1
|
Lim YW, Quinn R, Bharti K, Ferrer M, Zarkoob H, Song MJ. Development of immunocompetent full thickness skin tissue constructs to model skin fibrosis for high-throughput drug screening. Biofabrication 2024; 17:015033. [PMID: 39622178 PMCID: PMC11638742 DOI: 10.1088/1758-5090/ad998c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 10/30/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
The lack of the immune component in most of the engineered skin models remains a challenge to study the interplay between different immune and non-immune cell types of the skin. Immunocompetent humanin vitroskin models offer potential advantages in recapitulatingin vivolike behavior which can serve to accelerate translational research and therapeutics development for skin diseases. Here we describe a three-dimensional human full-thickness skin (FTS) equivalent incorporating polarized M1 and M2 macrophages from human peripheral CD14+monocytes. This macrophage-incorporated FTS model demonstrates discernible immune responses with physiologically relevant cytokine production and macrophage plasticity under homeostatic and lipopolysaccharide stimulation conditions. M2-incorporated FTS recapitulates skin fibrosis phenotypes with transforming growth factor-β1 treatment as reflected by significant collagen deposition and myofibroblast expression, demonstrating a M2 potentiation effect. In conclusion, we successfully biofabricated an immunocompetent FTS with functional macrophages in a high-throughput (HT) amenable format. This model is the first step towards a HT-assay platform to develop new therapeutics for skin diseases.
Collapse
Affiliation(s)
- Yi Wei Lim
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Russell Quinn
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Kapil Bharti
- National Eye Institute, National Institutes of Health, Bethesda, MD 20814, United States of America
| | - Marc Ferrer
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Hoda Zarkoob
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| | - Min Jae Song
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, United States of America
| |
Collapse
|
2
|
Michielon E, King L, Waaijman T, Veth M, Spiekstra S, van der Vliet H, Gibbs S, de Gruijl T. An organotypic human melanoma-in-skin model as an in vitro tool for testing Vγ9Vδ2-T cell-based immunotherapy. IMMUNO-ONCOLOGY TECHNOLOGY 2024; 24:100724. [PMID: 39220726 PMCID: PMC11363583 DOI: 10.1016/j.iotech.2024.100724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Despite considerable advancements in cancer immunotherapy, advanced melanoma still presents a substantial clinical challenge. In an effort to explore treatment options, we examined the immunotherapeutic potential of effector Vγ9Vδ2-T cells in vitro in a three-dimensional (3D) human organotypic melanoma-in-skin (Mel-RhS) model. Materials and methods Vγ9Vδ2-T cells were introduced into Mel-RhS via intradermal injection and cultured within the tissue microenvironment for up to 3 days. Results Vγ9Vδ2-T cells remained viable for up to 3 days and were in close proximity to or within tumor nests. Upon Mel-RhS dissociation, a fraction was shown to be decorated by melanoma-associated chondroitin sulfate proteoglycan (MCSP), demonstrating their ability to actively navigate the tumor microenvironment and trogocytose cancer cells. Investigation into the apparent trogocytosis revealed an enhanced activated state of MCSP-decorated Vγ9Vδ2-T cells, evidenced by increased expression levels of 4-1BB, NKp44, programmed cell death protein-1 (PD-1), and programmed death-ligand 1 (PD-L1), compared with their MCSP- counterpart. These findings suggest that Vγ9Vδ2-T cells, upon successfully contacting melanoma cells, actively recognize and acquire MCSP from these malignant cells. Evidence of actual tumor cell elimination, although not significant, was only obtained after preincubation of Mel-RhS with pamidronate, a phosphoantigen-inducing agent, indicating the need for additional T cell receptor-mediated signaling for Vγ9Vδ2-T cells to reach their full oncolytic potential. Conclusions This study highlights the viability and persistence of Vγ9Vδ2-T cells within the 3D microenvironment, their migratory and antitumor functionality, and the suitability of the model for testing T cell-based therapies, contributing both to the understanding of Vγ9Vδ2-T cell biology and their application in cancer immunotherapy.
Collapse
Affiliation(s)
- E. Michielon
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
| | - L.A. King
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Department of Medical Oncology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
| | - T. Waaijman
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
| | - M. Veth
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Department of Medical Oncology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
| | - S.W. Spiekstra
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
| | - H.J. van der Vliet
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Department of Medical Oncology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
- Lava Therapeutics NV, Utrecht
| | - S. Gibbs
- Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, The Netherlands
| | - T.D. de Gruijl
- Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Cancer Center Amsterdam, Cancer Biology and Immunology Program, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam
- Department of Medical Oncology, Amsterdam University Medical Center Location Vrije Universiteit Amsterdam, Amsterdam
| |
Collapse
|
3
|
Wang XY, Jia QN, Li J, Zheng HY. Organoids as Tools for Investigating Skin Aging: Mechanisms, Applications, and Insights. Biomolecules 2024; 14:1436. [PMID: 39595612 PMCID: PMC11591780 DOI: 10.3390/biom14111436] [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: 09/23/2024] [Revised: 11/04/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Organoids have emerged as transformative tools in biomedical research, renowned for their ability to replicate the complexity construct of human tissues. Skin aging is a multifaceted biological process, influenced by both intrinsic factors and extrinsic factors. Traditional models for studying skin aging often fall short in capturing the intricate dynamics of human skin. In contrast, skin organoids offer a more physiologically relevant system, reflecting the structural and functional characteristics of native skin. These characteristics make skin organoids highly suitable for studying the mechanisms of skin aging, identifying novel therapeutic targets, and testing anti-aging interventions. Despite their promise, challenges such as limited scalability, reproducibility, and ethical considerations remain. Addressing these hurdles through interdisciplinary research and technological advancements will be essential to maximizing the potential of skin organoids for dermatological research and personalized anti-aging therapies.
Collapse
Affiliation(s)
| | | | - Jun Li
- Peking Union Medical College Hospital (Dongdan Campus), No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing 100730, China; (X.-Y.W.); (Q.-N.J.)
| | - He-Yi Zheng
- Peking Union Medical College Hospital (Dongdan Campus), No. 1 Shuaifuyuan Wangfujing Dongcheng District, Beijing 100730, China; (X.-Y.W.); (Q.-N.J.)
| |
Collapse
|
4
|
Ferrara V, Perfili C, Artemi G, Iacolino B, Sciandra F, Perini G, Fusco L, Pogorielov M, Delogu LG, Papi M, De Spirito M, Palmieri V. Advanced approaches in skin wound healing - a review on the multifunctional properties of MXenes in therapy and sensing. NANOSCALE 2024; 16:18684-18714. [PMID: 39312211 DOI: 10.1039/d4nr02843k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2024]
Abstract
In recent years, the use of MXenes, a class of two-dimensional materials composed of transition metal carbides, nitrides, or carbonitrides, has shown significant promise in the field of skin wound healing. This review explores the multifunctional properties of MXenes, focusing on their electrical conductivity, photothermal effects, and biocompatibility in this field. MXenes have been utilized to develop advanced wound healing devices such as hydrogels, patches, and smart bandages for healing examination. These devices offer enhanced antibacterial activity, promote tissue regeneration, and provide real-time monitoring of parameters. The review highlights the synthesis methods, chemical features, and biological effects of MXenes, emphasizing their role in innovative skin repair strategies. Additionally, it discusses the potential of MXene-based sensors for humidity, pH, and temperature monitoring, which are crucial for preventing infections and complications in wound healing. The integration of MXenes into wearable devices represents a significant advancement in wound management, promising improved clinical outcomes and enhanced quality of life for patients.
Collapse
Affiliation(s)
- Valeria Ferrara
- Fondazione Policlinico Universitario "A. Gemelli" IRCSS, L.go Agostino Gemelli 8, 00136 RM, Rome, Italy.
| | - Caterina Perfili
- Fondazione Policlinico Universitario "A. Gemelli" IRCSS, L.go Agostino Gemelli 8, 00136 RM, Rome, Italy.
| | - Giulia Artemi
- Istituto dei Sistemi Complessi, ISC-CNR, Via dei Taurini 19, 00185, RM, Rome, Italy
| | - Brunella Iacolino
- Istituto dei Sistemi Complessi, ISC-CNR, Via dei Taurini 19, 00185, RM, Rome, Italy
| | - Francesca Sciandra
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", SCITEC-CNR, c/o Istituto Biochimica e Biochimica Clinica, Università Cattolica del Sacro Cuore, L.go Vito, 1, 00168 RM, Rome, Italy
| | - Giordano Perini
- Fondazione Policlinico Universitario "A. Gemelli" IRCSS, L.go Agostino Gemelli 8, 00136 RM, Rome, Italy.
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, L.go Francesco Vito, 1, 00168 RM, Rome, Italy
| | - Laura Fusco
- ImmuneNanoLab, Dipartimento di Scienze Biomediche, Università degli Studi di Padova, Padova, Italy
| | - Maksym Pogorielov
- Biomedical Research Centre, Sumy State University, Kharkivska Street, 116, 40007, Sumy, Ukraine
- Institute of Atomic Physics and Spectroscopy, University of Latvia, Jelgavas street, 3, Riga, 1004, Latvia
| | - Lucia Gemma Delogu
- RIC2D, Department of Biological Sciences, Khalifa University of Science & Technology, Abu Dhabi, United Arab Emirates
- ImmuneNanoLab, Dipartimento di Scienze Biomediche, Università degli Studi di Padova, Padova, Italy
| | - Massimiliano Papi
- Fondazione Policlinico Universitario "A. Gemelli" IRCSS, L.go Agostino Gemelli 8, 00136 RM, Rome, Italy.
- Istituto dei Sistemi Complessi, ISC-CNR, Via dei Taurini 19, 00185, RM, Rome, Italy
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, L.go Francesco Vito, 1, 00168 RM, Rome, Italy
| | - Marco De Spirito
- Fondazione Policlinico Universitario "A. Gemelli" IRCSS, L.go Agostino Gemelli 8, 00136 RM, Rome, Italy.
