1
|
Jung K, Ku JY, Kwon JS, Won G, Yoon H, Oh SI, Kim MH, Kim C, Yoon JS. IFN-γ/TNF-α Synergism Induces Pro-Inflammatory Cytokine and Chemokine Production by In Vitro Canine Keratinocytes. Vet Sci 2025; 12:55. [PMID: 39852930 PMCID: PMC11769522 DOI: 10.3390/vetsci12010055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/10/2025] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Activated keratinocytes play a crucial role in skin inflammation through the production of multiple inflammatory mediators; however, little is known about cytokine secretion by activated keratinocytes in dogs. This study aimed to investigate the effects of the Th1 and Th2 types of cytokines on the production of keratinocyte-derived inflammatory mediators. Canine progenitor epidermal keratinocytes (CPEKs) were incubated with canine recombinant IL-4, IL-13, an IL4/IL13 mixture, IFN-γ, TNF-α, or an IFN-γ/TNF-α mixture for 24 h following 100% confluency. Culture supernatants were analyzed for cytokine concentration, including chemokine ligand (CXCL) 8, IL-10, IL-6, IL-1ß, IL-12, and chemokine ligand 2 (CCL2) by enzyme-linked immunosorbent assay. CPEKs incubated with the IFN-γ/TNF-α mixture showed significantly increased IL-6 concentration. In addition, significantly increased concentrations of CXCL8 were detected in CPEKs incubated with TNF-α and with the IFN-γ/TNF-α mixture. CCL2 concentrations increased in cells incubated with IFN-γ, TNF-α, and the IFN-γ/TNF-α mixture. The IFN-γ/TNF-α mixture synergistically enhanced CCL2 production. Dose-dependent elevations were also observed in IL-6 in response to the IFN-γ/TNF-α mixture, and in CCL2 in response to IFN-γ, TNF-α, and the IFN-γ/TNF-α mixture. These findings indicate that IFN-γ and TNF-α synergistically increase pro-inflammatory cytokines and chemokines secreted by canine keratinocytes. This in vitro culture system could be useful to investigate cytokine-mediated crosstalk between keratinocytes and immune cells and new therapeutic strategies for keratinocyte-mediated inflammatory skin diseases.
Collapse
Affiliation(s)
- Kyungsook Jung
- Functional Biomaterials Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup 56212, Republic of Korea;
| | - Ji-Yeong Ku
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan 54896, Republic of Korea; (J.-Y.K.); (J.-S.K.); (G.W.); (H.Y.); (S.-I.O.)
| | - Je-Seong Kwon
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan 54896, Republic of Korea; (J.-Y.K.); (J.-S.K.); (G.W.); (H.Y.); (S.-I.O.)
| | - Gayeon Won
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan 54896, Republic of Korea; (J.-Y.K.); (J.-S.K.); (G.W.); (H.Y.); (S.-I.O.)
| | - Hakyoung Yoon
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan 54896, Republic of Korea; (J.-Y.K.); (J.-S.K.); (G.W.); (H.Y.); (S.-I.O.)
| | - Sang-Ik Oh
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan 54896, Republic of Korea; (J.-Y.K.); (J.-S.K.); (G.W.); (H.Y.); (S.-I.O.)
| | - Mi Hye Kim
- College of Korean Medicine, Woosuk University, Wanju 55338, Republic of Korea;
| | - Chongchan Kim
- Korea Thumb Vet Co., Ltd., 470-15 Seonhwa-ro, Iksan 54631, Republic of Korea;
| | - Ji-Seon Yoon
- Biosafety Research Institute and College of Veterinary Medicine, Jeonbuk National University, Iksan 54896, Republic of Korea; (J.-Y.K.); (J.-S.K.); (G.W.); (H.Y.); (S.-I.O.)
| |
Collapse
|
2
|
Park YJ, Grossman J, Robertson L. Severe atopic dermatitis treated with Dupilumab in a CTLA-4-deficient patient: A case report and review of the literature. SAGE Open Med Case Rep 2025; 13:2050313X241311042. [PMID: 39777027 PMCID: PMC11705363 DOI: 10.1177/2050313x241311042] [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: 10/13/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
Atopic dermatitis is a chronic inflammatory skin disease associated with immune dysregulation, particularly overexpression of T helper 2 cytokines. Cytotoxic T lymphocyte-associated antigen 4 deficiency, a primary immune disorder, can exacerbate atopic dermatitis. Dupilumab, an IL-4 and IL-13 receptor antagonist, has demonstrated efficacy in controlling severe, recalcitrant atopic dermatitis by mitigating T helper 2-driven inflammation. We present a case of a 24-year-old male with cytotoxic T lymphocyte-associated antigen 4 haploinsufficiency and severe atopic dermatitis successfully managed with Dupilumab. The patient showed marked improvement in eczema severity scores, including a sixfold reduction in the Eczema Area and Severity Index and a threefold reduction in the Dermatology Life Quality Index over 6 months, highlighting Dupilumab's potential role in cytotoxic T lymphocyte-associated antigen 4-deficient patients experiencing atopic dermatitis.
Collapse
Affiliation(s)
- Ye-Jean Park
- Temerty Faculty of Medicine, Division of Medicine, Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jennifer Grossman
- Division of Hematology, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Lynne Robertson
- Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Division of Dermatology, Department of Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
3
|
Mallick S, Duttaroy AK, Bose B. A Snapshot of Cytokine Dynamics: A Fine Balance Between Health and Disease. J Cell Biochem 2025; 126:e30680. [PMID: 39668456 DOI: 10.1002/jcb.30680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 10/23/2024] [Accepted: 11/08/2024] [Indexed: 12/14/2024]
Abstract
Health and disease are intricately intertwined and often determined by the delicate balance of biological processes. Cytokines, a family of small signalling molecules, are pivotal in maintaining this balance, ensuring the body's immune system functions optimally. In a healthy condition, cytokines act as potent mediators of immune responses. They orchestrate the activities of immune cells, coordinating their proliferation, differentiation, and migration. This intricate role of cytokine signalling enables the body to effectively combat infections, repair damaged tissues, and regulate inflammation. However, the delicate equilibrium of cytokine production is susceptible to disruption. Excessive or abnormal cytokine levels can lead to a cascade of pathological conditions, including autoimmune diseases, chronic inflammation, infections, allergies, and even cancer. Interestingly, from the bunch of cytokines, few cytokines play an essential role in maintaining the balance between normal physiological status and diseases. In this review, we have appraised key cytokines' potential role and feedback loops in augmenting the imbalances in the body's biological functions, presenting a critical link between inflammation and disease pathology. Moreover, we have also highlighted the significance of cytokines and their molecular interplay, particularly in the recent viral pandemic COVID-19 disease. Hence, understandings regarding the interplay between viral infection and cytokine responses are essential and fascinating for developing effective therapeutic strategies.
Collapse
Affiliation(s)
- Sumit Mallick
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, India
| | - Asim K Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Bipasha Bose
- Stem Cells and Regenerative Medicine Centre, Yenepoya Research Centre, Yenepoya (Deemed to be University), University Road, Deralakatte, Mangalore, Karnataka, India
| |
Collapse
|
4
|
Martínez-Navarrete M, Guillot AJ, Lobita MC, Recio MC, Giner R, Aparicio-Blanco J, Montesinos MC, Santos HA, Melero A. Cyclosporin A-loaded dissolving microneedles for dermatitis therapy: Development, characterisation and efficacy in a delayed-type hypersensitivity in vivo model. Drug Deliv Transl Res 2024; 14:3404-3421. [PMID: 38472726 PMCID: PMC11499354 DOI: 10.1007/s13346-024-01542-9] [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: 02/07/2024] [Indexed: 03/14/2024]
Abstract
Several drugs can be used for treating inflammatory skin pathologies like dermatitis and psoriasis. However, for the management of chronic and long-term cases, topical administration is preferred over oral delivery since it prevents certain issues due to systemic side effects from occurring. Cyclosporin A (CsA) has been used for this purpose; however, its high molecular weight (1202 Da) restricts the diffusion through the skin structure. Here, we developed a nano-in-micro device combining lipid vesicles (LVs) and dissolving microneedle array patches (DMAPs) for targeted skin delivery. CsA-LVs allowed the effective incorporation of CsA in the hydrophilic DMAP matrix despite the hydrophobicity of the drug. Polymeric matrix composed of poly (vinyl alcohol) (5% w/v), poly (vinyl pyrrolidine) (15% w/v) and CsA-LV dispersion (10% v/v) led to the formation of CsA-LVs@DMAPs with adequate mechanical properties to penetrate the stratum corneum barrier. The safety and biocompatibility were ensured in an in vitro viability test using HaCaT keratinocytes and L929 fibroblast cell lines. Ex vivo permeability studies in a Franz-diffusion cell setup showed effective drug retention in the skin structure. Finally, CsA-LVs@DMAPs were challenged in an in vivo murine model of delayed-type hypersensitivity to corroborate their potential to ameliorate skin inflammatory conditions. Different findings like photon emission reduction in bioluminescence study, normalisation of histological damage and decrease of inflammatory cytokines point out the effectivity of CsA-LVs@DMAPs to treat these conditions. Overall, our study demonstrates that CsA-LVs@DMAPs can downregulate the skin inflammatory environment which paves the way for their clinical translation and their use as an alternative to corticosteroid-based therapies.
Collapse
Affiliation(s)
- Miquel Martínez-Navarrete
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
| | - Antonio José Guillot
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain.
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| | - Maria C Lobita
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - María Carmen Recio
- Department of Pharmacology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
| | - Rosa Giner
- Department of Pharmacology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
| | - Juan Aparicio-Blanco
- Department of Pharmaceutics and Food Technology, Faculty of Pharmacy, Complutense University of Madrid, Plaza Ramón y Cajal s/n, 28040, Madrid, Spain
| | - María Carmen Montesinos
- Department of Pharmacology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
- Interuniversity Research Institute for Molecular Recognition and Technological Development (IDM), University of Valencia, Polytechnic University of Valencia, Valencia, Spain
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Ana Melero
- Department of Pharmacy and Pharmaceutical Technology and Parasitology, University of Valencia, Ave. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
| |
Collapse
|
5
|
Quílez C, Bebiano LB, Jones E, Maver U, Meesters L, Parzymies P, Petiot E, Rikken G, Risueño I, Zaidi H, Zidarič T, Bekeschus S, H van den Bogaard E, Caley M, Colley H, López NG, Letsiou S, Marquette C, Maver T, Pereira RF, Tobin DJ, Velasco D. Targeting the Complexity of In Vitro Skin Models: A Review of Cutting-Edge Developments. J Invest Dermatol 2024; 144:2650-2670. [PMID: 39127929 DOI: 10.1016/j.jid.2024.04.032] [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: 11/20/2023] [Revised: 02/29/2024] [Accepted: 04/10/2024] [Indexed: 08/12/2024]
Abstract
Skin in vitro models offer much promise for research, testing drugs, cosmetics, and medical devices, reducing animal testing and extensive clinical trials. There are several in vitro approaches to mimicking human skin behavior, ranging from simple cell monolayer to complex organotypic and bioengineered 3-dimensional models. Some have been approved for preclinical studies in cosmetics, pharmaceuticals, and chemicals. However, development of physiologically reliable in vitro human skin models remains in its infancy. This review reports on advances in in vitro complex skin models to study skin homeostasis, aging, and skin disease.
Collapse
Affiliation(s)
- Cristina Quílez
- Bioengineering Department, Universidad Carlos III de Madrid, Leganés, Spain; Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Luís B Bebiano
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal
| | - Eleri Jones
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Uroš Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia; Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Luca Meesters
- Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Piotr Parzymies
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Emma Petiot
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| | - Gijs Rikken
- Department of Dermatology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ignacio Risueño
- Bioengineering Department, Universidad Carlos III de Madrid, Leganés, Spain; Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Hamza Zaidi
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| | - Tanja Zidarič
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Sander Bekeschus
- Clinic and Policlinic for Dermatology and Venerology, Rostock University Medical Center, Rostock, Germany; ZIK plasmatis, Leibniz Institute for Plasma Science and Technology (INP Greifswald), Greifswald, Germany
| | | | - Matthew Caley
- Centre for Cell Biology and Cutaneous Research, Blizard Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Helen Colley
- School of Clinical Dentistry, University of Sheffield, Sheffield, United Kingdom
| | - Nuria Gago López
- Melanoma group, Molecular Oncology Programme, Spanish National Cancer Research Center (CNIO), Madrid, Spain
| | - Sophia Letsiou
- Department of Biomedical Sciences, University of West Attica, Athens, Greece; Department of Food Science and Technology, University of West Attica, Athens, Greece
| | - Christophe Marquette
- 3d.FAB, CNRS, INSA, Univ Lyon, CPE-Lyon, UMR5246, ICBMS, Université Lyon 1, Villeurbanne Cedex, France
| | - Tina Maver
- Institute of Biomedical Sciences, Faculty of Medicine, University of Maribor, Maribor, Slovenia; Department of Pharmacology, Faculty of Medicine, University of Maribor, Maribor, Slovenia
| | - Rúben F Pereira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal; Instituto de Engenharia Biomédica (INEB), Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Desmond J Tobin
- Charles Institute of Dermatology, University College Dublin, Dublin, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| | - Diego Velasco
- Bioengineering Department, Universidad Carlos III de Madrid, Leganés, Spain; Fundación Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain.
| |
Collapse
|
6
|
Zhou S, Li R, Sun J, Gu M, Gao D, Tang L, Zhu J. Construction of a pumpless gravity-driven vascularized Skin-on-a-Chip for the study of hepatocytotoxicity in percutaneous exposure to exogenous chemicals. Biomed Microdevices 2024; 26:40. [PMID: 39302507 DOI: 10.1007/s10544-024-00723-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/12/2024] [Indexed: 09/22/2024]
Abstract
The utilization of existing Skin-on-a-Chip (SoC) is constrained by the complex structures, the multiplicity of auxiliary devices, and the inability to evaluate exogenous chemicals that are hepatotoxic after percutaneous metabolism. In this study, a gravity-driven SoC without any auxiliary devices was constructed for the hepatocytotoxicity study of exogenous chemicals. The SoC possesses 3 layers of culture chambers, from top to bottom, for human skin equivalent (HSE), Human Umbilical Vein Endothelial Cells (HUVEC) and hepatocytes (HepG2), and the maintenance and expression capacity of the corresponding cells on the SoC were verified by specificity parameters. The reactivity of the SoC to exogenous chemicals was verified by 2-aminofluorene (2-AF). The SoC can realistically simulate the in vivo exposure process of exogenous chemicals that are percutaneously exposed and metabolized into the bloodstream and then to the liver to produce toxicity, and it can achieve the same effects on transcriptome as those of animal tests at lower exposure levels while examining multiple toxicological targets of the skin, vascular endothelial cells, and hepatocytes. Both in terms of species similarity, the principles of reduction, replacement and refinement (3R), or the level of exposure suggest that the present SoC has a degree of replacement for animal models in assessing exogenous chemicals, especially those that are hepatotoxic after percutaneous metabolism.
Collapse
Affiliation(s)
- Su Zhou
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China
| | - Rui Li
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai, 201203, China
| | - Jie Sun
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai, 201203, China
| | - Minyang Gu
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai, 201203, China
| | - Dan Gao
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai, 201203, China
| | - Liming Tang
- Pharmacology and Toxicology Department, Shanghai Institute for Food and Drug Control, Shanghai, 201203, China
| | - Jiangbo Zhu
- Department of Health Toxicology, College of Naval Medicine, Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
7
|
Cho SW, Malick H, Kim SJ, Grattoni A. Advances in Skin-on-a-Chip Technologies for Dermatological Disease Modeling. J Invest Dermatol 2024; 144:1707-1715. [PMID: 38493383 DOI: 10.1016/j.jid.2024.01.031] [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: 10/30/2023] [Revised: 01/19/2024] [Accepted: 01/29/2024] [Indexed: 03/18/2024]
Abstract
Skin-on-a-chip (SoC) technologies are emerging as a paradigm shift in dermatology research by replicating human physiology in a dynamic manner not achievable by current animal models. Although animal models have contributed to successful clinical trials, their ability to predict human outcomes remains questionable, owing to inherent differences in skin anatomy and immune response. Covering areas including infectious diseases, autoimmune skin conditions, wound healing, drug toxicity, aging, and antiaging, SoC aims to circumvent the inherent disparities created by traditional models. In this paper, we review current SoC technologies, highlighting their potential as an alternative to animal models for a deeper understanding of complex skin conditions.
Collapse
Affiliation(s)
- Seo Won Cho
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA; Texas A&M University School of Medicine, College Station, Texas, USA
| | - Hamza Malick
- Texas A&M University School of Medicine, College Station, Texas, USA
| | - Soo Jung Kim
- Department of Dermatology, Baylor College of Medicine, Houston, Texas, USA
| | - Alessandro Grattoni
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, Texas, USA; Department of Surgery, Houston Methodist Hospital, Houston, Texas, USA; Department of Radiation Oncology, Houston Methodist Hospital, Houston, Texas, USA.
| |
Collapse
|
8
|
De Felice B, De Luca P, Montanino C, Mallardo M, Babino G, Mattera E, Sorbo R, Ragozzino G, Argenziano G, Daniele A, Nigro E. LncRNA microarray profiling identifies novel circulating lncRNAs in hidradenitis suppurativa. Mol Med Rep 2024; 30:112. [PMID: 38757342 PMCID: PMC11094584 DOI: 10.3892/mmr.2024.13236] [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: 05/17/2023] [Accepted: 02/13/2024] [Indexed: 05/18/2024] Open
Abstract
Long noncoding RNAs (lncRNAs) have been demonstrated to be involved in biological processes, both physiological and pathological, including cancer, cardiovascular diseases, multiple sclerosis, autoimmune hepatitis and types I and II diabetes. LncRNAs are also known to have a critical role in the physiology of skin, and in the pathology of cutaneous diseases. LncRNAs are involved in a wide range of biological activities, including transcriptional post‑transcriptional processes, epigenetics, RNA splicing, gene activation and or silencing, modifications and/or editing; therefore, lncRNAs may be useful as potential targets for disease treatment. Hidradenitis suppurativa (HS), also termed acne inversa, is a major skin disease, being an inflammatory disorder that affects ~1% of global population in a chronic manner. Its pathogenesis, however, is only partly understood, although immune dysregulation is known to have an important role. To investigate the biological relevance of lncRNAs with HS, the most differentially expressed lncRNAs and mRNAs were first compared. Furthermore, the lncRNA‑microRNA regulatory network was also defined via reverse transcription‑quantitative PCR analysis, whereby a trio of lncRNA expression signatures, lncRNA‑TINCR, lncRNA‑RBM5‑ASI1 and lncRNA‑MRPL23‑AS1, were found to be significantly overexpressed in patients with HS compared with healthy controls. In conclusion, the three lncRNAs isolated in the present study may be useful for improving the prognostic prediction of HS, as well as contributing towards an improved understanding of the underlying pathogenic mechanisms, thereby potentially providing new therapeutic targets.
Collapse
Affiliation(s)
- Bruna De Felice
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, I-81100 Caserta, Italy
| | | | - Concetta Montanino
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, I-81100 Caserta, Italy
| | - Marta Mallardo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, I-81100 Caserta, Italy
- CEINGE-Franco Salvatore Advanced Biotechnology, I-80145 Naples, Italy
| | - Graziella Babino
- Dermatology Unit, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Edi Mattera
- Department of Internal and Experimental Medicine and Surgery Unit of Internal Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Raffaele Sorbo
- Department of Internal and Experimental Medicine and Surgery Unit of Internal Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Giovanni Ragozzino
- Department of Internal and Experimental Medicine and Surgery Unit of Internal Medicine, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Giuseppe Argenziano
- Dermatology Unit, University of Campania Luigi Vanvitelli, I-80131 Naples, Italy
| | - Aurora Daniele
- CEINGE-Franco Salvatore Advanced Biotechnology, I-80145 Naples, Italy
- Department of Molecular Medicine and Medical Biotechnology, University of Naples Federico II, I-80131 Naples, Italy
| | - Ersilia Nigro
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania Luigi Vanvitelli, I-81100 Caserta, Italy
- CEINGE-Franco Salvatore Advanced Biotechnology, I-80145 Naples, Italy
| |
Collapse
|
9
|
Kim YK, Cho M, Kang DJ. Anti-Inflammatory Response of New Postbiotics in TNF-α/IFN-γ-Induced Atopic Dermatitis-like HaCaT Keratinocytes. Curr Issues Mol Biol 2024; 46:6100-6111. [PMID: 38921035 PMCID: PMC11203040 DOI: 10.3390/cimb46060364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024] Open
Abstract
This study examines the synergistic interaction between the immunomodulatory functions of lactic acid bacteria postbiotics and the anti-inflammatory properties of Smilax china L. extract through a combined fermentation process. Using atopic dermatitis (AD) as a model, characterized by an immune imbalance that leads to skin inflammation, we developed a fermented product, MB-2006, and compared its effects to those of the heat-killed probiotics Lactobacillus acidophilus (LAC) and Lactobacillus rhamnosus (LRH). Our experiments focused on elucidating the mechanism of action of MB-2006 in AD-like HaCaT keratinocyte cells, particularly its impact on the NF-κB pathway, a pivotal regulator of inflammation. MB-2006 proved more effective in reducing inflammation markers, such as IL-4 and thymic stromal lymphopoietin (TSLP), and in inhibiting NF-κB activation compared to LAC and LRH. Significantly, MB-2006 also reduced the expression of thymus- and activation-regulated chemokine (TARC), highlighting a synergistic effect that enhances its therapeutic potential. These results suggest that the combined fermentation of Smilax china L. extract with lactic acid bacteria enhanced both the anti-inflammatory and immunomodulatory effects, presenting a promising integrative approach to treating conditions like AD. Further studies are needed to validate these results in clinical settings and fully explore the potential of this synergistic fermentation process.
Collapse
Affiliation(s)
| | | | - Dae-Jung Kang
- MNH Bio Co., Ltd., Dongtan-Biz-Tower 609, Dongtancheomdansaneop 1-ro, Hwaseong-si 18469, Gyeonggi-do, Republic of Korea; (Y.-K.K.); (M.C.)
| |
Collapse
|
10
|
Kim S, Song Y, Kim J, Jeong B, Park N, Park YM, Kim YT, Rho D, Lee SJ, Choi BG, Im SG, Lee KG. Nanotopology-Enabled On-Site Pathogen Detection for Managing Atopic Dermatitis. Adv Healthc Mater 2024; 13:e2303272. [PMID: 38412280 DOI: 10.1002/adhm.202303272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/19/2024] [Indexed: 02/29/2024]
Abstract
Atopic dermatitis (AD), a prevalent skin condition often complicated by microbial infection, poses a significant challenge in identifying the responsible pathogen for its effective management. However, a reliable, safe tool for pinpointing the source of these infections remains elusive. In this study, a novel on-site pathogen detection that combines chemically functionalized nanotopology with genetic analysis is proposed to capture and analyze pathogens closely associated with severe atopic dermatitis. The chemically functionalized nanotopology features a 3D hierarchical nanopillar array (HNA) with a functional polymer coating, tailored to isolate target pathogens from infected skin. This innovative nanotopology demonstrates superior pathogenic capture efficiency, favorable entrapment patterns, and non-cytotoxicity. An HNA-assembled stick is utilized to directly retrieve bacteria from infected skin samples, followed by extraction-free quantitative loop-mediated isothermal amplification (direct qLAMP) for validation. To mimic human skin conditions, porcine skin is employed to successfully capture Staphylococcus aureus, a common bacterium exacerbating AD cases. The on-site detection method exhibits an impressive detection limit of 103 cells mL-1. The HNA-assembled stick represents a promising tool for on-site detection of bacteria associated with atopic dermatitis. This innovative approach enables to deepen the understanding of AD pathogenesis and open avenues for more effective management strategies for chronic skin conditions.
Collapse
Affiliation(s)
- Seongeun Kim
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Younseong Song
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jueun Kim
- Department of Chemical Engineering, Kangwon National University, Samcheok, 25913, Republic of Korea
| | - Booseok Jeong
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Nahyun Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Yoo Min Park
- Center for NanoBio Development, National NanoFab Center, Daejeon, 34141, Republic of Korea
| | - Yong Tae Kim
- Department of Chemical Engineering & Biotechnology, Tech University of Korea, Siheung-si, 15073, Republic of Korea
| | - Donggee Rho
- Center for NanoBio Development, National NanoFab Center, Daejeon, 34141, Republic of Korea
| | - Seok Jae Lee
- Center for NanoBio Development, National NanoFab Center, Daejeon, 34141, Republic of Korea
| | - Bong Gill Choi
- Department of Chemical Engineering, Kangwon National University, Samcheok, 25913, Republic of Korea
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Kyoung G Lee
- Center for NanoBio Development, National NanoFab Center, Daejeon, 34141, Republic of Korea
| |
Collapse
|
11
|
Asbóth D, Bánfi B, Kocsis D, Erdő F. Rodent models of dermatological disorders. Ital J Dermatol Venerol 2024; 159:303-317. [PMID: 38287740 DOI: 10.23736/s2784-8671.23.07700-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2024]
Abstract
To assess the possible beneficial effects of drugs and drug candidates, different dermatological disease models are available in rodents. These models are able to mimic one or more characteristic features of the disorders, but not completely recapitulate the pathogenesis of the human skin diseases. Therefore, to improve the technology many new models have been developed both by genetic engineering and by chemical or physical induction. Currently the in vivo rodent models provide the physiologically most relevant approach to produce the pathology related to the majority of dermatological diseases. In this short review some widely used animal techniques (psoriasis, allergic contact dermatitis, atopic dermatitis, wound healing, melanoma and non-melanoma type skin cancers and UV erythema) are shown which are currently applied in pharmacological, pharmacokinetic, pharmaceutical and dermatological research. First the main points of the human pathomechanism are shown and afterwards the rodent models are briefly discussed. Finally critical evaluation is provided by the authors. However, according to the 3R rule the number of experimental animals is strongly suggested to be reduced, therefore the advanced in vitro and ex vivo techniques become more and more important contrary to in vivo preclinical methods also in dermatological research. As it is described in the outlook section, although the 2D/3D in vitro and skin on-a-chip techniques are promising and have many advantages they are not able to completely substitute the animal models in their vascular, immunological, secretory and neural complexity.
Collapse
Affiliation(s)
- Dorottya Asbóth
- Pediatric Dermatology Center, Szent János Center Hospital in North Buda, Budapest, Hungary
| | - Barnabás Bánfi
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
- Faculty of Science, Eötvös Loránd University, Budapest, Hungary
| | - Dorottya Kocsis
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary -
| |
Collapse
|
12
|
Migayron L, Bordes S, Closs B, Seneschal J, Boniface K. Type-2 immunity associated with type-1 related skin inflammatory diseases: friend or foe? Front Immunol 2024; 15:1405215. [PMID: 38868763 PMCID: PMC11167106 DOI: 10.3389/fimmu.2024.1405215] [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: 03/22/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
Chronic inflammatory skin diseases are multifactorial diseases that combine genetic predisposition, environmental triggers, and metabolic disturbances associated with abnormal immune responses. From an immunological perspective, the better understanding of their physiopathology has demonstrated a large complex network of immune cell subsets and related cytokines that interact with both epidermal and dermal cells. For example, in type-1-associated diseases such as alopecia areata, vitiligo, and localized scleroderma, recent evidence suggests the presence of a type-2 inflammation that is well known in atopic dermatitis. Whether this type-2 immune response has a protective or detrimental impact on the development and chronicity of these diseases remains to be fully elucidated, highlighting the need to better understand its involvement for the management of patients. This mini-review explores recent insights regarding the potential role of type-2-related immunity in alopecia areata, vitiligo, and localized scleroderma.
Collapse
Affiliation(s)
- Laure Migayron
- Univ. Bordeaux, CNRS, Immuno ConcEpT, UMR 5164, Bordeaux, France
- R&D Department, SILAB, Brive-la-Gaillarde, France
| | | | | | - Julien Seneschal
- Univ. Bordeaux, CNRS, Immuno ConcEpT, UMR 5164, Bordeaux, France
- CHU de Bordeaux, Dermatology and Pediatric Dermatology, National Reference Center for Rare Skin Disorders, Hôpital Saint-André, UMR 5164, Bordeaux, France
| | - Katia Boniface
- Univ. Bordeaux, CNRS, Immuno ConcEpT, UMR 5164, Bordeaux, France
| |
Collapse
|
13
|
Morrison AI, Sjoerds MJ, Vonk LA, Gibbs S, Koning JJ. In vitro immunity: an overview of immunocompetent organ-on-chip models. Front Immunol 2024; 15:1373186. [PMID: 38835750 PMCID: PMC11148285 DOI: 10.3389/fimmu.2024.1373186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 04/30/2024] [Indexed: 06/06/2024] Open
Abstract
Impressive advances have been made to replicate human physiology in vitro over the last few years due to the growth of the organ-on-chip (OoC) field in both industrial and academic settings. OoCs are a type of microphysiological system (MPS) that imitates functional and dynamic aspects of native human organ biology on a microfluidic device. Organoids and organotypic models, ranging in their complexity from simple single-cell to complex multi-cell type constructs, are being incorporated into OoC microfluidic devices to better mimic human physiology. OoC technology has now progressed to the stage at which it has received official recognition by the Food and Drug Administration (FDA) for use as an alternative to standard procedures in drug development, such as animal studies and traditional in vitro assays. However, an area that is still lagging behind is the incorporation of the immune system, which is a critical element required to investigate human health and disease. In this review, we summarise the progress made to integrate human immunology into various OoC systems, specifically focusing on models related to organ barriers and lymphoid organs. These models utilise microfluidic devices that are either commercially available or custom-made. This review explores the difference between the use of innate and adaptive immune cells and their role for modelling organ-specific diseases in OoCs. Immunocompetent multi-OoC models are also highlighted and the extent to which they recapitulate systemic physiology is discussed. Together, the aim of this review is to describe the current state of immune-OoCs, the limitations and the future perspectives needed to improve the field.
Collapse
Affiliation(s)
- Andrew I. Morrison
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| | - Mirthe J. Sjoerds
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Leander A. Vonk
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Susan Gibbs
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit, Amsterdam, Netherlands
| | - Jasper J. Koning
- Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Amsterdam Institute for Infection and Immunity, Inflammatory Diseases, Amsterdam, Netherlands
| |
Collapse
|
14
|
Al B, Traidl S, Holzscheck N, Freimooser S, Mießner H, Reuter H, Dittrich-Breiholz O, Werfel T, Seidel JA. Single-cell RNA sequencing reveals 2D cytokine assay can model atopic dermatitis more accurately than immune-competent 3D setup. Exp Dermatol 2024; 33:e15077. [PMID: 38711200 DOI: 10.1111/exd.15077] [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: 01/08/2024] [Revised: 03/24/2024] [Accepted: 03/30/2024] [Indexed: 05/08/2024]
Abstract
Modelling atopic dermatitis (AD) in vitro is paramount to understand the disease pathophysiology and identify novel treatments. Previous studies have shown that the Th2 cytokines IL-4 and IL-13 induce AD-like features in keratinocytes in vitro. However, it has not been systematically researched whether the addition of Th2 cells, their supernatants or a 3D structure is superior to model AD compared to simple 2D cell culture with cytokines. For the first time, we investigated what in vitro option most closely resembles the disease in vivo based on single-cell RNA sequencing data (scRNA-seq) obtained from skin biopsies in a clinical study and published datasets of healthy and AD donors. In vitro models were generated with primary fibroblasts and keratinocytes, subjected to cytokine treatment or Th2 cell cocultures in 2D/3D. Gene expression changes were assessed using qPCR and Multiplex Immunoassays. Of all cytokines tested, incubation of keratinocytes and fibroblasts with IL-4 and IL-13 induced the closest in vivo-like AD phenotype which was observed in the scRNA-seq data. Addition of Th2 cells to fibroblasts failed to model AD due to the downregulation of ECM-associated genes such as POSTN. While keratinocytes cultured in 3D showed better stratification than in 2D, changes induced with AD triggers did not better resemble AD keratinocyte subtypes observed in vivo. Taken together, our comprehensive study shows that the simple model using IL-4 or IL-13 in 2D most accurately models AD in fibroblasts and keratinocytes in vitro, which may aid the discovery of novel treatment options.
Collapse
Affiliation(s)
- Benjamin Al
- Discovery, Beiersdorf AG, Hamburg, Germany
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | - Stephan Traidl
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | | | - Sina Freimooser
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | | | | | | | - Thomas Werfel
- Department of Dermatology and Allergy, Hannover Medical School, Hannover, Germany
| | | |
Collapse
|
15
|
Farhang Doost N, Srivastava SK. A Comprehensive Review of Organ-on-a-Chip Technology and Its Applications. BIOSENSORS 2024; 14:225. [PMID: 38785699 PMCID: PMC11118005 DOI: 10.3390/bios14050225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/09/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
Organ-on-a-chip (OOC) is an emerging technology that simulates an artificial organ within a microfluidic cell culture chip. Current cell biology research focuses on in vitro cell cultures due to various limitations of in vivo testing. Unfortunately, in-vitro cell culturing fails to provide an accurate microenvironment, and in vivo cell culturing is expensive and has historically been a source of ethical controversy. OOC aims to overcome these shortcomings and provide the best of both in vivo and in vitro cell culture research. The critical component of the OOC design is utilizing microfluidics to ensure a stable concentration gradient, dynamic mechanical stress modeling, and accurate reconstruction of a cellular microenvironment. OOC also has the advantage of complete observation and control of the system, which is impossible to recreate in in-vivo research. Multiple throughputs, channels, membranes, and chambers are constructed in a polydimethylsiloxane (PDMS) array to simulate various organs on a chip. Various experiments can be performed utilizing OOC technology, including drug delivery research and toxicology. Current technological expansions involve multiple organ microenvironments on a single chip, allowing for studying inter-tissue interactions. Other developments in the OOC technology include finding a more suitable material as a replacement for PDMS and minimizing artefactual error and non-translatable differences.
Collapse
Affiliation(s)
| | - Soumya K. Srivastava
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA;
| |
Collapse
|
16
|
Guttman-Yassky E, Facheris P, Gomez-Arias PJ, Del Duca E, Da Rosa JC, Weidinger S, Bissonnette R, Armstrong AW, Seneschal J, Eyerich K, Estrada YD, Bose SN, Xu D, Chen A, Tatulych S, Güler E, Chan G, Page KM, Kerkmann U. Effect of abrocitinib on skin biomarkers in patients with moderate-to-severe atopic dermatitis. Allergy 2024; 79:1258-1270. [PMID: 38108208 DOI: 10.1111/all.15969] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023]
Abstract
BACKGROUND This is the first report on the effects of abrocitinib, a Janus kinase 1-selective inhibitor, on the expression of skin biomarkers in patients with moderate-to-severe atopic dermatitis (AD). METHODS JADE MOA (NCT03915496) was a double-blind Phase 2a trial. Adults were randomly assigned 1:1:1 to receive monotherapy with once-daily abrocitinib 200 mg, abrocitinib 100 mg, or placebo for 12 weeks. The primary endpoint was change from baseline in markers of inflammation (matrix metalloproteinase [MMP]-12), epidermal hyperplasia (keratin-16 [KRT16]), T-helper 2 (Th2) immune response (C-C motif chemokine ligand [CCL]17, CCL18, and CCL26), and Th22 immune response (S100 calcium binding protein A8, A9, and A12 [S100A8, S100A9, and S100A12]) in skin through 12 weeks. RESULTS A total of 46 patients received abrocitinib 200 mg (n = 14), abrocitinib 100 mg (n = 16), or placebo (n = 16). Abrocitinib improved AD clinical signs and reduced itch. Gene expression of MMP-12, KRT16, S100A8, S100A9, and S100A12 was significantly decreased from baseline with abrocitinib 200 mg (at Weeks 2, 4, and 12) and abrocitinib 100 mg (at Weeks 4 and 12) in a dose-dependent manner. Abrocitinib 200 mg resulted in significant decreases from baseline in CCL17 expression at Week 12 and CCL18 expression at Weeks 2, 4, and 12; no significant decreases were observed for CCL26. CONCLUSIONS Alongside improvements in clinical signs and symptoms of AD, 12 weeks of abrocitinib treatment resulted in downregulation of genes associated with inflammation, epidermal hyperplasia, and Th2 and Th22 immune responses in the skin of patients with moderate-to-severe AD.
Collapse
Affiliation(s)
- Emma Guttman-Yassky
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | - Paola Facheris
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | | | - Ester Del Duca
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | - Joel Correa Da Rosa
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | | | | | | | - Julien Seneschal
- Department of Dermatology and Pediatric Dermatology, National Reference Center for Rare Skin Disorders, Hospital Saint-André, Bordeaux, France
- Bordeaux University, CNRS UMR 5164, Immunoconcept, Bordeaux, France
| | | | - Yeriel D Estrada
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | - Swaroop N Bose
- Icahn School of Medicine at Mount Sinai Medical Center, New York, New York, USA
| | - Dan Xu
- Pfizer Inc., San Diego, California, USA
| | | | | | | | - Gary Chan
- Pfizer Inc., Groton, Connecticut, USA
| | | | | |
Collapse
|
17
|
Ponmozhi J, Dhinakaran S, Kocsis D, Iván K, Erdő F. Models for barrier understanding in health and disease in lab-on-a-chips. Tissue Barriers 2024; 12:2221632. [PMID: 37294075 PMCID: PMC11042069 DOI: 10.1080/21688370.2023.2221632] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 05/31/2023] [Indexed: 06/10/2023] Open
Abstract
The maintenance of body homeostasis relies heavily on physiological barriers. Dysfunction of these barriers can lead to various pathological processes, including increased exposure to toxic materials and microorganisms. Various methods exist to investigate barrier function in vivo and in vitro. To investigate barrier function in a highly reproducible manner, ethically, and high throughput, researchers have turned to non-animal techniques and micro-scale technologies. In this comprehensive review, the authors summarize the current applications of organ-on-a-chip microfluidic devices in the study of physiological barriers. The review covers the blood-brain barrier, ocular barriers, dermal barrier, respiratory barriers, intestinal, hepatobiliary, and renal/bladder barriers under both healthy and pathological conditions. The article then briefly presents placental/vaginal, and tumour/multi-organ barriers in organ-on-a-chip devices. Finally, the review discusses Computational Fluid Dynamics in microfluidic systems that integrate biological barriers. This article provides a concise yet informative overview of the current state-of-the-art in barrier studies using microfluidic devices.
Collapse
Affiliation(s)
- J. Ponmozhi
- Microfluidics Laboratory, Department of Mechanical Engineering, IPS Academy-Institute of Engineering Science, Indore, India
| | - S. Dhinakaran
- The Centre for Fluid Dynamics, Department of Mechanical Engineering, Indian Institute of Technology Indore, Indore, India
| | - Dorottya Kocsis
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Kristóf Iván
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| | - Franciska Erdő
- Faculty of Information Technology and Bionics, Pázmány Péter Catholic University, Budapest, Hungary
| |
Collapse
|
18
|
Tan SH, Liu S, Teoh SH, Bonnard C, Leavesley D, Liang K. A sustainable strategy for generating highly stable human skin equivalents based on fish collagen. BIOMATERIALS ADVANCES 2024; 158:213780. [PMID: 38280287 DOI: 10.1016/j.bioadv.2024.213780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 01/29/2024]
Abstract
Tissue engineered skin equivalents are increasingly recognized as potential alternatives to traditional skin models such as human ex vivo skin or animal skin models. However, most of the currently investigated human skin equivalents (HSEs) are constructed using mammalian collagen which can be expensive and difficult to extract. Fish skin is a waste product produced by fish processing industries and identified as a cost-efficient and sustainable source of type I collagen. In this work, we describe a method for generating highly stable HSEs based on fibrin fortified tilapia fish collagen. The fortified fish collagen (FFC) formulation is optimized to enable reproducible fabrication of full-thickness HSEs that undergo limited contraction, facilitating the incorporation of human donor-derived skin cells and formation of biomimetic dermal and epidermal layers. The morphology and barrier function of the FFC HSEs are compared with a commercial skin model and validated with immunohistochemical staining and transepithelial electrical resistance testing. Finally, the potential of a high throughput screening platform with FFC HSE is explored by scaling down its fabrication to 96-well format.
Collapse
Affiliation(s)
- Shi Hua Tan
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore
| | - Shaoqiong Liu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Swee Hin Teoh
- College of Materials Science and Engineering, Hunan University, People's Republic of China
| | - Carine Bonnard
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore; Skin Research Institute of Singapore (SRIS), Singapore
| | | | - Kun Liang
- A*STAR Skin Research Labs (A*SRL), Agency for Science, Technology and Research (A*STAR), Singapore; Skin Research Institute of Singapore (SRIS), Singapore.
| |
Collapse
|
19
|
Tan IJ, Podwojniak A, Parikh A, Cohen BA. Precision Dermatology: A Review of Molecular Biomarkers and Personalized Therapies. Curr Issues Mol Biol 2024; 46:2975-2990. [PMID: 38666916 PMCID: PMC11049353 DOI: 10.3390/cimb46040186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 03/17/2024] [Accepted: 03/22/2024] [Indexed: 04/28/2024] Open
Abstract
The evolution of personalized medicine in dermatology signifies a transformative shift towards individualized treatments, driven by the integration of biomarkers. These molecular indicators serve beyond diagnostics, offering insights into disease staging, prognosis, and therapeutic monitoring. Specific criteria guide biomarker selection, ensuring attributes like specificity, sensitivity, cost feasibility, stability, rapid detection, and reproducibility. This literature review, based on data from PubMed, SCOPUS, and Web of Science, explores biomarkers in Hidradenitis Suppurativa (HS), Psoriasis, Atopic Dermatitis (AD), Alopecia Areata (AA), Vitiligo, and Chronic Spontaneous Urticaria (CSU). In HS, TNF-α, IL-1β, and MMPs serve as biomarkers, influencing targeted therapies like adalimumab and anakinra. Psoriasis involves biomarkers such as TNF-α, IL-23, and HLA genes, shaping treatments like IL23 and IL17 inhibitors. AD biomarkers include ECP, IL-4, IL-13, guiding therapies like dupilumab and tralokinumab. For AA, lipocalin-2, cytokines, and genetic polymorphisms inform JAK inhibitors' use. Vitiligo biomarkers range from cytokines to genetic markers like TYR, TYRP1, guiding treatments like JAK inhibitors. CSU biomarkers encompass IgE, cytokines, and autologous serum tests, influencing therapies like omalizumab and cyclosporine. Comparing conditions, common proinflammatory markers reveal limited specificity. While some biomarkers aid diagnosis and standard treatments, others hold more scientific than clinical value. Precision medicine, driven by biomarkers, has shown success in skin malignancies. Future directions involve AI-powered algorithms, nanotechnology, and multi-omics integration for personalized dermatological care.
Collapse
Affiliation(s)
- Isabella J. Tan
- Rutgers Robert Wood Johnson Medical School, 125 Paterson Steet, New Brunswick, NJ 08901, USA; (I.J.T.); (A.P.)
| | - Alicia Podwojniak
- Rowan-Virtua School of Osteopathic Medicine, 113 E Laurel Road, Stratford, NJ 08084, USA;
| | - Aarushi Parikh
- Rutgers Robert Wood Johnson Medical School, 125 Paterson Steet, New Brunswick, NJ 08901, USA; (I.J.T.); (A.P.)
| | - Bernard A. Cohen
- Department of Dermatology, The Johns Hopkins Hospital, Baltimore, MD 21287, USA
| |
Collapse
|
20
|
Jeong S, Nam HM, Sung GY. Optimization of hair follicle spheroids for hair-on-a-chip. Biomater Sci 2024; 12:1693-1706. [PMID: 38372380 DOI: 10.1039/d3bm02012f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Currently, most models for hair follicle research have the limitation of not replicating some key features of the hair follicle microenvironment. To complement this, we transfected various factors for hair growth into dermal papilla cells (DPCs) by electroporation and cultured the spheroids with keratinocytes (KCs). We optimized the cell number and culture period for applying spheroids to hair-on-a-chip. Furthermore, we investigated the expression of hair growth factors in spheroids depending on the presence or absence of human umbilical vein endothelial cells (HUVECs) and transfection. In spheroids in which DPCs, KCs, and HUVECs were co-cultured for 21 days, the expression of lymphoid enhancer factor 1 (LEF1), T-cell factor 1 (TCF1), and keratin 25 (K25) in the center of the spheroid, the expression of keratin 17 (K17) on the outer surface of the spheroid, and the shape of hair extending outward from the spheroid surface were observed. From these results, it is expected that a hair-on-a-chip experiment in which short-term cultured TKH spheroids are injected into the dermis and co-cultured with KC will enable the production of full-thickness skin equivalents containing hair in vitro without transplantation into animals.
Collapse
Affiliation(s)
- Subin Jeong
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Republic of Korea.
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Hyeon-Min Nam
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Republic of Korea.
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Republic of Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Republic of Korea.
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Republic of Korea
- Major in Materials Science and Engineering, Hallym University, Chuncheon 24252, Republic of Korea
| |
Collapse
|
21
|
Pezzolo E, Sechi A, Tartaglia J, Naldi L. A critical evaluation of suitability of tralokinumab for treatment of moderate-to-severe atopic dermatitis in adolescents and adults. Expert Rev Clin Immunol 2024; 20:255-266. [PMID: 37955186 DOI: 10.1080/1744666x.2023.2283585] [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: 08/21/2023] [Accepted: 11/10/2023] [Indexed: 11/14/2023]
Abstract
INTRODUCTION Atopic dermatitis (AD) is a chronic, intensely pruritic disease associated with significant patient burden. Recent advancements in AD pathogenesis have expanded its therapeutics pipeline. Tralokinumab is a fully human monoclonal antibody that binds specifically Interleukin (IL)-13, inhibiting the downstream IL-13 signaling. Phase 3 clinical trials and some real-world studies showed that tralokinumab, as monotherapy or in combination with topical corticosteroids, is efficacious and safe in adult patients with moderate-to-severe AD. Similar results were reported in a phase 3 trial in adolescents (aged ≥12 years). AREAS COVERED We review the role of IL-13 in AD and discuss the value of tralokinumab for treating moderate-to-severe AD, comparing efficacy and safety results derived from clinical trials and real-life data. EXPERT OPINION The role of IL-13 in AD supports a targeted therapeutic approach. Tralokinumab has proven efficacious and well-tolerated in a large proportion of patients confirming its value for treating moderate-to-severe AD from age 12 years onwards; it quickly improves itching and can maintain a high-level of response over time; it can be administered with flexible dosing schedules. Future studies will further clarify the role of IL-13 pathway and which patients would be best suited to tralokinumab, shifting AD treatment into an era of precision medicine.
Collapse
Affiliation(s)
- Elena Pezzolo
- Dermatology Unit, San Bortolo Hospital, Vicenza, Italy
- Centro Studi GISED (Italian Group for Epidemiologic Research in Dermatology) - FROM (Research Foundation of Ospedale Maggiore Bergamo), Padiglione Mazzoleni - Presidio Ospedaliero Matteo Rota, Bergamo, Italy
| | - Andrea Sechi
- Dermatology Unit, San Bortolo Hospital, Vicenza, Italy
| | - Jacopo Tartaglia
- Dermatology Unit, Department of Medicine, University of Padova, Padova, Italy
| | - Luigi Naldi
- Dermatology Unit, San Bortolo Hospital, Vicenza, Italy
- Centro Studi GISED (Italian Group for Epidemiologic Research in Dermatology) - FROM (Research Foundation of Ospedale Maggiore Bergamo), Padiglione Mazzoleni - Presidio Ospedaliero Matteo Rota, Bergamo, Italy
| |
Collapse
|
22
|
Akhtar S, Alsayed RKME, Ahmad F, AlHammadi A, Al-Khawaga S, AlHarami SMAM, Alam MA, Al Naama KAHN, Buddenkotte J, Uddin S, Steinhoff M, Ahmad A. Epigenetic control of inflammation in Atopic Dermatitis. Semin Cell Dev Biol 2024; 154:199-207. [PMID: 37120405 DOI: 10.1016/j.semcdb.2023.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 04/12/2023] [Accepted: 04/16/2023] [Indexed: 05/01/2023]
Abstract
Atopic dermatitis (AD), also known as atopic eczema, is a common but also complex chronic, itchy skin condition with underlying inflammation of the skin. This skin ailment is prevalent worldwide and affects people of all ages, particularly children below five years of age. The itching and resulting rashes in AD patients are often the result of inflammatory signals, thus necessitating a closer look at the inflammation-regulating mechanisms for putative relief, care and therapy. Several chemical- as well as genetically-induced animal models have established the importance of targeting pro-inflammatory AD microenvironment. Epigenetic mechanisms are gaining attention towards a better understanding of the onset as well as the progression of inflammation. Several physiological processes with implications in pathophysiology of AD, such as, barrier dysfunction either due to reduced filaggrin / human β-defensins or altered microbiome, reprograming of Fc receptors with resulting overexpression of high affinity IgE receptors, elevated eosinophil numbers or the elevated IL-22 production by CD4 + T cells have underlying epigenetic mechanisms that include differential promoter methylation and/or regulation by non-coding RNAs. Reversing these epigenetic changes has been verified to reduce inflammatory burden through altered secretion of cytokines IL-6, IL-4, IL-13, IL-17, IL-22 etc, with benefit against AD progression in experimental models. A thorough understanding of epigenetic remodeling of inflammation in AD has the potential of opening avenues for novel diagnostic, prognostic and therapeutic options.
Collapse
Affiliation(s)
- Sabah Akhtar
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Reem Khaled M E Alsayed
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar
| | - Fareed Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Ayda AlHammadi
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Sara Al-Khawaga
- Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | | | - Majid Ali Alam
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | | | - Joerg Buddenkotte
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar
| | - Shahab Uddin
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Laboratory Animal Research Center, Qatar University, Doha 2713, Qatar
| | - Martin Steinhoff
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar; Weill Cornell Medicine-Qatar, Medical School, Doha 24144, Qatar; Dept. of Dermatology, Weill Cornell Medicine, New York 10065, NY, USA.
| | - Aamir Ahmad
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Dermatology Institute, Academic Health System, Hamad Medical Corporation, Doha 3050, Qatar; Department of Dermatology and Venereology, Rumailah Hospital, Hamad Medical Corporation, Doha 3050, Qatar.
| |
Collapse
|
23
|
Galvan A, Pellicciari C, Calderan L. Recreating Human Skin In Vitro: Should the Microbiota Be Taken into Account? Int J Mol Sci 2024; 25:1165. [PMID: 38256238 PMCID: PMC10816982 DOI: 10.3390/ijms25021165] [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: 11/28/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024] Open
Abstract
Skin plays crucial roles in the human body: besides protecting the organism from external threats, it acts as a thermal regulator, is responsible for the sense of touch, hosts microbial communities (the skin microbiota) involved in preventing the invasion of foreign pathogens, contains immunocompetent cells that maintain a healthy immunogenic/tolerogenic balance, and is a suitable route for drug administration. In the skin, four defense levels can be identified: besides the physical, chemical, and immune barriers that are inherent to the tissue, the skin microbiota (i.e., the numerous microorganisms living on the skin surface) provides an additional barrier. Studying the skin barrier function or the effects of drugs or cosmetic agents on human skin is a difficult task since snapshot evidence can only be obtained using bioptic samples where dynamic processes cannot properly be followed. To overcome these limitations, many different in vitro models of human skin have been developed that are characterized by diverse levels of complexity in terms of chemical, structural, and cellular composition. The aim of this review is to summarize and discuss the advantages and disadvantages of the different human skin models so far available and to underline how the insertion of a proper microbiota would positively impact an in vitro human skin model in an attempt to better mimic conditions in vivo.
Collapse
Affiliation(s)
- Andrea Galvan
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (A.G.); (L.C.)
| | - Carlo Pellicciari
- Department of Biology and Biotechnology, University of Pavia, Via A. Ferrata 9, 27100 Pavia, Italy
| | - Laura Calderan
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134 Verona, Italy; (A.G.); (L.C.)
| |
Collapse
|
24
|
Kim K, Jang H, Kim E, Kim H, Sung GY. Recent advances in understanding the role of the skin microbiome in the treatment of atopic dermatitis. Exp Dermatol 2023; 32:2048-2061. [PMID: 37767872 DOI: 10.1111/exd.14940] [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: 06/16/2023] [Revised: 08/31/2023] [Accepted: 09/16/2023] [Indexed: 09/29/2023]
Abstract
The skin is the largest organ in the human body, and histologically consists of the epidermis, dermis and subcutaneous tissue. Humans maintain a cooperative symbiotic relationship with their skin microbiota, a complex community of bacteria, fungi and viruses that live on the surface of the skin, and which act as a barrier to protect the body from the inside and outside. The skin is a 'habitat' and vast 'ecosystem' inhabited by countless microbes; as such, relationships have been forged through millions of years of coevolution. It is not surprising then that microbes are key participants in shaping and maintaining essential physiological processes. In addition to maintaining barrier function, the unique symbiotic microbiota that colonizes the skin increases the immune response and provides protection against pathogenic microbes. This review examines our current understanding of skin microbes in shaping and enhancing the skin barrier, as well as skin microbiome-host interactions and their roles in skin diseases, such as atopic dermatitis (AD). We also report on the current status of AD therapeutic drugs that target the skin microbiome, related research on current therapeutic strategies, and the limitations and future considerations of skin microbiome research. In particular, as a future strategy, we discuss the need for a skin-on-a-chip-based microphysiological system research model amenable to biomimetic in vitro studies and human skin equivalent models, including skin appendages.
Collapse
Affiliation(s)
- Kyunghee Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Hyeji Jang
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Eunyul Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Hyeju Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon, Korea
- Major in Materials Science and Engineering, Hallym University, Chuncheon, Korea
| |
Collapse
|
25
|
Jang HJ, Lee JB, Yoon JK. Advanced In Vitro Three-Dimensional Skin Models of Atopic Dermatitis. Tissue Eng Regen Med 2023; 20:539-552. [PMID: 36995643 PMCID: PMC10313606 DOI: 10.1007/s13770-023-00532-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/11/2023] [Accepted: 02/19/2023] [Indexed: 03/31/2023] Open
Abstract
Atopic dermatitis (AD) is one of the most prevalent inflammatory skin diseases that is characterized by eczematous rashes, intense itching, dry skin, and sensitive skin. Although AD significantly impacts the quality of life and the number of patients keeps increasing, its pathological mechanism is still unknown because of its complexity. The importance of developing new in vitro three-dimensional (3D) models has been underlined in order to understand the mechanisms for the development of therapeutics since the limitations of 2D models or animal models have been repeatedly reported. Thus, the new in vitro AD models should not only be created in 3D structure, but also reflect the pathological characteristics of AD, which are known to be associated with Th2-mediated inflammatory responses, epidermal barrier disruption, increased dermal T-cell infiltration, filaggrin down-regulation, or microbial imbalance. In this review, we introduce various types of in vitro skin models including 3D culture methods, skin-on-a-chips, and skin organoids, as well as their applications to AD modeling for drug screening and mechanistic studies.
Collapse
Affiliation(s)
- Hye-Jeong Jang
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea
| | - Jung Bok Lee
- Department of Biological Sciences, Research Institute of Women's Health, Sookmyung Women's University, Seoul, 04310, Republic of Korea.
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do, 17546, Republic of Korea.
| |
Collapse
|
26
|
Hua X, Blosch CD, Dorsey H, Ficaro MK, Wallace NL, Hsung RP, Dai J. Epidermal Loss of RORα Enhances Skin Inflammation in a MC903-Induced Mouse Model of Atopic Dermatitis. Int J Mol Sci 2023; 24:10241. [PMID: 37373387 DOI: 10.3390/ijms241210241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Atopic dermatitis (AD) is a chronic inflammatory skin disease featuring skin barrier dysfunction and immune dysregulation. Previously, we reported that the retinoid-related orphan nuclear receptor RORα was highly expressed in the epidermis of normal skin. We also found that it positively regulated the expression of differentiation markers and skin barrier-related genes in human keratinocytes. In contrast, epidermal RORα expression was downregulated in the skin lesions of several inflammatory skin diseases, including AD. In this study, we generated mouse strains with epidermis-specific Rora ablation to understand the roles of epidermal RORα in regulating AD pathogenesis. Although Rora deficiency did not cause overt macroscopic skin abnormalities at the steady state, it greatly amplified MC903-elicited AD-like symptoms by intensifying skin scaliness, increasing epidermal hyperproliferation and barrier impairment, and elevating dermal immune infiltrates, proinflammatory cytokines, and chemokines. Despite the normal appearance at the steady state, Rora-deficient skin showed microscopic abnormalities, including mild epidermal hyperplasia, increased TEWL, and elevated mRNA expression of Krt16, Sprr2a, and Tslp genes, indicating subclinical impairment of epidermal barrier functions. Our results substantiate the importance of epidermal RORα in partially suppressing AD development by maintaining normal keratinocyte differentiation and skin barrier function.
Collapse
Affiliation(s)
- Xiangmei Hua
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Conrad Dean Blosch
- Biomedical Research Model Services, University of Wisconsin, Madison, WI 53705, USA
| | - Hannah Dorsey
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Maria K Ficaro
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Nicole L Wallace
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Richard P Hsung
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Jun Dai
- School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
- UW Carbone Cancer Center, University of Wisconsin, Madison, WI 53705, USA
| |
Collapse
|
27
|
Freer M, Darling N, Goncalves K, Mills KJ, Przyborski S. Development of a mammalian neurosensory full-thickness skin equivalent and its application to screen sensitizing stimuli. Bioeng Transl Med 2023; 8:e10484. [PMID: 37206205 PMCID: PMC10189474 DOI: 10.1002/btm2.10484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 12/06/2022] [Accepted: 12/21/2022] [Indexed: 01/04/2023] Open
Abstract
Human skin equivalents (HSEs) are an increasingly popular research tool due to limitations associated with animal testing for dermatological research. They recapitulate many aspects of skin structure and function, however, many only contain two basic cell types to model dermal and epidermal compartments, which limits their application. We describe advances in the field skin tissue modeling to produce a construct containing sensory-like neurons that is responsive to known noxious stimuli. Through incorporation of mammalian sensory-like neurons, we were able to recapitulate aspects of the neuroinflammatory response including secretion of substance P and a range of pro-inflammatory cytokines in response to a well-characterized neurosensitizing agent: capsaicin. We observed that neuronal cell bodies reside in the upper dermal compartment with neurites extending toward the keratinocytes of the stratum basale where they exist in close proximity to one another. These data suggest that we are able to model aspects of the neuroinflammatory response that occurs during exposure to dermatological stimuli including therapeutics and cosmetics. We propose that this skin construct can be considered a platform technology with a wide range of applications including screening of actives, therapeutics, modeling of inflammatory skin diseases, and fundamental approaches to probe underlying cell and molecular mechanisms.
Collapse
Affiliation(s)
| | | | | | | | - Stefan Przyborski
- Department of BiosciencesDurham UniversityDurhamUK
- Reprocell Europe LtdGlasgowUK
| |
Collapse
|
28
|
Nishimoto R, Kodama C, Yamashita H, Hattori F. Human Induced Pluripotent Stem Cell-Derived Keratinocyte-Like Cells for Research on Protease-Activated Receptor 2 in Nonhistaminergic Cascades of Atopic Dermatitis . J Pharmacol Exp Ther 2023; 384:248-253. [PMID: 36351795 DOI: 10.1124/jpet.122.001412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 09/13/2022] [Accepted: 10/06/2022] [Indexed: 11/11/2022] Open
Abstract
Keratinocytes are the most abundant cells in the epidermis, and as part of the frontline immunologic defense system, keratinocytes function as a barrier to exogenous attacks. Protease-activated receptor 2 (PAR2) is expressed in human keratinocytes and activated in several inflammatory conditions, such as atopic dermatitis (AD). In this study, we demonstrated the differentiation of human induced pluripotent stem cell into keratinocytes by the improved, robust differentiation procedure and confirmed that human induced pluripotent stem cell-derived keratinocyte-like cells (iKera) express PAR2, which is activated by external addition of the ligand peptide and trypsin. The activation of PAR2 led to the release of calcium from intracellular calcium storage, followed by the release of the proinflammatory cytokine tumor necrosis factor α Moreover, PAR2 antagonist I-191 (CAS No. 1690172-25-8) inhibited calcium release in a dose-dependent manner. This is the first study to demonstrate that iKera expresses a functional PAR2 protein. Furthermore, our results indicate crosstalk between the PAR2- and IL-4-mediated inflammatory axes in iKera, suggesting that iKera can be used as a platform for a broad range of mechanism-targeted drug screening in AD. SIGNIFICANCE STATEMENT: This is the first study to provide evidence that human induced pluripotent stem cell-derived keratinocyte-like cells (iKera) express functional protease-activated receptor 2 (PAR2). Furthermore, this study demonstrated in iKera that the IL-4 inflammatory axis can crosstalk with the PAR2-mediated inflammatory axis in keratinocytes. To the best of our knowledge, this is the first report to indicate that iKera can be used for research and as a drug screening platform for atopic dermatitis.
Collapse
Affiliation(s)
- Rio Nishimoto
- Innovative Regenerative Medicine, Kansai Medical University, Graduate School of Medicine, Osaka, Japan (R.N., C.K., H.Y., F.H.) and Osaka College of High-Technology, Osaka, Japan (R.N., C.K.)
| | - Chinatsu Kodama
- Innovative Regenerative Medicine, Kansai Medical University, Graduate School of Medicine, Osaka, Japan (R.N., C.K., H.Y., F.H.) and Osaka College of High-Technology, Osaka, Japan (R.N., C.K.)
| | - Hiromi Yamashita
- Innovative Regenerative Medicine, Kansai Medical University, Graduate School of Medicine, Osaka, Japan (R.N., C.K., H.Y., F.H.) and Osaka College of High-Technology, Osaka, Japan (R.N., C.K.)
| | - Fumiyuki Hattori
- Innovative Regenerative Medicine, Kansai Medical University, Graduate School of Medicine, Osaka, Japan (R.N., C.K., H.Y., F.H.) and Osaka College of High-Technology, Osaka, Japan (R.N., C.K.)
| |
Collapse
|
29
|
Giacomelli R, Fargnoli MC. In Vitro Trials: The Dawn of a New Era for Drug Discovery in Atopic Dermatitis? J Pharmacol Exp Ther 2023; 384:245-247. [PMID: 36653181 DOI: 10.1124/jpet.122.001469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 12/02/2022] [Indexed: 01/20/2023] Open
Affiliation(s)
- R Giacomelli
- Clinical and Research Unit of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Rome, Italy (R.G.); Rheumatology and Clinical Immunology, Department of Medicine, University Campus Bio-Medico, School of Medicine, Rome, Italy (R.G.); Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, School of Medicine, L'Aquila, Italy (M.C.F.); and Dermatology Unit, Ospedale San Salvatore, L'Aquila, Italy (M.C.F.)
| | - M C Fargnoli
- Clinical and Research Unit of Rheumatology and Clinical Immunology, Fondazione Policlinico Campus Bio-Medico, Rome, Italy (R.G.); Rheumatology and Clinical Immunology, Department of Medicine, University Campus Bio-Medico, School of Medicine, Rome, Italy (R.G.); Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, School of Medicine, L'Aquila, Italy (M.C.F.); and Dermatology Unit, Ospedale San Salvatore, L'Aquila, Italy (M.C.F.)
| |
Collapse
|
30
|
Hyaluronic Acid Hydrogel Containing Resveratrol-Loaded Chitosan Nanoparticles as an Adjuvant in Atopic Dermatitis Treatment. J Funct Biomater 2023; 14:jfb14020082. [PMID: 36826881 PMCID: PMC9959248 DOI: 10.3390/jfb14020082] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Revised: 01/14/2023] [Accepted: 01/29/2023] [Indexed: 02/04/2023] Open
Abstract
Atopic dermatitis (AD) is a common disease-causing skin inflammation, redness, and irritation, which can eventually result in infection that drastically impacts patient quality of life. Resveratrol (Res) is a natural phytochemical famed for its excellent anti-inflammatory and antioxidant activities. However, it is poorly bioavailable. Thus, a drug delivery system is needed to enhance in vivo bioactivity. Herein, we report the preparation of hyaluronic acid (HA) hydrogels containing resveratrol-loaded chitosan (CS) nanoparticles, their physicochemical analysis, and their potential therapeutic effects in the treatment of AD. Positively charged CS nanoparticles prepared by tripolyphosphate (TPP) gelation showed sizes ranging from 120 to around 500 nm and Res encapsulation efficiency as high as 80%. Embedding the nanoparticles in HA retarded their hydrolytic degradation and also slowed resveratrol release. Resveratrol released from nanoparticle-loaded hydrogel counteracted the oxidative damage induced by ROS generation in TNF-α/INF-γ-treated human keratinocytes (HaCaT) used as an AD in vitro model. Moreover, pre-treatment with Res@gel reduced secretion and gene expression of proinflammatory cytokines in HaCaT cells. The physicochemical analysis and in vitro assay confirmed that the formulated hydrogel could be considered an efficient and sustained resveratrol delivery vector in AD treatment.
Collapse
|
31
|
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
|
32
|
Kim K, Jeong S, Sung GY. Effect of Periodical Tensile Stimulation on the Human Skin Equivalents by Magnetic Stretching Skin-on-a-Chip (MSSC). BIOCHIP JOURNAL 2022. [DOI: 10.1007/s13206-022-00092-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
33
|
Kim K, Kim H, Sung GY. Effects of Indole-3-Lactic Acid, a Metabolite of Tryptophan, on IL-4 and IL-13-Induced Human Skin-Equivalent Atopic Dermatitis Models. Int J Mol Sci 2022; 23:13520. [PMID: 36362303 PMCID: PMC9655012 DOI: 10.3390/ijms232113520] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 08/13/2023] Open
Abstract
Indole-3-lactic acid (I3LA) is a well-known metabolite involved in tryptophan metabolism. Indole derivatives are involved in the differentiation of immune cells and the synthesis of cytokines via the aryl hydrocarbon receptors for modulating immunity, and the indole derivatives may be involved in allergic responses. I3LA was selected as a candidate substance for the treatment of atopic dermatitis (AD), and its inhibitory effect on AD progression was investigated. Full-thickness human skin equivalents (HSEs) consisting of human-derived cells were generated on microfluidic chips and stimulated with major AD-inducing factors. The induced AD-HSEs were treated with I3LA for 7 days, and this affected the AD-associated genetic biomarkers and increased the expression of the major constituent proteins of the skin barrier. After the treatment for 14 days, the surface became rough and sloughed off, and there was no significant difference between the increased AD-related mRNA expression and the skin barrier protein expression. Therefore, the short-term use of I3LA for approximately one week is considered to be effective in suppressing AD.
Collapse
Affiliation(s)
- Kyunghee Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Hyeju Kim
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
| | - Gun Yong Sung
- Interdisciplinary Program of Nano-Medical Device Engineering, Hallym University, Chuncheon 24252, Korea
- Integrative Materials Research Institute, Hallym University, Chuncheon 24252, Korea
- Major in Materials Science and Engineering, Hallym University, Chuncheon 24252, Korea
| |
Collapse
|
34
|
Structural and Functional Analysis of Excised Skins and Human Reconstructed Epidermis with Confocal Raman Spectroscopy and in Microfluidic Diffusion Chambers. Pharmaceutics 2022; 14:pharmaceutics14081689. [PMID: 36015315 PMCID: PMC9415586 DOI: 10.3390/pharmaceutics14081689] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/04/2022] [Accepted: 08/11/2022] [Indexed: 12/02/2022] Open
Abstract
Several ex vivo and in vitro skin models are available in the toolbox of dermatological and cosmetic research. Some of them are widely used in drug penetration testing. The excised skins show higher variability, while the in vitro skins provide more reproducible data. The aim of the current study was to compare the chemical composition of different skin models (excised rat skin, excised human skin and human-reconstructed epidermis) by measurement of ceramides, cholesterol, lactate, urea, protein and water at different depths of the tissues. The second goal was to compile a testing system, which includes a skin-on-a-chip diffusion setup and a confocal Raman spectroscopy for testing drug diffusion across the skin barrier and accumulation in the tissue models. A hydrophilic drug caffeine and the P-glycoprotein substrate quinidine were used in the study as topical cream formulations. The results indicate that although the transdermal diffusion of quinidine is lower, the skin accumulation was comparable for the two drugs. The various skin models showed different chemical compositions. The human skin was abundant in ceramides and cholesterol, while the reconstructed skin contained less water and more urea and protein. Based on these results, it can be concluded that skin-on-a-chip and confocal Raman microspectroscopy are suitable for testing drug penetration and distribution at different skin layers within an exposition window. Furthermore, obese human skin should be treated with caution for skin absorption testing due to its unbalanced composition.
Collapse
|
35
|
Phang SJ, Basak S, Teh HX, Packirisamy G, Fauzi MB, Kuppusamy UR, Neo YP, Looi ML. Advancements in Extracellular Matrix-Based Biomaterials and Biofabrication of 3D Organotypic Skin Models. ACS Biomater Sci Eng 2022; 8:3220-3241. [PMID: 35861577 DOI: 10.1021/acsbiomaterials.2c00342] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Over the last decades, three-dimensional (3D) organotypic skin models have received enormous attention as alternative models to in vivo animal models and in vitro two-dimensional assays. To date, most organotypic skin models have an epidermal layer of keratinocytes and a dermal layer of fibroblasts embedded in an extracellular matrix (ECM)-based biomaterial. The ECM provides mechanical support and biochemical signals to the cells. Without advancements in ECM-based biomaterials and biofabrication technologies, it would have been impossible to create organotypic skin models that mimic native human skin. In this review, the use of ECM-based biomaterials in the reconstruction of skin models, as well as the study of complete ECM-based biomaterials, such as fibroblasts-derived ECM and decellularized ECM as a better biomaterial, will be highlighted. We also discuss the benefits and drawbacks of several biofabrication processes used in the fabrication of ECM-based biomaterials, such as conventional static culture, electrospinning, 3D bioprinting, and skin-on-a-chip. Advancements and future possibilities in modifying ECM-based biomaterials to recreate disease-like skin models will also be highlighted, given the importance of organotypic skin models in disease modeling. Overall, this review provides an overview of the present variety of ECM-based biomaterials and biofabrication technologies available. An enhanced organotypic skin model is expected to be produced in the near future by combining knowledge from previous experiences and current research.
Collapse
Affiliation(s)
- Shou Jin Phang
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Soumyadeep Basak
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee-247 667, Uttarakhand, India
| | - Huey Xhin Teh
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Gopinath Packirisamy
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee-247 667, Uttarakhand, India
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, 56000 Kuala Lumpur, Malaysia
| | - Umah Rani Kuppusamy
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| | - Yun Ping Neo
- School of Biosciences, Faculty of Health and Medical Sciences, Taylor's University, 47500 Selangor, Malaysia
| | - Mee Lee Looi
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
36
|
Modeling an Optimal 3D Skin-on-Chip within Microfluidic Devices for Pharmacological Studies. Pharmaceutics 2022; 14:pharmaceutics14071417. [PMID: 35890312 PMCID: PMC9316928 DOI: 10.3390/pharmaceutics14071417] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/27/2022] [Accepted: 07/05/2022] [Indexed: 02/05/2023] Open
Abstract
Preclinical research remains hampered by an inadequate representation of human tissue environments which results in inaccurate predictions of a drug candidate’s effects and target’s suitability. While human 2D and 3D cell cultures and organoids have been extensively improved to mimic the precise structure and function of human tissues, major challenges persist since only few of these models adequately represent the complexity of human tissues. The development of skin-on-chip technology has allowed the transition from static 3D cultures to dynamic 3D cultures resembling human physiology. The integration of vasculature, immune system, or the resident microbiome in the next generation of SoC, with continuous detection of changes in metabolism, would potentially overcome the current limitations, providing reliable and robust results and mimicking the complex human skin. This review aims to provide an overview of the biological skin constituents and mechanical requirements that should be incorporated in a human skin-on-chip, permitting pharmacological, toxicological, and cosmetic tests closer to reality.
Collapse
|
37
|
Nosratabadi R, Khajepour F, Zangouyee M, Khosravimashizi A, Afgar A, Abdollahi V, Dabiri S. Caraway extract alleviates atopic dermatitis by regulating oxidative stress, suppressing Th2 cells, and upregulating Th1 cells in mice. Asian Pac J Trop Biomed 2022. [DOI: 10.4103/2221-1691.357741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|