Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros











Intervalo de año de publicación
1.
Front Bioeng Biotechnol ; 12: 1386896, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38646012

RESUMEN

Introduction: Autologous cell suspension (ACS)-based therapy represents a highly promising approach for burns and chronic wounds. However, existing technologies have not achieved the desired clinical success due to several limitations. To overcome practical and cost-associated obstacles of existing ACS methods, we have established a novel methodology for rapid, enzymatic disaggregation of human skin cells and their isolation using a procedure that requires no specialist laboratory instrumentation and is performed at room temperature. Methods: Cells were isolated using enzymatic disaggregation of split-thickness human skin followed by several filtration steps for isolation of cell populations, and cell viability was determined. Individual population recovery was confirmed in appropriate culture medium types, and the presence of epidermal stem cells (EpSCs) within keratinocyte sub-populations was defined by flow cytometry via detection of CD49 and CD71. Positive mediators of wound healing secreted by ACS-derived cultures established on a collagen-based wound-bed mimic were detected by proteome arrays and quantified by ELISA, and the role of such mediators was determined by cell proliferation assays. The effect of ACS-derived conditioned-medium on myofibroblasts was investigated using an in-vitro model of myofibroblast differentiation via detection of α-SMA using immunoblotting and immunofluorescence microscopy. Results: Our methodology permitted efficient recovery of keratinocytes, fibroblasts and melanocytes, which remained viable upon long-term culture. ACS-derivatives comprised sub-populations with the CD49-high/CD71-low expression profile known to demarcate EpSCs. Via secretion of mitogenic factors and wound healing-enhancing mediators, the ACS secretome accelerated keratinocyte proliferation and markedly curtailed cytodifferentiation of myofibroblasts, the latter being key mediators of fibrosis and scarring. Discussion: The systematic characterisation of the cell types within our ACS isolates provided evidence for their superior cell viability and the presence of EpSCs that are critical drivers of wound healing. We defined the biological properties of ACS-derived keratinocytes, which include ability to secrete positive mediators of wound healing as well as suppression of myofibroblast cytodifferentiation. Thus, our study provides several lines of evidence that the established ACS isolates comprise highly-viable cell populations which can physically support wound healing and possess biological properties that have the potential to enhance not only the speed but also the quality of wound healing.

2.
Australas J Dermatol ; 65(4): 311-318, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38419202

RESUMEN

BACKGROUND: Atrophic acne scarring is a common sequela of inflammatory acne, causing significant problems for affected patients. Although prolonged inflammation and subsequent aberrant tissue regeneration are considered the underlying pathogenesis, the role of epidermal stem cells, which are crucial to the regeneration of pilosebaceous units, remains unknown. OBJECTIVES: To examine the changes occurring in epidermal stem cells in atrophic acne scars. METHODS: Changes in collagen, elastic fibre and human leukocyte antigen (HLA)-DR expression were analysed in normal skin and inflammatory acne lesions at days 1, 3 and 7 after development. The expression of epidermal stem cell markers and proliferation markers was compared between normal skin and mature atrophic acne scar tissue. RESULTS: In acne lesions, inflammation had invaded into pilosebaceous units over time. Their normal structure had been destructed and replaced with a reduced amount of collagen and elastic fibre. Expression of stem cell markers including CD34, p63, leucine-rich repeat-containing G protein-coupled receptor (LGR)6 and LGR5, which are expressed in the interfollicular epidermis, isthmus and bulge of hair follicles, significantly decreased in atrophic acne scar tissue compared to normal skin. Epidermal proliferation was significantly reduced in scar tissue. CONCLUSIONS: These findings suggest that as inflammatory acne lesions progress, inflammation gradually infiltrates the pilosebaceous unit and affects the resident stem cells. This disruption impedes the normal regeneration of the interfollicular epidermis and adnexal structures, resulting in atrophic acne scars.