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, L.go Francesco Vito, 1, 00168 RM, Rome, Italy
| | - Valentina Palmieri
- Fondazione Policlinico Universitario "A. Gemelli" IRCSS, L.go Agostino Gemelli 8, 00136 RM, Rome, Italy.
- Istituto dei Sistemi Complessi, ISC-CNR, Via dei Taurini 19, 00185, RM, Rome, Italy
- Dipartimento di Neuroscienze, Università Cattolica del Sacro Cuore, L.go Francesco Vito, 1, 00168 RM, Rome, Italy
| |
Collapse
|
5
|
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
|
6
|
Bhar B, Das E, Manikumar K, Mandal BB. 3D Bioprinted Human Skin Model Recapitulating Native-Like Tissue Maturation and Immunocompetence as an Advanced Platform for Skin Sensitization Assessment. Adv Healthc Mater 2024; 13:e2303312. [PMID: 38478847 DOI: 10.1002/adhm.202303312] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Revised: 03/08/2024] [Indexed: 03/28/2024]
Abstract
Physiologically-relevant in vitro skin models hold the utmost importance for efficacy assessments of pharmaceutical and cosmeceutical formulations, offering valuable alternatives to animal testing. Here, an advanced immunocompetent 3D bioprinted human skin model is presented to assess skin sensitization. Initially, a photopolymerizable bioink is formulated using silk fibroin methacrylate, gelatin methacrylate, and photoactivated human platelet releasate. The developed bioink shows desirable physicochemical and rheological attributes for microextrusion bioprinting. The tunable physical and mechanical properties of bioink are modulated through variable photocuring time for optimization. Thereafter, the bioink is utilized to 3D bioprint "sandwich type" skin construct where an artificial basement membrane supports a biomimetic epidermal layer on one side and a printed pre-vascularized dermal layer on the other side within a transwell system. The printed construct is further cultured in the air-liquid interface for maturation. Immunofluorescence staining demonstrated a differentiated keratinocyte layer and dermal extracellular matrix (ECM)-remodeling by fibroblasts and endothelial cells. The biochemical estimations and gene-expression analysis validate the maturation of the printed model. The incorporation of macrophages further enhances the physiological relevance of the model. This model effectively classifies skin irritative and non-irritative substances, thus establishing itself as a suitable pre-clinical screening platform for sensitization tests.
Collapse
Affiliation(s)
- Bibrita Bhar
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Eshani Das
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Kodieswaran Manikumar
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| | - Biman B Mandal
- Biomaterials and Tissue Engineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Centre for Nanotechnology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
- Jyoti and Bhupat Mehta School of Health Sciences and Technology, Indian Institute of Technology Guwahati, Guwahati, Assam, 781039, India
| |
Collapse
|
7
|
Li M, Liu Z, Shen Z, Han L, Wang J, Sang S. A heparin-functionalized bioink with sustained delivery of vascular endothelial growth factor for 3D bioprinting of prevascularized dermal constructs. Int J Biol Macromol 2024; 262:130075. [PMID: 38340924 DOI: 10.1016/j.ijbiomac.2024.130075] [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: 07/31/2023] [Revised: 02/06/2024] [Accepted: 02/07/2024] [Indexed: 02/12/2024]
Abstract
Skin tissue engineering faces challenges due to the absence of vascular architecture, impeding the development of permanent skin replacements. To address this, a heparin-functionalized 3D-printed bioink (GH/HepMA) was formulated to enable sustained delivery of vascular endothelial growth factor (VEGF), comprising 0.3 % (w/v) hyaluronic acid (HA), 10 % (w/v) gelatin methacrylate (GelMA), and 0.5 % (w/v) heparin methacrylate (HepMA). The bioink was then used to print dermal constructs with angiogenic functions, including fibroblast networks and human umbilical vein endothelial cell (HUVEC) networks. GH/HepMA, with its covalently cross-linked structure, exhibits enhanced mechanical properties and heparin stability, allowing for a 21-day sustained delivery of VEGF. Cytocompatibility experiments showed that the GH/HepMA bioink supported fibroblast proliferation and promoted collagen I production. With VEGF present, the GH/HepMA bioink promoted HUVEC proliferation, migration, as well as the formation of a richer capillary-like network. Furthermore, HA within the GH/HepMA bioink enhanced rheological properties and printability. Additionally, 3D-bioprinted dermal constructs showed significant deposition of collagen I and III and mature stable capillary-like structures along the axial direction. In summary, this study offers a promising approach for constructing biomimetic multicellular skin substitutes with angiogenesis-induced functions.
Collapse
Affiliation(s)
- Meng Li
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Zixian Liu
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, PR China
| | - Zhizhong Shen
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan 030031, PR China
| | - Lu Han
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Shanxi Research Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan 030031, PR China
| | - Jianming Wang
- General Hospital of TISCO, North Street, Xinghualing District, Taiyuan 030809, PR China
| | - Shengbo Sang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Electronic Information and Optical Engineering, Taiyuan University of Technology, Taiyuan 030024, PR China; Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, PR China.
| |
Collapse
|
8
|
Yan J, Wu T, Zhang J, Gao Y, Wu JM, Wang S. Revolutionizing the female reproductive system research using microfluidic chip platform. J Nanobiotechnology 2023; 21:490. [PMID: 38111049 PMCID: PMC10729361 DOI: 10.1186/s12951-023-02258-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Accepted: 12/07/2023] [Indexed: 12/20/2023] Open
Abstract
Comprehensively understanding the female reproductive system is crucial for safeguarding fertility and preventing diseases concerning women's health. With the capacity to simulate the intricate physio- and patho-conditions, and provide diagnostic platforms, microfluidic chips have fundamentally transformed the knowledge and management of female reproductive health, which will ultimately promote the development of more effective assisted reproductive technologies, treatments, and drug screening approaches. This review elucidates diverse microfluidic systems in mimicking the ovary, fallopian tube, uterus, placenta and cervix, and we delve into the culture of follicles and oocytes, gametes' manipulation, cryopreservation, and permeability especially. We investigate the role of microfluidics in endometriosis and hysteromyoma, and explore their applications in ovarian cancer, endometrial cancer and cervical cancer. At last, the current status of assisted reproductive technology and integrated microfluidic devices are introduced briefly. Through delineating the multifarious advantages and challenges of the microfluidic technology, we chart a definitive course for future research in the woman health field. As the microfluidic technology continues to evolve and advance, it holds great promise for revolutionizing the diagnosis and treatment of female reproductive health issues, thus propelling us into a future where we can ultimately optimize the overall wellbeing and health of women everywhere.
Collapse
Affiliation(s)
- Jinfeng Yan
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Tong Wu
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Jinjin Zhang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Yueyue Gao
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China
| | - Jia-Min Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China.
| | - Shixuan Wang
- National Clinical Research Center for Obstetrical and Gynecological Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Cancer Invasion and Metastasis, Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095, Jiefang Avenue, Wuhan, 430030, China.
| |
Collapse
|
9
|
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
|
10
|
Mulder PP, Vlig M, Elgersma A, Rozemeijer L, Mastenbroek LS, Middelkoop E, Joosten I, Koenen HJ, Boekema BK. Monocytes and T cells incorporated in full skin equivalents to study innate or adaptive immune reactions after burn injury. Front Immunol 2023; 14:1264716. [PMID: 37901218 PMCID: PMC10611519 DOI: 10.3389/fimmu.2023.1264716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 10/03/2023] [Indexed: 10/31/2023] Open
Abstract
Introduction Thermal injury often leads to prolonged and excessive inflammation, which hinders the recovery of patients. There is a notable absence of suitable animal-free models for investigating the inflammatory processes following burn injuries, thereby impeding the development of more effective therapies to improve burn wound healing in patients. Methods In this study, we established a human full skin equivalent (FSE) burn wound model and incorporated human peripheral blood-derived monocytes and T cells. Results Upon infiltration into the FSEs, the monocytes differentiated into macrophages within a span of 7 days. Burn-injured FSEs exhibited macrophages with increased expression of HLA-DR+ and elevated production of IL-8 (CXCL8), in comparison to uninjured FSEs. Among the T cells that actively migrated into the FSEs, the majority were CD4+ and CD25+. These T cells demonstrated augmented expression of markers associated with regulatory T cell, Th1, or Th17 activity, which coincided with significant heightened cytokine production, including IFN-γ, IL-4, IL-6, IL-8, IL-10, IL-12p70, IL-17A, IP-10 (CXCL10), and TGF-β1. Burn injury did not impact the studied effector T cell subsets or cytokine levels. Discussion Collectively, this study represents a significant advancement in the development of an immunocompetent human skin model, specifically tailored for investigating burn-induced innate or adaptive immune reactions at the site of burn injury.
Collapse
Affiliation(s)
- Patrick P.G. Mulder
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Marcel Vlig
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
| | - Anouk Elgersma
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
| | - Lotte Rozemeijer
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
| | | | - Esther Middelkoop
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, Netherlands
- Tissue Function and Regeneration, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Irma Joosten
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Hans J.P.M. Koenen
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Bouke K.H.L. Boekema
- Preclinical Research, Association of Dutch Burn Centres (ADBC), Beverwijk, Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, VU University Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
11
|
Attiogbe E, Larochelle S, Chaib Y, Mainzer C, Mauroux A, Bordes S, Closs B, Gilbert C, Moulin VJ. An in vitro autologous, vascularized, and immunocompetent Tissue Engineered Skin model obtained by the self-assembled approach. Acta Biomater 2023; 168:361-371. [PMID: 37419164 DOI: 10.1016/j.actbio.2023.06.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/09/2023]
Abstract
A complete in vitro skin model, containing resident cell types is needed to understand physiology and to consider the role of immune and endothelial cells in dermal drug testing. In this study, a cell extraction technique was developed to isolate resident skin cells from the same human donor while preserving the immune and endothelial cells. Then those cells were used to reconstruct an autologous, vascularized, and immunocompetent Tissue-Engineered Skin model, aviTES. Phenotypic characterization of the viable cells was performed on freshly isolated cells and after thawing through flow cytometry. Dermal cell extracts were characterized as fibroblasts, endothelial and immune cells, and the average amount of each cell type represents 4, 0.5, and 1 million viable cells per g of the dermis, respectively. The 3D models, TES and aviTES, were characterized by a fully differentiated epidermis that showed an increase in the presence of Ki67+ cells in the basolateral layer of the aviTES model. Capillary-like network formation, through the self-assembly of endothelial cells, and the presence of functional immune cells were identified through immunofluorescence staining in aviTES. In addition, the aviTES model was immunocompetent, as evidenced by its capacity to increase the production of pro-inflammatory cytokines TNF-α, MIP-1α, and GM-CSF following LPS stimulation. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate resident skin cell interactions and drug development. STATEMENT OF SIGNIFICANCE: There is an urgent need for a complete in vitro skin model containing the resident cell types to better understand the role of immune and endothelial cells in skin and to be able to use it for drug testing. Actual 3D models of human skin most often contain only fibroblasts and keratinocytes with a limited number of models containing endothelial cells or a limited variety of immune cells. This study describes an autologous skin model containing a functional resident skin immune system and a capillary network. It provides a relevant tool to study the contribution of the immune system to skin diseases and inflammatory responses and to investigate interactions between resident skin cell, improving our capacity to develop new drugs.
