Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.683
Filter
1.
Sci Rep ; 14(1): 10400, 2024 05 06.
Article in English | MEDLINE | ID: mdl-38710823

ABSTRACT

Without the protective shielding of Earth's atmosphere, astronauts face higher doses of ionizing radiation in space, causing serious health concerns. Highly charged and high energy (HZE) particles are particularly effective in causing complex and difficult-to-repair DNA double-strand breaks compared to low linear energy transfer. Additionally, chronic cortisol exposure during spaceflight raises further concerns, although its specific impact on DNA damage and repair remains unknown. This study explorers the effect of different radiation qualities (photons, protons, carbon, and iron ions) on the DNA damage and repair of cortisol-conditioned primary human dermal fibroblasts. Besides, we introduce a new measure, the Foci-Integrated Damage Complexity Score (FIDCS), to assess DNA damage complexity by analyzing focus area and fluorescent intensity. Our results show that the FIDCS captured the DNA damage induced by different radiation qualities better than counting the number of foci, as traditionally done. Besides, using this measure, we were able to identify differences in DNA damage between cortisol-exposed cells and controls. This suggests that, besides measuring the total number of foci, considering the complexity of the DNA damage by means of the FIDCS can provide additional and, in our case, improved information when comparing different radiation qualities.


Subject(s)
DNA Breaks, Double-Stranded , DNA Repair , Fibroblasts , Hydrocortisone , Humans , Fibroblasts/radiation effects , Fibroblasts/metabolism , DNA Breaks, Double-Stranded/radiation effects , Hydrocortisone/pharmacology , Radiation, Ionizing , Cells, Cultured , DNA Damage
2.
J Drugs Dermatol ; 23(5): 366-375, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38709706

ABSTRACT

OBJECTIVE:   This study aimed to investigate the ultraviolet (UV) protection/repair benefits of a patented Amino Acid Complex (AAComplex). METHODS: I) AAComplex was incubated with dermal fibroblasts, with/without UVA, and collagen I was measured with a GlasBoxPlus device. II) A lotion, with/without AAComplex (1%) was applied topically to skin explants, following UVA irradiation, and quantified for health-related biomarkers (TNFalpha, histamine, and MMP-1). III) A broad spectrum sunscreen with SPF 46 and a skincare serum containing AAComplex (2%) were assessed using epidermal equivalents, in the presence of UV irradiation, for effects on IL-1alpha, thymine dimers, Ki-67, filaggrin and Nrf2. RESULTS: I) Collagen I synthesis in dermal fibroblasts was significantly decreased after UVA compared to without UV. The presence of AAComplex prevented this decrease. II) UVA irradiation of skin explants increased histamine, TNFα, and MMP-1. Hydrocortisone aceponate cream significantly decreases all 3 biomarkers. AAComplex contained lotion also significantly decreased all 3 biomarkers, the no AAComplex control lotion only reduced histamine. III) With the regimen of sunscreen + AAComplex contained skincare serum, the significant reduction in IL-1alpha was observed along with a complete recovery of Ki-67 and stimulation of filaggrin and Nrf2T. No thymine dimer positive cell was observed indicating the most positive skin impact from the regiment.  Conclusion: This research using different human skin models demonstrated that AAComplex can provide protection and damage repair caused by UV, at the ingredient level also when formulated in a serum or lotion formula. Skin may be best protected from UV damage when the regimen is used.   J Drugs Dermatol. 2024;23(5):366-375. doi:10.36849/JDD.7916.


Subject(s)
Fibroblasts , Filaggrin Proteins , Matrix Metalloproteinase 1 , NF-E2-Related Factor 2 , Tumor Necrosis Factor-alpha , Ultraviolet Rays , Humans , Ultraviolet Rays/adverse effects , Fibroblasts/drug effects , Fibroblasts/radiation effects , Fibroblasts/metabolism , Matrix Metalloproteinase 1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Skin/radiation effects , Skin/drug effects , Skin/metabolism , Sunscreening Agents/administration & dosage , Sunscreening Agents/chemistry , Sunscreening Agents/pharmacology , Amino Acids/administration & dosage , Amino Acids/pharmacology , Amino Acids/chemistry , Interleukin-1alpha/metabolism , Histamine/blood , Skin Cream/administration & dosage , Biomarkers/metabolism , Collagen Type I , Intermediate Filament Proteins/metabolism , Ki-67 Antigen/metabolism , Pyrimidine Dimers , Cells, Cultured
3.
Aging (Albany NY) ; 16(9): 7511-7522, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38700499