Asunto(s)
Acné Vulgar , Cicatriz , Folículo Piloso , Células Madre , Humanos , Acné Vulgar/complicaciones , Acné Vulgar/patología , Cicatriz/patología , Cicatriz/etiología , Células Madre/metabolismo , Células Madre/patología , Folículo Piloso/patología , Atrofia , Colágeno/metabolismo , Tejido Elástico/patología , Masculino , Femenino , Antígenos HLA-DR/metabolismo , Proliferación Celular , Adulto Joven , Adulto , Células Epidérmicas/metabolismo , Epidermis/patología , Epidermis/metabolismo
3.
Artículo en Chino | WPRIM (Pacífico Occidental) | ID: wpr-1021521

RESUMEN

BACKGROUND:Hypertrophic scar is a skin fibrosis disease characterized by excessive proliferation of fibroblasts,epidermal thickening,and stratum corneum dysfunction.At present,the pathogenesis of Hypertrophic scar is still unclear. OBJECTIVE:To screen the core(Hub)genes and important signaling pathways in hypertrophic scar-related datasets based on bioinformatics,and then verify them by cell experiments to predict small molecule drugs that may have therapeutic effects on hypertrophic scar. METHODS:Datasets related to hypertrophic scar were searched from Gene Expression Omnibus(GEO)database,and differentially expressed genes were identified by R software analysis.Gene ontology and KEGG enrichment analyses were performed for differentially expressed genes.Protein-protein interaction network of differentially expressed genes was constructed using String online platform.Then,the key genes and core modules in the protein-protein interaction network were screened by Cytohubba and MCODE plugin-in Cytoscape software respectively,and the Hub genes were obtained by the intersection of the above key genes and the genes that formed the core module.Real-time fluorescent quantitative PCR was used to verify the difference in Hub gene mRNA expression between human hypertrophic scar and normal skin epidermal stem cells.The histological data from the Human Protein Atlas were used to verify the differences in the expression and distribution of Hub gene-encoded proteins in the two kinds of human tissues.Finally,the potential drugs for hypertrophic scar were predicted by the connectivity map database. RESULTS AND CONCLUSION:Among the identified differentially expressed genes,102 genes were up-regulated and 702 genes were down-regulated.Gene ontology and KEGG analysis showed that the enriched signaling pathways and biological processes were mainly involved in tight junction,arachidonic acid metabolism,extracellular matrix receptor interaction,epidermal development and keratinization.Eight Hub genes were found to be closely related to the mevalonate pathway that regulates cholesterol metabolism,including HMGCS1,DHCR7,MSMO1,FDPS,MVK,HMGCR,MVD and ACAT2.Compared with the normal skin group,the mRNA expression of HMGCS1,DHCR7,MSMO1,FDPS,HMGCR,MVD and ACAT2 in the hypertrophic scar group decreased significantly(P<0.05),while MVK mRNA expression had no significant change(P>0.05).Except for MVK,the expression levels of other Hub gene-encoded proteins in normal skin tissue were higher than those in hypertrophic scar tissue(P<0.05).The top 10 candidate drugs included protein kinase A inhibitor(H-89),serine protease inhibitor(Dabigatran-Etexilate),FLT3 inhibitor(sunitinib),among which resveratrol and β-sitosterol are plant extracts.To conclude,Hub genes closely related to mevalonate metabolism may affect the structure and function of the epidermis by regulating lipid metabolism,which may an important pathogenesis of hypertrophic scar.The small-molecule compounds identified in this study can be used as candidate drugs for the treatment of hypertrophic scar.

