Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
Histochem Cell Biol ; 162(4): 311-321, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38997526

ABSTRACT

Long-term radiofrequency radiation (RFR) exposure, which adversely affects organisms, deteriorates testicular functions. Misfolding or unfolding protein accumulation in the endoplasmic reticulum (ER) initiates an intracellular reaction known as ER stress (ERS), which activates the unfolded protein response (UPR) for proteostasis. Since both RFR exposure and ERS can cause male infertility, we hypothesized that RFR exposure causes ERS to adversely affect testicular functions in rats. To investigate role of ERS in mediating RFR effects on rat testis, we established five experimental groups in male rats: control, short-term 2100-megahertz (MHz) RFR (1-week), short-term sham (sham/1-week), long-term 2100-MHz RFR (10-week), and long-term sham (sham/10-week). ERS markers Grp78 and phosphorylated PERK (p-Perk) levels and ERS-related apoptosis markers Chop and caspase 12 were investigated by immunohistochemistry, immunoblotting, and quantitative real-time polymerase chain reaction (qPCR). Long-term RFR exposure increased Grp78, p-Perk, and Chop levels, while short-term RFR exposure elevated Chop and caspase 12 levels. Chop expression was not observed in spermatogonia and primary spermatocytes, which may protect spermatogonia and primary spermatocytes against RFR-induced ERS-mediated apoptosis, thereby allowing transmission of genetic material to next generations. While short and long-term RFR exposures trigger ERS and ERS-related apoptotic pathways, further functional analyses are needed to elucidate whether this RFR-induced apoptosis has long-term male infertility effects.


Subject(s)
Endoplasmic Reticulum Stress , Rats, Sprague-Dawley , Testis , Animals , Male , Endoplasmic Reticulum Stress/radiation effects , Testis/radiation effects , Testis/metabolism , Rats , Radio Waves/adverse effects , Apoptosis/radiation effects
2.
J Dermatol Sci ; 115(2): 75-84, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38969533

ABSTRACT

BACKGROUND: Few reports have confirmed whether exosomes derived from fibroblasts can regulate the process of melanogenesis. We wondered whether exosomes derived from fibroblasts could have a potent regulatory effect on melanogenesis and explored the underlying mechanisms. OBJECTIVE: This study aimed to find the role of fibroblasts in melanocytes and revealed the related mechanisms. METHODS: RT-qPCR, Western blot analysis were conducted to measure the RNA and protein expression level of various related genes. miRNA sequencing, mass spectrum analysis and subsequent bioinformatics analysis were employed to find the underlying targets. Zebrafish were employed to measure the melanin synthesis related process in vivo. Furthermore, electron microscopy, ROS measurement and dual-luciferase reporter assay were adopted to investigate the relationship between these processes. RESULTS: We found that exosomes derived from human primary dermal fibroblasts were internalized by human primary melanocytes and MNT1 cells and that the melanin content and the expression of melanin synthesis-related proteins TYR and MITF was inhibited by exosomes derived from UVB-induced human primary dermal fibroblasts. The miRNA expression profile in secreted exosomes changed significantly, with miR-25-5p identified as capable of regulating TSC2 expression via the CDS region. The miR-25-5p-TSC2 axis could affect the melanin content through subsequent cellular organelle dysfunction, such as mitochondrial dysfunction, endoplasmic reticulum stress and dysregulation of lysosomal cysteine proteases. CONCLUSION: We unveiled a novel regulatory role of fibroblasts in melanocytes, facilitated by the secretion of exosomes. miR-25-5p within exosomes plays a pivotal role in regulating melanogenesis via TSC2-induced cellular organelle dysfunction.


Subject(s)
Exosomes , Fibroblasts , Melanins , Melanocytes , MicroRNAs , Tuberous Sclerosis Complex 2 Protein , Ultraviolet Rays , Zebrafish , Humans , Exosomes/metabolism , Exosomes/radiation effects , MicroRNAs/metabolism , MicroRNAs/genetics , Fibroblasts/radiation effects , Fibroblasts/metabolism , Melanins/biosynthesis , Melanins/metabolism , Melanocytes/radiation effects , Melanocytes/metabolism , Animals , Tuberous Sclerosis Complex 2 Protein/metabolism , Tuberous Sclerosis Complex 2 Protein/genetics , Ultraviolet Rays/adverse effects , Cells, Cultured , Endoplasmic Reticulum Stress/radiation effects , Primary Cell Culture , Microphthalmia-Associated Transcription Factor/metabolism , Microphthalmia-Associated Transcription Factor/genetics , Mitochondria/radiation effects , Mitochondria/metabolism , Melanogenesis
3.
J Photochem Photobiol B ; 257: 112963, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38908147

ABSTRACT

The therapeutic potential of blue light photobiomodulation in cancer treatment, particularly in inhibiting cell proliferation and promoting cell death, has attracted significant interest. Oral squamous cell carcinoma (OSCC) is a prevalent form of oral cancer, necessitating innovative treatment approaches to improve patient outcomes. In this study, we investigated the effects of 420 nm blue LED light on OSCC and explored the underlying mechanisms. Our results demonstrated that 420 nm blue light effectively reduced OSCC cell viability and migration, and induced G2/M arrest. Moreover, we observed that 420 nm blue light triggered endoplasmic reticulum (ER) stress and mitochondrial dysfunction in OSCC cells, leading to activation of the CHOP signal pathway and alterations in the levels of Bcl-2 and Bax proteins, ultimately promoting cell apoptosis. Additionally, blue light suppressed mitochondrial gene expression, likely due to its damage to mitochondrial DNA. This study highlights the distinct impact of 420 nm blue light on OSCC cells, providing valuable insights into its potential application as a clinical treatment for oral cancer.