ABSTRACT

The battle against the COVID-19 pandemic has spurred a heightened state of vigilance in global healthcare, leading to the proliferation of diverse sanitization methods. Among these approaches, germicidal lamps utilizing ultraviolet (UV) rays, particularly UV-C (wavelength ranging from 280 to 100 nm), have gained prominence for domestic use. These light-emitting diode (LED) lamps are designed to sanitize the air, objects, and surfaces. However, the prevailing concern is that these UV lamps are often introduced into the market without adequate accompanying information to ensure their safe utilization. Importantly, exposure to absorbed UV light can potentially trigger adverse biological responses, encompassing cell death and senescence. Our research encompassed a series of investigations aimed at comprehending the biological repercussions of UV-C radiation exposure from readily available domestic lamps. Our focus centered on epithelial retinal cells, keratinocytes, and fibroblasts, components of the skin and ocular targets frequently exposed to UV irradiation. Our findings underscore the potential harm associated with even brief exposure to UV, leading to irreversible and detrimental alterations in both skin cells and retinal cells of the eye. Notably, epithelial retinal cells exhibited heightened sensitivity, marked by substantial apoptosis. In contrast, keratinocytes demonstrated resilience to apoptosis even at elevated UV doses, though they were prone to senescence. Meanwhile, fibroblasts displayed a gradual amplification of both senescence and apoptosis as radiation doses escalated. In summary, despite the potential benefits offered by UV-C in deactivating pathogens like SARS-CoV-2, it remains evident that the concurrent risks posed by UV-C to human health cannot be ignored.


Subject(s)
Apoptosis , COVID-19 , Cellular Senescence , SARS-CoV-2 , Ultraviolet Rays , Ultraviolet Rays/adverse effects , Apoptosis/radiation effects , Humans , Cellular Senescence/radiation effects , SARS-CoV-2/radiation effects , Keratinocytes/radiation effects , Fibroblasts/radiation effects
5.
Sci Rep ; 14(1): 9444, 2024 04 24.
Article in English | MEDLINE | ID: mdl-38658667

ABSTRACT

One of the biggest challenges in tissue engineering and regenerative medicine is to ensure oxygen supply of cells in the (temporary) absence of vasculature. With the vision to exploit photosynthetic oxygen production by microalgae, co-cultivated in close vicinity to oxygen-consuming mammalian cells, we are searching for culture conditions that are compatible for both sides. Herein, we investigated the impact of long-term illumination on mammalian cells which is essential to enable photosynthesis by microalgae: four different cell types-primary human fibroblasts, dental pulp stem cells, and osteoblasts as well as the murine beta-cell line INS-1-were continuously exposed to warm white light, red or blue light over seven days. We observed that illumination with red light has no adverse effects on viability, metabolic activity and growth of the cells whereas exposure to white light has deleterious effects that can be attributed to its blue light portion. Quantification of intracellular glutathione did not reveal a clear correlation of this effect with an enhanced production of reactive oxygen species. Finally, our data indicate that the cytotoxic effect of short-wavelength light is predominantly a direct effect of cell illumination; photo-induced changes in the cell culture media play only a minor role.


Subject(s)
Fibroblasts , Light , Reactive Oxygen Species , Humans , Animals , Fibroblasts/metabolism , Fibroblasts/radiation effects , Fibroblasts/cytology , Mice , Reactive Oxygen Species/metabolism , Cell Survival/radiation effects , Dental Pulp/cytology , Dental Pulp/radiation effects , Osteoblasts/metabolism , Osteoblasts/radiation effects , Osteoblasts/cytology , Cells, Cultured , Cell Line , Stem Cells/metabolism , Stem Cells/radiation effects , Stem Cells/cytology , Glutathione/metabolism
6.
Radiother Oncol ; 194: 110198, 2024 May.
Article in English | MEDLINE | ID: mdl-38438016

ABSTRACT

BACKGROUND AND PURPOSE: Ionizing radiation (IR) induces DNA double-strand breaks (DSBs), leading to micronuclei formation, which has emerged as a key mediator of inflammatory responses after IR. This study aimed to investigate the signaling cascade in inflammatory gene expression using fibroblasts harboring DNA damage response deficiency after exposure to IR. MATERIALS AND METHODS: Micronuclei formation was examined in human dermal fibroblasts derived from patients with deficiencies in ATM, ATR, MRE11, XLF, Artemis, or BRCA2 after IR. RNA-sequencing analysis was performed to assess gene expression, pathway mapping, and the balance of transcriptional activity using the transcription factor-based downstream gene expression mapping (TDEM) method developed in this study. RESULTS: Deficiencies in ATM, ATR, or MRE11 led to increased micronuclei formation after IR compared to normal cells. RNA-seq analysis revealed significant upregulation of inflammatory expression in cells deficient in ATM, ATR, or MRE11 following IR. Pathway mapping analysis identified the upregulation of RIG-I, MDA-5, IRF7, IL6, and interferon stimulated gene expression after IR. These changes were pronounced in cells deficient in ATM, ATR, or MRE11. TDEM analysis suggested the differential activation of STAT1/3-pathway between ATM and ATR deficiency. CONCLUSION: Enhanced micronuclei formation upon ATM, ATR, or MRE11 deficiency activated the cGAS/STING, RIG-I-MDA-5-IRF7-IL6 pathway, resulting in its downstream interferon stimulated gene expression following exposure to IR. Our study provides comprehensive information regarding the status of inflammation-related gene expression under DSB repair deficiency after IR. The generated dataset may be useful in developing functional biomarkers to accurately identify patients sensitive to radiotherapy.


