattering.
Collapse
Affiliation(s)
- Akhil Kallepalli
- Sensors Group, Centre for Electronic Warfare, Information and Cyber, Defence Academy of the United Kingdom, Cranfield University, Shrivenham Campus, Shrivenham, UK
| | - Blake McCall
- Aston Institute of Materials Research, Engineering and Applied Sciences, Aston University, Birmingham, UK
| | - David B James
- Sensors Group, Centre for Electronic Warfare, Information and Cyber, Defence Academy of the United Kingdom, Cranfield University, Shrivenham Campus, Shrivenham, UK
| | - Sarah Junaid
- Aston Institute of Materials Research, Engineering and Applied Sciences, Aston University, Birmingham, UK
| | - James Halls
- Department of Radiology, The Great Western Hospital, Swindon, UK
| | - Mark A Richardson
- Sensors Group, Centre for Electronic Warfare, Information and Cyber, Defence Academy of the United Kingdom, Cranfield University, Shrivenham Campus, Shrivenham, UK
| |
Collapse
|
61
|
Bal-Öztürk A, Miccoli B, Avci-Adali M, Mogtader F, Sharifi F, Çeçen B, Yaşayan G, Braeken D, Alarcin E. Current Strategies and Future Perspectives of Skin-on-a-Chip Platforms: Innovations, Technical Challenges and Commercial Outlook. Curr Pharm Des 2019; 24:5437-5457. [PMID: 30727878 DOI: 10.2174/1381612825666190206195304] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 02/02/2019] [Indexed: 01/09/2023]
Abstract
The skin is the largest and most exposed organ in the human body. Not only it is involved in numerous biological processes essential for life but also it represents a significant endpoint for the application of pharmaceuticals. The area of in vitro skin tissue engineering has been progressing extensively in recent years. Advanced in vitro human skin models strongly impact the discovery of new drugs thanks to the enhanced screening efficiency and reliability. Nowadays, animal models are largely employed at the preclinical stage of new pharmaceutical compounds development for both risk assessment evaluation and pharmacokinetic studies. On the other hand, animal models often insufficiently foresee the human reaction due to the variations in skin immunity and physiology. Skin-on-chips devices offer innovative and state-of-the-art platforms essential to overcome these limitations. In the present review, we focus on the contribution of skin-on-chip platforms in fundamental research and applied medical research. In addition, we also highlighted the technical and practical difficulties that must be overcome to enhance skin-on-chip platforms, e.g. embedding electrical measurements, for improved modeling of human diseases as well as of new drug discovery and development.
Collapse
Affiliation(s)
- Ayça Bal-Öztürk
- Department of Analytical Chemistry, Faculty of Pharmacy, İstinye University, 34010, Zeytinburnu, Istanbul, Turkey,Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey
| | - Beatrice Miccoli
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium,Katholieke Universiteit Leuven, 3000 Leuven, Belgium
| | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tuebingen, Calwerstraße 7/1, 72076 Tuebingen, Germany
| | - Ferzaneh Mogtader
- Department of Stem Cell and Tissue Engineering, Institute of Health Sciences, Istinye University, 34010 Istanbul, Turkey,NanoBMT, Cyberpark, Bilkent 06800, Ankara, Turkey
| | - Fatemeh Sharifi
- Department of Mechanical Engineering, Sharif University of Technology, Tehran 11365-11155, Iran
| | - Berivan Çeçen
- Biomechanics Department, Institute of Health Science, Dokuz Eylul University, 35340, Inciraltı, Izmir, Turkey; Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, Istanbul, Turkey
| | - Gökçen Yaşayan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34668, Haydarpaşa, Istanbul, Turkey
| | - Dries Braeken
- Imec, Department of Life Sciences and Imaging, 3001 Heverlee, Belgium
| | - Emine Alarcin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Marmara University, 34668, Haydarpaşa, Istanbul, Turkey
| |
Collapse
|
62
|
Naito C, Yamaguchi T, Katsumi H, Kimura S, Kamei S, Morishita M, Sakane T, Kawabata K, Yamamoto A. Utility of Three-Dimensional Skin From Human-Induced Pluripotent Stem Cells as a Tool to Evaluate Transdermal Drug Permeation. J Pharm Sci 2019; 108:3524-3527. [DOI: 10.1016/j.xphs.2019.07.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/08/2019] [Accepted: 07/17/2019] [Indexed: 11/29/2022]
|
63
|
Li J, Wang J, Wang Z, Xia Y, Zhou M, Zhong A, Sun J. Experimental models for cutaneous hypertrophic scar research. Wound Repair Regen 2019; 28:126-144. [PMID: 31509318 DOI: 10.1111/wrr.12760] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 08/29/2019] [Accepted: 09/06/2019] [Indexed: 01/07/2023]
Abstract
Human skin wound repair may result in various outcomes with most of them leading to scar formation. Commonly seen in many cutaneous wound healing cases, hypertrophic scars are considered as phenotypes of abnormal wound repair. To prevent the formation of hypertrophic scars, efforts have been made to understand the mechanism of scarring following wound closure. Numerous in vivo and in vitro models have been created to facilitate investigations into cutaneous scarring and the development of antiscarring treatments. To select the best model for a specific study, background knowledge of the current models of hypertrophic scars is necessary. In this review, we describe in vivo and in vitro models for studying hypertrophic scars, as well as the distinct characteristics of these models. The choice of models for a specific study should be based on the characteristics of the model and the goal of the study. In general, in vivo animal models are often used in phenotypical scar formation analysis, development of antiscarring treatment, and functional analyses of individual genes. In contrast, in vitro models are chosen to pathway identification during scar formation as well as in high-throughput analysis in drug development. Besides helping investigators choose the best scarring model for their research, the goal of this review is to provide knowledge for improving the existing models and development of new models. These will contribute to the progress of scarring studies.
Collapse
Affiliation(s)
- Jialun Li
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jiecong Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yun Xia
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Muran Zhou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Aimei Zhong
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
64
|
Direct 3D bioprinted full-thickness skin constructs recapitulate regulatory signaling pathways and physiology of human skin. ACTA ACUST UNITED AC 2019. [DOI: 10.1016/j.bprint.2019.e00051] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
65
|
Beckert B, Panico F, Pollmann R, Eming R, Banning A, Tikkanen R. Immortalized Human hTert/KER-CT Keratinocytes a Model System for Research on Desmosomal Adhesion and Pathogenesis of Pemphigus Vulgaris. Int J Mol Sci 2019; 20:ijms20133113. [PMID: 31247885 PMCID: PMC6651391 DOI: 10.3390/ijms20133113] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 06/24/2019] [Accepted: 06/24/2019] [Indexed: 12/18/2022] Open
Abstract
Pemphigus Vulgaris is an autoimmune disease that results in blister formation in the epidermis and in mucosal tissues due to antibodies recognizing desmosomal cadherins, mainly desmoglein-3 and -1. Studies on the molecular mechanisms of Pemphigus have mainly been carried out using the spontaneously immortalized human keratinocyte cell line HaCaT or in primary keratinocytes. However, both cell systems have suboptimal features, with HaCaT cells exhibiting a large number of chromosomal aberrations and mutated p53 tumor suppressor, whereas primary keratinocytes are short-lived, heterogeneous and not susceptible to genetic modifications due to their restricted life-span. We have here tested the suitability of the commercially available human keratinocyte cell line hTert/KER-CT as a model system for research on epidermal cell adhesion and Pemphigus pathomechanisms. We here show that hTert cells exhibit a calcium dependent expression of desmosomal cadherins and are well suitable for typical assays used for studies on Pemphigus, such as sequential detergent extraction and Dispase-based dissociation assay. Treatment with Pemphigus auto-antibodies results in loss of monolayer integrity and altered localization of desmoglein-3, as well as loss of colocalization with flotillin-2. Our findings demonstrate that hTert cells are well suitable for studies on epidermal cell adhesion and Pemphigus pathomechanisms.
Collapse
Affiliation(s)
- Benedikt Beckert
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Francesca Panico
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Robert Pollmann
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Rüdiger Eming
- Department of Dermatology and Allergology, Philipps-Universität Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Antje Banning
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany
| | - Ritva Tikkanen
- Institute of Biochemistry, Medical Faculty, University of Giessen, Friedrichstrasse 24, 35392 Giessen, Germany.
| |
Collapse
|
66
|
Roger M, Fullard N, Costello L, Bradbury S, Markiewicz E, O'Reilly S, Darling N, Ritchie P, Määttä A, Karakesisoglou I, Nelson G, von Zglinicki T, Dicolandrea T, Isfort R, Bascom C, Przyborski S. Bioengineering the microanatomy of human skin. J Anat 2019; 234:438-455. [PMID: 30740672 PMCID: PMC6422806 DOI: 10.1111/joa.12942] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/07/2019] [Indexed: 12/12/2022] Open
Abstract
Recreating the structure of human tissues in the laboratory is valuable for fundamental research, testing interventions, and reducing the use of animals. Critical to the use of such technology is the ability to produce tissue models that accurately reproduce the microanatomy of the native tissue. Current artificial cell-based skin systems lack thorough characterisation, are not representative of human skin, and can show variation. In this study, we have developed a novel full thickness model of human skin comprised of epidermal and dermal compartments. Using an inert porous scaffold, we created a dermal construct using human fibroblasts that secrete their own extracellular matrix proteins, which avoids the use of animal-derived materials. The dermal construct acts as a foundation upon which epidermal keratinocytes were seeded and differentiated into a stratified keratinised epithelium. In-depth morphological analyses of the model demonstrated very close similarities with native human skin. Extensive immunostaining and electron microscopy analysis revealed ultrastructural details such as keratohyalin granules and lamellar bodies within the stratum granulosum, specialised junctional complexes, and the presence of a basal lamina. These features reflect the functional characteristics and barrier properties of the skin equivalent. Robustness and reproducibility of in vitro models are important attributes in experimental practice, and we demonstrate the consistency of the skin construct between different users. In summary, a new model of full thickness human skin has been developed that possesses microanatomical features reminiscent of native tissue. This skin model platform will be of significant interest to scientists researching the structure and function of human skin.
Collapse
Affiliation(s)
| | | | | | | | | | - Steven O'Reilly
- Department of Health and Life SciencesNorthumbria UniversityNewcastleUK
| | | | | | - Arto Määttä
- Department of BiosciencesDurham UniversityDurhamUK
| | | | - Glyn Nelson
- Institute for Ageing and HealthUniversity of NewcastleNewcastleUK
| | | | | | - Robert Isfort
- Mason Business Centre, Procter & GambleMason, CincinnatiOHUSA
| | - Charles Bascom
- Mason Business Centre, Procter & GambleMason, CincinnatiOHUSA
| | - Stefan Przyborski
- Department of BiosciencesDurham UniversityDurhamUK
- Reprocell EuropeSedgefieldUK
| |
Collapse
|
67
|
Muhsen M, Protschka M, Schneider LE, Müller U, Köhler G, Magin TM, Büttner M, Alber G, Siegemund S. Orf virus (ORFV) infection in a three-dimensional human skin model: Characteristic cellular alterations and interference with keratinocyte differentiation. PLoS One 2019; 14:e0210504. [PMID: 30699132 PMCID: PMC6353139 DOI: 10.1371/journal.pone.0210504] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Accepted: 12/24/2018] [Indexed: 01/01/2023] Open
Abstract
ORF virus (ORFV) is the causative agent of contagious ecthyma, a pustular dermatitis of small ruminants and humans. Even though the development of lesions caused by ORFV was extensively studied in animals, only limited knowledge exists about the lesion development in human skin. The aim of the present study was to evaluate a three-dimensional (3D) organotypic culture (OTC) as a human skin model for ORFV infection considering lesion development, replication of the virus, viral gene transcription and modulation of differentiation of human keratinocytes by ORFV. ORFV infection of OTC was performed using the ORFV isolate B029 derived from a human patient. The OTC sections showed a similar structure of stratified epidermal keratinocytes as human foreskin and a similar expression profile of the differentiation markers keratin 1 (K1), K10, and loricrin. Upon ORFV infection, OTCs exhibited histological cytopathic changes including hyperkeratosis and ballooning degeneration of the keratinocytes. ORFV persisted for 10 days and was located in keratinocytes of the outer epidermal layers. ORFV-specific early, intermediate and late genes were transcribed, but limited viral spread and restricted cell infection were noticed. ORFV infection resulted in downregulation of K1, K10, and loricrin at the transcriptional level without affecting proliferation as shown by PCNA or Ki-67 expression. In conclusion, OTC provides a suitable model to study the interaction of virus with human keratinocytes in a similar structural setting as human skin and reveals that ORFV infection downregulates several differentiation markers in the epidermis of the human skin, a hitherto unknown feature of dermal ORFV infection in man.
Collapse
Affiliation(s)
- Mahmod Muhsen
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Martina Protschka
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Laura E. Schneider
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Uwe Müller
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | | | - Thomas M. Magin
- Institute of Biology, Division of Cell and Developmental Biology, University of Leipzig, Leipzig, Germany
| | - Mathias Büttner
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Gottfried Alber
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| | - Sabine Siegemund
- Institute of Immunology/Molecular Pathogenesis, Center for Biotechnology and Biomedicine, College of Veterinary Medicine, University of Leipzig, Leipzig, Germany
| |
Collapse
|
68
|
Sriram G, Bigliardi PL, Bigliardi-Qi M. Full-Thickness Human Skin Equivalent Models of Atopic Dermatitis. Methods Mol Biol 2019; 1879:367-383. [PMID: 29790095 DOI: 10.1007/7651_2018_163] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Atopic dermatitis is a chronic inflammatory skin disease caused by complex multifactorial etiology. In the recent years, there have been significant advances in tissue engineering and the generation of in vitro skin models representative of healthy and diseased states. This chapter describes the methodology for the fabrication of in vitro human skin equivalent (HSE) from human keratinocytes and fibroblasts using a fibrin-based dermal matrix and serum-free culture conditions. Modification of the culture conditions with the supplementation of Th2 cytokines such as interleukin-4 induces the development of atopic dermatitis-like skin model. The chapter also describes the histological and immunohistochemical tools for characterization of the HSE model. The reconstruction of tissue-engineered HSE models that recapitulate the essential features of atopic dermatitis provides powerful tools for deeper understanding of the underlying pathological mechanisms on epidermal level, identification and testing of novel treatment options, and safety and toxicological evaluation in a pathophysiologically relevant system.
Collapse
Affiliation(s)
- Gopu Sriram
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore.
| | | | - Mei Bigliardi-Qi
- Department of Dermatology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
69
|
di Luca M, Curcio M, Valli E, Cirillo G, Voli F, Butini ME, Farfalla A, Pantuso E, Leggio A, Nicoletta FP, Tavanti A, Iemma F, Vittorio O. Combining antioxidant hydrogels with self-assembled microparticles for multifunctional wound dressings. J Mater Chem B 2019. [DOI: 10.1039/c9tb00871c] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
A multi-functional composite to be employed as a dressing material was prepared by combining hydrogel and microparticle systems.
Collapse
|
70
|
Engineering a Multilayered Skin Equivalent: The Importance of Endogenous Extracellular Matrix Maturation to Provide Robustness and Reproducibility. Methods Mol Biol 2019; 1993:107-122. [PMID: 31148082 DOI: 10.1007/978-1-4939-9473-1_9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Human skin equivalents (HSEs) are a valuable tool for both academic and industrial laboratories to further the understanding of skin physiology and associated diseases. Over the last few decades, there have been many advances in the development of HSEs that successfully recapitulate the structure of human skin in vitro; however a main limitation is variability due to the use of complex protocols and exogenous extracellular matrix (ECM) proteins. We have developed a robust and unique full-thickness skin equivalent that is highly reproducible due to the use of a consistent scaffold, commercially available cells, and defined low-serum media. The Alvetex® scaffold technology allows fibroblasts to produce their own endogenous ECM proteins within the scaffold, which alleviates the need for exogenous collagen, and supports the differentiation and stratification of the epidermis. Our full-thickness skin equivalent is generated using a detailed step-by-step protocol, which sequentially forms the multilayered structure of human skin in vitro. This model can be adapted for many downstream applications such as disease modeling and testing of active compounds for cosmetics.
Collapse
|
71
|
|
72
|
Kwak BS, Choi W, Jeon JW, Won JI, Sung GY, Kim B, Sung JH. In vitro 3D skin model using gelatin methacrylate hydrogel. J IND ENG CHEM 2018. [DOI: 10.1016/j.jiec.2018.05.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
73
|
In Vitro Models for Studying Transport Across Epithelial Tissue Barriers. Ann Biomed Eng 2018; 47:1-21. [DOI: 10.1007/s10439-018-02124-w] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022]
|
74
|
Jung JP, Lin WH, Riddle MJ, Tolar J, Ogle BM. A 3D in vitro model of the dermoepidermal junction amenable to mechanical testing. J Biomed Mater Res A 2018; 106:3231-3238. [PMID: 30208260 PMCID: PMC6283247 DOI: 10.1002/jbm.a.36519] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 07/20/2018] [Accepted: 07/31/2018] [Indexed: 01/06/2023]
Abstract
Recessive dystrophic Epidermolysis Bullosa (RDEB) is caused by mutations in collagen‐type VII gene critical for the dermoepidermal junction (DEJ) formation. Neither tissues of animal models nor currently available in vitro models are amenable to the quantitative assessment of mechanical adhesion between dermal and epidermal layers. Here, we created a 3D in vitro DEJ model using extracellular matrix (ECM) proteins of the DEJ anchored to a poly(ethylene glycol)‐based slab (termed ECM composites) and seeded with human keratinocytes and dermal fibroblasts. Keratinocytes and fibroblasts of healthy individuals were well maintained in the ECM composite and showed the expression of collagen type VII over a 2‐week period. The ECM composites with healthy keratinocytes and fibroblasts exhibited yield stress associated with the separation of the model DEJ at 0.268 ± 0.057 kPa. When we benchmarked this measure of adhesive strength with that of the model DEJ fabricated with cells of individuals with RDEB, the yield stress was significantly lower (0.153 ± 0.064 kPa) consistent with our current mechanistic understanding of RDEB. In summary, a 3D in vitro model DEJ was developed for quantification of mechanical adhesion between epidermal‐ and dermal‐mimicking layers, which can be utilized for assessment of mechanical adhesion of the model DEJ applicable for Epidermolysis Bullosa‐associated therapeutics. © 2018 The Authors. Journal Of Biomedical Materials Research Part A Published By Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 3231–3238, 2018.
Collapse
Affiliation(s)
- Jangwook P Jung
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Department of Biological Engineering, Louisiana State University, Baton Rouge, Louisiana
| | - Wei-Han Lin
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Megan J Riddle
- Department of Pediatrics, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Jakub Tolar
- Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Department of Pediatrics, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| | - Brenda M Ogle
- Department of Biomedical Engineering, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Stem Cell Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Masonic Cancer Center, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Lillehei Heart Institute, University of Minnesota-Twin Cities, Minneapolis, Minnesota.,Institute for Engineering in Medicine, University of Minnesota-Twin Cities, Minneapolis, Minnesota
| |
Collapse
|
75
|
Pupovac A, Senturk B, Griffoni C, Maniura-Weber K, Rottmar M, McArthur SL. Toward Immunocompetent 3D Skin Models. Adv Healthc Mater 2018. [PMID: 29542274 DOI: 10.1002/adhm.201701405] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
3D human skin models provide a platform for toxicity testing, biomaterials evaluation, and investigation of fundamental biological processes. However, the majority of current in vitro models lack an inflammatory system, vasculature, and other characteristics of native skin, indicating scope for more physiologically complex models. Looking at the immune system, there are a variety of cells that could be integrated to create novel skin models, but to do this effectively it is also necessary to understand the interface between skin biology and tissue engineering as well as the different roles the immune system plays in specific health and disease states. Here, a progress report on skin immunity and current immunocompetent skin models with a focus on construction methods is presented; scaffold and cell choice as well as the requirements of physiologically relevant models are elaborated. The wide range of technological and fundamental challenges that need to be addressed to successfully generate immunocompetent skin models and the steps currently being made globally by researchers as they develop new models are explored. Induced pluripotent stem cells, microfluidic platforms to control the model environment, and new real-time monitoring techniques capable of probing biochemical processes within the models are discussed.
Collapse
Affiliation(s)
- Aleta Pupovac
- Faculty of Science; Engineering and Technology; Swinburne University of Technology; Hawthorn Victoria 3122 Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO); Probing Biosystems Future Science Platform and Manufacturing; Clayton Victoria 3168 Australia
| | - Berna Senturk
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Chiara Griffoni
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Markus Rottmar
- Laboratory for Biointerfaces; Empa; Swiss Federal Laboratories for Materials Science and Technology; 9014 St. Gallen Switzerland
| | - Sally L. McArthur
- Faculty of Science; Engineering and Technology; Swinburne University of Technology; Hawthorn Victoria 3122 Australia
- Commonwealth Scientific and Industrial Research Organization (CSIRO); Probing Biosystems Future Science Platform and Manufacturing; Clayton Victoria 3168 Australia
| |
Collapse
|
76
|
Jedrusik N, Meyen C, Finkenzeller G, Stark GB, Meskath S, Schulz SD, Steinberg T, Eberwein P, Strassburg S, Tomakidi P. Nanofibered Gelatin-Based Nonwoven Elasticity Promotes Epithelial Histogenesis. Adv Healthc Mater 2018. [PMID: 29529354 DOI: 10.1002/adhm.201700895] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Regarding tissue regeneration, mechanics of biomaterials gains progressive importance. Therefore, this study reports on in situ crosslinked electrospun gelatin nonwoven mats (NWMs) whose distinct modulus of elasticity (ME) promotes epithelial tissue formation in a graded manner. NWMs, comprising fiber diameters in various distributions, yield an ME of about 2.1, 3.2, and 10.9 kPa. A two-step approach of preclinical in vitro validation identifies the elasticity of 3.2 kPa as superior to the other, regarding the histogenetic epithelial outcome. Hence, this 3.2 kPa candidate NWM is colonized with oral mucosal epithelial keratinocytes in the absence or presence of mesenchymal fibroblasts and/or endothelial cells. Evaluation of epithelial histogenesis at days 1 to 10 occurs by colorimetric and fluorescence-based immunohistochemistry (IHCH) of specific biomarkers. These include cytokeratins (CK) 14, CK1, and involucrin that indicate different stages of epithelial differentiation, as well as the basement membrane constituent collagen type IV and Ki-67 as a proliferation marker. Intriguingly, histogenesis and IHCH reveal the best resemblance of the native epithelium by the NWM alone, irrespective of other cell counterparts. These findings prove the gelatin NWM a convenient cell matrix, and evidence that NWM mechanics is important to promote epithelial histogenesis in view of prospective clinical applications.
Collapse
Affiliation(s)
- Nicole Jedrusik
- Division of Oral Biotechnology; Center for Dental Medicine; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Christoph Meyen
- Department of Plastic and Hand Surgery; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Günter Finkenzeller
- Department of Plastic and Hand Surgery; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - G. Björn Stark
- Department of Plastic and Hand Surgery; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Stephan Meskath
- Department of Orthopedics and Trauma Surgery; Medical Center - University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Simon Daniel Schulz
- Division of Oral Biotechnology; Center for Dental Medicine; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Thorsten Steinberg
- Division of Oral Biotechnology; Center for Dental Medicine; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Philipp Eberwein
- Eye Center; Medical Center - University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Sandra Strassburg
- Department of Plastic and Hand Surgery; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| | - Pascal Tomakidi
- Division of Oral Biotechnology; Center for Dental Medicine; Medical Center-; University of Freiburg; Faculty of Medicine; University of Freiburg; 79106 Freiburg Germany
| |
Collapse
|
77
|
Skin corrosion test: a comparison between reconstructed human epidermis and full thickness skin models. Eur J Pharm Biopharm 2018; 125:51-57. [DOI: 10.1016/j.ejpb.2018.01.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 11/23/2022]
|
78
|
|
79
|
Löwa A, Vogt A, Kaessmeyer S, Hedtrich S. Generation of full-thickness skin equivalents using hair follicle-derived primary human keratinocytes and fibroblasts. J Tissue Eng Regen Med 2018; 12:e2134-e2146. [PMID: 29377584 DOI: 10.1002/term.2646] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 12/15/2017] [Accepted: 01/16/2018] [Indexed: 02/03/2023]
Abstract
Skin equivalents are increasingly used as human-based test systems for basic and preclinical research. Most of the established skin equivalents are composed of primary keratinocytes and fibroblasts, isolated either from excised human skin or juvenile foreskin following circumcisions. Although the potential of hair follicle-derived cells for the generation of skin equivalents has been shown, this approach normally requires microdissections from the scalp for which there is limited subject compliance or ethical approval. In the present study, we report a novel method to isolate and cultivate keratinocytes and fibroblasts from plucked hair follicles that were then used to generate skin equivalents. The procedure is non-invasive, inflicts little-pain, and may allow easy access to patient-derived cells without taking punch biopsies. Overall, minor differences in morphology, ultrastructure, expression of important structural proteins, or barrier function were observed between skin equivalents generated from hair follicle-derived or interfollicular keratinocytes and fibroblasts. Interestingly, improved basal lamina formation was seen in the hair follicle-derived skin equivalents. The presented method here allows easy and non-invasive access to keratinocytes and fibroblasts from plucked hair follicles that may be useful particularly for the generation of skin disease equivalents.
Collapse
Affiliation(s)
- Anna Löwa
- Institute for Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| | - Annika Vogt
- Experimental Research Unit Clinical Research Center for Hair and Skin Sciences, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Sabine Kaessmeyer
- Department of Veterinary Medicine, Institute for Veterinary Anatomy, Freie Universität Berlin, Berlin, Germany
| | - Sarah Hedtrich
- Institute for Pharmacy, Pharmacology and Toxicology, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
80
|
Pridgeon CS, Schlott C, Wong MW, Heringa MB, Heckel T, Leedale J, Launay L, Gryshkova V, Przyborski S, Bearon RN, Wilkinson EL, Ansari T, Greenman J, Hendriks DFG, Gibbs S, Sidaway J, Sison-Young RL, Walker P, Cross MJ, Park BK, Goldring CEP. Innovative organotypic in vitro models for safety assessment: aligning with regulatory requirements and understanding models of the heart, skin, and liver as paradigms. Arch Toxicol 2018; 92:557-569. [PMID: 29362863 PMCID: PMC5818581 DOI: 10.1007/s00204-018-2152-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 11/27/2017] [Indexed: 01/02/2023]
Abstract
The development of improved, innovative models for the detection of toxicity of drugs, chemicals, or chemicals in cosmetics is crucial to efficiently bring new products safely to market in a cost-effective and timely manner. In addition, improvement in models to detect toxicity may reduce the incidence of unexpected post-marketing toxicity and reduce or eliminate the need for animal testing. The safety of novel products of the pharmaceutical, chemical, or cosmetics industry must be assured; therefore, toxicological properties need to be assessed. Accepted methods for gathering the information required by law for approval of substances are often animal methods. To reduce, refine, and replace animal testing, innovative organotypic in vitro models have emerged. Such models appear at different levels of complexity ranging from simpler, self-organized three-dimensional (3D) cell cultures up to more advanced scaffold-based co-cultures consisting of multiple cell types. This review provides an overview of recent developments in the field of toxicity testing with in vitro models for three major organ types: heart, skin, and liver. This review also examines regulatory aspects of such models in Europe and the UK, and summarizes best practices to facilitate the acceptance and appropriate use of advanced in vitro models.
Collapse
Affiliation(s)
- Chris S Pridgeon
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Constanze Schlott
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Min Wei Wong
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Minne B Heringa
- National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Tobias Heckel
- Dr. Johannes Heidenhain GmbH, Dr.-Johannes-Heidenhain-Straße 5, 83301, Traunreut, Germany
| | - Joe Leedale
- Department of Mathematical Sciences, University of Liverpool, Liverpool, L69 7ZL, UK
| | | | - Vitalina Gryshkova
- Investigative Toxicology, Department of Non-Clinical Development, UCB Biopharma SPRL, 1420, Braine L'Alleud, Belgium
| | | | - Rachel N Bearon
- Department of Mathematical Sciences, University of Liverpool, Liverpool, L69 7ZL, UK
| | - Emma L Wilkinson
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Tahera Ansari
- Northwick Park Institute for Medical Research, Northwick Park and St Mark's Hospital, Middlesex, HA1 3UJ, UK
| | - John Greenman
- School of Life Sciences, University of Hull, Hull, HU6 7RX, UK
| | - Delilah F G Hendriks
- Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Sue Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands
| | | | - Rowena L Sison-Young
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Paul Walker
- Cyprotex Discovery Ltd, Cheshire, SK10 4TG, UK
| | - Mike J Cross
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - B Kevin Park
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK
| | - Chris E P Goldring
- Department of Molecular and Clinical Pharmacology, University of Liverpool, Liverpool, L69 3GE, UK.
| |
Collapse
|
81
|
Mieremet A, Rietveld M, van Dijk R, Bouwstra JA, El Ghalbzouri A. Recapitulation of Native Dermal Tissue in a Full-Thickness Human Skin Model Using Human Collagens. Tissue Eng Part A 2017; 24:873-881. [PMID: 29130419 DOI: 10.1089/ten.tea.2017.0326] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Full-thickness skin models comprise a three-dimensional dermal equivalent based on an animal-derived collagen matrix that harbors fibroblasts and an epidermal equivalent formed by keratinocytes. The functionality of both equivalents is influenced by many factors, including extracellular matrix composition and resident cell type. Animal-derived collagens differ in amino acid composition and physicochemical properties from human collagens. This composition could alter the functionality of the dermal equivalent and epidermal morphogenesis with the barrier formation in full-thickness models (FTMs). By replacement of animal-derived collagen for human collagen, we generated and characterized the animal material-free human collagen full-thickness models (hC-FTMs) that better mimic native dermal tissue. MATERIALS AND METHODS An isolation procedure to obtain soluble collagen from human abdominal dermis was developed. Both FTMs and hC-FTMs were generated with primary human fibroblasts and keratinocytes. Immunohistochemical analyses with biomarkers for the dermal matrix composition, basement membrane (BM) formation, epidermal proliferation, differentiation, and activation were performed. The stratum corneum (SC) lipid composition was studied with liquid chromatography-mass spectrometry. Lipid lamellar organization was determined by small-angle X-ray diffraction. RESULTS The FTMs and hC-FTMs exhibit many similarities, including the dermal matrix structure, BM formation, epidermal basal layer proliferation, and execution of differentiation programs. The SC contains a similar number of corneocyte layers and the same level of lipids. The ceramide chain length distribution and ceramide subclass profile showed only minor differences. Subsequently, this led to an unaltered lamellar organization. CONCLUSION The animal material-free hC-FTM is generated successfully using collagens isolated from human abdominal dermis. Utilization of human collagens revealed that (epi-)dermal morphogenesis and lipid barrier formation resembled that of original FTMs. The hC-FTMs contain a dermal equivalent that mimics the native stromal tissue to a higher extent. Therefore these in vitro skin models can be used as promising tool for research purposes that contribute to animal-free experimentation.
Collapse
Affiliation(s)
- Arnout Mieremet
- 1 Department of Dermatology, Leiden University Medical Centre , Leiden, The Netherlands
| | - Marion Rietveld
- 1 Department of Dermatology, Leiden University Medical Centre , Leiden, The Netherlands
| | - Rianne van Dijk
- 2 Division of Drug Delivery Technology, LACDR, Leiden University , Leiden, The Netherlands
| | - Joke A Bouwstra
- 2 Division of Drug Delivery Technology, LACDR, Leiden University , Leiden, The Netherlands
| | | |
Collapse
|
82
|
Grayson AK, Hearnden V, Bolt R, Jebreel A, Colley HE, Murdoch C. Use of a Rho kinase inhibitor to increase human tonsil keratinocyte longevity for three-dimensional, tissue engineered tonsil epithelium equivalents. J Tissue Eng Regen Med 2017; 12:e1636-e1646. [PMID: 29048773 DOI: 10.1002/term.2590] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 09/18/2017] [Accepted: 10/09/2017] [Indexed: 12/25/2022]
Abstract
The generation of tissue-engineered epithelial models is often hampered by the limited proliferative capacity of primary epithelial cells. This study aimed to isolate normal tonsillar keratinocytes (NTK) from human tonsils, increase the lifespan of these cells using the Rho kinase inhibitor Y-27632 and to develop tissue-engineered equivalents of healthy and infected tonsil epithelium. The proliferation rate of isolated NTK and expression of c-MYC and p16INK4A were measured in the absence or presence of the inhibitor. Y-27632-treated NTK were used to generate tissue-engineered tonsil epithelium equivalents using de-epidermised dermis that were then incubated with Streptococcus pyogenes to model bacterial tonsillitis, and the expression of pro-inflammatory cytokines was measured by cytokine array and ELISA. NTK cultured in the absence of Y-27632 rapidly senesced whereas cells cultured in the presence of this inhibitor proliferated for over 30 population doublings without changing their phenotype. Y-27632-treated NTK produced a multi-layered differentiated epithelium that histologically resembled normal tonsillar surface epithelium and responded to S. pyogenes infection by increased expression of pro-inflammatory cytokines including CXCL5 and IL-6. NTK can be isolated and successfully cultured in vitro with Y-27632 leading to a markedly prolonged lifespan without any deleterious consequences to cell morphology. This functional tissue-engineered equivalent of tonsil epithelium will provide a valuable tool for studying tonsil biology and host-pathogen interactions in a more physiologically relevant manner.
Collapse
Affiliation(s)
- Amy K Grayson
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, UK
| | - Vanessa Hearnden
- Department of Materials Science and Engineering, University of Sheffield, UK
| | - Robert Bolt
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, UK
| | - Ala Jebreel
- Sheffield Teaching Hospitals NHS Foundation Trust, Royal Hallamshire Hospital, Sheffield, UK
| | - Helen E Colley
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, UK
| | - Craig Murdoch
- School of Clinical Dentistry, Claremont Crescent, University of Sheffield, UK
| |
Collapse
|
83
|
Buskermolen JK, Reijnders CMA, Spiekstra SW, Steinberg T, Kleverlaan CJ, Feilzer AJ, Bakker AD, Gibbs S. Development of a Full-Thickness Human Gingiva Equivalent Constructed from Immortalized Keratinocytes and Fibroblasts. Tissue Eng Part C Methods 2017; 22:781-91. [PMID: 27406216 PMCID: PMC4991602 DOI: 10.1089/ten.tec.2016.0066] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Organotypic models make it possible to investigate the unique properties of oral mucosa in vitro. For gingiva, the use of human primary keratinocytes (KC) and fibroblasts (Fib) is limited due to the availability and size of donor biopsies. The use of physiologically relevant immortalized cell lines would solve these problems. The aim of this study was to develop fully differentiated human gingiva equivalents (GE) constructed entirely from cell lines, to compare them with the primary cell counterpart (Prim), and to test relevance in an in vitro wound healing assay. Reconstructed gingiva epithelium on a gingiva fibroblast-populated collagen hydrogel was constructed from cell lines (keratinocytes: TERT or HPV immortalized; fibroblasts: TERT immortalized) and compared to GE-Prim and native gingiva. GE were characterized by immunohistochemical staining for proliferation (Ki67), epithelial differentiation (K10, K13), and basement membrane (collagen type IV and laminin 5). To test functionality of GE-TERT, full-thickness wounds were introduced. Reepithelialization, fibroblast repopulation of hydrogel, metabolic activity (MTT assay), and (pro-)inflammatory cytokine release (enzyme-linked immunosorbent assay) were assessed during wound closure over 7 days. Significant differences in basal KC cytokine secretion (IL-1α, IL-18, and CXCL8) were only observed between KC-Prim and KC-HPV. When Fib-Prim and Fib-TERT were stimulated with TNF-α, no differences were observed regarding cytokine secretion (IL-6, CXCL8, and CCL2). GE-TERT histology, keratin, and basement membrane protein expression very closely represented native gingiva and GE-Prim. In contrast, the epithelium of GE made with HPV-immortalized KC was disorganized, showing suprabasal proliferating cells, limited keratinocyte differentiation, and the absence of basement membrane proteins. When a wound was introduced into the more physiologically relevant GE-TERT model, an immediate inflammatory response (IL-6, CCL2, and CXCL8) was observed followed by complete reepithelialization. Seven days after wounding, tissue integrity, metabolic activity, and cytokine levels had returned to the prewounded state. In conclusion, immortalized human gingiva KC and fibroblasts can be used to make physiologically relevant GE, which resemble either the healthy gingiva or a neoplastic disease model. These organotypic models will provide valuable tools to investigate oral mucosa biology and can also be used as an animal alternative for drug targeting, vaccination studies, microbial biofilm studies, and testing new therapeutics.
Collapse
Affiliation(s)
- Jeroen K Buskermolen
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands .,2 Department of Dental Materials Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | | | - Sander W Spiekstra
- 3 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| | - Thorsten Steinberg
- 4 Department of Oral Biotechnology, University Medical Center Freiburg , Freiburg, Germany
| | - Cornelis J Kleverlaan
- 2 Department of Dental Materials Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Albert J Feilzer
- 2 Department of Dental Materials Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Astrid D Bakker
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands
| | - Susan Gibbs
- 1 Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam , MOVE Research Institute Amsterdam, Amsterdam, The Netherlands .,3 Department of Dermatology, VU University Medical Centre , Amsterdam, The Netherlands
| |
Collapse
|
84
|
Ewart L, Dehne EM, Fabre K, Gibbs S, Hickman J, Hornberg E, Ingelman-Sundberg M, Jang KJ, Jones DR, Lauschke VM, Marx U, Mettetal JT, Pointon A, Williams D, Zimmermann WH, Newham P. Application of Microphysiological Systems to Enhance Safety Assessment in Drug Discovery. Annu Rev Pharmacol Toxicol 2017; 58:65-82. [PMID: 29029591 DOI: 10.1146/annurev-pharmtox-010617-052722] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Enhancing the early detection of new therapies that are likely to carry a safety liability in the context of the intended patient population would provide a major advance in drug discovery. Microphysiological systems (MPS) technology offers an opportunity to support enhanced preclinical to clinical translation through the generation of higher-quality preclinical physiological data. In this review, we highlight this technological opportunity by focusing on key target organs associated with drug safety and metabolism. By focusing on MPS models that have been developed for these organs, alongside other relevant in vitro models, we review the current state of the art and the challenges that still need to be overcome to ensure application of this technology in enhancing drug discovery.
Collapse
Affiliation(s)
- Lorna Ewart
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| | | | - Kristin Fabre
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451, USA
| | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, 1081 HZ Amsterdam, The Netherlands.,Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, 1081 LA Amsterdam, The Netherlands
| | - James Hickman
- NanoScience Technology Center, University of Central Florida, Orlando, Florida 32826, USA
| | - Ellinor Hornberg
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, 431 83 Mölndal, Sweden
| | - Magnus Ingelman-Sundberg
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - David R Jones
- Medicines & Healthcare Products Regulatory Agency, London SW1W 9SZ, United Kingdom
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Section of Pharmacogenetics, Karolinska Institutet, 171 77 Stockholm, Sweden
| | | | - Jerome T Mettetal
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Waltham, Massachusetts 02451, USA
| | - Amy Pointon
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| | - Dominic Williams
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| | - Wolfram-Hubertus Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen, Goettingen 37075, Germany.,German Center for Cardiovascular Research (DZHK), Goettingen 37075, Germany
| | - Peter Newham
- Drug Safety and Metabolism, Innovative Medicines and Early Development, AstraZeneca, Cambridge CB4 0WG, United Kingdom;
| |
Collapse
|
85
|
Smits JPH, Niehues H, Rikken G, van Vlijmen-Willems IMJJ, van de Zande GWHJF, Zeeuwen PLJM, Schalkwijk J, van den Bogaard EH. Immortalized N/TERT keratinocytes as an alternative cell source in 3D human epidermal models. Sci Rep 2017; 7:11838. [PMID: 28928444 PMCID: PMC5605545 DOI: 10.1038/s41598-017-12041-y] [Citation(s) in RCA: 130] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 09/01/2017] [Indexed: 12/24/2022] Open
Abstract
The strong societal urge to reduce the use of experimental animals, and the biological differences between rodent and human skin, have led to the development of alternative models for healthy and diseased human skin. However, the limited availability of primary keratinocytes to generate such models hampers large-scale implementation of skin models in biomedical, toxicological, and pharmaceutical research. Immortalized cell lines may overcome these issues, however, few immortalized human keratinocyte cell lines are available and most do not form a fully stratified epithelium. In this study we compared two immortalized keratinocyte cell lines (N/TERT1, N/TERT2G) to human primary keratinocytes based on epidermal differentiation, response to inflammatory mediators, and the development of normal and inflammatory human epidermal equivalents (HEEs). Stratum corneum permeability, epidermal morphology, and expression of epidermal differentiation and host defence genes and proteins in N/TERT-HEE cultures was similar to that of primary human keratinocytes. We successfully generated N/TERT-HEEs with psoriasis or atopic dermatitis features and validated these models for drug-screening purposes. We conclude that the N/TERT keratinocyte cell lines are useful substitutes for primary human keratinocytes thereby providing a biologically relevant, unlimited cell source for in vitro studies on epidermal biology, inflammatory skin disease pathogenesis and therapeutics.
Collapse
Affiliation(s)
- Jos P H Smits
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands
| | - Hanna Niehues
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands
| | - Gijs Rikken
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands
| | - Ivonne M J J van Vlijmen-Willems
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands
| | - Guillaume W H J F van de Zande
- Department of Human Genetics, Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands
| | - Patrick L J M Zeeuwen
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands
| | - Joost Schalkwijk
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands
| | - Ellen H van den Bogaard
- Department of Dermatology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), PO BOX 9101, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
86
|
Klaka P, Grüdl S, Banowski B, Giesen M, Sättler A, Proksch P, Welss T, Förster T. A novel organotypic 3D sweat gland model with physiological functionality. PLoS One 2017; 12:e0182752. [PMID: 28796813 PMCID: PMC5552089 DOI: 10.1371/journal.pone.0182752] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Accepted: 07/23/2017] [Indexed: 11/18/2022] Open
Abstract
Dysregulated human eccrine sweat glands can negatively impact the quality-of-life of people suffering from disorders like hyperhidrosis. Inability of sweating can even result in serious health effects in humans affected by anhidrosis. The underlying mechanisms must be elucidated and a reliable in vitro test system for drug screening must be developed. Here we describe a novel organotypic three-dimensional (3D) sweat gland model made of primary human eccrine sweat gland cells. Initial experiments revealed that eccrine sweat gland cells in a two-dimensional (2D) culture lose typical physiological markers. To resemble the in vivo situation as close as possible, we applied the hanging drop cultivation technology regaining most of the markers when cultured in its natural spherical environment. To compare the organotypic 3D sweat gland model versus human sweat glands in vivo, we compared markers relevant for the eccrine sweat gland using transcriptomic and proteomic analysis. Comparing the marker profile, a high in vitro-in vivo correlation was shown. Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), muscarinic acetylcholine receptor M3 (CHRM3), Na+-K+-Cl- cotransporter 1 (NKCC1), calcium-activated chloride channel anoctamin-1 (ANO1/TMEM16A), and aquaporin-5 (AQP5) are found at significant expression levels in the 3D model. Moreover, cholinergic stimulation with acetylcholine or pilocarpine leads to calcium influx monitored in a calcium flux assay. Cholinergic stimulation cannot be achieved with the sweat gland cell line NCL-SG3 used as a sweat gland model system. Our results show clear benefits of the organotypic 3D sweat gland model versus 2D cultures in terms of the expression of essential eccrine sweat gland key regulators and in the physiological response to stimulation. Taken together, this novel organotypic 3D sweat gland model shows a good in vitro-in vivo correlation and is an appropriate alternative for screening of potential bioactives regulating the sweat mechanism.
Collapse
Affiliation(s)
- Patricia Klaka
- Henkel AG & Co. KGaA, Düsseldorf, Germany
- * E-mail: (PK); (TW)
| | | | | | | | | | - Peter Proksch
- Institute of Pharmaceutical Biology and Biotechnology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Thomas Welss
- Henkel AG & Co. KGaA, Düsseldorf, Germany
- * E-mail: (PK); (TW)
| | | |
Collapse
|
87
|
Pereira RF, Sousa A, Barrias CC, Bayat A, Granja PL, Bártolo PJ. Advances in bioprinted cell-laden hydrogels for skin tissue engineering. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/s40898-017-0003-8] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
88
|
Lombardi B, Casale C, Imparato G, Urciuolo F, Netti PA. Spatiotemporal Evolution of the Wound Repairing Process in a 3D Human Dermis Equivalent. Adv Healthc Mater 2017; 6. [PMID: 28407433 DOI: 10.1002/adhm.201601422] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 03/14/2017] [Indexed: 01/01/2023]
Abstract
Several skin equivalent models have been developed to investigate in vitro the re-epithelialization process occurring during wound healing. Although these models recapitulate closure dynamics of epithelial cells, they fail to capture how a wounded connective tissue rebuilds its 3D architecture until the evolution in a scar. Here, the in vitro tissue repair dynamics of a connective tissue is replicated by using a 3D human dermis equivalent (3D-HDE) model composed of fibroblasts embedded in their own extracellular matrix (ECM). After inducing a physical damage, 3D-HDE undergoes a series of cellular and extracellular events quite similar to those occurring in the native dermis. In particular, fibroblasts differentiation toward myofibroblasts phenotype and neosynthesis of hyaluronic acid, fibronectin, and collagen during the repair process are assessed. Moreover, tissue reorganization after physical damage is investigated by measuring the diameter of bundles and the orientation of fibers of the newly formed ECM network. Finally, the ultimate formation of a scar-like tissue as physiological consequence of the repair and closure process is demonstrated. Taking together, the results highlight that the presence of cell-assembled and responsive stromal components enables quantitative and qualitative in vitro evaluation of the processes involved in scarring during wound healing.
Collapse
Affiliation(s)
- Bernadette Lombardi
- Center for Advanced Biomaterials for HealthCare@CRIB; Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci 53 80125 Naples Italy
- Department of Chemical, Materials and Industrial Production (DICMAPI); University of Naples Federico II; P.leTecchio 80 80125 Naples Italy
| | - Costantino Casale
- Interdisciplinary Research Centre on Biomaterials (CRIB); University of Naples Federico II; P.leTecchio 80 80125 Naples Italy
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB; Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci 53 80125 Naples Italy
| | - Francesco Urciuolo
- Center for Advanced Biomaterials for HealthCare@CRIB; Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci 53 80125 Naples Italy
| | - Paolo Antonio Netti
- Center for Advanced Biomaterials for HealthCare@CRIB; Istituto Italiano di Tecnologia; Largo Barsanti e Matteucci 53 80125 Naples Italy
- Department of Chemical, Materials and Industrial Production (DICMAPI); University of Naples Federico II; P.leTecchio 80 80125 Naples Italy
- Interdisciplinary Research Centre on Biomaterials (CRIB); University of Naples Federico II; P.leTecchio 80 80125 Naples Italy
| |
Collapse
|
89
|
van den Broek LJ, Bergers LIJC, Reijnders CMA, Gibbs S. Progress and Future Prospectives in Skin-on-Chip Development with Emphasis on the use of Different Cell Types and Technical Challenges. Stem Cell Rev Rep 2017; 13:418-429. [PMID: 28536890 PMCID: PMC5486511 DOI: 10.1007/s12015-017-9737-1] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Understanding the healthy and diseased state of skin is important in many areas of basic and applied research. Although the field of skin tissue engineering has advanced greatly over the last years, current in vitro skin models still do not mimic the complexity of the human skin. Skin-on-chip and induced pluripotent stem cells (iPSC) might be key technologies to improve in vitro skin models. This review summarizes the state of the art of in vitro skin models with regard to cell sources (primary, cell line, iPSC) and microfluidic devices. It can be concluded that iPSC have the potential to be differentiated into many kinds of immunologically matched cells and skin-on-chip technology might lead to more physiologically relevant skin models due to the controlled environment, possible exchange of immune cells, and an increased barrier function. Therefore the combination of iPSC and skin-on-chip is expected to lead to superior healthy and diseased in vitro skin models.
Collapse
Affiliation(s)
| | | | | | - Susan Gibbs
- Department of Dermatology, VU University Medical Center, Amsterdam, The Netherlands.
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam, University of Amsterdam and VU University, Amsterdam, The Netherlands.
| |
Collapse
|
90
|
Jennings LR, Colley HE, Ong J, Panagakos F, Masters JG, Trivedi HM, Murdoch C, Whawell S. Development and Characterization of In Vitro Human Oral Mucosal Equivalents Derived from Immortalized Oral Keratinocytes. Tissue Eng Part C Methods 2016; 22:1108-1117. [PMID: 27846777 DOI: 10.1089/ten.tec.2016.0310] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Tissue-engineered oral mucosal equivalents (OME) are being increasingly used to measure toxicity, drug delivery, and to model oral diseases. Current OME mainly comprise normal oral keratinocytes (NOK) cultured on top of a normal oral fibroblasts-containing matrix. However, the commercial supply of NOK is limited, restricting widespread use of these mucosal models. In addition, NOK suffer from poor longevity and donor-to-donor variability. Therefore, we constructed, characterized, and tested the functionality of OME based on commercial TERT2-immortalized oral keratinocytes (FNB6) to produce a more readily available alternative to NOK-based OME. FNB6 OME cultured at an air-to-liquid interface for 14 days exhibited expression of differentiation markers cytokeratin 13 in the suprabasal layers and cytokeratin 14 in basal layer of the epithelium. Proliferating cells were restricted to the basal epithelium, and there was immuno-positive expression of E-cadherin confirming the presence of established cell-to-cell contacts. The histology and expression of these structural markers paralleled those observed in the normal oral mucosa and NOK-based models. On stimulation with TNFα and IL-1, FNB6 OME displayed a similar global gene expression profile to NOK-based OME, with increased expression of many common pro-inflammatory molecules such as chemokines (CXCL8), cytokines (IL-6), and adhesion molecules (ICAM-1) when analyzed by gene array and quantitative PCR. Similarly, pathway analysis showed that both FNB6 and NOK models initiated similar intracellular signaling on stimulation. Gene expression in FNB6 OME was more consistent than NOK-based OME that suffered from donor variation in response to stimuli. Mucosal equivalents based on immortalized FNB6 cells are accessible, reproducible and will provide an alternative animal experimental system for studying mucosal drug delivery systems, host-pathogen interactions, and drug-induced toxicity.
Collapse
Affiliation(s)
- Luke R Jennings
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| | - Helen E Colley
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| | - Jane Ong
- 2 Colgate-Palmolive Company , Piscataway, New Jersey
| | | | | | | | - Craig Murdoch
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| | - Simon Whawell
- 1 School of Clinical Dentistry, University of Sheffield , Sheffield, United Kingdom
| |
Collapse
|
91
|
Skin-on-a-chip model simulating inflammation, edema and drug-based treatment. Sci Rep 2016; 6:37471. [PMID: 27869150 PMCID: PMC5116589 DOI: 10.1038/srep37471] [Citation(s) in RCA: 212] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 10/26/2016] [Indexed: 12/25/2022] Open
Abstract
Recent advances in microfluidic cell cultures enable the construction of in vitro human skin models that can be used for drug toxicity testing, disease study. However, current in vitro skin model have limitations to emulate real human skin due to the simplicity of model. In this paper, we describe the development of ‘skin-on-a-chip’ to mimic the structures and functional responses of the human skin. The proposed model consists of 3 layers, on which epidermal, dermal and endothelial components originated from human, were cultured. The microfluidic device was designed for co-culture of human skin cells and each layer was separated by using porous membranes to allow interlayer communication. Skin inflammation and edema were induced by applying tumor necrosis factor alpha on dermal layer to demonstrate the functionality of the system. The expression levels of proinflammatory cytokines were analyzed to illustrate the feasibility. In addition, we evaluated the efficacy of therapeutic drug testing model using our skin chip. The function of skin barrier was evaluated by staining tight junctions and measuring a permeability of endothelium. Our results suggest that the skin-on-a-chip model can potentially be used for constructing in vitro skin disease models or for testing the toxicity of cosmetics or drugs.
Collapse
|
92
|
Planz V, Lehr CM, Windbergs M. In vitro models for evaluating safety and efficacy of novel technologies for skin drug delivery. J Control Release 2016; 242:89-104. [PMID: 27612408 DOI: 10.1016/j.jconrel.2016.09.002] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 08/22/2016] [Accepted: 09/05/2016] [Indexed: 12/14/2022]
Abstract
For preclinical testing of novel therapeutics, predictive in vitro models of the human skin are required to assess efficacy, absorption and safety. Simple as well as more sophisticated three-dimensional organotypic models of the human skin emerged as versatile and powerful tools simulating healthy as well as diseased skin states. Besides addressing the demands of research and industry, such models serve as valid alternative to animal testing. Recently, the acceptance of several models by regulatory authorities corroborates their role as important building block for preclinical development. However, valid assessment of readout parameters derived from these models requires suitable analytical techniques. Standard analytical methods are mostly destructive and limited regarding in-depth investigation on molecular level. The combination of adequate in vitro models with modern non-invasive analytical modalities bears a great potential to address important skin drug delivery related questions. Topics of interest are for instance the assessment of repeated dosing effects and xenobiotic biotransformation, which cannot be analyzed by destructive techniques. This review provides a comprehensive overview of current in vitro skin models differing in functional complexity and mimicking healthy as well as diseased skin states. Further, benefits and limitations regarding analytical evaluation of efficacy, absorption and safety of novel drug carrier systems applied to such models are discussed along with a prospective view of anticipated future directions. In addition, emerging non-invasive imaging modalities are introduced and their significance and potential to advance current knowledge in the field of skin drug delivery is explored.
Collapse
Affiliation(s)
- Viktoria Planz
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; PharmBioTec GmbH, 66123 Saarbrücken, Germany
| | - Claus-Michael Lehr
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; PharmBioTec GmbH, 66123 Saarbrücken, Germany
| | - Maike Windbergs
- Helmholtz Centre for Infection Research (HZI), Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Department of Drug Delivery (DDEL), 66123 Saarbrücken, Germany; Department of Pharmacy, Saarland University, 66123 Saarbrücken, Germany; PharmBioTec GmbH, 66123 Saarbrücken, Germany.
| |
Collapse
|
93
|
Surrao DC, Boon K, Borys B, Sinha S, Kumar R, Biernaskie J, Kallos MS. Large-scale expansion of human skin-derived precursor cells (hSKPs) in stirred suspension bioreactors. Biotechnol Bioeng 2016; 113:2725-2738. [PMID: 27345530 DOI: 10.1002/bit.26040] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Revised: 06/06/2016] [Accepted: 06/20/2016] [Indexed: 01/07/2023]
Abstract
Human skin-derived precursor cells (hSKPs) are multipotent adult stem cells found in the dermis of human skin. Incorporation of hSKPs into split-thickness skin grafts (STSGs), the current gold standard to treat severe burns or tissue resections, has been proposed as a treatment option to enhance skin wound healing and tissue function. For this approach to be clinically viable substantial quantities of hSKPs are required, which is the rate-limiting step, as only a few thousand hSKPs can be isolated from an autologous skin biopsy without causing donor site morbidity. In order to produce sufficient quantities of clinically viable cells, we have developed a bioprocess capable of expanding hSKPs as aggregates in stirred suspension bioreactors (SSBs). In this study, we found hSKPs from adult donors to expand significantly more (P < 0.05) at 60 rpm in SSBs than in static cultures. Furthermore, the utility of the SSBs, at 60 rpm is demonstrated by serial passaging of hSKPs from a small starting population, which can be isolated from an autologous skin biopsy without causing donor site morbidity. At 60 rpm, aggregates were markedly smaller and did not experience oxygen diffusional limitations, as seen in hSKPs cultured at 40 rpm. While hSKPs also grew at 80 rpm (0.74 Pa) and 100 rpm (1 Pa), they produced smaller aggregates due to high shear stress. The pH of the media in all the SSBs was closer to biological conditions and significantly different (P < 0.05) from static cultures, which recorded acidic pH conditions. The nutrient concentrations of the media in all the SSBs and static cultures did not drop below acceptable limits. Furthermore, there was no significant build-up of waste products to limit hSKP expansion in the SSBs. In addition, hSKP markers were maintained in the 60 rpm SSB as demonstrated by immunocytochemistry. This method of growing hSKPs in a batch culture at 60 rpm in a SSB represents an important first step in developing an automated bioprocess to produce substantial numbers of clinically viable hSKPs aimed at regenerating the dermis to improve healing of severe skin wounds. Biotechnol. Bioeng. 2016;113: 2725-2738. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Denver C Surrao
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Kathryn Boon
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada
| | - Breanna Borys
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada.,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada
| | - Sarthak Sinha
- Faculty of Veterinary Medicine, Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ranjan Kumar
- Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, Department of Comparative Biology and Experimental Medicine, University of Calgary, Calgary, Alberta, Canada.,Hotchkiss Brain Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada.,Alberta Children's Hospital Research Institute, University of Calgary, Calgary, Alberta, Canada
| | - Michael S Kallos
- Pharmaceutical Production Research Facility (PPRF), Schulich School of Engineering, University of Calgary, Calgary, Alberta, Canada. .,Department of Chemical and Petroleum Engineering, Schulich School of Engineering, University of Calgary, 2500 University Drive NW, Calgary, Alberta, T2N 1N4, Canada. .,Biomedical Engineering Graduate Program, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
94
|
Methods to study differences in cell mobility during skin wound healing in vitro. J Biomech 2016; 49:1381-1387. [DOI: 10.1016/j.jbiomech.2016.01.040] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Revised: 01/19/2016] [Accepted: 01/28/2016] [Indexed: 01/19/2023]
|
95
|
Modeling Barrier Tissues In Vitro: Methods, Achievements, and Challenges. EBioMedicine 2016; 5:30-9. [PMID: 27077109 PMCID: PMC4816829 DOI: 10.1016/j.ebiom.2016.02.023] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 02/09/2016] [Accepted: 02/11/2016] [Indexed: 12/24/2022] Open
Abstract
Organ-on-a-chip devices have gained attention in the field of in vitro modeling due to their superior ability in recapitulating tissue environments compared to traditional multiwell methods. These constructed growth environments support tissue differentiation and mimic tissue-tissue, tissue-liquid, and tissue-air interfaces in a variety of conditions. By closely simulating the in vivo biochemical and biomechanical environment, it is possible to study human physiology in an organ-specific context and create more accurate models of healthy and diseased tissues, allowing for observations in disease progression and treatment. These chip devices have the ability to help direct, and perhaps in the distant future even replace animal-based drug efficacy and toxicity studies, which have questionable relevance to human physiology. Here, we review recent developments in the in vitro modeling of barrier tissue interfaces with a focus on the use of novel and complex microfluidic device platforms.
Collapse
|