1
|
Muñoz-Torres JR, Garza-Veloz I, Velasco-Elizondo P, Martinez-Fierro ML. HEALS-A and GRADES: Novel Histological and Clinical Scales for Assessing Skin Regeneration in Murine Wound Healing Models. Diagnostics (Basel) 2025; 15:387. [PMID: 39941317 PMCID: PMC11816375 DOI: 10.3390/diagnostics15030387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Background: Wounds affect approximately 15 out of every 1000 individuals, representing a significant healthcare challenge. The preclinical evaluation of novel wound treatments is important for advancing therapies that promote effective skin regeneration and improve healing outcomes. Methods: In this study, we integrated existing knowledge from the literature on murine wound healing models, histological features of the skin, and clinical scores described in humans to propose two complementary assessment tools: the HEALS-A histological score (healing, epithelialization, angiogenesis, leukocytes, scar tissue, appendages) and the GRADES clinical score (granulation tissue, redness/edema, appearance of wound, devitalized tissue). Results: These scales combine real-time clinical observation with detailed histological analysis, providing a practical and comprehensive approach to assessing wound healing. Unlike existing wound assessing approaches, HEALS-A does not require specialized software and considers regenerated tissue structures, ensuring a broader and more-detailed evaluation. Conclusions: The assessment of wound closure over time, combined with clinical evaluation and histological analysis of skin, provides a comprehensive approach to determining the true impact of new treatments on skin regeneration and the recovery of its functions in wounds.
Collapse
Affiliation(s)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autonoma de Zacatecas, Zacatecas 98160, Mexico; (J.R.M.-T.); (P.V.-E.)
| | | | - Margarita L. Martinez-Fierro
- Molecular Medicine Laboratory, Academic Unit of Human Medicine and Health Sciences, Universidad Autonoma de Zacatecas, Zacatecas 98160, Mexico; (J.R.M.-T.); (P.V.-E.)
| |
Collapse
|
2
|
Mansor NI, Balqis TN, Lani MN, Lye KL, Nor Muhammad NA, Ismail WIW, Abidin SZ. Nature's Secret Neuro-Regeneration Pathway in Axolotls, Polychaetes and Planarians for Human Therapeutic Target Pathways. Int J Mol Sci 2024; 25:11904. [PMID: 39595973 PMCID: PMC11593954 DOI: 10.3390/ijms252211904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 11/28/2024] Open
Abstract
Despite significant improvements in the comprehension of neuro-regeneration, restoring nerve injury in humans continues to pose a substantial therapeutic difficulty. In the peripheral nervous system (PNS), the nerve regeneration process after injury relies on Schwann cells. These cells play a crucial role in regulating and releasing different extracellular matrix proteins, including laminin and fibronectin, which are essential for facilitating nerve regeneration. However, during regeneration, the nerve is required to regenerate for a long distance and, subsequently, loses its capacity to facilitate regeneration during this progression. Meanwhile, it has been noted that nerve regeneration has limited capabilities in the central nervous system (CNS) compared to in the PNS. The CNS contains factors that impede the regeneration of axons following injury to the axons. The presence of glial scar formation results from this unfavourable condition, where glial cells accumulate at the injury site, generating a physical and chemical barrier that hinders the regeneration of neurons. In contrast to humans, several species, such as axolotls, polychaetes, and planarians, possess the ability to regenerate their neural systems following amputation. This ability is based on the vast amount of pluripotent stem cells that have the remarkable capacity to differentiate and develop into any cell within their body. Although humans also possess these cells, their numbers are extremely limited. Examining the molecular pathways exhibited by these organisms has the potential to offer a foundational understanding of the human regeneration process. This review provides a concise overview of the molecular pathways involved in axolotl, polychaete, and planarian neuro-regeneration. It has the potential to offer a new perspective on therapeutic approaches for neuro-regeneration in humans.
Collapse
Affiliation(s)
- Nur Izzati Mansor
- Department of Nursing, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras 56000, Kuala Lumpur, Malaysia;
| | - Tengku Nabilatul Balqis
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
| | - Mohd Nizam Lani
- Faculty of Fisheries and Food Science, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia;
| | - Kwan Liang Lye
- ME Scientifique Sdn Bhd, Taman Universiti Indah, Seri Kembangan 43300, Selangor, Malaysia;
| | - Nor Azlan Nor Muhammad
- Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, Bangi 43600, Selangor, Malaysia;
| | - Wan Iryani Wan Ismail
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| | - Shahidee Zainal Abidin
- Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia; (T.N.B.); (W.I.W.I.)
- Research Interest Group Biological Security and Sustainability (BIOSES), Faculty of Science and Marine Environment, Universiti Malaysia Terengganu, Kuala Nerus 21030, Terengganu, Malaysia
| |
Collapse
|
3
|
Ferrarese L, Koch M, Baumann A, Bento-Lopes L, Wüst D, Berest I, Kopf M, Werner S. Inflammatory Mediators Suppress FGFR2 Expression in Human Keratinocytes to Promote Inflammation. Mol Cell Biol 2024; 44:489-504. [PMID: 39340759 PMCID: PMC11529413 DOI: 10.1080/10985549.2024.2399766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/30/2024] [Accepted: 08/20/2024] [Indexed: 09/30/2024] Open
Abstract
Fibroblast growth factors (FGFs) are key orchestrators of development, tissue homeostasis and repair. FGF receptor (FGFR) deficiency in mouse keratinocytes causes an inflammatory skin phenotype with similarities to atopic dermatitis, but the human relevance is unclear. Therefore, we generated human keratinocytes with a CRISPR/Cas9-induced knockout of FGFR2. Loss of this receptor promoted the expression of interferon-stimulated genes and pro-inflammatory cytokines under homeostatic conditions and in particular in response to different inflammatory mediators. Expression of FGFR2 itself was strongly downregulated in cultured human keratinocytes exposed to various pro-inflammatory stimuli. This is relevant in vivo, because bioinformatics analysis of bulk and single-cell RNA-seq data showed strongly reduced expression of FGFR2 in lesional skin of atopic dermatitis patients, which likely aggravates the inflammatory phenotype. These results reveal a key function of FGFR2 in human keratinocytes in the suppression of inflammation and suggest a role of FGFR2 downregulation in the pathogenesis of atopic dermatitis and possibly other inflammatory diseases.
Collapse
Affiliation(s)
- Luca Ferrarese
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Michael Koch
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Artemis Baumann
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Liliana Bento-Lopes
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Daria Wüst
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Ivan Berest
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Manfred Kopf
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zürich, Zürich, Switzerland
| |
Collapse
|
4
|
Peña OA, Martin P. Cellular and molecular mechanisms of skin wound healing. Nat Rev Mol Cell Biol 2024; 25:599-616. [PMID: 38528155 DOI: 10.1038/s41580-024-00715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/27/2024]
Abstract
Wound healing is a complex process that involves the coordinated actions of many different tissues and cell lineages. It requires tight orchestration of cell migration, proliferation, matrix deposition and remodelling, alongside inflammation and angiogenesis. Whereas small skin wounds heal in days, larger injuries resulting from trauma, acute illness or major surgery can take several weeks to heal, generally leaving behind a fibrotic scar that can impact tissue function. Development of therapeutics to prevent scarring and successfully repair chronic wounds requires a fuller knowledge of the cellular and molecular mechanisms driving wound healing. In this Review, we discuss the current understanding of the different phases of wound healing, from clot formation through re-epithelialization, angiogenesis and subsequent scar deposition. We highlight the contribution of different cell types to skin repair, with emphasis on how both innate and adaptive immune cells in the wound inflammatory response influence classically studied wound cell lineages, including keratinocytes, fibroblasts and endothelial cells, but also some of the less-studied cell lineages such as adipocytes, melanocytes and cutaneous nerves. Finally, we discuss newer approaches and research directions that have the potential to further our understanding of the mechanisms underpinning tissue repair.
Collapse
Affiliation(s)
- Oscar A Peña
- School of Biochemistry, University of Bristol, Bristol, UK.
| | - Paul Martin
- School of Biochemistry, University of Bristol, Bristol, UK.
| |
Collapse
|
5
|
Clark JF, Soriano P. Diverse Fgfr1 signaling pathways and endocytic trafficking regulate mesoderm development. Genes Dev 2024; 38:393-414. [PMID: 38834239 PMCID: PMC11216173 DOI: 10.1101/gad.351593.124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/21/2024] [Indexed: 06/06/2024]
Abstract
The fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1-null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identified processes regulating early mesoderm development by mechanisms involving both canonical and noncanonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.
Collapse
Affiliation(s)
- James F Clark
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
6
|
Clarke RM, Meier M, Wilson MJ. Genome-wide analysis of early vascular tunic repair and regeneration for Botrylloides digenesis reveals striking similarities to human wound healing. Dev Biol 2024; 509:28-42. [PMID: 38342399 DOI: 10.1016/j.ydbio.2024.02.003] [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: 06/19/2023] [Revised: 10/31/2023] [Accepted: 02/09/2024] [Indexed: 02/13/2024]
Abstract
The early stages of regeneration after injury are similar to those of wound healing. The ascidian Botrylloides diegensis can regenerate an entire adult from a small fragment of vascular tunic following the removal of all zooids in an injury-induced regeneration model. We investigated the molecular and cellular changes following injury to determine the differences between the healing process and the initiation of whole-body regeneration (WBR). We conducted transcriptome analysis at specific time points during regeneration and wound healing to identify differentially expressed genes (DEGs) and the unique biological processes associated with each state. Our findings revealed 296 DEGs at 10 h post-injury (hpi), with 71 highly expressed in healed tissue and 225 expressed during the WBR process. These DEGs were predicted to play roles in tissue reorganization, integrin signaling, extracellular matrix organization, and the innate immune system. Pathway analysis of the upregulated genes in the healed tunic indicated functional enrichment related to tissue repair, as has been observed in other species. Additionally, we examined the cell types in the tunic and ampullae in both tissue states using histology and in situ hybridization for six genes identified by transcriptome analysis. We observed strong mRNA expression in cells within the WBR tunic, and in small RNA-positive granules near the tunic edge. We hypothesized that many of these genes function in the compaction of the ampullae tunic, which is a pivotal process for WBR and dormancy in B. diegensis, and in an immune response. These findings establish surprising similarities between ascidian regeneration and human wound healing, emphasizing the potential for future investigations into human regenerative and repair mechanisms. This study provides valuable insights into the gene sets specifically activated during regeneration compared to wound healing, shedding light on the divergent activities of these processes.
Collapse
Affiliation(s)
- Rebecca M Clarke
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Michael Meier
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand
| | - Megan J Wilson
- Developmental Genomics Laboratory, Department of Anatomy, School of Biomedical Sciences, University of Otago, PO Box 56, Dunedin, 9054, New Zealand.
| |
Collapse
|
7
|
Kaelin CB, McGowan KA, Hutcherson AD, Delay JM, Li JH, Kiener S, Jagannathan V, Leeb T, Murphy WJ, Barsh GS. Ancestry dynamics and trait selection in a designer cat breed. Curr Biol 2024; 34:1506-1518.e7. [PMID: 38531359 PMCID: PMC11162505 DOI: 10.1016/j.cub.2024.02.075] [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: 07/31/2023] [Revised: 01/10/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024]
Abstract
The Bengal cat breed was developed from intercrosses between the Asian leopard cat, Prionailurus bengalensis, and the domestic cat, Felis catus, with a last common ancestor approximately 6 million years ago. Predicted to derive ∼94% of their genome from domestic cats, regions of the leopard cat genome are thought to account for the unique pelage traits and ornate color patterns of the Bengal breed, which are similar to those of ocelots and jaguars. We explore ancestry distribution and selection signatures in the Bengal breed by using reduced representation and whole-genome sequencing from 947 cats. The mean proportion of leopard cat DNA in the Bengal breed is 3.48%, lower than predicted from breed history, and is broadly distributed, covering 93% of the Bengal genome. Overall, leopard cat introgressions do not show strong signatures of selection across the Bengal breed. However, two popular color traits in Bengal cats, charcoal and pheomelanin intensity, are explained by selection of leopard cat genes whose expression is reduced in a domestic cat background, consistent with genetic incompatibility resulting from hybridization. We characterize several selective sweeps in the Bengal genome that harbor candidate genes for pelage and color pattern and that are associated with domestic, rather than leopard, cat haplotypes. We identify the molecular and phenotypic basis of one selective sweep as reduced expression of the Fgfr2 gene, which underlies glitter, a trait desired by breeders that affects hair texture and light reflectivity.
