1
|
Dong X, Xiang H, Li J, Hao A, Wang H, Gou Y, Li A, Rahaman S, Qiu Y, Li J, Mei O, Zhong J, You W, Shen G, Wu X, Li J, Shu Y, Shi LL, Zhu Y, Reid RR, He TC, Fan J. Dermal fibroblast-derived extracellular matrix (ECM) synergizes with keratinocytes in promoting re-epithelization and scarless healing of skin wounds: Towards optimized skin tissue engineering. Bioact Mater 2025; 47:1-17. [PMID: 39872210 PMCID: PMC11762682 DOI: 10.1016/j.bioactmat.2024.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/24/2024] [Accepted: 12/27/2024] [Indexed: 01/30/2025] Open
Abstract
Skin serves as the first-order protective barrier against the environment and any significant disruptions in skin integrity must be promptly restored. Despite significant advances in therapeutic strategies, effective management of large chronic skin wounds remains a clinical challenge. Dermal fibroblasts are the primary cell type responsible for remodeling the extracellular matrix (ECM) in wound healing. Here, we investigated whether ECM derived from exogenous fibroblasts, in combination with keratinocytes, promoted scarless cutaneous wound healing. To overcome the limited lifespan of primary dermal fibroblasts, we established reversibly immortalized mouse dermal fibroblasts (imDFs), which were non-tumorigenic, expressed dermal fibroblast markers, and were responsive to TGF-β1 stimulation. The decellularized ECM prepared from both imDFs and primary dermal fibroblasts shared similar expression profiles of extracellular matrix proteins and promoted the proliferation of keratinocyte (iKera) cells. The imDFs-derived ECM solicited no local immune response. While the ECM and to a lesser extent imDFs enhanced skin wound healing with excessive fibrosis, a combination of imDFs-derived ECM and iKera cells effectively promoted the re-epithelization and scarless healing of full-thickness skin wounds. These findings strongly suggest that dermal fibroblast-derived ECM, not fibroblasts themselves, may synergize with keratinocytes in regulating scarless healing and re-epithelialization of skin wounds. Given its low immunogenic nature, imDFs-derived ECM should be a valuable resource of skin-specific biomaterial for wound healing and skin tissue engineering.
Collapse
Affiliation(s)
- Xiangyu Dong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Han Xiang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiajia Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ailing Hao
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Hao Wang
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yannian Gou
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Aohua Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Saidur Rahaman
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Yiheng Qiu
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Jiahao Li
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
| | - Ou Mei
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopedic Surgery, Jiangxi Hospital of Traditional Chinese Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang, 330006, China
| | - Jiamin Zhong
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Wulin You
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedic Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, 214071, China
| | - Guowei Shen
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Orthopaedic Surgery, BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Nanjing, 210019, China
| | - Xingye Wu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jingjing Li
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Department of Oncology, The Affiliated Hospital of Shandong Second Medical University, Weifang, 261053, China
| | - Yi Shu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Stem Cell Biology and Therapy Laboratory of the Pediatric Research Institute, the National Clinical Research Center for Child Health and Disorders, and Ministry of Education Key Laboratory of Child Development and Disorders, the Children's Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lewis L. Shi
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Yi Zhu
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Russell R. Reid
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Tong-Chuan He
- Molecular Oncology Laboratory, Department of Orthopaedic Surgery and Rehabilitation Medicine, The University of Chicago Medical Center, Chicago, IL, 60637, USA
- Laboratory of Craniofacial Biology and Development, Section of Plastic and Reconstructive Surgery, Department of Surgery, The University of Chicago Medical Center, Chicago, IL, 60637, USA
| | - Jiaming Fan
- Ministry of Education Key Laboratory of Diagnostic Medicine, and Department of Clinical Biochemistry, School of Clinical Laboratory Medicine, Chongqing Medical University, Chongqing, 400016, China
- Western Institute of Digital-Intelligent Medicine, Chongqing, 401329, China
| |
Collapse
|
2
|
Bordoni B, Escher AR. Fascial Manual Medicine: The Concept of Fascial Continuum. Cureus 2025; 17:e82136. [PMID: 40226146 PMCID: PMC11992952 DOI: 10.7759/cureus.82136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/12/2025] [Indexed: 04/15/2025] Open
Abstract
Fascial tissue ubiquitously pervades the body system, becoming the target of many disciplines that use manual techniques for patient treatment. It is a much-debated topic as there is currently no univocal definition among different authors. Due to the non-discontinuity of the fascia, we can speak of a fascial continuum; this principle is the basis of the osteopathic perspective. This vision, which seems banal, is not always applied in manual fascial medicine, where, often, it is conditioned by a reductionist (layers) and mechanistic (compartments) approach, forgetting that the body is not a machine but an organism. This continuity teaches that manual treatment does not only reverberate in the area where the operator's hands rest but creates a series of local and systemic adaptations. This narrative review revises the concept of the fascial continuum by highlighting that fascia is a tissue system (different tissues working in harmony), multi-organ (capable of behaving like an organ), whose macroscopic functional expression (movement) and microscopic (with cellular adaptations) derives from a nanoscopic coherence (electromagnetic behaviors). This means that the body acts as a unit, and makes the manual approach never local but always systemic. The aim of the article is to highlight the fact that the fascial continuum is a single biological entity (solid and fluid), and that manual fascial medicine does not approach a single segment, but the entire person.
Collapse
Affiliation(s)
- Bruno Bordoni
- Physical Medicine and Rehabilitation, Foundation Don Carlo Gnocchi, Milan, ITA
| | - Allan R Escher
- Oncologic Sciences, University of South Florida Morsani College of Medicine, Tampa, USA
- Anesthesiology/Pain Medicine, H. Lee Moffitt Cancer Center and Research Institute, Tampa, USA
| |
Collapse
|
3
|
Shestakova VA, Smirnova EI, Atiakshin DA, Kisel AA, Koryakin SN, Litun EV, Saburov VO, Demyashkin GA, Lagoda TS, Yakimova AO, Kabakov AE, Ignatyuk MA, Yatsenko EM, Kudlay DA, Ivanov SA, Shegay PV, Kaprin AD, Baranovskii DS, Komarova LN, Klabukov ID. Impact of Minimally Manipulated Cell Therapy on Immune Responses in Radiation-Induced Skin Wound Healing. Int J Mol Sci 2025; 26:1994. [PMID: 40076619 PMCID: PMC11900442 DOI: 10.3390/ijms26051994] [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: 12/30/2024] [Revised: 02/07/2025] [Accepted: 02/18/2025] [Indexed: 03/14/2025] Open
Abstract
The current treatment of radiation-induced skin wounds utilizes mainly conventional therapies, including topical steroids, creams, ointments, and hydrogel dressings, which do not take into account the immunologic changes that occur in the skin after radiation exposure. Therefore, it is relevant to consider alternative therapies and their impact on changes in the immune landscape of the skin. The aim of this study was to investigate the effect of allogeneic minimally manipulated keratinocytes and fibroblasts on rat skin repair and the development of immune responses. We found that the use of cell therapy compared to treatment with syntazone ointment and no treatment resulted in faster healing and a reduction in the size of radiation-induced skin wounds, area of inflammation, and edema. Additionally, in the group receiving the cell therapy application, there was an observed increase in the number of mast cells (MCs), activation of MC interaction with M2 macrophages, a reduction in the direct contact of MCs with the vascular bed, an increase in the content of collagen fibers due to the intensification of collagen fibrillogenesis, and a restoration of their histotopographic organization. Thus, the positive effect of cell therapy based on allogeneic minimally manipulated keratinocytes and fibroblasts on skin regeneration indicated that it can be used in clinical practice to improve the effectiveness of rehabilitation after radiation therapy.
Collapse
Affiliation(s)
- Victoria A. Shestakova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Ekaterina I. Smirnova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Dmitrii A. Atiakshin
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anastas A. Kisel
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Sergey N. Koryakin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Evgeniy V. Litun
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Vyacheslav O. Saburov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Grigory A. Demyashkin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Institute for Translational Medicine and Biotechnology, Sechenov First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia
| | - Tatyana S. Lagoda
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Anna O. Yakimova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Alexander E. Kabakov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Michael A. Ignatyuk
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Elena M. Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Dmitry A. Kudlay
- Immunology Department, Institute of Immunology FMBA of Russia, 115552 Moscow, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Sergey A. Ivanov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Peter V. Shegay
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Scientific and Educational Resource Center “Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis”, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Denis S. Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- University Hospital Basel, Basel University, 4001 Basel, Switzerland
- Research and Educational Resource Center for Cellular Technologies, Patrice Lumumba Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Lyudmila N. Komarova
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Ilya D. Klabukov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia; (V.A.S.)
- Department of Biotechnology, Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| |
Collapse
|
4
|
Song Y, Cheng W, Wang Z, Zhou T, Wu F, Yin Y, Xu D, Liu Y. Mechanism of PAVA-induced toxicity and inflammation in a cocultured skin cell model. Front Pharmacol 2025; 16:1531459. [PMID: 40041491 PMCID: PMC11876130 DOI: 10.3389/fphar.2025.1531459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/27/2025] [Indexed: 03/06/2025] Open
Abstract
Background Pelargonic acid vanillyl amide (PAVA), a stable synthetic analog of capsaicin, exhibits potential for therapeutic applications; however, it may present cytotoxic and pro-inflammatory risks. This study aims to investigate the injury effects of PAVA on a cocultured skin cell model in vitro. Methods Human keratinocytes and dermal fibroblasts were co-cultured and exposed to PAVA at concentrations ranging from 12.5 to 200 µM for durations of 5, 24, and 48 h. Cell proliferation was quantified using MTS assays. Morphological changes were observed through microscopy, reactive oxygen species (ROS) production was evaluated via fluorescence analysis, apoptosis was assessed using flow cytometry and Western blotting techniques, while inflammatory cytokines (IL-6, IL-8) were quantified by ELISA. Results The proliferation of cells was significantly inhibited by PAVA in a dose- and time-dependent manner, with concentrations of 100 µM and above inducing substantiazl cytotoxicity. Morphological analysis revealed an increase in cell dispersion, irregular morphology, and apoptosis, particularly after prolonged exposure. Treatment with PAVA led to elevated levels of ROS, indicating the presence of oxidative stress. Apoptosis was initiated through both extrinsic pathways (NF-κB, Caspase-8) at an early stage and intrinsic pathways (Caspase-3/9, Bax) at a later period. Furthermore, PAVA markedly increased the secretion of IL-6 and IL-8, suggesting a robust pro-inflammatory response. Conclusion 100 μM PAVA elicits pronounced cytotoxic, oxidative, and pro-inflammatory effects on cocultured skin cell model, particularly at higher concentrations and prolonged exposure durations. These findings underscore the necessity of exercising caution when employing PAVA for therapeutic purposes and highlight the imperative for further research to mitigate its adverse consequences as a riot control agent.