4.
Acta Biochim Biophys Sin (Shanghai) ; 55(8): 1265-1274, 2023 Jul 03.
Artículo en Inglés | MEDLINE | ID: mdl-37394884

RESUMEN

Proliferation and migration of epidermal stem cells (EpSCs) are essential for epithelialization during skin wound healing. Angiopoietin-like 4 (ANGPTL4) has been reported to play an important role in wound healing, but the mechanisms involved are not fully understood. Here, we investigate the contribution of ANGPTL4 to full-thickness wound re-epithelialization and the underlying mechanisms using Angptl4-knockout mice. Immunohistochemical staining reveals that ANGPTL4 is significantly upregulated in the basal layer cells of the epidermis around the wound during cutaneous wound healing. ANGPTL4 deficiency impairs wound healing. H&E staining shows that ANGPTL4 deficiency significantly reduces the thickness, length and area of the regenerated epidermis postwounding. Immunohistochemical staining for markers of EpSCs (α6 integrin and ß1 integrin) and cell proliferation (PCNA) shows that the number and proliferation of EpSCs in the basal layer of the epidermis are reduced in ANGPTL4-deficient mice. In vitro studies show that ANGPTL4 deficiency impedes EpSC proliferation, causes cell cycle arrest at the G1 phase and reduces the expressions of cyclins D1 and A2, which can be reversed by ANGPTL4 overexpression. ANGPTL4 deletion suppresses EpSC migration, which is also rescued by ANGPTL4 overexpression. Overexpression of ANGPTL4 in EpSCs accelerates cell proliferation and migration. Collectively, our results indicate that ANGPTL4 promotes EpSC proliferation by upregulating cyclins D1 and A2 expressions and accelerating the cell cycle transition from G1 to S phase and that ANGPTL4 promotes skin wound re-epithelialization by stimulating EpSC proliferation and migration. Our study reveals a novel mechanism underlying EpSC activation and re-epithelialization during cutaneous wound healing.


Asunto(s)
Epidermis , Repitelización , Animales , Ratones , Angiopoyetinas/metabolismo , Movimiento Celular , Proliferación Celular/genética , Ciclinas/metabolismo , Epidermis/metabolismo , Ratones Noqueados , Células Madre/metabolismo
5.
Inflamm Regen ; 43(1): 22, 2023 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-36973725

RESUMEN

BACKGROUND: Despite progress in developing wound care strategies, there is currently no treatment that promotes the self-tissue repair capabilities. H2 has been shown to effectively protect cells and tissues from oxidative and inflammatory damage. While comprehensive effects and how H2 functions in wound healing remains unknown, especially for the link between H2 and extracellular matrix (ECM) deposition and epidermal stem cells (EpSCs) activation. METHODS: Here, we established a cutaneous aseptic wound model and applied a high concentration of H2 (66% H2) in a treatment chamber. Molecular mechanisms and the effects of healing were evaluated by gene functional enrichment analysis, digital spatial profiler analysis, blood perfusion/oxygen detection assay, in vitro tube formation assay, enzyme-linked immunosorbent assay, immunofluorescent staining, non-targeted metabonomic analysis, flow cytometry, transmission electron microscope, and live-cell imaging. RESULTS: We revealed that a high concentration of H2 (66% H2) greatly increased the healing rate (3 times higher than the control group) on day 11 post-wounding. The effect was not dependent on O2 or anti-reactive oxygen species functions. Histological and cellular experiments proved the fast re-epithelialization in the H2 group. ECM components early (3 days post-wounding) deposition were found in the H2 group of the proximal wound, especially for the dermal col-I, epidermal col-III, and dermis-epidermis-junction col-XVII. H2 accelerated early autologous EpSCs proliferation (1-2 days in advance) and then differentiation into myoepithelial cells. These epidermal myoepithelial cells could further contribute to ECM deposition. Other beneficial outcomes include sustained moist healing, greater vascularization, less T-helper-1 and T-helper-17 cell-related systemic inflammation, and better tissue remodelling. CONCLUSION: We have discovered a novel pattern of wound healing induced by molecular hydrogen treatment. This is the first time to reveal the direct link between H2 and ECM deposition and EpSCs activation. These H2-induced multiple advantages in healing may be related to the enhancement of cell viability in various cells and the maintenance of mitochondrial functions at a basic level in the biological processes of life.

