Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 509
Filter
1.
Cell Death Dis ; 15(6): 404, 2024 Jun 10.
Article in English | MEDLINE | ID: mdl-38858355

ABSTRACT

Senescent cells exhibit a diverse spectrum of changes in their morphology, proliferative capacity, senescence-associated secretory phenotype (SASP) production, and mitochondrial homeostasis. These cells often manifest with elongated mitochondria, a hallmark of cellular senescence. However, the precise regulatory mechanisms orchestrating this phenomenon remain predominantly unexplored. In this study, we provide compelling evidence for decreases in TIA-1, a pivotal regulator of mitochondrial dynamics, in models of both replicative senescence and ionizing radiation (IR)-induced senescence. The downregulation of TIA-1 was determined to trigger mitochondrial elongation and enhance the expression of senescence-associated ß-galactosidase, a marker of cellular senescence, in human foreskin fibroblast HS27 cells and human keratinocyte HaCaT cells. Conversely, the overexpression of TIA-1 mitigated IR-induced cellular senescence. Notably, we identified the miR-30-5p family as a novel factor regulating TIA-1 expression. Augmented expression of the miR-30-5p family was responsible for driving mitochondrial elongation and promoting cellular senescence in response to IR. Taken together, our findings underscore the significance of the miR-30-5p/TIA-1 axis in governing mitochondrial dynamics and cellular senescence.


Subject(s)
Cellular Senescence , MicroRNAs , Mitochondria , Mitochondrial Dynamics , T-Cell Intracellular Antigen-1 , Humans , MicroRNAs/metabolism , MicroRNAs/genetics , Cellular Senescence/radiation effects , Cellular Senescence/genetics , Mitochondrial Dynamics/genetics , T-Cell Intracellular Antigen-1/metabolism , T-Cell Intracellular Antigen-1/genetics , Mitochondria/metabolism , Fibroblasts/metabolism , Fibroblasts/radiation effects , Cell Line , Keratinocytes/metabolism , Keratinocytes/radiation effects , Keratinocytes/cytology , Signal Transduction , Radiation, Ionizing
2.
Cancer Med ; 13(12): e7381, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38888415

ABSTRACT

INTRODUCTION: Therapy-induced senescent cancer and stromal cells secrete cytokines and growth factors to promote tumor progression. Therefore, senescent cells may be novel targets for tumor treatment. Near-infrared photoimmunotherapy (NIR-PIT) is a highly tumor-selective therapy that employs conjugates of a molecular-targeting antibody and photoabsorber. Thus, NIR-PIT has the potential to be applied as a novel senolytic therapy. This study aims to investigate the efficacy of NIR-PIT treatment on senescent cancer and stromal cells. METHODS: Two cancer cell lines (human lung adenocarcinoma A549 cells and human pancreatic cancer MIA PaCa-2 cells) and two normal cell lines (mouse fibroblast transfected with human epidermal growth factor receptor 2 [HER2] cells and human fibroblast WI38 cells) were used. The cytotoxicity of NIR-PIT was evaluated using anti-epidermal growth factor receptor (EGFR) antibody panitumumab and anti-HER2 antibody transtuzumab. RESULTS: Cellular senescence was induced in A549 and MIA PaCa-2 cells by 10 Gy γ-irradiation. The up-regulation of cellular senescence markers and characteristic morphological changes in senescent cells, including enlargement, flattening, and multinucleation, were observed in cancer cells after 5 days of γ-irradiation. Then, NIR-PIT targeting EGFR was performed on these senescent cancer cells. The NIR-PIT induced morphological changes, including bleb formation, swelling, and the inflow of extracellular fluid, and induced a significant decrease in cellular viability. These results suggested that NIR-PIT may induce cytotoxicity using the same mechanism in senescent cancer cells. In addition, similar morphological changes were also induced in radiation-induced senescent 3T3-HER2 fibroblasts by NIR-PIT targeting human epidermal growth factor receptor 2. CONCLUSION: NIR-PIT eliminates both senescent cancer and stromal cells in vitro suggesting it may be a novel strategy for tumor treatment.


Subject(s)
Cellular Senescence , ErbB Receptors , Immunotherapy , Phototherapy , Stromal Cells , Humans , Cellular Senescence/radiation effects , Animals , Mice , Immunotherapy/methods , Stromal Cells/metabolism , Phototherapy/methods , ErbB Receptors/metabolism , Cell Line, Tumor , Infrared Rays/therapeutic use , Receptor, ErbB-2/metabolism , Lung Neoplasms/therapy , Lung Neoplasms/pathology , Trastuzumab/pharmacology , Panitumumab/pharmacology , A549 Cells , Gamma Rays
3.
Discov Med ; 36(184): 1080-1090, 2024 May.
Article in English | MEDLINE | ID: mdl-38798266