Subject(s)
Apoptosis , Carcinoma, Squamous Cell , Cell Survival , Endoplasmic Reticulum Stress , Light , Mitochondria , Mouth Neoplasms , Humans , Endoplasmic Reticulum Stress/radiation effects , Mitochondria/radiation effects , Mitochondria/metabolism , Mouth Neoplasms/radiotherapy , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Cell Line, Tumor , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Apoptosis/radiation effects , Cell Survival/radiation effects , Cell Proliferation/radiation effects , Cell Movement/radiation effects , Signal Transduction/radiation effects , bcl-2-Associated X Protein/metabolism , bcl-2-Associated X Protein/genetics , Transcription Factor CHOP/metabolism , Transcription Factor CHOP/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Blue Light
4.
Biochem Biophys Res Commun ; 724: 150226, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-38865815

ABSTRACT

In patients with high-level radiation exposure, gastrointestinal injury is the main cause of death. Despite the severity of damage to the gastrointestinal tract, no specific therapeutic option is available. Tauroursodeoxycholic acid (TUDCA) is a conjugated form of ursodeoxycholic acid that suppresses endoplasmic reticulum (ER) stress and regulates various cell-signaling pathways. We investigated the effect of TUDCA premedication in alleviating intestinal damage and enhancing the survival of C57BL/6 mice administered a lethal dose (15Gy) of focal abdominal irradiation. TUDCA was administered to mice 1 h before radiation exposure, and reduced apoptosis of the jejunal crypts 12 h after irradiation. At later timepoint (3.5 days), irradiated mice manifested intestinal morphological changes that were detected via histological examination. TUDCA decreased the inflammatory cytokine levels and attenuated the decrease in serum citrulline levels after radiation exposure. Although radiation induced ER stress, TUDCA pretreatment decreased ER stress in the irradiated intestinal cells. The effect of TUDCA indicates the possibility of radiation therapy for cancer in tumor cells. TUDCA did not affect cell proliferation and apoptosis in the intestinal epithelium. TUDCA decreased the invasive ability of the CT26 metastatic colon cancer cell line. Reduced invasion after TUDCA treatment was associated with decreased matrix metalloproteinase (MMP)-7 and MMP-13 expression, which play important roles in invasion and metastasis. This study shows a potential role of TUDCA in protecting against radiation-induced intestinal damage and inhibiting tumor cell migration without any radiation and radiation therapy effect.


Subject(s)
Apoptosis , Endoplasmic Reticulum Stress , Mice, Inbred C57BL , Radiation-Protective Agents , Taurochenodeoxycholic Acid , Animals , Taurochenodeoxycholic Acid/pharmacology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/radiation effects , Apoptosis/drug effects , Apoptosis/radiation effects , Radiation-Protective Agents/pharmacology , Mice , Male , Intestines/radiation effects , Intestines/drug effects , Intestines/pathology , Disease Models, Animal , Intestinal Mucosa/drug effects , Intestinal Mucosa/radiation effects , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Radiation Injuries, Experimental/prevention & control , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/drug therapy , Radiation Injuries, Experimental/metabolism , Matrix Metalloproteinase 13/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects
5.
J Dermatol Sci ; 114(1): 24-33, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38448340

ABSTRACT

BACKGROUND: The unfolded protein response (UPR) is one of the cytoprotective mechanisms against various stresses and essential for the normal function of skin. Skin injury caused by ionizing radiation (IR) is a common side effect of radiotherapy and it is unclear how UPR affects IR-induced skin injury. OBJECTIVES: To verify the effect of UPR on IR-induced DNA damage in keratinocytes and the relation between an endoplasmic reticulum (ER) protein KTN1 and UPR. METHODS: All experiments were performed on keratinocytes models: HaCaT and HEK-A. ER lumen and the expression levels of KTN1 and UPR pathway proteins (PERK, IRE1α and ATF6) were examined by transmission electron microscopy and immunoblotting, respectively. 4-PBA, an UPR inhibitor, was used to detected its effects on DNA damage and cell proliferation. Subsequently, the effects of KTN1 deletion on UPR, DNA damage and cell proliferation after IR were detected. Tunicamycin was used to reactivate UPR and then we examined its effects on DNA damage. RESULTS: UPR was activated by IR in keratinocytes. Inhibition of UPR aggravated DNA damage and suppressed cell proliferation after IR. KTN1 expression was upregulated by IR and KTN1 depletion reduced ER expansion and the expression of UPR-related proteins. Moreover, KTN1 depletion aggravated DNA damage and suppressed cell proliferation after IR could reversed by reactivation of UPR. CONCLUSION: KTN1 deletion aggravates IR-induced keratinocyte DNA damage via inhibiting UPR. Our findings provide new insights into the mechanisms of keratinocytes in response to IR-induced damage.