6.
J Dermatol Sci ; 106(3): 150-158, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35610160

RESUMEN

BACKGROUND: Age-related thinning and reduced cell proliferation in the human epidermis are associated with the accumulation of senescent cells and decreases in the number and function of epidermal stem cells. OBJECTIVE: This study examined the expression of INHBA/Activin-A in human epidermis and expression differences with age, and the effect of Activin-A on epidermal stem/progenitor cells. METHODS: Immunohistochemical staining was used to analyze age-related changes in the expression of INHBA/Activin-A in the epidermal tissue of young and old subjects. Epidermal INHBA/Activin-A expression levels, epidermal morphology, and the number of epidermal stem/progenitor cells or proliferating cells were investigated using older abdominal skin samples. The effects of Activin-A on the development of a three-dimensional (3D) reconstructed epidermis and cell proliferation were also assessed. RESULTS: INHBA/Activin-A expression levels in the human epidermis increased with age, although they varied among individuals. In the epidermis of older abdominal skin samples, INHBA/Activin-A expression levels negatively correlated with epidermal thickness, the rete ridge depth and the interdigitation index. The proportion of epidermal stem/progenitor cells and proliferating cells decreased with increases in INHBA/Activin-A expression levels. Activin-A had no effect on the differentiation of keratinocytes in the 3D-reconstructed epidermis; however, thinning of the 3D epidermis was noted. Moreover, the addition of Activin-A inhibited the proliferation of epidermal stem/progenitor cells in a concentration-dependent manner. CONCLUSIONS: Age-related increased in INHBA/Activin-A expression levels were observed in the human epidermis, and may contribute to epidermal thinning and decreases in the number of epidermal stem/progenitor cells and proliferative activity.


Asunto(s)
Activinas , Epidermis , Activinas/metabolismo , Envejecimiento , Proliferación Celular , Epidermis/metabolismo , Humanos , Subunidades beta de Inhibinas , Células Madre/metabolismo
7.
Front Genet ; 12: 705019, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34539738

RESUMEN

Epidermolysis bullosa (EB) is a group of devastating genetic diseases characterized by skin and mucosal fragility and formation of blisters, which develop either spontaneously or in response to minor mechanical trauma. There is no definitive therapy for any form of EB. Intermediate junctional EB (JEB) caused by mutations in the gene LAMB3 has been the first genetic skin disease successfully tackled by ex vivo gene therapy. Here, we present a multicenter, open-label, uncontrolled phase II/III study that aims at confirming the efficacy of Hologene 5, a graft consisting of cultured transgenic keratinocytes and epidermal stem cells and meant to combine cell and gene therapy for the treatment of LAMB3-related JEB. Autologous clonogenic keratinocytes will be isolated from patients' skin biopsies, genetically corrected with a gamma-retroviral vector (γRV) carrying the full-length human LAMB3 cDNA and plated onto a fibrin support (144cm2). The transgenic epidermis will be transplanted onto surgically prepared selected skin areas of at least six JEB patients (four pediatric and two adults). Evaluation of clinical efficacy will include, as primary endpoint, a combination of clinical parameters, such as percentage of re-epithelialization, cellular, molecular, and functional parameters, mechanical stress tests, and patient-reported outcome (PRO), up to 12months after transplantation. Safety and further efficacy endpoints will also be assessed during the clinical trial and for additional 15years in an interventional non-pharmacological follow-up study. If successful, this clinical trial would provide a therapeutic option for skin lesions of JEB patients with LAMB3 mutations and pave the way to a combined cell and gene therapy platform tackling other forms of EB and different genodermatoses. Clinical Trial Registration: EudraCT Number: 2018-000261-36.