ABSTRACT

BACKGROUND: Skin photoaging is a complex process of skin aging caused by continuous exposure to ultraviolet (UV) radiation through oxidative stress and other pathways, yet effective treatments are scarce. Metformin is a drug with both anti-senescence and antioxidant functions; however, there are fewer studies on photoaging. The study aimed to investigate the role of needle-free injection of metformin in alleviating ultraviolet radiation B (UVB) induced skin photoaging, and to explore the mechanisms through which metformin alleviates fibroblast photoaging by inhibiting ferroptosis and oxidative stress. METHODS: In our study, we initially performed bioinformatic analysis on the gene expression profile (GSE38308), and our RNA sequencing (RNA-Seq) found that photoaging is associated with ferroptosis. We investigated the potential skin-protective mechanism of metformin by utilizing a UVB-induced rat skin photoaging model and human skin fibroblasts (HSF) treated with UVB. For in vitro experiments, cellular senescence was detected using SA-ß-galactosidase staining and p16 in western blot. Ferroptosis and oxidative stress were assessed via western blot (glutathione Peroxidase 4 (GPX4) and nuclear factor erythroid-2-related factor 2 (Nrf2)), reactive oxygen species (ROS) levels, transmission electron microscope, Lillie's staining, and immunofluorescence staining. During in vivo experiments, metformin was administered by needle-free jet injectors injected into the backs of rats. The effectiveness of metformin was detected using the Masson staining and western blot. RESULTS: We found that the ferroptosis pathway was closely associated with photoaging through bioinformatics analysis. In the UVB-induced photoaging HSF cells, treatment with metformin exhibits the following effects: a reduction in blue-stained granules in SA-ß-galactosidase staining and a decrease in the expression of p16, indicating a reduction in cellular senescence. Moreover, metformin leads to decreased ROS levels and increased expression of the oxidative stress-related protein Nrf2, suggesting inhibition of oxidative stress within the cells. Additionally, metformin results in an elevation of GPX4 expression, a decrease in blue-stained granules in Lillie's staining, and a reduction in ferroptosis-associated mitochondrial damage, indicating a decline in ferroptosis. Needle-free injection of metformin could directly achieve therapeutic effects by affecting HSF cells in the dermis. The needle-free injection of metformin treatment effectively improved the photoaging skin in rats compared to the photoaging group, ameliorated oxidative stress, and reduced ferroptosis. CONCLUSIONS: Our data highlights a novel needle-free injection of metformin that improves photoaging and has good therapeutic potential.


Subject(s)
Ferroptosis , Metformin , Oxidative Stress , Skin Aging , Ultraviolet Rays , Metformin/pharmacology , Metformin/administration & dosage , Oxidative Stress/drug effects , Oxidative Stress/radiation effects , Animals , Skin Aging/drug effects , Skin Aging/radiation effects , Ferroptosis/drug effects , Ferroptosis/radiation effects , Rats , Humans , Ultraviolet Rays/adverse effects , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Reactive Oxygen Species/metabolism , Skin/drug effects , Skin/pathology , Skin/radiation effects , Skin/metabolism , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Rats, Sprague-Dawley , Male , NF-E2-Related Factor 2/metabolism
4.
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.
Cancer Lett ; 593: 216930, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38705566

ABSTRACT

Radiotherapy (RT) in non-small cell lung cancer (NSCLC) triggers cellular senescence, complicating tumor microenvironments and affecting treatment outcomes. This study examines the role of lymphocyte immunoglobulin-like receptor B2 (LILRB2) in modulating RT-induced senescence and radiosensitivity in NSCLC. Through methodologies including irradiation, lentivirus transfection, and various molecular assays, we assessed LILRB2's expression and its impact on cellular senescence levels and tumor cell behaviors. Our findings reveal that RT upregulates LILRB2, facilitating senescence and a senescence-associated secretory phenotype (SASP), which in turn enhances tumor proliferation and resistance to radiation. Importantly, LILRB2 silencing attenuates these effects by inhibiting the JAK2/STAT3 pathway, significantly increasing radiosensitivity in NSCLC models. Clinical data correlate high LILRB2 expression with reduced RT response and poorer prognosis, suggesting LILRB2's pivotal role in RT-induced senescence and its potential as a therapeutic target to improve NSCLC radiosensitivity.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Cellular Senescence , Lung Neoplasms , Radiation Tolerance , Receptors, Immunologic , Humans , Carcinoma, Non-Small-Cell Lung/radiotherapy , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , Cellular Senescence/radiation effects , Radiation Tolerance/genetics , Lung Neoplasms/radiotherapy , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Cell Line, Tumor , Cell Proliferation/radiation effects , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Animals , Janus Kinase 2/metabolism , Janus Kinase 2/genetics , Mice , Signal Transduction , Gene Expression Regulation, Neoplastic/radiation effects , Senescence-Associated Secretory Phenotype/genetics , A549 Cells , Female
6.
Phytomedicine ; 130: 155676, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38820663