Collapse
Affiliation(s)
- Yunyang Song
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Wenjie Cheng
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Zhen Wang
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Tianqi Zhou
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Fanghui Wu
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Yifeng Yin
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| | - Dan Xu
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
- Key Laboratory of Freshwater Aquatic Genetic Resources, Ministry of Agriculture, Shanghai Ocean University, Shanghai, China
| | - Yanli Liu
- State Key Laboratory of NBC Protection for Civilian, Beijing, China
| |
Collapse
|
5
|
Daugaard ND, Tholstrup R, Tornby JR, Bendixen SM, Larsen FT, De Zio D, Barnkob MB, Ravnskjaer K, Brewer JR. Characterization of human melanoma skin cancer models: A step towards model-based melanoma research. Acta Biomater 2025; 191:308-324. [PMID: 39549863 DOI: 10.1016/j.actbio.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 10/27/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024]
Abstract
Advancing 3D in vitro human tissue models is crucial for biomedical research and drug development to address the ethical and biological limitations of animal testing. Recently, 3D skin models have proven to be effective for studying serious skin conditions, such as melanoma. For these advanced models to be applicable in preclinical studies, thorough characterization is essential to understand their applicability and limitations. In this study, we used bioimaging and RNA sequencing to assess the architecture and transcriptomic profiles of skin models, including models with melanoma. Our results indicated that these models closely mimicked skin morphology and gene expression patterns. The full-thickness (FT) model shows a superior resemblance to the human skin, particularly in basement membrane formation and cellular interactions. The integrity of the skin-like properties and gene expression signatures of both skin and melanoma cells were preserved upon the integration of melanoma cells, establishing these models as robust platforms for cancer research. The responsiveness of the FT melanoma models to vemurafenib treatment was successfully monitored, demonstrating their validity as a reliable, reproducible, and humane tool for pharmacological testing and drug development. Furthermore, the transcriptomic data showed that skin models with cancer spheroids had upregulated genes linked to aggressive and resilient cancer behavior compared to spheroids alone. This emphasizes the importance of the microenvironment in cancer progression and suggests that 3D skin models can serve to uncover mechanisms and therapeutic targets that are not detectable in simpler systems. STATEMENT OF SIGNIFICANCE: This study introduces advanced, ethically sound skin and melanoma models as alternatives to animal testing in drug discovery. By thoroughly characterizing these models using bioimaging and RNA sequencing, we demonstrate their close resemblance to human skin, particularly in full-thickness models. These models not only replicate the complex cellular interactions and gene expression patterns of human tissue but also maintain robustness after melanoma integration. Our findings highlight the potential of these models in revealing cancer mechanisms and therapeutic targets, offering a significant impact on melanoma research and preclinical testing.
Collapse
Affiliation(s)
- Nicoline Dorothea Daugaard
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Rikke Tholstrup
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Jakob Rask Tornby
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Sofie Marchsteiner Bendixen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Frederik Tibert Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Daniela De Zio
- Melanoma Research Team, Center for Autophagy, Recycling and Disease (CARD), Danish Cancer Institute, Copenhagen, Denmark; Department of Cancer and Inflammation Research, Institute of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Mike Bogetofte Barnkob
- Centre for Cellular Immunotherapy of Haematological Cancer Odense (CITCO), Department of Clinical Immunology, Odense University Hospital, University of Southern Denmark, Odense, Denmark
| | - Kim Ravnskjaer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark
| | - Jonathan R Brewer
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| |
Collapse
|
6
|
van der Vloet L, Ducarne Z, Heeren RMA, Berends AC, Vandenbosch M. Lipid analysis of human primary dermal fibroblasts and epidermal keratinocytes after near-infrared exposure using mass spectrometry imaging. J Biotechnol 2024; 396:53-61. [PMID: 39426412 DOI: 10.1016/j.jbiotec.2024.10.009] [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: 07/08/2024] [Revised: 10/11/2024] [Accepted: 10/15/2024] [Indexed: 10/21/2024]
Abstract
Photobiomodulation (PBM) therapy is the application of near-infrared (NIR) exposure to injuries or lesions to (among others) improve wound healing, reduce inflammation, and decreases acute and chronic pain. However, the understanding of the molecular mechanism of PBM, more specifically the effects of NIR on skin cells is still lacking behind. Lipids are essential components of cellular membranes that are integral to skin structure and function. This study aims to elucidate the impact of NIR exposure on the skin's lipidome by investigating the molecular effect of NIR exposure on single skin cells. Primary human dermal fibroblasts (NHDFa) and human epidermal keratinocytes (HEKa) were exposed to NIR (850 nm) with a dose of 6.5 J/cm2 for 5 consecutive days between 09.00 and 12.00 am. A workflow utilizing matrix-assisted laser desorption/ionization mass spectrometry imaging combined with liquid chromatography tandem mass spectrometry for lipidomics analysis was performed. This study provides evidence that adequate exposure of NIR influences lipid metabolism in NHDFa, whereas no alterations were found in HEKa. This work lays the groundwork in explaining the beneficial properties on both skin-related effects and systemic health benefits as seen in clinical studies.
Collapse
Affiliation(s)
- Laura van der Vloet
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands.
| | - Zoé Ducarne
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands.
| | - Ron M A Heeren
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands.
| | - Anne C Berends
- Seaborough Life Science, Amsterdam 1098 XG, The Netherlands.
| | - Michiel Vandenbosch
- The Maastricht MultiModal Molecular Imaging (M4I) institute, Division of Imaging Mass Spectrometry (IMS), Maastricht University, Maastricht 6229 ER, The Netherlands.
| |
Collapse
|
7
|
Jia YY, Atwood SX. Diversity of human skin three-dimensional organotypic cultures. Curr Opin Genet Dev 2024; 89:102275. [PMID: 39536613 DOI: 10.1016/j.gde.2024.102275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/17/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Recently, significant strides have been made in the development of high-fidelity skin organoids, encompassing techniques such as 3D bioprinting, skin-on-a-chip systems, and models derived from pluripotent stem cells (PSCs), replicating appendage structures and diverse skin cell types. Despite the emergence of these state-of-the-art skin engineering models, human organotypic cultures (OTCs), initially proposed in the 1970s, continue to reign as the predominant in vitro cultured three-dimensional skin model in the field of tissue engineering. This enduring prevalence is owed to their cost-effectiveness, straight forward setup, time efficiency, and faithful representation of native human skin. In this review, we systematically delineate recent advances in skin OTC models, aiming to inform future efforts to enhance in vitro skin model fidelity and reproducibility.
Collapse
Affiliation(s)
- Yunlong Y Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Dermatology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
8
|
Klein B, Nguyen NTK, Moallemian R, Kahlenberg JM. Keratinocytes - Amplifiers of Immune Responses in Systemic Lupus Erythematosus. Curr Rheumatol Rep 2024; 27:1. [PMID: 39570551 DOI: 10.1007/s11926-024-01168-3] [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] [Accepted: 10/22/2024] [Indexed: 11/22/2024]
Abstract
PURPOSE OF REVIEW Epithelial cells have been acknowledged as important players in autoimmune diseases by directing and enhancing inflammatory responses. Here, we summarize recent publications that examine keratinocyte (KC) dysfunction and its contribution to cutaneous and systemic disease in systemic lupus erythematosus patients. RECENT FINDINGS Chronic upregulation of type I interferon (IFN) in KCs is a feature of both lesional and nonlesional lupus skin. This IFN rich environment modulates epidermal cell death responses and promotes inflammatory responses to UV light exposure. In addition, newer technologies such as single cell RNA-seq are informing our understanding of lupus-specific intercellular crosstalk and how this contributes to disease. Recent discoveries in KC dysfunction in lupus skin include aberrant IFN responses to environmental stress, enhanced cytokine and chemokine secretion and epigenetic changes leading to increased cell death. Further research will enable precision therapies for lupus treatment.
Collapse
Affiliation(s)
- Benjamin Klein
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Nguyen Thi Kim Nguyen
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - Rezvan Moallemian
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA
| | - J Michelle Kahlenberg
- Division of Rheumatology, Department of Internal Medicine, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109, USA.
- Department of Dermatology, University of Michigan, Ann Arbor, USA.
| |
Collapse
|
9
|
Morgner B, Werz O, Wiegand C, Tittelbach J. Bilayered skin equivalent mimicking psoriasis as predictive tool for preclinical treatment studies. Commun Biol 2024; 7:1529. [PMID: 39558145 PMCID: PMC11574237 DOI: 10.1038/s42003-024-07226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 11/07/2024] [Indexed: 11/20/2024] Open
Abstract
Psoriasis is a prevalent, inflammatory skin disease without cure. Further research is required to unravel dysregulated processes and develop new therapeutic interventions. The lack of suitable in vivo and in vitro preclinical models is an impediment in the psoriasis research. Recently, the development of 3D skin models has progressed including replicas with disease-like features. To investigate the use of in vitro models as preclinical test tools, the study focused on treatment responses of 3D skin replicas. Cytokine-priming of skin organoids induced psoriatic features like inflammation, antimicrobial peptides (AMP), hyperproliferation and impaired differentiation. Topical application of dexamethasone (DEX) or celastrol (CEL), a natural anti-inflammatory compound reduced the secretion of pro-inflammatory cytokines. DEX and CEL decreased the gene expression of inflammatory mediators. DEX barely affected the psoriatic AMP transcription but CEL downregulated psoriasis-driven AMP genes. Subcutaneous application of adalimumab (ADM) or bimekizumab (BMM) showed anti-psoriatic effects via protein induction of the differentiation marker keratin-10. Dual blockage of TNF-α and IL-17A repressed the inflammatory psoriasis phenotype. BMM inhibited the psoriatic expression of AMP genes and induced KRT10 and cell-cell contact genes. The present in vitro model provides a 3D environment with in vivo-like cutaneous responses and represents a promising tool for preclinical investigations.