8.
Stem Cell Res Ther ; 12(1): 341, 2021 06 10.
Artículo en Inglés | MEDLINE | ID: mdl-34112252

RESUMEN

BACKGROUND: Epidermal stem cells (EpSCs) play a vital role in wound healing and skin renewal. Although biomaterial scaffolds have been used for transplantation of EpSCs in wound healing, the ex vivo differentiation of EpSCs limits their application. METHODS: To inhibit the differentiation of EpSCs and maintain their stemness, we developed an electrospun polycaprolactone (PCL)+cellulose acetate (CA) micro/nanofiber for the culture and transplantation of EpSCs. The modulation effect on EpSCs of the scaffold and the underlying mechanism were explored. Liquid chromatography-tandem mass spectrometry for label-free quantitative proteomics was used to analyze proteomic changes in EpSCs cultured on scaffolds. In addition, the role of transplanted undifferentiated EpSCs in wound healing was also studied. RESULTS: In this study, we found that the PCL+CA micro/nanofiber scaffold can inhibit the differentiation of EpSCs through YAP activation-mediated inhibition of the Notch signaling pathway. Significantly differentially expressed proteomics was observed in EpSCs cultured on scaffolds and IV collagen-coated culture dishes. Importantly, differential expression levels of ribosome-related proteins and metabolic pathway-related proteins were detected. Moreover, undifferentiated EpSCs transplanted with the PCL+CA scaffold can promote wound healing through the activation of the Notch signaling pathway in rat full-thickness skin defect models. CONCLUSIONS: Overall, our study demonstrated the role of the PCL+CA micro-nanofiber scaffold in maintaining the stemness of EpSCs for wound healing, which can be helpful for the development of EpSCs maintaining scaffolds and exploration of interactions between biomaterials and EpSCs.


Asunto(s)
Nanofibras , Animales , Biomimética , Poliésteres , Proteómica , Ratas , Transducción de Señal , Células Madre , Andamios del Tejido , Cicatrización de Heridas
9.
Front Genet ; 12: 658295, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995490

RESUMEN

Epidermolysis bullosa (EB) is a genodermatosis, characterized by the formation of extended blisters and lesions on the skin and mucous membranes upon minimal mechanical trauma. The disease is caused by mutations in genes encoding proteins that are essential for skin stability. Functional impairment, reduction, or absence of one of these proteins results in skin fragility due to reduced connectivity between dermis and epidermis. Currently, gene therapy represents the only treatment option with the potential to cure this severe blistering skin disease. Two promising forms of gene therapy are potentially feasible for EB: gene replacement and genome editing. While genome editing for genodermatoses remains at the preclinical stage, gene replacement approaches are clinically advanced and have been applied already to a small number of patients with junctional and dystrophic forms of EB. Here, the viral transduction of the "wild-type" transgene into skin stem cells, followed by autologous grafting of corrected epidermal sheets, led to the regeneration of stable skin. Recent developments regarding designer nuclease-based gene editing strategies enable the establishment of alternative options to restore the gene function in genodermatoses. This is particularly true in cases wherein genetic constellation hinders gene therapy-based gene replacement.

10.
Biomolecules ; 11(5)2021 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-33946143

RESUMEN

Adult stem cells (ASCs) are the undifferentiated cells that possess self-renewal and differentiation abilities. They are present in all major organ systems of the body and are uniquely reserved there during development for tissue maintenance during homeostasis, injury, and infection. They do so by promptly modulating the dynamics of proliferation, differentiation, survival, and migration. Any imbalance in these processes may result in regeneration failure or developing cancer. Hence, the dynamics of these various behaviors of ASCs need to always be precisely controlled. Several genetic and epigenetic factors have been demonstrated to be involved in tightly regulating the proliferation, differentiation, and self-renewal of ASCs. Understanding these mechanisms is of great importance, given the role of stem cells in regenerative medicine. Investigations on various animal models have played a significant part in enriching our knowledge and giving In Vivo in-sight into such ASCs regulatory mechanisms. In this review, we have discussed the recent In Vivo studies demonstrating the role of various genetic factors in regulating dynamics of different ASCs viz. intestinal stem cells (ISCs), neural stem cells (NSCs), hematopoietic stem cells (HSCs), and epidermal stem cells (Ep-SCs).


Asunto(s)
Células Madre Adultas/fisiología , Diferenciación Celular , Proliferación Celular , Regulación de la Expresión Génica , Transducción de Señal , Animales , Movimiento Celular , Células Madre Hematopoyéticas , Humanos , Modelos Animales , Células-Madre Neurales
11.
Exp Dermatol ; 30(1): 62-67, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32970880