ABSTRACT

BACKGROUND: Prolonged exposure to sun radiation may result in harmful skin photoaging. Therefore, discovering novel anti-photoaging treatment modalities is critical. An active component isolated from Salvia miltiorrhiza (SM), Salvianolic acid B (Sal-B), is a robust antioxidant and anti-inflammatory agent. This investigation aimed to discover the therapeutic impact and pathways of salvianolic acid B for UVB-induced skin photoaging, an area that remains unexplored. METHODS: We conducted in vitro experiments on human dermal fibroblasts (HDFs) exposed to UVB radiation, assessing cellular senescence, superoxide dismutase (SOD) activity, cell viability, proliferation, migration, levels of reactive oxygen species (ROS), and mitochondrial health. The potential mechanism of Sal-B was analyzed using RNA sequencing, with further validation through Western blotting, PCR, and nuclear factor erythroid 2-related factor 2 (NRF2) silencing methods. In vivo, a model of skin photoaging induced by UVB in nude mice was employed. The collagen fiber levels were assessed utilizing hematoxylin and eosin (H&E), Masson, and Sirus red staining. Additionally, NRF2 and related gene and protein expression levels were identified utilizing PCR and Western blotting. RESULTS: Sal-B was found to significantly counteract photoaging in UVB-exposed skin fibroblasts, reducing aging-related decline in fibroblast proliferation and an increase in apoptosis. It was observed that Sal-B aids in protecting mitochondria from excessive ROS production by promoting NRF2 nuclear translocation. NRF2 knockdown experiments established its necessity for Sal-B's anti-photoaging effects. The in vivo studies also verified Sal-B's anti-photoaging efficacy, surpassing that of tretinoin (Retino-A). These outcomes offer novel insights into the contribution of Sal-B in developing clinical treatment modalities for UVB-induced photodamage in skin fibroblasts. CONCLUSION: In this investigation, we identified the Sal-B protective impact on the senescence of dermal fibroblasts and skin photoaging induced by radiation of UVB. The outcomes suggest Sal-B as a potential modulator of the NRF2 signaling pathway.


Subject(s)
Benzofurans , Fibroblasts , Mice, Nude , NF-E2-Related Factor 2 , Skin Aging , Ultraviolet Rays , NF-E2-Related Factor 2/metabolism , Skin Aging/drug effects , Skin Aging/radiation effects , Animals , Ultraviolet Rays/adverse effects , Humans , Fibroblasts/drug effects , Fibroblasts/radiation effects , Benzofurans/pharmacology , Reactive Oxygen Species/metabolism , Mice , Salvia miltiorrhiza/chemistry , Cell Proliferation/drug effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Antioxidants/pharmacology , Cell Survival/drug effects , Skin/drug effects , Skin/radiation effects , Superoxide Dismutase/metabolism , Depsides
7.
Biochem Pharmacol ; 224: 116234, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38670436

ABSTRACT

Ionizing radiation, a standard therapeutic approach for lung cancer, often leads to cellular senescence and the induction of epithelial-mesenchymal transition (EMT), posing significant challenges in treatment efficacy and cancer progression. Overcoming these obstacles is crucial for enhancing therapeutic outcomes in lung cancer management. This study investigates the effects of ionizing radiation and gemcitabine on lung cancer cells, with a focus on induced senescence, EMT, and apoptosis. Human-derived A549, PC-9, and mouse-derived Lewis lung carcinoma cells exposed to 10 Gy X-ray irradiation exhibited senescence, as indicated by morphological changes, ß-galactosidase staining, and cell cycle arrest through the p53-p21 pathway. Ionizing radiation also promoted EMT via TGFß/SMAD signaling, evidenced by increased TGFß1 levels, altered EMT marker expressions, and enhanced cell migration. Gemcitabine, a first-line lung cancer treatment, was shown to enhance apoptosis in senescent cells caused by radiation. It inhibited cell proliferation, induced mitochondrial damage, and triggered caspase-mediated apoptosis, thus mitigating EMT in vitro. Furthermore, in vivo studies using a lung cancer mouse model revealed that gemcitabine, combined with radiation, significantly reduced tumor volume and weight, extended survival, and suppressed malignancy indices in irradiated tumors. Collectively, these findings demonstrate that gemcitabine enhances the therapeutic efficacy against radiation-resistant lung cancer cells, both by inducing apoptosis in senescent cells and inhibiting EMT, offering potential improvements in lung cancer treatment strategies.


Subject(s)
Antimetabolites, Antineoplastic , Cellular Senescence , Deoxycytidine , Epithelial-Mesenchymal Transition , Gemcitabine , Lung Neoplasms , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Animals , Humans , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Mice , Antimetabolites, Antineoplastic/pharmacology , Cell Line, Tumor , Mice, Inbred C57BL , A549 Cells , Radiation, Ionizing , Apoptosis/drug effects , Apoptosis/radiation effects
8.
Aging (Albany NY) ; 16(8): 6673-6693, 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38683123

ABSTRACT

PURPOSE: The objective of this study was to investigate the senescent phenotypes of human corneal endothelial cells (hCEnCs) upon treatment with ultraviolet (UV)-A. METHODS: We assessed cell morphology, senescence-associated ß-galactosidase (SA-ß-gal) activity, cell proliferation and expression of senescence markers (p16 and p21) in hCEnCs exposed to UV-A radiation, and senescent hCEnCs induced by ionizing radiation (IR) were used as positive controls. We performed RNA sequencing and proteomics analyses to compare gene and protein expression profiles between UV-A- and IR-induced senescent hCEnCs, and we also compared the results to non-senescent hCEnCs. RESULTS: Cells exposed to 5 J/cm2 of UV-A or to IR exhibited typical senescent phenotypes, including enlargement, increased SA-ß-gal activity, decreased cell proliferation and elevated expression of p16 and p21. RNA-Seq analysis revealed that 83.9% of the genes significantly upregulated and 82.6% of the genes significantly downregulated in UV-A-induced senescent hCEnCs overlapped with the genes regulated in IR-induced senescent hCEnCs. Proteomics also revealed that 93.8% of the proteins significantly upregulated in UV-A-induced senescent hCEnCs overlapped with those induced by IR. In proteomics analyses, senescent hCEnCs induced by UV-A exhibited elevated expression levels of several factors part of the senescence-associated secretory phenotype. CONCLUSIONS: In this study, where senescence was induced by UV-A, a more physiological stress for hCEnCs compared to IR, we determined that UV-A modulated the expression of many genes and proteins typically altered upon IR treatment, a more conventional method of senescence induction, even though UV-A also modulated specific pathways unrelated to IR.


Subject(s)
Cell Proliferation , Cellular Senescence , Endothelial Cells , Ultraviolet Rays , Humans , Cellular Senescence/radiation effects , Ultraviolet Rays/adverse effects , Cell Proliferation/radiation effects , Endothelial Cells/radiation effects , Endothelial Cells/metabolism , Endothelium, Corneal/radiation effects , Endothelium, Corneal/metabolism , Cells, Cultured , Proteomics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , beta-Galactosidase/metabolism , beta-Galactosidase/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics
9.
Int J Radiat Oncol Biol Phys ; 118(3): 801-816, 2024 Mar 01.
Article in English | MEDLINE | ID: mdl-37758068

ABSTRACT

PURPOSE: Histone variant H2A.J is associated with premature senescence after ionizing radiation (IR) and modulates senescence-associated secretory phenotype (SASP). Using constitutive H2A.J knock-out mice, the role of H2A.J was investigated in radiation dermatitis. METHODS AND MATERIALS: H2A.J wild-type (WT) and knock-out (KO) mice were exposed to moderate or high IR doses (≤20 Gy, skinfold IR). Radiation-induced skin reactions were investigated up to 2 weeks post-IR at macroscopic and microscopic levels. H2A.J and other senescence markers, as well as DNA damage and proliferation markers, were studied by immunohistochemistry, immunofluorescence, and electron microscopy. After high-dose IR, protein-coding transcriptomes were analyzed by RNA sequencing, immune cell infiltration by flow cytometry, and gene expression by reverse transcription polymerase chain reaction in (non-) irradiated WT versus KO skin. RESULTS: In WT skin, epidermal keratinocytes showed time- and dose-dependent H2A.J accumulation after IR exposure. Unexpectedly, stronger inflammatory reactions with increased epidermal thickness and progressive hair follicle loss were observed in irradiated KO versus WT skin. Clearly more radiation-induced senescence was observed in keratinocyte populations of KO skin after moderate and high doses, with hair follicle stem cells being particularly badly damaged, leading to follicle atrophy. After high-dose IR, transcriptomic analysis revealed enhanced senescence-associated signatures in irradiated KO skin, with intensified release of SASP factors. Flow cytometric analysis indicated increased immune cell infiltration in both WT and KO skin; however, specific chemokine-mediated signaling in irradiated KO skin led to more neutrophil recruitment, thereby aggravating radiation toxicities. Increased skin damage in irradiated KO skin led to hyperproliferation, abnormal differentiation, and cornification of keratinocytes, accompanied by increased upregulation of transcription-factor JunB. CONCLUSIONS: Lack of radiation-induced H2A.J expression in keratinocytes is associated with increased senescence induction, modulation of SASP expression, and exacerbated inflammatory skin reactions. Hence, epigenetic H2A.J-mediated gene expression in response to IR regulates keratinocyte immune functions and plays an essential role in balancing the inflammatory response during radiation dermatitis.


Subject(s)
Histones , Radiodermatitis , Animals , Mice , Histones/metabolism , Skin/radiation effects , Keratinocytes/physiology , Radiation, Ionizing , Cellular Senescence/radiation effects
10.
Radiother Oncol ; 190: 109984, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37926332

ABSTRACT

BACKGROUND & AIM: Irradiation of the salivary glands during head and neck cancer treatment induces cellular senescence in response to DNA damage and contributes to radiation-induced hyposalivation by affecting the salivary gland stem/progenitor cell (SGSC) niche. Cellular senescence, such as that induced by radiation, is a state of cell-cycle arrest, accompanied by an altered pro-inflammatory secretome known as the senescence-associated secretory phenotype (SASP) with potential detrimental effects on the surrounding microenvironment. We hypothesized that the pro-regenerative properties of mesenchymal stem cells (MSCs) may attenuate cellular senescence post-irradiation. Therefore, here we evaluated the effects of adipose-derived MSCs (ADSCs) on the radiation-induced response of salivary gland organoids (SGOs). METHODS: Proteomic analyses to identify soluble mediators released by ADSCs co-cultured with SGOS revealed secretion of hepatocyte growth factor (HGF) in ADSCs, suggesting a possible role in the stem cell crosstalk. Next, the effect of recombinant HGF in the culture media of ex vivo grown salivary gland cells was tested in 2D monolayers and 3D organoid models. RESULTS: Treatment with HGF robustly increased salivary gland cell proliferation. Importantly, HGF supplementation post-irradiation enhanced proliferation at lower doses of radiation (0, 3, 7 Gy), but not at higher doses (10, 14 Gy) where most cells stained positive for senescence-associated beta-galactosidase. Furthermore, HGF had no effect on the senescence-associated secretory phenotype (SASP) of irradiated SGOs, suggesting there may be compensatory proliferation by cell-division competent cells instead of a reversal of cellular senescence after irradiation. CONCLUSION: ADSCs may positively influence radiation recovery through HGF secretion and can promote the ex vivo expansion of salivary gland stem/progenitor cells to enhance the effects of co-transplanted SGSC.


Subject(s)
Hepatocyte Growth Factor , Mesenchymal Stem Cells , Humans , Hepatocyte Growth Factor/pharmacology , Proteomics , Salivary Glands , Cellular Senescence/radiation effects , Cell Proliferation
11.
Radiother Oncol ; 190: 110028, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38007043

ABSTRACT

BACKGROUND AND PURPOSE: Patients undergoing radiotherapy for head and neck cancer often experience a decline in their quality of life due to the co-irradiation of salivary glands. Radiation-induced cellular senescence is a key factor contributing to salivary gland dysfunction. Interestingly, mitochondrial dysfunction and cellular senescence have been reported to be strongly interconnected and thus implicated in several aging-related diseases. This study aims to investigate the role of mitochondrial dysfunction in senescence induction in salivary gland stem/progenitor cells after irradiation. MATERIALS AND METHODS: A dose of 7 Gy photons was used to irradiate mouse salivary gland organoids. Senescent markers and mitochondrial function were assessed using rt-qPCR, western blot analysis, SA-ß-Gal staining and flow cytometry analysis. Mitochondrial dynamics-related proteins were detected by western blot analysis while Mdivi-1 and MFI8 were used to modulate the mitochondrial fission process. To induce mitophagy, organoids were treated with Urolithin A and PMI and subsequently stem/progenitor cell self-renewal capacity was assessed as organoid forming efficiency. RESULTS: Irradiation led to increased senescence and accumulation of dysfunctional mitochondria. This was accompanied by a strong downregulation of mitochondrial fission-related proteins and mitophagy-related genes. After irradiation, treatment with the mitophagy inducer Urolithin A attenuated the senescent phenotype and improved organoid growth and stem/progenitor cell self-renewal capacity. CONCLUSION: This study shows the important interplay between senescence and mitochondrial dysfunction after irradiation. Importantly, activation of mitophagy improved salivary gland stem/progenitor cell function thereby providing a novel therapeutic strategy to restore the regenerative capacity of salivary glands following irradiation.


Subject(s)
Mitochondrial Diseases , Quality of Life , Animals , Mice , Cellular Senescence/radiation effects , Mitochondrial Diseases/metabolism , Mitophagy , Salivary Glands , Stem Cells/radiation effects
12.
Int J Mol Sci ; 23(17)2022 Aug 29.
Article in English | MEDLINE | ID: mdl-36077226

ABSTRACT

PURPOSE: Precise molecular and cellular mechanisms of radiation-induced dermatitis are incompletely understood. Histone variant H2A.J is associated with cellular senescence and modulates senescence-associated secretory phenotype (SASP) after DNA-damaging insults, such as ionizing radiation (IR). Using ex vivo irradiated cultured foreskin, H2A.J was analyzed as a biomarker of radiation-induced senescence, potentially initiating the inflammatory cascade of radiation-induced skin injury. METHODS: Human foreskin explants were collected from young donors, irradiated ex vivo with 10 Gy, and cultured in air-liquid interphase for up to 72 h. At different time-points after ex vivo IR exposure, the foreskin epidermis was analyzed for proliferation and senescence markers by immunofluorescence and immunohistochemical staining of sectioned tissue. Secretion of cytokines was measured in supernatants by ELISA. Using our mouse model with fractionated in vivo irradiation, H2A.J expression was analyzed in epidermal stem/progenitor cell populations localized in different regions of murine hair follicles (HF). RESULTS: Non-vascularized foreskin explants preserved their tissue homeostasis up to 72 h (even after IR exposure), but already non-irradiated foreskin epithelium expressed high levels of H2A.J in all epidermal layers and secreted high amounts of cytokines. Unexpectedly, no further increase in H2A.J expression and no obvious upregulation of cytokine secretion was observed in the foreskin epidermis after ex vivo IR. Undifferentiated keratinocytes in murine HF regions, by contrast, revealed low H2A.J expression in non-irradiated skin and significant radiation-induced H2A.J upregulations at different time-points after IR exposure. Based on its staining characteristics, we presume that H2A.J may have previously underestimated the importance of the epigenetic regulation of keratinocyte maturation. CONCLUSIONS: Cultured foreskin characterized by highly keratinized epithelium and specific immunological features is not an appropriate model for studying H2A.J-associated tissue reactions during radiation-induced dermatitis.


Subject(s)
Foreskin , Radiodermatitis , Animals , Cells, Cultured , Cellular Senescence/radiation effects , Cytokines , Epigenesis, Genetic , Histones , Humans , Male , Mice , Radiation, Ionizing
13.
J Photochem Photobiol B ; 235: 112568, 2022 Oct.
Article in English | MEDLINE | ID: mdl-36137302

ABSTRACT

The human corneal endothelial cells (HCEnCs) play a vital role in the maintenance of corneal transparency and visual acuity. In our daily life, HCEnCs are inevitably exposed to ultraviolet B (UVB) radiation leading to decreases of visual acuity and corneal transparency resulting in visual loss eventually. Therefore, understanding the UVB-induced cytotoxicity in HCEnCs is of importance for making efficient strategies to protect our vision from UVB-damage. However, in-depth knowledge about UVB-induced cytotoxicity in HCEnCs is missing. Herein, we pulse-irradiated the HCEnCs in vitro with 150 mJ/cm2 UVB (the environmental dose) at each subculture for 4 passages to explore the insights into UVB-induced phototoxicity. The results showed that the UVB-treated HCEnCs exhibit typical senescent characteristics, including significantly enlarged relative cell area, increased senescence-associated ß-galactosidase positive staining, and upregulated p16INK4A and senescence associated secretory phenotypes (SASPs) such as CCL-27, IL-1α/6/8/10, TGF-ß1 and TNF-α, as well as decreased cell proliferation and Lamin B1 expression, and translocation of Lamin B1. Furthermore, we explored the causative mechanisms of senescence and found that 150 mJ/cm2 UVB pulse-irradiation impairs DNA to activate DNA damage response (DDR) pathway of ATM-p53-p21WAF1/CIP1 with downregulated DNA repair enzyme PARP1, leading to cell cycle arrest resulting in DDR-mediated senescence. Meanwhile, UVB pulse-irradiation also elicits a consistent increase of ROS production to aggravate DNA damage and impose oxidative stress on energy metabolism leading to metabolic disturbance resulting in metabolic disturbance-mediated senescence. Altogether, the repeated pulse-irradiation of 150 mJ/cm2 UVB induces HCEnC senescence via both DDR pathway and energy metabolism disturbance.


Subject(s)
Cellular Senescence , DNA Damage , Endothelial Cells , Oxidative Stress , Ultraviolet Rays , Cells, Cultured , Cellular Senescence/radiation effects , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Endothelial Cells/cytology , Endothelial Cells/radiation effects , Humans , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Suppressor Protein p53/metabolism , Ultraviolet Rays/adverse effects , beta-Galactosidase/metabolism
14.
J Eur Acad Dermatol Venereol ; 36(7): 1125-1135, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35274377

ABSTRACT

BACKGROUND: Skin ageing is caused by numerous factors that result in structural and functional changes in cutaneous components. Research has shown that senescent cells are known to accumulate in skin ageing, however, the role of senescent cells in skin ageing has not been defined. OBJECTIVES: To elucidate the role of the senescent cell in skin ageing, we evaluated the effect of known senolytic drugs on senescent dermal fibroblasts. METHODS: Primary human dermal fibroblasts (HDFs) were induced to senescence by long-term passaging, UV irradiation, and H2 O2 treatment. Cell viability was measured after treatment of ABT-263 and ABT-737 on HDFs. Young and aged hairless mice were intradermally injected with drugs or vehicle on the dorsal skin for 10 days. Skin specimens were obtained and reverse-transcription quantitative PCR, western blotting, and histological analysis were performed. RESULTS: We found that ABT-263 and ABT-737 induced selective clearance of senescent dermal fibroblasts, regardless of the method of senescence induction. Aged mouse skin treated with ABT-263 or ABT-737 showed increased collagen density, epidermal thickness, and proliferation of keratinocytes, as well as decreased senescence-associated secretory phenotypes, such as MMP-1 and IL-6. CONCLUSIONS: Taken together, our results indicate that selective clearance of senescent skin cells can attenuate and improve skin ageing phenotypes and that senolytic drugs may be of potential use as new therapeutic agents for treating ageing of the skin.


Subject(s)
Senotherapeutics , Skin Aging , Animals , Cellular Senescence/genetics , Cellular Senescence/radiation effects , Fibroblasts , Humans , Mice , Skin/pathology
15.
Mol Oncol ; 16(6): 1365-1383, 2022 03.
Article in English | MEDLINE | ID: mdl-35122388

ABSTRACT

Radiation therapy can induce cellular senescence in cancer cells, leading to short-term tumor growth arrest but increased long-term recurrence. To better understand the molecular mechanisms involved, we developed a model of radiation-induced senescence in cultured cancer cells. The irradiated cells exhibited a typical senescent phenotype, including upregulation of p53 and its main target, p21, followed by a sustained reduction in cellular proliferation, changes in cell size and cytoskeleton organization, and senescence-associated beta-galactosidase activity. Mass spectrometry-based proteomic profiling of the senescent cells indicated downregulation of proteins involved in cell cycle progression and DNA repair, and upregulation of proteins associated with malignancy. A functional siRNA screen using a cell death-related library identified mitochondrial serine protease HtrA2 as being necessary for sustained growth arrest of the senescent cells. In search of direct HtrA2 substrates following radiation, we determined that HtrA2 cleaves the intermediate filament protein vimentin, affecting its cytoplasmic organization. Ectopic expression of active cytosolic HtrA2 resulted in similar changes to vimentin filament assembly. Thus, HtrA2 is involved in the cytoskeletal reorganization that accompanies radiation-induced senescence and the continuous maintenance of proliferation arrest.


Subject(s)
Cellular Senescence , High-Temperature Requirement A Serine Peptidase 2 , Neoplasms , Proteomics , Apoptosis , Cellular Senescence/physiology , Cellular Senescence/radiation effects , High-Temperature Requirement A Serine Peptidase 2/genetics , High-Temperature Requirement A Serine Peptidase 2/metabolism , Humans , Mitochondrial Proteins/metabolism , Neoplasms/genetics , Neoplasms/radiotherapy , Tumor Cells, Cultured , Vimentin/metabolism
16.
Cells ; 11(2)2022 01 13.
Article in English | MEDLINE | ID: mdl-35053389

ABSTRACT

Irreparable DNA damage following ionizing radiation (IR) triggers prolonged DNA damage response and induces premature senescence. Cellular senescence is a permanent state of cell-cycle arrest characterized by chromatin restructuring, altered nuclear morphology and acquisition of secretory phenotype, which contributes to senescence-related inflammation. However, the mechanistic connections for radiation-induced DNA damage that trigger these senescence-associated hallmarks are poorly understood. In our in vitro model of radiation-induced senescence, mass spectrometry-based proteomics was combined with high-resolution imaging techniques to investigate the interrelations between altered chromatin compaction, nuclear envelope destabilization and nucleo-cytoplasmic chromatin blebbing. Our findings confirm the general pathophysiology of the senescence-response, with disruption of nuclear lamin organization leading to extensive chromatin restructuring and destabilization of the nuclear membrane with release of chromatin fragments into the cytosol, thereby activating cGAS-STING-dependent interferon signaling. By serial block-face scanning electron microscopy (SBF-SEM) whole-cell datasets were acquired to investigate the morphological organization of senescent fibroblasts. High-resolution 3-dimensional (3D) reconstruction of the complex nuclear shape allows us to precisely visualize the segregation of nuclear blebs from the main nucleus and their fusion with lysosomes. By multi-view 3D electron microscopy, we identified nanotubular channels formed in lamin-perturbed nuclei of senescent fibroblasts; the potential role of these nucleo-cytoplasmic nanotubes for expulsion of damaged chromatin has to be examined.


Subject(s)
Cell Nucleus/radiation effects , Cell Nucleus/ultrastructure , Cellular Senescence/radiation effects , Fibroblasts/radiation effects , Fibroblasts/ultrastructure , Imaging, Three-Dimensional , Microscopy, Electron , Radiation, Ionizing , Cell Line , Cell Nucleus/pathology , Cell Shape/radiation effects , Chromatin Assembly and Disassembly , Fibroblasts/pathology , Humans , Nanotubes/ultrastructure , Proteomics
17.
Biochim Biophys Acta Mol Basis Dis ; 1868(4): 166352, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35041996

ABSTRACT

Cancer survivorship has remarkably improved over the past decades; nevertheless, cancer survivors are burdened with multiple health complications primarily caused by their cancer therapy. Therapy-induced senescence is recognized as a fundamental mechanism contributing to adverse health complications in cancer survivors. In this mini-review, we will discuss the recent literature describing the mechanisms of cancer therapy-induced senescence. We will focus on endothelial cell senescence since it has been shown to be a key player in numerous cardiovascular complications. We will also discuss novel senotherapeutic approaches that have the potential to combat therapy-induced endothelial cell senescence.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiovascular Diseases/etiology , Cellular Senescence , Anthracyclines/adverse effects , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Endothelial Cells/cytology , Endothelial Cells/metabolism , Humans , Neoplasms/drug therapy , Neoplasms/pathology , Neoplasms/radiotherapy , Radiation, Ionizing
18.
Bull Exp Biol Med ; 172(2): 228-235, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855083

ABSTRACT

We studied the effect of neural stem cells (NSC) and mesenchymal stem cells (MSC) from mouse adipose tissue on survival, clonogenic activity, and senescence of NSC after exposure to γ-radiation. It was found that survival and clonogenic activity of NSC irradiated in doses of 1 and 2 Gy was enhanced when irradiated cells were co-cultured with non-irradiated NSC and MSC in permeable Transwell inserts. The proportion of senescent NSC (cells with high ß-galactosidase activity) increased with increasing irradiation dose. Co-culturing with non-irradiated NSC in 3 days after irradiation in a dose of 1 Gy led to a decrease in the proportion of senescent cells among irradiated NSC. Factors secreted by NSC and MSC can become the basis for the development of means for prevention and treatment of damage to brain cells resulting from radiation therapy of head and neck cancer.


Subject(s)
Gamma Rays/adverse effects , Mesenchymal Stem Cells/cytology , Neural Stem Cells/radiation effects , Adipose Tissue/cytology , Animals , Apoptosis/radiation effects , Cell Survival/radiation effects , Cells, Cultured , Cellular Senescence/physiology , Cellular Senescence/radiation effects , Coculture Techniques , Colony-Forming Units Assay , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Neural Stem Cells/cytology , Neural Stem Cells/physiology
19.
Cells ; 10(12)2021 12 17.
Article in English | MEDLINE | ID: mdl-34944078

ABSTRACT

Population aging is occurring rapidly worldwide, challenging the global economy and healthcare services. Brain aging is a significant contributor to various age-related neurological and neuropsychological disorders, including Alzheimer's disease and Parkinson's disease. Several extrinsic factors, such as exposure to ionizing radiation, can accelerate senescence. Multiple human and animal studies have reported that exposure to ionizing radiation can have varied effects on organ aging and lead to the prolongation or shortening of life span depending on the radiation dose or dose rate. This paper reviews the effects of radiation on the aging of different types of brain cells, including neurons, microglia, astrocytes, and cerebral endothelial cells. Further, the relevant molecular mechanisms are discussed. Overall, this review highlights how radiation-induced senescence in different cell types may lead to brain aging, which could result in the development of various neurological and neuropsychological disorders. Therefore, treatment targeting radiation-induced oxidative stress and neuroinflammation may prevent radiation-induced brain aging and the neurological and neuropsychological disorders it may cause.


Subject(s)
Brain/pathology , Cellular Senescence/radiation effects , Radiation, Ionizing , Animals , Autophagy/radiation effects , Humans , Mitochondria/pathology , Mitochondria/radiation effects , Oxidative Stress/radiation effects
20.
Int J Mol Sci ; 22(22)2021 Nov 22.
Article in English | MEDLINE | ID: mdl-34830482

ABSTRACT

Radiotherapy promotes tumor cell death and senescence through the induction of oxidative damage. Recent work has highlighted the importance of lipid peroxidation for radiotherapy efficacy. Excessive lipid peroxidation can promote ferroptosis, a regulated form of cell death. In this review, we address the evidence supporting a role of ferroptosis in response to radiotherapy and discuss the molecular regulators that underlie this interaction. Finally, we postulate on the clinical implications for the intersection of ferroptosis and radiotherapy.


Subject(s)
Lipid Metabolism/radiation effects , Lipid Peroxidation/radiation effects , Neoplasms/radiotherapy , Cell Death/radiation effects , Cellular Senescence/genetics , Cellular Senescence/radiation effects , Ferroptosis/genetics , Ferroptosis/radiation effects , Humans , Lipid Metabolism/genetics , Neoplasms/genetics , Neoplasms/pathology , Oxidative Stress/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...