Collapse
Affiliation(s)
- Bianka Morgner
- University Hospital Jena, Department of Dermatology, Friedrich Schiller University Jena, Jena, Germany.
| | - Oliver Werz
- Department of Pharmaceutical/Medicinal Chemistry, Institute of Pharmacy, Friedrich Schiller University Jena, Jena, Germany
| | - Cornelia Wiegand
- University Hospital Jena, Department of Dermatology, Friedrich Schiller University Jena, Jena, Germany
| | - Jörg Tittelbach
- University Hospital Jena, Department of Dermatology, Friedrich Schiller University Jena, Jena, Germany
| |
Collapse
|
10
|
Haga M, Iida K, Okada M. Positive and negative feedback regulation of the TGF-β1 explains two equilibrium states in skin aging. iScience 2024; 27:109708. [PMID: 38706856 PMCID: PMC11066433 DOI: 10.1016/j.isci.2024.109708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 02/05/2024] [Accepted: 04/06/2024] [Indexed: 05/07/2024] Open
Abstract
During aging, skin homeostasis is essential for maintaining appearance, as well as biological defense of the human body. In this study, we identified thrombospondin-1 (THBS1) and fibromodulin (FMOD) as positive and negative regulators, respectively, of the TGF-β1-SMAD4 axis in human skin aging, based on in vitro and in vivo omics analyses and mathematical modeling. Using transcriptomic and epigenetic analyses of senescent dermal fibroblasts, TGF-β1 was identified as the key upstream regulator. Bifurcation analysis revealed a binary high-/low-TGF-β1 switch, with THBS1 as the main controller. Computational simulation of the TGF-β1 signaling pathway indicated that THBS1 expression was sensitively regulated, whereas FMOD was regulated robustly. Results of sensitivity analysis and validation showed that inhibition of SMAD4 complex formation was a promising method to control THBS1 production and senescence. Therefore, this study demonstrated the potential of combining data-driven target discovery with mathematical approaches to determine the mechanisms underlying skin aging.
Collapse
Affiliation(s)
- Masatoshi Haga
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
- Basic Research Development Division, ROHTO Pharmaceutical Co., Ltd, Osaka 544-8666, Japan
| | - Keita Iida
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Mariko Okada
- Institute for Protein Research, Osaka University, Suita, Osaka 565-0871, Japan
- Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
11
|
Liu R, Wang Y, Kuai W, Li W, Wang Z, Xiao L, Wu J. Troxerutin suppress inflammation response and oxidative stress in jellyfish dermatitis by activating Nrf2/HO-1 signaling pathway. Front Immunol 2024; 15:1369849. [PMID: 38779681 PMCID: PMC11109374 DOI: 10.3389/fimmu.2024.1369849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/19/2024] [Indexed: 05/25/2024] Open
Abstract
Background Stomolophus meleagris envenomation causes severe cutaneous symptoms known as jellyfish dermatitis. The potential molecule mechanisms and treatment efficiency of dermatitis remain elusive because of the complicated venom components. The biological activity and molecular regulation mechanism of Troxerutin (TRX) was firstly examined as a potential treatment for jellyfish dermatitis. Methods We examined the inhibit effects of the TRX on tentacle extract (TE) obtained from S. meleagris in vivo and in vitro using the mice paw swelling models and corresponding assays for Enzyme-Linked Immunosorbent Assay (ELISA) Analysis, cell counting kit-8 assay, flow cytometry, respectively. The mechanism of TRX on HaCaT cells probed the altered activity of relevant signaling pathways by RNA sequencing and verified by RT-qPCR, Western blot to further confirm protective effects of TRX against the inflammation and oxidative damage caused by TE. Results TE significantly induced the mice paw skin toxicity and accumulation of inflammatory cytokines and reactive oxygen species in vivo and vitro. Moreover, a robust increase in the phosphorylation of mitogen-activated protein kinase (MAPKs) and nuclear factor-kappa B (NF-κB) signaling pathways was observed. While, the acute cutaneous inflammation and oxidative stress induced by TE were significantly ameliorated by TRX treatment. Notablly, TRX suppressed the phosphorylation of MAPK and NF-κB by initiating the nuclear factor erythroid 2-related factor 2 signaling pathway, which result in decreasing inflammatory cytokine release. Conclusion TRX inhibits the major signaling pathway responsible for inducing inflammatory and oxidative damage of jellyfish dermatitis, offering a novel therapy in clinical applications.
Collapse
Affiliation(s)
- Ran Liu
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Yulian Wang
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Wenhao Kuai
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Wenting Li
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| | - Zengfa Wang
- Faculty of Naval Medicine, Naval Medical University, Shanghai, China
- College of Traditional Chinese Medicine, Jilin Agricultural University, Changchun, China
| | - Liang Xiao
- Faculty of Naval Medicine, Naval Medical University, Shanghai, China
| | - Jianhua Wu
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Navy Medical University, Shanghai, China
| |
Collapse
|
12
|
Rimal R, Muduli S, Desai P, Marquez AB, Möller M, Platzman I, Spatz J, Singh S. Vascularized 3D Human Skin Models in the Forefront of Dermatological Research. Adv Healthc Mater 2024; 13:e2303351. [PMID: 38277705 PMCID: PMC11468127 DOI: 10.1002/adhm.202303351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/04/2023] [Indexed: 01/28/2024]
Abstract
In vitro engineered skin models are emerging as an alternative platform to reduce and replace animal testing in dermatological research. Despite the progress made in recent years, considerable challenges still exist for the inclusion of diverse cell types within skin models. Blood vessels, in particular, are essential in maintaining tissue homeostasis and are one of many primary contributors to skin disease inception and progression. Substantial efforts in the past have allowed the successful fabrication of vascularized skin models that are currently utilized for disease modeling and drugs/cosmetics testing. This review first discusses the need for vascularization within tissue-engineered skin models, highlighting their role in skin grafting and disease pathophysiology. Second, the review spotlights the milestones and recent progress in the fabrication and utilization of vascularized skin models. Additionally, advances including the use of bioreactors, organ-on-a-chip devices, and organoid systems are briefly explored. Finally, the challenges and future outlook for vascularized skin models are addressed.
Collapse
Affiliation(s)
- Rahul Rimal
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Saradaprasan Muduli
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| | - Prachi Desai
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Andrea Bonnin Marquez
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Martin Möller
- DWI Leibniz Institute for Interactive Materials e.VRWTH Aachen UniversityForckenbeckstrasse 5052074AachenGermany
| | - Ilia Platzman
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
| | - Joachim Spatz
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
- Institute for Molecular Systems Engineering and Advanced Materials (IMSEAM)Heidelberg UniversityIm Neuenheimer Feld 22569120HeidelbergGermany
- Max Planck School Matter to LifeJahnstrasse 2969120HeidelbergGermany
| | - Smriti Singh
- Max‐Planck‐Institute for Medical ResearchJahnstrasse 2969120HeidelbergGermany
| |
Collapse
|
13
|
Roh YJ, Choi YH, Shin SH, Lee MK, Won YJ, Lee JH, Cho BS, Park KY, Seo SJ. Adipose tissue-derived exosomes alleviate particulate matter-induced inflammatory response and skin barrier damage in atopic dermatitis-like triple-cell model. PLoS One 2024; 19:e0292050. [PMID: 38241278 PMCID: PMC10798485 DOI: 10.1371/journal.pone.0292050] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 09/11/2023] [Indexed: 01/21/2024] Open
Abstract
Recently, particulate matter (PM) has been shown to exacerbate atopic dermatitis (AD) by inducing an inflammatory response. Meanwhile, several studies revealed that exosomes derived from adipose tissue-derived mesenchymal stem cells promote wound healing and alleviate inflammation via their regenerative and immunomodulatory capacities. Our study aimed to investigate the effects of human adipose tissue-derived mesenchymal stem cell-derived (ASC)-exosomes in PM-induced AD. An AD-like triple-cell model was established by treating human keratinocytes, dermal fibroblasts, and mast cells with polyinosinic:polycytidylic acid (Poly I:C) and interleukin 1 alpha (IL-1α). The effects of PM and ASC-exosomes on the expression of pro-inflammatory cytokines and skin barrier proteins were examined using quantitative real-time polymerase chain reaction, western blotting, and immunofluorescence. PM increased pro-inflammatory cytokines (IL-6, IL-1β, and IL-1α) and decreased the anti-inflammatory cytokine IL-10, while the mRNA expression of skin barrier proteins (loricrin and filaggrin) decreased. However, when the cells were treated with ASC-exosomes, the PM-induced effects on pro-inflammatory cytokines and skin barrier proteins were reversed. Our results confirmed that PM-induced inflammation and skin barrier damage were alleviated by ASC-exosomes in our AD-like triple-cell model. These data suggest that ASC-exosomes can serve as a therapeutic agent for PM-exacerbated AD.
Collapse
Affiliation(s)
- Yoon Jin Roh
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yong Hee Choi
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Sun Hye Shin
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Mi-Kyung Lee
- Department of Laboratory Medicine, Chung-Ang University College of Medicine, Seoul, Korea
| | - Yu Jin Won
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul, Korea
| | - Jun Ho Lee
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul, Korea
| | - Byong Seung Cho
- ExoCoBio Exosome Institute (EEI), ExoCoBio Inc., Seoul, Korea
| | - Kui Young Park
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Seong Jun Seo
- Department of Dermatology, Chung-Ang University College of Medicine, Seoul, Korea
| |
Collapse
|
14
|
Wang L, Wang B, Kou E, Du L, Zhu Y. New insight into the role of fibroblasts in the epithelial immune microenvironment in the single-cell era. Front Immunol 2023; 14:1259515. [PMID: 37809065 PMCID: PMC10556469 DOI: 10.3389/fimmu.2023.1259515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2023] [Accepted: 09/11/2023] [Indexed: 10/10/2023] Open
Abstract
The skin is exposed to environmental challenges and contains heterogeneous cell populations such as epithelial cells, stromal cells, and skin-resident immune cells. As the most abundant type of stromal cells, fibroblasts have been historically considered silent observers in the immune responses of the cutaneous epithelial immune microenvironment (EIME), with little research conducted on their heterogeneity and immune-related functions. Single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST) have overcome the limitations of bulk RNA sequencing and help recognize the functional and spatial heterogeneity of fibroblasts, as well as their crosstalk with other types of cells in the cutaneous EIME. Recently, emerging single-cell sequencing data have demonstrated that fibroblasts notably participate in the immune responses of the EIME and impact the initiation and progression of inflammatory skin diseases. Here, we summarize the latest advances in the role of fibroblasts in the cutaneous EIME of inflammatory skin diseases and discuss the distinct functions and molecular mechanisms of activated fibroblasts in fibrotic skin diseases and non-fibrotic inflammatory skin diseases. This review help unveil the multiple roles of fibroblasts in the cutaneous EIME and offer new promising therapeutic strategies for the management of inflammatory skin diseases by targeting fibroblasts or the fibroblast-centered EIME.