RESUMEN

Type XVII collagen (COL17) is a transmembrane protein expressed in the basal epidermis. COL17 serves as a niche for epidermal stem cells, and although its reduction has been implicated in altering cell polarity and ageing of the epidermis, it is unknown how COL17 affects epidermal cell polarity. Here, we uncovered COL17 as a binding partner of the aPKC-PAR complex, which is a key regulating factor of cell polarity. Immunoprecipitation-immunoblot assay and protein-protein binding assay revealed that COL17 interacts with aPKC and PAR3. COL17 deficiency or epidermis-specific aPKCλ deletion destabilized PAR3 distribution in the epidermis, while aPKCζ knockout did not. Asymmetrical cell division was pronounced in COL17-null neonatal paw epidermis. These results show that COL17 is pivotal for maintaining epidermal cell polarity. Our study highlights the previously unrecognized role of COL17 in the basal keratinocytes.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Autoantígenos/metabolismo , Proteínas de Ciclo Celular/metabolismo , Polaridad Celular , Epidermis/metabolismo , Colágenos no Fibrilares/metabolismo , Proteína Quinasa C/metabolismo , Animales , Autoantígenos/genética , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Colágenos no Fibrilares/genética , Isoformas de Proteínas/metabolismo , Colágeno Tipo XVII
12.
J Tissue Eng ; 11: 2041731420972310, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33224464

RESUMEN

Biomaterial scaffolds are increasingly being used to drive tissue regeneration. The limited success so far in human tissues rebuilding and therapy application may be due to inadequacy of the functionality biomaterial scaffold. We developed a new decellularized method to obtain complete anatomical skin biomatrix scaffold in situ with extracellular matrix (ECM) architecture preserved, in this study. We described a skin scaffold map by integrated proteomics and systematically analyzed the interaction between ECM proteins and epidermal cells in skin microenvironment on this basis. They were used to quantify structure and function of the skin's Matrisome, comprised of core ECM components and ECM-associated soluble signals that are key regulators of epidermal development. We especially revealed that ECM played a role in determining the fate of epidermal stem cells through hemidesmosome components. These concepts not only bring us a new understanding of the role of the skin ECM niche, they also provide an attractive combinational strategy based on tissue engineering principles with skin biomatrix scaffold materials for the acceleration and enhancement of tissue regeneration.

13.
Antioxidants (Basel) ; 9(10)2020 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-33036398

RESUMEN

Antioxidants may modulate the microenvironment of epidermal stem cells by reducing the production of reactive oxygen species or by regulating the expression of extracellular matrix protein. The extracellular membrane is an important component of the stem cell niche, and microRNAs regulate extracellular membrane-mediated basal keratinocyte proliferation. In this narrative review, we will discuss several antioxidants such as ascorbic acid, plant extracts, peptides and hyaluronic acid, and their effect on the epidermal stem cell niche and the proliferative potential of interfollicular epidermal stem cells in 3D skin equivalent models.

14.
Stem Cell Res Ther ; 11(1): 415, 2020 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-32967725

RESUMEN

Millions suffer from skin diseases. Functional interfollicular epidermal stem cells are needed in skin therapy or drug screening in vitro. We obtained functional interfollicular epidermal stem cells with intact stemness and cell junctions by treating them with Wnt3a. Moreover, epidermal stem cell-derived extracellular vesicles were useful in epidermal cell growth. Finally, functional epidermal 3D organoids with polarity were cultured using Wnt3a and the supernatant derived from interfollicular epidermal stem cells and fresh medium in a 1:1 ratio. These results provide novel directions for the improvement of skin organoids and their potential in clinical application.


Asunto(s)
Vesículas Extracelulares , Vía de Señalización Wnt , Células Epidérmicas , Epidermis , Células Madre
15.
Acta Biochim Biophys Sin (Shanghai) ; 52(10): 1102-1110, 2020 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-32840291