Collapse
Affiliation(s)
- Christopher B Kaelin
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kelly A McGowan
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - John M Delay
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA
| | | | - Sarah Kiener
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; Dermfocus, University of Bern, 3001 Bern, Switzerland
| | - Vidhya Jagannathan
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; Dermfocus, University of Bern, 3001 Bern, Switzerland
| | - Tosso Leeb
- Institute of Genetics, Vetsuisse Faculty, University of Bern, 3001 Bern, Switzerland; Dermfocus, University of Bern, 3001 Bern, Switzerland
| | - William J Murphy
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine, Texas A&M University, College Station, TX 77843, USA
| | - Gregory S Barsh
- HudsonAlpha Institute for Biotechnology, Huntsville, AL 35806, USA; Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
8
|
Clark JF, Soriano P. Diverse Fgfr1 signaling pathways and endocytic trafficking regulate early mesoderm development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.16.580629. [PMID: 38405698 PMCID: PMC10888970 DOI: 10.1101/2024.02.16.580629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
The Fibroblast growth factor (FGF) pathway is a conserved signaling pathway required for embryonic development. Activated FGF receptor 1 (FGFR1) drives multiple intracellular signaling cascade pathways, including ERK/MAPK and PI3K/AKT, collectively termed canonical signaling. However, unlike Fgfr1 null embryos, embryos containing hypomorphic mutations in Fgfr1 lacking the ability to activate canonical downstream signals are still able to develop to birth, but exhibit severe defects in all mesodermal-derived tissues. The introduction of an additional signaling mutation further reduces the activity of Fgfr1, leading to earlier lethality, reduced somitogenesis, and more severe changes in transcriptional outputs. Genes involved in migration, ECM-interaction, and phosphoinositol signaling were significantly downregulated, proteomic analysis identified changes in interactions with endocytic pathway components, and cells expressing mutant receptors show changes in endocytic trafficking. Together, we identify processes regulating early mesoderm development by mechanisms involving both canonical and non-canonical Fgfr1 pathways, including direct interaction with cell adhesion components and endocytic regulation.
Collapse
Affiliation(s)
- James F. Clark
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
9
|
Noble A, Qubrosi R, Cariba S, Favaro K, Payne SL. Neural dependency in wound healing and regeneration. Dev Dyn 2024; 253:181-203. [PMID: 37638700 DOI: 10.1002/dvdy.650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/29/2023] Open
Abstract
In response to injury, humans and many other mammals form a fibrous scar that lacks the structure and function of the original tissue, whereas other vertebrate species can spontaneously regenerate damaged tissues and structures. Peripheral nerves have been identified as essential mediators of wound healing and regeneration in both mammalian and nonmammalian systems, interacting with the milieu of cells and biochemical signals present in the post-injury microenvironment. This review examines the diverse functions of peripheral nerves in tissue repair and regeneration, specifically during the processes of wound healing, blastema formation, and organ repair. We compare available evidence in mammalian and nonmammalian models, identifying critical nerve-mediated mechanisms for regeneration and providing future perspectives toward integrating these mechanisms into a therapeutic framework to promote regeneration.
Collapse
Affiliation(s)
- Alexandra Noble
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Rozana Qubrosi
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Solsa Cariba
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Kayla Favaro
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Samantha L Payne
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
10
|
Arockiaraj AI, Johnson MA, Munir A, Ekambaram P, Lucas PC, McAllister-Lucas LM, Kemaladewi DU. CRISPRa-induced upregulation of human LAMA1 compensates for LAMA2-deficiency in Merosin-deficient congenital muscular dystrophy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.06.531347. [PMID: 36945402 PMCID: PMC10028808 DOI: 10.1101/2023.03.06.531347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/09/2023]
Abstract
Merosin-deficient congenital muscular dystrophy (MDC1A) is an autosomal recessive disorder caused by mutations in the LAMA2 gene, resulting in a defective form of the extracellular matrix protein laminin-α2 (LAMA2). Individuals diagnosed with MDC1A exhibit progressive muscle wasting and declining neuromuscular functions. No treatments for this disorder are currently available. We previously showed that postnatal Lama1 upregulation, achieved through CRISPR activation (CRISPRa), compensates for Lama2 deficiency and prevents neuromuscular pathophysiology in a mouse model of MDC1A. In this study, we assessed the feasibility of upregulating human LAMA1 as a potential therapeutic strategy for individuals with MDC1A, regardless of their mutations. We hypothesized that CRISPRa-mediated upregulation of human LAMA1 would compensate for the lack of LAMA2 and rescue cellular abnormalities in MDC1A fibroblasts. Global transcriptomic and pathway enrichment analyses of fibroblasts collected from individuals carrying pathogenic LAMA2 mutations, compared with healthy controls, indicated higher expression of transcripts encoding proteins that contribute to wound healing, including Transforming Growth Factor-β (TGF-β) and Fibroblast Growth Factor (FGF). These findings were supported by wound-healing assays indicating that MDC1A fibroblasts migrated significantly more rapidly than the controls. Subsequently, we treated the MDC1A fibroblasts with SadCas9-2XVP64 and sgRNAs targeting the LAMA1 promoter. We observed robust LAMA1 expression, which was accompanied by significant decreases in cell migration and expression of FGFR2, TGF-β2, and ACTA2, which are involved in the wound-healing mechanism in MDC1A fibroblasts. Collectively, our data suggest that CRISPRa-mediated LAMA1 upregulation may be a feasible mutation-independent therapeutic approach for MDC1A. This strategy might be adapted to address other neuromuscular diseases and inherited conditions in which strong compensatory mechanisms have been identified.
Collapse
Affiliation(s)
- Annie I. Arockiaraj
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Marie A. Johnson
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Anushe Munir
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Prasanna Ekambaram
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | - Peter C. Lucas
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, USA
| | | | - Dwi U. Kemaladewi
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, USA
| |
Collapse
|
11
|
Secretome of Adipose-Derived Stem Cells Cultured in Platelet Lysate Improves Migration and Viability of Keratinocytes. Int J Mol Sci 2023; 24:ijms24043522. [PMID: 36834932 PMCID: PMC9962933 DOI: 10.3390/ijms24043522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/05/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
Chronic wounds depict a silent epidemic challenging medical professionals worldwide. Regenerative medicine uses adipose-derived stem cells (ADSC) in promising new therapies. In this study, platelet lysate (PL) as a xenogen-free substitute for foetal bovine serum (FBS) in ADSC culture was used to create an ADSC secretome containing cytokines for optimal wound healing conditions. The ADSC secretome was tested on keratinocytes for migrational behaviour and viability. Therefore, human ADSC were characterized under FBS (10%) and PL (5% and 10%) substitution, regarding morphology, differentiation, viability, gene and protein expression. ADSC were then cultured in 5% PL and their secretome was used for stimulation of keratinocyte migration and viability. To enhance the effect, ADSC were treated with Epithelial Growth Factor (EGF, 100 ng/mL) and hypoxia (1% O₂). In both PL and FBS groups, ADSC expressed typical stem cell markers. PL induced a significantly higher increase in cell viability compared to FBS substitution. ADSC secretome contained various beneficial proteins which enhance the wound healing capacity of keratinocytes. This could be optimized treating ADSC with hypoxia and EGF. In conclusion, the study shows that ADSC cultivated in 5% PL can effectively support wound healing conditions and can be considered as a promising new therapy for individual treatment of chronic wound disorders.
Collapse
|
12
|
Morozov AM, Sergeev AN, Sungurova AV, Morozov DV, Belyak MA, Domracheva AS. Computer simulation of the wound process (review of literature). BULLETIN OF THE MEDICAL INSTITUTE "REAVIZ" (REHABILITATION, DOCTOR AND HEALTH) 2022. [DOI: 10.20340/vmi-rvz.2023.1.ictm.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Relevance. Computer simulation is a mathematical modeling process performed on a computer that is designed to predict the behavior or results of a real or physical system. Computer simulation has a number of advantages over classical models of animal experiments: the cheapness of the method (the need to acquire and maintain animals disappears by itself), the speed of obtaining results, the absence of bioethical problems, the ability to change the conditions of the experiment, etc.he purpose of this study is to review the methods of computer simulation of the wound process, to identify the shortcomings of the models and propose ways to solve them, as well as to select the best existing model for describing wound regeneration.Material and methods. In the course of this work, an analysis was made of foreign and domestic literature on the problem of computer modeling of the wound process.Results. After analyzing the relevant literature on this topic, the problem is seen precisely in the insufficiently studied process of wound regeneration, since many different cells, cytokines, growth factors, enzymes, fibrillar proteins, etc. take part in it. The models that currently exist describe wound regeneration only in an extremely generalized way, which does not allow us to apply them in clinical situations. Analyzing literature sources, we came to the conclusion that both numerical approaches, both cellular-biochemical (the first type of models) and phenomenological (the second type) are applicable in the case of wound modeling and can be used very successfully. The problem is that on the basis of one approach it is impossible to display a complete picture of wound healing, in this way it is possible to predict only individual regeneration parameters necessary for certain purposes due to the complexity and versatility of this typical pathophysiological process.Conclusion. Computer modeling of wounds is still a controversial and complex topic. Existing models are not intended to describe all the processes occurring in a healing wound. It is much more productive to describe the various phenomena during healing separately. This is due to the fact that many elements are involved in the regeneration of the skin, which are almost impossible to take into account in full. The available models are of exclusively scientific value, consisting in attempts to understand all complex processes and interactions. Practical application is difficult, since existing models require specific input data that require highly specialized equipment. If we abstract from all this, then the best existing model of the first type is the model of the authors Yangyang Wang, Christian F. Guerrero-Juarez, Yuchi Qiu and co-authors, in addition to it, any of the described phenomenological models will do.
Collapse
|
13
|
Maier M, Olthoff S, Hill K, Zosel C, Magauer T, Wein LA, Schaefer M. KS0365, a novel activator of the transient receptor potential vanilloid 3 (TRPV3) channel, accelerates keratinocyte migration. Br J Pharmacol 2022; 179:5290-5304. [PMID: 35916168 DOI: 10.1111/bph.15937] [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: 10/22/2021] [Revised: 06/09/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND AND PURPOSE Ca2+ signalling mediated by the thermosensitive, non-selective, Ca2+ -permeable transient receptor potential channel TRPV3 is assumed to play a critical role in regulating several aspects of skin functions, such as keratinocyte proliferation, differentiation, skin barrier formation and wound healing. Studying the function of TRPV3 in skin homeostasis, however, is still constrained by a lack of potent and selective pharmacological modulators of TRPV3. EXPERIMENTAL APPROACH By screening an in-house compound library using fluorometric intracellular Ca2+ assays, we identified two chemically related hits. The more potent and efficient TRPV3 activator KS0365 was further evaluated in fluo-4-assisted Ca2+ assays, different Ca2+ imaging approaches, electrophysiological studies, cytotoxicity and migration assays. KEY RESULTS KS0365 activated recombinant and native mouse TRPV3 more potently and with a higher efficacy compared to 2-APB and did not activate TRPV1, TRPV2 or TRPV4 channels. The activation of TRPV3 by KS0365 super-additively accelerated the EGF-induced keratinocyte migration, which was inhibited by the TRP channel blocker ruthenium red or by siRNA-mediated TRPV3 knockdown. Moreover, KS0365 induced strong Ca2+ responses in migrating front cells and in leading edges of keratinocytes. CONCLUSIONS AND IMPLICATIONS The selective TRPV3 activator KS0365 triggers increases in [Ca2+ ]i with most prominent signals in the leading edge, and accelerates migration of keratinocytes. TRPV3 activators may promote reepithelialization upon skin wounding.