Subject(s)
Cell Proliferation , DNA Damage , HaCaT Cells , Keratinocytes , Radiation, Ionizing , Unfolded Protein Response , Humans , Cell Line , Cell Proliferation/radiation effects , Cell Proliferation/drug effects , DNA Damage/radiation effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/radiation effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Stress/radiation effects , Endoplasmic Reticulum Stress/drug effects , Keratinocytes/radiation effects , Keratinocytes/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Skin/radiation effects , Skin/pathology , Skin/cytology , Skin/drug effects , Skin/metabolism , Unfolded Protein Response/radiation effects , Unfolded Protein Response/drug effects
6.
ACS Nano ; 18(9): 7267-7286, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38382065

ABSTRACT

Cancer progression and treatment-associated cellular stress impairs therapeutic outcome by inducing resistance. Endoplasmic reticulum (ER) stress is responsible for core events. Aberrant activation of stress sensors and their downstream components to disrupt homeostasis have emerged as vital regulators of tumor progression as well as response to cancer therapy. Here, an orchestrated nanophotoinducer (ERsNP) results in specific tumor ER-homing, induces hyperthermia and mounting oxidative stress associated reactive oxygen species (ROS), and provokes intense and lethal ER stress upon near-infrared laser irradiation. The strengthened "dying" of ER stress and ROS subsequently induce apoptosis for both primary and abscopal B16F10 and GL261 tumors, and promote damage-associated molecular patterns to evoke stress-dependent immunogenic cell death effects and release "self-antigens". Thus, there is a cascade to activate maturation of dendritic cells, reprogram myeloid-derived suppressor cells to manipulate immunosuppression, and recruit cytotoxic T lymphocytes and effective antitumor response. The long-term protection against tumor recurrence is realized through cascaded combinatorial preoperative and postoperative photoimmunotherapy including the chemokine (C-C motif) receptor 2 antagonist, ERsNP upon laser irradiation, and an immune checkpoint inhibitor. The results highlight great promise of the orchestrated nanophotoinducer to exert potent immunogenic cell stress and death by reinforcing ER stress and oxidative stress to boost cancer photoimmunotherapy.


Subject(s)
Neoplasms , Humans , Reactive Oxygen Species/metabolism , Neoplasms/therapy , Endoplasmic Reticulum Stress/radiation effects , Oxidative Stress , Apoptosis , Cell Line, Tumor
7.
J Mol Histol ; 53(1): 75-83, 2022 Feb.
Article in English | MEDLINE | ID: mdl-34676487

ABSTRACT

Although endoplasmic reticulum (ER) stress is thought to be involved in various diseases such as cancer, metabolic, and inflammatory disorders, the relationship between ER stress and bone diseases, are remains unclear. Tunicamycin-treated MC3T3-E1 osteoblasts were used as the ER stress model in this study. 635 nm light-emitting diode irradiation (635 nm-IR) was carried out for 1 h before and after inducing ER stress. To investigate the effects of 635 nm-IR on ER stress-induced MC3T3-E1 osteoblasts and the underlying mechanism, western blot, reverse transcription polymerase chain reaction, alkaline phosphatase and Alizarin red staining, 2',7'-dichlorodyhydrofluorescein diacetate assay, Fluo-3AM and immunocytochemistry were performed. Pretreatment with 635 nm-IR effectively prevented intracellular reactive oxygen species production and alleviated ER stress through the pancreatic ER kinase (PERK)-eukaryotic initiation factor 2 (eIF2)-activating transcription factor 4 (ATF4)-nuclear factor-like 2 (Nrf2) signaling pathway. Hence, 635 nm-IR may serve a protective role in the treatment of ER stress-related bone diseases.


Subject(s)
Endoplasmic Reticulum Stress/radiation effects , Lasers, Semiconductor , Osteoblasts/radiation effects , 3T3 Cells , Activating Transcription Factor 4/metabolism , Alkaline Phosphatase/metabolism , Animals , Blotting, Western , Cell Survival , Cells, Cultured , Eukaryotic Initiation Factor-2/metabolism , Mice , NF-E2-Related Factor 2/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Reactive Oxygen Species/metabolism , Real-Time Polymerase Chain Reaction , Signal Transduction
8.
Reprod Sci ; 29(3): 944-954, 2022 03.
Article in English | MEDLINE | ID: mdl-34642916

ABSTRACT

Today, infertility affects 15% of couples and half of this rate is due to reproductive problems in men. Radiation-induced damage to the testicles causes sterility depending on the dose. Radiation causes endoplasmic reticulum (ER) stress and ER stress induces apoptosis. In this study, the effect of human amniotic membrane-derived mesenchymal stem cells (hAMSCs) and conditioned medium (hAMSCs-CM) on testicular damage induced by ionizing radiation is aimed to be elucidated through ER stress and apoptosis mechanisms. Six gray scrotal irradiation was used to create a testicular injury model. hAMSCs isolated and characterized with immunofluorescence and flow cytometry, while 2.5 × 105 hAMSCs were transplanted into testis and hAMSCs-CM was applied. Fertility assessment was performed. Expressions of ER stress markers GRP78, Ire1, Chop and Caspase-12, and Caspase-3 were determined. TUNEL was performed. Serum FSH, LH, and testosterone were measured. After hAMSC transplantation and administration of hAMSCs-CM, offsprings were obtained. Seminiferous tubule diameter and seminiferous epithelial height increased. The expression of GRP78, IRE1α, CHOP, Caspase-12, and Caspase-3 decreased. Percentages of tunel positive cells decreased. While FSH and LH levels decreased, testosterone increased. After irradiation, both hAMSCs transplantation and paracrine activity of hAMSCs may have a role in reducing ER stress by suppressing the UPR response. Decrease in FSH and LH and increase in testosterone level after MSCs transplantation may have contributed to the improvement of spermatogenesis. Thus, it can be said that MSCs derived from human amniotic membrane can improve ionized radiation-induced testicular damage by reducing ER stress and apoptosis.