Subject(s)
Ataxia Telangiectasia Mutated Proteins , Fibroblasts , Radiation, Ionizing , Signal Transduction , Humans , Fibroblasts/radiation effects , Fibroblasts/metabolism , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/deficiency , Ataxia Telangiectasia Mutated Proteins/metabolism , MRE11 Homologue Protein/genetics , Inflammation/etiology , DNA Breaks, Double-Stranded
7.
J Cosmet Dermatol ; 23(6): 2256-2269, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38497297

ABSTRACT

BACKGROUND: Research has demonstrated the anti-photoaging properties of glabridin and bakuchiol. METHODS: The impact of glabridin, glabridin + bakuchiol, and bakuchiol on the levels of tumor necrosis factor alpha (TNF-α) and interleukin-1 beta (IL-1ß) in mice skin fibroblasts was observed. Furthermore, we investigated the potential roles of fibronectin (FN), interferon-γ (IFN-γ), interleukin-22 (IL-22), and transforming growth factor-ß (TGF-ß) in the tissues, and evaluated their impact on the enzymatic levels in the skin. In conjunction with transcriptomic analysis, metabolomic profiling, and network pharmacology, all samples underwent comprehensive metabolomic and principal component analysis. The Venny2.1 method was utilized to identify variances in shared metabolites between the treatment group and the UVB group, as well as between the UVB group and the control group. Subsequently, a cluster heat map was generated to forecast and analyze metabolic pathways and targets. RESULTS: The outcomes from the hematoxylin and eosin and toluidine blue staining revealed that glabridin and bakuchiol markedly decreased dermal thickness and suppressed mast cell infiltration in photoaged mice. Immunohistochemistry and Elisa analysis revealed that glabridin and bakuchiol effectively attenuated the levels of pro-inflammatory factors, including IL-1ß, tumor necrosis factor-α, IL-22, and IFN-γ. Furthermore, an increase in the levels of anti-inflammatory factors such as FN and TGF-ß was also observed. The determination of the contents of superoxide dismutase, hydroxypropyltransferase and malondialdehyde in mice dorsal skin revealed that glabridin and bakuchiol not only elevated the levels of superoxide dismutase and hydroxyproline, but also reduced malondialdehyde content. Due to the limited number of shared differential metabolites exclusively within Kyoto Encyclopedia of Genes and Genomes, comprehensive pathway enrichment analysis was not feasible. CONCLUSION: This study demonstrates that glabridin and bakuchiol effectively impede photoaging and alleviate skin inflammation in mice.


Subject(s)
Isoflavones , Phenols , Skin Aging , Skin , Ultraviolet Rays , Animals , Phenols/pharmacology , Mice , Skin Aging/drug effects , Skin Aging/radiation effects , Ultraviolet Rays/adverse effects , Isoflavones/pharmacology , Skin/drug effects , Skin/radiation effects , Skin/pathology , Skin/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Interleukin-1beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Interleukins/metabolism , Fibronectins/metabolism , Interleukin-22 , Female , Interferon-gamma/metabolism , Transforming Growth Factor beta/metabolism
8.
J Cosmet Dermatol ; 23(5): 1850-1861, 2024 May.
Article in English | MEDLINE | ID: mdl-38327116

ABSTRACT

BACKGROUND: The oxidative stress induced by ultraviolet (UV) radiation is a pivotal factor in skin aging and can even contribute to the development of skin cancer. AIM: This study explored the antioxidant effect and mechanism of water-soluble intracellular extract (WIE) of Desmodesmus sp.YT (YT), aiming to develop a natural antioxidant suitable for incorporation into cosmetics. METHODS: The study evaluated the scavenging capacity of YT-WIE against free radicals and assessed its impact on human skin fibroblasts (HSF) cell viability and UV resistance using Cell Counting Kit-8 (CCK-8). Transcriptome sequencing was employed to elucidate the mechanism of action, while RT-qPCR and western blot were used to validate the expression of key genes. RESULTS: YT-WIE displayed robust antioxidant activity, demonstrating potent scavenging abilities against 2,2-diphenyl-1-picrylhydrazyl (DPPH; IC50 = 0.55 mg mL-1), 2,2'-Azino-bis (3 ethylbenzothiazoline-6-sulfonic acid; ABTS; IC50 = 3.11 mg mL-1), Hydroxyl (·OH; IC50 = 2.21 mg mL-1), and Superoxide anion (O2 •-; IC50 = 0.98 mg mL-1). Furthermore, compared to the control group, the YT-WIE group exhibited an 89.30% enhancement in HSF viability and a 44.63% increase in survival rate post-UV irradiation. Significant upregulation of antioxidant genes (GCLC, GCLM, TXNRD1, HMOX1, NQO1) was observed with YT-WIE treatment at 400 µg mL-1, with fold increases ranging from 1.13 to 5.85 times. CONCLUSION: YT-WIE demonstrated considerable potential as an antioxidant, shielding human cells from undue oxidative stress triggered by external stimuli such as UV radiation. This suggests its promising application in cosmetics antioxidants.