Collapse
Affiliation(s)
- Marion Maier
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Stefan Olthoff
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Kerstin Hill
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Carolin Zosel
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| | - Thomas Magauer
- Leopold-Franzens-University Innsbruck, Institute of Organic Chemistry and Center for Molecular Biosciences, Innsbruck, Austria
| | - Lukas Anton Wein
- Leopold-Franzens-University Innsbruck, Institute of Organic Chemistry and Center for Molecular Biosciences, Innsbruck, Austria
| | - Michael Schaefer
- Leipzig University, Medical Faculty, Rudolf-Boehm-Institute of Pharmacology and Toxicology, Leipzig, Germany
| |
Collapse
|
14
|
Ornitz DM, Itoh N. New developments in the biology of fibroblast growth factors. WIREs Mech Dis 2022; 14:e1549. [PMID: 35142107 PMCID: PMC10115509 DOI: 10.1002/wsbm.1549] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/28/2023]
Abstract
The fibroblast growth factor (FGF) family is composed of 18 secreted signaling proteins consisting of canonical FGFs and endocrine FGFs that activate four receptor tyrosine kinases (FGFRs 1-4) and four intracellular proteins (intracellular FGFs or iFGFs) that primarily function to regulate the activity of voltage-gated sodium channels and other molecules. The canonical FGFs, endocrine FGFs, and iFGFs have been reviewed extensively by us and others. In this review, we briefly summarize past reviews and then focus on new developments in the FGF field since our last review in 2015. Some of the highlights in the past 6 years include the use of optogenetic tools, viral vectors, and inducible transgenes to experimentally modulate FGF signaling, the clinical use of small molecule FGFR inhibitors, an expanded understanding of endocrine FGF signaling, functions for FGF signaling in stem cell pluripotency and differentiation, roles for FGF signaling in tissue homeostasis and regeneration, a continuing elaboration of mechanisms of FGF signaling in development, and an expanding appreciation of roles for FGF signaling in neuropsychiatric diseases. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Neurological Diseases > Molecular and Cellular Physiology Congenital Diseases > Stem Cells and Development Cancer > Stem Cells and Development.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Nobuyuki Itoh
- Kyoto University Graduate School of Pharmaceutical Sciences, Sakyo, Kyoto, Japan
| |
Collapse
|
15
|
Xue Y, Reddy SK, Garza LA. Toward Understanding Wound Immunology for High-Fidelity Skin Regeneration. Cold Spring Harb Perspect Biol 2022; 14:a041241. [PMID: 35667792 PMCID: PMC9248820 DOI: 10.1101/cshperspect.a041241] [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] [Indexed: 11/24/2022]
Abstract
Effective tissue repair is vital for the survival of organisms. Yet, how the immune system coordinates with tissue stem cells (SCs) to effect postnatal tissue restoration remains elusive. This review presents current knowledge surrounding wound-induced SC and immune signaling that favors tissue repair, including wound healing and regeneration. We discuss factors that affect regenerative capacities among organisms and the dynamics of local immune cells and SCs during reepithelialization. We also present recent insights into how immune niches communicate with SCs or other body systems to restore the epithelial architecture. Additionally, we summarize our findings on functional wound regeneration, specifically how alarmin (double-stranded RNA [dsRNA])-activated Toll-like receptor signaling and host-microbe interaction-related immune pathways alter the regenerative property of skin SCs. Last, we touch on mechanisms by which known immunologic cellular and molecular signaling might boost the skin's regenerative property. Overall, this review will provide insights into how therapeutically modulating immune signaling could enhance postnatal tissue regeneration.
Collapse
Affiliation(s)
| | - Sashank K Reddy
- Department of Plastic and Reconstructive Surgery
- Department of Biomedical Engineering
- Institute for NanoBioTechnology
| | - Luis A Garza
- Department of Dermatology
- Department of Cell Biology
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland 21231, USA
| |
Collapse
|
16
|
Takaya K, Aramaki-Hattori N, Sakai S, Okabe K, Asou T, Kishi K. Fibroblast Growth Factor 7 Suppresses Fibrosis and Promotes Epithelialization during Wound Healing in Mouse Fetuses. Int J Mol Sci 2022; 23:ijms23137087. [PMID: 35806092 PMCID: PMC9266578 DOI: 10.3390/ijms23137087] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 02/07/2023] Open
Abstract
Adult mammalian wounds leave visible scars, whereas skin wounds in developing mouse fetuses are scarless until a certain point in development when complete regeneration occurs, including the structure of the dermis and skin appendages. Analysis of the molecular mechanisms at this transition will provide clues for achieving scarless wound healing. The fibroblast growth factor (FGF) family is a key regulator of inflammation and fibrosis during wound healing. We aimed to determine the expression and role of FGF family members in fetal wound healing. ICR mouse fetuses were surgically wounded at embryonic day 13 (E13), E15, and E17. Expression of FGF family members and FGF receptor (FGFR) in tissue samples from these fetuses was evaluated using in situ hybridization and reverse transcription-quantitative polymerase chain reaction. Fgfr1 was downregulated in E15 and E17 wounds, and its ligand Fgf7 was upregulated in E13 and downregulated in E15 and E17. Recombinant FGF7 administration in E15 wounds suppressed fibrosis and promoted epithelialization at the wound site. Therefore, the expression level of Fgf7 may correlate with scar formation in late mouse embryos, and external administration of FGF7 may represent a therapeutic option to suppress fibrosis and reduce scarring.
Collapse
Affiliation(s)
- Kento Takaya
- Correspondence: ; Tel.: +81-3-5363-3814; Fax: +81-3-3352-1054
| | | | | | | | | | | |
Collapse
|
17
|
Teplyakova O, Vinnik Y, Drobushevskaya A, Malinovskaya N, Kirichenko A, Ponedelnik D. Ozone improved the wound healing in type 2 diabetics via down-regulation of IL- 8, 10 and induction of FGFR expression. ACTA BIO-MEDICA : ATENEI PARMENSIS 2022; 93:e2022060. [PMID: 35546010 PMCID: PMC9171882 DOI: 10.23750/abm.v93i2.12291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 10/01/2021] [Indexed: 01/13/2023]
Abstract
Background and aim: We aimed to investigate the effect of ozonated autohaemotherapy (OA) on the wound healing, serum values of interleukin (IL) - 6, 8, 10, tumor necrosis factor-alpha (TNF-α), basic fibroblast growth factor (bFGF) and local expression of fibroblast growth factor receptors (FGFR) in type 2 diabetics with the acute soft-tissue infections. Methods: Patients in the first cohort (n-30) received a basic comprehensive treatment (BCT-group), and the second (n=28) also received OA (OA-group). Blood samples for ELISA and tissue specimens for the immunohistochemical examinations were collected at admission (day 0) and at the 9th day of inpatient treatment. Results: The additional using of OA has accelerated the timing of a single and the complete wound granulation and the timing to marginal epithelization, compared with the results of the standard treatment. The use of OA has significantly reduced the production of IL-8, 10 at 9th day. OA-group patients were characterized by consistently high levels of bFGF production in contrast to the BCT-group, where the decreasing in the serum bFGF level was observed. The maximum number of bFGFR - immunopositive labels was observed in OA-group out to 9th day (319,45 (249,90-348,43) versus baseline 192,65 (171,93-207,72), versus BCT-group 123,30 (105,23- 141,10), p<0,001). Conclusions: Application of OA in the complex treatment of the acute soft-tissue infections in diabetics makes it possible to achieve the significant reductions in the duration of the wound inflammation and regeneration phases by eliminating of overproduction of IL- 8, 10 and induction of expression of bFGF and its receptors. (www.actabiomedica.it)
Collapse
Affiliation(s)
- Olga Teplyakova
- Department of General Surgery named after Professor M. I. Gulman, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation.
| | - Yurii Vinnik
- Department of General Surgery named after Professor M. I. Gulman, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation.
| | - Anna Drobushevskaya
- Department of General Surgery named after Professor M. I. Gulman, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation; Center for collective use «Molecular & cell technologies», Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation .
| | - Natalia Malinovskaya
- Department of Biological Chemistry with the Course of Medical, Pharmaceutical and Toxicological Chemistry, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation; Research Institute of Molecular Medicine and Pathobiochemistry, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation; Center for collective use «Molecular & cell technologies», Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation.
| | - Andrey Kirichenko
- Department of Pathological Anatomy named after Professor P. G. Podzolkov, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation; Department of Pathological Anatomy, Clinical hospital «RZD-Medicine» city Krasnoyarsk, Krasnoyarsk, Russian Federation.
| | - Darya Ponedelnik
- Department of General Surgery named after Professor M. I. Gulman, Professor V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, Krasnoyarsk, Russian Federation.
| |
Collapse
|
18
|
Joanna S, Anna N, Drzewiecka K, Monika KM. miR-26a-5p and miR-125b-5p affect trophoblast genes and cell functions important during early pregnancy†. Biol Reprod 2022; 107:590-604. [PMID: 35416938 DOI: 10.1093/biolre/ioac071] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 01/18/2022] [Accepted: 04/01/2022] [Indexed: 11/14/2022] Open
Abstract
The most critical stage of pregnancy is embryo implantation, which relies on the synchronised developmental capacity of the embryo and uterine receptivity to implantation. In early pregnancy, conceptus and uterus release several factors enabling successful implantation and placentation. Molecules involved in embryo-maternal crosstalk include, but are not limited to, hormones, growth factors and cytokines. The discovery of microRNAs (miRNAs, small non-coding RNAs regulating gene expression) has revolutionized our understanding of many biological processes, including pregnancy. To date, numerous miRNAs have been detected in different species during pregnancy, both at the endometrial and embryonic sites. Thus, miRNAs are considered important regulators of early pregnancy events. Here, we report miR-26a-5p and miR-125b-5p effects on human and pig trophoblast cell function. Both miRNAs change the level of several genes and proteins important for proper embryo development. Moreover, miR-26a-5p stimulates porcine trophoblast proliferation and has a negative impact on its affinity to laminin. However, miR-125b-5p decreases porcine trophoblast cell migration. Our studies suggest that miR-26a-5p and miR-125b-5p can affect early pregnancy functions by regulating genes and processes important for proper conceptuses' development and progression through the implantation process.
Collapse
Affiliation(s)
- Szuszkiewicz Joanna
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
| | - Nitkiewicz Anna
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
| | - Klaudia Drzewiecka
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
| | - Kaczmarek M Monika
- Department of Hormonal Action Mechanisms, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland.,Molecular Biology Laboratory, Institute of Animal Reproduction and Food Research, Polish Academy of Sciences, Tuwima 10, 10-748 Olsztyn, Poland
| |
Collapse
|
19
|
Chen K, Rao Z, Dong S, Chen Y, Wang X, Luo Y, Gong F, Li X. Roles of the fibroblast growth factor signal transduction system in tissue injury repair. BURNS & TRAUMA 2022; 10:tkac005. [PMID: 35350443 PMCID: PMC8946634 DOI: 10.1093/burnst/tkac005] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Revised: 12/13/2021] [Indexed: 12/13/2022]
Abstract
Following injury, tissue autonomously initiates a complex repair process, resulting in either partial recovery or regeneration of tissue architecture and function in most organisms. Both the repair and regeneration processes are highly coordinated by a hierarchy of interplay among signal transduction pathways initiated by different growth factors, cytokines and other signaling molecules under normal conditions. However, under chronic traumatic or pathological conditions, the reparative or regenerative process of most tissues in different organs can lose control to different extents, leading to random, incomplete or even flawed cell and tissue reconstitution and thus often partial restoration of the original structure and function, accompanied by the development of fibrosis, scarring or even pathogenesis that could cause organ failure and death of the organism. Ample evidence suggests that the various combinatorial fibroblast growth factor (FGF) and receptor signal transduction systems play prominent roles in injury repair and the remodeling of adult tissues in addition to embryonic development and regulation of metabolic homeostasis. In this review, we attempt to provide a brief update on our current understanding of the roles, the underlying mechanisms and clinical application of FGFs in tissue injury repair.
Collapse
Affiliation(s)
| | | | - Siyang Dong
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
- Department of breast surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yajing Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xulan Wang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Yongde Luo
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Fanghua Gong
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| | - Xiaokun Li
- Correspondence. Xiaokun Li, ; Fanghua Gong, ; Yongde Luo,
| |
Collapse
|
20
|
Caballero-Solares A, Umasuthan N, Xue X, Katan T, Kumar S, Westcott JD, Chen Z, Fast MD, Skugor S, Taylor RG, Rise ML. Interacting Effects of Sea Louse (Lepeophtheirus salmonis) Infection and Formalin-Killed Aeromonas salmonicida on Atlantic Salmon Skin Transcriptome. Front Immunol 2022; 13:804987. [PMID: 35401509 PMCID: PMC8987027 DOI: 10.3389/fimmu.2022.804987] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Lepeophtheirus salmonis (sea lice) and bacterial co-infection threatens wild and farmed Atlantic salmon performance and welfare. In the present study, pre-adult L. salmonis-infected and non-infected salmon were intraperitoneally injected with either formalin-killed Aeromonas salmonicida bacterin (ASAL) or phosphate-buffered saline (PBS). Dorsal skin samples from each injection/infection group (PBS/no lice, PBS/lice, ASAL/no lice, and ASAL/lice) were collected at 24 h post-injection and used for transcriptome profiling using a 44K salmonid microarray platform. Microarray results showed no clear inflammation gene expression signatures and revealed extensive gene repression effects by pre-adult lice (2,189 down and 345 up-regulated probes) in the PBS-injected salmon (PBS/lice vs. PBS/no lice), which involved basic cellular (e.g., RNA and protein metabolism) processes. Lice repressive effects were not observed within the group of ASAL-injected salmon (ASAL/lice vs. ASAL/no lice); on the contrary, the observed skin transcriptome changes –albeit of lesser magnitude (82 up and 1 down-regulated probes)– suggested the activation in key immune and wound healing processes (e.g., neutrophil degranulation, keratinocyte differentiation). The molecular skin response to ASAL was more intense in the lice-infected (ASAL/lice vs. PBS/lice; 272 up and 11 down-regulated probes) than in the non-infected fish (ASAL/no lice vs. PBS/no lice; 27 up-regulated probes). Regardless of lice infection, the skin’s response to ASAL was characterized by the putative activation of both antibacterial and wound healing pathways. The transcriptomic changes prompted by ASAL+lice co-stimulation (ASAL/lice vs. PBS/no lice; 1878 up and 3120 down-regulated probes) confirmed partial mitigation of lice repressive effects on fundamental cellular processes and the activation of pathways involved in innate (e.g., neutrophil degranulation) and adaptive immunity (e.g., antibody formation), as well as endothelial cell migration. The qPCR analyses evidenced immune-relevant genes co-stimulated by ASAL and lice in an additive (e.g., mbl2b, bcl6) and synergistic (e.g., hampa, il4r) manner. These results provided insight on the physiological response of the skin of L. salmonis-infected salmon 24 h after ASAL stimulation, which revealed immunostimulatory properties by the bacterin with potential applications in anti-lice treatments for aquaculture. As a simulated co-infection model, the present study also serves as a source of candidate gene biomarkers for sea lice and bacterial co-infection.
Collapse
Affiliation(s)
- Albert Caballero-Solares
- Department of Ocean Sciences, Memorial University, St. John’s, NL, Canada
- *Correspondence: Albert Caballero-Solares,
| | | | - Xi Xue
- Department of Ocean Sciences, Memorial University, St. John’s, NL, Canada
| | - Tomer Katan
- Department of Ocean Sciences, Memorial University, St. John’s, NL, Canada
| | - Surendra Kumar
- Department of Ocean Sciences, Memorial University, St. John’s, NL, Canada
| | | | - Zhiyu Chen
- Department of Ocean Sciences, Memorial University, St. John’s, NL, Canada
- Fisheries and Marine Institute, Memorial University, St. John’s, NL, Canada
| | - Mark D. Fast
- Department of Pathology and Microbiology, Atlantic Veterinary College, University of Prince Edward Island, Charlottetown, PE, Canada
| | - Stanko Skugor
- Cargill Aqua Nutrition, Cargill, Sea Lice Research Center (SLRC), Sandnes, Norway
| | | | - Matthew L. Rise
- Department of Ocean Sciences, Memorial University, St. John’s, NL, Canada
| |
Collapse
|
21
|
Tsai YJ, Ma MC, Wu PH, Wu WB. Novel involvement of PLD-PKCδ-CREB axis in regulating FGF-2-mediated pentraxin 3 production in human nasal fibroblast cells. J Cell Physiol 2021; 237:1871-1887. [PMID: 34897684 DOI: 10.1002/jcp.30657] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 01/27/2023]
Abstract
A higher expression level of mitogenic fibroblast growth factor-2 (FGF-2) has been reported in human nasal mucus of both chronic rhinosinusitis (CRS) with nasal polyps (CRSwNP) and CRS without nasal polyps (CRSsNP). Meanwhile, we have shown that long pentraxin 3 (PTX3), an essential component of humoral innate immunity that is produced at sites of infection and inflammation, was overproduced in human nasal mucosae and secretions of CRSsNP. Therefore, this study was aimed to investigate how FGF-2 regulates PTX3 expression in human CRSsNP nasal mucosa-derived fibroblast cells (hNMDFs). The FGF-2 treatment caused ptx3 mRNA expression and PTX3 protein induction and secretion. In parallel, a differential expression of FGF receptor (FGFR)-1 to FGFR-4 was observed in hNMDFs and human nasal tissues. While conventionally known PI3K/Akt/mTOR and AP-1 pathways following FGFR activation were shown to be involved, the protein kinase Cδ (PKCδ) and cAMP response element-binding protein (CREB) were also found to be as critical signaling molecules in FGF-2-induced PTX3 induction. The PKCδ and CREB activation could be detected in total cells and in the cell nucleus. Accordingly, a novel CREB binding sequence was detected in the human ptx3 promoter region and could interact with activated CREB in cells challenged with FGF-2. Surprisingly, the phospholipase D (PLD), but not phosphoinositide- and phosphatidylcholine-phospholipase C, was necessarily required for PKCδ and CREB activation. Therefore, we demonstrated here for the first time that FGF-2 mediates PTX3 production not only through PI-3K/Akt/mTOR and AP-1 activation, but also through a novel FGFR-PLD-PKCδ-CREB cellular signaling pathway.
Collapse
Affiliation(s)
- Yih-Jeng Tsai
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.,Department of Otolaryngology Head and Neck Surgery, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ming-Chieh Ma
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Pi-Hui Wu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| | - Wen-Bin Wu
- School of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan.,Graduate Institute of Biomedical and Pharmaceutical Science, College of Medicine, Fu Jen Catholic University, New Taipei City, Taiwan
| |
Collapse
|
22
|
Merecz-Sadowska A, Sitarek P, Zajdel K, Kucharska E, Kowalczyk T, Zajdel R. The Modulatory Influence of Plant-Derived Compounds on Human Keratinocyte Function. Int J Mol Sci 2021; 22:12488. [PMID: 34830374 PMCID: PMC8618348 DOI: 10.3390/ijms222212488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/16/2021] [Accepted: 11/16/2021] [Indexed: 11/16/2022] Open
Abstract
The plant kingdom is a rich source of secondary metabolites with numerous properties, including the potential to modify keratinocyte biology. Keratinocytes are important epithelial cells that play a protective role against various chemical, physical and biological stimuli, and participate in reactive oxygen scavenging and inflammation and wound healing processes. The epidermal cell response may be modulated by phytochemicals via changes in signal transduction pathways. Plant extracts and single secondary compounds can possess a high antioxidant capacity and may suppress reactive oxygen species release, inhibit pro-apoptotic proteins and apoptosis and activate antioxidant enzymes in keratinocytes. Moreover, selected plant extracts and single compounds also exhibit anti-inflammatory properties and exposure may result in limited production of adhesion molecules, pro-inflammatory cytokines and chemokines in keratinocytes. In addition, plant extracts and single compounds may promote keratinocyte motility and proliferation via the regulation of growth factor production and enhance wound healing. While such plant compounds may modulate keratinocyte functions, further in vitro and in vivo studies are needed on their mechanisms of action, and more specific toxicity and clinical studies are needed to ensure their effectiveness and safety for use on human skin.
Collapse
Affiliation(s)
- Anna Merecz-Sadowska
- Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland;
| | - Przemysław Sitarek
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, 90-151 Lodz, Poland;
| | - Karolina Zajdel
- Department of Medical Informatics and Statistics, Medical University of Lodz, 90-645 Lodz, Poland;
| | - Ewa Kucharska
- Chair of Gerontology, Geriatrics and Social Work at the Faculty of Pedagogy, Ignatianum Academy in Cracow, 31-501 Cracow, Poland;
| | - Tomasz Kowalczyk
- Department of Molecular Biotechnology and Genetics, University of Lodz, 90-237 Lodz, Poland;
| | - Radosław Zajdel
- Department of Computer Science in Economics, University of Lodz, 90-214 Lodz, Poland;
| |
Collapse
|
23
|
Farooq M, Khan AW, Kim MS, Choi S. The Role of Fibroblast Growth Factor (FGF) Signaling in Tissue Repair and Regeneration. Cells 2021; 10:cells10113242. [PMID: 34831463 PMCID: PMC8622657 DOI: 10.3390/cells10113242] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2021] [Revised: 11/10/2021] [Accepted: 11/16/2021] [Indexed: 02/06/2023] Open
Abstract
Fibroblast growth factors (FGFs) are a large family of secretory molecules that act through tyrosine kinase receptors known as FGF receptors. They play crucial roles in a wide variety of cellular functions, including cell proliferation, survival, metabolism, morphogenesis, and differentiation, as well as in tissue repair and regeneration. The signaling pathways regulated by FGFs include RAS/mitogen-activated protein kinase (MAPK), phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K)–protein kinase B (AKT), phospholipase C gamma (PLCγ), and signal transducer and activator of transcription (STAT). To date, 22 FGFs have been discovered, involved in different functions in the body. Several FGFs directly or indirectly interfere with repair during tissue regeneration, in addition to their critical functions in the maintenance of pluripotency and dedifferentiation of stem cells. In this review, we summarize the roles of FGFs in diverse cellular processes and shed light on the importance of FGF signaling in mechanisms of tissue repair and regeneration.
Collapse
Affiliation(s)
- Mariya Farooq
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Abdul Waheed Khan
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Moon Suk Kim
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
| | - Sangdun Choi
- Department of Molecular Science and Technology, Ajou University, Suwon 16499, Korea; (M.F.); (A.W.K.); (M.S.K.)
- S&K Therapeutics, Ajou University Campus Plaza 418, 199 Worldcup-ro, Yeongtong-gu, Suwon 16502, Korea
- Correspondence:
| |
Collapse
|
24
|
Bock FJ, Sedov E, Koren E, Koessinger AL, Cloix C, Zerbst D, Athineos D, Anand J, Campbell KJ, Blyth K, Fuchs Y, Tait SWG. Apoptotic stress-induced FGF signalling promotes non-cell autonomous resistance to cell death. Nat Commun 2021; 12:6572. [PMID: 34772930 PMCID: PMC8590049 DOI: 10.1038/s41467-021-26613-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 10/08/2021] [Indexed: 11/13/2022] Open
Abstract
Damaged or superfluous cells are typically eliminated by apoptosis. Although apoptosis is a cell-autonomous process, apoptotic cells communicate with their environment in different ways. Here we describe a mechanism whereby cells under apoptotic stress can promote survival of neighbouring cells. We find that upon apoptotic stress, cells release the growth factor FGF2, leading to MEK-ERK-dependent transcriptional upregulation of pro-survival BCL-2 proteins in a non-cell autonomous manner. This transient upregulation of pro-survival BCL-2 proteins protects neighbouring cells from apoptosis. Accordingly, we find in certain cancer types a correlation between FGF-signalling, BCL-2 expression and worse prognosis. In vivo, upregulation of MCL-1 occurs in an FGF-dependent manner during skin repair, which regulates healing dynamics. Importantly, either co-treatment with FGF-receptor inhibitors or removal of apoptotic stress restores apoptotic sensitivity to cytotoxic therapy and delays wound healing. These data reveal a pathway by which cells under apoptotic stress can increase resistance to cell death in surrounding cells. Beyond mediating cytotoxic drug resistance, this process also provides a potential link between tissue damage and repair.
Collapse
Affiliation(s)
- Florian J Bock
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
- Department of Radiotherapy (MAASTRO), GROW-School for Oncology and Developmental Biology, Maastricht University, 6229 ER, Maastricht, The Netherlands.
| | - Egor Sedov
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Anna L Koessinger
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Catherine Cloix
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Désirée Zerbst
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Dimitris Athineos
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Jayanthi Anand
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Kirsteen J Campbell
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, 3200003, Israel
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK.
- Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1QH, UK.
| |
Collapse
|
25
|
Sasidharan V, Sánchez Alvarado A. The Diverse Manifestations of Regeneration and Why We Need to Study Them. Cold Spring Harb Perspect Biol 2021; 14:a040931. [PMID: 34750171 PMCID: PMC9438785 DOI: 10.1101/cshperspect.a040931] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
For hundreds of years, the question of why some organisms can regenerate missing body parts while others cannot has remained poorly understood. This has been due in great part to the inability to genetically, molecularly, and cellularly dissect this problem for most of the history of the field. It has only been in the past 20-30 years that important mechanistic advances have been made in methodologies that introduce loss and gain of gene function in animals that can regenerate. However, we still have a very incomplete understanding of how broadly regenerative abilities may be dispersed across species and whether or not such properties share a common evolutionary origin, which may have emerged independently or both. Understanding regeneration, therefore, will require rigorously practiced fundamental, curiosity-driven, discovery research. Expanding the number of research organisms used to study regeneration allows us to uncover aspects of this problem we may not yet know exist and simultaneously increases our chances of solving this long-standing problem of biology.
Collapse
|
26
|
Rauschendorfer T, Gurri S, Heggli I, Maddaluno L, Meyer M, Inglés-Prieto Á, Janovjak H, Werner S. Acute and chronic effects of a light-activated FGF receptor in keratinocytes in vitro and in mice. Life Sci Alliance 2021; 4:4/11/e202101100. [PMID: 34548382 PMCID: PMC8473723 DOI: 10.26508/lsa.202101100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 09/06/2021] [Accepted: 09/07/2021] [Indexed: 11/24/2022] Open
Abstract
Optogenetic activation of FGFR2 allowed temporally precise induction of signaling and behavioural changes, but counter-regulation at multiple levels prevented a sustained response in keratinocytes. FGFs and their high-affinity receptors (FGFRs) play key roles in development, tissue repair, and disease. Because FGFRs bind overlapping sets of ligands, their individual functions cannot be determined using ligand stimulation. Here, we generated a light-activated FGFR2 variant (OptoR2) to selectively activate signaling by the major FGFR in keratinocytes. Illumination of OptoR2-expressing HEK 293T cells activated FGFR signaling with remarkable temporal precision and promoted cell migration and proliferation. In murine and human keratinocytes, OptoR2 activation rapidly induced the classical FGFR signaling pathways and expression of FGF target genes. Surprisingly, multi-level counter-regulation occurred in keratinocytes in vitro and in transgenic mice in vivo, including OptoR2 down-regulation and loss of responsiveness to light activation. These results demonstrate unexpected cell type–specific limitations of optogenetic FGFRs in long-term in vitro and in vivo settings and highlight the complex consequences of transferring optogenetic cell signaling tools into their relevant cellular contexts.
Collapse
Affiliation(s)
- Theresa Rauschendorfer
- Department of Biology, Institute of Molecular Health Sciences, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Selina Gurri
- Department of Biology, Institute of Molecular Health Sciences, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Irina Heggli
- Department of Biology, Institute of Molecular Health Sciences, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Luigi Maddaluno
- Department of Biology, Institute of Molecular Health Sciences, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Michael Meyer
- Department of Biology, Institute of Molecular Health Sciences, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | | | - Harald Janovjak
- Institute of Science and Technology (IST) Austria, Klosterneuburg, Austria .,Australian Regenerative Medicine Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Australia.,European Molecular Biology Laboratory Australia, Monash University, Clayton, Australia
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| |
Collapse
|
27
|
Tang X, Hao M, Chang C, Bhatia A, O'Brien K, Chen M, Armstrong DG, Li W. Wound Healing Driver Gene and Therapeutic Development: Political and Scientific Hurdles. Adv Wound Care (New Rochelle) 2021; 10:415-435. [PMID: 32966158 PMCID: PMC8236301 DOI: 10.1089/wound.2019.1143] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 09/02/2020] [Indexed: 12/12/2022] Open
Abstract
Significance: Since the last Food and Drug Administration (FDA) approval of a wound healing therapeutic in 1997, no new therapeutic candidate (excluding physical therapies, devices, dressings, and antimicrobial agents) has advanced to clinical applications. During this period, the FDA drug approvals for tumors, which have been referred to as "wounds that do not heal," have reached a total of 284 (by end of 2018). Both political and scientific factors may explain this large discrepancy in drug approvals for the two seemingly related and equally complex pathophysiological conditions. Recent Advances: Using the current research funding ratio of 1:150 for wound healing to cancer and the 5% FDA drug approval rate for oncology, we reach a crude estimate of a 0.03% success rate for wound healing therapeutics. Unless a drastic improvement of the current situation, we express a pessimistic outlook toward new and effective wound healing drugs. Critical Issues: We argue that successful development of wound healing therapeutics will rely on identification of wound healing driver genes (WDGs), and the focus should be on WDGs for the wound closure phase of wound healing. Therefore, WDGs must be both necessary and sufficient for wound closure; the absence of a WDG disrupts wound closure, while its supplementation alone is sufficient to restore full wound closure. Successful translation of a WDG into therapeutics requires availability of well-defined animal models with a high degree of relevance to humans. This review discusses the main hurdles faced by the wound healing research community behind the development of so-called "rescuing drugs" for wound healing. Future Directions: Given the lack of new wound healing drugs for the past 23 years, there is a need for a wide range of fresh, innovative, and thorough debates on wound healing drug development, including an organized movement to raise public support for wound healing research.
Collapse
Affiliation(s)
- Xin Tang
- Department of Dermatology and The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Michelle Hao
- Department of Dermatology and The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Cheng Chang
- Department of Dermatology and The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Ayesha Bhatia
- Department of Dermatology and The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Kathrine O'Brien
- Department of Dermatology and The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Mei Chen
- Department of Dermatology and The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - David G. Armstrong
- Department of Surgery, University of Southern California Keck Medical Center, Los Angeles, California, USA
| | - Wei Li
- Department of Dermatology and The USC-Norris Comprehensive Cancer Center, University of Southern California Keck Medical Center, Los Angeles, California, USA
| |
Collapse
|
28
|
Prudovsky I. Cellular Mechanisms of FGF-Stimulated Tissue Repair. Cells 2021; 10:cells10071830. [PMID: 34360000 PMCID: PMC8304273 DOI: 10.3390/cells10071830] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/15/2021] [Accepted: 07/16/2021] [Indexed: 01/10/2023] Open
Abstract
Growth factors belonging to the FGF family play important roles in tissue and organ repair after trauma. In this review, I discuss the regulation by FGFs of the aspects of cellular behavior important for reparative processes. In particular, I focus on the FGF-dependent regulation of cell proliferation, cell stemness, de-differentiation, inflammation, angiogenesis, cell senescence, cell death, and the production of proteases. In addition, I review the available literature on the enhancement of FGF expression and secretion in damaged tissues resulting in the increased FGF supply required for tissue repair.
Collapse
Affiliation(s)
- Igor Prudovsky
- Maine Medical Center Research Institute, 81 Research Dr., Scarborough, ME 04074, USA
| |
Collapse
|
29
|
Guasto A, Cormier-Daire V. Signaling Pathways in Bone Development and Their Related Skeletal Dysplasia. Int J Mol Sci 2021; 22:4321. [PMID: 33919228 PMCID: PMC8122623 DOI: 10.3390/ijms22094321] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/19/2021] [Indexed: 12/19/2022] Open
Abstract
Bone development is a tightly regulated process. Several integrated signaling pathways including HH, PTHrP, WNT, NOTCH, TGF-β, BMP, FGF and the transcription factors SOX9, RUNX2 and OSX are essential for proper skeletal development. Misregulation of these signaling pathways can cause a large spectrum of congenital conditions categorized as skeletal dysplasia. Since the signaling pathways involved in skeletal dysplasia interact at multiple levels and have a different role depending on the time of action (early or late in chondrogenesis and osteoblastogenesis), it is still difficult to precisely explain the physiopathological mechanisms of skeletal disorders. However, in recent years, significant progress has been made in elucidating the mechanisms of these signaling pathways and genotype-phenotype correlations have helped to elucidate their role in skeletogenesis. Here, we review the principal signaling pathways involved in bone development and their associated skeletal dysplasia.
Collapse
Affiliation(s)
- Alessandra Guasto
- Imagine Institute, Université de Paris, Clinical Genetics, INSERM UMR 1163, Necker Enfants Malades Hospital, 75015 Paris, France;
| | - Valérie Cormier-Daire
- Imagine Institute, Université de Paris, Clinical Genetics, INSERM UMR 1163, Necker Enfants Malades Hospital, 75015 Paris, France;
- Centre de Référence Pour Les Maladies Osseuses Constitutionnelles, Service de Génétique Clinique, AP-HP, Hôpital Necker-Enfants Malades, 75015 Paris, France
| |
Collapse
|
30
|
Kim J, Lee KM, Han SH, Ko EA, Yoon DS, Park IK, Shin HC, Park KH, Lee JW. Development of stabilized dual growth factor-loaded hyaluronate collagen dressing matrix. J Tissue Eng 2021; 12:2041731421999750. [PMID: 33796249 PMCID: PMC7970265 DOI: 10.1177/2041731421999750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Accepted: 02/13/2021] [Indexed: 12/26/2022] Open
Abstract
Patients with diabetes experience impaired growth factor production such as
epidermal growth factor (EGF) and basic fibroblast growth factor (bFGF), and
they are reportedly involved in wound healing processes. Here, we report dual
growth factor-loaded hyaluronate collagen dressing (Dual-HCD) matrix, using
different ratios of the concentration of stabilized growth
factors—stabilized-EGF (S-EGF) and stabilized-bFGF (S-bFGF). At first, the
optimal concentration ratio of S-EGF to S-bFGF in the Dual-HCD matrix is
determined to be 1:2 in type I diabetic mice. This Dual-HCD matrix does not
cause cytotoxicity and can be used in vivo. The wound-healing effect of this
matrix is confirmed in type II diabetic mice. Dual HCD enhances angiogenesis
which promotes wound healing and thus, it shows a significantly greater
synergistic effect than the HCD matrix loaded with a single growth factor.
Overall, we conclude that the Dual-HCD matrix represents an effective
therapeutic agent for impaired diabetic wound healing.
Collapse
Affiliation(s)
- Jihyun Kim
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyoung-Mi Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Hwan Han
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Gangnam Severance Hospital, Seoul, South Korea
| | - Eun Ae Ko
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Suk Yoon
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Ik Kyu Park
- R&D Center, Genewel Co., Ltd., Sungnam, South Korea
| | - Hang-Cheol Shin
- School of Systems Biomedical Science, Soongsil University, Seoul, South Korea
| | - Kwang Hwan Park
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin Woo Lee
- Department of Orthopaedic Surgery, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, South Korea.,Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
31
|
BET bromodomain inhibitors regulate keratinocyte plasticity. Nat Chem Biol 2021; 17:280-290. [PMID: 33462494 DOI: 10.1038/s41589-020-00716-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/23/2020] [Indexed: 01/29/2023]
Abstract
Although most acute skin wounds heal rapidly, non-healing skin ulcers represent an increasing and substantial unmet medical need that urgently requires effective therapeutics. Keratinocytes resurface wounds to re-establish the epidermal barrier by transitioning to an activated, migratory state, but this ability is lost in dysfunctional chronic wounds. Small-molecule regulators of keratinocyte plasticity with the potential to reverse keratinocyte malfunction in situ could offer a novel therapeutic approach in skin wound healing. Utilizing high-throughput phenotypic screening of primary keratinocytes, we identify such small molecules, including bromodomain and extra-terminal domain (BET) protein family inhibitors (BETi). BETi induce a sustained activated, migratory state in keratinocytes in vitro, increase activation markers in human epidermis ex vivo and enhance skin wound healing in vivo. Our findings suggest potential clinical utility of BETi in promoting keratinocyte re-epithelialization of skin wounds. Importantly, this novel property of BETi is exclusively observed after transient low-dose exposure, revealing new potential for this compound class.
Collapse
|
32
|
Hou Q, Chen H, Liu Q, Yan X. FGF10 Attenuates Experimental Traumatic Brain Injury through TLR4/MyD88/NF-κB Pathway. Cells Tissues Organs 2021; 209:248-256. [PMID: 33440393 DOI: 10.1159/000511381] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 08/27/2020] [Indexed: 11/19/2022] Open
Abstract
Traumatic brain injury (TBI) can induce neuronal apoptosis and neuroinflammation, resulting in substantial neuronal damage and behavioral disorders. Fibroblast growth factors (FGFs) have been shown to be critical mediators in tissue repair. However, the role of FGF10 in experimental TBI remains unknown. In this study, mice with TBI were established via weight-loss model and validated by increase of modified neurological severity scores (mNSS) and brain water content. Secondly, FGF10 levels were elevated in mice after TBI, whereas intraventricular injection of Ad-FGF10 decreased mNSS score and brain water content, indicating the remittance of neurological deficit and cerebral edema in TBI mice. In addition, neuronal damage could also be ameliorated by stereotactic injection of Ad-FGF10. Overexpression of FGF10 increased protein expression of Bcl-2, while it decreased Bax and cleaved caspase-3/PARP, and improved neuronal apoptosis in TBI mice. In addition, Ad-FGF10 relieved neuroinflammation induced by TBI and significantly reduced the level of interleukin 1β/6, tumor necrosis factor α, and monocyte chemoattractant protein-1. Moreover, Ad-FGF10 injection decreased the protein expression level of Toll-like receptor 4 (TLR4), MyD88, and phosphorylation of NF-κB (p-NF-κB), suggesting the inactivation of the TLR4/MyD88/NF-κB pathway. In conclusion, overexpression of FGF10 could ameliorate neurological deficit, neuronal apoptosis, and neuroinflammation through inhibition of the TLR4/MyD88/NF-κB pathway, providing a potential therapeutic strategy for brain injury in the future.
Collapse
Affiliation(s)
- Qinhan Hou
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Hongmou Chen
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China,
| | - Quan Liu
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| | - Xianlei Yan
- Department of Neurosurgery, The Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou City, China
| |
Collapse
|
33
|
Cocetta V, Cadau J, Saponaro M, Giacomini I, Dall'Acqua S, Sut S, Catanzaro D, Orso G, Miolo G, Menilli L, Pagetta A, Ragazzi E, Montopoli M. Further assessment of Salvia haenkei as an innovative strategy to counteract skin photo-aging and restore the barrier integrity. Aging (Albany NY) 2021; 13:89-103. [PMID: 33424011 PMCID: PMC7835004 DOI: 10.18632/aging.202464] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 12/09/2020] [Indexed: 01/04/2023]
Abstract
Skin is the essential barrier of the human body which performs multiple functions. Endogenous factors, in concert with external assaults, continuously affect skin integrity, leading to distinct structural changes that influence not only the skin appearance but also its various physiological functions. Alterations of the barrier functions lead to an increased risk of developing disease and side reactions, thus the importance of maintaining the integrity of the epidermal barrier and slowing down the skin aging process is evident. Salvia haenkei (SH) has been recently identified as a potential anti-senescence agent; its extract is able to decrease the level of senescent cells by affecting the IL1α release and reducing reactive oxygen species (ROS) generation. In this study, SH extract was tested on human keratinocyte cell line (HaCaT) exposed to stress factors related to premature aging of cells such as free radicals and ultraviolet B radiation. We confirmed that SH acts as scavenger of ROS and found its ability to restore the skin barrier integrity by reinforcing the cytoskeleton structure, sealing the tight junctions and increasing the migration rate of cells. Given these results, this work becomes relevant, identifying Salvia haenkei as a compound useful for anti-aging skin treatment in clinical performance.
Collapse
Affiliation(s)
- Veronica Cocetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Jessica Cadau
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Miriam Saponaro
- Veneto Institute of Molecular Medicine (VIMM), Padova 35121, Italy.,Department of Medicine, University of Padova, Padova 35128, Italy
| | - Isabella Giacomini
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Stefano Dall'Acqua
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Stefania Sut
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Daniela Catanzaro
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Genny Orso
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Giorgia Miolo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Luca Menilli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Andrea Pagetta
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Eugenio Ragazzi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy
| | - Monica Montopoli
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova 35121, Italy.,Veneto Institute of Molecular Medicine (VIMM), Padova 35121, Italy
| |
Collapse
|
34
|
Ray AT, Mazot P, Brewer JR, Catela C, Dinsmore CJ, Soriano P. FGF signaling regulates development by processes beyond canonical pathways. Genes Dev 2020; 34:1735-1752. [PMID: 33184218 PMCID: PMC7706708 DOI: 10.1101/gad.342956.120] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/13/2020] [Indexed: 01/06/2023]
Abstract
FGFs are key developmental regulators that engage a signal transduction cascade through receptor tyrosine kinases, prominently engaging ERK1/2 but also other pathways. However, it remains unknown whether all FGF activities depend on this canonical signal transduction cascade. To address this question, we generated allelic series of knock-in Fgfr1 and Fgfr2 mouse strains, carrying point mutations that disrupt binding of signaling effectors, and a kinase dead allele of Fgfr2 that broadly phenocopies the null mutant. When interrogated in cranial neural crest cells, we identified discrete functions for signaling pathways in specific craniofacial contexts, but point mutations, even when combined, failed to recapitulate the single or double null mutant phenotypes. Furthermore, the signaling mutations abrogated established FGF-induced signal transduction pathways, yet FGF functions such as cell-matrix and cell-cell adhesion remained unaffected, though these activities did require FGFR kinase activity. Our studies establish combinatorial roles of Fgfr1 and Fgfr2 in development and uncouple novel FGFR kinase-dependent cell adhesion properties from canonical intracellular signaling.
Collapse
MESH Headings
- Animals
- Cell Adhesion/genetics
- Cell Death/genetics
- Cells, Cultured
- Fibroblast Growth Factors/physiology
- Gene Expression Regulation, Developmental/genetics
- Mice
- Mutation
- Neural Crest/cytology
- Protein Kinases/metabolism
- Receptor, Fibroblast Growth Factor, Type 1/genetics
- Receptor, Fibroblast Growth Factor, Type 1/metabolism
- Receptor, Fibroblast Growth Factor, Type 2/genetics
- Receptor, Fibroblast Growth Factor, Type 2/metabolism
- Receptors, Fibroblast Growth Factor/genetics
- Receptors, Fibroblast Growth Factor/metabolism
- Signal Transduction/genetics
Collapse
Affiliation(s)
- Ayan T Ray
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Pierre Mazot
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - J Richard Brewer
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Catarina Catela
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Colin J Dinsmore
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Philippe Soriano
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| |
Collapse
|
35
|
Nickol ME, Lyle SM, Dennehy B, Kindrachuk J. Dysregulated Host Responses Underlie 2009 Pandemic Influenza-Methicillin Resistant Staphylococcus aureus Coinfection Pathogenesis at the Alveolar-Capillary Barrier. Cells 2020; 9:E2472. [PMID: 33202895 PMCID: PMC7696554 DOI: 10.3390/cells9112472] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 11/11/2020] [Accepted: 11/11/2020] [Indexed: 02/04/2023] Open
Abstract
Influenza viruses are a continual public health concern resulting in 3-5 million severe infections annually despite intense vaccination campaigns and messaging. Secondary bacterial infections, including Staphylococcus aureus, result in increased morbidity and mortality during seasonal epidemics and pandemics. While coinfections can result in deleterious pathologic consequences, including alveolar-capillary barrier disruption, the underlying mechanisms are poorly understood. We have characterized host- and pathogen-centric mechanisms contributing to influenza-bacterial coinfections in a primary cell coculture model of the alveolar-capillary barrier. Using 2009 pandemic influenza (pH1N1) and methicillin-resistant S. aureus (MRSA), we demonstrate that coinfection resulted in dysregulated barrier function. Preinfection with pH1N1 resulted in modulation of adhesion- and invasion-associated MRSA virulence factors during lag phase bacterial replication. Host response modulation in coinfected alveolar epithelial cells were primarily related to TLR- and inflammatory response-mediated cell signaling events. While less extensive in cocultured endothelial cells, coinfection resulted in changes to cellular stress response- and TLR-related signaling events. Analysis of cytokine expression suggested that cytokine secretion might play an important role in coinfection pathogenesis. Taken together, we demonstrate that coinfection pathogenesis is related to complex host- and pathogen-mediated events impacting both epithelial and endothelial cell regulation at the alveolar-capillary barrier.
Collapse
Affiliation(s)
- Michaela E. Nickol
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Sarah M. Lyle
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Brendan Dennehy
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
| | - Jason Kindrachuk
- Laboratory of Emerging and Re-Emerging Viruses, Department of Medical Microbiology, University of Manitoba, Winnipeg, MB R3E 0J9, Canada; (M.E.N.); (S.M.L.); (B.D.)
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, Saskatoon, SK S7N 5E3, Canada
| |
Collapse
|
36
|
Goldstein NB, Steel A, Barbulescu CC, Koster MI, Wright MJ, Jones KL, Gao B, Ward B, Woessner B, Trottier Z, Pakieser J, Hu J, Lambert KA, Shellman YG, Fujita M, Robinson WA, Roop DR, Norris DA, Birlea SA. Melanocyte Precursors in the Hair Follicle Bulge of Repigmented Vitiligo Skin Are Controlled by RHO-GTPase, KCTD10, and CTNNB1 Signaling. J Invest Dermatol 2020; 141:638-647.e13. [PMID: 32800877 DOI: 10.1016/j.jid.2020.07.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 06/18/2020] [Accepted: 07/06/2020] [Indexed: 01/09/2023]
Abstract
In repigmentation of human vitiligo, the melanocyte (MC) precursors in the hair follicle bulge proliferate, migrate, and differentiate to repopulate the depigmented epidermis. Here, we present a comprehensive characterization of pathways and signals in the bulge that control the repigmentation process. Using biopsies from patients with vitiligo, we have selectively harvested, by laser capture microdissection, MC and keratinocyte precursors from the hair follicle bulge of untreated vitiligo skin and vitiligo skin treated with narrow-band UVB. The captured material was subjected to whole transcriptome RNA-sequencing. With this strategy, we found that repigmentation in the bulge MC precursors is driven by KCTD10, a signal with unknown roles in the skin, and CTNNB1 (encoding β-catenin) and RHO guanosine triphosphatase [RHO GTPase, RHO], two signaling pathways previously shown to be involved in pigmentation biology. Knockdown studies in cultured human MCs of RHOJ, the upmost differentially expressed RHO family component, corroborated with our findings in patients with vitiligo, identified RHOJ involvement in UV response and melanization, and confirmed previously identified roles in melanocytic cell migration and apoptosis. A better understanding of mechanisms that govern repigmentation in MC precursors will enable the discovery of molecules that induce robust repigmentation phenotypes in vitiligo.
Collapse
Affiliation(s)
| | - Andrea Steel
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | | | - Maranke I Koster
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Michael J Wright
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Kenneth L Jones
- Department of Hematology, University of Colorado, Aurora, Colorado, USA; Department of Pediatrics, University of Colorado, Aurora, Colorado, USA
| | - Bifeng Gao
- Sequencing and Microarray Core, University of Colorado, Aurora, Colorado, USA
| | - Brian Ward
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Brian Woessner
- Sequencing and Microarray Core, University of Colorado, Aurora, Colorado, USA
| | - Zachary Trottier
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Jen Pakieser
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - Junxiao Hu
- Department of Pediatrics, University of Colorado, Aurora, Colorado, USA; Cancer Center Biostatistics Core, University of Colorado, Aurora, Colorado, USA
| | - Karoline A Lambert
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Yiqun G Shellman
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA
| | - Mayumi Fujita
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA; Denver Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | | | - Dennis R Roop
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA
| | - David A Norris
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA; Denver Department of Veterans Affairs Medical Center, Denver, Colorado, USA
| | - Stanca A Birlea
- Department of Dermatology, University of Colorado, Aurora, Colorado, USA; Gates Center for Regenerative Medicine, University of Colorado, Aurora, Colorado, USA; Human Medical Genetics and Genomics Program, Aurora, Colorado, USA.
| |
Collapse
|
37
|
Sengal AT, Patch AM, Snell CE, Smith DS, Leung SCY, Talhouk A, Williams ED, McAlpine JN, Pollock PM. FGFR2c Mesenchymal Isoform Expression Is Associated with Poor Prognosis and Further Refines Risk Stratification within Endometrial Cancer Molecular Subtypes. Clin Cancer Res 2020; 26:4569-4580. [PMID: 32414751 DOI: 10.1158/1078-0432.ccr-19-4088] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 04/17/2020] [Accepted: 05/11/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE The two most common molecular subtypes of endometrial cancers, mismatch repair deficient (MMRd) and p53 wild-type (p53wt) comprise the majority of endometrial cancers and have intermediate prognoses where additional risk stratification biomarkers are needed. Isoform switching of FGFR2 from FGFR2b to FGFR2c (normally expressed in mesenchymal cells), has been reported in other solid carcinomas. The objective of this study was to investigate the role of FGFR2c in risk stratification of endometrial cancer. EXPERIMENTAL DESIGN We have developed and optimized a BaseScope RNA ISH assay to detect FGFR2c. FGFR2c expression was determined in a preliminary screening cohort of 78 endometrial cancers and a clinically and molecularly annotated Vancouver cohort (n = 465). Cox regression model analyses were performed to assess the prognostic value of FGFR2c. RESULTS Univariate and multivariate analyses revealed FGFR2c expression was significantly associated with shorter disease-specific survival (DSS) and progression-free survival (PFS) in endometrioid endometrial cancer (EEC, n = 302). Notably, FGFR2c expression was significantly associated with shorter PFS and DSS in patients with grade 3 EECs (P < 0.003 and P < 0.002) and the European Society Medical Oncology (ESMO) high-risk group (P < 0.0001 and P < 0.002), respectively. Moreover, within the MMRd subtype, FGFR2c expression was significantly associated with shorter PFS (P < 0.048) and DSS (P < 0.001). CONCLUSIONS FGFR2c expression appears an independent prognostic biomarker in patients with EEC and further discerns the outcomes within grade 3 tumors, ESMO high-risk groups, as well as within the MMRd and p53wt subtypes. FGFR2c inclusion into future molecular subtyping can further refine risk stratification of EEC.
Collapse
Affiliation(s)
- Asmerom T Sengal
- Queensland University of Technology, School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, located at the Translational Research Institute, PA Hospital Campus, 37 Kent St Woolloongabba, Brisbane, Queensland, Australia
| | - Ann-Marie Patch
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Cameron E Snell
- Mater Pathology, Mater Research and University of Queensland, Brisbane, Queensland, Australia
| | - Deborah S Smith
- Mater Pathology, Mater Research and University of Queensland, Brisbane, Queensland, Australia
| | - Samuel C Y Leung
- Department of Pathology and Laboratory Medicine, Genetic Pathology Evaluation Centre, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aline Talhouk
- Division of Gynaecologic Oncology, Department of Gynaecology and Obstetrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Elizabeth D Williams
- Queensland University of Technology, School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, located at the Translational Research Institute, PA Hospital Campus, 37 Kent St Woolloongabba, Brisbane, Queensland, Australia
| | - Jessica N McAlpine
- Division of Gynaecologic Oncology, Department of Gynaecology and Obstetrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Pamela M Pollock
- Queensland University of Technology, School of Biomedical Sciences, Faculty of Health, Institute of Health and Biomedical Innovation, located at the Translational Research Institute, PA Hospital Campus, 37 Kent St Woolloongabba, Brisbane, Queensland, Australia.
| |
Collapse
|
38
|
Rascu AM, Otelea MR, Mambet C, Handra C, Neagu AI, Rascu A, Giurcaneanu C, Diaconu CC. Modified Plasma Cytokine Profile in Occupational Dermatitis. Endocr Metab Immune Disord Drug Targets 2020; 20:1295-1302. [PMID: 32515314 DOI: 10.2174/1871530320666200607194021] [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: 10/31/2019] [Revised: 02/03/2020] [Accepted: 04/29/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND Despite significant progress in the diagnosis of contact dermatitis, the identification by specific tests or biomarkers remains an unsolved issue, particularly when needed for the confirmation of the occupational origin of the disease. OBJECTIVE To characterize the plasma proteome profile in occupational dermatitis in workers of paint industry. METHODS The study has a case-control design, comparing exposed workers with and without occupational contact dermatitis, matched for age, gender, occupational history, and comorbidities. An immunological assay (Human XL Cytokine Array Kit - ARY022B, R&D Systems) was used to measure the plasma levels of 105 cytokines and chemokines in a pooled sample of the cases and a pooled sample of the controls. RESULTS A 1.5-fold increase was noticed for interleukin 3, interleukin 10, and leptin in cases, as compared to controls. Fibroblast growth factor-7 and growth/differentiation factor-15 showed a 1.4-fold increase, while interleukin 19, interleukin 31, and macrophage inflammatory protein 3a.had only a 1.3- fold increase. The leukemia inhibitory factor was the only plasma cytokine that showed a 1.3-fold decrease. All other cytokines had a variation of less than 1.2-fold between cases and controls. CONCLUSION The recognition of the molecular signatures is very important for an accurate and indisputable diagnosis of occupational contact dermatitis. In workers from the paint industry, plasma levels of interleukins 3, 10, 13 and 19, fibroblast growth factor-7, and growth/differentiation factor-15, together with leukemia inducible factor, may differentiate subjects with contact dermatitis from those without skin lesions.
Collapse
Affiliation(s)
- Alexandra M Rascu
- Clinic Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Marina R Otelea
- Clinic Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | | | - Claudia Handra
- Clinic Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
| | - Ana I Neagu
- Stefan S. Nicolau Institute of Virology, Bucharest, Romania
| | - Agripina Rascu
- Clinic Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- Colentina Clinical Hospital, Bucharest, Romania
| | - Calin Giurcaneanu
- Clinic Department, "Carol Davila" University of Medicine and Pharmacy, Bucharest, Romania
- Dermatology Clinic, Elias Hospital, Bucharest, Romania
| | | |
Collapse
|
39
|
Meyer M, Ben-Yehuda Greenwald M, Rauschendorfer T, Sänger C, Jukic M, Iizuka H, Kubo F, Chen L, Ornitz DM, Werner S. Mouse genetics identifies unique and overlapping functions of fibroblast growth factor receptors in keratinocytes. J Cell Mol Med 2019; 24:1774-1785. [PMID: 31830366 PMCID: PMC6991627 DOI: 10.1111/jcmm.14871] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/07/2019] [Accepted: 11/11/2019] [Indexed: 12/13/2022] Open
Abstract
Fibroblast growth factors (FGFs) are key regulators of tissue development, homeostasis and repair, and abnormal FGF signalling is associated with various human diseases. In human and murine epidermis, FGF receptor 3 (FGFR3) activation causes benign skin tumours, but the consequences of FGFR3 deficiency in this tissue have not been determined. Here, we show that FGFR3 in keratinocytes is dispensable for mouse skin development, homeostasis and wound repair. However, the defect in the epidermal barrier and the resulting inflammatory skin disease that develops in mice lacking FGFR1 and FGFR2 in keratinocytes were further aggravated upon additional loss of FGFR3. This caused fibroblast activation and fibrosis in the FGFR1/FGFR2 double‐knockout mice and even more in mice lacking all three FGFRs, revealing functional redundancy of FGFR3 with FGFR1 and FGFR2 for maintaining the epidermal barrier. Taken together, our study demonstrates that FGFR1, FGFR2 and FGFR3 act together to maintain epidermal integrity and cutaneous homeostasis, with FGFR2 being the dominant receptor.
Collapse
Affiliation(s)
- Michael Meyer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | | | - Theresa Rauschendorfer
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Catharina Sänger
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Marko Jukic
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Haruka Iizuka
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Fumimasa Kubo
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| | - Lin Chen
- Center of Bone Metabolism and Repair, Department of Rehabilitation Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Third Military Medical University, Chongqing, China
| | - David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri
| | - Sabine Werner
- Department of Biology, Institute of Molecular Health Sciences, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
40
|
Kon E, Calvo-Jiménez E, Cossard A, Na Y, Cooper JA, Jossin Y. N-cadherin-regulated FGFR ubiquitination and degradation control mammalian neocortical projection neuron migration. eLife 2019; 8:47673. [PMID: 31577229 PMCID: PMC6786859 DOI: 10.7554/elife.47673] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2019] [Accepted: 10/01/2019] [Indexed: 12/18/2022] Open
Abstract
The functions of FGF receptors (FGFRs) in early development of the cerebral cortex are well established. Their functions in the migration of neocortical projection neurons, however, are unclear. We have found that FGFRs regulate multipolar neuron orientation and the morphological change into bipolar cells necessary to enter the cortical plate. Mechanistically, our results suggest that FGFRs are activated by N-Cadherin. N-Cadherin cell-autonomously binds FGFRs and inhibits FGFR K27- and K29-linked polyubiquitination and lysosomal degradation. Accordingly, FGFRs accumulate and stimulate prolonged Erk1/2 phosphorylation. Neurons inhibited for Erk1/2 are stalled in the multipolar zone. Moreover, Reelin, a secreted protein regulating neuronal positioning, prevents FGFR degradation through N-Cadherin, causing Erk1/2 phosphorylation. These findings reveal novel functions for FGFRs in cortical projection neuron migration, suggest a physiological role for FGFR and N-Cadherin interaction in vivo and identify Reelin as an extracellular upstream regulator and Erk1/2 as downstream effectors of FGFRs during neuron migration.
Collapse
Affiliation(s)
- Elif Kon
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Elisa Calvo-Jiménez
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Alexia Cossard
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| | - Youn Na
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Jonathan A Cooper
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Yves Jossin
- Laboratory of Mammalian Development & Cell Biology, Institute of Neuroscience, Université Catholique de Louvain, Brussels, Belgium
| |
Collapse
|
41
|
Seltmann K, Meyer M, Sulcova J, Kockmann T, Wehkamp U, Weidinger S, Auf dem Keller U, Werner S. Humidity-regulated CLCA2 protects the epidermis from hyperosmotic stress. Sci Transl Med 2019; 10:10/440/eaao4650. [PMID: 29743348 DOI: 10.1126/scitranslmed.aao4650] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Accepted: 04/16/2018] [Indexed: 12/12/2022]
Abstract
Low environmental humidity aggravates symptoms of the inflammatory skin disease atopic dermatitis (AD). Using mice that develop AD-like signs, we show that an increase in environmental humidity rescues their cutaneous inflammation and associated epidermal abnormalities. Quantitative proteomics analysis of epidermal lysates of mice kept at low or high humidity identified humidity-regulated proteins, including chloride channel accessory 3A2 (CLCA3A2), a protein with previously unknown function in the skin. The epidermis of patients with AD, organotypic skin cultures under dry conditions, and cultured keratinocytes exposed to hyperosmotic stress showed up-regulation of the nonorthologous human homolog CLCA2. Hyperosmolarity-induced CLCA2 expression occurred via p38/c-Jun N-terminal kinase-activating transcription factor 2 signaling. CLCA2 knockdown promoted keratinocyte apoptosis induced by hyperosmotic stress through impairment of cell-cell adhesion. These findings provide a mechanistic explanation for the beneficial effect of high environmental humidity for AD patients and identify CLCA3A2/CLCA2 up-regulation as a mechanism to protect keratinocytes from damage induced by low humidity.
Collapse
Affiliation(s)
- Kristin Seltmann
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Michael Meyer
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Jitka Sulcova
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Tobias Kockmann
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.,Functional Genomics Center Zurich, ETH Zurich/University of Zurich, 8057 Zurich, Switzerland
| | - Ulrike Wehkamp
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Stephan Weidinger
- Department of Dermatology, Allergology and Venereology, University Hospital Schleswig-Holstein, Campus Kiel, Germany
| | - Ulrich Auf dem Keller
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
42
|
Hirano YU, Suzuki K, Iguchi T, Yamada G, Miyagawa S. The Role of Fgf Signaling on Epithelial Cell Differentiation in Mouse Vagina. In Vivo 2019; 33:1499-1505. [PMID: 31471398 DOI: 10.21873/invivo.11630] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Revised: 06/30/2019] [Accepted: 07/03/2019] [Indexed: 12/28/2022]
Abstract
BACKGROUND/AIM The mouse vagina exhibits stratified squamous epithelium, which is comprised of multiple cell layers. We previously showed that erbB signaling, induced by epithelial estrogen receptor 1 (ESR1), is required for the initial differentiation of the epithelium. However, the downstream effector that mediates terminal differentiation in the apical layers remains elusive. The contribution of fibroblast growth factor (FGF) to vaginal epithelial cell differentiation was investigated. MATERIALS AND METHODS Vaginas from wild-type or epithelium-specific Esr1 conditional knockout (cKO) mice were analyzed using immunohistochemistry and quantitative real-time RT-PCR. RESULTS Of the FGF ligands examined, Fgf22 mRNA was significantly induced following estrogen treatment. Furthermore, FGF downstream signaling, phosphorylated FRS2 and ERK1/2 were exclusively expressed in the apical layers of the vaginal epithelium. No changes in such expression were observed in the Esr1 cKO mice. CONCLUSION FGF-ERK/MAPK pathway may be a main inducer of terminal differentiation in the mouse vaginal epithelium.
Collapse
Affiliation(s)
- Y U Hirano
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Kentaro Suzuki
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Taisen Iguchi
- Graduate School of Nanobioscience, Yokohama City University, Yokohama, Japan
| | - Gen Yamada
- Department of Developmental Genetics, Institute of Advanced Medicine, Wakayama Medical University, Wakayama, Japan
| | - Shinichi Miyagawa
- Faculty of Industrial Science and Technology, Tokyo University of Science, Tokyo, Japan
| |
Collapse
|
43
|
The Morphopathogenetic Aspects of Intraabdominal Adhesions in Children under One Year of Age. ACTA ACUST UNITED AC 2019; 55:medicina55090556. [PMID: 31480453 PMCID: PMC6780280 DOI: 10.3390/medicina55090556] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/13/2019] [Accepted: 08/14/2019] [Indexed: 01/20/2023]
Abstract
Background and Objectives: The morphopathogenesis of adhesions is a complex process, characterized by the accumulation of an extracellular matrix, inflammation and hypoxia. The regulatory role between morphopathogenic factors in adhesions has not yet been defined. The aim was to investigate the appearance of transforming growth factor beta (TGFβ), basic fibroblast growth factor (FGF-2), fibroblast growth factor receptor 1 (FGFR1), protein gene product 9.5 (PGP 9.5), chromogranin A (CgA), interleukin-1 alpha (IL-1α), interleukin-4 (IL-4), interleukin-6 (IL-6), interleukin-7 (IL-7), interleukin-8 (IL-8), interleukin-10 (IL-10), tumor necrosis factor alpha (TNFα), matrix metaloproteinase-2 (MMP-2) and matrix metaloproteinase-2 tissue inhibitor (TIMP-2) in intraabdominal adhesions. Materials and Methods: The study material was obtained from 49 patients under one year of age with total or partial bowel obstruction. All factors were detected using immunohistochemistry methods and their relative distribution was evaluated by means of the semiquantitative counting method. Results: Intraabdominal adhesions are characterized by increased TGFβ, FGFR1 and decreased FGF-2, PGP 9.5, IL-1, IL-4, IL-8, TIMP-2 findings. The most significant changes observed were the remodulation of the extracellular matrix, promotion of neoangiogenesis and the maintenance of a prolonged inflammation. Conclusions: The increase in TGFβ, as well as the disbalance between MMP-2 and TIMP-2 proves an increased fibrosis in intraabdominal adhesions. Less detected FGF-2 and more prominent FGR1 findings points out a compensatory receptor stimulation in response to the lacking same factor. The decrease in PGP 9.5 indicate hypoxic injury and proves the stimulation of neoangiogenesis. An unpronounced IL-1 and marked IL-10 finding indicate the local tissue protection reaction, the decrease in IL-4 could be the direct cause of giant cells, but the decrease of IL-8 could confirm a delayed chemotaxis of inflammatory cells.
Collapse
|
44
|
Nourian Dehkordi A, Mirahmadi Babaheydari F, Chehelgerdi M, Raeisi Dehkordi S. Skin tissue engineering: wound healing based on stem-cell-based therapeutic strategies. Stem Cell Res Ther 2019; 10:111. [PMID: 30922387 PMCID: PMC6440165 DOI: 10.1186/s13287-019-1212-2] [Citation(s) in RCA: 305] [Impact Index Per Article: 50.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Normal wound healing is a dynamic and complex multiple phase process involving coordinated interactions between growth factors, cytokines, chemokines, and various cells. Any failure in these phases may lead wounds to become chronic and have abnormal scar formation. Chronic wounds affect patients' quality of life, since they require repetitive treatments and incur considerable medical costs. Thus, much effort has been focused on developing novel therapeutic approaches for wound treatment. Stem-cell-based therapeutic strategies have been proposed to treat these wounds. They have shown considerable potential for improving the rate and quality of wound healing and regenerating the skin. However, there are many challenges for using stem cells in skin regeneration. In this review, we present some sets of the data published on using embryonic stem cells, induced pluripotent stem cells, and adult stem cells in healing wounds. Additionally, we will discuss the different angles whereby these cells can contribute to their unique features and show the current drawbacks.
Collapse
Affiliation(s)
- Azar Nourian Dehkordi
- Department of Stem Cell and Regenerative Medicine, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Fatemeh Mirahmadi Babaheydari
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mohammad Chehelgerdi
- Biotechnology Research Center, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | | |
Collapse
|
45
|
Chadaeva I, Ponomarenko P, Rasskazov D, Sharypova E, Kashina E, Kleshchev M, Ponomarenko M, Naumenko V, Savinkova L, Kolchanov N, Osadchuk L, Osadchuk A. Natural Selection Equally Supports the Human Tendencies in Subordination and Domination: A Genome-Wide Study With in silico Confirmation and in vivo Validation in Mice. Front Genet 2019; 10:73. [PMID: 30873204 PMCID: PMC6404730 DOI: 10.3389/fgene.2019.00073] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 01/28/2019] [Indexed: 12/29/2022] Open
Abstract
We proposed the following heuristic decision-making rule: "IF {an excess of a protein relating to the nervous system is an experimentally known physiological marker of low pain sensitivity, fast postinjury recovery, or aggressive, risk/novelty-seeking, anesthetic-like, or similar agonistic-intolerant behavior} AND IF {a single nucleotide polymorphism (SNP) causes overexpression of the gene encoding this protein} THEN {this SNP can be a SNP marker of the tendency in dominance} WHILE {underexpression corresponds to subordination} AND vice versa." Using this decision-making rule, we analyzed 231 human genes of neuropeptidergic, non-neuropeptidergic, and neurotrophinergic systems that encode neurotrophic and growth factors, interleukins, neurotransmitters, receptors, transporters, and enzymes. These proteins are known as key factors of human social behavior. We analyzed all the 5,052 SNPs within the 70 bp promoter region upstream of the position where the protein-coding transcript starts, which were retrieved from databases Ensembl and dbSNP using our previously created public Web service SNP_TATA_Comparator (http://beehive.bionet.nsc.ru/cgi-bin/mgs/tatascan/start.pl). This definition of the promoter region includes all TATA-binding protein (TBP)-binding sites. A total of 556 and 552 candidate SNP markers contributing to the dominance and the subordination, respectively, were uncovered. On this basis, we determined that 231 human genes under study are subject to natural selection against underexpression (significance p < 0.0005), which equally supports the human tendencies in domination and subordination such as the norm of a reaction (plasticity) of the human social hierarchy. These findings explain vertical transmission of domination and subordination traits previously observed in rodent models. Thus, the results of this study equally support both sides of the century-old unsettled scientific debate on whether both aggressiveness and the social hierarchy among humans are inherited (as suggested by Freud and Lorenz) or are due to non-genetic social education, when the children are influenced by older individuals across generations (as proposed by Berkowitz and Fromm).
Collapse
Affiliation(s)
- Irina Chadaeva
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | | | | | | | | | - Maxim Kleshchev
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Mikhail Ponomarenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Vladimir Naumenko
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | | | - Nikolay Kolchanov
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Ludmila Osadchuk
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexandr Osadchuk
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
46
|
Miranda JP, Camões SP, Gaspar MM, Rodrigues JS, Carvalheiro M, Bárcia RN, Cruz P, Cruz H, Simões S, Santos JM. The Secretome Derived From 3D-Cultured Umbilical Cord Tissue MSCs Counteracts Manifestations Typifying Rheumatoid Arthritis. Front Immunol 2019; 10:18. [PMID: 30804924 PMCID: PMC6370626 DOI: 10.3389/fimmu.2019.00018] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Accepted: 01/04/2019] [Indexed: 01/23/2023] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disorder whose treatment is mostly restricted to pain and symptom management and to the delay of joint destruction. Mesenchymal stem/stromal cells from the umbilical cord tissue (UC-MSCs) have previously been proven to be immunomodulatory and more efficient than bone marrow-derived MSCs in causing remission of local and systemic arthritic manifestations in vivo. Given the paracrine nature of UC-MSC activity, their application as active substances can be replaced by their secretome, thus avoiding allogeneic rejection and safety issues related to unwanted grafting. In this work, we aimed at demonstrating the viability of applying the 3D-primed UC-MSC secretome for the amelioration of arthritic signs. A proteomic analysis was performed to both, media conditioned by UC-MSC monolayer (CM2D) and 3D cultures (CM3D). The analysis of relevant trophic factors confirmed secretome profiles with very significant differences in terms of therapeutic potential. Whereas, CM3D was characterised by a prevailing expression of anti-inflammatory cytokines such as IL-10 and LIF, along with trophic factors involved in different mechanisms leading to tissue regeneration, such as PDGF-BB, FGF-2, I-309, SCF, and GM-CSF; CM2D presented relatively higher levels of IL-6, MCP-1, and IL-21, with recognised pro-inflammatory roles in joint disease and pleiotropic effects in the progression of rheumatoid arthritis (RA). Accordingly, different motogenic effects over mouse chondrocytes and distinct capacities of inducing glycosaminoglycan synthesis in vitro were observed between CM3D and CM2D. Finally, the evaluation of arthritic manifestations in vivo, using an adjuvant-induced model for arthritis (AIA), suggested a significantly higher therapeutic potential of CM3D over CM2D and even UC-MSCs. Histological analysis confirmed a faster remission of local and systemic arthritic manifestations of CM3D-treated animals. Overall, the results show that the use of UC-MSC CM3D is a viable and better strategy than direct UC-MSC administration for counteracting AIA-related signs. This strategy represents a novel MSC-based but nonetheless cell-free treatment for arthritic conditions such as those characterising RA.
Collapse
Affiliation(s)
- Joana P Miranda
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Sérgio P Camões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Maria M Gaspar
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Joana S Rodrigues
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Manuela Carvalheiro
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | | | | | | | - Sandra Simões
- Faculty of Pharmacy, Research Institute for Medicines (iMed.ULisboa), University of Lisbon, Lisbon, Portugal
| | - Jorge M Santos
- ECBio S.A., Amadora, Portugal.,Centro de Estudos de Ciência Animal, Instituto de Ciências, Tecnologias e Agroambiente, Universidade do Porto, Porto, Portugal.,Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| |
Collapse
|
47
|
Mellers AP, Tenorio CA, Lacatusu DA, Powell BD, Patel BN, Harper KM, Blaber M. Fine-Sampled Photographic Quantitation of Dermal Wound Healing Senescence in Aged BALB/cByJ Mice and Therapeutic Intervention with Fibroblast Growth Factor-1. Adv Wound Care (New Rochelle) 2018; 7:409-418. [PMID: 31741752 DOI: 10.1089/wound.2018.0801] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 06/24/2018] [Indexed: 11/13/2022] Open
Abstract
Objective: To determine quantitative parameters of dermal wound healing senescence in aged BALB/cByJ mice (an important animal model of aging) and to evaluate the potential for therapeutic intervention by fibroblast growth factor-1 (FGF-1). Approach: Utilize a novel noninvasive fine-sampled photographic methodology to quantify wound healing parameters for healing phases from wounding through to wound closure. Results: Parameters associated with key healing phases were quantified and compared between nonaged and aged cohorts of both genders. The results identify a sexual dimorphism in dermal wound healing, with nonaged females exhibiting a greater overall healing efficiency than males. This enhanced healing in females, however, senesces with age such that healing parameters for aged males and females are statistically indistinguishable. Topical application of FGF-1 was identified as an effective therapeutic intervention to treat dermal healing senescence in aged females. Innovation: The FGF intervention is being analyzed using a new recently published model. This approach significantly increases the amount of preclinical animal data obtainable in wound healing studies, minimizes cohort number compared with (lethal) histological studies, and permits a direct statistical comparison between different healing studies. Conclusion: Quantitative parameters of dermal wound healing, obtained from noninvasive fine-sampled photographic data, identify topical FGF-1 as an effective therapeutic to treat the senescence of dermal healing present in aged female BALB/cByJ mice.
Collapse
Affiliation(s)
- Alana P. Mellers
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| | - Connie A. Tenorio
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| | - Diana A. Lacatusu
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| | - Brett D. Powell
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| | - Bhavi N. Patel
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| | - Kathleen M. Harper
- Biomedical Research Laboratory Animal Resources, Florida State University, Tallahassee, Florida
| | - Michael Blaber
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida
| |
Collapse
|
48
|
Andasari V, Lü D, Swat M, Feng S, Spill F, Chen L, Luo X, Zaman M, Long M. Computational model of wound healing: EGF secreted by fibroblasts promotes delayed re-epithelialization of epithelial keratinocytes. Integr Biol (Camb) 2018; 10:605-634. [PMID: 30206629 PMCID: PMC6571173 DOI: 10.1039/c8ib00048d] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It is widely agreed that keratinocyte migration plays a crucial role in wound re-epithelialization. Defects in this function contribute to wound reoccurrence causing significant clinical problems. Several in vitro studies have shown that the speed of migrating keratinocytes can be regulated by epidermal growth factor (EGF) which affects keratinocyte's integrin expression. The relationship between integrin expression (through cell-matrix adhesion) stimulated by EGF and keratinocyte migration speed is not linear since increased adhesion, due to increased integrin expression, has been experimentally shown to slow down cell migration due to the biphasic dependence of cell speed on adhesion. In our previous work we showed that keratinocytes that were co-cultured with EGF-enhanced fibroblasts formed an asymmetric migration pattern, where, the cumulative distances of keratinocytes migrating toward fibroblasts were smaller than those migrating away from fibroblasts. This asymmetric pattern is thought to be provoked by high EGF concentration secreted by fibroblasts. The EGF stimulates the expression of integrin receptors on the surface of keratinocytes migrating toward fibroblasts via paracrine signaling. In this paper, we present a computational model of keratinocyte migration that is controlled by EGF secreted by fibroblasts using the Cellular Potts Model (CPM). Our computational simulation results confirm the asymmetric pattern observed in experiments. These results provide a deeper insight into our understanding of the complexity of keratinocyte migration in the presence of growth factor gradients and may explain re-epithelialization failure in impaired wound healing.
Collapse
Affiliation(s)
- Vivi Andasari
- Boston University, Department of Biomedical Engineering, 44 Cummington Mall, Boston, MA 02215, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Nrf3 promotes UV-induced keratinocyte apoptosis through suppression of cell adhesion. Cell Death Differ 2018; 25:1749-1765. [PMID: 29487353 PMCID: PMC6179989 DOI: 10.1038/s41418-018-0074-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 12/19/2017] [Accepted: 01/22/2018] [Indexed: 01/13/2023] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a key regulator of the cellular stress response, but the biological functions of the related Nrf3 protein are largely unknown. Here we demonstrate a novel pro-apoptotic function of Nrf3 in mouse and human keratinocytes. In response to UV irradiation, Nrf3-deficient keratinocytes were protected from apoptosis in vitro and in vivo. The protective function was also seen under oxidative or hyperosmotic stress conditions, but not when apoptosis was induced by disruption of cell–matrix interactions. Mechanistically, we show that Nrf3-deficient keratinocytes exhibit stronger cell–cell and cell-matrix adhesion, which correlates with higher cell surface integrin levels and enhanced activation of focal adhesion kinase. Nrf3-deficient cells also formed more and larger focal adhesions and exhibited a higher motility. These results suggest that the strong expression of Nrf3 in basal keratinocytes promotes their elimination in response to DNA damage-inducing agents, thereby preventing accumulation of mutated stem and transit amplifying cells in the epidermis.
Collapse
|
50
|
Maddaluno L, Urwyler C, Werner S. Fibroblast growth factors: key players in regeneration and tissue repair. Development 2017; 144:4047-4060. [PMID: 29138288 DOI: 10.1242/dev.152587] [Citation(s) in RCA: 178] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tissue injury initiates a complex repair process, which in some organisms can lead to the complete regeneration of a tissue. In mammals, however, the repair of most organs is imperfect and results in scar formation. Both regeneration and repair are orchestrated by a highly coordinated interplay of different growth factors and cytokines. Among the key players are the fibroblast growth factors (FGFs), which control the migration, proliferation, differentiation and survival of different cell types. In addition, FGFs influence the expression of other factors involved in the regenerative response. Here, we summarize current knowledge on the roles of endogenous FGFs in regeneration and repair in different organisms and in different tissues and organs. Gaining a better understanding of these FGF activities is important for appropriate modulation of FGF signaling after injury to prevent impaired healing and to promote organ regeneration in humans.
Collapse
Affiliation(s)
- Luigi Maddaluno
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Corinne Urwyler
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| | - Sabine Werner
- Institute of Molecular Health Sciences, Department of Biology, Swiss Federal Institute of Technology (ETH) Zurich, Otto-Stern-Weg 7, 8093 Zürich, Switzerland
| |
Collapse
|