Collapse
Affiliation(s)
| | | | | | - Lin Du
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai, China
| | - Yuanjie Zhu
- Department of Dermatology, Naval Medical Center, Naval Medical University, Shanghai, China
| |
Collapse
|
15
|
Stabell AR, Lee GE, Jia Y, Wong KN, Wang S, Ling J, Nguyen SD, Sen GL, Nie Q, Atwood SX. Single-cell transcriptomics of human-skin-equivalent organoids. Cell Rep 2023; 42:112511. [PMID: 37195865 PMCID: PMC10348600 DOI: 10.1016/j.celrep.2023.112511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 03/07/2023] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Several methods for generating human-skin-equivalent (HSE) organoid cultures are in use to study skin biology; however, few studies thoroughly characterize these systems. To fill this gap, we use single-cell transcriptomics to compare in vitro HSEs, xenograft HSEs, and in vivo epidermis. By combining differential gene expression, pseudotime analyses, and spatial localization, we reconstruct HSE keratinocyte differentiation trajectories that recapitulate known in vivo epidermal differentiation pathways and show that HSEs contain major in vivo cellular states. However, HSEs also develop unique keratinocyte states, an expanded basal stem cell program, and disrupted terminal differentiation. Cell-cell communication modeling shows aberrant epithelial-to-mesenchymal transition (EMT)-associated signaling pathways that alter upon epidermal growth factor (EGF) supplementation. Last, xenograft HSEs at early time points post transplantation significantly rescue many in vitro deficits while undergoing a hypoxic response that drives an alternative differentiation lineage. This study highlights the strengths and limitations of organoid cultures and identifies areas for potential innovation.
Collapse
Affiliation(s)
- Adam R Stabell
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Grace E Lee
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Yunlong Jia
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - Kirsten N Wong
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA
| | - Shuxiong Wang
- Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA
| | - Ji Ling
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Sandrine D Nguyen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA
| | - George L Sen
- Department of Dermatology, University of California, San Diego, La Jolla, CA 92093, USA; Department of Cellular and Molecular Medicine, UCSD Stem Cell Program, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Department of Mathematics, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Scott X Atwood
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA 92697, USA; NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA 92697, USA; Center for Complex Biological Systems, Chao Family Comprehensive Cancer Center, University of California, Irvine, Irvine, CA 92697, USA; Department of Dermatology, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
16
|
Li T, Sun Y, Wang J, Zhang C, Sun Y. Promoted Skin Wound Healing by Tail-Amputated Eisenia foetida Proteins via the Ras/Raf/MEK/ERK Signaling Pathway. ACS OMEGA 2023; 8:13935-13943. [PMID: 37091432 PMCID: PMC10116500 DOI: 10.1021/acsomega.3c00317] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 03/22/2023] [Indexed: 05/03/2023]
Abstract
Skin wound healing is an important fundamental problem in biological and medical fields. This study aimed to investigate wound healing promotion of protein extract from tail-amputated Eisenia foetida (E. foetida) and reveal the mechanism correlated with the Ras/Raf/MEK/ERK signaling pathway. Proteins extracted from tail-amputated E. foetida were applied on rats' full-thickness excisional wounds to evaluate their regenerative efficacy. Rat skin tissues around surgical defects were analyzed by immunofluorescence staining and Western blot methods. The Ras/Raf/MEK/ERK signaling pathway was further investigated in vitro using the NIH3T3 cell line. A tail-amputated protein extract (ES2) from E. foetida significantly accelerated rat wound healing ability via higher re-epithelialization and ECM deposition in the tissue section compared to the blank control and un-amputated earthworm extract groups. Furthermore, ES2 treatment dramatically accumulated the expressions of platelet-derived growth factor (PDGF), transforming growth factor-β (TGF-β), and hydroxyproline (HYP) in wound areas on day 7 without their accumulation on day 21 post-wounding, diminishing excessive scar formation. Accelerated wound healing ability with the ES2 was proved to correlate with the up-regulation of the Ras/Raf/MEK/ERK signaling pathway. The mRNA expression of this pathway increased significantly in NIH3T3 cells after being treated with the ES2 at an appropriate concentration. The tail-amputated E. foetida proteins (ES2) can significantly promote skin wound healing better than the un-amputated earthworm tissue extract without excessive scar tissue formation. This effect was related to the up-regulation of the Ras/Raf/MEK/ERK signaling pathway.
Collapse
Affiliation(s)
- Tianyi Li
- School
of Chinese Materia Medica, Beijing University
of Chinese Medicine, Yangguang South Road, Fangshan District, Beijing 100029, China
| | - Yujie Sun
- School
of Chinese Materia Medica, Beijing University
of Chinese Medicine, Yangguang South Road, Fangshan District, Beijing 100029, China
| | - Jiaqi Wang
- School
of Chinese Materia Medica, Beijing University
of Chinese Medicine, Yangguang South Road, Fangshan District, Beijing 100029, China
| | - Chenning Zhang
- School
of Chinese Materia Medica, Beijing University
of Chinese Medicine, Yangguang South Road, Fangshan District, Beijing 100029, China
- Department
of Pharmacy, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
- . Phone: +07103420011
| | - Yikun Sun
- School
of Chinese Materia Medica, Beijing University
of Chinese Medicine, Yangguang South Road, Fangshan District, Beijing 100029, China
- . Phone: +01084738619
| |
Collapse
|
17
|
Andl T, Zhou L, Zhang Y. The dermal papilla dilemma and potential breakthroughs in bioengineering hair follicles. Cell Tissue Res 2023; 391:221-233. [PMID: 36562864 PMCID: PMC9898212 DOI: 10.1007/s00441-022-03730-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
The generation and growing of de novo hair follicles is the most daring hair replacement approach to treat alopecia. This approach has been explored at least since the 1960s without major success. Latest in the 1980s, the realization that the mesenchymal compartment of hair follicles, the dermal papilla (DP), is the crucial signaling center and element required for fulfilling this vision of hair follicle engineering, propelled research into the fibroblasts that occupy the DP. However, working with DP fibroblasts has been stubbornly frustrating. Decades of work in understanding the nature of DP fibroblasts in vitro and in vivo have led to the appreciation that hair follicle biology is complex, and the dermal papilla is an enigma. Functional DP fibroblasts tend to aggregate in 2D culture, while impaired DP cells do not. This fact has stimulated recent approaches to overcome the hurdles to DP cell culture by mimicking their natural habitat, such as growing DP fibroblasts in three dimensions (3D) by their self-aggregation, adopting 3D matrix scaffold, or bioprinting 3D microstructures. Furthermore, including keratinocytes in the mix to form hair follicle-like composite structures has been explored but remains a far cry from a useful and affordable method to generate human hair follicles in sufficient quantity and quality in a practical time frame for patients. This suggests that the current strategies may have reached their limitations in achieving successful hair follicle bioengineering for clinical applications. Novel approaches are required to overcome these barriers, such as focusing on embryonic cell types and processes in combination with emerging techniques.
Collapse
Affiliation(s)
- Thomas Andl
- Burnett School of Biological Sciences, University of Central Florida, Orlando, FL, 32816, USA
| | - Linli Zhou
- Division of Pharmaceutical Science, College of Pharmacy, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Yuhang Zhang
- Division of Pharmaceutical Science, College of Pharmacy, University of Cincinnati, Cincinnati, OH, 45267, USA.
| |
Collapse
|
18
|
Induction of psoriasis- and atopic dermatitis-like phenotypes in 3D skin equivalents with a fibroblast-derived matrix. Sci Rep 2023; 13:1807. [PMID: 36720910 PMCID: PMC9889787 DOI: 10.1038/s41598-023-28822-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 01/25/2023] [Indexed: 02/01/2023] Open
Abstract
Skin homeostasis is a complex regulated process relying on the crosstalk of keratinocytes, fibroblasts and immune cells. Imbalances of T-cell subsets and the cytokine environment can lead to inflammatory skin diseases such as psoriasis (Ps) and atopic dermatitis (AD). Modern tissue engineering provides several in vitro models mimicking Ps and AD phenotypes. However, these models are either limited in their pathological features, life span, sample availability, reproducibility, controlled handling or simplicity. Some models further lack intensive characterization as they solely focus on differentiation and proliferation aspects. This study introduces a self-assembly model in which the pathological T-cell-signalling of Ps and AD was simulated by subcutaneous Th1 and Th2 cytokine stimulation. The self-established dermal fibroblast-derived matrices of these models were hypothesized to be beneficial for proximal cytokine signalling on epidermal keratinocytes. Comprehensive histological and mRNA analyses of the diseased skin models showed a weakened barrier, distinct differentiation defects, reduced cellular adhesion, inflammation and parakeratosis formation. A keratin shift of declining physiological cytokeratin-10 (CK10) towards increasing inflammatory CK16 was observed upon Th1 or Th2 stimulation. Antimicrobial peptides (AMPs) were upregulated in Ps and downregulated in AD models. The AD biomarker genes CA2, NELL2 and CCL26 were further induced in AD. While Ps samples featured basal hyperproliferation, cells in AD models displayed apoptotic signs. In accordance, these well-controllable three-dimensional in vitro models exhibited Ps and AD-like phenotypes with a high potential for disease research and therapeutic drug testing.
Collapse
|
19
|
Mulder PPG, Raktoe RS, Vlig M, Elgersma A, Middelkoop E, Boekema BKHL. Full Skin Equivalent Models for Simulation of Burn Wound Healing, Exploring Skin Regeneration and Cytokine Response. J Funct Biomater 2023; 14:29. [PMID: 36662076 PMCID: PMC9864292 DOI: 10.3390/jfb14010029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023] Open
Abstract
Healing of burn injury is a complex process that often leads to the development of functional and aesthetic complications. To study skin regeneration in more detail, organotypic skin models, such as full skin equivalents (FSEs) generated from dermal matrices, can be used. Here, FSEs were generated using de-epidermalized dermis (DED) and collagen matrices MatriDerm® and Mucomaix®. Our aim was to validate the MatriDerm- and Mucomaix-based FSEs for the use as in vitro models of wound healing. Therefore, we first characterized the FSEs in terms of skin development and cell proliferation. Proper dermal and epidermal morphogenesis was established in all FSEs and was comparable to ex vivo human skin models. Extension of culture time improved the organization of the epidermal layers and the basement membrane in MatriDerm-based FSE but resulted in rapid degradation of the Mucomaix-based FSE. After applying a standardized burn injury to the models, re-epithelization occurred in the DED- and MatriDerm-based FSEs at 2 weeks after injury, similar to ex vivo human skin. High levels of pro-inflammatory cytokines were present in the culture media of all models, but no significant differences were observed between models. We anticipate that these animal-free in vitro models can facilitate research on skin regeneration and can be used to test therapeutic interventions in a preclinical setting to improve wound healing.
Collapse
Affiliation(s)
- Patrick P. G. Mulder
- Preclinical Research, Association of Dutch Burn Centres (ADBC), P.O. Box 1015, 1940 AE Beverwijk, The Netherlands
- Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud University Medical Center, Geert Grooteplein Zuid 10, 6525 GA Nijmegen, The Netherlands
| | - Rajiv S. Raktoe
- Preclinical Research, Association of Dutch Burn Centres (ADBC), P.O. Box 1015, 1940 AE Beverwijk, The Netherlands
| | - Marcel Vlig
- Preclinical Research, Association of Dutch Burn Centres (ADBC), P.O. Box 1015, 1940 AE Beverwijk, The Netherlands
| | - Anouk Elgersma
- Preclinical Research, Association of Dutch Burn Centres (ADBC), P.O. Box 1015, 1940 AE Beverwijk, The Netherlands
| | - Esther Middelkoop
- Preclinical Research, Association of Dutch Burn Centres (ADBC), P.O. Box 1015, 1940 AE Beverwijk, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
- Tissue Function and Regeneration, Amsterdam Movement Sciences, De Boelelaan 1105, 1081 HV Amsterdam, The Netherlands
| | - Bouke K. H. L. Boekema
- Preclinical Research, Association of Dutch Burn Centres (ADBC), P.O. Box 1015, 1940 AE Beverwijk, The Netherlands
- Department of Plastic, Reconstructive and Hand Surgery, Amsterdam UMC, Vrije Universiteit Amsterdam, De Boelelaan 1118, 1081 HV Amsterdam, The Netherlands
| |
Collapse
|
20
|
Zhou S, Xie M, Su J, Cai B, Li J, Zhang K. New insights into balancing wound healing and scarless skin repair. J Tissue Eng 2023; 14:20417314231185848. [PMID: 37529248 PMCID: PMC10388637 DOI: 10.1177/20417314231185848] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 06/17/2023] [Indexed: 08/03/2023] Open
Abstract
Scars caused by skin injuries after burns, wounds, abrasions and operations have serious physical and psychological effects on patients. In recent years, the research of scar free wound repair has been greatly expanded. However, understanding the complex mechanisms of wound healing, in which various cells, cytokines and mechanical force interact, is critical to developing a treatment that can achieve scarless wound healing. Therefore, this paper reviews the types of wounds, the mechanism of scar formation in the healing process, and the current research progress on the dual consideration of wound healing and scar prevention, and some strategies for the treatment of scar free wound repair.
Collapse
Affiliation(s)
- Shengxi Zhou
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Mengbo Xie
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Jingjing Su
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Bingjie Cai
- Department of Dermatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Jingan Li
- School of Materials Science and Engineering, Zhengzhou University, Zhengzhou, Henan, P. R. China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, Henan, P. R. China
| |
Collapse
|
21
|
Saraiva MM, Campelo MDS, Câmara Neto JF, Lima ABN, Silva GDA, Dias ATDFF, Ricardo NMPS, Kaplan DL, Ribeiro MENP. Alginate/polyvinyl alcohol films for wound healing: Advantages and challenges. J Biomed Mater Res B Appl Biomater 2023; 111:220-233. [PMID: 35959858 DOI: 10.1002/jbm.b.35146] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 07/08/2022] [Accepted: 07/25/2022] [Indexed: 11/06/2022]
Abstract
The skin is the largest organ in the human body and its physical integrity must be maintained for body homeostasis and to prevent the entry of pathogenic microorganisms. Sodium alginate (SA) and polyvinyl alcohol (PVA) are two polymers widely used in films for wound dressing applications. Furthermore, blends between SA and PVA improve physical, mechanical and biological properties of the final wound healing material when compared to the individual polymers. Different drugs have been incorporated into SA/PVA-based films to improve wound healing activity. It is noteworthy that SA/PVA films can be crosslinked with Ca2+ or other agents, which improves physicochemical and biological properties. Thus, SA/PVA associations are promising for the biomedical field, as a potential alternative for wound treatment. This review focuses on the main techniques for obtaining SA/PVA films, their physical-chemical characterization, drug incorporation, and the advantages and challenges of these films for wound healing.
Collapse
Affiliation(s)
- Matheus Morais Saraiva
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| | - Matheus da Silva Campelo
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| | - João Francisco Câmara Neto
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| | - Ana Beatriz Nogueira Lima
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| | - George de Almeida Silva
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| | - Andre Tavares de Freitas Figueredo Dias
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| | - Nágila Maria Pontes Silva Ricardo
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| | - David L Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts, USA
| | - Maria Elenir Nobre Pinho Ribeiro
- Department of Organic and Inorganic Chemistry, Sciences Center, Laboratory of Polymers and Materials Innovation, Federal University of Ceará, Fortaleza, Brazil
| |
Collapse
|
22
|
Tan SH, Chua DAC, Tang JRJ, Bonnard C, Leavesley D, Liang K. Design of Hydrogel-based Scaffolds for in vitro Three-dimensional Human Skin Model Reconstruction. Acta Biomater 2022; 153:13-37. [DOI: 10.1016/j.actbio.2022.09.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/01/2022] [Accepted: 09/26/2022] [Indexed: 11/01/2022]
|
23
|
Bu T, Zhang M, Lee SH, Cheong YE, Park Y, Kim KH, Kim D, Kim S. GC-TOF/MS-Based Metabolomics for Comparison of Volar and Non-Volar Skin Types. Metabolites 2022; 12:metabo12080717. [PMID: 36005589 PMCID: PMC9415232 DOI: 10.3390/metabo12080717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 07/30/2022] [Accepted: 08/01/2022] [Indexed: 12/10/2022] Open
Abstract
Skin has heterogenous identities on different body sites despite similar cellular compositions. There are two types of skin, volar (palmoplantar) and non-volar (dorsal), which are characterized by epidermal thickness, pigmentation, and presence of hair follicles. However, the mechanisms underlying the development of these different skin types remain unclear. To investigate these, we profiled the cellular metabolites of volar and non-volar skin in mice using gas chromatography-time-of-flight/mass spectrometry (GC-TOF/MS), and further assessed the metabolic differences between them. In total, 96 metabolites from both volar and non-volar skin of mice were identified using the BinBase database system. Metabolomics analysis revealed important differences associated with amino acid metabolism (phenylalanine, tyrosine, and tryptophan biosynthesis; aspartate and glutamate metabolism), sugar metabolism (pentose phosphate pathway), and nucleotide metabolism (pyrimidine metabolism) in volar skin. Fifty metabolites were identified as potential biomarkers differentiating the physiological characteristics of these skin types. Of these, nine were highly increased whereas 41 were significantly decreased in volar skin compared with those in non-volar skin. Overall, these results provide valuable information for understanding the metabolic differences between volar and non-volar skin.
Collapse
Affiliation(s)
- Ting Bu
- Department of Environment Science & Biotechnology, Jeonju University, Jeonju 55069, Korea; (T.B.); (M.Z.)
- University Provincial Key Laboratory for Protection and Utilization of Longdong Bio-Resources in Gan-Su Province, College of Life Sciences and Technology, Longdong University, Qingyang 745000, China
| | - Ming Zhang
- Department of Environment Science & Biotechnology, Jeonju University, Jeonju 55069, Korea; (T.B.); (M.Z.)
| | - Sun-Hee Lee
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Korea; (S.-H.L.); (Y.E.C.); (K.H.K.)
| | - Yu Eun Cheong
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Korea; (S.-H.L.); (Y.E.C.); (K.H.K.)
| | - Yukyung Park
- Graduate School of Energy/Biotechnology, Dongseo University, Busan 47011, Korea;
| | - Kyoung Heon Kim
- Department of Biotechnology, Graduate School, Korea University, Seoul 02841, Korea; (S.-H.L.); (Y.E.C.); (K.H.K.)
| | - Dongwon Kim
- Graduate School of Energy/Biotechnology, Dongseo University, Busan 47011, Korea;
- Department of Bio-Pharmaceutical Engineering, Dongseo University, Busan 47011, Korea
- Correspondence: (D.K.); (S.K.); Tel.: +82-51-320-1972 (D.K.); +82-63-220-2384 (S.K.)
| | - Sooah Kim
- Department of Environment Science & Biotechnology, Jeonju University, Jeonju 55069, Korea; (T.B.); (M.Z.)