Subject(s)
Antioxidants , Fibroblasts , Oxidative Stress , Skin , Ultraviolet Rays , Humans , Fibroblasts/radiation effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Ultraviolet Rays/adverse effects , Antioxidants/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Skin/radiation effects , Skin/drug effects , Skin/cytology , Cell Survival/drug effects , Cell Survival/radiation effects , Skin Aging/drug effects , Skin Aging/radiation effects , Water , Cells, Cultured
9.
Int J Radiat Oncol Biol Phys ; 118(4): 1105-1109, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-37956734

ABSTRACT

PURPOSE: This study aimed to demonstrate for the first time the possibility of irradiating biological cells with gray (Gy)-scale doses delivered over single bursts of picosecond-scale electron beams, resulting in unprecedented dose rates of 1010 to 1011 Gy/s. METHODS AND MATERIALS: Cancer stem cells and human skin fibroblasts were irradiated with MeV-scale electron beams from a laser-driven source. Doses up to 3 Gy per pulse with a high spatial uniformity (coefficient of variance, 3%-6%) and within a timescale range of 10 to 20 picoseconds were delivered. Doses were characterized during irradiation and were found to be in agreement with Monte Carlo simulations. Cell survival and DNA double-strand break repair dynamics were studied for both cell lines using clonogenic assay and 53BP1 foci formation. The results were compared with reference x-rays at a dose rate of 0.49 Gy/min. RESULTS: Results from clonogenic assays of both cell lines up to 3 Gy were well fitted by a linear quadratic model with α = (0.68 ± 0.08) Gy-1 and ß = (0.01 ± 0.01) Gy-2 for human skin fibroblasts and α = (0.51 ± 0.14) Gy-1 and ß = (0.01 ± 0.01) Gy-2 for cancer stem cells. Compared with irradiation at 0.49 Gy/min, our experimental results indicate no statistically significant difference in cell survival rate for doses up to 3 Gy despite a significant increase in the α parameter, which may reflect more complex damage. Foci measurements showed no significant difference between irradiation at 1011 Gy/s and at 0.49 Gy/min. CONCLUSIONS: This study demonstrates the possibility of performing radiobiological studies with picosecond-scale laser-generated electron beams at ultrahigh dose rates of 1010 to1011 Gy/s. Preliminary results indicate, within statistical uncertainties, a significant increase of the α parameter, a possible indication of more complex damage induced by a higher density of ionizing tracks.


Subject(s)
Electrons , Neoplasms , Humans , Dose-Response Relationship, Radiation , DNA Repair , Fibroblasts/radiation effects , Neoplastic Stem Cells , Neoplasms/metabolism
10.
J Biophotonics ; 17(2): e202300166, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37975254

ABSTRACT

We investigated the effects of low-level Er:YAG laser irradiation on proliferation and alternations in early gene expression of gingival fibroblasts. Mice primary gingival fibroblasts were irradiated with an Er:YAG laser (1.8, 3.9, and 5.8 J/cm2 ). Irradiation at 3.9 J/cm2 promoted cell proliferation without significant changes in lactate dehydrogenase or Hspa1a expression. Three hours after irradiation at 3.9 J/cm2 , the Fn1 expression level was significantly increased. RNA-seq identified 15 differentially expressed genes between irradiated and non-irradiated cells, some of which belonged to immediate early genes (IEGs). Kyoto Encyclopedia of Genes and Genomes pathway analysis indicated MAPK pathway enhancement, and gene set enrichment analysis showed enrichment in the TGF-ß signaling gene set. Enhanced proliferation via laser irradiation disappeared upon inhibition of Dusp4, Dusp5, and Tgfr1 expression. Low-level Er:YAG laser irradiation, especially at 3.9 J/cm2 without a major temperature elevation, enhanced fibroblast proliferation, via TGF-ß and the MAPK signaling pathway following IEG expression.


Subject(s)
Lasers, Solid-State , Mice , Animals , Maxilla , Cell Proliferation/radiation effects , Transforming Growth Factor beta , Fibroblasts/radiation effects , Gene Expression
11.
Photochem Photobiol ; 100(1): 225-232, 2024.
Article in English | MEDLINE | ID: mdl-37254280

ABSTRACT

There are limited data on comparison of pulsed and continuous wave in photobiomodulation therapy (PBM). This study aimed to investigate the effect of PBM with 980 nm laser in pulsed and continuous wave on the proliferation and migration of human gingival fibroblasts (HGF) cells. Cultured HGF were divided into three main groups: (1) irradiated in pulsed mode (frequencies of 50 and 25 KHz; energy densities of 3 and 5 J/cm2 ), (2) irradiated in continuous mode (energy densities of 3.2 and 5.2 J/cm2 ), and (3) no irradiation as control group. HGF proliferation rate was measured by MTT assay at 24, 48, and 72 h post irradiation. In addition, HGF migration rate was measured by scratch test at 24 h post PBM. At 24 h, the group received continuous irradiation at 5.2 J/cm2 showed significantly higher proliferation compared with the control group (p = 0.012). At 48 and 72 h, the groups received continuous, and 50 Hz pulsed irradiation at energy densities of 5.2 and 5 J/cm2 respectively, had significantly higher HGF proliferation rates compared to the control (p < 0.05). Only the continuous irradiations were effective in significant increase of the cell migration. In conclusion, continuous PBM at energy density of 5.2 J/cm2 showed promising effect on HGF proliferation and migration.


Subject(s)
Low-Level Light Therapy , Humans , Cell Proliferation/radiation effects , Cell Survival , Lasers , Fibroblasts/radiation effects
12.
Photochem Photobiol ; 100(1): 146-158, 2024.
Article in English | MEDLINE | ID: mdl-37477119

ABSTRACT

The effect of terahertz (THz) radiation has been studied in medicine. However, there is a lack of scientific information regarding its possible mutagenicity. Therefore, the present study aimed to assess the mutagenicity of 1.6 THz laser irradiation. The Ames test was conducted using five bacterial tester strains. The bacteria were subjected to (i) 1.6 THz laser irradiation at 3.8 mW/cm2 for 60 min using a tabletop THz pulse laser system, (ii) ultraviolet irradiation, (iii) treatment with positive control chemicals (positive control) or (iv) treatment with the solvent used in the positive control (negative control). After treatment, the bacterial suspensions were cultured on minimal glucose agar to determine the number of revertant colonies. In addition, the comet assay was performed using fibroblasts (V79) to assess possible DNA damage caused by the THz laser irradiation. The Ames test demonstrated that the THz laser irradiation did not increase the number of revertant colonies compared to that in the negative control group, whereas the ultraviolet irradiation and positive control treatment increased the number of revertant colonies. Thus, 1.6 THz laser irradiation is unlikely to be mutagenic. The comet assay additionally suggests that the THz laser irradiation unlikely induce cellular DNA damage.


Subject(s)
DNA Damage , Mutagens , Mutagens/toxicity , Comet Assay , Mutagenesis , Fibroblasts/radiation effects , Mutagenicity Tests
13.
J Biophotonics ; 17(2): e202300388, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38019097

ABSTRACT

Photobiomodulation (PBM) can be used to treat a range of conditions in dermatology. PBM refers to the changes induced by red (RL, 620-700 nm) and near-infrared (NIR, 700-1440 nm) light. Light radiation-induced DNA damage is a major contributor to aging and skin cancer. It is crucial to study the effects of PBM on DNA to ensure safety. Our lab previously demonstrated that RL (633 ± 6 nm) did not result in human dermal fibroblasts (HDFs) DNA damage. This study employed similar methods to investigate NIR effects. Commercially available LED-NIR (830 ± 5 nm) panels (66, 132, and 264 J/cm2 ) did not result in DNA damage measured by cyclobutane pyrimidine dimers and pyrimidine-6,4-pyrimidone photoproducts in HDFs compared to temperature-matched controls immediately, 3 h, and 24 h following irradiation and compared to positive and negative controls. This demonstrates that LED-NIR does not damage DNA in HDFs in vitro.


Subject(s)
DNA Damage , Skin , Humans , Skin/radiation effects , Infrared Rays , Fibroblasts/radiation effects , DNA
14.
Int J Radiat Oncol Biol Phys ; 118(1): 218-230, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37586613

ABSTRACT

PURPOSE: Radiation-induced pulmonary fibrosis (RIPF) is a common side effect of radiation therapy for thoracic tumors without effective prevention and treatment methods at present. The aim of this study was to explore whether glycyrrhetinic acid (GA) has a protective effect on RIPF and the underlying mechanism. METHODS AND MATERIALS: A RIPF mouse model administered GA was used to determine the effect of GA on RIPF. The cocultivation of regulatory T (Treg) cells with mouse lung epithelial-12 cells or mouse embryonic fibroblasts and intervention with GA or transforming growth factor-ß1 (TGF-ß1) inhibitor to block TGF-ß1 was conducted to study the mechanism by which GA alleviates RIPF. Furthermore, injection of Treg cells into GA-treated RIPF mice to upregulate TGF-ß1 levels was performed to verify the roles of TGF-ß1 and Treg cells. RESULTS: GA intervention improved the damage to lung tissue structure and collagen deposition and inhibited Treg cell infiltration, TGF-ß1 levels, epithelial mesenchymal transition (EMT), and myofibroblast (MFB) transformation in mice after irradiation. Treg cell-induced EMT and MFB transformation in vitro were prevented by GA, as well as a TGF-ß1 inhibitor, by decreasing TGF-ß1. Furthermore, reinfusion of Treg cells upregulated TGF-ß1 levels and exacerbated RIPF in GA-treated RIPF mice. CONCLUSIONS: GA can improve RIPF in mice, and the corresponding mechanisms may be related to the inhibition of TGF-ß1 secreted by Treg cells to induce EMT and MFB transformation. Therefore, GA may be a promising therapeutic candidate for the clinical treatment of RIPF.


Subject(s)
Glycyrrhetinic Acid , Lung Injury , Pulmonary Fibrosis , Radiation Injuries , Animals , Mice , Epithelial-Mesenchymal Transition , Fibroblasts/radiation effects , Glycyrrhetinic Acid/pharmacology , Lung/radiation effects , Lung Injury/pathology , Pulmonary Fibrosis/drug therapy , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/prevention & control , Radiation Injuries/pathology , T-Lymphocytes, Regulatory , Transforming Growth Factor beta1
15.
Arch Oral Biol ; 159: 105874, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38147800

ABSTRACT

OBJECTIVE: Photobiomodulation therapy (PBMT) has proven to reduce inflammation and pain and increase wound healing. Thus, the aim of this study was to analyze the effects of PBMT parameters on migration, proliferation, and gene expression after ionizing radiation and bacterial-induced stress in an in vitro study. DESIGN: Keratinocytes (HaCaT) and Fibroblasts (HGFs) were grown in DMEM with 10 % fetal bovine serum until stressful condition induction with lipopolysaccharide (LPS) of Escherichia coli (1 µg/mL), Porphyromonas gingivalis protein extract (5 µg/mL) and ionizing radiation (8 Gy). Low-laser irradiation (660 nm, 30 mW) was carried out in four sessions, with 6 h intervals, and energy density of 2, 3, 4, and 5 J/cm². Scratch assays, immunofluorescence, and RT-qPCR were performed. RESULTS: Treated fibroblasts and keratinocytes showed significant response in proliferation and migration after scratch assays (p < 0.05). Higher expressions of α-SMA in fibroblasts and F-actin in keratinocytes were observed in cells subjected to 3 J/cm². PI3K-pathway genes expression tended to enhance in fibroblasts, presenting a higher relative expression when compared to keratinocytes. In keratinocytes, PBMT groups demonstrated deregulated expression for all inflammatory cytokines' genes tested while fibroblasts presented a tendency to enhance those genes expression in a dose dependent way. CONCLUSIONS: The present study showed that delivering 660 nm, 30 mW was effective to stimulate cell migration, proliferation and to accelerate wound healing. PBMT can modulate cytokines and pathways involved in wound repair. The different energy densities delivering distinct responses in vitro highlights that understanding laser parameters is fundamental to improve treatment strategies.


Subject(s)
Low-Level Light Therapy , Phosphatidylinositol 3-Kinases , Keratinocytes , Fibroblasts/radiation effects , Cell Proliferation/radiation effects , Radiation, Ionizing
16.
Lasers Med Sci ; 39(1): 9, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38110767

ABSTRACT

An in vitro study was designed to evaluate the effects of photobiomodulation (PBM) with 915-nm diode laser on human gingival fibroblast (HGF) cells under hyperglycemic condition. The HGF cells were cultured in Dulbecco's modified eagle medium (DMEM) medium containing 30 mM glucose concentration for 48 h to mimic the hyperglycemic condition. Subsequently, the cells received three sessions of PBM (915 nm, continuous emission mode, 200 mW, energy density values of 3.2, 6, and 9.2 J/cm2). Twenty-four hours post-irradiation, cell proliferation, expression of interleukin 6 (IL-6), and vascular endothelial growth factor (VEGF) were assessed with MTT and real-time polymerase chain reaction (PCR) tests, respectively. Also, reactive oxygen species (ROS) production was measured using CM-H2DCFDA fluorimetry. No changes were detected in the cell proliferation rate between the high glucose control group and laser-treated cells, while VEGF and IL-6 gene expression levels increased significantly after PBM in the high glucose-treated cells group. ROS level was significantly decreased in the irradiated cells in high-glucose medium compared with the high glucose control group. Our study revealed the inductive role of 915-nm-mediated PBM on VEGF and the inflammatory response while concurrently reducing reactive oxygen species production in HGF cells in hyperglycemic conditions.


Subject(s)
Interleukin-6 , Low-Level Light Therapy , Humans , Interleukin-6/genetics , Vascular Endothelial Growth Factor A/genetics , Reactive Oxygen Species/metabolism , Blood Glucose , Fibroblasts/radiation effects , Cells, Cultured
17.
J Photochem Photobiol B ; 249: 112801, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37897855

ABSTRACT

Skin photoaging, resulting from prolonged exposure to sunlight, especially UVA rays, has been identified as a key contributor to age-related skin degeneration. However, the mechanism by which UVA radiation induces skin cell senescence has not been fully elucidated. In this investigation, bioinformatics technology was employed to identify SIRT6 as the core hub gene involved in the progression of skin photoaging. The study evinced that prolonged exposure of cutaneous fibroblasts to UVA radiation results in a marked reduction in the expression of SIRT6, both in vivo and in vitro. Knockdown of SIRT6 in skin fibroblasts resulted in the upregulation of genes associated with cellular aging, thereby exacerbating the effects of UVA radiation-induced photoaging. Conversely, overexpression of SIRT6 decreased the expression of cell aging-related genes, indicating that SIRT6 plays a role in the regulation of senescence in skin fibroblasts induced by UVA radiation. We proffer substantiation that overexpression of SIRT6 protects skin fibroblasts from UVA-induced oxidative stress by activating the NRF2/HO-1 signaling cascade. Moreover, SIRT6 overexpression also reduced UVA-induced type I collagen degradation by inhibiting NF-κB signaling cascade. In summary, our findings showed that overexpression of SIRT6 inhibits UVA-induced senescence phenotype and type I collagen degradation in skin fibroblasts by modulating the NRF2/HO-1 and NF-κB signaling pathways. And the regulation of these signaling pathways by SIRT6 may be achieved through its deacetylase activity. Therefore, SIRT6 is a novel and promising therapeutic target for skin aging related to age and UV.


Subject(s)
Sirtuins , Skin Aging , Skin Diseases , Humans , Collagen Type I/genetics , Collagen Type I/metabolism , Fibroblasts/radiation effects , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Signal Transduction , Sirtuins/genetics , Sirtuins/metabolism , Sirtuins/pharmacology , Skin/radiation effects , Ultraviolet Rays
18.
J Biol Chem ; 299(7): 104900, 2023 07.
Article in English | MEDLINE | ID: mdl-37301510

ABSTRACT

Nucleotide excision repair (NER) eliminates highly genotoxic solar UV-induced DNA photoproducts that otherwise stimulate malignant melanoma development. Here, a genome-wide loss-of-function screen, coupling CRISPR/Cas9 technology with a flow cytometry-based DNA repair assay, was used to identify novel genes required for efficient NER in primary human fibroblasts. Interestingly, the screen revealed multiple genes encoding proteins, with no previously known involvement in UV damage repair, that significantly modulate NER uniquely during S phase of the cell cycle. Among these, we further characterized Dyrk1A, a dual specificity kinase that phosphorylates the proto-oncoprotein cyclin D1 on threonine 286 (T286), thereby stimulating its timely cytoplasmic relocalization and proteasomal degradation, which is required for proper regulation of the G1-S phase transition and control of cellular proliferation. We demonstrate that in UV-irradiated HeLa cells, depletion of Dyrk1A leading to overexpression of cyclin D1 causes inhibition of NER uniquely during S phase and reduced cell survival. Consistently, expression/nuclear accumulation of nonphosphorylatable cyclin D1 (T286A) in melanoma cells strongly interferes with S phase NER and enhances cytotoxicity post-UV. Moreover, the negative impact of cyclin D1 (T286A) overexpression on repair is independent of cyclin-dependent kinase activity but requires cyclin D1-dependent upregulation of p21 expression. Our data indicate that inhibition of NER during S phase might represent a previously unappreciated noncanonical mechanism by which oncogenic cyclin D1 fosters melanomagenesis.


Subject(s)
Cyclin D1 , Cyclin-Dependent Kinase Inhibitor p21 , DNA Repair , Protein Serine-Threonine Kinases , Protein-Tyrosine Kinases , Humans , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/radiation effects , HeLa Cells , Protein-Tyrosine Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/pathology , Fibroblasts/radiation effects , S Phase , G1 Phase , Melanoma/genetics , Melanoma/pathology , Cells, Cultured , Ultraviolet Rays/adverse effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinogenesis/radiation effects , Dyrk Kinases
19.
Geriatr Gerontol Int ; 23(4): 304-312, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36807963

ABSTRACT

AIM: Molecular hydrogen is not only expected to be used as an energy-generating resource, but also to have preventive effects on a variety of clinical manifestations related to oxidative stress through scavenging radicals or regulating gene expression. In the current study, we investigated the influence of intermittent environmental exposure to hydrogen gas at a safe concentration (1.3%) on photoaging using an ultraviolet A (UVA)-irradiated murine model. METHODS: To mimic the expected human daily activity cycle, UVA exposure in the daytime and hydrogen exposure in the night-time, an original design, UVA-transmission, hydrogen-exposure system was established. Mice were bred under experimental conditions of UVA irradiation and normal air for 8 h (outdoor time 09.00-17.00 hours), and UVA non-irradiation and inhalation of hydrogen gas for 16 h (indoor time 17.00-09.00 hours), and the daily cycle was continued for up to 6 weeks. The progression of photoaging, including morphological changes, collagen degradation and UVA-related DNA damage, was evaluated. RESULTS: Intermittent administration of hydrogen gas by our system prevented UVA-induced epidermal signs, such as hyperplasia, melanogenesis and appearance of senescence cells, and UVA-induced dermal signs, such as collagen degradation. In addition, we detected attenuation of DNA damage in the hydrogen exposure group as indirect evidence that intermittent exposure to hydrogen gas reduced oxidative stress. CONCLUSIONS: Our findings support the notion that long-term, intermittent environmental exposure to hydrogen gas in daily life has a beneficial effect on UVA-induced photoaging. Geriatr Gerontol Int 2023; 23: 304-312.


Subject(s)
Skin Aging , Skin Diseases , Humans , Animals , Mice , Skin/metabolism , Cells, Cultured , Oxidative Stress , Collagen/metabolism , Environmental Exposure , Fibroblasts/metabolism , Fibroblasts/radiation effects , Ultraviolet Rays/adverse effects
20.
J Photochem Photobiol B ; 241: 112670, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36841175

ABSTRACT

BACKGROUND: Antimicrobial blue light (aBL) kills a variety of bacteria, including Porphyromonas gingivalis. However, little is known about the transcriptomic response of P. gingivalis to aBL therapy. This study was designed to evaluate the selective cytotoxicity of aBL against P. gingivalis over human cells and to further investigate the genetic response of P. gingivalis to aBL at the transcriptome level. METHODS: Colony forming unit (CFU) testing, confocal laser scanning microscopy (CLSM), and scanning electron microscopy (SEM) were used to investigate the antimicrobial effectiveness of blue light against P. gingivalis. The temperatures of the irradiated targets were measured to prevent overheating. Multiple fluorescent probes were used to quantify reactive oxygen species (ROS) generation after blue-light irradiation. RNA sequencing (RNA-seq) was used to investigate the changes in global gene expression. Following the screening of target genes, real-time quantitative polymerase chain reaction (RT-qPCR) was performed to confirm the regulation of gene expression. RESULTS: A 405 nm aBL at 100 mW/cm2 significantly killed P. gingivalis within 5 min while sparing human gingival fibroblasts (HGFs). No obvious temperature changes were detected in the irradiated surface under our experimental conditions. RNA-seq showed that the transcription of multiple genes was regulated, and RT-qPCR revealed that the expression levels of the genes RgpA and RgpB, which may promote heme uptake, as well as the genes Ftn and FetB, which are related to iron homeostasis, were significantly upregulated. The expression levels of the FeoB-2 and HmuR genes, which are related to hydroxyl radical scavenging, were significantly downregulated. CONCLUSIONS: aBL strengthens the heme uptake and iron export gene pathways while reducing the ROS scavenging pathways in P. gingivalis, thus improving the accumulation of endogenous photosensitizers and enhancing oxidative damage to P. gingivalis.


Subject(s)
Color , Gene Expression Regulation, Bacterial , Genes, Bacterial , Iron , Light , Porphyrins , Porphyromonas gingivalis , Porphyrins/metabolism , Iron/metabolism , Porphyromonas gingivalis/cytology , Porphyromonas gingivalis/genetics , Porphyromonas gingivalis/metabolism , Porphyromonas gingivalis/radiation effects , Biological Transport/genetics , Biological Transport/radiation effects , Humans , Gingiva/cytology , Fibroblasts/cytology , Fibroblasts/radiation effects , Hydroxyl Radical/metabolism , Heme/metabolism , Up-Regulation/radiation effects , Homeostasis/radiation effects , Down-Regulation/radiation effects , Microbial Viability/radiation effects , Reactive Oxygen Species/metabolism , Aerobiosis , Genes, Bacterial/radiation effects , Gene Expression Regulation, Bacterial/genetics , Gene Expression Regulation, Bacterial/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...