Subject(s)
Amnion/cytology , Apoptosis/radiation effects , Endoplasmic Reticulum Stress/radiation effects , Infertility, Male/etiology , Infertility, Male/therapy , Mesenchymal Stem Cell Transplantation , Testis/radiation effects , Animals , Culture Media, Conditioned , Female , Humans , Male , Rats
9.
Cell Death Dis ; 12(11): 1029, 2021 10 29.
Article in English | MEDLINE | ID: mdl-34716300

ABSTRACT

Breast cancer is a major threat to women's health and estrogen receptor-positive (ER+) breast cancer exhibits the highest incidence among these cancers. As the primary estrogen, estradiol strongly promotes cellular proliferation and radiotherapy, as a standard treatment, exerts an excellent therapeutic effect on ER+ breast cancer. Therefore, we herein wished to explore the mechanism(s) underlying the inhibitory effects of radiation on the proliferation of ER+ breast cancer cells. We used the ER+ breast cancer cell lines MCF7 and T47D, and their complementary tamoxifen-resistant cell lines in our study. The aforementioned cells were irradiated at different doses of X-rays with or without exogenous estradiol. CCK8 and clone-formation assays were used to detect cellular proliferation, enzyme-linked immunosorbent assay (ELISA) to determine estradiol secretion, western immunoblotting analysis and quantitative real-time PCR to evaluate the expression of proteins, and immunofluorescence to track endoplasmic reticulum stress-related processes. Finally, BALB/C tumor-bearing nude mice were irradiated with X-rays to explore the protein expression in tumors using immunohistochemistry. We found that ionizing radiation significantly reduced the phosphorylation of estrogen receptors and the secretion of estradiol by ER+ breast cancer cells. CYP19A (aromatase) is an enzyme located in the endoplasmic reticulum, which plays a critical role in estradiol synthesis (aromatization), and we further demonstrated that ionizing radiation could induce endoplasmic reticulum stress with or without exogenous estradiol supplementation, and that it downregulated the expression of CYP19A through ER-phagy. In addition, ionizing radiation also promoted lysosomal degradation of CYP19A, reduced estradiol synthesis, and inhibited the proliferation of tamoxifen-resistant ER+ breast cancer cells. We concluded that ionizing radiation downregulated the expression of CYP19A and reduced estradiol synthesis by inducing endoplasmic reticulum stress in ER+ breast cancer cells, thereby ultimately inhibiting cellular proliferation.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/radiotherapy , Cell Proliferation/radiation effects , Down-Regulation/radiation effects , Endoplasmic Reticulum Stress/radiation effects , Estradiol/biosynthesis , Radiation, Ionizing , Receptors, Estrogen/metabolism , Signal Transduction/radiation effects , Animals , Aromatase/metabolism , Breast Neoplasms/pathology , Drug Resistance, Neoplasm/radiation effects , Estradiol/pharmacology , Female , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Nude , Phosphorylation/radiation effects , Signal Transduction/drug effects , Tamoxifen/pharmacology , Treatment Outcome , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
10.
J Radiat Res ; 62(5): 782-792, 2021 Sep 13.
Article in English | MEDLINE | ID: mdl-34265852

ABSTRACT

The mammalian target of rapamycin (mTOR) is a sensor of nutrient status and plays an important role in cell growth and metabolism. Although inhibition of mTOR signaling promotes tumor cell death and several mTOR inhibitors have been used clinically, recent reports have shown that co-treatment with MHY1485, an mTOR activator, enhances the anti-cancer effects of anti-PD-1 antibody and 5-fluorouracil. However, it remains unclear whether MHY1485 treatment alters the effects of radiation on tumor cells. In this study, the radiosensitizing effects of MHY1485 were investigated using murine CT26 and LLC cell lines. We examined mTOR signaling, tumor cell growth, colony formation, apoptosis, senescence, oxidative stress, p21 accumulation and endoplasmic reticulum (ER) stress levels in cells treated with MHY1485 and radiation, either alone or together. We found that MHY1485 treatment inhibited growth and colony formation in both cell lines under irradiation and no-irradiation conditions, results that were not fully consistent with MHY1485's known role in activating mTOR signaling. Furthermore, we found that combined treatment with MHY1485 and radiation significantly increased apoptosis and senescence in tumor cells in association with oxidative stress, ER stress and p21 stabilization, compared to radiation treatment alone. Our results suggested that MHY1485 enhances the radiosensitivity of tumor cells by a mechanism that may differ from MHY1485's role in mTOR activation.