Collapse
Affiliation(s)
- Emilie Attiogbe
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Sébastien Larochelle
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Yanis Chaib
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | | | | | | | | | - Caroline Gilbert
- Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Véronique J Moulin
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval (LOEX), Québec, QC, Canada; Centre de Recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
12
|
Frisch E, Clavier L, Belhamdi A, Vrana NE, Lavalle P, Frisch B, Heurtault B, Gribova V. Preclinical in vitro evaluation of implantable materials: conventional approaches, new models and future directions. Front Bioeng Biotechnol 2023; 11:1193204. [PMID: 37576997 PMCID: PMC10416115 DOI: 10.3389/fbioe.2023.1193204] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 07/12/2023] [Indexed: 08/15/2023] Open
Abstract
Nowadays, implants and prostheses are widely used to repair damaged tissues or to treat different diseases, but their use is associated with the risk of infection, inflammation and finally rejection. To address these issues, new antimicrobial and anti-inflammatory materials are being developed. Aforementioned materials require their thorough preclinical testing before clinical applications can be envisaged. Although many researchers are currently working on new in vitro tissues for drug screening and tissue replacement, in vitro models for evaluation of new biomaterials are just emerging and are extremely rare. In this context, there is an increased need for advanced in vitro models, which would best recapitulate the in vivo environment, limiting animal experimentation and adapted to the multitude of these materials. Here, we overview currently available preclinical methods and models for biological in vitro evaluation of new biomaterials. We describe several biological tests used in biocompatibility assessment, which is a primordial step in new material's development, and discuss existing challenges in this field. In the second part, the emphasis is made on the development of new 3D models and approaches for preclinical evaluation of biomaterials. The third part focuses on the main parameters to consider to achieve the optimal conditions for evaluating biocompatibility; we also overview differences in regulations across different geographical regions and regulatory systems. Finally, we discuss future directions for the development of innovative biomaterial-related assays: in silico models, dynamic testing models, complex multicellular and multiple organ systems, as well as patient-specific personalized testing approaches.
Collapse
Affiliation(s)
- Emilie Frisch
- Université de Strasbourg, CNRS UMR 7199, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Strasbourg, France
| | - Lisa Clavier
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR_S 1121 Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| | | | | | - Philippe Lavalle
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR_S 1121 Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
- SPARTHA Medical, Strasbourg, France
| | - Benoît Frisch
- Université de Strasbourg, CNRS UMR 7199, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Strasbourg, France
| | - Béatrice Heurtault
- Université de Strasbourg, CNRS UMR 7199, 3Bio Team, Laboratoire de Conception et Application de Molécules Bioactives, Faculté de Pharmacie, Strasbourg, France
| | - Varvara Gribova
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR_S 1121 Biomaterials and Bioengineering, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
- Université de Strasbourg, Faculté de Chirurgie Dentaire, Strasbourg, France
| |
Collapse
|
13
|
Hofmann E, Fink J, Pignet AL, Schwarz A, Schellnegger M, Nischwitz SP, Holzer-Geissler JCJ, Kamolz LP, Kotzbeck P. Human In Vitro Skin Models for Wound Healing and Wound Healing Disorders. Biomedicines 2023; 11:biomedicines11041056. [PMID: 37189674 DOI: 10.3390/biomedicines11041056] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023] Open
Abstract
Skin wound healing is essential to health and survival. Consequently, high amounts of research effort have been put into investigating the cellular and molecular components involved in the wound healing process. The use of animal experiments has contributed greatly to the knowledge of wound healing, skin diseases, and the exploration of treatment options. However, in addition to ethical concerns, anatomical and physiological inter-species differences often influence the translatability of animal-based studies. Human in vitro skin models, which include essential cellular and structural components for wound healing analyses, would improve the translatability of results and reduce animal experiments during the preclinical evaluation of novel therapy approaches. In this review, we summarize in vitro approaches, which are used to study wound healing as well as wound healing-pathologies such as chronic wounds, keloids, and hypertrophic scars in a human setting.
Collapse
Affiliation(s)
- Elisabeth Hofmann
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Julia Fink
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna-Lisa Pignet
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna Schwarz
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Marlies Schellnegger
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Sebastian P Nischwitz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Judith C J Holzer-Geissler
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Kotzbeck
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
14
|
Hofmann E, Schwarz A, Fink J, Kamolz LP, Kotzbeck P. Modelling the Complexity of Human Skin In Vitro. Biomedicines 2023; 11:biomedicines11030794. [PMID: 36979772 PMCID: PMC10045055 DOI: 10.3390/biomedicines11030794] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/08/2023] Open
Abstract
The skin serves as an important barrier protecting the body from physical, chemical and pathogenic hazards as well as regulating the bi-directional transport of water, ions and nutrients. In order to improve the knowledge on skin structure and function as well as on skin diseases, animal experiments are often employed, but anatomical as well as physiological interspecies differences may result in poor translatability of animal-based data to the clinical situation. In vitro models, such as human reconstructed epidermis or full skin equivalents, are valuable alternatives to animal experiments. Enormous advances have been achieved in establishing skin models of increasing complexity in the past. In this review, human skin structures are described as well as the fast evolving technologies developed to reconstruct the complexity of human skin structures in vitro.
Collapse
Affiliation(s)
- Elisabeth Hofmann
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna Schwarz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Julia Fink
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Kotzbeck
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Correspondence:
| |
Collapse
|
15
|
Liu T, Zhang J, Lu B, Wang H, Zhan J, Tan X, Wu C, Liu S, Wang Z, Zhang J, Zhang J. Highly efficient conotoxin delivery enabled by a bio-derived ionic liquid. J Mol Liq 2022. [DOI: 10.1016/j.molliq.2022.120529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
16
|
Urciuolo F, Passariello R, Imparato G, Casale C, Netti PA. Bioengineered Wound Healing Skin Models: The Role of Immune Response and Endogenous ECM to Fully Replicate the Dynamic of Scar Tissue Formation In Vitro. Bioengineering (Basel) 2022; 9:233. [PMID: 35735476 PMCID: PMC9219817 DOI: 10.3390/bioengineering9060233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/24/2022] [Accepted: 05/25/2022] [Indexed: 12/27/2022] Open
Abstract
The healing of deep skin wounds is a complex phenomenon evolving according with a fine spatiotemporal regulation of different biological events (hemostasis, inflammation, proliferation, remodeling). Due to the spontaneous evolution of damaged human dermis toward a fibrotic scar, the treatment of deep wounds still represents a clinical concern. Bioengineered full-thickness skin models may play a crucial role in this direction by providing a deep understanding of the process that leads to the formation of fibrotic scars. This will allow (i) to identify new drugs and targets/biomarkers, (ii) to test new therapeutic approaches, and (iii) to develop more accurate in silico models, with the final aim to guide the closure process toward a scar-free closure and, in a more general sense, (iv) to understand the mechanisms involved in the intrinsic and extrinsic aging of the skin. In this work, the complex dynamic of events underlaying the closure of deep skin wound is presented and the engineered models that aim at replicating such complex phenomenon are reviewed. Despite the complexity of the cellular and extracellular events occurring during the skin wound healing the gold standard assay used to replicate such a process is still represented by planar in vitro models that have been largely used to identify the key factors regulating the involved cellular processes. However, the lack of the main constituents of the extracellular matrix (ECM) makes these over-simplistic 2D models unable to predict the complexity of the closure process. Three-dimensional bioengineered models, which aim at recreating the closure dynamics of the human dermis by using exogenous biomaterials, have been developed to fill such a gap. Although interesting mechanistic effects have been figured out, the effect of the inflammatory response on the ECM remodelling is not replicated yet. We discuss how more faithful wound healing models can be obtained by creating immunocompetent 3D dermis models featuring an endogenous ECM.
Collapse
Affiliation(s)
- Francesco Urciuolo
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; (C.C.); (P.A.N.)
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy;
| | - Roberta Passariello
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy;
- Center for Advanced Biomaterials for HealthCare@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy;
| | - Giorgia Imparato
- Center for Advanced Biomaterials for HealthCare@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy;
| | - Costantino Casale
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; (C.C.); (P.A.N.)
| | - Paolo Antonio Netti
- Interdisciplinary Research Centre on Biomaterials (CRIB), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy; (C.C.); (P.A.N.)