- Correspondence: (D.K.); (S.K.); Tel.: +82-51-320-1972 (D.K.); +82-63-220-2384 (S.K.)
| |
Collapse
|
24
|
Holzknecht J, Dubrac S, Hedtrich S, Galgóczy L, Marx F. Small, Cationic Antifungal Proteins from Filamentous Fungi Inhibit Candida albicans Growth in 3D Skin Infection Models. Microbiol Spectr 2022; 10:e0029922. [PMID: 35499318 PMCID: PMC9241769 DOI: 10.1128/spectrum.00299-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/01/2022] [Indexed: 12/19/2022] Open
Abstract
The emerging resistance of human-pathogenic fungi to antifungal drugs urges the development of alternative therapeutic strategies. The small, cationic antifungal proteins (AFPs) from filamentous ascomycetes represent promising candidates for next-generation antifungals. These bio-molecules need to be tested for tolerance in the host and efficacy against fungal pathogens before they can be safely applied in humans. Testing of the efficacy and possible adverse effects of new drug candidates in three-dimensional (3D) human-cell based models represents an advantageous alternative to animal experiments. In, this study, as a proof-of-principle, we demonstrate the usefulness of 3D skin infection models for screening new antifungal drug candidates for topical application. We established a cutaneous infection with the opportunistic human-pathogenic yeast Candida albicans in a commercially available 3D full-thickness (FT) skin model to test the curative potential of distinct AFPs from Penicillium chrysogenum (PAFopt, PAFB, and PAFC) and Neosartorya (Aspergillus) fischeri (NFAP2) in vitro. All tested AFPs were comparably well tolerated by the skin models. The infected 3D models exhibited reduced epidermal permeability barriers, allowing C. albicans to colonize the epidermal and dermal layers, and showed increased secretion of the pro-inflammatory cytokine IL-6 and the chemokine IL-8. AFP treatment diminished the fungal burden and penetration depth of C. albicans in the infected models. The epidermal permeability barrier was restored and the secretion of IL-8 was decreased following AFP treatment. In summary, our study proves that the tested AFPs exhibit antifungal potential against cutaneous C. albicans infection in a 3D FT skin model. IMPORTANCE Candida albicans represents one of the most prevalent opportunistic fungal pathogens, causing superficial skin and mucosal infections in humans with certain predisposing health conditions and life-threatening systemic infections in immunosuppressed patients. The emerging drug resistance of this human-pathogenic yeast and the limited number of antifungal drugs for prevention and treatment of infections urgently demands the identification of new antifungal compounds with novel mechanisms of action. Small, cationic antifungal proteins (AFPs) from filamentous fungi represent promising candidates for next-generation antifungals for topical application. These bio-molecules need to be tested for tolerance by the host and efficacy in pathogen clearance prior to being involved in clinical trials. In a proof-of-principle study, we provide evidence for the suitability of 3D human-cell based models as advantageous alternatives to animal experiments. We document the tolerance of specific AFPs and their curative efficacy against cutaneous C. albicans infection in a 3D skin model.
Collapse
Affiliation(s)
- Jeanett Holzknecht
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sandrine Dubrac
- Department of Dermatology, Venereology and Allergology, Medical University of Innsbruck, Innsbruck, Austria
| | - Sarah Hedtrich
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, British Columbia, Canada
| | - László Galgóczy
- Department of Biotechnology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
- Institute of Biochemistry, Biological Research Centre, Eötvös Loránd Research Network, Szeged, Hungary
| | - Florentine Marx
- Biocenter, Institute of Molecular Biology, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
25
|
Sutterby E, Thurgood P, Baratchi S, Khoshmanesh K, Pirogova E. Evaluation of in vitro human skin models for studying effects of external stressors and stimuli and developing treatment modalities. VIEW 2022. [DOI: 10.1002/viw.20210012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Affiliation(s)
- Emily Sutterby
- School of Engineering RMIT University Melbourne Victoria Australia
| | - Peter Thurgood
- School of Engineering RMIT University Melbourne Victoria Australia
| | - Sara Baratchi
- School of Health and Biomedical Sciences RMIT University Bundoora Victoria Australia
| | | | - Elena Pirogova
- School of Engineering RMIT University Melbourne Victoria Australia
| |
Collapse
|
26
|
Mallick S, Nag M, Lahiri D, Pandit S, Sarkar T, Pati S, Nirmal NP, Edinur HA, Kari ZA, Ahmad Mohd Zain MR, Ray RR. Engineered Nanotechnology: An Effective Therapeutic Platform for the Chronic Cutaneous Wound. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:778. [PMID: 35269266 PMCID: PMC8911807 DOI: 10.3390/nano12050778] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Revised: 02/02/2022] [Accepted: 02/06/2022] [Indexed: 12/27/2022]
Abstract
The healing of chronic wound infections, especially cutaneous wounds, involves a complex cascade of events demanding mutual interaction between immunity and other natural host processes. Wound infections are caused by the consortia of microbial species that keep on proliferating and produce various types of virulence factors that cause the development of chronic infections. The mono- or polymicrobial nature of surface wound infections is best characterized by its ability to form biofilm that renders antimicrobial resistance to commonly administered drugs due to poor biofilm matrix permeability. With an increasing incidence of chronic wound biofilm infections, there is an urgent need for non-conventional antimicrobial approaches, such as developing nanomaterials that have intrinsic antimicrobial-antibiofilm properties modulating the biochemical or biophysical parameters in the wound microenvironment in order to cause disruption and removal of biofilms, such as designing nanomaterials as efficient drug-delivery vehicles carrying antibiotics, bioactive compounds, growth factor antioxidants or stem cells reaching the infection sites and having a distinct mechanism of action in comparison to antibiotics-functionalized nanoparticles (NPs) for better incursion through the biofilm matrix. NPs are thought to act by modulating the microbial colonization and biofilm formation in wounds due to their differential particle size, shape, surface charge and composition through alterations in bacterial cell membrane composition, as well as their conductivity, loss of respiratory activity, generation of reactive oxygen species (ROS), nitrosation of cysteines of proteins, lipid peroxidation, DNA unwinding and modulation of metabolic pathways. For the treatment of chronic wounds, extensive research is ongoing to explore a variety of nanoplatforms, including metallic and nonmetallic NPs, nanofibers and self-accumulating nanocarriers. As the use of the magnetic nanoparticle (MNP)-entrenched pre-designed hydrogel sheet (MPS) is found to enhance wound healing, the bio-nanocomposites consisting of bacterial cellulose and magnetic nanoparticles (magnetite) are now successfully used for the healing of chronic wounds. With the objective of precise targeting, some kinds of "intelligent" nanoparticles are constructed to react according to the required environment, which are later incorporated in the dressings, so that the wound can be treated with nano-impregnated dressing material in situ. For the effective healing of skin wounds, high-expressing, transiently modified stem cells, controlled by nano 3D architectures, have been developed to encourage angiogenesis and tissue regeneration. In order to overcome the challenge of time and dose constraints during drug administration, the approach of combinatorial nano therapy is adopted, whereby AI will help to exploit the full potential of nanomedicine to treat chronic wounds.
Collapse
Affiliation(s)
- Suhasini Mallick
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia 741249, India;
| | - Moupriya Nag
- Department of Biotechnology, University of Engineering & Management, Kolkata 700156, India; (M.N.); (D.L.)
| | - Dibyajit Lahiri
- Department of Biotechnology, University of Engineering & Management, Kolkata 700156, India; (M.N.); (D.L.)
| | - Soumya Pandit
- Department of Life Sciences, Sharda University, Noida 201310, India;
| | - Tanmay Sarkar
- Department of Food Processing Technology, Malda Polytechnic, West Bengal State Council of Technical Education, Government of West Bengal, Malda 732102, India;
| | - Siddhartha Pati
- NatNov Bioscience Private Limited, Balasore 756001, India;
- Skills Innovation & Academic Network (SIAN) Institute, Association for Biodiversity Conservation & Research (ABC), Balasore 756001, India
| | - Nilesh Prakash Nirmal
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand;
| | - Hisham Atan Edinur
- School of Health Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian 16150, Malaysia;
| | - Zulhisyam Abdul Kari
- Department of Agricultural Science, Faculty of Agro-Based Industry, Universiti Malaysia Kelantan, Jeli 17600, Malaysia
| | | | - Rina Rani Ray
- Department of Biotechnology, Maulana Abul Kalam Azad University of Technology, Nadia 741249, India;
| |
Collapse
|
27
|
Jayasinghe AMK, Kirindage KGIS, Fernando IPS, Han EJ, Oh GW, Jung WK, Ahn G. Fucoidan Isolated from Sargassum confusum Suppresses Inflammatory Responses and Oxidative Stress in TNF-α/IFN-γ- Stimulated HaCaT Keratinocytes by Activating Nrf2/HO-1 Signaling Pathway. Mar Drugs 2022; 20:117. [PMID: 35200646 PMCID: PMC8880602 DOI: 10.3390/md20020117] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/19/2022] [Accepted: 01/27/2022] [Indexed: 02/06/2023] Open
Abstract
Recent studies have revealed that marine brown seaweeds contain numerous bioactive compounds which exhibit various bioactivities. The present study investigated the effect of low molecular weight fucoidan (SCF) isolated from Sargassum confusum, a brown alga, on inflammatory responses and oxidative stress in HaCaT keratinocytes stimulated by tumor necrosis factor (TNF)-α/interferon (IFN)-γ. SCF significantly increased the cell viability while decreasing the intracellular reactive oxygen species (ROS) production in TNF-α/IFN-γ-stimulated HaCaT keratinocytes. In addition, SCF effectively reduced inflammatory cytokines (interleukin (IL)-1β, IL-6, IL-8, IL-13, TNF-α, and IFN-γ) and chemokines (Eotaxin, macrophage-derived chemokine (MDC), regulated on activation, normal T cell expressed and secreted (RANTES), and thymus and activation-regulated chemokine (TARC)) expression, by down-regulating the expression of epithelial and epidermal innate cytokines (IL-25, IL-33, and thymic stromal lymphopoietin (TSLP)). Furthermore, SCF suppressed the activation of TNF-α/IFN-γ-stimulated mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) signaling pathways, while activating the nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) signaling pathway. The cytoprotective effect of SCF against TNF-α/IFN-γ stimulation was considerably reduced upon inhibition of HO-1 activity by ZnPP. Overall, these results suggest that SCF effectively suppressed inflammatory responses and oxidative stress in TNF-α/IFN-γ-stimulated HaCaT keratinocytes via activating the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
| | | | | | - Eui Jeong Han
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu 59626, Korea; (A.M.K.J.); (K.G.I.S.K.); (E.J.H.)