RESUMEN

Skin epidermal stem cells (EpSCs) play an important role in wound healing. Quercetin is a phytoestrogen reported to accelerate skin wound healing, but its effect on EpSCs is unknown. In this study, we investigated the effect of quercetin on human EpSC proliferation and explored the underlying mechanisms. We found that quercetin at 0.1~1 µM significantly promoted EpSC proliferation and increased the number of cells in S phase. The pro-proliferative effect of quercetin on EpSCs was confirmed in cultured human skin tissue. Mechanistic studies showed that quercetin significantly upregulated the expressions of ß-catenin, c-Myc, and cyclins A2 and E1. Inhibitor for ß-catenin or c-Myc significantly inhibited quercetin-induced EpSC proliferation. The ß-catenin inhibitor XAV-939 suppressed quercetin-induced expressions of ß-catenin, c-Myc, and cyclins A2 and E1. The c-Myc inhibitor 10058-F4 inhibited the upregulation of c-Myc and cyclin A2 by quercetin. Pretreatment of EpSCs with estrogen receptor (ER) antagonist ICI182780, but not the G protein-coupled ER1 antagonist G15, reversed quercetin-induced cell proliferation and upregulation of ß-catenin, c-Myc, and cyclin A2. Collectively, these results indicate that quercetin promotes EpSC proliferation through ER-mediated activation of ß-catenin/c-Myc/cyclinA2 signaling pathway and ER-independent upregulation of cyclin E1 and that quercetin may accelerate skin wound healing through promoting EpSC proliferation. As EpSCs are used not only in clinic to treat skin wounds but also as seed cells in skin tissue engineering, quercetin is a useful reagent to expand EpSCs for basic research, skin wound treatment, and skin tissue engineering.


Asunto(s)
Células Epidérmicas/metabolismo , Quercetina/farmacología , Transducción de Señal/efectos de los fármacos , Células Madre/metabolismo , Adulto , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Ciclina A2/metabolismo , Ciclina E/metabolismo , Células Epidérmicas/efectos de los fármacos , Humanos , Masculino , Proteínas Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Receptores de Estrógenos/metabolismo , Piel/citología , Piel/efectos de los fármacos , Piel/metabolismo , Piel/patología , Células Madre/efectos de los fármacos , beta Catenina/metabolismo
16.
Stem Cell Res Ther ; 11(1): 232, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32527289

RESUMEN

The skin, which serves as the first barrier of the human body, is particularly susceptible to exogenous injuries. Skin wounds, including acute burns and chronic non-healing ulcers, are commonly observed in clinics. Healing of skin wounds is a complex process, consisting of infiltration of inflammatory cells, cellular proliferation, and tissue remodeling phases, which restore the integrity and functions of the skin. Epithelialization is involved in wound healing through re-establishing an intact keratinocyte layer. Epidermal stem cells are indispensable for epithelialization, and they are regulated by multiple proinflammatory cytokines or growth factors. In this review, we summarize recent advances in the effect of these cytokines on migration, proliferation, and differentiation processes of epidermal stem cells. We also introduce promising therapeutic strategies targeting epidermal stem cells or related proinflammatory cytokines for patients with skin wounds.


Asunto(s)
Citocinas , Repitelización , Células Epidérmicas , Humanos , Queratinocitos , Células Madre , Cicatrización de Heridas
17.
Cells ; 9(1)2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31936599

RESUMEN

The stem cells located in the hair follicle bulge area are critical for skin regeneration and repair. To date, little is known about the evolution of the transcriptome of these cells across time. Here, we have combined genome-wide expression analyses and a variety of in silico tools to determine the age-dependent evolution of the transcriptome of those cells. Our results reveal that the transcriptome of skin stem cells fluctuates extensively along the lifespan of mice. The use of both unbiased and pathway-centered in silico approaches has also enabled the identification of biological programs specifically regulated at those specific time-points. It has also unveiled hubs of highly transcriptionally interconnected genes and transcriptional factors potentially located at the core of those age-specific changes.


Asunto(s)
Envejecimiento/genética , Simulación por Computador , Evolución Molecular , Piel/citología , Células Madre/metabolismo , Transcriptoma/genética , Animales , Secuencia de Bases , Sitios de Unión , Masculino , Ratones Endogámicos C57BL , Transducción de Señal/genética , Factores de Tiempo , Factores de Transcripción/metabolismo , Transcripción Genética
18.
J Comput Biol ; 27(5): 769-778, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31502860

RESUMEN

We aimed to identify differentially expressed genes (DEGs) in epidermal stem cells (epiSCs) in response to high fat diet (HFD). DEGs were identified by time-series analysis of the gene expression profile (GSE84510) in Gene Expression Omnibus (GEO) database. Functions and pathways affected by HFD were identified by functional annotation of DEGs. Key factors responding to HFD was identified by protein-protein interaction (PPI) network analysis. Two groups of genes with the same tendency in response to HFD were identified. ECM-related processes and PI3K pathway were altered in the early stage of obesity. A PPI network was constructed to delineate the interactions among proteins encoded by DEGs and ICAM1 and RELA were key epiSC factors respond to HFD. Our studies may provide valuable insights into the molecular mechanisms underlying how obesity affects the functions of epiSC.


