1
|
Cheng LC, You S, Ren T, Qiu J, Hua K. Development and characterization of a novel immortalized human vaginal fibroblast cell line for advanced applications in reproductive health. Reprod Biol Endocrinol 2025; 23:56. [PMID: 40211367 PMCID: PMC11984242 DOI: 10.1186/s12958-025-01393-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 04/02/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Reproductive health issues related to the vagina, face significant challenges due to the lack of standardized research models. Vaginal fibroblasts, which constitute approximately 55% of the vaginal wall's cellular composition, are crucial for tissue repair, remodelling, and reproductive health. These fibroblasts have broad applications in regenerative medicine and gynaecological treatments. Despite their importance, current research relies primarily on epithelial cells or primary vaginal fibroblasts, but primary fibroblasts are limited by their short lifespan, donor-to-donor variability, and susceptibility to senescence. Immortalized fibroblast lines offer a solution by extending the lifespan and enabling reproducible studies. However, a well-characterized immortalized human vaginal fibroblast line has not been established, highlighting the need for novel models to better understand and address vaginal-associated conditions. METHODS Primary human vaginal fibroblasts were immortalized via the lentiviral transfection of human telomerase reverse transcriptase. The resulting cell line was characterized through histological, immunofluorescent, RT-qPCR and flow cytometry analyses. Proliferation, senescence, gene expression, hormone responsiveness and genomic stability were assessed via quantitative polymerase chain reaction, transcriptome sequencing, gene set enrichment analysis, short tandem repeat profiling, and karyotype analysis. RESULTS The immortalized human vaginal fibroblasts (ihVFs) retained typical spindle-shaped fibroblast morphology and fibroblast-specific marker expression. Compared with primary vaginal fibroblasts, ihVF exhibited significantly reduced senescence, maintained sustained growth through extended culture passages, and preserved genetic stability. Transcriptome sequencing revealed high gene expression similarity between immortalized and primary fibroblasts, with no significant alterations in oncogenic pathways. PCR and immunofluorescent analyses revealed that ihVFs are responsive to estrogen and progesterone stimulation. Short tandem repeat analysis confirmed the novelty of the immortalized cell line, with no overlap with existing cell databases. CONCLUSIONS The novel ihVF cell line retains key phenotypic, functional, and genetic characteristics of primary vaginal fibroblasts, providing a stable, reproducible, and physiologically relevant model for reproductive health research. This cell line addresses the limitations of primary fibroblasts and has broad applications in tissue engineering, gynaecological disorder research, and drug screening, advancing our understanding of vaginal fibroblast biology and therapeutic interventions.
Collapse
Affiliation(s)
- Leong Chi Cheng
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, 200011, China
| | - Shuoming You
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, 200011, China
| | - Tingting Ren
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, 200011, China
| | - Junjun Qiu
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, 200011, China.
| | - Keqin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 419 Fangxie Road, Shanghai, 200011, China.
- Shanghai Key Laboratory of Female Reproductive Endocrine-Related Diseases, Fudan University, Shanghai, 200011, China.
| |
Collapse
|
2
|
Bi O, Caballero‐Lima D, Sikkink S, Westgate G, Kauser S, Elies J, Thornton MJ. Do Melanocytes Have a Role in Controlling Epidermal Bacterial Colonisation and the Skin Microbiome? Exp Dermatol 2025; 34:e70071. [PMID: 40051134 PMCID: PMC11885897 DOI: 10.1111/exd.70071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 02/09/2025] [Accepted: 02/17/2025] [Indexed: 03/10/2025]
Abstract
In addition to producing melanin to protect epidermal keratinocytes against DNA damage, melanocytes may have important roles in strengthening innate immunity against pathogens. We have developed a functional, pigmented, human full-thickness 3D skin equivalent to determine whether the presence of melanocytes impacts epidermal bacterial growth and regulates the expression of genes involved in the immune response. We introduced primary epidermal melanocytes to construct a 3-cell full-thickness skin equivalent with primary dermal fibroblasts and epidermal keratinocytes. Immunohistochemistry verified the appropriate ratio and spatial organisation of melanocytes. Alpha-MSH induced melanogenesis, confirming an appropriate physiological response. We compared this 3-cell skin equivalent with the 2-cell version without melanocytes in response to inoculation with 3 species of bacteria: Staphylococcus epidermidis, Corynebacterium striatum, and Cutibacterium acnes. There was a significant decrease in the colonisation of bacteria in the skin equivalents containing functional melanocytes. There was increased expression of immune-response genes (S100A9, DEFB4A, IL-4R) following microorganism exposure; however, there were marked differences between the unpigmented and pigmented skin equivalents. This physiologically relevant human 3D-skin equivalent opens up new avenues for studying complex skin pigmentation disorders, melanoma, and UV damage, as well as the rapidly evolving field of the skin microbiome and the balance between commensal and pathogenic species.
Collapse
Affiliation(s)
- Omera Bi
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
- Labskin UK, York Biotech CampusSand HuttonUK
| | | | - Stephen Sikkink
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| | - Gill Westgate
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| | - Sobia Kauser
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| | - Jacobo Elies
- Faculty of Life SciencesUniversity of BradfordBradfordUK
| | - M. Julie Thornton
- Centre for Skin Sciences, Faculty of Life ScienceUniversity of BradfordBradfordUK
| |
Collapse
|
3
|
Hughes BK, Davis A, Milligan D, Wallis R, Mossa F, Philpott MP, Wainwright LJ, Gunn DA, Bishop CL. SenPred: a single-cell RNA sequencing-based machine learning pipeline to classify deeply senescent dermal fibroblast cells for the detection of an in vivo senescent cell burden. Genome Med 2025; 17:2. [PMID: 39810225 PMCID: PMC11731430 DOI: 10.1186/s13073-024-01418-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Senescence classification is an acknowledged challenge within the field, as markers are cell-type and context dependent. Currently, multiple morphological and immunofluorescence markers are required. However, emerging scRNA-seq datasets have enabled an increased understanding of senescent cell heterogeneity. METHODS Here we present SenPred, a machine-learning pipeline which identifies fibroblast senescence based on single-cell transcriptomics from fibroblasts grown in 2D and 3D. RESULTS Using scRNA-seq of both 2D and 3D deeply senescent fibroblasts, the model predicts intra-experimental fibroblast senescence to a high degree of accuracy (> 99% true positives). Applying SenPred to in vivo whole skin scRNA-seq datasets reveals that cells grown in 2D cannot accurately detect fibroblast senescence in vivo. Importantly, utilising scRNA-seq from 3D deeply senescent fibroblasts refines our ML model leading to improved detection of senescent cells in vivo. This is context specific, with the SenPred pipeline proving effective when detecting senescent human dermal fibroblasts in vivo, but not the senescence of lung fibroblasts or whole skin. CONCLUSIONS We position this as a proof-of-concept study based on currently available scRNA-seq datasets, with the intention to build a holistic model to detect multiple senescent triggers using future emerging datasets. The development of SenPred has allowed for the detection of an in vivo senescent fibroblast burden in human skin, which could have broader implications for the treatment of age-related morbidities. All code for the SenPred pipeline is available at the following URL: https://github.com/bethk-h/SenPred_HDF .
Collapse
Affiliation(s)
- Bethany K Hughes
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Andrew Davis
- Unilever R&D, Merritt Blvd, Trumbull, CT, 06611, USA
| | - Deborah Milligan
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Ryan Wallis
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Federica Mossa
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Michael P Philpott
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK
| | - Linda J Wainwright
- Unilever R&D, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - David A Gunn
- Unilever R&D, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Cleo L Bishop
- Blizard Institute, Barts and The London Faculty of Medicine and Dentistry, Queen Mary University of London, London, E1 2AT, UK.
| |
Collapse
|
4
|
De Los Santos Gomez P, Goncalves K, Maltman V, Smith L, Przyborski S. Engineering a Pigmented Skin Equivalent That Is Responsive to External Stimuli. Methods Mol Biol 2025; 2922:115-134. [PMID: 40208531 DOI: 10.1007/978-1-0716-4510-9_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Abstract
Recently, dermatological research has more widely adopted the use of human skin equivalents (HSEs) to better recreate skin structure and function in vitro, which can act as a preclinical tool. However, many popular HSEs contain only two main cell types: keratinocytes and fibroblasts to model epidermal and dermal compartments respectively. This lack of supporting cell types can be associated with a wide range of limitations, most notably an inaccurate representation of skin's innate response to exogenous stressors such as UV radiation. We have adapted our novel full-thickness skin platform to incorporate human melanocytes that produce melanin and transfer melanin to neighboring keratinocytes, where it is located apically to the nucleus, forming typical supranuclear protective caps. The use of Alvetex® scaffold and the ability of dermal fibroblasts to secrete their own endogenous extracellular matrix (ECM) proteins provide a robust dermal foundation for the construction of a pigmented epidermis and are essential for supranuclear cap formation, a physiological phenomenon essential to melanin's protective function. This model system is responsive both to up and downregulation of melanogenesis, therefore providing an in vitro platform for a wide array of applications, ranging from industrial active testing for cosmetic formulations to academic insights into the cellular response to environmental stressors.
Collapse
Affiliation(s)
| | | | | | - Lucy Smith
- Department of Biosciences, Durham University, Durham, UK
| | - Stefan Przyborski
- Department of Biosciences, Durham University, Durham, UK.
- Reprocell Europe Ltd, Glasgow, UK.
| |
Collapse
|
5
|
Daugaard ND, Tholstrup R, Tornby JR, Bendixen SM, Larsen FT, De Zio D, Barnkob MB, Ravnskjaer K, Brewer JR. Characterization of human melanoma skin cancer models: A step towards model-based melanoma research. Acta Biomater 2025; 191:308-324. [PMID: 39549863 DOI: 10.1016/j.actbio.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/27/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Advancing 3D in vitro human tissue models is crucial for biomedical research and drug development to address the ethical and biological limitations of animal testing. Recently, 3D skin models have proven to be effective for studying serious skin conditions, such as melanoma. For these advanced models to be applicable in preclinical studies, thorough characterization is essential to understand their applicability and limitations. In this study, we used bioimaging and RNA sequencing to assess the architecture and transcriptomic profiles of skin models, including models with melanoma. Our results indicated that these models closely mimicked skin morphology and gene expression patterns. The full-thickness (FT) model shows a superior resemblance to the human skin, particularly in basement membrane formation and cellular interactions. The integrity of the skin-like properties and gene expression signatures of both skin and melanoma cells were preserved upon the integration of melanoma cells, establishing these models as robust platforms for cancer research. The responsiveness of the FT melanoma models to vemurafenib treatment was successfully monitored, demonstrating their validity as a reliable, reproducible, and humane tool for pharmacological testing and drug development. Furthermore, the transcriptomic data showed that skin models with cancer spheroids had upregulated genes linked to aggressive and resilient cancer behavior compared to spheroids alone. This emphasizes the importance of the microenvironment in cancer progression and suggests that 3D skin models can serve to uncover mechanisms and therapeutic targets that are not detectable in simpler systems. STATEMENT OF SIGNIFICANCE: This study introduces advanced, ethically sound skin and melanoma models as alternatives to animal testing in drug discovery. By thoroughly characterizing these models using bioimaging and RNA sequencing, we demonstrate their close resemblance to human skin, particularly in full-thickness models. These models not only replicate the complex cellular interactions and gene expression patterns of human tissue but also maintain robustness after melanoma integration. Our findings highlight the potential of these models in revealing cancer mechanisms and therapeutic targets, offering a significant impact on melanoma research and preclinical testing.
Collapse
Affiliation(s)
- Nicoline Dorothea Daugaard
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Rikke Tholstrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Jakob Rask Tornby
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Sofie Marchsteiner Bendixen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Frederik Tibert Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Daniela De Zio
- Melanoma Research Team, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Institute, Copenhagen, Denmark; Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Mike Bogetofte Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Department of Clinical Immunology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Kim Ravnskjaer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Jonathan R Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| |
Collapse
|
6
|
Jia YY, Atwood SX. Diversity of human skin three-dimensional organotypic cultures. Curr Opin Genet Dev 2024; 89:102275. [PMID: 39536613 DOI: 10.1016/j.gde.2024.102275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/17/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Recently, significant strides have been made in the development of high-fidelity skin organoids, encompassing techniques such as 3D bioprinting, skin-on-a-chip systems, and models derived from pluripotent stem cells (PSCs), replicating appendage structures and diverse skin cell types. Despite the emergence of these state-of-the-art skin engineering models, human organotypic cultures (OTCs), initially proposed in the 1970s, continue to reign as the predominant in vitro cultured three-dimensional skin model in the field of tissue engineering. This enduring prevalence is owed to their cost-effectiveness, straight forward setup, time efficiency, and faithful representation of native human skin. In this review, we systematically delineate recent advances in skin OTC models, aiming to inform future efforts to enhance in vitro skin model fidelity and reproducibility.