- Department of Chemical, Materials and Industrial Production Engineering (DICMAPI), University of Naples Federico II, P.le Tecchio 80, 80125 Naples, Italy;
- Center for Advanced Biomaterials for HealthCare@CRIB Istituto Italiano di Tecnologia, Largo Barsanti e Matteucci 53, 80125 Naples, Italy;
| |
Collapse
|
17
|
Caipa Garcia AL, Arlt VM, Phillips DH. Organoids for toxicology and genetic toxicology: applications with drugs and prospects for environmental carcinogenesis. Mutagenesis 2022; 37:143-154. [PMID: 34147034 PMCID: PMC9071088 DOI: 10.1093/mutage/geab023] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 06/17/2021] [Indexed: 12/19/2022] Open
Abstract
Advances in three-dimensional (3D) cell culture technology have led to the development of more biologically and physiologically relevant models to study organ development, disease, toxicology and drug screening. Organoids have been derived from many mammalian tissues, both normal and tumour, from adult stem cells and from pluripotent stem cells. Tissue organoids can retain many of the cell types and much of the structure and function of the organ of origin. Organoids derived from pluripotent stem cells display increased complexity compared with organoids derived from adult stem cells. It has been shown that organoids express many functional xenobiotic-metabolising enzymes including cytochrome P450s (CYPs). This has benefitted the drug development field in facilitating pre-clinical testing of more personalised treatments and in developing large toxicity and efficacy screens for a range of compounds. In the field of environmental and genetic toxicology, treatment of organoids with various compounds has generated responses that are close to those obtained in primary tissues and in vivo models, demonstrating the biological relevance of these in vitro multicellular 3D systems. Toxicological investigations of compounds in different tissue organoids have produced promising results indicating that organoids will refine future studies on the effects of environmental exposures and carcinogenic risk to humans. With further development and standardised procedures, advancing our understanding on the metabolic capabilities of organoids will help to validate their use to investigate the modes of action of environmental carcinogens.
Collapse
Affiliation(s)
- Angela L Caipa Garcia
- Department of Analytical, Environmental and Forensic Sciences, School of Population Health and Environmental Sciences, King’s College London, London, SE1 9NH, UK
| | - Volker M Arlt
- Department of Analytical, Environmental and Forensic Sciences, School of Population Health and Environmental Sciences, King’s College London, London, SE1 9NH, UK
| | - David H Phillips
- Department of Analytical, Environmental and Forensic Sciences, School of Population Health and Environmental Sciences, King’s College London, London, SE1 9NH, UK
| |
Collapse
|
18
|
Papi M, Pozzi D, Palmieri V, Caracciolo G. Principles for optimization and validation of mRNA lipid nanoparticle vaccines against COVID-19 using 3D bioprinting. NANO TODAY 2022; 43:101403. [PMID: 35079274 PMCID: PMC8776405 DOI: 10.1016/j.nantod.2022.101403] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/10/2022] [Accepted: 01/19/2022] [Indexed: 05/03/2023]
Abstract
BioNTech/Pfizer's Comirnaty and Moderna's SpikeVax vaccines consist in mRNA encapsulated in lipid nanoparticles (LNPs). The modularity of the delivery platform and the manufacturing possibilities provided by microfluidics let them look like an instant success, but they are the product of decades of intense research. There is a multitude of considerations to be made when designing an optimal mRNA-LNPs vaccine. Herein, we provide a brief overview of what is presently known and what still requires investigation to optimize mRNA LNPs vaccines. Lastly, we give our perspective on the engineering of 3D bioprinted validation systems that will allow faster, cheaper, and more predictive vaccine testing in the future compared with animal models.
Collapse
Affiliation(s)
- Massimiliano Papi
- Department of Neuroscience, Catholic University of Sacred Heart, L.go Francesco Vito 1, 00168 Rome, Italy
| | - Daniela Pozzi
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| | - Valentina Palmieri
- Institute for Complex Systems, National Research Council of Italy, Via dei Taurini 19, 00185 Rome, Italy
| | - Giulio Caracciolo
- Department of Molecular Medicine, Sapienza University of Rome, Viale Regina Elena 291, 00161 Rome, Italy
| |
Collapse
|
19
|
Antezana PE, Municoy S, Álvarez-Echazú MI, Santo-Orihuela PL, Catalano PN, Al-Tel TH, Kadumudi FB, Dolatshahi-Pirouz A, Orive G, Desimone MF. The 3D Bioprinted Scaffolds for Wound Healing. Pharmaceutics 2022; 14:464. [PMID: 35214197 PMCID: PMC8875365 DOI: 10.3390/pharmaceutics14020464] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/09/2022] [Accepted: 02/11/2022] [Indexed: 02/01/2023] Open
Abstract
Skin tissue engineering and regeneration aim at repairing defective skin injuries and progress in wound healing. Until now, even though several developments are made in this field, it is still challenging to face the complexity of the tissue with current methods of fabrication. In this review, short, state-of-the-art on developments made in skin tissue engineering using 3D bioprinting as a new tool are described. The current bioprinting methods and a summary of bioink formulations, parameters, and properties are discussed. Finally, a representative number of examples and advances made in the field together with limitations and future needs are provided.
Collapse
Affiliation(s)
- Pablo Edmundo Antezana
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| | - Sofia Municoy
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| | - María Inés Álvarez-Echazú
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| | - Pablo Luis Santo-Orihuela
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
- Centro de Investigaciones en Plagas e Insecticidas (CIPEIN), Instituto de Investigaciones Científicas y Técnicas para la Defensa CITEDEF/UNIDEF, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina (CONICET), Juan B. de La Salle 4397, Villa Martelli, Buenos Aires 1603, Argentina
| | - Paolo Nicolás Catalano
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
- Departamento de Micro y Nanotecnología, Instituto de Nanociencia y Nanotecnología, CNEA-CONICET, Av. General Paz 1499, San Martín 1650, Argentina
| | - Taleb H Al-Tel
- Sharjah Institute for Medical Research and College of Pharmacy, University of Sharjah, Sharjah P.O. Box 27272, United Arab Emirates
| | - Firoz Babu Kadumudi
- Department of Health Technology, Technical University of Denmark, 2800 Kongens Lyngby, Denmark
| | | | - Gorka Orive
- Laboratory of Pharmaceutics, NanoBioCel Group, School of Pharmacy, University of the Basque Country UPV/EHU, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain
- Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
- Bioaraba, NanoBioCel Research Group, 01006 Vitoria-Gasteiz, Spain
- University Institute for Regenerative Medicine and Oral Implantology-UIRMI (UPV/EHU-Fundación Eduardo Anitua), 01007 Vitoria-Gasteiz, Spain
- Singapore Eye Research Institute, The Academia, 20 College Road, Discovery Tower, Singapore 169856, Singapore
| | - Martin Federico Desimone
- Facultad de Farmacia y Bioquímica, Instituto de Química y Metabolismo del Fármaco (IQUIMEFA), Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Junín 956, Buenos Aires 1113, Argentina
| |
Collapse
|
20
|
Bioengineered Efficacy Models of Skin Disease: Advances in the Last 10 Years. Pharmaceutics 2022; 14:pharmaceutics14020319. [PMID: 35214050 PMCID: PMC8877988 DOI: 10.3390/pharmaceutics14020319] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/24/2021] [Accepted: 01/25/2022] [Indexed: 12/19/2022] Open
Abstract
Models of skin diseases, such as psoriasis and scleroderma, must accurately recapitulate the complex microenvironment of human skin to provide an efficacious platform for investigation of skin diseases. Skin disease research has been shifting from less complex and less relevant 2D (two-dimensional) models to significantly more relevant 3D (three-dimensional) models. Three-dimensional modeling systems are better able to recapitulate the complex cell–cell and cell–matrix interactions that occur in vivo within skin. Three-dimensional human skin equivalents (HSEs) have emerged as an advantageous tool for the study of skin disease in vitro. These 3D HSEs can be highly complex, containing both epidermal and dermal compartments with integrated adnexal structures. The addition of adnexal structures to 3D HSEs has allowed researchers to gain more insight into the complex pathology of various hereditary and acquired skin diseases. One method of constructing 3D HSEs, 3D bioprinting, has emerged as a versatile and useful tool for generating highly complex HSEs. The development of commercially available 3D bioprinters has allowed researchers to create highly reproducible 3D HSEs with precise integration of multiple adnexal structures. While the field of bioengineered models for study of skin disease has made tremendous progress in the last decade, there are still significant efforts necessary to create truly biomimetic skin disease models. In future studies utilizing 3D HSEs, emphasis must be placed on integrating all adnexal structures relevant to the skin disease under investigation. Thorough investigation of the intricate pathology of skin diseases and the development of effective treatments requires use of highly efficacious models of skin diseases.
Collapse
|
21
|
Varga-Medveczky Z, Kocsis D, Naszlady MB, Fónagy K, Erdő F. Skin-on-a-Chip Technology for Testing Transdermal Drug Delivery-Starting Points and Recent Developments. Pharmaceutics 2021; 13:1852. [PMID: 34834264 PMCID: PMC8619496 DOI: 10.3390/pharmaceutics13111852] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 01/22/2023] Open
Abstract
During the last decades, several technologies were developed for testing drug delivery through the dermal barrier. Investigation of drug penetration across the skin can be important in topical pharmaceutical formulations and also in cosmeto-science. The state-of- the-art in the field of skin diffusion measurements, different devices, and diffusion platforms used, are summarized in the introductory part of this review. Then the methodologies applied at Pázmány Péter Catholic University are shown in detail. The main testing platforms (Franz diffusion cells, skin-on-a-chip devices) and the major scientific projects (P-glycoprotein interaction in the skin; new skin equivalents for diffusion purposes) are also presented in one section. The main achievements of our research are briefly summarized: (1) new skin-on-a-chip microfluidic devices were validated as tools for drug penetration studies for the skin; (2) P-glycoprotein transport has an absorptive orientation in the skin; (3) skin samples cannot be used for transporter interaction studies after freezing and thawing; (4) penetration of hydrophilic model drugs is lower in aged than in young skin; (5) mechanical sensitization is needed for excised rodent and pig skins for drug absorption measurements. Our validated skin-on-a-chip platform is available for other research groups to use for testing and for utilizing it for different purposes.