Subject(s)
Apoptosis/drug effects , Cellular Senescence/drug effects , Morpholines/pharmacology , Neoplasm Proteins/agonists , TOR Serine-Threonine Kinases/drug effects , Triazines/pharmacology , Animals , Apoptosis/radiation effects , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/pathology , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Cellular Senescence/radiation effects , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Drug Screening Assays, Antitumor , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/radiation effects , Genes, p53 , Genes, ras , Lipid Peroxidation/drug effects , Lipid Peroxidation/radiation effects , Mice , Mitochondria/drug effects , Mitochondria/radiation effects , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tumor Stem Cell Assay
11.
Biochem Biophys Res Commun ; 552: 183-190, 2021 05 07.
Article in English | MEDLINE | ID: mdl-33751936

ABSTRACT

Malignant melanoma is a critical and aggressive skin tumor with a steeply rising incidence and a less favorable prognosis due to the lack of efficient treatment. Photodynamic therapy (PDT) is a new promising treatment for this tumor through photosensitizers-mediated oxidative cytotoxicity. In this study, we explored the role of berberine-mediated PDT (BBR-PDT) in the anti-proliferative effect on human malignant melanoma cells (MMCs). We found that there were significant differences between MMCs with BBR-PDT and MMCs with BBR or PDT only. Further research showed that BBR-PDT induced apoptosis via up-regulating the expression of cleaved caspase-3 protein. We also observed that LC3-related autophagy level was upregulated in MMCs with BBR-PDT. Besides, it was also found that BBR-PDT activated endoplasmic reticulum (ER) stress, involving a dramatic increase in reactive oxygen species (ROS). Interestingly, the knockdown of CHOP protein expression inhibited apoptosis, autophagy and ER stress levels caused by BBR-PDT, suggesting that CHOP protein may be related to apoptosis, autophagy and ER stress in MMCs with BBR-PDT. Collectively, our results indicated that BBR-PDT had an essential impact on MMCs' growth inhibition, and therefore may reveal the possibility of developing BBR-PDT into human malignant melanoma.


Subject(s)
Apoptosis/drug effects , Autophagy/drug effects , Berberine/pharmacology , Endoplasmic Reticulum Stress/drug effects , Melanoma/therapy , Photochemotherapy/methods , Transcription Factor CHOP/metabolism , Apoptosis/radiation effects , Autophagy/radiation effects , Berberine/chemistry , Blotting, Western , Cell Line, Tumor , Endoplasmic Reticulum Stress/radiation effects , Humans , Light , Melanoma/metabolism , Melanoma/pathology , Molecular Structure , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Signal Transduction/radiation effects
12.
Radiat Res ; 194(3): 236-245, 2020 09 16.
Article in English | MEDLINE | ID: mdl-32942301

ABSTRACT

Autophagy has been reported to play a radioresistance role in high-dose-rate irradiation. However, its mechanisms and roles in continuous low-dose-rate (CLDR) irradiation have not been clearly understood. Iodine-125 (I-125) seed brachytherapy is a modality of CLDR irradiation and has been used in the treatment of various cancers. In this study, we investigated the mechanisms and roles of autophagy induced by I-125 seed radiation in human esophageal squamous cell carcinoma (ESCC) cell lines (Eca-109 and EC-109) and a xenograft mouse model. The results of this work showed that I-125 seed radiation induced a dose-dependent increase in autophagy in both cell lines. In Eca-109 cells, I-125 seed radiation-induced endoplasmic reticulum (ER) stress, manifesting as the increased levels of intracellular Ca2+ and Grp78/BiP, and activated PERK-eIF2α, IRE1, and ATF6 pathways of the unfolded protein response. Knockdown of PERK led to the decreased expression of autophagy marker, LC3B-II. Inhibition of autophagy by chloroquine or knockdown of ATG5 enhanced I-125 seed radiation-induced cell proliferation inhibition and apoptosis. Interestingly, chloroquine did not aggravate ER stress but promoted apoptosis via the mitochondrial pathway. The animal experiment showed that inhibition of autophagy by chloroquine improved the efficacy of I-125 seed radiation. In summary, our data demonstrate that I-125 seed CLDR radiation induces ER stress-mediated autophagy in ESCC. Autophagy plays a pro-survival role in I-125 seed CLDR irradiation, and chloroquine is a potential candidate for use in combination therapy with I-125 seed radiation treatment to improve efficacy against ESCC.


Subject(s)
Autophagy/radiation effects , Endoplasmic Reticulum Stress/radiation effects , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/radiotherapy , Iodine Radioisotopes/therapeutic use , Cell Line, Tumor , Cell Survival/radiation effects , Endoplasmic Reticulum Chaperone BiP , Humans , Mitochondria/metabolism , Mitochondria/radiation effects
13.
Aging (Albany NY) ; 12(16): 16579-16596, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32858529