Asunto(s)
Molécula 1 de Adhesión Intercelular/genética , Obesidad/genética , Mapas de Interacción de Proteínas/genética , Células Madre/metabolismo , Factor de Transcripción ReIA/genética , Animales , Biología Computacional , Dieta Alta en Grasa/efectos adversos , Células Epidérmicas/metabolismo , Regulación de la Expresión Génica/genética , Ontología de Genes , Redes Reguladoras de Genes/genética , Humanos , Ratones , Obesidad/patología , Transcriptoma/genética
19.
Biomedicine (Taipei) ; 10(2): 5-11, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33854915

RESUMEN

BACKGROUND: Burn wounds are one of the causes of cutaneous injury that involve both epidermal and dermal layers of skin. Silver sulfadiazine (SSD) has been widely used to treat burn wounds, however recent studies have found the treatment to have some drawbacks, such as cellular toxicity effects. Cutaneous wound regeneration is known to start from the basal layer of the epidermal epithelial cells, which are enriched with highly proliferative cells. Keratin-19 (K19) is one of the epidermal stem cell biomarkers found in the skin. This study aims to explore the expression of K19 in burn wound tissue and to investigate the effect of SSD on its expression. METHODS: We created a burn wound model in Sprague Dawley rats and randomly divided them into control and SSD groups. Wound closure was evaluated (visitrak) overtime series followed by histological evaluation of K19 expression in the wound tissue (immunohistochemistry staining). RESULTS: Our model successfully represents full-thickness damage caused by a burn wound. The SSD group showed a faster reduction of wound surface area (wound closure) compared to the control group with the peak at day 18 post wounding (p < 0.05). K19 expression was found in both groups and was distributed on epidermal layers, hair follicles and dermis of granulation tissue showing similar patterns. CONCLUSION: Topical application of SSD on burn wounds showed superiority in wound closure and is likely to have no harmful effect on epidermal stem cells. However, further study is required to investigate the effect of silver species on cell viability and toxicity effects during long term treatment.

20.
Braz. j. med. biol. res ; 53(11): e10009, 2020. tab, graf
Artículo en Inglés | LILACS, Coleciona SUS | ID: biblio-1132495

RESUMEN

The epidermis, the outermost layer of the skin, is the first barrier that comes into contact with the external environment. It plays an important role in resisting the invasion of harmful substances and microbial infections. The skin changes with age and external environmental factors. This study aimed to investigate epidermal stem cells during the process of aging. This study enrolled 9 volunteers with benign pigmented nevus for clinical dermatologic surgery. The phenotypes associated with skin aging changes such as skin wrinkles and elasticity of the unexposed/healthy parts near benign pigmented skin were measured, and epidermal stem cells from this region were isolated for transcriptome sequencing. The results showed that epidermal stem cells could be obtained by magnetic activated cell sorting (MACS) with high purity. Results of the transcriptome sequencing revealed that aquaporin (AQP)5 significantly decreased in the epidermal stem cells with age, and further functional experiments revealed that AQP5 could promote the proliferation and dedifferentiation of HaCaT, but did not influence cell apoptosis. In summary, AQP5 regulated the proliferation and differentiation of epidermal stem cells in skin aging, and it may play an important role in the balance of proliferation and differentiation. However, further studies are needed to determine the mechanism by which AQP5 regulates the proliferation and differentiation of epidermal skin cells in aging.


Asunto(s)
Humanos , Envejecimiento de la Piel , Acuaporina 5/metabolismo , Células Madre , Diferenciación Celular , Proliferación Celular , Epidermis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA