1
|
Schneider AM, Feehan RP, Sennett ML, Wills CA, Garner C, Cong Z, Billingsley EM, Flamm AF, Shantz LM, Nelson AM. TLR3 activation mediates partial epithelial-to-mesenchymal transition in human keratinocytes. Life Sci Alliance 2024; 7:e202402777. [PMID: 39348939 PMCID: PMC11443013 DOI: 10.26508/lsa.202402777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 09/23/2024] [Accepted: 09/23/2024] [Indexed: 10/02/2024] Open
Abstract
TLR3 is expressed in human skin and keratinocytes, and given its varied role in skin inflammation, development, and regeneration, we sought to determine the cellular response in normal human keratinocytes to TLR3 activation. We investigated this mechanism by treating primary human keratinocytes with both UVB, an endogenous and physiologic TLR3 activator, and poly(I:C), a synthetic and selective TLR3 ligand. TLR3 activation with either UVB or poly(I:C) altered keratinocyte morphology, coinciding with the key features of epithelial-to-mesenchymal transition: increased epithelial-to-mesenchymal transition gene expression, enhanced migration, and increased invasion properties. These results confirm and extend previous studies demonstrating that in addition to its classical role in the innate immune response, TLR3 signaling also regulates stem cell-like properties and developmental programs.
Collapse
Affiliation(s)
- Andrea M Schneider
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
| | - Robert P Feehan
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
| | - Mackenzie L Sennett
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
| | - Carson A Wills
- Department of Pediatrics, Penn State College of Medicine, Hershey, PA, USA
| | - Charlotte Garner
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
| | - Zhaoyuan Cong
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
| | | | - Alexandra F Flamm
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
| | - Lisa M Shantz
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
- Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA
| | - Amanda M Nelson
- Department of Dermatology, Penn State Health Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
2
|
Zhang M, Huang M, Dong X, Wang Y, Zhang L, Wang Z, Cao J. Rotating cell culture system-induced injectable self-assembled microtissues with epidermal stem cells for full-thickness skin repair. PeerJ 2024; 12:e18418. [PMID: 39494298 PMCID: PMC11531757 DOI: 10.7717/peerj.18418] [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: 03/26/2024] [Accepted: 10/07/2024] [Indexed: 11/05/2024] Open
Abstract
Epidermal stem cells (EpSCs) are crucial for wound healing and tissue regeneration, and traditional culture methods often lead to their inactivation. It is urgent to increase the yield of high quality EpSCs. In this study, primary EpSCs were isolated and cultured in a serum-free, feeder-free culture system. EpSCs are then expanded in a dynamic 3D environment using a rotating cell culture system (RCCS) with biodegradable porous microcarriers (MC). Over a period of 14 days, the cells self-assembled into microtissues with superior cell proliferation compared to 3D static culture. Immunofluorescence and qPCR analyses consistently showed that the stemness of the 3D microtissues was preserved, especially the COL17A1 associated with anti-aging was highly expressed in RCCS induced microtissues. In vivo experiments demonstrated that the group treated with 3D microtissues loaded with EpSCs showed enhanced early wound healing, and the injectable 3D microtissues were more conducive to maintaining cell viability and differentiation potential. Our study provides valuable insights into the dynamic 3D culture of EpSCs and introduces an injectable therapy using 3D microtissues loaded with EpSCs, which provides a new and effective approach for cell delivery and offering a promising strategy for guiding the regeneration of full-thickness skin defects.
Collapse
Affiliation(s)
- Min Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Meng Huang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Xixi Dong
- Department of Stomatology, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Yibo Wang
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Luyue Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Zhaoxiang Wang
- Medical School of Chinese PLA, Beijing, China
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Junkai Cao
- Department of Stomatology, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
3
|
Huang Y, Qin P, Zhou P, Long B, Zhang S, Gao R, Zhu B, Li Y, Li Q. Non-pharmacological interventions of intermittent fasting and pulsed radiofrequency energy (PRFE) combination therapy promote diabetic wound healing. Nutr Diabetes 2024; 14:83. [PMID: 39375333 PMCID: PMC11458794 DOI: 10.1038/s41387-024-00344-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 09/18/2024] [Accepted: 09/24/2024] [Indexed: 10/09/2024] Open
Abstract
OBJECTIVE This study aims to conduct an unbiased assessment of the synergistic effects of non-pharmacological Interventions of intermittent fasting and pulsed radiofrequency energy (PRFE) combination therapy on the facilitation of diabetic wound healing, while also exploring the underlying mechanisms. The findings of this research will provide a theoretical framework and innovative strategy for unconventional therapeutic interventions aimed at enhancing the healing process of diabetes-related wounds. METHODS In vivo experiments involved the induction of diabetic models in C57 mice through streptozotocin injection. To simulate a combined therapeutic approach, diabetic mice underwent fasting on days 2 and 6, accompanied by twice daily PRFE applications for 8 days. In vitro experiments were conducted using a serum-free culture medium to replicate fasting conditions. The investigation encompassed wound healing rate, proliferation, migration, angiogenesis, oxidative stress, fibrogenesis, and sensory nerve growth through histological analysis and functional assessments in vivo. Additionally, this study utilized quantitative reverse transcription polymerase chain reaction (qRT-PCR), western blotting (WB), and immunofluorescence staining techniques to elucidate the potential mechanisms underlying the effects of intermittent Fasting and PRFE combination therapy in diabetic wound healing, both in vitro and in vivo. RESULTS The intermittent fasting and PRFE combination therapy demonstrated superior efficacy in enhancing diabetic wound healing compared to either treatment alone. It harnessed the respective strengths of individual therapies, fostering migration, mitigating oxidative stress, and enhancing fibrogenesis. Furthermore, the combination therapy manifested a synergistic effect in promoting proliferation, tube formation, angiogenesis, and sensory nerve growth. CONCLUSION This study demonstrates that intermittent fasting and PRFE combination therapy enhance diabetic wound healing, effectively leveraging the strengths of both therapies and even yielding synergistic benefits. Moreover, it indicates the potential engagement of the P75/HIF1A/VEGFA axis in mediating these effects.
Collapse
Affiliation(s)
- Yating Huang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Peiliang Qin
- Department of Vascular Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Peng Zhou
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Binbin Long
- General Surgery Department, Taihe Hospital affiliated to Hubei University of Medicine, Shiyan, Hubei, 430000, China
| | - Shan Zhang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Ruikang Gao
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Bingjie Zhu
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China.
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China.
| |
Collapse
|
4
|
Fuchs C, Stalnaker KJ, Dalgard CL, Sukumar G, Hupalo D, Dreyfuss JM, Pan H, Wang Y, Pham L, Wu X, Jozic I, Anderson RR, Cho S, Meyerle JH, Tam J. Plantar Skin Exhibits Altered Physiology, Constitutive Activation of Wound-Associated Phenotypes, and Inherently Delayed Healing. J Invest Dermatol 2024; 144:1633-1648.e14. [PMID: 38237729 DOI: 10.1016/j.jid.2023.12.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 12/16/2023] [Accepted: 12/19/2023] [Indexed: 06/24/2024]
Abstract
Wound research has typically been performed without regard for where the wounds are located on the body, despite well-known heterogeneities in physical and biological properties between different skin areas. The skin covering the palms and soles is highly specialized, and plantar ulcers are one of the most challenging and costly wound types to manage. Using primarily the porcine model, we show that plantar skin is molecularly and functionally more distinct from nonplantar skin than previously recognized, with unique gene and protein expression profiles, broad alterations in cellular functions, constitutive activation of many wound-associated phenotypes, and inherently delayed healing. This unusual physiology is likely to play a significant but underappreciated role in the pathogenesis of plantar ulcers as well as the last 25+ years of futility in therapy development efforts. By revealing this critical yet unrecognized pitfall, we hope to contribute to the development of more effective therapies for these devastating nonhealing wounds.
Collapse
Affiliation(s)
- Christiane Fuchs
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Katherine J Stalnaker
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Clifton L Dalgard
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Gauthaman Sukumar
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Daniel Hupalo
- The American Genome Center, Uniformed Services University, Bethesda, Maryland, USA; Department of Anatomy, Physiology & Genetics, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA
| | - Jonathan M Dreyfuss
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Hui Pan
- Bioinformatics and Biostatistics Core, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts, USA
| | - Ying Wang
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Linh Pham
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Xunwei Wu
- Cutaneous Biology Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Ivan Jozic
- Wound Healing and Regenerative Medicine Research Program, Dr. Phillp Frost Department of Dermatology & Cutaneous Surgery, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - R Rox Anderson
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA
| | - Sunghun Cho
- Department of Dermatology, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA; Department of Dermatology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Jon H Meyerle
- Department of Dermatology, F. Edward Hebert School of Medicine, Uniformed Services University, Bethesda, Maryland, USA; Department of Dermatology, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Joshua Tam
- Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, Massachusetts, USA; Department of Dermatology, Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
5
|
Bingnan W, Jiao T, Ghorbani A, Baghei S. Enhancing regenerative potential: A comprehensive review of stem cell transplantation for sports-related neuronal injuries, with a focus on spinal cord injuries and peripheral nervous system damage. Tissue Cell 2024; 88:102429. [PMID: 38833939 DOI: 10.1016/j.tice.2024.102429] [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: 01/24/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/06/2024]
Abstract
Neuronal injuries, as one of the consequences of sports-related incidents, exert a profound influence on the athletes' future, potentially leading to complete immobility and impeding their athletic pursuits. In cases of severe damage inflicted upon the spinal cord (SC) and peripheral nervous systems (PNS), the regenerative process is notably compromised, rendering it essentially inefficient. Among the pivotal therapeutic approaches for the enhancement and prevention of secondary SC injuries (SCI), stem cell transplantation (SCT) stands out prominently. Stem cells, whether directly involved in replacement and reconstruction or indirectly through modification and secretion of crucial bioenvironmental factors, engage in the intricate process of tissue regeneration. Stem cells, through the secretion of neurotrophic factors (NTFs) (aiming to modulate the immune system), reduction of inflammation, axonal growth stimulation, and myelin formation, endeavor to facilitate the regeneration of damaged SC tissue. The fundamental challenges of this approach encompass the proper selection of suitable stem cell candidates for transplantation and the establishment of an appropriate microenvironment conducive to SC repair. In this article, an attempt has been made to explore sports-related injuries, particularly SCI, to comprehensively review innovative methods for treating SCI, and to address the existing challenges. Additionally, some of the stem cells used in neural injuries and the process of their utilization have been discussed.
Collapse
Affiliation(s)
- Wang Bingnan
- Department of P.E, Central South University, Changsha 410083, China
| | - Tong Jiao
- The High School Attached to Hunan Normal University Bocai Experimental Middle School,Changsha 410208, China.
| | - A Ghorbani
- Biotechnology Department, Islamic Azad University, Isfahan, Iran
| | - Sh Baghei
- Biotechnology Department, Islamic Azad University, Isfahan, Iran.
| |
Collapse
|
6
|
De Silva WGM, Sequeira VB, Yang C, Dixon KM, Holland AJA, Mason RS, Rybchyn MS. 1,25-Dihydroxyvitamin D 3 Suppresses UV-Induced Poly(ADP-Ribose) Levels in Primary Human Keratinocytes, as Detected by a Novel Whole-Cell ELISA. Int J Mol Sci 2024; 25:5583. [PMID: 38891771 PMCID: PMC11171802 DOI: 10.3390/ijms25115583] [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: 04/05/2024] [Revised: 05/13/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Photoprotective properties of 1,25-dihydroxyvitamin D3 (1,25(OH)2D3) to reduce UV-induced DNA damage have been established in several studies. UV-induced DNA damage in skin such as single or double strand breaks is known to initiate several cellular mechanisms including activation of poly(ADP-ribose) (pADPr) polymerase-1 (PARP-1). DNA damage from UV also increases extracellular signal-related kinase (ERK) phosphorylation, which further increases PARP activity. PARP-1 functions by using cellular nicotinamide adenine dinucleotide (NAD+) to synthesise pADPr moieties and attach these to target proteins involved in DNA repair. Excessive PARP-1 activation following cellular stress such as UV irradiation may result in excessive levels of cellular pADPr. This can also have deleterious effects on cellular energy levels due to depletion of NAD+ to suboptimal levels. Since our previous work indicated that 1,25(OH)2D3 reduced UV-induced DNA damage in part through increased repair via increased energy availability, the current study investigated the effect of 1,25(OH)2D3 on UV-induced PARP-1 activity using a novel whole-cell enzyme- linked immunosorbent assay (ELISA) which quantified levels of the enzymatic product of PARP-1, pADPr. This whole cell assay used around 5000 cells per replicate measurement, which represents a 200-400-fold decrease in cell requirement compared to current commercial assays that measure in vitro pADPr levels. Using our assay, we observed that UV exposure significantly increased pADPr levels in human keratinocytes, while 1,25(OH)2D3 significantly reduced levels of UV-induced pADPr in primary human keratinocytes to a similar extent as a known PARP-1 inhibitor, 3-aminobenzamide (3AB). Further, both 1,25(OH)2D3 and 3AB as well as a peptide inhibitor of ERK-phosphorylation significantly reduced DNA damage in UV-exposed keratinocytes. The current findings support the proposal that reduction in pADPr levels may be critical for the function of 1,25(OH)2D3 in skin to reduce UV-induced DNA damage.
Collapse
Affiliation(s)
| | - Vanessa Bernadette Sequeira
- Department of Physiology, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Chen Yang
- Department of Physiology, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Katie Marie Dixon
- Department of Anatomy and Histology and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia;
| | - Andrew J. A. Holland
- Douglas Cohen Department of Paediatric Surgery, The Children’s Hospital at Westmead Clinical School, The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rebecca Sara Mason
- Department of Physiology, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
- School of Life and Environmental Sciences and Charles Perkins Centre, The University of Sydney, Sydney, NSW 2006, Australia
| | - Mark Stephen Rybchyn
- Department of Physiology, School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
7
|
Moreno JA, Dudchenko O, Feigin CY, Mereby SA, Chen Z, Ramos R, Almet AA, Sen H, Brack BJ, Johnson MR, Li S, Wang W, Gaska JM, Ploss A, Weisz D, Omer AD, Yao W, Colaric Z, Kaur P, Leger JS, Nie Q, Mena A, Flanagan JP, Keller G, Sanger T, Ostrow B, Plikus MV, Kvon EZ, Aiden EL, Mallarino R. Emx2 underlies the development and evolution of marsupial gliding membranes. Nature 2024; 629:127-135. [PMID: 38658750 PMCID: PMC11062917 DOI: 10.1038/s41586-024-07305-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 03/13/2024] [Indexed: 04/26/2024]
Abstract
Phenotypic variation among species is a product of evolutionary changes to developmental programs1,2. However, how these changes generate novel morphological traits remains largely unclear. Here we studied the genomic and developmental basis of the mammalian gliding membrane, or patagium-an adaptative trait that has repeatedly evolved in different lineages, including in closely related marsupial species. Through comparative genomic analysis of 15 marsupial genomes, both from gliding and non-gliding species, we find that the Emx2 locus experienced lineage-specific patterns of accelerated cis-regulatory evolution in gliding species. By combining epigenomics, transcriptomics and in-pouch marsupial transgenics, we show that Emx2 is a critical upstream regulator of patagium development. Moreover, we identify different cis-regulatory elements that may be responsible for driving increased Emx2 expression levels in gliding species. Lastly, using mouse functional experiments, we find evidence that Emx2 expression patterns in gliders may have been modified from a pre-existing program found in all mammals. Together, our results suggest that patagia repeatedly originated through a process of convergent genomic evolution, whereby regulation of Emx2 was altered by distinct cis-regulatory elements in independently evolved species. Thus, different regulatory elements targeting the same key developmental gene may constitute an effective strategy by which natural selection has harnessed regulatory evolution in marsupial genomes to generate phenotypic novelty.
Collapse
Affiliation(s)
- Jorge A Moreno
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Stowers Institute for Medical Research, Kansas City, MO, USA
| | - Olga Dudchenko
- The Center for Genome Architecture, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- The Center for Theoretical Biological Physics, Rice University, Houston, TX, USA
| | - Charles Y Feigin
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- School of BioSciences, The University of Melbourne, Parkville, Victoria, Australia
- Department of Environment and Genetics, La Trobe University, Bundoora, Victoria, Australia
| | - Sarah A Mereby
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Zhuoxin Chen
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Raul Ramos
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Axel A Almet
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
| | - Harsha Sen
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Benjamin J Brack
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Matthew R Johnson
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sha Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Wei Wang
- Lewis Sigler Center for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Jenna M Gaska
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - David Weisz
- The Center for Genome Architecture, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Arina D Omer
- The Center for Genome Architecture, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Weijie Yao
- The Center for Genome Architecture, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Zane Colaric
- The Center for Genome Architecture, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Parwinder Kaur
- The University of Western Australia, Crawley, Western Australia, Australia
| | - Judy St Leger
- Cornell University College of Veterinary Medicine, Ithaca, NY, USA
| | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, Irvine, CA, USA
| | | | | | - Greta Keller
- Department of Biology, Loyola University, Chicago, IL, USA
| | - Thomas Sanger
- Department of Biology, Loyola University, Chicago, IL, USA
| | - Bruce Ostrow
- Department of Biology, Grand Valley State University, Allendale, MI, USA
| | - Maksim V Plikus
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Evgeny Z Kvon
- Department of Developmental and Cell Biology, University of California, Irvine, Irvine, CA, USA
| | - Erez Lieberman Aiden
- The Center for Genome Architecture, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- The Center for Theoretical Biological Physics, Rice University, Houston, TX, USA.
| | - Ricardo Mallarino
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
8
|
Pazzin DB, Previato TTR, Budelon Gonçalves JI, Zanirati G, Xavier FAC, da Costa JC, Marinowic DR. Induced Pluripotent Stem Cells and Organoids in Advancing Neuropathology Research and Therapies. Cells 2024; 13:745. [PMID: 38727281 PMCID: PMC11083827 DOI: 10.3390/cells13090745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/19/2024] [Accepted: 03/19/2024] [Indexed: 05/13/2024] Open
Abstract
This review delves into the groundbreaking impact of induced pluripotent stem cells (iPSCs) and three-dimensional organoid models in propelling forward neuropathology research. With a focus on neurodegenerative diseases, neuromotor disorders, and related conditions, iPSCs provide a platform for personalized disease modeling, holding significant potential for regenerative therapy and drug discovery. The adaptability of iPSCs, along with associated methodologies, enables the generation of various types of neural cell differentiations and their integration into three-dimensional organoid models, effectively replicating complex tissue structures in vitro. Key advancements in organoid and iPSC generation protocols, alongside the careful selection of donor cell types, are emphasized as critical steps in harnessing these technologies to mitigate tumorigenic risks and other hurdles. Encouragingly, iPSCs show promising outcomes in regenerative therapies, as evidenced by their successful application in animal models.
Collapse
Affiliation(s)
- Douglas Bottega Pazzin
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (D.B.P.); (T.T.R.P.); (J.I.B.G.); (G.Z.); (F.A.C.X.); (J.C.d.C.)
- Graduate Program in Pediatrics and Child Health, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619-900, Brazil
| | - Thales Thor Ramos Previato
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (D.B.P.); (T.T.R.P.); (J.I.B.G.); (G.Z.); (F.A.C.X.); (J.C.d.C.)
- Graduate Program in Biomedical Gerontology, School of Medicine, Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90619-900, Brazil
| | - João Ismael Budelon Gonçalves
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (D.B.P.); (T.T.R.P.); (J.I.B.G.); (G.Z.); (F.A.C.X.); (J.C.d.C.)
| | - Gabriele Zanirati
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (D.B.P.); (T.T.R.P.); (J.I.B.G.); (G.Z.); (F.A.C.X.); (J.C.d.C.)
| | - Fernando Antonio Costa Xavier
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (D.B.P.); (T.T.R.P.); (J.I.B.G.); (G.Z.); (F.A.C.X.); (J.C.d.C.)
| | - Jaderson Costa da Costa
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (D.B.P.); (T.T.R.P.); (J.I.B.G.); (G.Z.); (F.A.C.X.); (J.C.d.C.)
| | - Daniel Rodrigo Marinowic
- Brain Institute of Rio Grande do Sul (BraIns), Pontifical Catholic University of Rio Grande do Sul, Porto Alegre 90610-000, Brazil; (D.B.P.); (T.T.R.P.); (J.I.B.G.); (G.Z.); (F.A.C.X.); (J.C.d.C.)
| |
Collapse
|
9
|
Kim D, Yadav D, Song M. An updated review on animal models to study attention-deficit hyperactivity disorder. Transl Psychiatry 2024; 14:187. [PMID: 38605002 PMCID: PMC11009407 DOI: 10.1038/s41398-024-02893-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Attention-deficit hyperactivity disorder (ADHD) is a neuropsychiatric disorder affecting both children and adolescents. Individuals with ADHD experience heterogeneous problems, such as difficulty in attention, behavioral hyperactivity, and impulsivity. Recent studies have shown that complex genetic factors play a role in attention-deficit hyperactivity disorders. Animal models with clear hereditary traits are crucial for studying the molecular, biological, and brain circuit mechanisms underlying ADHD. Owing to their well-managed genetic origins and the relative simplicity with which the function of neuronal circuits is clearly established, models of mice can help learn the mechanisms involved in ADHD. Therefore, in this review, we highlighting the important genetic animal models that can be used to study ADHD.
Collapse
Affiliation(s)
- Daegeon Kim
- Department of Life Science, Yeungnam University, Gyeongsan-si, South Korea
| | - Dhananjay Yadav
- Department of Life Science, Yeungnam University, Gyeongsan-si, South Korea
| | - Minseok Song
- Department of Life Science, Yeungnam University, Gyeongsan-si, South Korea.
| |
Collapse
|
10
|
Peake M, Dunnill C, Ibraheem K, Smith A, Clarke DJ, Georgopoulos NT. A novel method for the establishment of autologous skin cell suspensions: characterisation of cellular sub-populations, epidermal stem cell content and wound response-enhancing biological properties. Front Bioeng Biotechnol 2024; 12:1386896. [PMID: 38646012 PMCID: PMC11026634 DOI: 10.3389/fbioe.2024.1386896] [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: 02/16/2024] [Accepted: 03/25/2024] [Indexed: 04/23/2024] Open
Abstract
Introduction: Autologous cell suspension (ACS)-based therapy represents a highly promising approach for burns and chronic wounds. However, existing technologies have not achieved the desired clinical success due to several limitations. To overcome practical and cost-associated obstacles of existing ACS methods, we have established a novel methodology for rapid, enzymatic disaggregation of human skin cells and their isolation using a procedure that requires no specialist laboratory instrumentation and is performed at room temperature. Methods: Cells were isolated using enzymatic disaggregation of split-thickness human skin followed by several filtration steps for isolation of cell populations, and cell viability was determined. Individual population recovery was confirmed in appropriate culture medium types, and the presence of epidermal stem cells (EpSCs) within keratinocyte sub-populations was defined by flow cytometry via detection of CD49 and CD71. Positive mediators of wound healing secreted by ACS-derived cultures established on a collagen-based wound-bed mimic were detected by proteome arrays and quantified by ELISA, and the role of such mediators was determined by cell proliferation assays. The effect of ACS-derived conditioned-medium on myofibroblasts was investigated using an in-vitro model of myofibroblast differentiation via detection of α-SMA using immunoblotting and immunofluorescence microscopy. Results: Our methodology permitted efficient recovery of keratinocytes, fibroblasts and melanocytes, which remained viable upon long-term culture. ACS-derivatives comprised sub-populations with the CD49-high/CD71-low expression profile known to demarcate EpSCs. Via secretion of mitogenic factors and wound healing-enhancing mediators, the ACS secretome accelerated keratinocyte proliferation and markedly curtailed cytodifferentiation of myofibroblasts, the latter being key mediators of fibrosis and scarring. Discussion: The systematic characterisation of the cell types within our ACS isolates provided evidence for their superior cell viability and the presence of EpSCs that are critical drivers of wound healing. We defined the biological properties of ACS-derived keratinocytes, which include ability to secrete positive mediators of wound healing as well as suppression of myofibroblast cytodifferentiation. Thus, our study provides several lines of evidence that the established ACS isolates comprise highly-viable cell populations which can physically support wound healing and possess biological properties that have the potential to enhance not only the speed but also the quality of wound healing.
Collapse
Affiliation(s)
- Michael Peake
- School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Centre for Dermatology Research, Division of Musculoskeletal and Dermatological Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, NIHR Manchester Biomedical Research Centre, Manchester, United Kingdom
| | - Chris Dunnill
- School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
| | - Khalidah Ibraheem
- School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
| | - Adrian Smith
- Department of General Surgery, Calderdale and Huddersfield NHS Foundation Trust, Huddersfield, United Kingdom
| | - Douglas J. Clarke
- School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
| | - Nikolaos T. Georgopoulos
- School of Applied Sciences, University of Huddersfield, Huddersfield, United Kingdom
- Biomolecular Sciences Research Centre, Industry and Innovation Research Institute, Sheffield Hallam University, Sheffield, United Kingdom
| |
Collapse
|
11
|
Benchaprathanphorn K, Muangman P, Chinaroonchai K, Namviriyachote N, Ampawong S, Angkhasirisap W, Kengkoom K, Viravaidya-Pasuwat K. Translational application of human keratinocyte-fibroblast cell sheets for accelerated wound healing in a clinically relevant type 2 diabetic rat model. Cytotherapy 2024; 26:360-371. [PMID: 38363247 DOI: 10.1016/j.jcyt.2024.01.003] [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: 08/09/2023] [Revised: 12/27/2023] [Accepted: 01/20/2024] [Indexed: 02/17/2024]
Abstract
BACKGROUND AIMS Despite advancements in wound care, wound healing remains a challenge, especially in individuals with type 2 diabetes. Cell sheet technology has emerged as an efficient and promising therapy for tissue regeneration and wound repair. Among these, bilayered human keratinocyte-fibroblast cell sheets constructed using temperature-responsive culture surfaces have been shown to mimic a normal tissue-like structure and secrete essential cytokines and growth factors that regulate the wound healing process. METHODS This study aimed to evaluate the safety and therapeutic potential of human skin cell sheets to treat full-thickness skin defects in a rat model of type 2 diabetes. RESULTS Our findings demonstrate that diabetic wounds transplanted with bilayered cell sheets resulted in accelerated re-epithelialization, increased angiogenesis, enhanced macrophage polarization and regeneration of tissue that closely resembled healthy skin. In contrast, the control group that did not receive cell sheet transplantation presented characteristic symptoms of impaired and delayed wound healing associated with type 2 diabetes. CONCLUSIONS The secretory cytokines and the upregulation of Nrf2 expression in response to cell sheet transplantation are believed to have played a key role in the improved wound healing observed in diabetic rats. Our study suggests that human keratinocyte-fibroblast cell sheets hold great potential as a therapeutic alternative for diabetic ulcers.
Collapse
Affiliation(s)
- Kanokaon Benchaprathanphorn
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand
| | - Pornprom Muangman
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Kusuma Chinaroonchai
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Nantaporn Namviriyachote
- Trauma Surgery Division, Department of Surgery, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wannee Angkhasirisap
- Research and Academic Support Office, National Laboratory Animal Center, Mahidol University, Nakorn Pathom, Thailand
| | - Kanchana Kengkoom
- Research and Academic Support Office, National Laboratory Animal Center, Mahidol University, Nakorn Pathom, Thailand
| | - Kwanchanok Viravaidya-Pasuwat
- Biological Engineering Program, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand; Chemical Engineering, Faculty of Engineering, King Mongkut's University of Technology Thonburi, Bangkok, Thailand.
| |
Collapse
|
12
|
Trinh TT, Choi JH, Yang J, Kim WH, Chien PN, Le LTT, Ngan‐Giang N, Nga PT, Nam S, Heo C. Effects on keratinocytes of the traditional combination of herb extract (Royal Oji Complex) implicated the improvement of young children's skin moisture and barrier. Skin Res Technol 2024; 30:e13682. [PMID: 38616504 PMCID: PMC11016816 DOI: 10.1111/srt.13682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/16/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND Natural products are often friendly and can be used on children's skin after systematic and careful research. Therefore, in this study, the Royal Oji Complex (ROC), a product with natural ingredients, was used to study their effectiveness on keratinocytes taken from the skin of children from 0 to 3 years old. METHOD Normal human epidermal keratinocytes and tissue-isolated keratinocytes (TIKC) from young donors were treated with three different concentrations of ROC: 0.1, 1, and 10 ppm. The mRNA expression of the epidermal barrier's essential genes, such as hyaluronic acid synthase 3 (Has3), involucrin (IVL), loricrin (LOR), and claudin-1 (CLD1) was investigated using qRT-PCR. Ceramide content was measured by ELISA, with retinoic acid (R.A.) and amarogentin (AMA) serving as positive controls. RESULTS ROC significantly elevated HAS3 gene expression in HEKn cells, especially at 10 ppm, indicating potential advantages for skin hydration in young infants. IVL increased at first but decreased as ROC concentrations increased. LOR was upregulated at lower ROC concentrations but reduced at higher doses. CLD1 gene expression increased considerably in HEKn but reduced with increasing ROC doses. Ceramide concentration increased somewhat but not significantly at 10 ppm. CONCLUSION ROC shows potential in altering keratinocyte gene expression, with unique responses in HEKn and TIKC from young donors. While changes in ceramide content were insignificant, these results help to comprehend ROC's multiple effects on young children's skin.
Collapse
Affiliation(s)
- Thuy‐Tien Thi Trinh
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
| | | | - Jee‐Eun Yang
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
- Korean Institute of Nonclinical StudySeongnamRepublic of Korea
| | | | - Pham Ngoc Chien
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
- Korean Institute of Nonclinical StudySeongnamRepublic of Korea
| | - Linh Thi Thuy Le
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
- Department of Biomedical ScienceCollege of MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Nguyen Ngan‐Giang
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
- Department of Medical Device DevelopmentCollege of MedicineSeoul National UniversitySeoulRepublic of Korea
| | - Pham Thi Nga
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
- Korean Institute of Nonclinical StudySeongnamRepublic of Korea
| | - Sun‐Young Nam
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
| | - Chan‐Yeong Heo
- Department of Plastic and Reconstructive SurgerySeoul National University Bundang HospitalSeongnamRepublic of Korea
- Korean Institute of Nonclinical StudySeongnamRepublic of Korea
- Department of Medical Device DevelopmentCollege of MedicineSeoul National UniversitySeoulRepublic of Korea
| |
Collapse
|
13
|
Dubau M, Tripetchr T, Mahmoud L, Kral V, Kleuser B. Advancing skin model development: A focus on a self-assembled, induced pluripotent stem cell-derived, xeno-free approach. J Tissue Eng 2024; 15:20417314241291848. [PMID: 39502328 PMCID: PMC11536386 DOI: 10.1177/20417314241291848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/01/2024] [Indexed: 11/08/2024] Open
Abstract
The demand for skin models as alternatives to animal testing has grown due to ethical concerns and the need for accurate substance evaluation. These alternatives, known as New Approach Methodologies (NAMs), are increasingly used for regulatory decisions. Current skin models from primary human cells often rely on bovine collagen, raising ethical issues. This study explores self-assembled skin models (SASM) as a new method, utilizing hair follicle-derived keratinocytes reprogrammed into induced pluripotent stem cells (iPSC) and differentiated into fibroblasts and keratinocytes. The model relies on the ability of fibroblasts to secrete collagen to produce a xeno-free dermal layer and on the differentiation of keratinocytes to create a functional epidermal layer. These layers exhibited confirmed metabolic activity and the capability to withstand test substances. The successful development of SASM underscores the significance of accurate alternatives in dermatological research, providing an ethical and reliable option for substance evaluation and regulatory testing.
Collapse
Affiliation(s)
| | | | - Lava Mahmoud
- Department of Pharmacology and Toxicology, Institute for Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Vivian Kral
- Department of Pharmacology and Toxicology, Institute for Pharmacy, Freie Universität Berlin, Berlin, Germany
| | - Burkhard Kleuser
- Department of Pharmacology and Toxicology, Institute for Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
14
|
Karakaya T, Slaufova M, Di Filippo M, Hennig P, Fenini G, Kündig T, Beer HD. Efficient Generation of CRISPR/Cas9-Mediated Knockout Human Primary Keratinocytes by Electroporation. Methods Mol Biol 2024; 2849:73-86. [PMID: 38407798 DOI: 10.1007/7651_2024_518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Due to their full differentiation capacity in vitro, the culture of human primary keratinocytes (HPKs) represents a physiological model for answering basic biological and dermatological research questions, including those related to skin diseases and the investigation of treatment options. When modified with the CRISPR/Cas9 gene editing approach and cultivated in organotypic 3D epidermal equivalents (EEs), these human cells have the potential to replace established mouse models. However, even when cultivated on feeder cells, HPKs have only a low proliferation capacity in 2D culture, limiting their application potential. This is particularly true for CRISPR/Cas9-modified HPKs, whose generation commonly requires selection of targeted cells, negatively affecting their lifespan. Here, we describe a robust protocol for the rapid, simple, and efficient generation of single- and multi-gene CRISPR/Cas9 knockout HPKs by electroporation of ribonucleoprotein (RNP) complexes, which comprise one or multiple guide RNAs (gRNAs) and Cas9 protein. Unlike DNA transfection or virus-based targeting strategies, electroporation of RNPs represents a targeting approach that minimizes immunological and toxic side effects. Using efficient gRNAs results in the generation of HPKs with a high yield of knockout cells, allowing for their immediate use in experiments without requiring the laborious process of selecting targeted cells or maintaining a feeder cell culture. Furthermore, the use of RNPs and their delivery via electroporation minimizes off-target and other unspecific effects, preventing unintended genomic alterations. Most importantly, CRISPR/Cas9 knockout HPKs generated with this protocol have the ability to form a fully differentiated epidermis in 3D, thus facilitating the understanding of specific protein functions in a highly physiological human skin model. Alternatively, this approach proves valuable for generating models of mono- or polygenic skin diseases via knockouts, providing insights into the underlying molecular mechanisms and facilitating the development of novel therapeutic approaches.
Collapse
Affiliation(s)
- Tugay Karakaya
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Marta Slaufova
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Michela Di Filippo
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Paulina Hennig
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Gabriele Fenini
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
| | - Thomas Kündig
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Hans-Dietmar Beer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland.
- Faculty of Medicine, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
Johnson MR, Li S, Guerrero-Juarez CF, Miller P, Brack BJ, Mereby SA, Moreno JA, Feigin CY, Gaska J, Rivera-Perez JA, Nie Q, Ploss A, Shvartsman SY, Mallarino R. A multifunctional Wnt regulator underlies the evolution of rodent stripe patterns. Nat Ecol Evol 2023; 7:2143-2159. [PMID: 37813945 PMCID: PMC10839778 DOI: 10.1038/s41559-023-02213-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 08/27/2023] [Indexed: 10/11/2023]
Abstract
Animal pigment patterns are excellent models to elucidate mechanisms of biological organization. Although theoretical simulations, such as Turing reaction-diffusion systems, recapitulate many animal patterns, they are insufficient to account for those showing a high degree of spatial organization and reproducibility. Here, we study the coat of the African striped mouse (Rhabdomys pumilio) to uncover how periodic stripes form. Combining transcriptomics, mathematical modelling and mouse transgenics, we show that the Wnt modulator Sfrp2 regulates the distribution of hair follicles and establishes an embryonic prepattern that foreshadows pigment stripes. Moreover, by developing in vivo gene editing in striped mice, we find that Sfrp2 knockout is sufficient to alter the stripe pattern. Strikingly, mutants exhibited changes in pigmentation, revealing that Sfrp2 also regulates hair colour. Lastly, through evolutionary analyses, we find that striped mice have evolved lineage-specific changes in regulatory elements surrounding Sfrp2, many of which may be implicated in modulating the expression of this gene. Altogether, our results show that a single factor controls coat pattern formation by acting both as an orienting signalling mechanism and a modulator of pigmentation. More broadly, our work provides insights into how spatial patterns are established in developing embryos and the mechanisms by which phenotypic novelty originates.
Collapse
Affiliation(s)
- Matthew R Johnson
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sha Li
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Christian F Guerrero-Juarez
- Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, USA
| | - Pearson Miller
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
| | - Benjamin J Brack
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Sarah A Mereby
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Jorge A Moreno
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Charles Y Feigin
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Jenna Gaska
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | | | - Qing Nie
- Department of Developmental and Cell Biology, University of California, Irvine, CA, USA
- Department of Mathematics, University of California, Irvine, CA, USA
- Center for Complex Biological Systems, University of California, Irvine, CA, USA
- NSF-Simons Center for Multiscale Cell Fate Research, University of California, Irvine, CA, USA
| | - Alexander Ploss
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
| | - Stanislav Y Shvartsman
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA
- Center for Computational Biology, Flatiron Institute, New York, NY, USA
- The Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ, USA
| | - Ricardo Mallarino
- Department of Molecular Biology, Princeton University, Princeton, NJ, USA.
| |
Collapse
|
16
|
Neely AE, Zhang Y, Blumensaadt LA, Mao H, Brenner B, Sun C, Zhang HF, Bao X. Nucleoporin downregulation modulates progenitor differentiation independent of nuclear pore numbers. Commun Biol 2023; 6:1033. [PMID: 37853046 PMCID: PMC10584948 DOI: 10.1038/s42003-023-05398-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
Nucleoporins (NUPs) comprise nuclear pore complexes, gateways for nucleocytoplasmic transport. As primary human keratinocytes switch from the progenitor state towards differentiation, most NUPs are strongly downregulated, with NUP93 being the most downregulated NUP in this process. To determine if this NUP downregulation is accompanied by a reduction in nuclear pore numbers, we leveraged Stochastic Optical Reconstruction Microscopy. No significant changes in nuclear pore numbers were detected using three independent NUP antibodies; however, NUP reduction in other subcellular compartments such as the cytoplasm was identified. To investigate how NUP reduction influences keratinocyte differentiation, we knocked down NUP93 in keratinocytes in the progenitor-state culture condition. NUP93 knockdown diminished keratinocytes' clonogenicity and epidermal regenerative capacity, without drastically affecting nuclear pore numbers or permeability. Using transcriptome profiling, we identified that NUP93 knockdown induces differentiation genes related to both mechanical and immune barrier functions, including the activation of known NF-κB target genes. Consistently, keratinocytes with NUP93 knockdown exhibited increased nuclear localization of the NF-κB p65/p50 transcription factors, and increased NF-κB reporter activity. Taken together, these findings highlight the gene regulatory roles contributed by differential NUP expression levels in keratinocyte differentiation, independent of nuclear pore numbers.
Collapse
Affiliation(s)
- Amy E Neely
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Yang Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA.
- Molecular Analytics and Photonics (MAP) Lab, Department of Textile Engineering, Chemistry and Science, North Carolina State University, Raleigh, NC, 27606, USA.
| | - Laura A Blumensaadt
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA
| | - Hongjing Mao
- Molecular Analytics and Photonics (MAP) Lab, Department of Textile Engineering, Chemistry and Science, North Carolina State University, Raleigh, NC, 27606, USA
| | - Benjamin Brenner
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Cheng Sun
- Department of Mechanical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Hao F Zhang
- Department of Biomedical Engineering, Northwestern University, Evanston, IL, 60208, USA
| | - Xiaomin Bao
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, 60208, USA.
- Department of Dermatology, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
17
|
Niwetbowornchai N, Chaisirirat T, Sriswasdi S, Saithong S, Filbertine G, Wright HL, Edwards SW, Virakul S, Chiewchengchol D. Regulation of dermal fibroblasts by human neutrophil peptides. Sci Rep 2023; 13:17499. [PMID: 37840103 PMCID: PMC10577140 DOI: 10.1038/s41598-023-44889-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 10/13/2023] [Indexed: 10/17/2023] Open
Abstract
Human neutrophil peptides (HNPs) can induce cell proliferation and activation so their growth promoting activities may have potential clinical benefit. This study investigated the effects of HNPs on human dermal fibroblasts. Differential gene expression in HNP-treated cells and genes involved in regulating intracellular pathways were explored. Dermal fibroblasts were isolated from healthy neonatal foreskin and treated with HNPs in 2D and 3D cell culture systems. The expression of cell proliferation (Ki-67) gene and cell activation (COL1A1) gene plus their proteins was measured. Differential gene expression was determined using RNA-seq, and upregulated and downregulated genes were mapped onto intracellular pathways by KEGG analysis and Gene Ontology databases. HNPs significantly increased cell proliferation without cytotoxicity whilst HNP1 enhanced expression of COL1A1 and type I collagen production in 2D cells and 3D spheroids. RNA-sequencing analysis showed gene clustering with clear separation between HNP1-treated and control groups. A heatmap of top 50 differentially expressed genes was consistent among HNP1-treated samples. Most upregulated genes were associated with cell proliferation and activation as mapped into intracellular pathways whilst most downregulated genes belonged to steroid/arachidonic acid metabolism and inflammatory signaling pathways. HNP1 increased cell proliferation and activation but reduced lipid metabolism and inflammation.
Collapse
Affiliation(s)
- Nattarika Niwetbowornchai
- Center of Excellence in Translational Research in Inflammation and Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Immunology Unit, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Thanawat Chaisirirat
- Center of Excellence in Computational Molecular Biology, Chulalongkorn University, Bangkok, Thailand
| | - Sira Sriswasdi
- Center of Excellence in Computational Molecular Biology, Chulalongkorn University, Bangkok, Thailand
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Supichcha Saithong
- Center of Excellence in Translational Research in Inflammation and Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Immunology Unit, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Grace Filbertine
- Center of Excellence in Translational Research in Inflammation and Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Immunology Unit, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Helen L Wright
- Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Steven W Edwards
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, UK
| | - Sita Virakul
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Immunology Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Microbiology, Faculty of Science, Chulalongkorn University, Bangkok, Thailand
| | - Direkrit Chiewchengchol
- Center of Excellence in Translational Research in Inflammation and Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.