Collapse
Affiliation(s)
| | | | | | | | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Práter u. 50a, H-1083 Budapest, Hungary; (Z.V.-M.); (D.K.); (M.B.N.); (K.F.)
| |
Collapse
|
22
|
Cryomicroneedles for transdermal cell delivery. Nat Biomed Eng 2021; 5:1008-1018. [PMID: 33941895 DOI: 10.1038/s41551-021-00720-1] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2018] [Accepted: 03/26/2021] [Indexed: 02/01/2023]
Abstract
Cell therapies for the treatment of skin disorders could benefit from simple, safe and efficient technology for the transdermal delivery of therapeutic cells. Conventional cell delivery by hypodermic-needle injection is associated with poor patient compliance, requires trained personnel, generates waste and has non-negligible risks of injury and infection. Here, we report the design and proof-of-concept application of cryogenic microneedle patches for the transdermal delivery of living cells. The microneedles are fabricated by stepwise cryogenic micromoulding of cryogenic medium with pre-suspended cells, and can be easily inserted into porcine skin and dissolve after deployment of the cells. In mice, cells delivered by the cryomicroneedles retained their viability and proliferative capability. In mice with subcutaneous melanoma tumours, the delivery of ovalbumin-pulsed dendritic cells via the cryomicroneedles elicited higher antigen-specific immune responses and led to slower tumour growth than intravenous and subcutaneous injections of the cells. Biocompatible cryomicroneedles may facilitate minimally invasive cell delivery for a range of cell therapies.
Collapse
|
23
|
Boero E, Mnich ME, Manetti AGO, Soldaini E, Grimaldi L, Bagnoli F. Human Three-Dimensional Models for Studying Skin Pathogens. Curr Top Microbiol Immunol 2021; 430:3-27. [PMID: 32601967 DOI: 10.1007/82_2020_219] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Skin is the most exposed surface of the human body, separating the microbe-rich external environment, from the sterile inner part. When skin is breached or its homeostasis is perturbed, bacterial, fungal and viral pathogens can cause local infections or use the skin as an entry site to spread to other organs. In the last decades, it has become clear that skin provides niches for permanent microbial colonization, and it actively interacts with microorganisms. This crosstalk promotes skin homeostasis and immune maturation, preventing expansion of harmful organisms. Skin commensals, however, are often found to be skin most prevalent and dangerous pathogens. Despite the medical interest, mechanisms of colonization and invasion for most skin pathogens are poorly understood. This limitation is due to the lack of reliable skin models. Indeed, animal models do not adequately mimic neither the anatomy nor the immune response of human skin. Human 3D skin models overcome these limitations and can provide new insights into the molecular mechanisms of microbial pathogenesis. Herein, we address the strengths and weaknesses of different types of human skin models and we review the main findings obtained using these models to study skin pathogens.
Collapse
Affiliation(s)
| | | | | | | | - Luca Grimaldi
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | | |
Collapse
|
24
|
Fritsche E, Haarmann-Stemmann T, Kapr J, Galanjuk S, Hartmann J, Mertens PR, Kämpfer AAM, Schins RPF, Tigges J, Koch K. Stem Cells for Next Level Toxicity Testing in the 21st Century. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2006252. [PMID: 33354870 DOI: 10.1002/smll.202006252] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/13/2020] [Indexed: 06/12/2023]
Abstract
The call for a paradigm change in toxicology from the United States National Research Council in 2007 initiates awareness for the invention and use of human-relevant alternative methods for toxicological hazard assessment. Simple 2D in vitro systems may serve as first screening tools, however, recent developments infer the need for more complex, multicellular organotypic models, which are superior in mimicking the complexity of human organs. In this review article most critical organs for toxicity assessment, i.e., skin, brain, thyroid system, lung, heart, liver, kidney, and intestine are discussed with regards to their functions in health and disease. Embracing the manifold modes-of-action how xenobiotic compounds can interfere with physiological organ functions and cause toxicity, the need for translation of such multifaceted organ features into the dish seems obvious. Currently used in vitro methods for toxicological applications and ongoing developments not yet arrived in toxicity testing are discussed, especially highlighting the potential of models based on embryonic stem cells and induced pluripotent stem cells of human origin. Finally, the application of innovative technologies like organs-on-a-chip and genome editing point toward a toxicological paradigm change moves into action.
Collapse
Affiliation(s)
- Ellen Fritsche
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
- Medical Faculty, Heinrich-Heine University Düsseldorf, Düsseldorf, 40225, Germany
| | | | - Julia Kapr
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Saskia Galanjuk
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Hartmann
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Peter R Mertens
- Department of Nephrology and Hypertension, Diabetes and Endocrinology, Otto-von-Guericke-University Magdeburg, Magdeburg, 39106, Germany
| | - Angela A M Kämpfer
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Roel P F Schins
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Julia Tigges
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| | - Katharina Koch
- IUF - Leibniz Research Institute for Environmental Medicine, Düsseldorf, 40225, Germany
| |
Collapse
|
25
|
Griffoni C, Neidhart B, Yang K, Groeber-Becker F, Maniura-Weber K, Dandekar T, Walles H, Rottmar M. In vitro skin culture media influence the viability and inflammatory response of primary macrophages. Sci Rep 2021; 11:7070. [PMID: 33782484 PMCID: PMC8007571 DOI: 10.1038/s41598-021-86486-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 03/16/2021] [Indexed: 11/09/2022] Open
Abstract
The replacement of animal models for investigation of inflammation and wound healing has been advancing by means of in vitro skin equivalents with increasing levels of complexity. However, the current in vitro skin models still have a limited pre-clinical relevance due to their lack of immune cells. So far, few steps have been made towards the incorporation of immune cells into in vitro skin and the requirements for immunocompetent co-cultures remain unexplored. To establish suitable conditions for incorporating macrophages into skin models, we evaluated the effects of different media on primary keratinocytes, fibroblasts and macrophages. Skin maturation was affected by culture in macrophage medium, while macrophages showed reduced viability, altered cell morphology and decreased response to pro- and anti-inflammatory stimuli in skin differentiation media, both in 2D and 3D. The results indicate that immunocompetent skin models have specific, complex requirements for supporting an accurate detection of immune responses, which point at the identification of a suitable culture medium as a crucial pre-requisite for the development of physiologically relevant models.
Collapse
Affiliation(s)
- Chiara Griffoni
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland.,Department Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Berna Neidhart
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Ke Yang
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Florian Groeber-Becker
- Department Tissue Engineering & Regenerative Medicine, University Hospital Würzburg, Würzburg, Germany.,Translational Center for Regenerative Therapies, Fraunhofer-Institute for Silicate Research ISC, Würzburg, Germany
| | - Katharina Maniura-Weber
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland
| | - Thomas Dandekar
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
| | - Heike Walles
- Translational Center for Regenerative Therapies, Fraunhofer-Institute for Silicate Research ISC, Würzburg, Germany.,Core Facility Tissue Engineering, Otto-Von-Guericke-University, Magdeburg, Germany
| | - Markus Rottmar
- Laboratory for Biointerfaces, Empa - Swiss Federal Laboratories for Materials Science and Technology, St. Gallen, Switzerland.
| |
Collapse
|
26
|
Ejiugwo M, Rochev Y, Gethin G, O'Connor G. Toward Developing Immunocompetent Diabetic Foot Ulcer-on-a-Chip Models for Drug Testing. Tissue Eng Part C Methods 2021; 27:77-88. [PMID: 33406980 DOI: 10.1089/ten.tec.2020.0331] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Bioengineering of skin has been significantly explored, ranging from the use of traditional cell culture systems to the most recent organ-on-a-chip (OoC) technology that permits skin modeling on physiological scales among other benefits. This article presents key considerations for developing physiologically relevant immunocompetent diabetic foot ulcer (DFU) models. Diabetic foot ulceration affects hundreds of millions of individuals globally, especially the elderly, and constitutes a major socioeconomic burden. When DFUs are not treated and managed in a timely manner, 15-50% of patients tend to undergo partial or complete amputation of the affected limb. Consequently, at least 40% of such patients die within 5 years postamputation. Currently, therapeutic strategies are actively sought and developed. However, present-day preclinical platforms (animals and in vitro models) are not robust enough to provide reliable data for clinical trials. Insights from published works on immunocompetent skin-on-a-chip models and bioengineering considerations, presented in this article, can inform researchers on how to develop robust OoC models for testing topical therapies such as growth factor-based therapies for DFUs. We propose that immunocompetent DFU-on-a-chip models should be bioengineered using diseased cells derived from individuals; in particular, the pathophysiological contribution of macrophages in diabetic wound healing, along with the typical fibroblasts and keratinocytes, needs to be recapitulated. The ideal model should consist of the following components: diseased cells embedded in reproducible scaffolds, which permit endogenous "diseased" extracellular matrix deposition, and the integration of the derived immunocompetent DFU model onto a microfluidic platform. The proposed DFU platforms will eventually facilitate reliable and robust drug testing of wound healing therapeutics, coupled with reduced clinical trial failure rates. Impact statement Current animal and cell-based systems are not physiologically relevant enough to retrieve reliable results for clinical translation of diabetic foot ulcer (DFU) therapies. Organ-on-a-chip (OoC) technology offers desirable features that could finally enable the vision of modeling DFU for pathophysiological studies and drug testing at a microscale. This article brings together the significant recent findings relevant to developing a minimally functional immunocompetent DFU-on-a-chip model, as wound healing cannot occur without a proper functioning immune response. It looks feasible in the future to recapitulate the stagnant inflammation in DFU (thought to impede wound healing) using OoC, diseased cells, and an endogenously produced extracellular matrix.
Collapse
Affiliation(s)
- Mirella Ejiugwo
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Physics, and National University of Ireland Galway, Galway City, Ireland
| | - Yury Rochev
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Physics, and National University of Ireland Galway, Galway City, Ireland
| | - Georgina Gethin
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Nursing and Midwifery, National University of Ireland Galway, Galway City, Ireland
| | - Gerard O'Connor
- SFI CÚRAM Centre for Research in Medical Devices, National University of Ireland Galway, Galway City, Ireland.,School of Physics, and National University of Ireland Galway, Galway City, Ireland
| |
Collapse
|
27
|
Transcriptome Profiling Analyses in Psoriasis: A Dynamic Contribution of Keratinocytes to the Pathogenesis. Genes (Basel) 2020; 11:genes11101155. [PMID: 33007857 PMCID: PMC7600703 DOI: 10.3390/genes11101155] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/28/2020] [Accepted: 09/29/2020] [Indexed: 02/08/2023] Open
Abstract
Psoriasis is an immune-mediated inflammatory skin disease with a complex etiology involving environmental and genetic factors. A better insight into related genomic alteration helps design precise therapies leading to better treatment outcome. Gene expression in psoriasis can provide relevant information about the altered expression of mRNA transcripts, thus giving new insights into the disease onset. Techniques for transcriptome analyses, such as microarray and RNA sequencing (RNA-seq), are relevant tools for the discovery of new biomarkers as well as new therapeutic targets. This review summarizes the findings related to the contribution of keratinocytes in the pathogenesis of psoriasis by an in-depth review of studies that have examined psoriatic transcriptomes in the past years. It also provides valuable information on reconstructed 3D psoriatic skin models using cells isolated from psoriatic patients for transcriptomic studies.
Collapse
|
28
|
Bédard P, Gauvin S, Ferland K, Caneparo C, Pellerin È, Chabaud S, Bolduc S. Innovative Human Three-Dimensional Tissue-Engineered Models as an Alternative to Animal Testing. Bioengineering (Basel) 2020; 7:E115. [PMID: 32957528 PMCID: PMC7552665 DOI: 10.3390/bioengineering7030115] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Revised: 09/11/2020] [Accepted: 09/15/2020] [Indexed: 12/12/2022] Open
Abstract
Animal testing has long been used in science to study complex biological phenomena that cannot be investigated using two-dimensional cell cultures in plastic dishes. With time, it appeared that more differences could exist between animal models and even more when translated to human patients. Innovative models became essential to develop more accurate knowledge. Tissue engineering provides some of those models, but it mostly relies on the use of prefabricated scaffolds on which cells are seeded. The self-assembly protocol has recently produced organ-specific human-derived three-dimensional models without the need for exogenous material. This strategy will help to achieve the 3R principles.
Collapse
Affiliation(s)
- Patrick Bédard
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Sara Gauvin
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Karel Ferland
- Faculté de Médecine, Sciences Biomédicales, Université Laval, Québec, QC G1V 0A6, Canada; (P.B.); (S.G.); (K.F.)
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Christophe Caneparo
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Ève Pellerin
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Chabaud
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
| | - Stéphane Bolduc
- Centre de Recherche en Organogénèse Expérimentale de l’Université Laval/LOEX, Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC G1J 1Z4, Canada; (C.C.); (È.P.); (S.C.)
- Département de Chirurgie, Faculté de Médecine, Université Laval, Québec, QC G1V 0A6, Canada
| |
Collapse
|
29
|
Emmert H, Rademacher F, Gläser R, Harder J. Skin microbiota analysis in human 3D skin models-"Free your mice". Exp Dermatol 2020; 29:1133-1139. [PMID: 32748435 DOI: 10.1111/exd.14164] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 07/08/2020] [Accepted: 07/17/2020] [Indexed: 12/22/2022]
Abstract
In the May issue of Experimental Dermatology 2018, we published a review article focusing on human 3D skin models in the context of microbiota research. The principal intention was to provide an overview of present and future concepts to use skin models in microbiota analyses. With the present viewpoint, we would like to draw the reader's attention again to the use of human skin models in microbiota research with the aim to highlight the benefits and necessity of human skin models to analyse the human skin-microbiota interaction. This is accompanied by a critical view on mice models that often are not suitable to analyse the functional impact of the human skin microbiota. In addition, we present novel and future concepts highlighting the benefits of human 3D skin models in microbiota research.
Collapse
Affiliation(s)
- Hila Emmert
- Department of Dermatology, Kiel University, Kiel, Germany
| | | | - Regine Gläser
- Department of Dermatology, Kiel University, Kiel, Germany
| | - Jürgen Harder
- Department of Dermatology, Kiel University, Kiel, Germany
| |
Collapse
|
30
|
Kaiser L, Weinschrott H, Quint I, Blaess M, Csuk R, Jung M, Kohl M, Deigner HP. Metabolite Patterns in Human Myeloid Hematopoiesis Result from Lineage-Dependent Active Metabolic Pathways. Int J Mol Sci 2020; 21:ijms21176092. [PMID: 32847028 PMCID: PMC7504406 DOI: 10.3390/ijms21176092] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 12/18/2022] Open
Abstract
Assessment of hematotoxicity from environmental or xenobiotic compounds is of notable interest and is frequently assessed via the colony forming unit (CFU) assay. Identification of the mode of action of single compounds is of further interest, as this often enables transfer of results across different tissues and compounds. Metabolomics displays one promising approach for such identification, nevertheless, suitability with current protocols is restricted. Here, we combined a hematopoietic stem and progenitor cell (HSPC) expansion approach with distinct lineage differentiations, resulting in formation of erythrocytes, dendritic cells and neutrophils. We examined the unique combination of pathway activity in glycolysis, glutaminolysis, polyamine synthesis, fatty acid oxidation and synthesis, as well as glycerophospholipid and sphingolipid metabolism. We further assessed their interconnections and essentialness for each lineage formation. By this, we provide further insights into active metabolic pathways during the differentiation of HSPC into different lineages, enabling profound understanding of possible metabolic changes in each lineage caused by exogenous compounds.
Collapse
Affiliation(s)
- Lars Kaiser
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 Villingen-Schwenningen, Germany; (L.K.); (H.W.); (I.Q.); (M.B.); (M.K.)
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, 79104 Freiburg i. Br., Germany;
| | - Helga Weinschrott
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 Villingen-Schwenningen, Germany; (L.K.); (H.W.); (I.Q.); (M.B.); (M.K.)
| | - Isabel Quint
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 Villingen-Schwenningen, Germany; (L.K.); (H.W.); (I.Q.); (M.B.); (M.K.)
| | - Markus Blaess
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 Villingen-Schwenningen, Germany; (L.K.); (H.W.); (I.Q.); (M.B.); (M.K.)
| | - René Csuk
- Organic Chemistry, Martin-Luther-University Halle-Wittenberg, Kurt-Mothes-Str. 2, 06120 Halle (Saale), Germany;
| | - Manfred Jung
- Institute of Pharmaceutical Sciences, University of Freiburg, Albertstraße 25, 79104 Freiburg i. Br., Germany;
- CIBSS—Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104 Freiburg, Germany
| | - Matthias Kohl
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 Villingen-Schwenningen, Germany; (L.K.); (H.W.); (I.Q.); (M.B.); (M.K.)
| | - Hans-Peter Deigner
- Institute of Precision Medicine, Medical and Life Sciences Faculty, Furtwangen University, Jakob-Kienzle-Straße 17, 78054 Villingen-Schwenningen, Germany; (L.K.); (H.W.); (I.Q.); (M.B.); (M.K.)
- Fraunhofer Institute IZI, Leipzig, EXIM Department, Schillingallee 68, 18057 Rostock, Germany
- Associated member of Tuebingen University, Faculty of Science, Auf der Morgenstelle 8, 72076 Tübingen, Germany
- Correspondence: ; Tel.: +49-7720-307-4232
| |
Collapse
|
31
|
Dijkhoff IM, Drasler B, Karakocak BB, Petri-Fink A, Valacchi G, Eeman M, Rothen-Rutishauser B. Impact of airborne particulate matter on skin: a systematic review from epidemiology to in vitro studies. Part Fibre Toxicol 2020; 17:35. [PMID: 32711561 PMCID: PMC7382801 DOI: 10.1186/s12989-020-00366-y] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/14/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Air pollution is killing close to 5 million people a year, and harming billions more. Air pollution levels remain extremely high in many parts of the world, and air pollution-associated premature deaths have been reported for urbanized areas, particularly linked to the presence of airborne nano-sized and ultrafine particles. MAIN TEXT To date, most of the research studies did focus on the adverse effects of air pollution on the human cardiovascular and respiratory systems. Although the skin is in direct contact with air pollutants, their damaging effects on the skin are still under investigation. Epidemiological data suggested a correlation between exposure to air pollutants and aggravation of symptoms of chronic immunological skin diseases. In this study, a systematic literature review was conducted to understand the current knowledge on the effects of airborne particulate matter on human skin. It aims at providing a deeper understanding of the interactions between air pollutants and skin to further assess their potential risks for human health. CONCLUSION Particulate matter was shown to induce a skin barrier dysfunction and provoke the formation of reactive oxygen species through direct and indirect mechanisms, leading to oxidative stress and induced activation of the inflammatory cascade in human skin. Moreover, a positive correlation was reported between extrinsic aging and atopic eczema relative risk with increasing particulate matter exposure.
Collapse
Affiliation(s)
- Irini M Dijkhoff
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Barbara Drasler
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Bedia Begum Karakocak
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Alke Petri-Fink
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland
| | - Giuseppe Valacchi
- Department of Biomedical and Specialist Surgical Sciences, University of Ferrara, Ferrara, Italy
- Department of Animal Sciences, PHHI NCRC, North Carolina State University, Kannapolis, NC, USA
| | | | - Barbara Rothen-Rutishauser
- Adolphe Merkle Institute, University of Fribourg, Chemin des Verdiers 4, CH-1700, Fribourg, Switzerland.
| |
Collapse
|
32
|
Barros N, Kim H, Goudie MJ, Lee K, Bandaru P, Banton EA, Sarikhani E, Sun W, Zhang S, Cho HJ, Hartel MC, Ostrovidov S, Ahadian S, Hussain S, Ashammakhi N, Dokmeci MR, Herculano RD, Lee J, Khademhosseini A. Biofabrication of endothelial cell, dermal fibroblast, and multilayered keratinocyte layers for skin tissue engineering. Biofabrication 2020; 13. [PMID: 32650324 DOI: 10.1088/1758-5090/aba503] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/10/2020] [Indexed: 01/10/2023]
Abstract
The skin serves a substantial number of physiological purposes and is exposed to numerous biological and chemical agents owing to its large surface area and accessibility. Yet, current skin models are limited in emulating the multifaceted functions of skin tissues due to a lack of effort on the optimization of biomaterials and techniques at different skin layers for building skin frameworks. Here, we use biomaterial-based approaches and bioengineered techniques to develop a 3D skin model with layers of endothelial cell networks, dermal fibroblasts, and multilayered keratinocytes. Analysis of mechanical properties of gelatin methacryloyl (GelMA)-based bioinks mixed with different portions of alginate revealed bioprinted endothelium could be better modeled to optimize endothelial cell viability with a mixture of 7.5% GelMA and 2% alginate. Matrix stiffness plays a crucial role in modulating produced levels of Pro-Collagen I alpha-1 and matrix metalloproteinase-1 in human dermal fibroblasts and affecting their viability, proliferation, and spreading. Moreover, seeding human keratinocytes with gelatin-coating multiple times proves helpful in reducing culture time to create multilayered keratinocytes while maintaining their viability. The ability to fabricate selected biomaterials for each layer of skin tissues has implications in the biofabrication of skin systems for regenerative medicine and disease modeling.