Collapse
Affiliation(s)
- Yunlong Y Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Dermatology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
7
|
Michielon E, Boninsegna M, Waaijman T, Fassini D, Spiekstra SW, Cramer J, Gaudriault P, Kodolányi J, de Gruijl TD, Homs-Corbera A, Gibbs S. Environmentally Controlled Microfluidic System Enabling Immune Cell Flow and Activation in an Endothelialised Skin-On-Chip. Adv Healthc Mater 2024; 13:e2400750. [PMID: 39370595 PMCID: PMC11582514 DOI: 10.1002/adhm.202400750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 09/17/2024] [Indexed: 10/08/2024]
Abstract
Integration of reconstructed human skin (RhS) into organ-on-chip (OoC) platforms addresses current limitations imposed by static culturing. This innovation, however, is not without challenges. Microfluidic devices, while powerful, often encounter usability, robustness, and gas bubble issues that hinder large-scale high-throughput setups. This study aims to develop a novel re-usable multi-well microfluidic adaptor (MMA) with the objective to provide a flexible tool for biologists implementing complex 3D biological models (e.g., skin) while enabling simultaneous user control over temperature, medium flow, oxigen (O2), nitrogen (N2), and carbon dioxide (CO2) without the need for an incubator. The presented MMA device is designed to be compatible with standard, commercially available 6-well multi-well plates (6MWPs) and 12-well transwells. This MMA-6MWP setup is employed to generate a skin-on-chip (SoC). RhS viability is maintained under flow for three days and their morphology closely resembles that of native human skin. A proof-of-concept study demonstrates the system's potential in toxicology applications by combining endothelialised RhS with flowing immune cells. This dynamic setting activates the monocyte-like MUTZ-3 cells (CD83 and CD86 upregulation) upon topical exposure of RhS to a sensitizer, revealing the MMA-6MWP's unique capabilities compared to static culturing, where such activation is absent.
Collapse
Affiliation(s)
- Elisabetta Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
| | - Matteo Boninsegna
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
- Department of Physics, Bielefeld University, Universitätsstr 25, 33615, Bielefeld, Germany
| | - Taco Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Dario Fassini
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Sander W Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
| | - Jeremy Cramer
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Pierre Gaudriault
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - János Kodolányi
- Department of Dental Material Science, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| | - Tanja D de Gruijl
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam UMC, Vrije Universiteit, Amsterdam, 1081 HV, The Netherlands
- Department of Medical Oncology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, 1081 HV, The Netherlands
| | - Antoni Homs-Corbera
- Cherry Biotech SAS, 14 Rue De La Beaune, Bâtiment A, 2ème Étage, Montreuil, 93100, France
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, De Boelelaan 1117, Amsterdam, 1081 HV, The Netherlands
- Amsterdam institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, 1081 LA, The Netherlands
| |
Collapse
|
8
|
Khalid A, Dremin V, El-Tamer A, Surnina M, Lancelot C, Rafailov E, Sokolovski S. Dual-mode OCT/fluorescence system for monitoring the morphology and metabolism of laser-printed 3D full-thickness skin equivalents. BIOMEDICAL OPTICS EXPRESS 2024; 15:6299-6312. [PMID: 39553855 PMCID: PMC11563319 DOI: 10.1364/boe.510610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 11/19/2024]
Abstract
The 3D structure of native human skin is fundamental for studying skin health, diseases, wound healing, and for testing the safety of skin care products, as well as personalized treatments for skin conditions. Tissue regeneration, driven by tissue engineering, often involves creating full-thickness skin equivalents (FSE), which are widely used for developing both healthy and diseased skin models. In this study, we utilized human skin cell lines to create FSE. We designed high-resolution 3D scaffolds to support the growth and maturation of these skin models. Additionally, we developed and validated a cost-effective, custom-built system combining fluorescence spectroscopy (FS) and optical coherence tomography (OCT) for non-destructive analysis of the metabolism and morphology of 3D FSEs. This system proved highly sensitive in detecting fluorescence from key metabolic co-enzymes (NADH and FAD) in solutions and cell suspensions, while OCT provided adequate resolution to observe the morphology of FSEs. As a result, both the 3D FSE model and the dual-mode optical system hold significant potential for use in 3D bioprinting of biological tissues, as well as in the development of cosmetics, drugs, and in monitoring their maturation over time.
Collapse
Affiliation(s)
- Arooj Khalid
- AIPT, College of Engineering and Physical Sciences, Aston University, B4 7 PH Birmingham, UK
| | - Viktor Dremin
- AIPT, College of Engineering and Physical Sciences, Aston University, B4 7 PH Birmingham, UK
| | | | | | - Celine Lancelot
- StratiCELL Ltd., Science Park Crealys, 5032 Les Isnes, Gembloux, Belgium
| | - Edik Rafailov
- AIPT, College of Engineering and Physical Sciences, Aston University, B4 7 PH Birmingham, UK
| | - Sergei Sokolovski
- AIPT, College of Engineering and Physical Sciences, Aston University, B4 7 PH Birmingham, UK
| |
Collapse
|
9
|
Elkordy AA, Hill D, Attia M, Chaw CS. Liposomes and Their Therapeutic Applications in Enhancing Psoriasis and Breast Cancer Treatments. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1760. [PMID: 39513840 PMCID: PMC11547384 DOI: 10.3390/nano14211760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Psoriasis and breast cancer are two examples of diseases where associated inflammatory pathways within the body's immune system are implicated. Psoriasis is a complex, chronic and incurable inflammatory skin disorder that is primarily recognized by thick, scaly plaques on the skin. The most noticeable pathophysiological effect of psoriasis is the abnormal proliferation of keratinocytes. Breast cancer is currently the most diagnosed cancer and the leading cause of cancer-related death among women globally. While treatments targeting the primary tumor have significantly improved, preventing metastasis with systemic treatments is less effective. Nanocarriers such as liposomes and lipid nanoparticles have emerged as promising drug delivery systems for drug targeting and specificity. Advances in technologies and drug combinations have emerged to develop more efficient lipid nanocarriers to include more than one drug in combinational therapy to enhance treatment outcomes and/or relief symptoms for better patients' quality of life. Although there are FDA-approved liposomes with anti-cancer drugs for breast cancer, there are still unmet clinical needs to reduce the side effects associated with those nanomedicines. Hence, combinational nano-therapy may eliminate some of the issues and challenges. Furthermore, there are no nanomedicines yet clinically available for psoriasis. Hence, this review will focus on liposomes encapsulated single and/or combinational therapy to augment treatment outcomes with an emphasis on the effectiveness of combinational therapy within liposomal-based nanoparticulate drug delivery systems to tackle psoriasis and breast cancer. This review will also include an overview of both diseases, challenges in delivering drug therapy and the roles of nanomedicines as well as psoriasis and breast cancer models used for testing therapeutic interventions to pave the way for effective in vivo testing prior to the clinical trials.
Collapse
Affiliation(s)
- Amal Ali Elkordy
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - David Hill
- School of Nursing and Health Sciences, Faculty of Health Sciences and Wellbeing, University of Sunderland, Sunderland SR1 3SD, UK;
| | - Mohamed Attia
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| | - Cheng Shu Chaw
- School of Pharmacy and Pharmaceutical Sciences, University of Sunderland, Sunderland SR1 3SD, UK; (A.A.E.); (M.A.)
| |
Collapse
|
10
|
Riabinin A, Pankratova M, Rogovaya O, Vorotelyak E, Terskikh V, Vasiliev A. Ideal Living Skin Equivalents, From Old Technologies and Models to Advanced Ones: The Prospects for an Integrated Approach. BIOMED RESEARCH INTERNATIONAL 2024; 2024:9947692. [PMID: 39184355 PMCID: PMC11343635 DOI: 10.1155/2024/9947692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 04/18/2024] [Accepted: 07/20/2024] [Indexed: 08/27/2024]
Abstract
The development of technologies for the generation and transplantation of living skin equivalents (LSEs) is a significant area of translational medicine. Such functional equivalents can be used to model and study the morphogenesis of the skin and its derivatives, to test drugs, and to improve the healing of chronic wounds, burns, and other skin injuries. The evolution of LSEs over the past 50 years has demonstrated the leap in technology and quality and the shift from classical full-thickness LSEs to principled new models, including modification of classical models and skin organoids with skin derived from human-induced pluripotent stem cells (iPSCs) (hiPSCs). Modern methods and approaches make it possible to create LSEs that successfully mimic native skin, including derivatives such as hair follicles (HFs), sebaceous and sweat glands, blood vessels, melanocytes, and nerve cells. New technologies such as 3D and 4D bioprinting, microfluidic systems, and genetic modification enable achievement of new goals, cost reductions, and the scaled-up production of LSEs.
Collapse
Affiliation(s)
- Andrei Riabinin
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Maria Pankratova
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Olga Rogovaya
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina Vorotelyak
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Vasiliy Terskikh
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| | - Andrey Vasiliev
- Department of Cell BiologyKoltzov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
11
|
Aranha ESP, Mendonça LDS, Almeida BDL, da Silva EL, Mesquita FP, Lima ES, Alves APNN, de Moraes MEA, Montenegro RC, de Vasconcellos MC. Three dimensional reconstruction of skin with melanoma: A model for study of invasion in vitro. Toxicol In Vitro 2024; 99:105883. [PMID: 38936442 DOI: 10.1016/j.tiv.2024.105883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 06/09/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Melanoma is a type of tumor skin with high metastatic potential. Reconstructed human skin, development for pre-clinic assay, are make using primary human cells, but with same limitations. The aim this study was to characterize a cell culture model, with structure similar to human skin containing melanoma cells entirely from cell lines. Reconstructed skin with melanoma were development using human fibroblasts (MRC5), human epidermal keratinocytes (HaCat), and human melanoma (SK-MEL-28) embedded in collagen type I. The structure was characterized by hematoxylin-eosin stained, as well as points of melanoma cell invasion, which was associated with activity of MMPs (MMP-2 and MMP-9) by zymographic method. Then, the gene expression of the target molecular mechanisms involved in melanoma progression were evaluated. Here, the model development showed a region epidermis organized and separated from the dermis, with fibroblast cells confined and melanoma cells form delimited area invasion. MMP-2 and MMP-9 were identified during of cell culture and gene expression of BRAF, NRAS, and Vimentin was confirmed. The proposed model provides one more opportunity to study in vitro tumor biology of melanoma and also to allows the study of new drugs with more reliable results then whats we would find in vivo.
Collapse
Affiliation(s)
- Elenn Suzany Pereira Aranha
- Faculty of Pharmaceutical Sciences, Post Graduate Program in Biodiversity and Biotechnology of the Amazon (Bionorte), Federal University of Amazonas, Manaus, Amazonas, Brazil
| | | | - Bianca de Lima Almeida
- Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | - Emerson Lucena da Silva
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Felipe Pantoja Mesquita
- Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, Ceará, Brazil
| | - Emersom Silva Lima
- Faculty of Pharmaceutical Sciences, Federal University of Amazonas, Manaus, Amazonas, Brazil
| | | | | | | | | |
Collapse
|
12
|
Ismayilzada N, Tarar C, Dabbagh SR, Tokyay BK, Dilmani SA, Sokullu E, Abaci HE, Tasoglu S. Skin-on-a-chip technologies towards clinical translation and commercialization. Biofabrication 2024; 16:042001. [PMID: 38964314 DOI: 10.1088/1758-5090/ad5f55] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 07/04/2024] [Indexed: 07/06/2024]
Abstract
Skin is the largest organ of the human body which plays a critical role in thermoregulation, metabolism (e.g. synthesis of vitamin D), and protection of other organs from environmental threats, such as infections, microorganisms, ultraviolet radiation, and physical damage. Even though skin diseases are considered to be less fatal, the ubiquity of skin diseases and irritation caused by them highlights the importance of skin studies. Furthermore, skin is a promising means for transdermal drug delivery, which requires a thorough understanding of human skin structure. Current animal andin vitrotwo/three-dimensional skin models provide a platform for disease studies and drug testing, whereas they face challenges in the complete recapitulation of the dynamic and complex structure of actual skin tissue. One of the most effective methods for testing pharmaceuticals and modeling skin diseases are skin-on-a-chip (SoC) platforms. SoC technologies provide a non-invasive approach for examining 3D skin layers and artificially creating disease models in order to develop diagnostic or therapeutic methods. In addition, SoC models enable dynamic perfusion of culture medium with nutrients and facilitate the continuous removal of cellular waste to further mimic thein vivocondition. Here, the article reviews the most recent advances in the design and applications of SoC platforms for disease modeling as well as the analysis of drugs and cosmetics. By examining the contributions of different patents to the physiological relevance of skin models, the review underscores the significant shift towards more ethical and efficient alternatives to animal testing. Furthermore, it explores the market dynamics ofin vitroskin models and organ-on-a-chip platforms, discussing the impact of legislative changes and market demand on the development and adoption of these advanced research tools. This article also identifies the existing obstacles that hinder the advancement of SoC platforms, proposing directions for future improvements, particularly focusing on the journey towards clinical adoption.