- Center of Excellence in Immunology and Immune-Mediated Diseases, Department of Microbiology, Faculty of Medicine, Immunology Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
18
|
Sierra-Sánchez Á, Barbier MA, Magne B, Larouche D, Arias-Santiago S, Germain L. Comparison of Two Human Skin Cell Isolation Protocols and Their Influence on Keratinocyte and Fibroblast Culture. Int J Mol Sci 2023; 24:14712. [PMID: 37834159 PMCID: PMC10572435 DOI: 10.3390/ijms241914712] [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: 08/22/2023] [Revised: 09/24/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
For the development of advanced therapies, the use of primary cells instead of cell lines is preferred. The manufacture of human tissue-engineered skin substitutes requires efficient isolation and culture protocols allowing a massive expansion of the cells in culture from an initial specimen of a minimal size. This study compared two skin cell isolation protocols, routinely applied in two clinical laboratories. Epithelial (keratinocytes) and dermal (fibroblasts) cells were isolated and cultured from three human skin biopsies (N = 3). The two-step digestion protocol (LOEX-Protocol) firstly used thermolysin to enzymatically disrupt the dermal-epidermal junction while, for the one-step digestion protocol (UPCIT-Protocol), mechanical detachment with scissors was applied. Then, the epidermal and dermal layers were digested, respectively, to achieve cell isolation. The cell size, viability, yield and growth were analyzed over five passages (P). The colony-forming efficiency (CFE) and Keratin 19 (K19) expression of epithelial cells were also assessed after P0 and P1. Regarding the dermal cells, no significant differences were observed in the tested parameters of isolation and culture. However, for the epithelial cells, viability was higher (93% vs. 85%) and the number of cells extracted per cm2 of skin was 3.4 times higher using the LOEX-Protocol compared to the UPCIT-Protocol. No significant difference was observed for any parameter once the keratinocytes were cultured from P1 to P4. The CFE and K19 expression decreased from P0 to P1 in both protocols, probably due to the culture process. This study shows that both protocols enable the efficient isolation of skin dermal and epithelial cells and subsequent culture to produce grafts destined for the treatment of patients.
Collapse
Affiliation(s)
- Álvaro Sierra-Sánchez
- LOEX Tissue Engineering Laboratory, Université Laval Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec City, QC G1J 1Z4, Canada
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Center, Québec City, QC G1J 1Z4, Canada
- Unidad de Producción Celular e Ingeniería Tisular (UPCIT), Virgen de las Nieves University Hospital, ibs.Granada, Andalusian Network of Design and Translation of Advanced Therapies, 18014 Granada, Spain
| | - Martin A Barbier
- LOEX Tissue Engineering Laboratory, Université Laval Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec City, QC G1J 1Z4, Canada
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Center, Québec City, QC G1J 1Z4, Canada
| | - Brice Magne
- LOEX Tissue Engineering Laboratory, Université Laval Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec City, QC G1J 1Z4, Canada
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Center, Québec City, QC G1J 1Z4, Canada
| | - Danielle Larouche
- LOEX Tissue Engineering Laboratory, Université Laval Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec City, QC G1J 1Z4, Canada
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Center, Québec City, QC G1J 1Z4, Canada
| | - Salvador Arias-Santiago
- Unidad de Producción Celular e Ingeniería Tisular (UPCIT), Virgen de las Nieves University Hospital, ibs.Granada, Andalusian Network of Design and Translation of Advanced Therapies, 18014 Granada, Spain
- Department of Dermatology, Virgen de las Nieves University Hospital, 18012 Granada, Spain
- Department of Dermatology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Lucie Germain
- LOEX Tissue Engineering Laboratory, Université Laval Research Center and Department of Surgery, Faculty of Medicine, Université Laval, Québec City, QC G1J 1Z4, Canada
- Division of Regenerative Medicine, CHU de Québec-Université Laval Research Center, Québec City, QC G1J 1Z4, Canada
| |
Collapse
|
19
|
Chen C, Guan MX. Induced pluripotent stem cells: ex vivo models for human diseases due to mitochondrial DNA mutations. J Biomed Sci 2023; 30:82. [PMID: 37737178 PMCID: PMC10515435 DOI: 10.1186/s12929-023-00967-7] [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: 04/05/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023] Open
Abstract
Mitochondria are essential organelles for cellular metabolism and physiology in eukaryotic cells. Human mitochondria have their own genome (mtDNA), which is maternally inherited with 37 genes, encoding 13 polypeptides for oxidative phosphorylation, and 22 tRNAs and 2 rRNAs for translation. mtDNA mutations are associated with a wide spectrum of degenerative and neuromuscular diseases. However, the pathophysiology of mitochondrial diseases, especially for threshold effect and tissue specificity, is not well understood and there is no effective treatment for these disorders. Especially, the lack of appropriate cell and animal disease models has been significant obstacles for deep elucidating the pathophysiology of maternally transmitted diseases and developing the effective therapy approach. The use of human induced pluripotent stem cells (iPSCs) derived from patients to obtain terminally differentiated specific lineages such as inner ear hair cells is a revolutionary approach to deeply understand pathogenic mechanisms and develop the therapeutic interventions of mitochondrial disorders. Here, we review the recent advances in patients-derived iPSCs as ex vivo models for mitochondrial diseases. Those patients-derived iPSCs have been differentiated into specific targeting cells such as retinal ganglion cells and eventually organoid for the disease modeling. These disease models have advanced our understanding of the pathophysiology of maternally inherited diseases and stepped toward therapeutic interventions for these diseases.
Collapse
Affiliation(s)
- Chao Chen
- Center for Mitochondrial Biomedicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Min-Xin Guan
- Center for Mitochondrial Biomedicine, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Division of Medical Genetics and Genomics, The Children's Hospital, Zhejiang University School of Medicine and National Clinical Research Center for Child Health, Hangzhou, Zhejiang, China.
- Institute of Genetics, Zhejiang University International School of Medicine, 866 Yuhangtang Road, Hangzhou, 310058, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Genetic and Developmental Disorders, Hangzhou, Zhejiang, China.
- Key Lab of Reproductive Genetics, Ministry of Education of PRC, Zhejiang University, Hangzhou, Zhejiang, China.
| |
Collapse
|
20
|
Witkowski TA, Li B, Andersen JG, Kumar B, Mroz EA, Rocco JW. Y-27632 acts beyond ROCK inhibition to maintain epidermal stem-like cells in culture. J Cell Sci 2023; 136:jcs260990. [PMID: 37698512 PMCID: PMC10508688 DOI: 10.1242/jcs.260990] [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: 01/17/2023] [Accepted: 07/24/2023] [Indexed: 09/13/2023] Open
Abstract
Conditional reprogramming is a cell culture technique that effectively immortalizes epithelial cells with normal genotypes by renewing epidermal stem cells. Y-27632, a compound that promotes conditional reprogramming through an unknown mechanism, was developed to inhibit the two Rho-associated kinase (ROCK) isoforms. We used human foreskin keratinocytes (HFKs) to study the role of Y-27632 in conditional reprogramming and learn how ROCKs control epidermal stem cell renewal. In conditional reprogramming, Y-27632 increased HFK adherence to culture dishes, progression through S, G2 and M phases of the cell cycle, and epidermal stem cell marker levels. Although this correlated with ROCK inhibition by Y-27632, we generated CRISPR-Cas9-mediated HFK ROCK knockouts to test the direct role of ROCK inhibition. Knockout of single ROCK isoforms was insufficient to disrupt ROCK activity or promote HFK propagation without Y-27632. Although ROCK activity was reduced, HFKs with double knockout of ROCK1 and ROCK2 still required Y-27632 to propagate. Y-27632 was the most effective among the ROCK inhibitors we tested at promoting HFK proliferation and epidermal stem cell marker expression. Thus, the ability of Y-27632 to promote an epidermal stem cell state in conditional reprogramming not only depends upon ROCK inhibition but also acts via as-yet-unidentified mechanisms. Epidermal stem cell renewal might in part be regulated by ROCKs, but also involves additional pathways.
Collapse
Affiliation(s)
- Travis A. Witkowski
- Department of Otolaryngology – Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Bin Li
- Department of Otolaryngology – Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jason G. Andersen
- Department of Otolaryngology – Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Bhavna Kumar
- Department of Otolaryngology – Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Edmund A. Mroz
- Department of Otolaryngology – Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - James W. Rocco
- Department of Otolaryngology – Head and Neck Surgery, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH 43210, USA
- The Ohio State University Comprehensive Cancer Center – James, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
21
|
De Silva WGM, McCarthy BY, Han J, Yang C, Holland AJA, Stern H, Dixon KM, Tang EKY, Tuckey RC, Rybchyn MS, Mason RS. The Over-Irradiation Metabolite Derivative, 24-Hydroxylumister-ol 3, Reduces UV-Induced Damage in Skin. Metabolites 2023; 13:775. [PMID: 37512482 PMCID: PMC10383208 DOI: 10.3390/metabo13070775] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/13/2023] [Accepted: 06/16/2023] [Indexed: 07/30/2023] Open
Abstract
The hormonal form of vitamin D3, 1,25(OH)2D3, reduces UV-induced DNA damage. UV exposure initiates pre-vitamin D3 production in the skin, and continued UV exposure photoisomerizes pre-vitamin D3 to produce "over-irradiation products" such as lumisterol3 (L3). Cytochrome P450 side-chain cleavage enzyme (CYP11A1) in skin catalyzes the conversion of L3 to produce three main derivatives: 24-hydroxy-L3 [24(OH)L3], 22-hydroxy-L3 [22(OH)L3], and 20,22-dihydroxy-L3 [20,22(OH)L3]. The current study investigated the photoprotective properties of the major over-irradiation metabolite, 24(OH)L3, in human primary keratinocytes and human skin explants. The results indicated that treatment immediately after UV with either 24(OH)L3 or 1,25(OH)2D3 reduced UV-induced cyclobutane pyrimidine dimers and oxidative DNA damage, with similar concentration response curves in keratinocytes, although in skin explants, 1,25(OH)2D3 was more potent. The reductions in DNA damage by both compounds were, at least in part, the result of increased DNA repair through increased energy availability via increased glycolysis, as well as increased DNA damage recognition proteins in the nucleotide excision repair pathway. Reductions in UV-induced DNA photolesions by either compound occurred in the presence of lower reactive oxygen species. The results indicated that under in vitro and ex vivo conditions, 24(OH)L3 provided photoprotection against UV damage similar to that of 1,25(OH)2D3.
Collapse
Affiliation(s)
| | - Bianca Yuko McCarthy
- School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Jeremy Han
- School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Chen Yang
- School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Andrew J A Holland
- Douglas Cohen Department of Paediatric Surgery, The Children's Hospital at Westmead Clinical School, The Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Harvey Stern
- Department of Plastic and Constructive Surgery, The Royal Prince Alfred Hospital, Sydney, NSW 2050, Australia
- Strathfield Private Hospital, Sydney, NSW 2042, Australia
| | - Katie Marie Dixon
- School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Edith Kai Yan Tang
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Robert Charles Tuckey
- School of Molecular Sciences, The University of Western Australia, Perth, WA 6009, Australia
| | - Mark Stephen Rybchyn
- School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
| | - Rebecca Sara Mason
- School of Medical Sciences and Bosch Institute, The University of Sydney, Sydney, NSW 2006, Australia
- School of Life and Environmental Sciences, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
22
|
Palmer MA, Dias IHK, Smart E, Benatzy Y, Haslam IS. Cholesterol homeostasis in hair follicle keratinocytes is disrupted by impaired ABCA5 activity. Biochim Biophys Acta Mol Cell Biol Lipids 2023:159361. [PMID: 37348644 DOI: 10.1016/j.bbalip.2023.159361] [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: 09/30/2022] [Revised: 05/27/2023] [Accepted: 06/09/2023] [Indexed: 06/24/2023]
Abstract
The importance of cholesterol in hair follicle biology is underscored by its links to the pathogenesis of alopecias and hair growth disorders. Reports have associated defects in ABCA5, a membrane transporter, with altered keratinocyte cholesterol distribution in individuals with a form of congenital hypertrichosis, yet the biological basis for this defect in hair growth remains unknown. This study aimed to determine the impact of altered ABCA5 activity on hair follicle keratinocyte behaviour. Primary keratinocytes isolated from the outer root sheath of plucked human hair follicles were utilised as a relevant cell model. Following exogenous cholesterol loading, an increase in ABCA5 co-localisation to intracellular organelles was seen. Knockdown of ABCA5 revealed a dysregulation in cholesterol homeostasis, with LXR agonism leading to partial restoration of the homeostatic response. Filipin staining and live BODIPY cholesterol immunofluorescence microscopy revealed a reduction in endo-lysosomal cholesterol following ABCA5 knockdown. Analysis of oxysterols showed a significant increase in the fold change of 25-hydroxycholesterol and 7-β-hydroxycholesterol following cholesterol loading in ORS keratinocytes, after ABCA5 knockdown. These data suggest a role for ABCA5 in the intracellular compartmentalisation of free cholesterol in primary hair follicle keratinocytes. The loss of normal homeostatic response, following the delivery of excess cholesterol after ABCA5 knockdown, suggests an impact on LXR-mediated transcriptional activity. The loss of ABCA5 in the hair follicle could lead to impaired endo-lysosomal cholesterol transport, impacting pathways known to influence hair growth. This avenue warrants further investigation.
Collapse
Affiliation(s)
- Megan A Palmer
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK; Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | | | - Eleanor Smart
- Centre for Dermatology Research, University of Manchester, UK
| | - Yvonne Benatzy
- Faculty of Medicine, Institute of Biochemistry I, Goethe-University Frankfurt, Frankfurt, Germany
| | - Iain S Haslam
- School of Applied Sciences, University of Huddersfield, Huddersfield, UK.
| |
Collapse
|
23
|
Agarwal R, Dittmar T, Beer HD, Kunz M, Müller S, Kappos EA, Contassot E, Navarini AA. Human epidermis organotypic cultures, a reproducible system recapitulating the epidermis in vitro. Exp Dermatol 2023. [PMID: 37114406 DOI: 10.1111/exd.14823] [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: 02/18/2022] [Revised: 03/01/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023]
Abstract
The translatability of research is highly dependent on models that recapitulate human tissues and organs. Here, we describe a procedure for the generation of human epidermis organotypic cultures (HEOCs) from primary keratinocytes isolated from foreskin and adult skin as well as from an immortalized keratinocyte cell line (KerTr). We tested several media conditions to develop a defined HEOC growing and expansion media. We characterized the HEOCs and show that in optimal culture conditions they express the proliferation marker Ki67, the basement membrane protein collagen 17 (col17) and the epidermal differentiation markers keratin 15 (K15), keratin 14 (K14), keratin 5 (K5), keratin 10 (K10), keratin 1 (K1), transglutaminase 1 (TGM1), transglutaminase 3 (TGM3) and filaggrin (FLG). Thus, they recapitulate the human epidermis and are stratified from the basal layer to the stratum corneum. These HEOC can be generated reproducibly on a large scale, making it an invaluable model for screening therapeutic compounds and also for the study of pathologies affecting the epidermis.