- Research Center for Healthcare and Biomedical Engineering, Chonnam National University, Yeosu 59626, Korea
| | - Gun-Woo Oh
- Department of Biomedical Engineering and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, 45, Yongso-ro, Nam-gu, Busan 48513, Korea;
| | - Won-Kyo Jung
- Department of Biomedical Engineering and Center for Marine-Integrated Biomedical Technology (BK21 Plus), Pukyong National University, 45, Yongso-ro, Nam-gu, Busan 48513, Korea;
| | - Ginnae Ahn
- Department of Food Technology and Nutrition, Chonnam National University, Yeosu 59626, Korea; (A.M.K.J.); (K.G.I.S.K.); (E.J.H.)
- Department of Marine Bio-Food Sciences, Chonnam National University, Yeosu 59626, Korea;
| |
Collapse
|
28
|
Samaka RM, Marae A, Faried M, Bazid HAS. Light chain 3 immunoexpression in psoriasis. J Immunoassay Immunochem 2022; 43:365-383. [PMID: 34996338 DOI: 10.1080/15321819.2021.2018708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Autophagy dysregulation is involved in many diseases. The implication of autophagy in psoriasis pathogenesis is still uncertain. To investigate the role of Light Chain 3 (LC3), a good marker for autophagy, in psoriatic skin based on immunohistochemical study and correlate its expression - for the first time to the best of our knowledge - to clinicopathological data Prospective case-control study was conducted on 60 subjects (30 control, 30 psoriasis patients). Skin biopsies from control, lesional, and perilesional skin were processed for routine histopathological examination and LC3 immunoreaction assessment. There was a significant upregulation of the epidermal and dermal LC3 immunoreaction in the lesional skin compared with the control and perilesional skin specimens (P < .001). A significant positive correlation between the epidermal and dermal LC3 H scores in the lesional and perilesional skin was recorded. There was a non-significant relationship between the H score in the lesional skin and disease severity. LC3 could be considered in psoriasis pathogenesis; however, LC3 was not related to the severity of the disease. The findings might offer a novel target therapy for psoriasis patients.
Collapse
Affiliation(s)
- Rehab M Samaka
- Pathology Department, Faculty of Medicine, Menoufia University, Shebin Elkom, Egypt
| | - Alaa Marae
- Dermatology and Andrology Department, Menoufia University, Faculty of Medicine, Shebin El-Kom, Egypt
| | - Manar Faried
- Dermatology and Andrology Department, Menoufia University, Faculty of Medicine, Shebin El-Kom, Egypt
| | - Heba A S Bazid
- Dermatology and Andrology Department, Menoufia University, Faculty of Medicine, Shebin El-Kom, Egypt
| |
Collapse
|
29
|
Kang X, Lei J, Yang C, Zhang P, Li X, Zheng S, Li Q, Zhang J. A hybrid hydrogel composed of chitin and β-glucan for effectively management of wound healing and scarring. Biomater Sci 2022; 10:6024-6036. [DOI: 10.1039/d2bm00935h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Non-functional scar commonly forms after the skin injury. At present, most of the clinical treatments for scar eradication are typically with long treatment courses, low curative effects and expensive. In...
Collapse
|
30
|
Gao G, Ahn M, Cho WW, Kim BS, Cho DW. 3D Printing of Pharmaceutical Application: Drug Screening and Drug Delivery. Pharmaceutics 2021; 13:1373. [PMID: 34575448 PMCID: PMC8465948 DOI: 10.3390/pharmaceutics13091373] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 08/20/2021] [Accepted: 08/29/2021] [Indexed: 12/22/2022] Open
Abstract
Advances in three-dimensional (3D) printing techniques and the development of tailored biomaterials have facilitated the precise fabrication of biological components and complex 3D geometrics over the past few decades. Moreover, the notable growth of 3D printing has facilitated pharmaceutical applications, enabling the development of customized drug screening and drug delivery systems for individual patients, breaking away from conventional approaches that primarily rely on transgenic animal experiments and mass production. This review provides an extensive overview of 3D printing research applied to drug screening and drug delivery systems that represent pharmaceutical applications. We classify several elements required by each application for advanced pharmaceutical techniques and briefly describe state-of-the-art 3D printing technology consisting of cells, bioinks, and printing strategies that satisfy requirements. Furthermore, we discuss the limitations of traditional approaches by providing concrete examples of drug screening (organoid, organ-on-a-chip, and tissue/organ equivalent) and drug delivery systems (oral/vaginal/rectal and transdermal/surgical drug delivery), followed by the introduction of recent pharmaceutical investigations using 3D printing-based strategies to overcome these challenges.
Collapse
Affiliation(s)
- Ge Gao
- Institute of Engineering Medicine, Beijing Institute of Technology, No. 5, South Street, Zhongguancun, Haidian District, Beijing 100081, China;
| | - Minjun Ahn
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Won-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| | - Byoung-Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, 49 Busandaehak-ro, Mulgeum-eup, Yangsan 50612, Kyungbuk, Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, POSTECH, 77 Cheongam-ro, Nam-gu, Pohang 37673, Kyungbuk, Korea; (M.A.); (W.-W.C.)
| |
Collapse
|
31
|
Enkhtaivan E, Lee CH. Role of Amine Neurotransmitters and Their Receptors in Skin Pigmentation: Therapeutic Implication. Int J Mol Sci 2021; 22:ijms22158071. [PMID: 34360837 PMCID: PMC8348573 DOI: 10.3390/ijms22158071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 07/09/2021] [Accepted: 07/24/2021] [Indexed: 12/27/2022] Open
Abstract
Skin pigmentation can occur due to increased melanin, including melanocyte proliferation, melanin biosynthesis, or melanocyte migration. There are many factors that influence the melanin production process, but the role of neurotransmitters in this process is still unclear. We found that histamine and serotonin influence the different stages of melanogenesis and melanogenesis, which increase melanogenesis. Since then, several related papers have been published, and from these papers, it has been recognised that the role of neurotransmitters in skin-pigment-related diseases needs to be summarised. By introducing the role of neurotransmitters in the regulation of various pigment disorders, including vitiligo and melasma, through this review, many researchers can be expected to try to apply neurotransmitter-related agonists and antagonists as treatments for skin pigment disorders.
Collapse
|
32
|
Murakami M, Akagi T, Sasano Y, Akashi M. Effect of 3D-Fibroblast Dermis Constructed by Layer-by-Layer Cell Coating Technique on Tight Junction Formation and Function in Full-Thickness Skin Equivalent. ACS Biomater Sci Eng 2021; 7:3835-3844. [PMID: 34286576 DOI: 10.1021/acsbiomaterials.1c00375] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human skin equivalents (HSEs) consisting of an epidermis and dermis have been used as promising tools for drug evaluation and for clinical applications in regenerative medicine. Normal human dermal fibroblasts (NHDFs) are essential for the fabrication of HSEs because they play an important role in the maturation of the epidermis. Recently, epidermal tight junctions (TJs), which are complex cell-cell junctions, have attracted much attention as a second barrier and regulator for other barrier functions. In a previous study, we revealed the expression of TJ-related proteins and the time course of formation of TJ structure in the HSE (layer-by-layer (LbL)-three-dimensional (3D) Skin) constructed by layer-by-layer (LbL) cell coating technique that have a unique dermis consisting of NHDFs only (3D-fibroblast dermis). However, the effect of the 3D-fibroblast dermis on the formation of functional epidermal TJs is unknown. In this study, we investigated the effect of the 3D-fibroblast dermis on the expression of TJ-related proteins and TJ function in LbL-3D Skin. We demonstrated that the 3D-fibroblast dermis affects the long-term expression of TJ-related proteins and the formation of TJ with barrier function in the epidermis. These results show that the 3D-fibroblast dermis in LbL-3D Skin contributes to the formation and maintenance of functional TJs as in native human skin by direct contact with KCs.
Collapse
Affiliation(s)
- Masato Murakami
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Takami Akagi
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| | - Yumi Sasano
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan.,Pharma-Medicals Division, Life & Healthcare Products Department, Nagase & Co., Ltd., 2-2-3 Murotani, Nishi-ku, Kobe, Hyogo 651-2241, Japan
| | - Mitsuru Akashi
- Department of Frontier Biosciences, Graduate School of Frontier Biosciences, Osaka University, 1-3 Yamadaoka, Suita, Osaka 565-0871, Japan
| |
Collapse
|
33
|
Banerjee K, Madhyastha R, Nakajima Y, Maruyama M, Madhyastha H. Nanoceutical Adjuvants as Wound Healing Material: Precepts and Prospects. Int J Mol Sci 2021; 22:4748. [PMID: 33947121 PMCID: PMC8124138 DOI: 10.3390/ijms22094748] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 02/07/2023] Open
Abstract
Dermal wound healing describes the progressive repair and recalcitrant mechanism of 12 damaged skin, and eventually, reformatting and reshaping the skin. Many probiotics, nutritional supplements, metal nanoparticles, composites, skin constructs, polymers, and so forth have been associated with the improved healing process of wounds. The exact mechanism of material-cellular interaction is a point of immense importance, particularly in pathological conditions such as diabetes. Bioengineered alternative agents will likely continue to dominate the outpatient and perioperative management of chronic, recalcitrant wounds as new products continue to cut costs and improve the wound healing process. This review article provides an update on the various remedies with confirmed wound healing activities of metal-based nanoceutical adjuvanted agents and also other nano-based counterparts from previous experiments conducted by various researchers.