Collapse
Affiliation(s)
- Natan Barros
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, UNITED STATES
| | - Hanjun Kim
- University of California Los Angeles, Los Angeles, California, 90095, UNITED STATES
| | - Marcus J Goudie
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, UNITED STATES
| | - KangJu Lee
- Bioengineering, UCLA, Los Angeles, California, UNITED STATES
| | - Praveen Bandaru
- Bioengineering, UCLA, Los Angeles, California, UNITED STATES
| | - Ethan A Banton
- Bioengineering, UCLA, Los Angeles, California, UNITED STATES
| | | | - Wujin Sun
- Bioengineering, UCLA, Los Angeles, California, UNITED STATES
| | - Shiming Zhang
- Bioengineering, UCLA, Los Angeles, California, UNITED STATES
| | - Hyun-Jong Cho
- Pharmacy, Kangwon National University, Chuncheon, Gangwon-do, Korea (the Republic of)
| | - Martin C Hartel
- Bioengineering, UCLA, Los Angeles, California, UNITED STATES
| | | | - Samad Ahadian
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, UNITED STATES
| | - Saber Hussain
- , Molecular Bioeffects Branch, Wright Patterson AFB, Ohio, UNITED STATES
| | - Nureddin Ashammakhi
- Center for Minimally Invasive Therapeutics (C-MIT), University of California - Los Angeles, Los Angeles, California, UNITED STATES
| | - Mehmet R Dokmeci
- Radiology, UCLA, CNSI 4528, 570 Westwood Plaza, Los Angeles, California, 90095, UNITED STATES
| | | | - Junmin Lee
- Terasaki Institute, Los Angeles, California, UNITED STATES
| | - Ali Khademhosseini
- Center for Minimally Invasive Therapeutics, UCLA, Los Angeles, California, UNITED STATES
| |
Collapse
|
33
|
Mnich ME, van Dalen R, van Sorge NM. C-Type Lectin Receptors in Host Defense Against Bacterial Pathogens. Front Cell Infect Microbiol 2020; 10:309. [PMID: 32733813 PMCID: PMC7358460 DOI: 10.3389/fcimb.2020.00309] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 05/22/2020] [Indexed: 12/11/2022] Open
Abstract
Antigen-presenting cells (APCs) are present throughout the human body—in tissues, at barrier sites and in the circulation. They are critical for processing external signals to instruct both local and systemic responses toward immune tolerance or immune defense. APCs express an extensive repertoire of pattern-recognition receptors (PRRs) to detect and transduce these signals. C-type lectin receptors (CLRs) comprise a subfamily of PRRs dedicated to sensing glycans, including those expressed by commensal and pathogenic bacteria. This review summarizes recent findings on the recognition of and responses to bacteria by membrane-expressed CLRs on different APC subsets, which are discussed according to the primary site of infection. Many CLR-bacterial interactions promote bacterial clearance, whereas other interactions are exploited by bacteria to enhance their pathogenic potential. The discrimination between protective and virulence-enhancing interactions is essential to understand which interactions to target with new prophylactic or treatment strategies. CLRs are also densely concentrated at APC dendrites that sample the environment across intact barrier sites. This suggests an–as yet–underappreciated role for CLR-mediated recognition of microbiota-produced glycans in maintaining tolerance at barrier sites. In addition to providing a concise overview of identified CLR-bacteria interactions, we discuss the main challenges and potential solutions for the identification of new CLR-bacterial interactions, including those with commensal bacteria, and for in-depth structure-function studies on CLR-bacterial glycan interactions. Finally, we highlight the necessity for more relevant tissue-specific in vitro, in vivo and ex vivo models to develop therapeutic applications in this area.
Collapse
Affiliation(s)
- Malgorzata E Mnich
- Medical Microbiology, UMC Utrecht, Utrecht University, Utrecht, Netherlands.,GSK, Siena, Italy
| | - Rob van Dalen
- Interfaculty Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen, Germany
| | - Nina M van Sorge
- Department of Medical Microbiology and Infection Prevention, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Netherlands Reference Laboratory for Bacterial Meningitis, Amsterdam University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
34
|
Ramadan Q, Zourob M. Organ-on-a-chip engineering: Toward bridging the gap between lab and industry. BIOMICROFLUIDICS 2020; 14:041501. [PMID: 32699563 PMCID: PMC7367691 DOI: 10.1063/5.0011583] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/22/2020] [Indexed: 05/03/2023]
Abstract
Organ-on-a-chip (OOC) is a very ambitious emerging technology with a high potential to revolutionize many medical and industrial sectors, particularly in preclinical-to-clinical translation in the pharmaceutical arena. In vivo, the function of the organ(s) is orchestrated by a complex cellular structure and physiochemical factors within the extracellular matrix and secreted by various types of cells. The trend in in vitro modeling is to simplify the complex anatomy of the human organ(s) to the minimal essential cellular structure "micro-anatomy" instead of recapitulating the full cellular milieu that enables studying the absorption, metabolism, as well as the mechanistic investigation of drug compounds in a "systemic manner." However, in order to reflect the human physiology in vitro and hence to be able to bridge the gap between the in vivo and in vitro data, simplification should not compromise the physiological relevance. Engineering principles have long been applied to solve medical challenges, and at this stage of organ-on-a-chip technology development, the work of biomedical engineers, focusing on device engineering, is more important than ever to accelerate the technology transfer from the academic lab bench to specialized product development institutions and to the increasingly demanding market. In this paper, instead of presenting a narrative review of the literature, we systemically present a synthesis of the best available organ-on-a-chip technology from what is found, what has been achieved, and what yet needs to be done. We emphasized mainly on the requirements of a "good in vitro model that meets the industrial need" in terms of the structure (micro-anatomy), functions (micro-physiology), and characteristics of the device that hosts the biological model. Finally, we discuss the biological model-device integration supported by an example and the major challenges that delay the OOC technology transfer to the industry and recommended possible options to realize a functional organ-on-a-chip system.
Collapse
Affiliation(s)
- Qasem Ramadan
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| | - Mohammed Zourob
- Alfaisal University, Al Zahrawi Street, Riyadh 11533, Kingdom of Saudi Arabia
| |
Collapse
|
35
|
Bolle ECL, Verderosa AD, Dhouib R, Parker TJ, Fraser JF, Dargaville TR, Totsika M. An in vitro Reconstructed Human Skin Equivalent Model to Study the Role of Skin Integration Around Percutaneous Devices Against Bacterial Infection. Front Microbiol 2020; 11:670. [PMID: 32477277 PMCID: PMC7240036 DOI: 10.3389/fmicb.2020.00670] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/24/2020] [Indexed: 01/19/2023] Open
Abstract
Percutaneous devices are a key technology in clinical practice, used to connect internal organs to external medical devices. Examples include prosthesis, catheters and electrical drivelines. Percutaneous devices breach the skin's natural barrier and create an entry point for pathogens, making device infections a widespread problem. Modification of the percutaneous implant surface to increase skin integration with the aim to reduce subsequent infection is attracting a great deal of attention. While novel surfaces have been tested in various in vitro models used to study skin integration around percutaneous devices, no skin model has been reported, for the study of bacterial infection around percutaneous devices. Here, we report the establishment of an in vitro human skin equivalent model for driveline infections caused by Staphylococcus aureus, the most common cause of driveline-related infections. Three types of mock drivelines manufactured using melt electrowriting (smooth or porous un-seeded and porous pre-seeded with human fibroblasts) were implanted in human skin constructs and challenged with S. aureus. Our results show a high and stable load of S. aureus in association with the skin surface and no signs of S. aureus-induced tissue damage. Furthermore, our results demonstrate that bacterial migration along the driveline surface occurs in micro-gaps caused by insufficient skin integration between the driveline and the surrounding skin consistent with clinical reports from explanted patient drivelines. Thus, the human skin-driveline infection model presented here provides a clinically-relevant and versatile experimental platform for testing novel device surfaces and infection therapeutics.
Collapse
Affiliation(s)
- Eleonore C. L. Bolle
- Tissue Repair and Translational Physiology Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
- The Innovative Cardiovascular Engineering and Technology Laboratory, Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia
- Infection and Immunity Research Program, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Anthony D. Verderosa
- Infection and Immunity Research Program, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Rabeb Dhouib
- Infection and Immunity Research Program, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| | - Tony J. Parker
- Tissue Repair and Translational Physiology Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - John F. Fraser
- The Innovative Cardiovascular Engineering and Technology Laboratory, Critical Care Research Group, The Prince Charles Hospital, Brisbane, QLD, Australia
| | - Tim R. Dargaville
- Tissue Repair and Translational Physiology Program, Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, QLD, Australia
| | - Makrina Totsika
- Infection and Immunity Research Program, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Faculty of Health, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
36
|
Vidal Yucha SE, Tamamoto KA, Kaplan DL. The importance of the neuro-immuno-cutaneous system on human skin equivalent design. Cell Prolif 2019; 52:e12677. [PMID: 31441145 PMCID: PMC6869210 DOI: 10.1111/cpr.12677] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Revised: 06/27/2019] [Accepted: 07/15/2019] [Indexed: 12/11/2022] Open
Abstract
The skin is a highly complex organ, responsible for sensation, protection against the environment (pollutants, foreign proteins, infection) and thereby linked to the immune and sensory systems in the neuro-immuno-cutaneous (NIC) system. Cutaneous innervation is a key part of the peripheral nervous system; therefore, the skin should be considered a sensory organ and an important part of the central nervous system, an 'active interface' and the first connection of the body to the outside world. Peripheral nerves are a complex class of neurons within these systems, subsets of functions are conducted, including mechanoreception, nociception and thermoception. Epidermal and dermal cells produce signalling factors (such as cytokines or growth factors), neurites influence skin cells (such as via neuropeptides), and peripheral nerves have a role in both early and late stages of the inflammatory response. One way this is achieved, specifically in the cutaneous system, is through neuropeptide release and signalling, especially via substance P (SP), neuropeptide Y (NPY) and nerve growth factor (NGF). Cutaneous, neuronal and immune cells play a central role in many conditions, including psoriasis, atopic dermatitis, vitiligo, UV-induced immunosuppression, herpes and lymphomas. Therefore, it is critical to understand the connections and interplay between the peripheral nervous system and the skin and immune systems, the NIC system. Relevant in vitro tissue models based on human skin equivalents can be used to gain insight and to address impact across research and clinical needs.