Collapse
Affiliation(s)
- Nilufar Ismayilzada
- Department of Mechanical Engineering, Koç University, Istanbul 34450, Turkey
| | - Ceren Tarar
- Department of Mechanical Engineering, Koç University, Istanbul 34450, Turkey
| | | | - Begüm Kübra Tokyay
- Koç University Research Center for Translational Medicine, Koç University, Istanbul 34450, Turkey
| | - Sara Asghari Dilmani
- Koç University Research Center for Translational Medicine, Koç University, Istanbul 34450, Turkey
| | - Emel Sokullu
- School of Medicine, Koç University, Istanbul 34450, Turkey
| | - Hasan Erbil Abaci
- Department of Dermatology, Columbia University, New York City, NY, United States of America
| | - Savas Tasoglu
- Department of Mechanical Engineering, Koç University, Istanbul 34450, Turkey
- Boğaziçi Institute of Biomedical Engineering, Boğaziçi University, Istanbul 34684, Turkey
- Koç University Research Center for Translational Medicine, Koç University, Istanbul 34450, Turkey
- Koç University Arçelik Research Center for Creative Industries (KUAR), Koç University, Istanbul 34450, Turkey
| |
Collapse
|
13
|
Zuniga K, Ghousifam N, Shaffer L, Brocklehurst S, Van Dyke M, Christy R, Natesan S, Rylander MN. Development of a Static Avascular and Dynamic Vascular Human Skin Equivalent Employing Collagen/Keratin Hydrogels. Int J Mol Sci 2024; 25:4992. [PMID: 38732209 PMCID: PMC11084893 DOI: 10.3390/ijms25094992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
One of the primary complications in generating physiologically representative skin tissue is the inability to integrate vasculature into the system, which has been shown to promote the proliferation of basal keratinocytes and consequent keratinocyte differentiation, and is necessary for mimicking representative barrier function in the skin and physiological transport properties. We created a 3D vascularized human skin equivalent (VHSE) with a dermal and epidermal layer, and compared keratinocyte differentiation (immunomarker staining), epidermal thickness (H&E staining), and barrier function (transepithelial electrical resistance (TEER) and dextran permeability) to a static, organotypic avascular HSE (AHSE). The VHSE had a significantly thicker epidermal layer and increased resistance, both an indication of increased barrier function, compared to the AHSE. The inclusion of keratin in our collagen hydrogel extracellular matrix (ECM) increased keratinocyte differentiation and barrier function, indicated by greater resistance and decreased permeability. Surprisingly, however, endothelial cells grown in a collagen/keratin extracellular environment showed increased cell growth and decreased vascular permeability, indicating a more confluent and tighter vessel compared to those grown in a pure collagen environment. The development of a novel VHSE, which incorporated physiological vasculature and a unique collagen/keratin ECM, improved barrier function, vessel development, and skin structure compared to a static AHSE model.
Collapse
Affiliation(s)
- Kameel Zuniga
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA;
- 59th Medical Wing Science and Technology, JBSA-Lackland, TX 78236, USA;
| | - Neda Ghousifam
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Lucy Shaffer
- 59th Medical Wing Science and Technology, JBSA-Lackland, TX 78236, USA;
| | - Sean Brocklehurst
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Mark Van Dyke
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ 85712, USA;
| | - Robert Christy
- Military Health Institute, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| | - Shanmugasundaram Natesan
- Extremity Trauma and Amputation Center of Excellence (EACE), Defense Health Agency, San Diego, CA 92134, USA;
| | - Marissa Nichole Rylander
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA;
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA;
| |
Collapse
|
14
|
Liu H, Xing F, Yu P, Zhe M, Duan X, Liu M, Xiang Z, Ritz U. A review of biomacromolecule-based 3D bioprinting strategies for structure-function integrated repair of skin tissues. Int J Biol Macromol 2024; 268:131623. [PMID: 38642687 DOI: 10.1016/j.ijbiomac.2024.131623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 04/09/2024] [Accepted: 04/13/2024] [Indexed: 04/22/2024]
Abstract
When skin is damaged or affected by diseases, it often undergoes irreversible scar formation, leading to aesthetic concerns and psychological distress for patients. In cases of extensive skin defects, the patient's life can be severely compromised. In recent years, 3D printing technology has emerged as a groundbreaking approach to skin tissue engineering, offering promising solutions to various skin-related conditions. 3D bioprinting technology enables the precise fabrication of structures by programming the spatial arrangement of cells within the skin tissue and subsequently printing skin replacements either in a 3D bioprinter or directly at the site of the defect. This study provides a comprehensive overview of various biopolymer-based inks, with a particular emphasis on chitosan (CS), starch, alginate, agarose, cellulose, and fibronectin, all of which are natural polymers belonging to the category of biomacromolecules. Additionally, it summarizes artificially synthesized polymers capable of enhancing the performance of these biomacromolecule-based bioinks, thereby composing hybrid biopolymer inks aimed at better application in skin tissue engineering endeavors. This review paper examines the recent advancements, characteristics, benefits, and limitations of biological 3D bioprinting techniques for skin tissue engineering. By utilizing bioinks containing seed cells, hydrogels with bioactive factors, and biomaterials, complex structures resembling natural skin can be accurately fabricated in a layer-by-layer manner. The importance of biological scaffolds in promoting skin wound healing and the role of 3D bioprinting in skin tissue regeneration processes is discussed. Additionally, this paper addresses the challenges and constraints associated with current 3D bioprinting technologies for skin tissue and presents future perspectives. These include advancements in bioink formulations, full-thickness skin bioprinting, vascularization strategies, and skin appendages bioprinting.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedic Surgery, Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fei Xing
- Department of Pediatric Surgery, Orthopedic Research Institute, West China Hospital, Sichuan University, 610041 Chengdu, China
| | - Peiyun Yu
- LIMES Institute, Department of Molecular Brain Physiology and Behavior, University of Bonn, Carl-Troll-Str. 31, 53115 Bonn, Germany
| | - Man Zhe
- Animal Experiment Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Duan
- Department of Orthopedic Surgery, Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ming Liu
- Department of Orthopedic Surgery, Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Zhou Xiang
- Department of Orthopedic Surgery, Orthopedic Research Institute, Laboratory of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China; Department of Orthopedics, Sanya People's Hospital, 572000 Sanya, Hainan, China.
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Langenbeckstr. 1, 55131 Mainz, Germany.
| |
Collapse
|
15
|
Labouchère A, Haselbach D, Michetti M, Pythoud C, Raffoul W, Applegate LA, Hirt-Burri N, de Buys Roessingh A. A New Ex Vivo Human Skin Burn Model. J Burn Care Res 2024; 45:308-317. [PMID: 37202124 PMCID: PMC10911692 DOI: 10.1093/jbcr/irad071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Indexed: 05/20/2023]
Abstract
Currently, most burn models for preclinical testing are on animals. For obvious ethical, anatomical, and physiological reasons, these models could be replaced with optimized ex vivo systems. The creation of a burn model on human skin using a pulsed dye laser could represent a relevant model for preclinical research. Six samples of excess human abdominal skin were obtained within one hour after surgery. Burn injuries were induced on small samples of cleaned skin using a pulsed dye laser on skin samples, at varying fluences, pulse numbers and illumination duration. In total, 70 burn injuries were performed on skin ex vivo before being histologically and dermato-pathologically analyzed. Irradiated burned skin samples were classified with a specified code representing burn degrees. Then, a selection of samples was inspected after 14 and 21 days to assess their capacity to heal spontaneously and re-epithelize. We determined the parameters of a pulsed dye laser inducing first, second, and third degree burns on human skin and with fixed parameters, especially superficial and deep second degree burns. After 21 days with the ex vivo model, neo-epidermis was formed. Our results showed that this simple, rapid, user-independent process creates reproducible and uniform burns of different, predictable degrees that are close to clinical reality. Human skin ex vivo models can be an alternative to and complete animal experimentation, particularly for preclinical large screening. This model could be used to foster the testing of new treatments on standardized degrees of burn injuries and thus improve therapeutic strategies.
Collapse
Affiliation(s)
- Ania Labouchère
- PlasticReconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Daniel Haselbach
- PlasticReconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Murielle Michetti
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Catherine Pythoud
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Wassim Raffoul
- PlasticReconstructive, and Hand Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Lee Ann Applegate
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Center for Applied Biotechnology and Molecular Medicine, University of Zurich, Zurich, Switzerland
| | - Nathalie Hirt-Burri
- Regenerative Therapy Unit, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Anthony de Buys Roessingh
- Lausanne Burn Center, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
- Children and Adolescent Surgery Service, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
16
|
Dubau M, Tripetchr T, Mahmoud L, Kral V, Kleuser B. Advancing skin model development: A focus on a self-assembled, induced pluripotent stem cell-derived, xeno-free approach. J Tissue Eng 2024; 15:20417314241291848. [PMID: 39502328 PMCID: PMC11536386 DOI: 10.1177/20417314241291848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/01/2024] [Indexed: 11/08/2024] Open
Abstract
The demand for skin models as alternatives to animal testing has grown due to ethical concerns and the need for accurate substance evaluation. These alternatives, known as New Approach Methodologies (NAMs), are increasingly used for regulatory decisions. Current skin models from primary human cells often rely on bovine collagen, raising ethical issues. This study explores self-assembled skin models (SASM) as a new method, utilizing hair follicle-derived keratinocytes reprogrammed into induced pluripotent stem cells (iPSC) and differentiated into fibroblasts and keratinocytes. The model relies on the ability of fibroblasts to secrete collagen to produce a xeno-free dermal layer and on the differentiation of keratinocytes to create a functional epidermal layer. These layers exhibited confirmed metabolic activity and the capability to withstand test substances. The successful development of SASM underscores the significance of accurate alternatives in dermatological research, providing an ethical and reliable option for substance evaluation and regulatory testing.
Collapse
Affiliation(s)
| | | | - Lava Mahmoud
- Department of Pharmacology and Toxicology, Institute for Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Vivian Kral
- Department of Pharmacology and Toxicology, Institute for Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Burkhard Kleuser
- Department of Pharmacology and Toxicology, Institute for Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
17
|
Jahn M, Lang V, Diehl S, Back R, Kaufmann R, Fauth T, Buerger C. Different immortalized keratinocyte cell lines display distinct capabilities to differentiate and reconstitute an epidermis in vitro. Exp Dermatol 2024; 33:e14985. [PMID: 38043130 DOI: 10.1111/exd.14985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 09/21/2023] [Accepted: 10/22/2023] [Indexed: 12/05/2023]
Abstract
Dermatological research relies on the availability of suitable models that most accurately reflect the in vivo situation. Primary keratinocytes obtained from skin reduction surgeries are not only limited by availability but have a short lifespan and show donor-specific variations, which hamper the understanding of general mechanisms. The spontaneously immortalized keratinocyte cell line HaCaT displays chromosomal aberrations and is known to differentiate in an abnormal manner. To overcome these issues, we validated different engineered immortalized cell lines created from primary human keratinocytes (NHK) as model systems to study epidermal function. Cell lines either immortalized by the expression of SV40 large T antigen and hTERT (NHK-SV/TERT) or by transduction with HPV E6/E7 (NHK-E6/E7) were analysed for their growth and differentiation behaviour using 2D and 3D culture systems and compared to primary keratinocytes. Both cell lines displayed a robust proliferative behaviour but were still sensitive to contact inhibition. NHK-E6/E7 could be driven into differentiation by Ca2+ switch, while NHK-SV/TERT needed withdrawal from any proliferative signal to initiate a delayed onset of differentiation. In 3D epidermal models both cell lines were able to reconstitute a stratified epidermis and functional epidermal barrier. However, only NHK-E6/E7 showed a degree of epidermal maturation and stratification that was comparable to primary keratinocytes.
Collapse
Affiliation(s)
- Magdalena Jahn
- Department of Dermatology, Venerology and Allergology, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Victoria Lang
- Department of Dermatology, Venerology and Allergology, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | - Sandra Diehl
- Department of Dermatology, Venerology and Allergology, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | | | - Roland Kaufmann
- Department of Dermatology, Venerology and Allergology, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| | | | - Claudia Buerger
- Department of Dermatology, Venerology and Allergology, Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany
| |
Collapse
|
18
|
Indolfo NDC, Ganzerla MD, Doratioto TR, Avelino TM, Tofani LB, Peroni LA, Rabelo RS, Arroteia KF, Figueira ACM. Combining a microphysiological system of three organ equivalents and transcriptomics to assess toxicological endpoints for cosmetic ingredients. LAB ON A CHIP 2023; 23:5092-5106. [PMID: 37921576 DOI: 10.1039/d3lc00546a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Animal testing for cosmetic ingredients and final products has been banned in Europe and is gaining legal force worldwide. However, the need for reliable testing methodologies remains for safety assessment of cosmetic ingredients. While new approach methodologies exist for many toxicological endpoints, some complex ones lack appropriate testing methods. Microphysiological systems (MPSs) have emerged as a promising tool to address this gap in pre-clinical testing, offering higher predictivity compared to animal models due to the phylogenetic distance between humans and animals. Moreover, they provide a more physiological approach than traditional in vitro testing by mimicking interconnections between different culture compartments as seen in complex organisms. This study presents a three-organ microfluidic MPS comprising skin, liver, and intestine equivalents. Combining this model with gene expression analysis, we evaluated toxicological endpoints of chemicals, demonstrating its potential for diverse applications. Our findings highlight the MPS model as a reliable and ethical method to be applied in an integrated approach for safety assessment in the cosmetic industry. It offers a promising strategy to evaluate toxicological endpoints for cosmetic ingredients and other chemicals, supporting the elimination of animal testing while ensuring consumer safety.
Collapse
Affiliation(s)
- Nathalia de Carvalho Indolfo
- Natura Cosméticos S.A., Cajamar, São Paulo, Brazil
- Graduate Program in Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, State University of Campinas, Brazil
| | - Melissa Dibbernn Ganzerla
- Graduate Program in Molecular and Morphofunctional Biology, Institute of Biology, State University of Campinas, Brazil
- Brazilian Biosciences National Laboratory (LNBio), Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | | | - Thayná Mendonça Avelino
- Brazilian Biosciences National Laboratory (LNBio), Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | - Larissa Bueno Tofani
- Brazilian Biosciences National Laboratory (LNBio), Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | - Luis Antonio Peroni
- Brazilian Biosciences National Laboratory (LNBio), Center for Research in Energy and Materials (CNPEM), Campinas, Brazil.
| | - Renata Santos Rabelo
- Brazilian Synchrotron Light Laboratory (LNLS), Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | | | | |
Collapse
|
19
|
Czyz CM, Kunth PW, Gruber F, Kremslehner C, Hammers CM, Hundt JE. Requisite instruments for the establishment of three-dimensional epidermal human skin equivalents-A methods review. Exp Dermatol 2023; 32:1870-1883. [PMID: 37605856 DOI: 10.1111/exd.14911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Revised: 07/30/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023]
Abstract
Human skin equivalents (HSEs) are three-dimensional skin organ culture models raised in vitro. This review gives an overview of common techniques for setting up HSEs. The HSE consists of an artificial dermis and epidermis. 3T3-J2 murine fibroblasts, purchased human fibroblasts or freshly isolated and cultured fibroblasts, together with other components, for example, collagen type I, are used to build the scaffold. Freshly isolated and cultured keratinocytes are seeded on top. It is possible to add other cell types, for example, melanocytes, to the HSE-depending on the research question. After several days and further steps, the 3D skin can be harvested. Additionally, we show possible markers and techniques for evaluation of artificial skin. Furthermore, we provide a comparison of HSEs to human skin organ culture, a model which employs human donor skin. We outline advantages and limitations of both models and discuss future perspectives in using HSEs.
Collapse
Affiliation(s)
- Christianna Marie Czyz
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Paul Werner Kunth
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
| | - Florian Gruber
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence - SKINMAGINE, Medical University of Vienna, Vienna, Austria
| | - Christopher Kremslehner
- Christian Doppler Laboratory for Skin Multimodal Analytical Imaging of Aging and Senescence - SKINMAGINE, Medical University of Vienna, Vienna, Austria
| | - Christoph Matthias Hammers
- Lübeck Institute of Experimental Dermatology (LIED), University of Lübeck, Lübeck, Germany
- Department of Dermatology, Venereology and Allergology, University of Kiel, Kiel, Germany
| | | |
Collapse
|
20
|
Voloshin N, Tyurin-Kuzmin P, Karagyaur M, Akopyan Z, Kulebyakin K. Practical Use of Immortalized Cells in Medicine: Current Advances and Future Perspectives. Int J Mol Sci 2023; 24:12716. [PMID: 37628897 PMCID: PMC10454025 DOI: 10.3390/ijms241612716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
In modern science, immortalized cells are not only a convenient tool in fundamental research, but they are also increasingly used in practical medicine. This happens due to their advantages compared to the primary cells, such as the possibility to produce larger amounts of cells and to use them for longer periods of time, the convenience of genetic modification, the absence of donor-to-donor variability when comparing the results of different experiments, etc. On the other hand, immortalization comes with drawbacks: possibilities of malignant transformation and/or major phenotype change due to genetic modification itself or upon long-term cultivation appear. At first glance, such issues are huge hurdles in the way of immortalized cells translation into medicine. However, there are certain ways to overcome such barriers that we describe in this review. We determined four major areas of usage of immortalized cells for practical medicinal purposes, and each has its own means to negate the drawbacks associated with immortalization. Moreover, here we describe specific fields of application of immortalized cells in which these problems are of much lesser concern, for example, in some cases where the possibility of malignant growth is not there at all. In general, we can conclude that immortalized cells have their niches in certain areas of practical medicine where they can successfully compete with other therapeutic approaches, and more preclinical and clinical trials with them should be expected.
Collapse
Affiliation(s)
- Nikita Voloshin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Pyotr Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Zhanna Akopyan
- Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Konstantin Kulebyakin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
- Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia;
| |
Collapse
|
21
|
Jaramillo V, Díaz E, Muñoz LN, González-Barrios AF, Rodríguez-Cortina J, Cruz JC, Muñoz-Camargo C. Enhancing Wound Healing: A Novel Topical Emulsion Combining CW49 Peptide and Lavender Essential Oil for Accelerated Regeneration and Antibacterial Protection. Pharmaceutics 2023; 15:1739. [PMID: 37376187 DOI: 10.3390/pharmaceutics15061739] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Wound healing is a complex process involving blood cells, extracellular matrix, and parenchymal cells. Research on biomimetics in amphibian skin has identified the CW49 peptide from Odorrana grahami, which has been demonstrated to promote wound regeneration. Additionally, lavender essential oil exhibits anti-inflammatory and antibacterial activities. Given these considerations, we propose an innovative emulsion that combines the CW49 peptide with lavender oil. This novel formulation could serve as a potent topical treatment, potentially fostering the regeneration of damaged tissues and providing robust antibacterial protection for skin wounds. This study investigates the physicochemical properties, biocompatibility, and in vitro regenerative capacity of the active components and the emulsion. The results show that the emulsion possesses appropriate rheological characteristics for topical application. Both the CW49 peptide and lavender oil exhibit high viability in human keratinocytes, indicating their biocompatibility. The emulsion induces hemolysis and platelet aggregation, an expected behavior for such topical treatments. Furthermore, the lavender-oil emulsion demonstrates antibacterial activity against both Gram-positive and Gram-negative bacterial strains. Finally, the regenerative potential of the emulsion and its active components is confirmed in a 2D wound model using human keratinocytes. In conclusion, the formulated emulsion, which combines the CW49 peptide and lavender oil, shows great promise as a topical treatment for wound healing. Further research is needed to validate these findings in more advanced in vitro models and in vivo settings, potentially leading to improved wound-care management and novel therapeutic options for patients with skin injuries.
Collapse
Affiliation(s)
- Valentina Jaramillo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Erika Díaz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | - Laura N Muñoz
- Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | | | - Jader Rodríguez-Cortina
- Corporación Colombiana de Investigación Agropecuaria-AGROSAVIA, Centro de Investigación Tibaitatá, Mosquera 250047, Colombia
| | - Juan C Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia
| | | |
Collapse
|
22
|
Stabell AR, Lee GE, Jia Y, Wong KN, Wang S, Ling J, Nguyen SD, Sen GL, Nie Q, Atwood SX. Single-cell transcriptomics of human-skin-equivalent organoids. Cell Rep 2023; 42:112511. [PMID: 37195865 PMCID: PMC10348600 DOI: 10.1016/j.celrep.2023.112511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 03/07/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Several methods for generating human-skin-equivalent (HSE) organoid cultures are in use to study skin biology; however, few studies thoroughly characterize these systems. To fill this gap, we use single-cell transcriptomics to compare in vitro HSEs, xenograft HSEs, and in vivo epidermis. By combining differential gene expression, pseudotime analyses, and spatial localization, we reconstruct HSE keratinocyte differentiation trajectories that recapitulate known in vivo epidermal differentiation pathways and show that HSEs contain major in vivo cellular states. However, HSEs also develop unique keratinocyte states, an expanded basal stem cell program, and disrupted terminal differentiation. Cell-cell communication modeling shows aberrant epithelial-to-mesenchymal transition (EMT)-associated signaling pathways that alter upon epidermal growth factor (EGF) supplementation. Last, xenograft HSEs at early time points post transplantation significantly rescue many in vitro deficits while undergoing a hypoxic response that drives an alternative differentiation lineage. This study highlights the strengths and limitations of organoid cultures and identifies areas for potential innovation.
Collapse
Affiliation(s)
- Adam R Stabell
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Grace E Lee
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yunlong Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Kirsten N Wong
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Shuxiong Wang
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Ling
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sandrine D Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - George L Sen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Dermatology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
23
|
Freer M, Darling N, Goncalves K, Mills KJ, Przyborski S. Development of a mammalian neurosensory full-thickness skin equivalent and its application to screen sensitizing stimuli. Bioeng Transl Med 2023; 8:e10484. [PMID: 37206205 PMCID: PMC10189474 DOI: 10.1002/btm2.10484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/06/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023] Open
Abstract
Human skin equivalents (HSEs) are an increasingly popular research tool due to limitations associated with animal testing for dermatological research. They recapitulate many aspects of skin structure and function, however, many only contain two basic cell types to model dermal and epidermal compartments, which limits their application. We describe advances in the field skin tissue modeling to produce a construct containing sensory-like neurons that is responsive to known noxious stimuli. Through incorporation of mammalian sensory-like neurons, we were able to recapitulate aspects of the neuroinflammatory response including secretion of substance P and a range of pro-inflammatory cytokines in response to a well-characterized neurosensitizing agent: capsaicin. We observed that neuronal cell bodies reside in the upper dermal compartment with neurites extending toward the keratinocytes of the stratum basale where they exist in close proximity to one another. These data suggest that we are able to model aspects of the neuroinflammatory response that occurs during exposure to dermatological stimuli including therapeutics and cosmetics. We propose that this skin construct can be considered a platform technology with a wide range of applications including screening of actives, therapeutics, modeling of inflammatory skin diseases, and fundamental approaches to probe underlying cell and molecular mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Przyborski
- Department of BiosciencesDurham UniversityDurhamUK
- Reprocell Europe LtdGlasgowUK
| |
Collapse
|
24
|
Yadav K, Singh D, Singh MR, Minz S, Princely Ebenezer Gnanakani S, Sucheta, Yadav R, Vora L, Sahu KK, Bagchi A, Singh Chauhan N, Pradhan M. Preclinical study models of psoriasis: State-of-the-art techniques for testing pharmaceutical products in animal and nonanimal models. Int Immunopharmacol 2023; 117:109945. [PMID: 36871534 DOI: 10.1016/j.intimp.2023.109945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/18/2023] [Accepted: 02/23/2023] [Indexed: 03/06/2023]
Abstract
Local and systemic treatments exist for psoriasis, but none can do more than control its symptoms because of its numerous unknown mechanisms. The lack of validated testing models or a defined psoriatic phenotypic profile hinders antipsoriatic drug development. Despite their intricacy, immune-mediated diseases have no improved and precise treatment. The treatment actions may now be predicted for psoriasis and other chronic hyperproliferative skin illnesses using animal models. Their findings confirmed that a psoriasis animal model could mimic a few disease conditions. However, their ethical approval concerns and inability to resemble human psoriasis rightly offer to look for more alternatives. Hence, in this article, we have reported various cutting-edge techniques for the preclinical testing of pharmaceutical products for the treatment of psoriasis.
Collapse
Affiliation(s)
- Krishna Yadav
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India; Raipur Institute of Pharmaceutical Education and Research, Sarona, Raipur, Chhattisgarh 492010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Manju Rawat Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur, Chhattisgarh 492010, India
| | - Sunita Minz
- Department of Pharmacy, Indira Gandhi National Tribal University, Amarkantak, India
| | | | - Sucheta
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | - Renu Yadav
- School of Medical and Allied Sciences, K. R. Mangalam University, Gurugram, Haryana 122103, India
| | - Lalitkumar Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Kantrol Kumar Sahu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh 281406, India
| | - Anindya Bagchi
- Tumor Initiation & Maintenance Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road La Jolla, CA 92037, USA
| | - Nagendra Singh Chauhan
- Drugs Testing Laboratory Avam Anusandhan Kendra (AYUSH), Government Ayurvedic College, Raipur, India
| | | |
Collapse
|
25
|
Goncalves K, De Los Santos Gomez P, Costello L, Smith L, Mead H, Simpson A, Przyborski S. Investigation into the effect of skin tone modulators and exogenous stress on skin pigmentation utilizing a novel bioengineered skin equivalent. Bioeng Transl Med 2023; 8:e10415. [PMID: 36925688 PMCID: PMC10013773 DOI: 10.1002/btm2.10415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/08/2022] [Accepted: 08/30/2022] [Indexed: 11/11/2022] Open
Abstract
Human skin equivalents (HSEs) are a popular technology due to limitations in animal testing, particularly as they recapitulate aspects of structure and function of human skin. Many HSEs contain two basic cell types to model dermal and epidermal compartments, however this limits their application, particularly when investigating the effect of exogenous stressors on skin health. We describe the development of a novel platform technology that accurately replicates skin pigmentation in vitro. Through incorporation of melanocytes, specialized pigment producing cells, into the basal layer of the epidermis we are able to re-create skin pigmentation in vitro. We observe apical distribution of melanin within keratinocytes and formation of supranuclear caps (SPNCs), only when the epidermal compartment is co-cultured with a dermal compartment, leading to the conclusion that fibroblast support is essential for correct pigment organization. We also evaluate the commonly observed phenomenon that pigmentation darkens with time in vitro, which we further explore through mechanical exfoliation to remove a build-up of melanin deposits in the stratum corneum. Finally, we demonstrate the application of a pigmented HSE to investigate drug modulation of skin tone and protection from UV-induced damage, highlighting the importance of such a model in the wider context of skin biology.
Collapse
Affiliation(s)
| | | | | | - Lucy Smith
- Department of BiosciencesDurham UniversityDurhamUK
| | - Hugh Mead
- Department of BiosciencesDurham UniversityDurhamUK
| | - Amy Simpson
- Department of BiosciencesDurham UniversityDurhamUK
| | - Stefan Przyborski
- Department of BiosciencesDurham UniversityDurhamUK
- Reprocell Europe LtdGlasgowUK
| |
Collapse
|
26
|
Multilayer In Vitro Human Skin Tissue Platforms for Quantitative Burn Injury Investigation. Bioengineering (Basel) 2023; 10:bioengineering10020265. [PMID: 36829759 PMCID: PMC9952576 DOI: 10.3390/bioengineering10020265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 02/01/2023] [Accepted: 02/06/2023] [Indexed: 02/19/2023] Open
Abstract
This study presents a multilayer in vitro human skin platform to quantitatively relate predicted spatial time-temperature history with measured tissue injury response. This information is needed to elucidate high-temperature, short-duration burn injury kinetics and enables determination of relevant input parameters for computational models to facilitate treatment planning. Multilayer in vitro skin platforms were constructed using human dermal keratinocytes and fibroblasts embedded in collagen I hydrogels. After three seconds of contact with a 50-100 °C burn tip, ablation, cell death, apoptosis, and HSP70 expression were spatially measured using immunofluorescence confocal microscopy. Finite element modeling was performed using the measured thermal characteristics of skin platforms to determine the temperature distribution within platforms over time. The process coefficients for the Arrhenius thermal injury model describing tissue ablation and cell death were determined such that the predictions calculated from the time-temperature histories fit the experimental burn results. The activation energy for thermal collagen ablation and cell death was found to be significantly lower for short-duration, high-temperature burns than those found for long-duration, low-temperature burns. Analysis of results suggests that different injury mechanisms dominate at higher temperatures, necessitating burn research in the temperature ranges of interest and demonstrating the practicality of the proposed skin platform for this purpose.
Collapse
|
27
|
van Gent ME, van Baaren T, Kłodzińska SN, Ali M, Dolezal N, van Doodewaerd BR, Bos E, de Waal AM, Koning RI, Drijfhout JW, Nielsen HM, Nibbering PH. Encapsulation of SAAP-148 in Octenyl Succinic Anhydride-Modified Hyaluronic Acid Nanogels for Treatment of Skin Wound Infections. Pharmaceutics 2023; 15:pharmaceutics15020429. [PMID: 36839751 PMCID: PMC9967827 DOI: 10.3390/pharmaceutics15020429] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/23/2023] [Accepted: 01/26/2023] [Indexed: 01/31/2023] Open
Abstract
Chronic wound infections colonized by bacteria are becoming more difficult to treat with current antibiotics due to the development of antimicrobial resistance (AMR) as well as biofilm and persister cell formation. Synthetic antibacterial and antibiofilm peptide (SAAP)-148 is an excellent alternative for treatment of such infections but suffers from limitations related to its cationic peptidic nature and thus instability and possible cytotoxicity, resulting in a narrow therapeutic window. Here, we evaluated SAAP-148 encapsulation in nanogels composed of octenyl succinic anhydride (OSA)-modified hyaluronic acid (HA) to circumvent these limitations. SAAP-148 was efficiently (>98%) encapsulated with high drug loading (23%), resulting in monodispersed anionic OSA-HA nanogels with sizes ranging 204-253 nm. Nanogel lyophilization in presence of polyvinyl alcohol maintained their sizes and morphology. SAAP-148 was sustainedly released from lyophilized nanogels (37-41% in 72 h) upon reconstitution. Lyophilized SAAP-148-loaded nanogels showed similar antimicrobial activity as SAAP-148 against planktonic and biofilm-residing AMR Staphylococcus aureus and Acinetobacter baumannii. Importantly, formulated SAAP-148 showed reduced cytotoxicity against human erythrocytes, primary human skin fibroblasts and human keratinocytes. Additionally, lyophilized SAAP-148-loaded nanogels eradicated AMR S. aureus and A. baumannii colonizing a 3D human epidermal model, without inducing any cytotoxicity in contrast to SAAP-148. These findings indicate that OSA-HA nanogels increase SAAP-148's therapeutic potential for treatment of skin wound infections.
Collapse
Affiliation(s)
- Miriam E. van Gent
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence:
| | - Tom van Baaren
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Sylvia N. Kłodzińska
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Muhanad Ali
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Natasja Dolezal
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Bjorn R. van Doodewaerd
- Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Erik Bos
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Amy M. de Waal
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Roman I. Koning
- Electron Microscopy Facility, Department of Cell and Chemical Biology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Jan Wouter Drijfhout
- Department of Immunology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | - Hanne Mørck Nielsen
- Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Peter H. Nibbering
- Department of Infectious Diseases, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| |
Collapse
|
28
|
Kim K, Jeong S, Sung GY. Effect of Periodical Tensile Stimulation on the Human Skin Equivalents by Magnetic Stretching Skin-on-a-Chip (MSSC). BIOCHIP JOURNAL 2022. [DOI: 10.1007/s13206-022-00092-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
29
|
Zuniga K, Ghousifam N, Sansalone J, Senecal K, Van Dyke M, Rylander MN. Keratin Promotes Differentiation of Keratinocytes Seeded on Collagen/Keratin Hydrogels. BIOENGINEERING (BASEL, SWITZERLAND) 2022; 9:bioengineering9100559. [PMID: 36290526 PMCID: PMC9598618 DOI: 10.3390/bioengineering9100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 10/07/2022] [Accepted: 10/09/2022] [Indexed: 11/05/2022]
Abstract
Keratinocytes undergo a complex process of differentiation to form the stratified stratum corneum layer of the skin. In most biomimetic skin models, a 3D hydrogel fabricated out of collagen type I is used to mimic human skin. However, native skin also contains keratin, which makes up 90% of the epidermis and is produced by the keratinocytes present. We hypothesized that the addition of keratin (KTN) in our collagen hydrogel may aid in the process of keratinocyte differentiation compared to a pure collagen hydrogel. Keratinocytes were seeded on top of a 100% collagen or 50/50 C/KTN hydrogel cultured in either calcium-free (Ca-free) or calcium+ (Ca+) media. Our study demonstrates that the addition of keratin and calcium in the media increased lysosomal activity by measuring the glucocerebrosidase (GBA) activity and lysosomal distribution length, an indication of greater keratinocyte differentiation. We also found that the presence of KTN in the hydrogel also increased the expression of involucrin, a differentiation marker, compared to a pure collagen hydrogel. We demonstrate that a combination (i.e., containing both collagen and kerateine or “C/KTN”) hydrogel was able to increase keratinocyte differentiation compared to a pure collagen hydrogel, and the addition of calcium further increased the differentiation of keratinocytes. This multi-protein hydrogel shows promise in future models or treatments to increase keratinocyte differentiation into the stratum corneum.
Collapse
Affiliation(s)
- Kameel Zuniga
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
- Correspondence:
| | - Neda Ghousifam
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - John Sansalone
- Department of Mechanical Engineering, The University of Texas at Austin, Austin, TX 78712, USA
| | - Kris Senecal
- Natick Soldier Center, U.S. Army Soldier & Biological Chemical Command, Natick, MA 01760, USA
| | - Mark Van Dyke
- College of Biomedical Engineering, The University of Arizona, Tucson, AZ 85721, USA
| | | |
Collapse
|
30
|
Khan I, Siddiqui MN, Jameel F, Qazi REM, Salim A, Aslam S, Zaidi MB. Potential of stem cell seeded three-dimensional scaffold for regeneration of full-thickness skin wounds. Interface Focus 2022; 12:20220017. [PMID: 35996740 PMCID: PMC9372646 DOI: 10.1098/rsfs.2022.0017] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/14/2022] [Indexed: 12/11/2022] Open
Abstract
Hypoxic wounds are tough to heal and are associated with chronicity, causing major healthcare burden. Available treatment options offer only limited success for accelerated and scarless healing. Traditional skin substitutes are widely used to improve wound healing, however, they lack proper vascularization. Mesenchymal stem cells (MSCs) offer improved wound healing; however, their poor retention, survival and adherence at the wound site negatively affect their therapeutic potential. The aim of this study is to enhance skin regeneration in a rat model of full-thickness dermal wound by transplanting genetically modified MSCs seeded on a three-dimensional collagen scaffold. Rat bone marrow MSCs were efficiently incorporated in the acellular collagen scaffold. Skin tissues with transplanted subcutaneous scaffolds were histologically analysed, while angiogenesis was assessed both at gene and protein levels. Our findings demonstrated that three-dimensional collagen scaffolds play a potential role in the survival and adherence of stem cells at the wound site, while modification of MSCs with jagged one gene provides a conducive environment for wound regeneration with improved proliferation, reduced inflammation and enhanced vasculogenesis. The results of this study represent an advanced targeted approach having the potential to be translated in clinical settings for targeted personalized therapy.
Collapse
Affiliation(s)
- Irfan Khan
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Marium Naz Siddiqui
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Fatima Jameel
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Rida-e-Maria Qazi
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Asmat Salim
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Shazmeen Aslam
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| | - Midhat Batool Zaidi
- Dr Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan
| |
Collapse
|
31
|
Kovacs D, Bastonini E, Briganti S, Ottaviani M, D’Arino A, Truglio M, Sciuto L, Zaccarini M, Pacifico A, Cota C, Iacovelli P, Picardo M. Altered epidermal proliferation, differentiation, and lipid composition: Novel key elements in the vitiligo puzzle. SCIENCE ADVANCES 2022; 8:eabn9299. [PMID: 36054352 PMCID: PMC10848961 DOI: 10.1126/sciadv.abn9299] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 07/18/2022] [Indexed: 06/15/2023]
Abstract
Vitiligo is an acquired skin depigmentation disease involving multiple pathogenetic mechanisms, which ultimately direct cytotoxic CD8+ cells to destroy melanocytes. Abnormalities have been described in several cells even in pigmented skin as an expression of a functional inherited defect. Keratinocytes regulate skin homeostasis by the assembly of a proper skin barrier and releasing and responding to cytokines and growth factors. Alterations in epidermal proliferation, differentiation, and lipid composition as triggers for immune response activation in vitiligo have not yet been investigated. By applying cellular and lipidomic approaches, we revealed a deregulated keratinocyte differentiation with altered lipid composition, associated with impaired energy metabolism and increased glycolytic enzyme expression. Vitiligo keratinocytes secreted inflammatory mediators, which further increased following mild mechanical stress, thus evidencing immune activation. These findings identify intrinsic alterations of the nonlesional epidermis, which can be the prime instigator of the local inflammatory milieu that stimulates immune responses targeting melanocytes.
Collapse
Affiliation(s)
- Daniela Kovacs
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Emanuela Bastonini
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Stefania Briganti
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Monica Ottaviani
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Andrea D’Arino
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Mauro Truglio
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Lorenzo Sciuto
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Marco Zaccarini
- Genetic Research, Molecular Biology and Dermatopathology Unit, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Alessia Pacifico
- Clinical Dermatology, Phototherapy Unit, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Carlo Cota
- Genetic Research, Molecular Biology and Dermatopathology Unit, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Paolo Iacovelli
- Clinical Dermatology, Phototherapy Unit, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| | - Mauro Picardo
- Laboratory of Cutaneous Physiopathology and Integrated Center of Metabolomics Research, San Gallicano Dermatological Institute, IRCCS, 00144 Rome, Italy
| |
Collapse
|
32
|
Motter Catarino C, Kaiser K, Baltazar T, Motter Catarino L, Brewer JR, Karande P. Evaluation of native and non-native biomaterials for engineering human skin tissue. Bioeng Transl Med 2022; 7:e10297. [PMID: 36176598 PMCID: PMC9472026 DOI: 10.1002/btm2.10297] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/02/2022] [Accepted: 01/07/2022] [Indexed: 11/09/2022] Open
Abstract
A variety of human skin models have been developed for applications in regenerative medicine and efficacy studies. Typically, these employ matrix molecules that are derived from non-human sources along with human cells. Key limitations of such models include a lack of cellular and tissue microenvironment that is representative of human physiology for efficacy studies, as well as the potential for adverse immune responses to animal products for regenerative medicine applications. The use of recombinant extracellular matrix proteins to fabricate tissues can overcome these limitations. We evaluated animal- and non-animal-derived scaffold proteins and glycosaminoglycans for the design of biomaterials for skin reconstruction in vitro. Screening of proteins from the dermal-epidermal junction (collagen IV, laminin 5, and fibronectin) demonstrated that certain protein combinations when used as substrates increase the proliferation and migration of keratinocytes compared to the control (no protein). In the investigation of the effect of components from the dermal layer (collagen types I and III, elastin, hyaluronic acid, and dermatan sulfate), the primary influence on the viability of fibroblasts was attributed to the source of type I collagen (rat tail, human, or bovine) used as scaffold. Furthermore, incorporation of dermatan sulfate in the dermal layer led to a reduction in the contraction of tissues compared to the control where the dermal scaffold was composed primarily of collagen type I. This work highlights the influence of the composition of biomaterials on the development of complex reconstructed skin models that are suitable for clinical translation and in vitro safety assessment.
Collapse
Affiliation(s)
- Carolina Motter Catarino
- Howard P. Isermann Department of Chemical and Biological EngineeringRensselaer Polytechnic InstituteTroyNew YorkUSA
- Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroyNew YorkUSA
| | - Katharina Kaiser
- Department of Biochemistry and Molecular BiologyUniversity of Southern DenmarkOdenseDenmark
| | - Tânia Baltazar
- Howard P. Isermann Department of Chemical and Biological EngineeringRensselaer Polytechnic InstituteTroyNew YorkUSA
- Present address:
Department of ImmunobiologyYale School of MedicineNew HavenConnecticutUSA
| | - Luiza Motter Catarino
- Howard P. Isermann Department of Chemical and Biological EngineeringRensselaer Polytechnic InstituteTroyNew YorkUSA
- Department of BiomedicinePositivo UniversityCuritibaBrazil
| | - Jonathan R. Brewer
- Department of Biochemistry and Molecular BiologyUniversity of Southern DenmarkOdenseDenmark
| | - Pankaj Karande
- Howard P. Isermann Department of Chemical and Biological EngineeringRensselaer Polytechnic InstituteTroyNew YorkUSA
- Center for Biotechnology and Interdisciplinary StudiesRensselaer Polytechnic InstituteTroyNew YorkUSA
| |
Collapse
|
33
|
Melnikov N, Kobel P, Detinis T, Segni AD, Leichtmann-Bardoogo Y, Haik J, Maoz BM. An automated high-throughput platform for experimental study of burn injuries - in vitro and ex vivo. Burns 2022:S0305-4179(22)00228-5. [DOI: 10.1016/j.burns.2022.08.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 08/04/2022] [Accepted: 08/29/2022] [Indexed: 11/02/2022]
|
34
|
Vahav I, Thon M, van den Broek LJ, Spiekstra SW, Atac B, Lindner G, Schimek K, Marx U, Gibbs S. Proof-of-Concept Organ-on-Chip Study: Topical Cinnamaldehyde Exposure of Reconstructed Human Skin with Integrated Neopapillae Cultured under Dynamic Flow. Pharmaceutics 2022; 14:pharmaceutics14081529. [PMID: 35893784 PMCID: PMC9330995 DOI: 10.3390/pharmaceutics14081529] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/01/2023] Open
Abstract
Pharmaceutical and personal care industries require human representative models for testing to ensure the safety of their products. A major route of penetration into our body after substance exposure is via the skin. Our aim was to generate robust culture conditions for a next generation human skin-on-chip model containing neopapillae and to establish proof-of-concept testing with the sensitizer, cinnamaldehyde. Reconstructed human skin consisting of a stratified and differentiated epidermis on a fibroblast populated hydrogel containing neopapillae spheroids (RhS-NP), were cultured air-exposed and under dynamic flow for 10 days. The robustness of three independent experiments, each with up to 21 intra-experiment replicates, was investigated. The epidermis was seen to invaginate into the hydrogel towards the neopapille spheroids. Daily measurements of lactate dehydrogenase (LDH) and glucose levels within the culture medium demonstrated high viability and stable metabolic activity throughout the culture period in all three independent experiments and in the replicates within an experiment. Topical cinnamaldehyde exposure to RhS-NP resulted in dose-dependent cytotoxicity (increased LDH release) and elevated cytokine secretion of contact sensitizer specific IL-18, pro-inflammatory IL-1β, inflammatory IL-23 and IFN-γ, as well as anti-inflammatory IL-10 and IL-12p70. This study demonstrates the robustness and feasibility of complex next generation skin models for investigating skin immunotoxicity.
Collapse
Affiliation(s)
- Irit Vahav
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
- Amsterdam Movement Sciences, Tissue Function & Regeneration, 1081 HV Amsterdam, The Netherlands
| | - Maria Thon
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, 1081 HV Amsterdam, The Netherlands
| | - Lenie J. van den Broek
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Sander W. Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, 1081 HV Amsterdam, The Netherlands
| | - Beren Atac
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
- Department of Biotechnology, Technische Universität Berlin, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Gerd Lindner
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
- Provio GmbH, Oranienburger Chaussee 2, 16548 Glienicke/Nordbahn, Germany
| | | | - Uwe Marx
- TissUse GmbH, Oudenarder Str. 16, 13347 Berlin, Germany
| | - Susan Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, 1081 HV Amsterdam, The Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, 1081 LA Amsterdam, The Netherlands
- Correspondence:
| |
Collapse
|
35
|
Boucard E, Vidal L, Coulon F, Mota C, Hascoët JY, Halary F. The degradation of gelatin/alginate/fibrin hydrogels is cell type dependent and can be modulated by targeting fibrinolysis. Front Bioeng Biotechnol 2022; 10:920929. [PMID: 35935486 PMCID: PMC9355319 DOI: 10.3389/fbioe.2022.920929] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 06/29/2022] [Indexed: 11/29/2022] Open
Abstract
In tissue engineering, cell origin is important to ensure outcome quality. However, the impact of the cell type chosen for seeding in a biocompatible matrix has been less investigated. Here, we investigated the capacity of primary and immortalized fibroblasts of distinct origins to degrade a gelatin/alginate/fibrin (GAF)-based biomaterial. We further established that fibrin was targeted by degradative fibroblasts through the secretion of fibrinolytic matrix-metalloproteinases (MMPs) and urokinase, two types of serine protease. Finally, we demonstrated that besides aprotinin, specific targeting of fibrinolytic MMPs and urokinase led to cell-laden GAF stability for at least forty-eight hours. These results support the use of specific strategies to tune fibrin-based biomaterials degradation over time. It emphasizes the need to choose the right cell type and further bring targeted solutions to avoid the degradation of fibrin-containing hydrogels or bioinks.
Collapse
Affiliation(s)
- Elea Boucard
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Luciano Vidal
- Rapid Manufacturing Platform, Institut de Recherche en Génie Civil et Mécanique (GeM), UMR 7 CNRS 6183 Ecole Centrale de Nantes, Nantes, France
| | - Flora Coulon
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
| | - Carlos Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Jean-Yves Hascoët
- Rapid Manufacturing Platform, Institut de Recherche en Génie Civil et Mécanique (GeM), UMR 7 CNRS 6183 Ecole Centrale de Nantes, Nantes, France
| | - Franck Halary
- Nantes Université, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, Nantes, France
- *Correspondence: Franck Halary,
| |
Collapse
|
36
|
Koskinen Holm C, Qu C. Engineering a 3D In Vitro Model of Human Gingival Tissue Equivalent with Genipin/Cytochalasin D. Int J Mol Sci 2022; 23:ijms23137401. [PMID: 35806407 PMCID: PMC9266888 DOI: 10.3390/ijms23137401] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 06/28/2022] [Accepted: 07/01/2022] [Indexed: 02/05/2023] Open
Abstract
Although three-dimensional (3D) co-culture of gingival keratinocytes and fibroblasts-populated collagen gel can mimic 3D structure of in vivo tissue, the uncontrolled contraction of collagen gel restricts its application in clinical and experimental practices. We here established a stable 3D gingival tissue equivalent (GTE) using hTERT-immortalized gingival fibroblasts (hGFBs)-populated collagen gel directly crosslinked with genipin/cytochalasin D and seeding hTERT-immortalized gingival keratinocytes (TIGKs) on the upper surface for a 2-week air–liquid interface co-culture. MTT assay was used to measure the cell viability of GTEs. GTE size was monitored following culture period, and the contraction was analyzed. Immunohistochemical assay was used to analyze GTE structure. qRT-PCR was conducted to examine the mRNA expression of keratinocyte-specific genes. Fifty µM genipin (G50) or combination (G + C) of G50 and 100 nM cytochalasin D significantly inhibited GTE contraction. Additionally, a higher cell viability appeared in GTEs crosslinked with G50 or G + C. GTEs crosslinked with genipin/cytochalasin D showed a distinct multilayered stratified epithelium that expressed keratinocyte-specific genes similar to native gingiva. Collagen directly crosslinked with G50 or G + C significantly reduced GTE contraction without damaging the epithelium. In summary, the TIGKs and hGFBs can successfully form organotypic multilayered cultures, which can be a valuable tool in the research regarding periodontal disease as well as oral mucosa disease. We conclude that genipin is a promising crosslinker with the ability to reduce collagen contraction while maintaining normal cell function in collagen-based oral tissue engineering.
Collapse
Affiliation(s)
- Cecilia Koskinen Holm
- Department of Odontology, Umeå University, 90185 Umeå, Sweden
- Wallenberg Center for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
- Correspondence: (C.K.H.); (C.Q.)
| | - Chengjuan Qu
- Department of Odontology, Umeå University, 90185 Umeå, Sweden
- Wallenberg Center for Molecular Medicine, Umeå University, 90187 Umeå, Sweden
- Correspondence: (C.K.H.); (C.Q.)
| |
Collapse
|
37
|
Sarmin AM, El Moussaid N, Suntornnond R, Tyler EJ, Kim YH, Di Cio S, Megone WV, Pearce O, Gautrot JE, Dawson J, Connelly JT. Multi-Scale Analysis of the Composition, Structure, and Function of Decellularized Extracellular Matrix for Human Skin and Wound Healing Models. Biomolecules 2022; 12:biom12060837. [PMID: 35740962 PMCID: PMC9221483 DOI: 10.3390/biom12060837] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 06/08/2022] [Accepted: 06/14/2022] [Indexed: 02/04/2023] Open
Abstract
The extracellular matrix (ECM) is a complex mixture of structural proteins, proteoglycans, and signaling molecules that are essential for tissue integrity and homeostasis. While a number of recent studies have explored the use of decellularized ECM (dECM) as a biomaterial for tissue engineering, the complete composition, structure, and mechanics of these materials remain incompletely understood. In this study, we performed an in-depth characterization of skin-derived dECM biomaterials for human skin equivalent (HSE) models. The dECM materials were purified from porcine skin, and through mass spectrometry profiling, we quantified the presence of major ECM molecules, including types I, III, and VI collagen, fibrillin, and lumican. Rheological analysis demonstrated the sol-gel and shear-thinning properties of dECM materials, indicating their physical suitability as a tissue scaffold, while electron microscopy revealed a complex, hierarchical structure of nanofibers in dECM hydrogels. The dECM materials were compatible with advanced biofabrication techniques, including 3D printing within a gelatin microparticle support bath, printing with a sacrificial material, or blending with other ECM molecules to achieve more complex compositions and structures. As a proof of concept, we also demonstrate how dECM materials can be fabricated into a 3D skin wound healing model using 3D printing. Skin-derived dECM therefore represents a complex and versatile biomaterial with advantageous properties for the fabrication of next-generation HSEs.
Collapse
Affiliation(s)
- Atiya M. Sarmin
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 4NS, UK; (A.M.S.); (N.E.M.); (R.S.)
| | - Nadia El Moussaid
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 4NS, UK; (A.M.S.); (N.E.M.); (R.S.)
| | - Ratima Suntornnond
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 4NS, UK; (A.M.S.); (N.E.M.); (R.S.)
| | - Eleanor J. Tyler
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (E.J.T.); (O.P.)
| | - Yang-Hee Kim
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK; (Y.-H.K.); (J.D.)
| | - Stefania Di Cio
- School of Engineering and Materials Sciences, Queen Mary University of London, London E1 4NS, UK; (S.D.C.); (W.V.M.); (J.E.G.)
| | - William V. Megone
- School of Engineering and Materials Sciences, Queen Mary University of London, London E1 4NS, UK; (S.D.C.); (W.V.M.); (J.E.G.)
| | - Oliver Pearce
- Barts Cancer Institute, Queen Mary University of London, London E1 4NS, UK; (E.J.T.); (O.P.)
| | - Julien E. Gautrot
- School of Engineering and Materials Sciences, Queen Mary University of London, London E1 4NS, UK; (S.D.C.); (W.V.M.); (J.E.G.)
| | - Jonathan Dawson
- Institute of Developmental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK; (Y.-H.K.); (J.D.)
| | - John T. Connelly
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Queen Mary University of London, London E1 4NS, UK; (A.M.S.); (N.E.M.); (R.S.)
- Correspondence:
| |
Collapse
|
38
|
Egu DT, Schmitt T, Waschke J. Mechanisms Causing Acantholysis in Pemphigus-Lessons from Human Skin. Front Immunol 2022; 13:884067. [PMID: 35720332 PMCID: PMC9205406 DOI: 10.3389/fimmu.2022.884067] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 04/12/2022] [Indexed: 11/13/2022] Open
Abstract
Pemphigus vulgaris (PV) is an autoimmune bullous skin disease caused primarily by autoantibodies (PV-IgG) against the desmosomal adhesion proteins desmoglein (Dsg)1 and Dsg3. PV patient lesions are characterized by flaccid blisters and ultrastructurally by defined hallmarks including a reduction in desmosome number and size, formation of split desmosomes, as well as uncoupling of keratin filaments from desmosomes. The pathophysiology underlying the disease is known to involve several intracellular signaling pathways downstream of PV-IgG binding. Here, we summarize our studies in which we used transmission electron microscopy to characterize the roles of signaling pathways in the pathogenic effects of PV-IgG on desmosome ultrastructure in a human ex vivo skin model. Blister scores revealed inhibition of p38MAPK, ERK and PLC/Ca2+ to be protective in human epidermis. In contrast, inhibition of Src and PKC, which were shown to be protective in cell cultures and murine models, was not effective for human skin explants. The ultrastructural analysis revealed that for preventing skin blistering at least desmosome number (as modulated by ERK) or keratin filament insertion (as modulated by PLC/Ca2+) need to be ameliorated. Other pathways such as p38MAPK regulate desmosome number, size, and keratin insertion indicating that they control desmosome assembly and disassembly on different levels. Taken together, studies in human skin delineate target mechanisms for the treatment of pemphigus patients. In addition, ultrastructural analysis supports defining the specific role of a given signaling molecule in desmosome turnover at ultrastructural level.
Collapse
|
39
|
Mistry R, Veres M, Issa F. A Systematic Review Comparing Animal and Human Scarring Models. Front Surg 2022; 9:711094. [PMID: 35529910 PMCID: PMC9073696 DOI: 10.3389/fsurg.2022.711094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 04/05/2022] [Indexed: 11/29/2022] Open
Abstract
Introduction A reproducible, standardised model for cutaneous scar tissue to assess therapeutics is crucial to the progress of the field. A systematic review was performed to critically evaluate scarring models in both animal and human research. Method All studies in which cutaneous scars are modelling in animals or humans were included. Models that were focused on the wound healing process or those in humans with scars from an existing injury were excluded. Ovid Medline® was searched on 25 February 2019 to perform two near identical searches; one aimed at animals and the other aimed at humans. Two reviewers independently screened the titles and abstracts for study selection. Full texts of potentially suitable studies were then obtained for analysis. Results The animal kingdom search yielded 818 results, of which 71 were included in the review. Animals utilised included rabbits, mice, pigs, dogs and primates. Methods used for creating scar tissue included sharp excision, dermatome injury, thermal injury and injection of fibrotic substances. The search for scar assessment in humans yielded 287 results, of which 9 met the inclusion criteria. In all human studies, sharp incision was used to create scar tissue. Some studies focused on patients before or after elective surgery, including bilateral breast reduction, knee replacement or midline sternotomy. Discussion The rabbit ear scar model was the most popular tool for scar research, although pigs produce scar tissue which most closely resembles that of humans. Immunodeficient mouse models allow for in vivo engraftment and study of human scar tissue, however, there are limitations relating to the systemic response to these xenografts. Factors that determine the use of animals include cost of housing requirements, genetic traceability, and ethical concerns. In humans, surgical patients are often studied for scarring responses and outcomes, but reproducibility and patient factors that impact healing can limit interpretation. Human tissue use in vitro may serve as a good basis to rapidly screen and assess treatments prior to clinical use, with the advantage of reduced cost and setup requirements.
Collapse
Affiliation(s)
- Riyam Mistry
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
- Correspondence: Riyam Mistry
| | - Mark Veres
- John Radcliffe Hospital, Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
40
|
Zoio P, Oliva A. Skin-on-a-Chip Technology: Microengineering Physiologically Relevant In Vitro Skin Models. Pharmaceutics 2022; 14:pharmaceutics14030682. [PMID: 35336056 PMCID: PMC8955316 DOI: 10.3390/pharmaceutics14030682] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/15/2022] [Accepted: 03/16/2022] [Indexed: 02/06/2023] Open
Abstract
The increased demand for physiologically relevant in vitro human skin models for testing pharmaceutical drugs has led to significant advancements in skin engineering. One of the most promising approaches is the use of in vitro microfluidic systems to generate advanced skin models, commonly known as skin-on-a-chip (SoC) devices. These devices allow the simulation of key mechanical, functional and structural features of the human skin, better mimicking the native microenvironment. Importantly, contrary to conventional cell culture techniques, SoC devices can perfuse the skin tissue, either by the inclusion of perfusable lumens or by the use of microfluidic channels acting as engineered vasculature. Moreover, integrating sensors on the SoC device allows real-time, non-destructive monitoring of skin function and the effect of topically and systemically applied drugs. In this Review, the major challenges and key prerequisites for the creation of physiologically relevant SoC devices for drug testing are considered. Technical (e.g., SoC fabrication and sensor integration) and biological (e.g., cell sourcing and scaffold materials) aspects are discussed. Recent advancements in SoC devices are here presented, and their main achievements and drawbacks are compared and discussed. Finally, this review highlights the current challenges that need to be overcome for the clinical translation of SoC devices.
Collapse
Affiliation(s)
- Patrícia Zoio
- Instituto de Tecnologia Química e Biológica (ITQB), Universidade Nova de Lisboa, Avenida da República, Estação Agronómica Nacional, 2780-157 Oeiras, Portugal;
| | - Abel Oliva
- Instituto de Tecnologia Química e Biológica (ITQB), Universidade Nova de Lisboa, Avenida da República, Estação Agronómica Nacional, 2780-157 Oeiras, Portugal;
- Instituto de Biologia Experimental e Tecnológica (IBET), 2781-901 Oeiras, Portugal
- Correspondence:
| |
Collapse
|
41
|
Sarmin AM, Connelly JT. Fabrication of Human Skin Equivalents Using Decellularized Extracellular Matrix. Curr Protoc 2022; 2:e393. [PMID: 35263039 PMCID: PMC9310708 DOI: 10.1002/cpz1.393] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Atiya M. Sarmin
- Centre for Cell Biology and Cutaneous Research Queen Mary University of London London UK
| | - John T. Connelly
- Centre for Cell Biology and Cutaneous Research Queen Mary University of London London UK
| |
Collapse
|
42
|
Nitsche KS, Müller I, Malcomber S, Carmichael PL, Bouwmeester H. Implementing organ-on-chip in a next-generation risk assessment of chemicals: a review. Arch Toxicol 2022; 96:711-741. [PMID: 35103818 PMCID: PMC8850248 DOI: 10.1007/s00204-022-03234-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 12/17/2022]
Abstract
Organ-on-chip (OoC) technology is full of engineering and biological challenges, but it has the potential to revolutionize the Next-Generation Risk Assessment of novel ingredients for consumer products and chemicals. A successful incorporation of OoC technology into the Next-Generation Risk Assessment toolbox depends on the robustness of the microfluidic devices and the organ tissue models used. Recent advances in standardized device manufacturing, organ tissue cultivation and growth protocols offer the ability to bridge the gaps towards the implementation of organ-on-chip technology. Next-Generation Risk Assessment is an exposure-led and hypothesis-driven tiered approach to risk assessment using detailed human exposure information and the application of appropriate new (non-animal) toxicological testing approaches. Organ-on-chip presents a promising in vitro approach by combining human cell culturing with dynamic microfluidics to improve physiological emulation. Here, we critically review commercial organ-on-chip devices, as well as recent tissue culture model studies of the skin, intestinal barrier and liver as the main metabolic organ to be used on-chip for Next-Generation Risk Assessment. Finally, microfluidically linked tissue combinations such as skin-liver and intestine-liver in organ-on-chip devices are reviewed as they form a relevant aspect for advancing toxicokinetic and toxicodynamic studies. We point to recent achievements and challenges to overcome, to advance non-animal, human-relevant safety studies.
Collapse
Affiliation(s)
- Katharina S Nitsche
- Division of Toxicology, Wageningen University, P.O. Box 8000, 6700 EA, Wageningen, The Netherlands.
| | - Iris Müller
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Sophie Malcomber
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Paul L Carmichael
- Division of Toxicology, Wageningen University, P.O. Box 8000, 6700 EA, Wageningen, The Netherlands
- Unilever Safety and Environmental Assurance Centre, Colworth Science Park, Sharnbrook, Bedfordshire, MK44 1LQ, UK
| | - Hans Bouwmeester
- Division of Toxicology, Wageningen University, P.O. Box 8000, 6700 EA, Wageningen, The Netherlands
| |
Collapse
|
43
|
Arab WT, Susapto HH, Alhattab D, Hauser CAE. Peptide nanogels as a scaffold for fabricating dermal grafts and 3D vascularized skin models. J Tissue Eng 2022; 13:20417314221111868. [PMID: 35923174 PMCID: PMC9340315 DOI: 10.1177/20417314221111868] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Millions of people worldwide suffer from skin injuries, which create significant problems in their lives and are costly to cure. Tissue engineering is a promising approach that aims to fabricate functional organs using biocompatible scaffolds. We designed ultrashort tetrameric peptides with promising properties required for skin tissue engineering. Our work aimed to test the efficacy of these scaffolds for the fabrication of dermal grafts and 3D vascularized skin tissue models. We found that the direct contact of keratinocytes and fibroblasts enhanced the proliferation of the keratinocytes. Moreover, the expression levels of TGF-β1, b-FGF, IL-6, and IL-1α is correlated with the growth of the fibroblasts and keratinocytes in the co-culture. Furthermore, we successfully produced a 3D vascularized skin co-culture model using these peptide scaffolds. We believe that the described results represent an advancement in the fabrication of skin tissue equivalent, thereby providing the opportunity to rebuild missing, failing, or damaged parts. Graphical abstract
Collapse
Affiliation(s)
- Wafaa T Arab
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Hepi H Susapto
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computational Bioscience Research Center (CBRC), KAUST, Thuwal, Saudi Arabia
| | - Dana Alhattab
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computational Bioscience Research Center (CBRC), KAUST, Thuwal, Saudi Arabia
| | - Charlotte A E Hauser
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and Engineering (BESE), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
- Computational Bioscience Research Center (CBRC), KAUST, Thuwal, Saudi Arabia
| |
Collapse
|
44
|
Comparative In Vitro Evaluation of Commercial Periodontal Gels on Antibacterial, Biocompatibility and Wound Healing Ability. Pharmaceutics 2021; 13:pharmaceutics13091502. [PMID: 34575578 PMCID: PMC8465455 DOI: 10.3390/pharmaceutics13091502] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/08/2021] [Accepted: 09/14/2021] [Indexed: 11/17/2022] Open
Abstract
In the last years, several studies testing commercial periodontal gels that contain chlorhexidine (CHX) or other antibacterial agents, have raised concerns regarding their cytotoxicity in periodontal tissues. We aimed at comparing the biocompatibility but also the efficacy as regards to the antibacterial and wound healing ability of different commercial periodontal gels. In vitro human gingival fibroblasts (GF) and a 3D model of human tissue equivalents of gingiva (GTE) were used under inflammatory conditions to evaluate wound closure, cytotoxicity and gene expression. Antibacterial effects were also investigated on Porphyromonas gingivalis growth, viability and gingipain activity. In GF and in the bacterial study, we found cytotoxic effects on GF and a high inhibition on bacterial growth rate in gels containing CHX, asiaticoside, enoxolone, cetylpyridinium chloride, propolis and eugenol. Of the two gels that were non-cytotoxic, Syntoss Biogel (containing chondrontin sulfate) and Emdogain (EMD, containing amelogenin and propylene glycol alginate), EMD showed the best wound closure, with no effect on P. gingivalis growth but decreased gingipain activity. On the other hand, Syntoss Biogel reduced viability and gingipain activity of P. gingivalis, but lack wound healing capacity. In the 3D GTE, Syntoss Biogel and EMD showed a good biocompatibility. Among all the tested gels, formulations containing CHX, asiaticoside, enoxolone, cetylpyridinium chloride, propolis and eugenol showed high antibacterial effect but also showed high cytotoxicity in eukaryotic cells. EMD was the one with the best biocompatibility and wound healing ability at the conditions tested.
Collapse
|
45
|
Fibroblast-derived matrices-based human skin equivalent as an in vitro psoriatic model for drug testing. J Biosci 2021. [DOI: 10.1007/s12038-021-00205-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
46
|
Plaza C, Meyrignac C, Botto JM, Capallere C. Characterization of a New Full-Thickness In Vitro Skin Model. Tissue Eng Part C Methods 2021; 27:411-420. [PMID: 34107746 DOI: 10.1089/ten.tec.2021.0035] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Since 30 years, bioengineering allowed to reconstruct human tissues using normal human cells. Skin is one of the first organ to be reconstructed thanks to the development of specific cell culture media and supports favoring the culture of human skin cells, such as fibroblasts, keratinocytes, or melanocytes. Skin models have evolved from epidermis to complex models including a dermis. The purpose of the present study was to design a reconstructed full-thickness (FT) skin suitable to perform in vitro testing of both molecules and plant extracts. First, we reconstructed epidermis with normal human keratinocytes displaying the expected multilayered morphology and expressing specific epidermal proteins (e-cadherin, claudin-1, p63, Ki67, Keratin 10, filaggrin, and loricrin). Then, a dermal equivalent was developed using a collagen matrix allowing the growth of fibroblasts. The functionality of the dermis was demonstrated by the measurement of skin parameters such as rigidity or elasticity with Ballistometer® and other parameters such as the contraction over time and the expression of dermal proteins. The combination of these two compartments (dermis and epidermis) allowed to reconstruct an FT model. This study model allowed to study the communication between compartments and with the establishment of a dermoepidermal junction showing the expression of specific proteins (collagen XVII, laminin, and collagen IV). Impact statement The objective of our research project was to design a three-dimensional human full-thickness (FT) skin suitable to perform in vitro testing of molecules and plant ingredients. The combination of these two reconstructed compartments (dermis and epidermis) allowed to reconstruct an FT model. This study model allowed to study the communication between compartments and with the establishment of a dermoepidermal junction showing the expression of specific proteins (collagen XVII, laminin, and collagen IV). This in vitro model can be use by cosmetic and pharmaceutical industries to study the effect of chemical or natural compounds on the skin.
Collapse
Affiliation(s)
- Christelle Plaza
- Department of Tissue Engineering and Toxicology In Vitro, Ashland Specialties France, Sophia Antipolis, France
| | - Celine Meyrignac
- Department of Tissue Engineering and Toxicology In Vitro, Ashland Specialties France, Sophia Antipolis, France
| | - Jean-Marie Botto
- Department of Tissue Engineering and Toxicology In Vitro, Ashland Specialties France, Sophia Antipolis, France
| | - Christophe Capallere
- Department of Tissue Engineering and Toxicology In Vitro, Ashland Specialties France, Sophia Antipolis, France
| |
Collapse
|
47
|
Murakami M, Akagi T, Sasano Y, Chiba T, Narita H, Shimoda H, Akashi M. Observation of a tight junction structure generated in LbL-3D skin reconstructed by layer-by-layer cell coating technique. J Tissue Eng Regen Med 2021; 15:798-803. [PMID: 34028181 DOI: 10.1002/term.3221] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 04/22/2021] [Accepted: 04/23/2021] [Indexed: 12/12/2022]
Abstract
Tissue-engineered skin equivalents are reconstructed the functions of human skin and can be used as an alternative to animal experiments in basic study or as cultured skin for regenerative medicine. Recent studies confirmed that epidermal tight junctions (TJs), which are complex intercellular junctions formed in the stratum granulosum of human skin, play an important part in the formation of the skin barrier function. In well-formed reconstructed human skin models, there are several reports on the expression of TJ proteins and their localization in epidermal layer, however, the morphological features of TJ, showing tight junctional contacts and the process of TJ formation have yet to be investigated. In this study, we systematically examined and identified TJ-related proteins and TJ structure in three-dimensional (3D) human skin equivalents reconstructed by layer-by-layer (LbL) cell coating technique (LbL-3D Skin). We demonstrate localization of TJ-related proteins and time course of formation of TJ structure with typical junctional morphology in LbL-3D Skin. These data provide evidence that the LbL-3D Skin is an in vitro model with structure and function extremely similar to living skin.
Collapse
Affiliation(s)
- Masato Murakami
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Takami Akagi
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| | - Yumi Sasano
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.,Life & Healthcare Products Department, Pharma-Medicals Division, Nagase & Co., Ltd., Kobe, Japan
| | - Tomohiro Chiba
- Department of Anatomical Science, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Hirokazu Narita
- Department of Anatomical Science, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Hiroshi Shimoda
- Department of Anatomical Science, Hirosaki University Graduate School of Medicine, Aomori, Japan
| | - Mitsuru Akashi
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan
| |
Collapse
|
48
|
Hirano M, Huang Y, Vela Jarquin D, De la Garza Hernández RL, Jodat YA, Luna Cerón E, García-Rivera LE, Shin SR. 3D bioprinted human iPSC-derived somatosensory constructs with functional and highly purified sensory neuron networks. Biofabrication 2021; 13. [PMID: 33962404 DOI: 10.1088/1758-5090/abff11] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Accepted: 05/07/2021] [Indexed: 12/20/2022]
Abstract
Engineering three-dimensional (3D) sensible tissue constructs, along with the complex microarchitecture wiring of the sensory nervous system, has been an ongoing challenge in the tissue engineering field. By combining 3D bioprinting and human pluripotent stem cell (hPSC) technologies, sensible tissue constructs could be engineered in a rapid, precise, and controllable manner to replicate 3D microarchitectures and mechanosensory functionalities of the native sensory tissue (e.g. response to external stimuli). Here, we introduce a biofabrication approach to create complex 3D microarchitecture wirings. We develop an hPSC-sensory neuron (SN) laden bioink using highly purified and functional SN populations to 3D bioprint microarchitecture wirings that demonstrate responsiveness to warm/cold sense-inducing chemicals and mechanical stress. Specifically, we tailor a conventional differentiation strategy to our purification method by utilizing p75 cell surface marker and DAPT treatment along with neuronal growth factors in order to selectively differentiate neural crest cells into SNs. To create spatial resolution in 3D architectures and grow SNs in custom patterns and directions, an induced pluripotent stem cell (iPSC)-SN-laden gelatin bioink was printed on laminin-coated substrates using extrusion-based bioprinting technique. Then the printed constructs were covered with a collagen matrix that guided SNs growing in the printed micropattern. Using a sacrificial bioprinting technique, the iPSC-SNs were seeded into the hollow microchannels created by sacrificial gelatin ink printed in the gelatin methacryloyl supporting bath, thereby demonstrating controllability over axon guidance in curved lines up to several tens of centimeters in length on 2D substrates and in straight microchannels in 3D matrices. Therefore, this biofabrication approach could be amenable to incorporate sensible SN networks into the engineered skin equivalents, regenerative skin implants, and augmented somatosensory neuro-prosthetics that have the potential to regenerate sensible functions by connecting host neuron systems in injured areas.
Collapse
Affiliation(s)
- Minoru Hirano
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America.,Future Vehicle Research Department, Toyota Research Institute North America, Toyota Motor North America Inc., 1555 Woodridge Ave, Ann Arbor, MI 48105, United States of America
| | - Yike Huang
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America.,Institute of Blood Transfusion, Chinese Academy of Medical Sciences and Peking Union Medical College, Chengdu 100730, People's Republic of China
| | - Daniel Vela Jarquin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America.,Instituto Tecnológico y de Estudios Superiores de Monterrey, Calle del Puente 222, Ejidos de Huipulco, Tlalpan, Ciudad de México, CDMX 14380, Mexico
| | - Rosakaren Ludivina De la Garza Hernández
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America.,Instituto Tecnológico y de Estudios Superiores de Monterrey, Av. Eugenio Garza Sada 2501 Sur, Tecnológico, 64849 Monterrey, NL, Mexico
| | - Yasamin A Jodat
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America
| | - Eder Luna Cerón
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America.,Instituto Tecnológico y de Estudios Superiores de Monterrey, Calle del Puente 222, Ejidos de Huipulco, Tlalpan, Ciudad de México, CDMX 14380, Mexico
| | - Luis Enrique García-Rivera
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America.,Instituto Tecnológico y de Estudios Superiores de Monterrey, Calle del Puente 222, Ejidos de Huipulco, Tlalpan, Ciudad de México, CDMX 14380, Mexico
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham Women's Hospital, Cambridge, MA 02139, United States of America
| |
Collapse
|
49
|
Ren X, Getschman AE, Hwang S, Volkman BF, Klonisch T, Levin D, Zhao M, Santos S, Liu S, Cheng J, Lin F. Investigations on T cell transmigration in a human skin-on-chip (SoC) model. LAB ON A CHIP 2021; 21:1527-1539. [PMID: 33616124 PMCID: PMC8058301 DOI: 10.1039/d0lc01194k] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
A microfluidics-based three-dimensional skin-on-chip (SoC) model is developed in this study to enable quantitative studies of transendothelial and transepithelial migration of human T lymphocytes in mimicked skin inflammatory microenvironments and to test new drug candidates. The keys results include 1) CCL20-dependent T cell transmigration is significantly inhibited by an engineered CCL20 locked dimer (CCL20LD), supporting the potential immunotherapeutic use of CCL20LD for treating skin diseases such as psoriasis; 2) transepithelial migration of T cells in response to a CXCL12 gradient mimicking T cell egress from the skin is significantly reduced by a sphingosine-1-phosphate (S1P) background, suggesting the role of S1P for T cell retention in inflamed skin tissues; and 3) T cell transmigration is induced by inflammatory cytokine stimulated epithelial cells in the SoC model. Collectively, the developed SoC model recreates a dynamic multi-cellular micro-environment that enables quantitative studies of T cell transmigration at a single cell level in response to physiological cutaneous inflammatory mediators and potential drugs.
Collapse
Affiliation(s)
- Xiaoou Ren
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada. and Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Anthony E Getschman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Samuel Hwang
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Thomas Klonisch
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - David Levin
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Min Zhao
- Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA and Department of Ophthalmology & Vision Science, California Davis School of Medicine, Sacramento, CA 95817, USA
| | - Susy Santos
- Victoria General Hospital, Winnipeg, MB R3T 2E8, Canada
| | - Song Liu
- Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Jasmine Cheng
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada.
| | - Francis Lin
- Department of Physics and Astronomy, University of Manitoba, 30A Sifton Rd, 301 Allen Bldg, Winnipeg, MB R3T 2N2, Canada. and Department of Biosystems Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada and Department of Dermatology, University of California Davis School of Medicine, Sacramento, CA 95816, USA and Department of Immunology, University of Manitoba, Winnipeg, MB R3E 0T5, Canada
| |
Collapse
|
50
|
Abstract
The skin is a passive and active barrier which protects the body from the environment. Its health is essential for the accomplishment of this role. Since several decades, the skin has aroused a strong interest in various fields (for e.g. cell biology, medicine, toxicology, cosmetology, and pharmacology). In contrast to other organs, 3D models were mostly and directly elaborated in humans due to its architectural simplicity and easy accessibility. The development of these models benefited from the societal pressure to reduce animal experiments. In this review, we first describe human and mouse skin structure and the major differences with other mammals and birds. Next, we describe the different 3D human skin models and their main applications. Finally, we review the available models for domestic animals and discuss the current and potential applications.
Collapse
Affiliation(s)
- Laurent Souci
- ISP, INRAE, Université de Tours, Equipe BioVA, Centre Val de Loire, 37380, Nouzilly, France
| | - Caroline Denesvre
- ISP, INRAE, Université de Tours, Equipe BioVA, Centre Val de Loire, 37380, Nouzilly, France.
| |
Collapse
|