Collapse
Affiliation(s)
- Kaushita Banerjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632014, India;
| | - Radha Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan; (R.M.); (Y.N.); (M.M.)
| | - Yuichi Nakajima
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan; (R.M.); (Y.N.); (M.M.)
| | - Masugi Maruyama
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan; (R.M.); (Y.N.); (M.M.)
| | - Harishkumar Madhyastha
- Department of Applied Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 8891692, Japan; (R.M.); (Y.N.); (M.M.)
| |
Collapse
|
34
|
Baltanás FC, Mucientes-Valdivieso C, Lorenzo-Martín LF, Fernández-Parejo N, García-Navas R, Segrelles C, Calzada N, Fuentes-Mateos R, Paramio JM, Bustelo XR, Santos E. Functional Specificity of the Members of the Sos Family of Ras-GEF Activators: Novel Role of Sos2 in Control of Epidermal Stem Cell Homeostasis. Cancers (Basel) 2021; 13:cancers13092152. [PMID: 33946974 PMCID: PMC8124217 DOI: 10.3390/cancers13092152] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/26/2021] [Accepted: 04/27/2021] [Indexed: 12/18/2022] Open
Abstract
Simple Summary The Sos Ras-GEFs are known to participate in a wide range of skin-related diseases including cutaneous cancers, cardio-facio-cutaneous syndromes, or hirsutism. However, the specific functional roles played by the Sos1 and/or Sos2 family members in specific skin compartments remain largely unknown. This report aimed at precisely characterizing the specific functions played by Sos1 and/or Sos2 in keratinocytes, an essential cellular component of the skin. Our data show that Sos1 and Sos2 make overlapping contributions to both keratinocyte proliferation and survival. However, Sos1 seems to have a preferential involvement in regulating the ERK axis, whereas Sos2 seems to control the signaling output from the PI3K axis. We also uncovered an essential role of Sos2 in the control of the population of epidermal stem cells. Abstract Prior reports showed the critical requirement of Sos1 for epithelial carcinogenesis, but the specific functionalities of the homologous Sos1 and Sos2 GEFs in skin homeostasis and tumorigenesis remain unclear. Here, we characterize specific mechanistic roles played by Sos1 or Sos2 in primary mouse keratinocytes (a prevalent skin cell lineage) under different experimental conditions. Functional analyses of actively growing primary keratinocytes of relevant genotypes—WT, Sos1-KO, Sos2-KO, and Sos1/2-DKO—revealed a prevalent role of Sos1 regarding transcriptional regulation and control of RAS activation and mechanistic overlapping of Sos1 and Sos2 regarding cell proliferation and survival, with dominant contribution of Sos1 to the RAS-ERK axis and Sos2 to the RAS-PI3K/AKT axis. Sos1/2-DKO keratinocytes could not grow under 3D culture conditions, but single Sos1-KO and Sos2-KO keratinocytes were able to form pseudoepidermis structures that showed disorganized layer structure, reduced proliferation, and increased apoptosis in comparison with WT 3D cultures. Remarkably, analysis of the skin of both newborn and adult Sos2-KO mice uncovered a significant reduction of the population of stem cells located in hair follicles. These data confirm that Sos1 and Sos2 play specific, cell-autonomous functions in primary keratinocytes and reveal a novel, essential role of Sos2 in control of epidermal stem cell homeostasis.
Collapse
Affiliation(s)
- Fernando C. Baltanás
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Correspondence: (F.C.B.); (E.S.)
| | - Cynthia Mucientes-Valdivieso
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - L. Francisco Lorenzo-Martín
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Natalia Fernández-Parejo
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Rósula García-Navas
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Carmen Segrelles
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Molecular Oncology Division, CIEMAT and Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, E-28040 Madrid, Spain
| | - Nuria Calzada
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Rocío Fuentes-Mateos
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Jesús M. Paramio
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Molecular Oncology Division, CIEMAT and Instituto de Investigación Sanitaria Hospital Universitario 12 de Octubre, E-28040 Madrid, Spain
| | - Xosé R. Bustelo
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
| | - Eugenio Santos
- Mechanisms of Cancer Program, Centro de Investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.M.-V.); (L.F.L.-M.); (N.F.-P.); (R.G.-N.); (N.C.); (R.F.-M.); (X.R.B.)
- Mechanisms of Tumor Progression Program, CIBERONC, University of Salamanca-CSIC, E-37007 Salamanca, Spain; (C.S.); (J.M.P.)
- Correspondence: (F.C.B.); (E.S.)
| |
Collapse
|
35
|
Engineering of diseased human skin equivalent using 3D cell printing for representing pathophysiological hallmarks of type 2 diabetes in vitro. Biomaterials 2021; 272:120776. [PMID: 33798956 DOI: 10.1016/j.biomaterials.2021.120776] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 02/26/2021] [Accepted: 03/21/2021] [Indexed: 02/06/2023]
Abstract
Despite many significant advances in 3D cell printing for skin, a disease model displaying the pathological processes present in the native skin has not been reported yet. Therefore, we were motivated for modeling a 3D diseased skin tissue with pathophysiological hallmarks of type 2 diabetes in vitro based on 3D cell printing technique. By stimulating epidermal-dermal intercellular crosstalk found in the native skin, it was hypothesized that normal keratinocytes would be differentiated as diabetic epidermis when interacting with the diabetic dermal compartment. To prove this, a novel wounded skin model was successfully devised during tissue maturation in vitro. Interestingly, the slow re-epithelization was observed in our diabetic model, which is a representative hallmark of diabetic skin. Using the versatility of 3D cell printing, the structural similarities and diabetic properties of the model were further augmented by addition of perfusable vascularized diabetic hypodermis. Insulin resistance, adipocyte hypertrophy, inflammatory reactions, and vascular dysfunction, as the typical hallmarks in diabetes, were found under hyperglycemia. Finally, the feasibility of this new disease model for drug development was successfully demonstrated through application of test drugs. We trust that this study provides a pioneering step towards 3D cell printing-based in vitro skin disease modeling.
Collapse
|
36
|
Serra F, Aielli L, Costantini E. The role of miRNAs in the inflammatory phase of skin wound healing. AIMS ALLERGY AND IMMUNOLOGY 2021. [DOI: 10.3934/allergy.2021020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
<abstract>
<p>Wound healing (WH) is a fundamental physiological process to keep the integrity of the skin, therefore impaired and chronic WH is a common and severe medical problem and represent one of the biggest challenges of public health. The resolution of the WH inflammatory phase is characterized by a complex series of events that involves many cellular types, especially neutrophils, macrophages and inflammatory mediators, which are crucial for a correct wound closure. MicroRNAs (miRNAs) play essential roles in wound repair. In fact, miR-142 is linked to inflammation modulating neutrophils' chemotaxis and polarization, while the polarization of M1 toward the M2 phenotype is driven by miR-223 and miR-132 is linked to chemokines and cytokines that activate endothelial cells and attract leukocytes and peripheral cells to the damage site. Thus, understanding the dysregulation of miRNAs in WH will be decisive for the development of new and more effective therapies for the management of chronic wounds.</p>
</abstract>
Collapse
|
37
|
Zhang S, Ke Z, Yang C, Zhou P, Jiang H, Chen L, Li Y, Li Q. High Glucose Causes Distinct Expression Patterns of Primary Human Skin Cells by RNA Sequencing. Front Endocrinol (Lausanne) 2021; 12:603645. [PMID: 33763026 PMCID: PMC7982678 DOI: 10.3389/fendo.2021.603645] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/04/2021] [Indexed: 11/30/2022] Open
Abstract
Diabetes-related skin problems represent the most common long-term complications in diabetes mellitus patients. These complications, which include diabetic dermopathy, diabetic blisters, necrobiosis lipoidica diabeticorum, and eruptive xanthomatosis, may dramatically impair patients' quality of life and cause long-lasting disability. However, the cellular and molecular mechanisms linking diabetes-related hyperglycemia and skin complications are still incompletely understood. To assess the role of the various skin-cell types in hyperglycemia-induced skin disorders, we performed RNA sequencing-based transcriptome analysis, measuring gene expression patterns in biological replicates in normal- and high glucose-stimulated skin cells. Three primary human skin-cell types were examined, i.e., epidermal keratinocytes, dermal fibroblasts, and dermal microvascular endothelial cells. For each separate cell type, we identified gene expression. Comparing gene abundances and expression levels revealed that transcription profiles exhibit distinct patterns in the three skin-cell types exposed to normal (i.e., physiological) glucose treatment and high (i.e., supraphysiological) glucose treatment. The obtained data indicate that high glucose induced differential gene expression and distinct activity patterns in signaling pathways in each skin-cell type. We are adding these data to the public database in the hope that they will facilitate future studies to develop novel targeted interventions for diabetic skin complications.
Collapse
Affiliation(s)
- Shan Zhang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zunxiang Ke
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chao Yang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Huanzong Jiang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Chen
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yiqing Li, ; Qin Li,
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Yiqing Li, ; Qin Li,
| |
Collapse
|
38
|
Mendibil U, Ruiz-Hernandez R, Retegi-Carrion S, Garcia-Urquia N, Olalde-Graells B, Abarrategi A. Tissue-Specific Decellularization Methods: Rationale and Strategies to Achieve Regenerative Compounds. Int J Mol Sci 2020; 21:E5447. [PMID: 32751654 PMCID: PMC7432490 DOI: 10.3390/ijms21155447] [Citation(s) in RCA: 160] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/25/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) is a complex network with multiple functions, including specific functions during tissue regeneration. Precisely, the properties of the ECM have been thoroughly used in tissue engineering and regenerative medicine research, aiming to restore the function of damaged or dysfunctional tissues. Tissue decellularization is gaining momentum as a technique to obtain potentially implantable decellularized extracellular matrix (dECM) with well-preserved key components. Interestingly, the tissue-specific dECM is becoming a feasible option to carry out regenerative medicine research, with multiple advantages compared to other approaches. This review provides an overview of the most common methods used to obtain the dECM and summarizes the strategies adopted to decellularize specific tissues, aiming to provide a helpful guide for future research development.
Collapse
Affiliation(s)
- Unai Mendibil
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 Donostia-San Sebastian, Spain; (N.G.-U.); (B.O.-G.)
| | - Raquel Ruiz-Hernandez
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
| | - Sugoi Retegi-Carrion
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
| | - Nerea Garcia-Urquia
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 Donostia-San Sebastian, Spain; (N.G.-U.); (B.O.-G.)
| | - Beatriz Olalde-Graells
- TECNALIA, Basque Research and Technology Alliance (BRTA), 20009 Donostia-San Sebastian, Spain; (N.G.-U.); (B.O.-G.)
| | - Ander Abarrategi
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), 20014 Donostia-San Sebastian, Spain; (U.M.); (R.R.-H.); (S.R.-C.)
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
| |
Collapse
|