ABSTRACT

Excessive light exposure is a principal environmental factor, which can cause damage to photoreceptors and retinal pigment epithelium (RPE) cells and may accelerate the progression of age-related macular degeneration (AMD). In this study, oxidative stress, endoplasmic reticulum (ER) stress and autophagy caused by light exposure were evaluated in vitro and in vivo. Light exposure caused severe photo-oxidative stress and ER stress in photoreceptors (661W cells) and RPE cells (ARPE-19 cells). Suppressing either oxidative stress or ER stress was protective against light damage in 661W and ARPE-19 cells and N-acetyl-L-cysteine treatment markedly inhibited the activation of ER stress caused by light exposure. Moreover, suppressing autophagy with 3-methyladenine significantly attenuated light-induced cell death. Additionally, inhibiting ER stress either by knocking down PERK signals or with GSK2606414 treatment remarkably suppressed prolonged autophagy and protected the cells against light injury. In vivo experiments verified neuroprotection via inhibiting ER stress-related autophagy in light-damaged retinas of mice. In conclusion, the above results suggest that light-induced photo-oxidative stress may trigger subsequent activation of ER stress and prolonged autophagy in photoreceptors and RPE cells. Suppressing ER stress may abrogate over-activated autophagy and protect the retina against light injury.


Subject(s)
Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Light/adverse effects , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Photoreceptor Cells, Vertebrate/drug effects , Retinal Pigment Epithelium/drug effects , Acetylcysteine/pharmacology , Adenine/analogs & derivatives , Adenine/pharmacology , Animals , Antioxidants/pharmacology , Autophagy/radiation effects , Cell Line , Endoplasmic Reticulum Stress/radiation effects , Humans , Indoles/pharmacology , Male , Mice, Inbred C57BL , Oxidative Stress/radiation effects , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Photoreceptor Cells, Vertebrate/radiation effects , Retinal Pigment Epithelium/metabolism , Retinal Pigment Epithelium/pathology , Retinal Pigment Epithelium/radiation effects , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
14.
J Dermatol Sci ; 98(1): 41-49, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32376153

ABSTRACT

BACKGROUND: Endoplasmic reticulum (ER) calcium depletion-induced ER stress is a crucial signal for keratinocyte differentiation and barrier homeostasis, but its effects on the epidermal tight junction (TJ) have not been characterized. Ultraviolet B (UVB) causes ER calcium release in keratinocytes and disrupts epidermal TJ, however, the involvement of ER stress in the UVB-induced TJ alterations remains unknown. OBJECTIVES: To investigate the effect of ER stress by pharmacological ER calcium depletion or UVB on the TJ integrity in normal human epidermal keratinocytes (NHEK). METHODS: NHEK were exposed to ER calcium pump inhibitor thapsigargin (Tg) or UVB. ER stress markers and TJ molecules expression, TJ and F-actin structures, and TJ barrier function were analyzed. RESULTS: Tg or UVB exposure dose-dependently triggered unfolded protein response (UPR) in NHEK. Low dose Tg induced the IRE1α-XBP1 pathway and strengthened TJ barrier. Contrary, high dose Tg activated PERK phosphorylation and disrupted TJ by F-actin disorganization. UVB disrupted TJ and F-actin structures dose dependently. IRE1α RNase inhibition induced or exacerbated TJ and F-actin disruption in the presence of low dose Tg or UVB. High dose Tg increased RhoA activity. 4-PBA or Rho kinase (ROCK) inhibitor partially prevented the disruption of TJ and F-actin following high dose Tg or UVB. CONCLUSIONS: ER stress has bimodal effects on the epidermal TJ depending on its intensity. The IRE1α pathway is critical for the maintenance of TJ integrity during mild ER stress. Severe ER stress-induced UPR or ROCK signalling mediates the disruption of TJ through cytoskeletal disorganization during severe ER stress.


Subject(s)
Calcium/metabolism , Endoplasmic Reticulum Stress/radiation effects , Keratinocytes/pathology , Tight Junctions/pathology , Ultraviolet Rays/adverse effects , Amides/pharmacology , Cell Line , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/radiation effects , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/radiation effects , Phenylbutyrates/pharmacology , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Signal Transduction/drug effects , Signal Transduction/radiation effects , Tight Junctions/drug effects , Tight Junctions/metabolism , Tight Junctions/radiation effects , Unfolded Protein Response/drug effects , Unfolded Protein Response/radiation effects , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
15.
Biochem Biophys Res Commun ; 524(4): 869-875, 2020 04 16.
Article in English | MEDLINE | ID: mdl-32051089

ABSTRACT

Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a neuroprotective factor produced in response to endoplasmic reticulum (ER) stress induced by various stressors, but its involvement in the radioresistance of tumor cells is unknown. Here, we found that MANF is released after γ-irradiation (2 Gy and 4 Gy) of B16 melanoma cells, and its release was suppressed by 4-phenylbutyric acid, an ER stress inhibitor. MANF was not released after low-dose (1 Gy) γ-irradiation, but pretreatment of 1 Gy-irradiated cells with recombinant MANF enhanced the cellular DNA damage response and attenuated reproductive cell death. In MANF-knockdown cells, the DNA damage response and p53 activation by γ-irradiation (2 Gy) were suppressed, and reproductive cell death was increased. MANF also activated the ERK signaling pathway. Our findings raise the possibility that MANF could be a new target for overcoming radioresistance.


Subject(s)
Endoplasmic Reticulum Stress/radiation effects , Endoplasmic Reticulum/radiation effects , Gene Expression Regulation, Neoplastic , Nerve Growth Factors/genetics , Radiation Tolerance/genetics , Animals , Cell Line, Tumor , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Gamma Rays , Melanoma, Experimental/genetics , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Melanoma, Experimental/radiotherapy , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Nerve Growth Factors/antagonists & inhibitors , Nerve Growth Factors/metabolism , Phenylbutyrates/pharmacology , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
16.
Nano Lett ; 20(3): 1928-1933, 2020 03 11.
Article in English | MEDLINE | ID: mdl-32073871

ABSTRACT

Immunogenic cell death (ICD) elicited by photodynamic therapy (PDT) is mediated through generation of reactive oxygen species (ROS) that induce endoplasmic reticulum (ER) stress. However, the half-life of ROS is very short and the intracellular diffusion depth is limited, which impairs ER localization and thus limits ER stress induction. To solve the problem, we synthesized reduction-sensitive Ds-sP NPs (PEG-s-s-1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[amino(polyethylene glycol)-2000] nanoparticles) loaded with an efficient ER-targeting photosensitizer TCPP-TER (4,4',4″,4'″-(porphyrin-5,10,15,20-tetrayl)tetrakis(N-(2-((4-methylphenyl)sulfonamido)ethyl)benzamide). The resulting Ds-sP/TCPP-TER NPs could selectively accumulate in the ER and locally generate ROS under near-infrared (NIR) laser irradiation, which induced ER stress, amplified ICD, and activated immune cells, leading to augmented immunotherapy effect. This study presents a novel ICD amplifying, ER-targeting PDT strategy that can effectively eradicate primary tumors under NIR exposure, as well as distant tumors through an abscopal effect.


Subject(s)
Drug Delivery Systems , Endoplasmic Reticulum Stress , Endoplasmic Reticulum , Immunotherapy , Infrared Rays , Neoplasms, Experimental , Animals , Cell Death/drug effects , Cell Death/immunology , Cell Line, Tumor , Endoplasmic Reticulum/immunology , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/immunology , Endoplasmic Reticulum Stress/radiation effects , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/immunology , Neoplasms, Experimental/pathology , Neoplasms, Experimental/therapy , Reactive Oxygen Species/immunology
17.
Mol Reprod Dev ; 87(1): 161-173, 2020 01.
Article in English | MEDLINE | ID: mdl-31793725

ABSTRACT

DNA damage associated with assisted reproductive technologies is an important factor affecting gamete fertility and embryo development. Activation of the TGR5 receptor by tauroursodeoxycholic acid (TUDCA) has been shown to reduce endoplasmic reticulum (ER) stress in embryos; however, its effect on genome damage responses (GDR) activation to facilitate DNA damage repair has not been examined. This study aimed to investigate the effect of TUDCA on DNA damage repair and embryo development. In a porcine model of ultraviolet light (UV)-induced nuclear stress, TUDCA reduced DNA damage and ER stress in developing embryos, as measured by γH2AX and glucose-regulated protein 78 immunofluorescence, respectively. TUDCA was equally able to rescue early embryo development. No difference in total cell number, DNA damage, or percentage of apoptotic cells, measured by cleaved caspase 3 immunofluorescence, was noted in embryos that reached the blastocyst stage. Interestingly, Dicer-substrate short interfering RNA-mediated disruption of TGR5 signaling abrogated the beneficial effects of TUDCA on UV-treated embryos. Quantitative PCR analysis revealed activation of the GDR, through increased messenger RNA abundance of DNAPK, 53BP1, and DNA ligase IV, as well as the ER stress response, through increased spliced XBP1 and X-linked inhibitor of apoptosis. Results from this study demonstrated that TUDCA activates TGR5-mediated signaling to reduce DNA damage and improve embryo development after UV exposure.


Subject(s)
DNA Damage/drug effects , DNA Repair/drug effects , Embryonic Development/drug effects , Receptors, G-Protein-Coupled/metabolism , Swine/embryology , Taurochenodeoxycholic Acid/pharmacology , Animals , Apoptosis/genetics , Apoptosis/radiation effects , Blastocyst/cytology , Blastocyst/radiation effects , Cells, Cultured , DNA Damage/genetics , DNA Damage/radiation effects , DNA Repair/genetics , DNA Repair/radiation effects , Embryonic Development/genetics , Embryonic Development/radiation effects , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/genetics , Endoplasmic Reticulum Stress/radiation effects , Female , Fertilization in Vitro/methods , Gene Knockdown Techniques , In Vitro Oocyte Maturation Techniques/methods , Oocyte Retrieval/methods , Ovary/cytology , Receptors, G-Protein-Coupled/genetics , Signal Transduction/genetics , Ultraviolet Rays , Unfolded Protein Response/genetics , Unfolded Protein Response/radiation effects , Zygote/radiation effects
18.
Metallomics ; 12(2): 290-300, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31872196