Collapse
Affiliation(s)
- Sarah E Vidal Yucha
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| | - Kasey A Tamamoto
- Department of Chemistry, Tufts University, Medford, Massachusetts
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts
| |
Collapse
|
37
|
Yamamoto K, Ioroi T, Harada K, Nishiyama S, Nishigori C, Yano I. Safety and Efficacy of Bis-Glyceryl Ascorbate (Amitose DGA) to Prevent Hand-Foot Skin Reaction in Patients With Renal Cell Carcinoma Receiving Sunitinib Therapy: Protocol for a Phase I/II, Uncontrolled, Single-Arm, Open-Label Trial. JMIR Res Protoc 2019; 8:e14636. [PMID: 31407671 PMCID: PMC6709566 DOI: 10.2196/14636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 12/14/2022] Open
Abstract
Background Hand-foot skin reaction (HFSR) is a serious side effect induced by multiple-tyrosine kinase inhibitors (TKIs). HFSR can cause treatment interruption or decreased dosing. HFSR also markedly decreases quality of life and is associated with the therapeutic efficacy of multiple-TKIs. Therefore, the management and prevention of HFSR is an important issue; however, an effective method for its prevention has not been established. Specific ascorbic acid derivatives can reverse multiple-TKI-induced keratinocyte growth and pathological changes in vitro. Objective This study was designed to evaluate the safety of bis-glyceryl ascorbate (Amitose DGA), a novel, hydrosoluble, and moisturizing ascorbic acid derivative, in patients with renal cell carcinoma (RCC) receiving sunitinib therapy. This study was also designed to evaluate Amitose DGA’s preventive efficacy for sunitinib-induced HFSR. Methods This is a Phase I/II, single-center, uncontrolled, single-arm, open-label trial. We will recruit a total of 30 patients with RCC receiving sunitinib therapy, with a 2-week-on and 1-week-off schedule. The participants will apply Amitose DGA-containing cream over both palmar and plantar surfaces within two treatment cycles (ie, 6 weeks) of sunitinib in combination with a general moisturizing agent, in addition to standard-of-care processes. Safety assessments will include dermatological abnormalities, clinical laboratory tests, and incidence of adverse events. Efficacy assessments will include development of HFSR and therapeutic outcomes associated with sunitinib. Results Recruitment to the study began in August 2017 and is ongoing in Japan. To date, 21 subjects have been recruited. Study completion is expected in 2021. Conclusions This is the first clinical study of Amitose DGA-containing cream in patients with RCC who are receiving sunitinib therapy. The single-center, single-arm, open-label design was selected to maximize subject exposure and increase the likelihood of achieving our study endpoints. The results will provide valuable and preliminary evidence of the effects of Amitose DGA-containing cream on HFSR. Trial Registration UMIN Clinical Trials Registry UMIN000027209; https://upload.umin.ac.jp/cgi-open-bin/ctr /ctr_view.cgi?recptno=R000031174 International Registered Report Identifier (IRRID) DERR1-10.2196/14636
Collapse
Affiliation(s)
| | - Takeshi Ioroi
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
| | - Kenichi Harada
- Department of Urology, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Satoshi Nishiyama
- Division of Dermatology, Department of Internal Related, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Chikako Nishigori
- Division of Dermatology, Department of Internal Related, Graduate School of Medicine, Kobe University, Kobe, Japan
| | - Ikuko Yano
- Department of Pharmacy, Kobe University Hospital, Kobe, Japan
| |
Collapse
|
38
|
|
39
|
Sung JH, Wang YI, Narasimhan Sriram N, Jackson M, Long C, Hickman JJ, Shuler ML. Recent Advances in Body-on-a-Chip Systems. Anal Chem 2019; 91:330-351. [PMID: 30472828 PMCID: PMC6687466 DOI: 10.1021/acs.analchem.8b05293] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jong Hwan Sung
- Department of Chemical Engineering , Hongik University , Seoul , 04066 , Republic of Korea
| | - Ying I Wang
- Nancy E. and Peter C. Meinig School of Biomedical Engineering , Cornell University , Ithaca , New York 14853 , United States
| | | | - Max Jackson
- Hesperos, Inc. Orlando , Florida 32836 , United States
| | | | - James J Hickman
- Hesperos, Inc. Orlando , Florida 32836 , United States
- NanoScience Technology Center , University of Central Florida , Orlando , Florida 32828 , United States
| | - Michael L Shuler
- Nancy E. and Peter C. Meinig School of Biomedical Engineering , Cornell University , Ithaca , New York 14853 , United States
- Hesperos, Inc. Orlando , Florida 32836 , United States
- Robert Frederick Smith School of Chemical and Biomolecular Engineering , Cornell University , Ithaca , New York 14853 , United States
| |
Collapse
|
40
|
Osswald A, Hedrich V, Sommergruber W. 3D-3 Tumor Models in Drug Discovery for Analysis of Immune Cell Infiltration. Methods Mol Biol 2019; 1953:151-162. [PMID: 30912021 DOI: 10.1007/978-1-4939-9145-7_10] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
The cross talk between tumor cells and other cells present in the tumor microenvironment such as stromal and immune cells highly influences the behavior and progression of disease. Understanding the underlying mechanisms of interaction is a prerequisite to develop new treatment strategies and to prevent or at least reduce therapy failure in the future. Specific reactivation of the patient's immune system is one of the major goals today. However, standard two-dimensional (2D) cell culture techniques lack the necessary complexity to address related questions. Novel three-dimensional (3D) in vitro models-embedded in a matrix or encapsulated in alginate-recapitulate the in vivo situation much better. Cross talk between different cell types can be studied starting from co-cultures. As cancer immune modulation is becoming a major research topic, 3D in vitro models represent an important tool to address immune regulatory/modulatory questions for T, NK, and other cells of the immune system. The 3D systems consisting of tumor cells, fibroblasts, and immune cells (3D-3) already proved as a reliable tool for us. For instance, we made use of those models to study the molecular mechanisms of the cross talk of non-small cell lung cancer (NSCLC) and fibroblasts, to unveil macrophage plasticity in the tumor microenvironment and to mirror drug responses in vivo. Generation of those 3D models and how to use them to study immune cell infiltration and activation will be described in the present book chapter.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Bioreactors
- Carcinoma, Non-Small-Cell Lung/drug therapy
- Carcinoma, Non-Small-Cell Lung/immunology
- Carcinoma, Non-Small-Cell Lung/pathology
- Cells, Immobilized/drug effects
- Cells, Immobilized/immunology
- Cells, Immobilized/pathology
- Coculture Techniques/methods
- Drug Discovery/methods
- Drug Screening Assays, Antitumor/methods
- Fibroblasts/drug effects
- Fibroblasts/immunology
- Fibroblasts/pathology
- Humans
- Immunity, Cellular/drug effects
- Lung Neoplasms/drug therapy
- Lung Neoplasms/immunology
- Lung Neoplasms/pathology
- Spheroids, Cellular/drug effects
- Spheroids, Cellular/immunology
- Spheroids, Cellular/pathology
- Tumor Cells, Cultured
- Tumor Microenvironment/drug effects
Collapse
Affiliation(s)
| | - Viola Hedrich
- Boehringer Ingelheim RCV GmbH & Co KG, Vienna, Austria
| | | |
Collapse
|
41
|
Vidal Yucha SE, Tamamoto KA, Nguyen H, Cairns DM, Kaplan DL. Human Skin Equivalents Demonstrate Need for Neuro-Immuno-Cutaneous System. ACTA ACUST UNITED AC 2018; 3:e1800283. [PMID: 32627348 DOI: 10.1002/adbi.201800283] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Indexed: 12/14/2022]
Abstract
A variety of human skin equivalents (HSEs) has been designed for clinical use or for exploratory skin research. In vitro HSE models have been used to target relationships between the skin and nervous or immune systems but have not yet considered the neuro-immuno-cutaneous (NIC) system. In this study, HSEs are described, with and without neural and immune components, to discern these types of effects. These systems are composed of only primary human cells and contain an epidermis, dermis, hypodermis (with immune cells), and human induced neural stem cells for the neuronal component. RNA sequencing is utilized to confirm differences between sample groups and to identify unique or important genes with respect to sample type. Only samples with both neural and immune components result in the upregulation of genes in all the key biological pathways explored. The analysis of protein secretion confirms that this group has measurable functions related to all key cell types. Overall, this novel skin tissue system confirms that designing HSEs that include the NIC system results in a tissue model that reflects key functions. These systems could be used to identify selected targets of interest in skin research related to healthy or diseased states.
Collapse
Affiliation(s)
- Sarah E Vidal Yucha
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - Kasey A Tamamoto
- Department of Chemistry, Tufts University, Medford, MA, 02155, USA
| | - Hanh Nguyen
- Department of Child Studies and Human Development, Tufts University, Medford, MA, 02155, USA
| | - Dana M Cairns
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby St., Medford, MA, 02155, USA
| |
Collapse
|