Collapse
Affiliation(s)
- Rishika Agarwal
- Dermatology Department, University Hospital of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Tanja Dittmar
- Dermatology Department, University Hospital of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Hans-Dietmar Beer
- Dermatology Department, University Hospital of Zurich, Zurich, Switzerland
| | - Michael Kunz
- Dermatology Department, University Hospital of Basel, Basel, Switzerland
| | - Simon Müller
- Dermatology Department, University Hospital of Basel, Basel, Switzerland
| | - Elisabeth A Kappos
- Department of Plastic, Reconstructive, Aesthetic and Hand Surgery, University Hospital Basel, Basel, Switzerland
| | - Emmanuel Contassot
- Dermatology Department, University Hospital of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alexander A Navarini
- Dermatology Department, University Hospital of Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| |
Collapse
|
24
|
Wijaya LK, Morici MV, Stumbles PA, Finch PM, Drummond PD. Stimulation of alpha-1 adrenoceptors may intensify cutaneous inflammation in complex regional pain syndrome. Pain 2023; 164:771-781. [PMID: 35994594 DOI: 10.1097/j.pain.0000000000002764] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 08/09/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT Alpha-1 adrenoceptors are overexpressed in the epidermis of a subgroup of patients with complex regional pain syndrome (CRPS). Activating α 1 -adrenoceptors in epidermal cells increases production of the proinflammatory cytokine interleukin-6 (IL-6), a mediator of inflammation. To investigate whether this might exacerbate inflammation in CRPS, primary keratinocytes or dermal fibroblasts were cultured from skin biopsies obtained from the affected limb of 25 patients and a similar site in 28 controls. The fundamental proinflammatory cytokine, tumor necrosis factor alpha, was administered for 24 hours to initiate inflammation. After this, cells were incubated for 6 hours with the α 1 -adrenoceptor agonist phenylephrine. Exposure to tumor necrosis factor alpha induced proinflammatory cytokine mRNA production and protein secretion in keratinocytes and fibroblasts and enhanced α 1B -adrenoceptor mRNA expression in keratinocytes. Additional stimulation of α 1 adrenoceptors with phenylephrine increased the production of IL-6 mRNA and protein secretion in both cell types. Under all conditions, gene and protein α 1 -adrenoceptor levels and cytokine gene expression and protein secretion were similar, overall, in patients and controls, except for abnormally high α 1 -adrenoceptor protein levels in the keratinocytes of 3 of 17 patients. These findings suggest that persistent inflammation in CRPS is not due to dysfunction of skin cells but is a normal response to extrinsic signals. After α 1 -adrenoceptor stimulation of keratinocytes, increases in IL-6 mRNA but not protein were proportional to basal α 1 -adrenoceptor protein levels. Skin cells play an important role in persistent inflammation in CRPS. Potentially, a positive feedback loop between α 1 -adrenoceptors and IL-6 production in skin cells contributes to this inflammatory state.
Collapse
Affiliation(s)
- Linda K Wijaya
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
- Telethon Kids Institute, Perth, Australia
| | - Michael V Morici
- Telethon Kids Institute, Perth, Australia
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia
| | - Philip A Stumbles
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
- Telethon Kids Institute, Perth, Australia
| | - Philip M Finch
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
| | - Peter D Drummond
- College of Science, Health, Engineering and Education, Murdoch University, Perth, Australia
| |
Collapse
|
25
|
Jin M, Bang JS, Ha DL, Kim JY, Park KD, Lee WJ, Lee SJ, Choi JK, Choi YA, Jang YH, Kim SH. Dermatophagoides farinae Extract Induces Interleukin 33-Mediated Atopic Skin Inflammation via Activation of RIP1. Int J Mol Sci 2023; 24:ijms24065228. [PMID: 36982304 PMCID: PMC10049056 DOI: 10.3390/ijms24065228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Receptor-interacting protein kinase (RIP) family 1 signaling has complex effects on inflammatory processes and cell death, but little is known concerning allergic skin diseases. We examined the role of RIP1 in Dermatophagoides farinae extract (DFE)-induced atopic dermatitis (AD)-like skin inflammation. RIP1 phosphorylation was increased in HKCs treated with DFE. Nectostatin-1, a selective and potent allosteric inhibitor of RIP1, inhibited AD-like skin inflammation and the expression of histamine, total IgE, DFE-specific IgE, IL-4, IL-5, and IL-13 in an AD-like mouse model. The expression of RIP1 was increased in ear skin tissue from a DFE-induced mouse model with AD-like skin lesions and in the lesional skin of AD patients with high house dust mite sensitization. The expression of IL-33 was down-regulated after RIP1 inhibition, and the levels of IL-33 were increased by over-expression of RIP1 in keratinocytes stimulated with DFE. Nectostatin-1 reduced IL-33 expression in vitro and in the DFE-induced mouse model. These results suggest that RIP1 can be one of the mediators that regulate IL-33-mediated atopic skin inflammation by house dust mites.
Collapse
Affiliation(s)
- Meiling Jin
- Department of Pharmacology, School of Medicine, Yanbian National University, Yanji 133002, China
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jin Seon Bang
- Department of Dermatology, School of Medicine, Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Dae-Lyong Ha
- Department of Dermatology, School of Medicine, Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jun Young Kim
- Department of Dermatology, School of Medicine, Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Kyung Duck Park
- Department of Dermatology, School of Medicine, Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Weon Ju Lee
- Department of Dermatology, School of Medicine, Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Seok-Jong Lee
- Department of Dermatology, School of Medicine, Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Jin Kyeong Choi
- Department of Immunology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Young-Ae Choi
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Yong Hyun Jang
- Department of Dermatology, School of Medicine, Bio-Medical Research Institute, Kyungpook National University, Daegu 41944, Republic of Korea
- Correspondence: (Y.H.J.); (S.-H.K.); Tel.: +82-53-200-5838 (Y.H.J.); +82-53-420-4838 (S.-H.K.)
| | - Sang-Hyun Kim
- Department of Pharmacology, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Correspondence: (Y.H.J.); (S.-H.K.); Tel.: +82-53-200-5838 (Y.H.J.); +82-53-420-4838 (S.-H.K.)
| |
Collapse
|
26
|
Solé C, Domingo S, Penzo E, Moliné T, Porres L, Aparicio G, Ferrer B, Cortés-Hernández J. Downregulation of miR-885-5p Promotes NF-κB Pathway Activation and Immune Recruitment in Cutaneous Lupus Erythematosus. J Invest Dermatol 2023; 143:209-219.e13. [PMID: 36049539 DOI: 10.1016/j.jid.2022.08.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/21/2022] [Accepted: 08/01/2022] [Indexed: 01/25/2023]
Abstract
Cutaneous lupus erythematosus (CLE) has a specific microRNA expression profile. MiR-885-5p has been found to be downregulated in the epidermis of CLE lesions; however, its biological role in the disease has not been studied. In this study, we show that miR-885-5p is markedly reduced in CLE keratinocytes (KCs) with IFN-α and UVB being strong miR-885-5p regulators in vitro. Microarray expression profiling of anti‒miR-885-5p‒transfected KCs identified PSMB5 as a direct target. Specific inhibition of miR-885-5p increased epidermal proliferation by modulating keratin 16 gene K16, BIRC5, TP63, and CDK4 proliferative genes and promoted NF-κB signaling pathway in human primary KCs by increasing IκBα degradation. Silencing PSMB5 rescued the effect of miR-885-5p inhibition, indicating that miR-885-5p regulates proliferation and NF-κB activation by targeting PSMB5 in KCs. In addition, inhibition of miR-885-5p increased the ability of KCs to attract leukocytes in a PSMB5-independent manner. We identified TRAF1 as another direct target, and its silencing reduced leukocyte migration. Collectively, our findings suggest that UVB and IFN-ɑ downregulate miR-885-5p in CLE KCs, leading to epidermal inflammation by NF-κB activity enhancement and proliferation through PSMB5 and immune recruitment through TRAF1. Our data indicate that miR-885-5p is a potential therapeutic target in CLE.
Collapse
Affiliation(s)
- Cristina Solé
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain.
| | - Sandra Domingo
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Eleonora Penzo
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Teresa Moliné
- Department of Pathology, Vall d'Hebrón University Hospital, Barcelona, Spain
| | - Laura Porres
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Gloria Aparicio
- Department of Dermatology, Vall d'Hebrón University Hospital, Barcelona, Spain
| | - Berta Ferrer
- Department of Pathology, Vall d'Hebrón University Hospital, Barcelona, Spain
| | - Josefina Cortés-Hernández
- Rheumatology Research Group - Lupus Unit, Vall d'Hebrón University Hospital, Vall d'Hebrón Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
27
|
Bhat A, Irizar H, Couch ACM, Raval P, Duarte RRR, Dutan Polit L, Hanger B, Powell T, Deans PJM, Shum C, Nagy R, McAlonan G, Iyegbe CO, Price J, Bramon E, Bhattacharyya S, Vernon AC, Srivastava DP. Attenuated transcriptional response to pro-inflammatory cytokines in schizophrenia hiPSC-derived neural progenitor cells. Brain Behav Immun 2022; 105:82-97. [PMID: 35716830 PMCID: PMC9810540 DOI: 10.1016/j.bbi.2022.06.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 05/29/2022] [Accepted: 06/13/2022] [Indexed: 01/07/2023] Open
Abstract
Maternal immune activation (MIA) during prenatal development is an environmental risk factor for psychiatric disorders including schizophrenia (SZ). Converging lines of evidence from human and animal model studies suggest that elevated cytokine levels in the maternal and fetal compartments are an important indication of the mechanisms driving this association. However, there is variability in susceptibility to the psychiatric risk conferred by MIA, likely influenced by genetic factors. How MIA interacts with a genetic profile susceptible to SZ is challenging to test in animal models. To address this gap, we examined whether differential gene expression responses occur in forebrain-lineage neural progenitor cells (NPCs) derived from human induced pluripotent stem cells (hiPSC) generated from three individuals with a diagnosis of schizophrenia and three healthy controls. Following acute (24 h) treatment with either interferon-gamma (IFNγ; 25 ng/μl) or interleukin (IL)-1β (10 ng/μl), we identified, by RNA sequencing, 3380 differentially expressed genes (DEGs) in the IFNγ-treated control lines (compared to untreated controls), and 1980 DEGs in IFNγ-treated SZ lines (compared to untreated SZ lines). Out of 4137 genes that responded significantly to IFNγ across all lines, 1223 were common to both SZ and control lines. The 2914 genes that appeared to respond differentially to IFNγ treatment in SZ lines were subjected to a further test of significance (multiple testing correction applied to the interaction effect between IFNγ treatment and SZ diagnosis), yielding 359 genes that passed the significance threshold. There were no differentially expressed genes in the IL-1β-treatment conditions after Benjamini-Hochberg correction. Gene set enrichment analysis however showed that IL-1β impacts immune function and neuronal differentiation. Overall, our data suggest that a) SZ NPCs show an attenuated transcriptional response to IFNγ treatment compared to controls; b) Due to low IL-1β receptor expression in NPCs, NPC cultures appear to be less responsive to IL-1β than IFNγ; and c) the genes differentially regulated in SZ lines - in the face of a cytokine challenge - are primarily associated with mitochondrial, "loss-of-function", pre- and post-synaptic gene sets. Our findings particularly highlight the role of early synaptic development in the association between maternal immune activation and schizophrenia risk.
Collapse
Affiliation(s)
- Anjali Bhat
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK; Division of Psychiatry, University College London, London, UK; Wellcome Centre for Human Neuroimaging, University College London, London, UK
| | - Haritz Irizar
- Division of Psychiatry, University College London, London, UK; Icahn School of Medicine, Mount Sinai Hospital, NY, USA
| | - Amalie C M Couch
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Pooja Raval
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Rodrigo R R Duarte
- Department of Social, Genetic & Developmental Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Medicine, Weill Cornell Medical College, Cornell University, NY, USA
| | - Lucia Dutan Polit
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Bjorn Hanger
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Timothy Powell
- Department of Social, Genetic & Developmental Psychiatry, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; Department of Medicine, Weill Cornell Medical College, Cornell University, NY, USA
| | - P J Michael Deans
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Carole Shum
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Roland Nagy
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Grainne McAlonan
- MRC Centre for Neurodevelopmental Disorders, King's College London, UK; Department of Forensic and Neurodevelopmental Sciences, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Conrad O Iyegbe
- Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | - Jack Price
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK
| | - Elvira Bramon
- Division of Psychiatry, University College London, London, UK; Department of Psychosis Studies, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, United Kingdom
| | | | - Anthony C Vernon
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK.
| | - Deepak P Srivastava
- Department of Basic & Clinical Neuroscience, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, UK.
| |
Collapse
|
28
|
Salik D, El Kaderi Y, Hans C, Lefort A, Libert F, Smits G. Comparative study of keratinocyte primary culture methods from pediatric skin biopsies for
RNA
‐sequencing. Exp Dermatol 2022; 31:1741-1747. [DOI: 10.1111/exd.14652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 07/02/2022] [Accepted: 07/21/2022] [Indexed: 11/28/2022]
Affiliation(s)
- D. Salik
- Department of Dermatology, CHU Saint‐Pierre, CHU Brugmann and Hôpital Universitaire des Enfants Reine Fabiola Université Libre de Bruxelles Brussels Belgium
| | - Y. El Kaderi
- Department of Dermatology, CHU Saint‐Pierre, CHU Brugmann and Hôpital Universitaire des Enfants Reine Fabiola Université Libre de Bruxelles Brussels Belgium
| | - C. Hans
- Cytogenetics Laboratory, Hôpital Erasme, ULB Center of Human Genetics Université Libre de Bruxelles (ULB) Brussels Belgium
| | - A. Lefort
- I.R.I.B.H.M, Campus Erasme Université Libre de Bruxelles 808 Route de Lennik, B‐1070 Brussels Belgium
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore), Campus Erasme Université Libre de Bruxelles 808 Route de Lennik, B‐1070 Brussels Belgium
| | - F. Libert
- I.R.I.B.H.M, Campus Erasme Université Libre de Bruxelles 808 Route de Lennik, B‐1070 Brussels Belgium
- Brussels Interuniversity Genomics High Throughput core (BRIGHTcore), Campus Erasme Université Libre de Bruxelles 808 Route de Lennik, B‐1070 Brussels Belgium
| | - G. Smits
- Department of Genetics, Hôpital Erasme, ULB Center of Human Genetics Université Libre de Bruxelles (ULB) Brussels Belgium
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics Université Libre de Bruxelles (ULB) Brussels Belgium
- Interuniversity Institute of Bioinformatics in Brussels Université Libre de Bruxelles Brussels Belgium
| |
Collapse
|
29
|
SerpinB7 deficiency contributes to development of psoriasis via calcium-mediated keratinocyte differentiation dysfunction. Cell Death Dis 2022; 13:635. [PMID: 35864103 PMCID: PMC9304369 DOI: 10.1038/s41419-022-05045-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Revised: 06/17/2022] [Accepted: 06/27/2022] [Indexed: 01/21/2023]
Abstract
Defective execution of proteases and protease inhibitors that mediate abnormal signaling cascades is emerging as a key contributor to skin diseases, such as psoriasis. SerpinB7 is identified as a skin-specific endogenous protease inhibitor, but the role and underlying mechanism in psoriasis are poorly understood. Here we found that SerpinB7 is highly expressed in psoriatic keratinocytes of patients and imiquimod-induced psoriatic lesions in mice. SerpinB7-/- mice showed abnormal epidermal barrier integrity and skin architecture in homeostasis, and aggravated psoriatic lesion with inhibiting terminal differentiation and increasing inflammatory cells infiltration compared to SerpinB7+/+ mice after Imiquimod treatment. Mechanistically, SerpinB7 deficiency results in excessive proliferation and impaired differentiation, as well as increased chemokines and antimicrobial peptide expression in normal human epidermal keratinocyte and mouse primary keratinocyte. Transcriptomics and proteomics results showed that the SeprinB7 deficiency affected keratinocyte differentiation and proinflammatory cytokines, possibly by affecting the calcium ion channel-related proteins. Notably, we demonstrated that SerpinB7 deficiency prevented the increase in intracellular Ca2+ influx, which was partly eliminated by the intracellular Ca2+ chelator BAPTA-AM. Our findings first described the critical role of SerpinB7 in the regulation of keratinocyte differentiation and psoriatic microenvironment mediated via keratinocytes' intracellular calcium flux, proposing a new candidate for therapeutic targets in psoriasis.
Collapse
|
30
|
Generating iPSCs with a High-Efficient, Non-Invasive Method-An Improved Way to Cultivate Keratinocytes from Plucked Hair for Reprogramming. Cells 2022; 11:cells11121955. [PMID: 35741085 PMCID: PMC9222083 DOI: 10.3390/cells11121955] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 06/01/2022] [Accepted: 06/14/2022] [Indexed: 11/16/2022] Open
Abstract
Various somatic cell types are suitable for induced pluripotency reprogramming, such as dermal fibroblasts, mesenchymal stem cells or hair keratinocytes. Harvesting primary epithelial keratinocytes from plucked human hair follicles (HFs) represents an easy and non-invasive alternative to a fibroblast culture from invasive skin biopsies. Nevertheless, to facilitate and simplify the process, which can be divided into three main steps (collecting, culturing and reprogramming), the whole procedure of generating hair keratinocytes has to be revised and upgraded continuously. In this study, we address advancements and approaches which improve the generation and handling of primary HF-derived keratinocytes tremendously, e.g., for iPSCs reprogramming. We not only evaluated different serum- and animal-origin-free media, but also supplements and coating solutions for an enhanced protocol. Here, we demonstrate the importance of speed and accuracy in the collecting step, as well as the choice of the right transportation medium. Our results lead to a more defined approach that further increases the reliability of downstream experiments and inter-laboratory reproducibility. These improvements will make it possible to obtain keratinocytes from plucked human hair for the generation of donor-specific iPSCs easier and more efficient than ever before, whilst preserving a non-invasive capability.
Collapse
|
31
|
Bailly A, Milhavet O, Lemaitre JM. RNA-Based Strategies for Cell Reprogramming toward Pluripotency. Pharmaceutics 2022; 14:317. [PMID: 35214051 PMCID: PMC8876983 DOI: 10.3390/pharmaceutics14020317] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/16/2022] [Accepted: 01/25/2022] [Indexed: 02/04/2023] Open
Abstract
Cell therapy approaches to treat a wide range of pathologies have greatly benefited from cell reprogramming techniques that allow the conversion of a somatic cell into a pluripotent cell. Many technological developments have been made since the initial major discovery of this biological process. Recently reprogramming methods based on the use of RNA have emerged and seem very promising. Thus, in this review we will focus on presenting the interest of such methods for cell reprogramming but also how these RNA-based strategies can be extended to eventually lead to medical applications to improve healthspan and longevity.
Collapse
Affiliation(s)
- Anaëlle Bailly
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- INGRAALYS, SA, IRMB, Incubator Cyborg, 34295 Montpellier, France
| | - Ollivier Milhavet
- IRMB, University Montpellier, INSERM, CNRS, 34295 Montpellier, France
- SAFE-iPSC Facility, CHU Montpellier, 34295 Montpellier, France
| | - Jean-Marc Lemaitre
- IRMB, University Montpellier, INSERM, 34295 Montpellier, France
- SAFE-iPSC Facility, CHU Montpellier, 34295 Montpellier, France
| |
Collapse
|
32
|
Shu F, Gao H, Wu W, Yu S, Zhang L, Liu H, Xiao S, Xia Z, Zheng Y. Amniotic epithelial cells accelerate diabetic wound healing by protecting keratinocytes and fibroblasts from high‐glucose‐induced senescence. Cell Biol Int 2022; 46:755-770. [PMID: 35077602 DOI: 10.1002/cbin.11771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/02/2022] [Accepted: 01/16/2022] [Indexed: 11/05/2022]
Affiliation(s)
- Futing Shu
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| | - Haojie Gao
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| | - Wenfeng Wu
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| | - Shaoshuo Yu
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| | - Lianjie Zhang
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
- Department of Burns and Plastic surgeryHongkou District of the Changhai Hospital, Naval Medical UniversityShanghai200081China
| | - Huazhen Liu
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| | - Shichu Xiao
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| | - Zhaofan Xia
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| | - Yongjun Zheng
- Department of Burn Surgerythe First Affiliated Hospital of Naval Medical UniversityShanghai200433China
| |
Collapse
|
33
|
Frese L, Darwiche SE, Gunning ME, Hoerstrup SP, von Rechenberg B, Giovanoli P, Calcagni M. Optimizing large-scale autologous human keratinocyte sheets for major burns-Toward an animal-free production and a more accessible clinical application. Health Sci Rep 2022; 5:e449. [PMID: 35028432 PMCID: PMC8738975 DOI: 10.1002/hsr2.449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 10/18/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
Background and Aims Autologous keratinocyte sheets constitute an important component of the burn wound treatment toolbox available to a surgeon and can be considered a life‐saving procedure for patients with severe burns over 50% of their total body surface area. Large‐scale keratinocyte sheet cultivation still fundamentally relies on the use of animal components such as inactivated murine 3T3 fibroblasts as feeders, animal‐derived enzymes such as trypsin, as well as media components such as fetal bovine serum (FBS). This study was therefore aimed to optimize autologous keratinocyte sheets by comparing various alternatives to critical components in their production. Methods Human skin samples were retrieved from remnant operative tissues. Cell isolation efficiency and viability were investigated by comparing the efficacy of porcine‐derived trypsin and animal‐free enzymes (Accutase and TrypLESelect). The subsequent expansion of the cells and the keratinocyte sheet formation was analyzed, comparing various cell culture substrates (inactivated murine 3T3 fibroblasts, inactivated human fibroblasts, Collagen I or plain tissue culture plastic), as well as media containing serum or chemically defined animal‐free media. Results The cell isolation step showed clear cell yield advantages when using porcine‐derived trypsin, compared to animal‐free alternatives. The keratinocyte sheets produced using animal‐free serum were similar to those produced using 3T3 feeder layer and FBS‐containing medium, particularly in mechanical integrity as all grafts were liftable. In addition, sheets grown on collagen in an animal‐free medium showed indications of advantages in homogeneity, speed, reduced variability, and differentiation status compared to the other growth conditions investigated. Most importantly, the procedure was compatible with the up‐scaling requirements of major burn wound treatments. Conclusion This study demonstrated that animal‐free components could be used successfully to reduce the risk profile of large‐scale autologous keratinocyte sheet production, and thereby increase clinical accessibility.
Collapse
Affiliation(s)
- Laura Frese
- Institute for Regenerative Medicine (IREM) University of Zurich Zurich Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM) University of Zurich Zurich Switzerland.,La Colline Sion Switzerland
| | - Salim Elias Darwiche
- Center for Applied Biotechnology and Molecular Medicine (CABMM) University of Zurich Zurich Switzerland.,Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty University of Zurich Zurich Switzerland
| | - Myrna Elisabeth Gunning
- Department of Plastic and Reconstructive Surgery University Hospital Zurich Zurich Switzerland
| | - Simon Philipp Hoerstrup
- Institute for Regenerative Medicine (IREM) University of Zurich Zurich Switzerland.,Center for Applied Biotechnology and Molecular Medicine (CABMM) University of Zurich Zurich Switzerland
| | - Brigitte von Rechenberg
- Center for Applied Biotechnology and Molecular Medicine (CABMM) University of Zurich Zurich Switzerland.,Musculoskeletal Research Unit (MSRU), Vetsuisse Faculty University of Zurich Zurich Switzerland
| | - Pietro Giovanoli
- Department of Plastic and Reconstructive Surgery University Hospital Zurich Zurich Switzerland
| | - Maurizio Calcagni
- Center for Applied Biotechnology and Molecular Medicine (CABMM) University of Zurich Zurich Switzerland.,Department of Plastic and Reconstructive Surgery University Hospital Zurich Zurich Switzerland
| |
Collapse
|
34
|
van Ee A, Kim D, Prizmic V, Rho H, Park Y, Evans B, Kim S, Lee S, Wang G, Yu J, Kane MA, Garza LA. CD14 Is Induced by Retinoic Acid and Is Required for Double Stranded Noncoding RNA-Induced Regeneration. J Invest Dermatol 2022; 142:2291-2294.e7. [PMID: 34999109 PMCID: PMC9259758 DOI: 10.1016/j.jid.2021.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/07/2021] [Accepted: 12/10/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Amy van Ee
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Dongwon Kim
- Department of Bio-Chemical Engineering, Dongseo University, Busan, Republic of Korea
| | - Vicky Prizmic
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Hyunyoung Rho
- Department of Bio-Chemical Engineering, Dongseo University, Busan, Republic of Korea
| | - Yukyung Park
- Department of Bio-Chemical Engineering, Dongseo University, Busan, Republic of Korea
| | - Benjamin Evans
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sooah Kim
- Department of Environment Science and Biotechnology, Jeonju University, Jeonju, Republic of Korea
| | - Sam Lee
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Gaofeng Wang
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA; Department of Plastic and Aesthetic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jianshi Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Maureen A Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Luis A Garza
- Department of Dermatology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
35
|
Yde Ohki CM, McNeill RV, Nieberler M, Radtke F, Kittel-Schneider S, Grünblatt E. Promising Developments in the Use of Induced Pluripotent Stem Cells in Research of ADHD. Curr Top Behav Neurosci 2022; 57:483-501. [PMID: 35543866 DOI: 10.1007/7854_2022_346] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Although research using animal models, peripheral and clinical biomarkers, multimodal neuroimaging techniques and (epi)genetic information has advanced our understanding of Attention-Deficit Hyperactivity Disorder (ADHD), the aetiopathology of this neurodevelopmental disorder has still not been elucidated. Moreover, as the primary affected tissue is the brain, access to samples is problematic. Alternative models are therefore required, facilitating cellular and molecular analysis. Recent developments in stem cell research have introduced the possibility to reprogram somatic cells from patients, in this case ADHD, and healthy controls back into their pluripotent state, meaning that they can then be differentiated into any cell or tissue type. The potential to translate patients' somatic cells into stem cells, and thereafter to use 2- and 3-dimensional (2D and 3D) neuronal cells to model neurodevelopmental disorders and/or test novel drug therapeutics, is discussed in this chapter.
Collapse
Affiliation(s)
- Cristine Marie Yde Ohki
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zürich, Switzerland
| | - Rhiannon V McNeill
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Matthias Nieberler
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Franziska Radtke
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, University Hospital Wuerzburg, Würzburg, Germany
| | - Sarah Kittel-Schneider
- Department of Psychiatry, Psychosomatic Medicine and Psychotherapy, University Hospital Wuerzburg, Wuerzburg, Germany
| | - Edna Grünblatt
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich (PUK), University of Zurich, Zürich, Switzerland.
- Neuroscience Center Zurich, University of Zurich and the ETH Zurich, Zürich, Switzerland.
- Zurich Center for Integrative Human Physiology, University of Zurich, Zürich, Switzerland.
| |
Collapse
|
36
|
Casalia ML, Casabona JC, García C, Cavaliere Candedo V, Quintá HR, Farías MI, Gonzalez J, Gonzalez Morón D, Córdoba M, Consalvo D, Mostoslavsky G, Urbano FJ, Pasquini J, Murer MG, Rela L, Kauffman MA, Pitossi FJ. A familiar study on self-limited childhood epilepsy patients using hIPSC-derived neurons shows a bias towards immaturity at the morphological, electrophysiological and gene expression levels. Stem Cell Res Ther 2021; 12:590. [PMID: 34823607 PMCID: PMC8620942 DOI: 10.1186/s13287-021-02658-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 10/31/2021] [Indexed: 12/28/2022] Open
Abstract
Background Self-limited Childhood Epilepsies are the most prevalent epileptic syndrome in children. Its pathogenesis is unknown. In this disease, symptoms resolve spontaneously in approximately 50% of patients when maturity is reached, prompting to a maturation problem. The purpose of this study was to understand the molecular bases of this disease by generating and analyzing induced pluripotent stem cell-derived neurons from a family with 7 siblings, among whom 4 suffer from this disease.
Methods Two affected siblings and, as controls, a healthy sister and the unaffected mother of the family were studied. Using exome sequencing, a homozygous variant in the FYVE, RhoGEF and PH Domain Containing 6 gene was identified in the patients as a putative genetic factor that could contribute to the development of this familial disorder. After informed consent was signed, skin biopsies from the 4 individuals were collected, fibroblasts were derived and reprogrammed and neurons were generated and characterized by markers and electrophysiology. Morphological, electrophysiological and gene expression analyses were performed on these neurons. Results Bona fide induced pluripotent stem cells and derived neurons could be generated in all cases. Overall, there were no major shifts in neuronal marker expression among patient and control-derived neurons. Compared to two familial controls, neurons from patients showed shorter axonal length, a dramatic reduction in synapsin-1 levels and cytoskeleton disorganization. In addition, neurons from patients developed a lower action potential threshold with time of in vitro differentiation and the amount of current needed to elicit an action potential (rheobase) was smaller in cells recorded from NE derived from patients at 12 weeks of differentiation when compared with shorter times in culture. These results indicate an increased excitability in patient cells that emerges with the time in culture. Finally, functional genomic analysis showed a biased towards immaturity in patient-derived neurons. Conclusions We are reporting the first in vitro model of self-limited childhood epilepsy, providing the cellular bases for future in-depth studies to understand its pathogenesis. Our results show patient-specific neuronal features reflecting immaturity, in resonance with the course of the disease and previous imaging studies. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02658-2.
Collapse
Affiliation(s)
| | | | - Corina García
- Institute Leloir Foundation- IIBBA-CONICET, Buenos Aires, Argentina
| | | | - Héctor Ramiro Quintá
- CONICET and Laboratorio de Medicina Experimental "Dr. J Toblli", Hospital Alemán, Buenos Aires, Argentina
| | | | - Joaquín Gonzalez
- Institute Leloir Foundation- IIBBA-CONICET, Buenos Aires, Argentina
| | - Dolores Gonzalez Morón
- Consultorio y Laboratorio de Neurogenética, Centro Universitario de Neurología "José María Ramos Mejía" Facultad de Medicina, UBA & Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina
| | - Marta Córdoba
- Consultorio y Laboratorio de Neurogenética, Centro Universitario de Neurología "José María Ramos Mejía" Facultad de Medicina, UBA & Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina
| | - Damian Consalvo
- Consultorio y Laboratorio de Neurogenética, Centro Universitario de Neurología "José María Ramos Mejía" Facultad de Medicina, UBA & Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina
| | - Gustavo Mostoslavsky
- Center For Regenerative Medicine (CReM) of Boston University and Boston Medical Center, Boston, USA
| | - Francisco J Urbano
- Department of Physiology, Molecular and Cellular Biology "Dr. Héctor Maldonado", Faculty of Exact and Natural Sciences, University of Buenos Aires, IFIBYNE-CONICET, Buenos Aires, Argentina
| | - Juana Pasquini
- Faculty of Pharmacy and Biochemistry, University of Buenos Aires, Buenos Aires, Argentina
| | - Mario Gustavo Murer
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina.,Universidad de Buenos Aires - CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO), Buenos Aires, Argentina
| | - Lorena Rela
- Universidad de Buenos Aires, Facultad de Medicina, Departamento de Ciencias Fisiológicas, Grupo de Neurociencia de Sistemas, Buenos Aires, Argentina.,Universidad de Buenos Aires - CONICET, Instituto de Fisiología y Biofísica Bernardo Houssay (IFIBIO), Buenos Aires, Argentina
| | - Marcelo A Kauffman
- Consultorio y Laboratorio de Neurogenética, Centro Universitario de Neurología "José María Ramos Mejía" Facultad de Medicina, UBA & Instituto de Investigaciones en Medicina Traslacional, Facultad de Ciencias Biomédicas, Universidad Austral-CONICET, Buenos Aires, Argentina.
| | | |
Collapse
|
37
|
Moniz T, Lima SAC, Reis S. Protocol for the Isolation of Stratum Corneum from Pig Ear Skin: Evaluation of the Trypsin Digestion Conditions. Methods Protoc 2021; 4:mps4040080. [PMID: 34842773 PMCID: PMC8628882 DOI: 10.3390/mps4040080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/01/2021] [Accepted: 11/01/2021] [Indexed: 11/22/2022] Open
Abstract
Stratum corneum (SC) represents the outermost layer of the skin, being an effective barrier against the entry of molecules and pathogens. Skin research has given particular focus to SC as it hampers effective drug delivery for cosmetical and therapeutical purposes. Following recommendations to develop alternative models to animals, the SC isolated from skin obtained from medical procedures or from pigs has gained extensive attention. Yet, there is still missing a standard and simple procedure accepted within the scientific community to avoid application of different isolated SC methodologies, a fact that may hamper progress in skin research. Considering this challenge, the present study evaluated different experimental conditions aiming to establish a useful and sustainable solvent-free procedure for the obtention of a realistic SC model. The studied trypsin digestion parameters included concentration, incubation period and temperature. Isolated SC was characterized using histological analysis and calcein’s permeability, after the procedure and during a 6-week storage period. Data recommend trypsin digestion at 4 °C for 20 h as the most effective procedure to isolate SC from pig ear skin. This work contributes to standardize the SC isolation procedure, and to obtain a valuable and reliable SC mimetic model for skin drug development.
Collapse
|
38
|
Sleiman Y, Lacampagne A, Meli AC. "Ryanopathies" and RyR2 dysfunctions: can we further decipher them using in vitro human disease models? Cell Death Dis 2021; 12:1041. [PMID: 34725342 PMCID: PMC8560800 DOI: 10.1038/s41419-021-04337-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 10/08/2021] [Accepted: 10/14/2021] [Indexed: 12/23/2022]
Abstract
The regulation of intracellular calcium (Ca2+) homeostasis is fundamental to maintain normal functions in many cell types. The ryanodine receptor (RyR), the largest intracellular calcium release channel located on the sarco/endoplasmic reticulum (SR/ER), plays a key role in the intracellular Ca2+ handling. Abnormal type 2 ryanodine receptor (RyR2) function, associated to mutations (ryanopathies) or pathological remodeling, has been reported, not only in cardiac diseases, but also in neuronal and pancreatic disorders. While animal models and in vitro studies provided valuable contributions to our knowledge on RyR2 dysfunctions, the human cell models derived from patients’ cells offer new hope for improving our understanding of human clinical diseases and enrich the development of great medical advances. We here discuss the current knowledge on RyR2 dysfunctions associated with mutations and post-translational remodeling. We then reviewed the novel human cellular technologies allowing the correlation of patient’s genome with their cellular environment and providing approaches for personalized RyR-targeted therapeutics.
Collapse
Affiliation(s)
- Yvonne Sleiman
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Alain Lacampagne
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France
| | - Albano C Meli
- PhyMedExp, University of Montpellier, INSERM, CNRS, Montpellier, France.
| |
Collapse
|
39
|
Ray A, Joshi JM, Sundaravadivelu PK, Raina K, Lenka N, Kaveeshwar V, Thummer RP. An Overview on Promising Somatic Cell Sources Utilized for the Efficient Generation of Induced Pluripotent Stem Cells. Stem Cell Rev Rep 2021; 17:1954-1974. [PMID: 34100193 DOI: 10.1007/s12015-021-10200-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/02/2021] [Indexed: 01/19/2023]
Abstract
Human induced Pluripotent Stem Cells (iPSCs) have enormous potential in understanding developmental biology, disease modeling, drug discovery, and regenerative medicine. The initial human iPSC studies used fibroblasts as a starting cell source to reprogram them; however, it has been identified to be a less appealing somatic cell source by numerous studies due to various reasons. One of the important criteria to achieve efficient reprogramming is determining an appropriate starting somatic cell type to induce pluripotency since the cellular source has a major influence on the reprogramming efficiency, kinetics, and quality of iPSCs. Therefore, numerous groups have explored various somatic cell sources to identify the promising sources for reprogramming into iPSCs with different reprogramming factor combinations. This review provides an overview of promising easily accessible somatic cell sources isolated in non-invasive or minimally invasive manner such as keratinocytes, urine cells, and peripheral blood mononuclear cells used for the generation of human iPSCs derived from healthy and diseased subjects. Notably, iPSCs generated from one of these cell types derived from the patient will offer ethical and clinical advantages. In addition, these promising somatic cell sources have the potential to efficiently generate bona fide iPSCs with improved reprogramming efficiency and faster kinetics. This knowledge will help in establishing strategies for safe and efficient reprogramming and the generation of patient-specific iPSCs from these cell types.
Collapse
Affiliation(s)
- Arnab Ray
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Jahnavy Madhukar Joshi
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India
| | - Pradeep Kumar Sundaravadivelu
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Khyati Raina
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India
| | - Nibedita Lenka
- National Centre for Cell Science, S. P. Pune University Campus, Pune - 411007, Ganeshkhind, Maharashtra, India
| | - Vishwas Kaveeshwar
- Central Research Laboratory, SDM College of Medical Sciences and Hospital, Shri Dharmasthala Manjunatheshwara University, Dharwad, 580009, Karnataka, India.
| | - Rajkumar P Thummer
- Laboratory for Stem Cell Engineering and Regenerative Medicine, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 781039, Assam, India.
| |
Collapse
|
40
|
Harada N, Kataoka M, Nakanosho M, Uyama H. Penetration of Singlet Oxygen into Films with Oxygen Permeability Coefficient Close to that of Skin. Photochem Photobiol 2021; 97:971-979. [PMID: 33973245 DOI: 10.1111/php.13446] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 05/05/2021] [Indexed: 12/30/2022]
Abstract
Although its antiviral and antibacterial functions help prevent infection, singlet oxygen (1 O2 )-which is generated by the action of light on an endogenous photosensitizer-is cytotoxic. In the present study, we investigated the ability of 1 O2 -generated by the action of visible light on a photosensitizer-to penetrate skin. We used two polymer films with oxygen permeability coefficients similar to that of skin-i.e. cellulose acetate (CA) and ethyl cellulose (EC). Both films contained 1,3-diphenylisobenzofuran (DPBF), which was used as an 1 O2 probe. 1 O2 generated externally did not permeate the films by mere contact. Therefore, we conclude that the potential for 1 O2 to penetrate the skin is very low, and films that generate 1 O2 are safe and useful for preventing infections by contact. We also proved that 1 O2 can move between the layers of integrated polymer films when they are joined together.
Collapse
Affiliation(s)
- Nobuyuki Harada
- Nippon Shokubai Research Alliance Laboratories, Osaka University, Osaka, Japan
| | - Mika Kataoka
- Nippon Shokubai Research Alliance Laboratories, Osaka University, Osaka, Japan
| | | | - Hiroshi Uyama
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Osaka, Japan
| |
Collapse
|
41
|
Spontaneous Cell Detachment and Reattachment in Cancer Cell Lines: An In Vitro Model of Metastasis and Malignancy. Int J Mol Sci 2021; 22:ijms22094929. [PMID: 34066490 PMCID: PMC8124513 DOI: 10.3390/ijms22094929] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 11/16/2022] Open
Abstract
There is an unmet need for simplified in vitro models of malignancy and metastasis that facilitate fast, affordable and scalable gene and compound analysis. "Adherent" cancer cell lines frequently release "free-floating" cells into suspension that are viable and can reattach. This, in a simplistic way, mimics the metastatic process. We compared the gene expression profiles of naturally co-existing populations of floating and adherent cells in SW620 (colon), C33a (cervix) and HeLa (cervix) cancer cells. We found that 1227, 1367 and 1333 genes were at least 2-fold differentially expressed in the respective cell lines, of which 122 were shared among the three cell lines. As proof of principle, we focused on the anti-metastatic gene NM23-H1, which was downregulated both at the RNA and protein level in the floating cell populations of all three cell lines. Knockdown of NM23-H1 significantly increased the number of floating (and viable) cells, whereas overexpression of NM23-H1 significantly reduced the proportion of floating cells. Other potential regulators of these cellular states were identified through pathway analysis, including hypoxia, mTOR (mechanistic target of rapamycin), cell adhesion and cell polarity signal transduction pathways. Hypoxia, a condition linked to malignancy and metastasis, reduced NM23-H1 expression and significantly increased the number of free-floating cells. Inhibition of mTOR or Rho-associated protein kinase (ROCK) significantly increased cell death specifically in the floating and not the adherent cell population. In conclusion, our study suggests that dynamic subpopulations of free-floating and adherent cells is a useful model to screen and identify genes, drugs and pathways that regulate the process of cancer metastasis, such as cell detachment and anoikis.
Collapse
|
42
|
Orvain C, Lin YL, Jean-Louis F, Hocini H, Hersant B, Bennasser Y, Ortonne N, Hotz C, Wolkenstein P, Boniotto M, Tisserand P, Lefebvre C, Lelièvre JD, Benkirane M, Pasero P, Lévy Y, Hüe S. Hair follicle stem cell replication stress drives IFI16/STING-dependent inflammation in hidradenitis suppurativa. J Clin Invest 2021; 130:3777-3790. [PMID: 32240121 DOI: 10.1172/jci131180] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 03/31/2020] [Indexed: 12/28/2022] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic, relapsing, inflammatory skin disease. HS appears to be a primary abnormality in the pilosebaceous-apocrine unit. In this work, we characterized hair follicle stem cells (HFSCs) isolated from HS patients and more precisely the outer root sheath cells (ORSCs). We showed that hair follicle cells from HS patients had an increased number of proliferating progenitor cells and lost quiescent stem cells. Remarkably, we also showed that the progression of replication forks was altered in ORSCs from hair follicles of HS patients, leading to activation of the ATR/CHK1 pathway. These alterations were associated with an increased number of micronuclei and with the presence of cytoplasmic ssDNA, leading to the activation of the IFI16/STING pathway and the production of type I IFNs. This mechanistic analysis of the etiology of HS in the HFSC compartment establishes a formal link between genetic predisposition and skin inflammation observed in HS.
Collapse
Affiliation(s)
- Cindy Orvain
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Yea-Lih Lin
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Francette Jean-Louis
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Hakim Hocini
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Barbara Hersant
- Service de chirurgie plastique et reconstructive.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Yamina Bennasser
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Nicolas Ortonne
- Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'anatomopathologie
| | - Claire Hotz
- Service de chirurgie plastique et reconstructive.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service de Dermatologie
| | - Pierre Wolkenstein
- Service de chirurgie plastique et reconstructive.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service de Dermatologie
| | - Michele Boniotto
- INSERM U955, Equipe 16, Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Pascaline Tisserand
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Cécile Lefebvre
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France
| | - Jean-Daniel Lelièvre
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'Immunologie Clinique, and
| | - Monsef Benkirane
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Philippe Pasero
- Equipe Labellisée Ligue contre le Cancer, Institut de Génétique Humaine, CNRS, Université de Montpellier, Montpellier France
| | - Yves Lévy
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'Immunologie Clinique, and
| | - Sophie Hüe
- INSERM U955, Equipe 16, Créteil, France.,Vaccine Research Institute (VRI), Université Paris Est Créteil, Faculté de Médecine, Créteil, France.,Groupe Hospitalier Henri Mondor, Assistance publique - Hôpitaux de Paris (AP-HP), Créteil, France.,Faculté de Médecine, Université Paris-Est Créteil (UPEC), Créteil, France.,Service d'Immunologie Biologique, Groupe Hospitalier Henri Mondor, AP-HP, Créteil, France
| |
Collapse
|
43
|
Culturing of Melanocytes from the Equine Hair Follicle Outer Root Sheath. Processes (Basel) 2021. [DOI: 10.3390/pr9010177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Hair follicles harbor a heterogeneous regenerative cell pool and represent a putative low-to-non-invasively available source of stem cells. We previously reported a technology for culturing human melanocytes from the hair follicle outer root sheath (ORS) for autologous pigmentation of tissue engineered skin equivalents. This study translated the ORS technology to horses. We de-veloped a culture of equine melanocytes from the ORS (eMORS) from equine forelock hair follicles cultured by means of an analogue human hair follicle-based in vitro methodology. The procedure was adjusted to equine physiology by addition of equine serum to the culture medium. The hair follicles were isolated by macerating forelock skin rests, enzymatically digested and subjected to air-medium-interface cultivation method. The procedure resulted in differentiated equine melanocytes, which exhibited typical morphology, presence of melanosomes, expression of cytoskeleton proteins vimentin, α-SMA, Sox2, S100ß and tyrosinase as well as tyrosinase activity followed by production of melanin. According to all assessed parameters, eMORS could be ranked as partially melanotic melanocytes. The results of the study offer an experimental base for further insight into hair follicle biology in equine and for comparative studies of hair follicles across different species.
Collapse
|
44
|
Epidermal progenitors suppress GRHL3-mediated differentiation through intronic polyadenylation promoted by CPSF-HNRNPA3 collaboration. Nat Commun 2021; 12:448. [PMID: 33469008 PMCID: PMC7815847 DOI: 10.1038/s41467-020-20674-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 12/11/2020] [Indexed: 01/29/2023] Open
Abstract
In self-renewing somatic tissue such as skin epidermis, terminal differentiation genes must be suppressed in progenitors to sustain regenerative capacity. Here we show that hundreds of intronic polyadenylation (IpA) sites are differentially used during keratinocyte differentiation, which is accompanied by downregulation of the Cleavage and Polyadenylation Specificity Factor (CPSF) complex. Sustained CPSF expression in undifferentiated keratinocytes requires the contribution from the transcription factor MYC. In keratinocytes cultured in undifferentiation condition, CSPF knockdown induces premature differentiation and partially affects dynamically used IpA sites. These sites include an IpA site located in the first intron of the differentiation activator GRHL3. CRISPR knockout of GRHL3 IpA increased full-length GRHL3 mRNA expression. Using a targeted genetic screen, we identify that HNRNPA3 interacts with CPSF and enhances GRHL3 IpA. Our data suggest a model where the interaction between CPSF and RNA-binding proteins, such as HNRNPA3, promotes site-specific IpA and suppresses premature differentiation in progenitors.
Collapse
|
45
|
Abreu CM, Pirraco RP, Reis RL, Cerqueira MT, Marques AP. Interfollicular epidermal stem-like cells for the recreation of the hair follicle epithelial compartment. Stem Cell Res Ther 2021; 12:62. [PMID: 33451331 PMCID: PMC7811263 DOI: 10.1186/s13287-020-02104-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 12/14/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Hair follicle (HF) development and growth are dependent on epithelial-mesenchymal interactions (EMIs). Dermal papilla (DP) cells are recognized as the key inductive mesenchymal player, but the ideal source of receptive keratinocytes for human HF regeneration is yet to be defined. We herein investigated whether human interfollicular epidermal keratinocytes with stem-like features (EpSlKCs), characterized by a α6bri/CD71dim expression, can replace human hair follicular keratinocytes (HHFKCs) for the recreation of the HF epithelium and respective EMIs. METHODS The α6bri/CD71dim cellular fraction was selected from the whole interfollicular keratinocyte population through fluorescence-activated cell sorting and directly compared with follicular keratinocytes in terms of their proliferative capacity and phenotype. The crosstalk with DP cells was studied in an indirect co-culture system, and EpSlKC hair forming capacity tested in a hair reconstitution assay when combined with DP cells. RESULTS EpSlKCs exhibited a phenotypic profile similar to follicular keratinocytes and were capable of increasing DP cell proliferation and, for short co-culture times, the number of alkaline phosphatase-active cells, suggesting an improvement of their inductivity. Moreover, the recreation of immature HFs and sebaceous glands was observed after EpSlKC and DP cell co-grafting in nude mice. CONCLUSIONS Our results suggest that EpSlKCs are akin to follicular keratinocytes and can crosstalk with DP cells, contributing to HF morphogenesis in vivo, thus representing an attractive epithelial cell source for hair regeneration strategies.
Collapse
Affiliation(s)
- Carla M Abreu
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rogério P Pirraco
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Mariana T Cerqueira
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Alexandra P Marques
- 3B's Research Group - Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Guimarães, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
46
|
Glycocalyx disruption enhances motility, proliferation and collagen synthesis in diabetic fibroblasts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:118955. [PMID: 33421533 DOI: 10.1016/j.bbamcr.2021.118955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 11/22/2022]
Abstract
Impaired wound healing represents one of the most debilitating side effects of Diabetes mellitus. Though the role of fibroblasts in wound healing is well-known, the extent to which their function is altered in the context of diabetes remains incompletely understood. Here, we address this question by comparing the phenotypes of healthy dermal fibroblasts (HDFs) and diabetic dermal fibroblasts (DDFs). We show that DDFs are more elongated but less motile and less contractile than HDFs. Reduced motility of DDFs is attributed to formation of larger focal adhesions stabilized by a bulky glycocalyx, associated with increased expression of the cell surface glycoprotein mucin 16 (MUC 16). Disruption of the glycocalyx not only restored DDF motility to levels comparable to that of HDFs, but also led to increased proliferation and collagen synthesis. Collectively, our results illustrate the influence of glycocalyx disruption on mechanics of diabetic fibroblasts relevant to cell motility.
Collapse
|
47
|
Localisation and regulation of cholesterol transporters in the human hair follicle: mapping changes across the hair cycle. Histochem Cell Biol 2021; 155:529-545. [PMID: 33404706 PMCID: PMC8134313 DOI: 10.1007/s00418-020-01957-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Cholesterol has long been suspected of influencing hair biology, with dysregulated homeostasis implicated in several disorders of hair growth and cycling. Cholesterol transport proteins play a vital role in the control of cellular cholesterol levels and compartmentalisation. This research aimed to determine the cellular localisation, transport capability and regulatory control of cholesterol transport proteins across the hair cycle. Immunofluorescence microscopy in human hair follicle sections revealed differential expression of ATP-binding cassette (ABC) transporters across the hair cycle. Cholesterol transporter expression (ABCA1, ABCG1, ABCA5 and SCARB1) reduced as hair follicles transitioned from growth to regression. Staining for free cholesterol (filipin) revealed prominent cholesterol striations within the basement membrane of the hair bulb. Liver X receptor agonism demonstrated active regulation of ABCA1 and ABCG1, but not ABCA5 or SCARB1 in human hair follicles and primary keratinocytes. These results demonstrate the capacity of human hair follicles for cholesterol transport and trafficking. Future studies examining the role of cholesterol transport across the hair cycle may shed light on the role of lipid homeostasis in human hair disorders.
Collapse
|
48
|
Wang X, Wang S, Guo B, Su Y, Tan Z, Chang M, Diao J, Zhao Y, Wang Y. Human primary epidermal organoids enable modeling of dermatophyte infections. Cell Death Dis 2021; 12:35. [PMID: 33414472 PMCID: PMC7790817 DOI: 10.1038/s41419-020-03330-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 11/15/2020] [Accepted: 11/17/2020] [Indexed: 12/12/2022]
Abstract
Technology of generating human epidermal derivatives with physiological relevance to in vivo epidermis is continuously investigated for improving their effects on modeling of human natural dermatological status in basic and clinical studies. Here, we report a method of robust establishment and expansion of human primary epidermal organoids (hPEOs) under a chemically defined condition. hPEOs reconstruct morphological, molecular, and functional features of human epidermis and can expand for 6 weeks. Remarkably, hPEOs are permissive for dermatophyte infections caused by Trichophyton Rubrum (T. rubrum). The T. rubrum infections on hPEOs reflect many aspects of known clinical pathological reactions and reveal that the repression on IL-1 signaling may contribute to chronic and recurrent infections with the slight inflammation caused by T. rubrum in human skin. Thus, our present study provides a new insight into the pathogenesis of T. rubrum infections and indicates that hPEOs are a potential ex vivo model for both basic studies of skin diseases and clinical studies of testing potential antifungal drugs.
Collapse
Affiliation(s)
- Xuan Wang
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Shuyong Wang
- Army Tuberculosis Prevention and Control Key Laboratory, Beijing Key Laboratory of New Techniques of Tuberculosis Diagnosis and Treatment, Institute for Tuberculosis Research, the 8th Medical Center of Chinese PLA General Hospital, Beijing, 100091, China
| | - Baolin Guo
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Yuxin Su
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Zuolong Tan
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Mingyang Chang
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China
| | - Jinmei Diao
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China
| | - Yi Zhao
- Department of Dermatology, Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Yunfang Wang
- Translational Medicine Research Center, Beijing Tsinghua Chang Gung Hospital, Beijing, 102218, China.
- Department of Stem Cell and Regenerative Medicine, Beijing Institute of Health Service and Transfusion Medicine, Beijing, 100850, China.
| |
Collapse
|
49
|
Wang Z, Wang Y, Bradbury N, Gonzales Bravo C, Schnabl B, Di Nardo A. Skin wound closure delay in metabolic syndrome correlates with SCF deficiency in keratinocytes. Sci Rep 2020; 10:21732. [PMID: 33303806 PMCID: PMC7728784 DOI: 10.1038/s41598-020-78244-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/23/2020] [Indexed: 12/15/2022] Open
Abstract
Poor wound closure due to diabetes, aging, stress, obesity, alcoholism, and chronic disease affects millions of people worldwide. Reasons wounds will not close are still unclear, and current therapies are limited. Although stem cell factor (SCF), a cytokine, is known to be important for wound repair, the cellular and molecular mechanisms of SCF in wound closure remain poorly understood. Here, we found that SCF expression in the epidermis is decreased in mouse models of delayed wound closure intended to mimic old age, obesity, and alcoholism. By using SCF conditionally knocked out mice, we demonstrated that keratinocytes' autocrine production of SCF activates a transient c-kit receptor in keratinocytes. Transient activation of the c-kit receptor induces the expression of growth factors and chemokines to promote wound re-epithelialization by increasing migration of skin cells (keratinocytes and fibroblasts) and immune cells (neutrophils) to the wound bed 24-48 h post-wounding. Our results demonstrate that keratinocyte-produced SCF is essential to wound closure due to the increased recruitment of a unique combination of skin cells and immune cells in the early phase after wounding. This discovery is imperative for developing clinical strategies that might improve the body's natural repair mechanisms for treating patients with wound-closure pathologies.
Collapse
Affiliation(s)
- Zhenping Wang
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yanhan Wang
- Division of Gastroenterology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Nicholas Bradbury
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Carolina Gonzales Bravo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Bernd Schnabl
- Division of Gastroenterology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Anna Di Nardo
- Department of Dermatology, School of Medicine, University of California, San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
50
|
Bankoti K, Rameshbabu AP, Datta S, Goswami P, Roy M, Das D, Ghosh SK, Das AK, Mitra A, Pal S, Maulik D, Su B, Ghosh P, Basu B, Dhara S. Dual Functionalized Injectable Hybrid Extracellular Matrix Hydrogel for Burn Wounds. Biomacromolecules 2020; 22:514-533. [PMID: 33289564 DOI: 10.1021/acs.biomac.0c01400] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Low strength and rapid biodegradability of acellular dermal matrix (ADM) restrict its wider clinical application as a rapid cell delivery platform in situ for management of burn wounds. Herein, the extracted ADM was modified by a dual cross-linking approach with ionic crosslinking using chitosan and covalent cross-linking using an iodine-modified 2,5-dihydro-2,5-dimethoxy-furan cross-linker, termed as CsADM-Cl. In addition, inherent growth factors and cytokines were found to be preserved in CsADM-Cl, irrespective of ionic/covalent crosslinking. CsADM-Cl demonstrated improvement in post crosslinking stiffness with a decreased biodegradation rate. This hybrid crosslinked hydrogel supported adhesion, proliferation, and migration of human foreskin-derived fibroblasts and keratinocytes. Also, the angiogenic potential of CsADM-Cl was manifested by chick chorioallantoic membrane assay. CsADM-Cl showed excellent antibacterial activity against Escherichia coli and Staphylococcus aureus. Moreover, CsADM-Cl treated full thickness burn wounds and demonstrated rapid healing marked with superior angiogenesis, well-defined dermal-epidermal junctions, mature basket weave collagen deposition, and development of more pronounced secondary appendages. Altogether, the bioactive CsADM-Cl hydrogel established significant clinical potential to support wound healing as an apt injectable antibacterial matrix to encounter unmet challenges concerning critical burn wounds.
Collapse
Affiliation(s)
- Kamakshi Bankoti
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Arun Prabhu Rameshbabu
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Sayanti Datta
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Piyali Goswami
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Madhurima Roy
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Dipankar Das
- Polymer Chemistry Laboratory, Department of Applied Chemistry, Indian Institute of Technology (Indian School of Mines), Dhanbad 826004, India
| | - Sudip Kumar Ghosh
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Amit Kumar Das
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Analava Mitra
- Natural Products Research Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | - Sagar Pal
- Polymer Chemistry Laboratory, Department of Applied Chemistry, Indian Institute of Technology (Indian School of Mines), Dhanbad 826004, India
| | - Dhrubajyoti Maulik
- Department of Surgery, Bankura Sammilani Medical College, Bankura 722102, India
| | - Bo Su
- Bristol Dental School, University of Bristol, Bristol BS1 2LY, U.K
| | - Paulomi Ghosh
- Structural Biology and Bioinformatics, CSIR-Indian Institute of Chemical Biology (CSIR-IICB), 4, Raja S C Mullick Road, Kolkata 700032, India
| | - Bikramajit Basu
- Materials Research Center, Indian Institute of Science, Bangalore 560012, India
| | - Santanu Dhara
- Biomaterials and Tissue Engineering Laboratory, School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|