ABSTRACT

Resveratrol displays cardioprotective activity; however, its mechanism of action remains unclear. In the current study, resveratrol-induced myocardial protection from endoplasmic reticulum stress (ERS) was investigated, focusing on the roles of Zn2+ and the mitochondrial permeability transition pore (mPTP). We found, using the MTT/LDH kit, that 2-DG-induced ERS significantly decreased H9c2 cell viability. Resveratrol markedly inhibited the expression of endoplasmic reticulum chaperone GRP 78/94 and ERS-related apoptosis proteins CHOP, Caspase12, and JNK induced by 2-DG. The zinc ion chelator TPEN, and ERK/GSK-3ß inhibitors PD98059 and SB216763 and their siRNAs blocked resveratrol function. The AKT inhibitor LY294002 and siRNA did not alter the action of resveratrol. In addition, resveratrol significantly increased the phosphorylation of ERK and GSK-3ß. Resveratrol prevented 2-DG-induced mPTP opening and increased intracellular Zn2+ concentration indicated by TMRE and Newport Green DCF fluorescence intensity, which were further abrogated by ERK/GSK-3ß inhibitors and siRNAs. Our data suggested that resveratrol protected cardiac cells from ERS by mobilizing intracellular Zn2+ and preventing mPTP opening through the ERK/GSK-3ß but not PI3K/AKT signaling pathway.


Subject(s)
Cardiotonic Agents/pharmacology , Endoplasmic Reticulum Stress/radiation effects , Mitochondrial Permeability Transition Pore/metabolism , Myocytes, Cardiac/drug effects , Resveratrol/pharmacology , Zinc/metabolism , Animals , Cell Line , Chromones/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Morpholines/metabolism , Myocytes, Cardiac/metabolism , Rats , Signal Transduction
19.
J Photochem Photobiol B ; 202: 111720, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31841988

ABSTRACT

It has been widely reported that ultraviolet-B (UV-B) radiation is the main extrinsic etiological agent that causes skin photodamage. UV-B exposure mediated photodamage (photo-aging/photo-carcinogenesis) to human skin is caused due to several physiological events at tissue, cellular and molecular levels that lead to impairment of skin function and integrity. In the present study, we investigated the protective role of Trigonelline (TG) against UV-B induced photo-damage in Human Dermal Fibroblasts (Hs68 cells) and Balb/C mice. We exposed human skin fibroblasts and Balb/C mice to UV-B radiation and evaluated various parameters of cellular damage, including, oxidative stress, cytosolic calcium (Ca2+) levels, apoptotic and ER-stress marker proteins. We found that UV-B irradiation induced ROS generation lead to the depletion of endoplasmic reticulum (ER) calcium and increased the expression of ER stress protein markers (phosphorylated elf2α, CHOP, ATF4) as well as apoptotic protein markers (Bcl2, Bax and caspase-9) in a dose and time dependent manner in Hs68 cells. We then determined the effect of TG treatment on UV-B -induced cell death in Hs68 cells and observed that cells exposed to UV-B radiation and treated with TG had a significantly higher survival rate compared to cells exposed to UV-B radiation alone. TG treatment successfully reduced oxidative stress; restored Ca2+ homeostasis and re-established the ER function and prevented apoptotic cell death process. Our results suggest that TG can be used as a potential therapeutic/cosmeceutic agent in preventing skin photo-damage.


Subject(s)
Alkaloids/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Endoplasmic Reticulum Stress/drug effects , Oxidative Stress/drug effects , Protective Agents/pharmacology , Ultraviolet Rays , Animals , Apoptosis/radiation effects , Caspase 9/genetics , Caspase 9/metabolism , Cell Line , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/radiation effects , Eukaryotic Initiation Factor-1/genetics , Eukaryotic Initiation Factor-1/metabolism , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gene Expression/drug effects , Gene Expression/radiation effects , Humans , Mice , Mice, Inbred BALB C , Oxidative Stress/radiation effects , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
20.
Biochem Biophys Res Commun ; 522(3): 612-617, 2020 02 12.
Article in English | MEDLINE | ID: mdl-31785812

ABSTRACT

Endoplasmic reticulum (ER) stress is a conserved cellular process for cells to clear unfolded or misfolded proteins and maintain cell homeostasis under stress conditions. Autophagy may act as a pro-survival strategy to cope with multiple stress conditions in tumor progression and distant metastasis. Although many studies have demonstrated that there is a close correlation between radiation-induced ER stress and autophagy, the molecular mechanisms currently remain unclear. In the present study, we performed an in vivo study concerning the effect of autophagy induced by ER stress on the radiosensitivity of mouse sarcoma using X-rays. Our results documented that X-rays could induce ER stress in sarcoma and then autophagy was activated by unfolded protein response (UPR) through the IRE1-JNK-pBcl2-Beclin1 signaling axis. The induction of autophagy caused a decline in cell apoptosis while inhibiting the autophagy resulted in increased apoptosis and inhibition of tumor progression. Combined treatment of X-ray exposure and chloroquine increased ER stress-related apoptosis and enhanced the radiosensitivity of mouse sarcoma that was not sensitive to X-ray irradiation alone. Thus, our study indicates that inhibition of ER stress-induced autophagy might be a novel strategy to improve the efficacy of radiotherapy against radioresistant sarcoma.


Subject(s)
Antimalarials/therapeutic use , Apoptosis/radiation effects , Autophagy/radiation effects , Chloroquine/therapeutic use , Endoplasmic Reticulum Stress/radiation effects , Sarcoma/radiotherapy , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Endoplasmic Reticulum Stress/drug effects , Female , Mice , Mice, Inbred BALB C , Radiation Tolerance/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL