Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
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
2.
Aging (Albany NY) ; 13(5): 7300-7313, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33640883

ABSTRACT

mTOR is well known to promote tumor growth but its roles in enhancing chemotherapy and radiotherapy have not been well studied. mTOR inhibition by rapamycin can sensitize cancer cells to radiotherapy. Here we show that Maf1 is required for rapamycin to increase radio-sensitivity in A549 lung cancer cells. In response to ionizing radiation (IR), Maf1 is inhibited by Akt-dependent re-phosphorylation, which activates mitochondrial unfolded protein response (UPRmt) through ATF5. Rapamycin suppresses IR-induced Maf1 re-phosphorylation and UPRmt activation in A549 cells, resulting in increased sensitivity to IR-mediated cytotoxicity. Consistently, Maf1 knockdown activates ATF5-transcription of mtHSP70 and HSP60, enhances mitochondrial membrane potential, reduces intracellular ROS levels and dampens rapamycin's effect on increasing IR-mediated cytotoxicity. In addition, Maf1 overexpression suppresses ethidium bromide-induced UPRmt and enhances IR-mediated cytotoxicity. Supporting our cell-based studies, elevated expression of UPRmt makers (mtHSP70 and HSP60) are associated with poor prognosis in patients with lung adenocarcinoma (LAUD). Together, our study reveals a novel role of Maf1-UPRmt axis in mediating rapamycin's enhancing effect on IR sensitivity in A549 lung cancer cells.


Subject(s)
A549 Cells/metabolism , Activating Transcription Factors/metabolism , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Repressor Proteins/metabolism , Sirolimus/pharmacology , Unfolded Protein Response/drug effects , A549 Cells/drug effects , A549 Cells/radiation effects , Blotting, Western , Flow Cytometry , Humans , Lung Neoplasms/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Mitochondria/radiation effects , Phosphorylation , Real-Time Polymerase Chain Reaction , Unfolded Protein Response/radiation effects
3.
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
4.
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
5.
Int J Mol Sci ; 20(11)2019 May 30.
Article in English | MEDLINE | ID: mdl-31151143

ABSTRACT

Head and neck squamous cell carcinomas (HNSCC) encompass a heterogeneous group of solid tumors that arise from the upper aerodigestive tract. The tumor cells face multiple challenges including an acute demand of protein synthesis often driven by oncogene activation, limited nutrient and oxygen supply and exposure to chemo/radiotherapy, which forces them to develop adaptive mechanisms such as the Unfolded Protein Response (UPR). It is now well documented that the UPR, a homeostatic mechanism, is induced at different stages of cancer progression in response to intrinsic (oncogenic activation) or extrinsic (microenvironment) perturbations. This review will discuss the role of the UPR in HNSCC as well as in the key processes that characterize the physiology of HNSCC. The role of the UPR in the clinical context of HNSCC will also be addressed.


Subject(s)
Squamous Cell Carcinoma of Head and Neck/metabolism , Unfolded Protein Response , Animals , Biomarkers , Cell Transformation, Neoplastic/metabolism , Combined Modality Therapy , Humans , Molecular Targeted Therapy , Prognosis , Signal Transduction , Squamous Cell Carcinoma of Head and Neck/etiology , Squamous Cell Carcinoma of Head and Neck/therapy , Treatment Outcome , Unfolded Protein Response/drug effects , Unfolded Protein Response/radiation effects
6.
J Photochem Photobiol B ; 194: 84-95, 2019 May.
Article in English | MEDLINE | ID: mdl-30933875

ABSTRACT

Ultraviolet (UV)-B radiation is a major environmental risk factor that is responsible for the development and progression of many skin cancers. Apigenin, a type of bioflavonoid, has been reported to inhibit UVB-induced skin cancer. However, how apigenin functions in keratinocytes with UV damage remains unclear. In this study, by lactate dehydrogenase (LDH) release assay, we found that apigenin treatment increased cell death in the primary human epidermal keratinocytes (HEKs) and the cutaneous squamous cell carcinoma cell line COLO-16. Apigenin treatment reduced microtubule-associated protein 1 light chain 3 (LC3)-II turnover, acridine orange staining and GFP-LC3 puncta in both cell types, suggesting autophagy inhibition. However, apigenin treatment restored the inhibition of autophagy in UVB-challenged HEKs. Moreover, apigenin treatment restored the UVB-induced downregulation of ataxia-telangiectasia mutated (ATM), ataxia-telangiectasia, Rad3-related (ATR) and the unfolded protein response (UPR) regulatory proteins, BiP, IRE1α and PERK in HEKs. Apigenin treatment also inhibited UVB-induced apoptosis and cell death in HEKs. In addition, autophagy inhibition by autophagy-related gene (ATG) 5 RNA interference interrupted apigenin-induced restoration of ATR, ATM and BiP, which were downregulated in HEKs exposed to UVB radiation. Our findings indicate that apigenin exhibits a novel protective effect in keratinocytes with UVB damage, suggesting potential application as a photoprotective agent.


Subject(s)
Apigenin/pharmacology , Autophagy/drug effects , Down-Regulation/drug effects , Keratinocytes/drug effects , Keratinocytes/radiation effects , Ultraviolet Rays/adverse effects , Unfolded Protein Response/drug effects , Autophagy/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Down-Regulation/radiation effects , HEK293 Cells , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Radiation-Protective Agents/pharmacology , Unfolded Protein Response/radiation effects
7.
Autophagy ; 15(8): 1391-1406, 2019 08.
Article in English | MEDLINE | ID: mdl-30773986

ABSTRACT

Cellular effects of ionizing radiation include oxidative damage to macromolecules, unfolded protein response (UPR) and metabolic imbalances. Oxidative stress and UPR have been shown to induce macroautophagy/autophagy in a context-dependent manner and are crucial factors in determining the fate of irradiated cells. However, an in-depth analysis of the relationship between radiation-induced damage and autophagy has not been explored. In the present study, we investigated the relationship between radiation-induced oxidative stress, UPR and autophagy in murine macrophage cells. A close association was observed between radiation-induced oxidative burst, UPR and induction of autophagy, with the possible involvement of EIF2AK3/PERK (eukaryotic translation initiation factor 2 alpha kinase 3) and ERN1/IRE1 (endoplasmic reticulum [ER] to nucleus signaling 1). Inhibitors of either UPR or autophagy reduced the cell survival indicating the importance of these processes after radiation exposure. Moreover, modulation of autophagy affected lethality in the whole body irradiated C57BL/6 mouse. These findings indicate that radiation-induced autophagy is a pro-survival response initiated by oxidative stress and mediated by EIF2AK3 and ERN1. Abbreviations: ACTB: actin, beta; ATF6: activating transcription factor 6; ATG: autophagy-related; BafA1: bafilomycin A1; CQ: chloroquine; DBSA: 3,5-dibromosalicylaldehyde; EIF2AK3: eukaryotic translation initiation factor 2 alpha kinase 3; ERN1: endoplasmic reticulum (ER) to nucleus signaling 1; IR: ionizing radiation; MAP1LC3/LC3: microtubule-associated protein 1 light chain 3; 3-MA: 3-methyladenine; MTOR: mechanistic target of rapamycin kinase; NAC: N-acetyl-L-cysteine; PARP1: poly (ADP-ribose) polymerase family, member 1; 4-PBA: 4-phenylbutyrate; Rap: rapamycin; ROS: reactive oxygen species; UPR: unfolded protein response; XBP1: x-box binding protein 1.


Subject(s)
Autophagy , Endoribonucleases/metabolism , Protein Serine-Threonine Kinases/metabolism , Radiation, Ionizing , eIF-2 Kinase/metabolism , Animals , Apoptosis/radiation effects , Autophagy/radiation effects , Cell Survival , Endoplasmic Reticulum Stress/radiation effects , Female , Mice , Mice, Inbred C57BL , Oxidative Stress/radiation effects , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Unfolded Protein Response/radiation effects
8.
Cell Mol Life Sci ; 76(7): 1255-1273, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30535789

ABSTRACT

This article reviews the current knowledge on the mechanisms of adaptive response to low doses of ionizing radiation or chemical exposure. A better knowledge of these mechanisms is needed to improve our understanding of health risks at low levels of environmental or occupational exposure and their involvement in cancer or non-cancer diseases. This response is orchestrated through a multifaceted cellular program involving the concerted action of diverse stress response pathways. These evolutionary highly conserved defense mechanisms determine the cellular response to chemical and physical aggression. They include DNA damage repair (p53, ATM, PARP pathways), antioxidant response (Nrf2 pathway), immune/inflammatory response (NF-κB pathway), cell survival/death pathway (apoptosis), endoplasmic response to stress (UPR response), and other cytoprotective processes including autophagy, cell cycle regulation, and the unfolded protein response. The coordinated action of these processes induced by low-dose radiation or chemicals produces biological effects that are currently estimated with the linear non-threshold model. These effects are controversial. They are difficult to detect because of their low magnitude, the scarcity of events in humans, and the difficulty of corroborating associations over the long term. Improving our understanding of these biological consequences should help humans and their environment by enabling better risk estimates, the revision of radiation protection standards, and possible therapeutic advances.


Subject(s)
Environmental Pollutants/toxicity , Radiation, Ionizing , Signal Transduction/drug effects , Antioxidants/metabolism , Autophagy/drug effects , Autophagy/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Environmental Pollutants/chemistry , Humans , Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Signal Transduction/radiation effects , Unfolded Protein Response/drug effects , Unfolded Protein Response/radiation effects
9.
Arch Dermatol Res ; 309(7): 541-550, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28631090

ABSTRACT

Stratum corneum forms the UV barrier. The effect of ultraviolet B (UVB) on normal skin was extensively studied; however, its effect on barrier perturbed skin remains undefined. Both barrier perturbation and UVB irradiation induce endoplasmic reticulum (ER) stress and unfolded protein response (UPR) in keratinocytes. Mild ER stress activates homeostatic UPR, while severe ER stress leads to abnormal UPR, promoting apoptosis and inflammation. Here, we investigated UV sensitivity and UVB-induced UPR in barrier-disrupted human skin and the effects of pseudoceramide-dominant emollient on UVB-induced skin responses. Tape-stripped skin of healthy volunteers showed enhanced susceptibility to erythema and augmented proinflammatory cytokines induction following suberythemal UVB irradiation. Suberythemal UVB activated XBP1 in normal skin, while increased CHOP transcription in barrier perturbed skin. After tape stripping, pseudoceramide-dominant emollient was applied for 3 days, and then, the areas were irradiated with suberythemal UVB. Pretreatment with topical pseudoceramide protected against UVB-induced upregulation of IL-1ß, IL-6, and TNF-α transcription and reduced susceptibility to erythema following UVB. Topical pseudoceramide also suppressed suberythemal UVB-induced CHOP transcription in barrier-disrupted skin. Taken together, these data indicate that permeability barrier disruption increases UV sensitivity in human skin, partly via switch the UVB-induced UPR, from homeostatic signals to pro-apoptotic and proinflammatory signals. In addition, we conclude that pseudoceramide-dominant emollient suppresses excessive ER stress induction and CHOP activation following UVB in barrier damaged skin, providing evidence that pseudoceramide-dominant emollients can be promising strategies for photoprotection of the barrier damaged skin.


Subject(s)
Endoplasmic Reticulum Stress/radiation effects , Erythema/drug therapy , Keratinocytes/radiation effects , Spermine/analogs & derivatives , Tyrosine/analogs & derivatives , Ultraviolet Rays/adverse effects , Unfolded Protein Response/radiation effects , Adult , CCAAT-Enhancer-Binding Proteins/biosynthesis , Erythema/prevention & control , Female , Humans , Interleukin-1beta/biosynthesis , Interleukin-1beta/genetics , Interleukin-6/biosynthesis , Interleukin-6/genetics , Keratinocytes/metabolism , RNA, Messenger/biosynthesis , Skin/pathology , Skin/radiation effects , Spermine/pharmacology , Tight Junctions/physiology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics , Tyrosine/pharmacology
10.
Mol Vis ; 23: 52-59, 2017.
Article in English | MEDLINE | ID: mdl-28331281

ABSTRACT

PURPOSE: Blue light is a high-energy emitting light with a short wavelength in the visible light spectrum. Blue light induces photoreceptor apoptosis and causes age-related macular degeneration or retinitis pigmentosa. In the present study, we investigated the roles of endoplasmic reticulum (ER) stress induced by blue light-emitting diode (LED) light exposure in murine photoreceptor cells. METHODS: The murine photoreceptor cell line was incubated and exposed to blue LED light (464 nm blue LED light, 450 lx, 3 to 24 h). The expression of the factors involved in the unfolded protein response pathway was examined using quantitative real-time reverse transcription (RT)-PCR and immunoblot analysis. The aggregation of short-wavelength opsin (S-opsin) in the murine photoreceptor cells was observed with immunostaining. The effect of S-opsin knockdown on ATF4 expression in the murine photoreceptor cell line was also investigated. RESULTS: Exposure to blue LED light increased the bip, atf4, and grp94 mRNA levels, induced the expression of ATF4 protein, and increased the levels of ubiquitinated proteins. Exposure to blue LED light in combination with ER stress inducers (tunicamycin and dithiothreitol) induced the aggregation of S-opsin. S-opsin mRNA knockdown prevented the induction of ATF4 expression in response to exposure to blue LED light. CONCLUSIONS: These findings indicate that the aggregation of S-opsin induced by exposure to blue LED light causes ER stress, and ATF4 activation in particular.


Subject(s)
Activating Transcription Factor 4/metabolism , Light , Photoreceptor Cells, Vertebrate/metabolism , Photoreceptor Cells, Vertebrate/pathology , Rod Opsins/metabolism , Animals , Cell Line , Dithiothreitol/pharmacology , Gene Knockdown Techniques , Mice, Inbred C57BL , Photoreceptor Cells, Vertebrate/drug effects , Photoreceptor Cells, Vertebrate/radiation effects , Polyubiquitin/metabolism , Protein Aggregates/drug effects , Protein Aggregates/radiation effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Tunicamycin/pharmacology , Ubiquitination/drug effects , Ubiquitination/radiation effects , Unfolded Protein Response/drug effects , Unfolded Protein Response/radiation effects
11.
Mol Oncol ; 10(7): 949-65, 2016 08.
Article in English | MEDLINE | ID: mdl-27106131

ABSTRACT

Development of therapeutic resistance is responsible for most prostate cancer (PCa) related mortality. Resistance has been attributed to an acquired or selected cancer stem cell phenotype. Here we report the histone deacetylase inhibitor apicidin (APC) or ER stressor thapsigargin (TG) potentiate paclitaxel (TXL)-induced apoptosis in PCa cells and limit accumulation of cancer stem cells. TXL-induced responses were modulated in the presence of TG with increased accumulation of cells at G1-phase, rearrangement of the cytoskeleton, and changes in cytokine release. Cytoskeletal rearrangement was associated with modulation of the cytoplasmic and mitochondrial unfolded protein response leading to mitochondrial dysfunction and release of proapoptotic proteins from mitochondria. TXL in combination with APC or TG enhanced caspase activation. Importantly, TXL in combination with TG induced caspase activation and apoptosis in X-ray resistant LNCaP cells. Increased release of transforming growth factor-beta (TGF-ß) was observed while phosphorylated ß-catenin level was suppressed with TXL combination treatments. This was accompanied by a decrease in the CD44(+)CD133(+) cancer stem cell-like population, suggesting treatment affects cancer stem cell properties. Taken together, combination treatment with TXL and either APC or TG induces efficient apoptosis in both proliferating and cancer stem cells, suggesting this therapeutic combination may overcome drug resistance and recurrence in PCa.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis , Cytoskeleton/metabolism , Mitochondria/metabolism , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/metabolism , Unfolded Protein Response , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Apoptosis/drug effects , Apoptosis/radiation effects , Caspases/metabolism , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Death/drug effects , Cell Death/radiation effects , Cell Line, Tumor , Cytoskeleton/drug effects , Cytoskeleton/radiation effects , Enzyme Activation/drug effects , G1 Phase/drug effects , G1 Phase/radiation effects , G2 Phase/drug effects , G2 Phase/radiation effects , HSP70 Heat-Shock Proteins/metabolism , Humans , Interferon-gamma/metabolism , Interleukin-8/metabolism , Male , Matrix Metalloproteinases/metabolism , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/radiation effects , Mitochondria/drug effects , Mitochondria/radiation effects , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Stem Cells/radiation effects , Paclitaxel , Peptides, Cyclic/pharmacology , Peptides, Cyclic/therapeutic use , Phosphorylation/drug effects , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Reactive Oxygen Species/metabolism , Thapsigargin/pharmacology , Thapsigargin/therapeutic use , Transforming Growth Factor beta/metabolism , Unfolded Protein Response/drug effects , Unfolded Protein Response/radiation effects , X-Rays , beta Catenin/metabolism
13.
Sci Rep ; 5: 13815, 2015 Sep 04.
Article in English | MEDLINE | ID: mdl-26338671

ABSTRACT

Heavy ion beams have advantages over conventional radiation in radiotherapy due to their superb biological effectiveness and dose conformity. However, little information is currently available concerning the cellular and molecular basis for heavy ion radiation-induced autophagy. In this study, human glioblastoma SHG44 and cervical cancer HeLa cells were irradiated with carbon ions of different linear energy transfers (LETs) and X-rays. Our results revealed increased LC3-II and decreased p62 levels in SHG44 and HeLa cells post-irradiation, indicating marked induction of autophagy. The autophagic level of tumor cells after irradiation increased in a LET-dependent manner and was inversely correlated with the sensitivity to radiations of various qualities. Furthermore, we demonstrated that high-LET carbon ions stimulated the unfolded protein response (UPR) and mediated autophagy via the UPR-eIF2α-CHOP-Akt signaling axis. High-LET carbon ions more severely inhibited Akt-mTOR through UPR to effectively induce autophagy. Thus, the present data could serve as an important radiobiological basis to further understand the molecular mechanisms by which high-LET radiation induces cell death.


Subject(s)
Autophagy/drug effects , Heavy Ions , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/pathology , Oncogene Protein v-akt/metabolism , Unfolded Protein Response/radiation effects , Carbon , Down-Regulation/radiation effects , Enzyme Activation/radiation effects , HeLa Cells , Humans , Neoplasms, Experimental/radiotherapy , Phosphorylation/radiation effects
14.
J Invest Dermatol ; 135(10): 2502-2511, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25950825

ABSTRACT

This study delineates the mechanisms by which UVB regulates protein synthesis in human keratinocytes and the importance of translational control in cell survival. Translation initiation is regulated by phosphorylation of eukaryotic initiation factor 2 (eIF2-P) that causes decreased global protein synthesis coincident with enhanced translation of selected stress-related transcripts, such as activating transcription factor 4 (ATF4). ATF4 is a transcriptional activator of the integrated stress response (ISR) that has cytoprotective functions as well as apoptotic signals through the downstream transcriptional regulator C/EBP homologous protein (CHOP; GADD153/DDIT3). We determined that UVB irradiation is a potent inducer of eIF2-P in keratinocytes, leading to decreased levels of translation initiation. However, expression of ATF4 or CHOP was not induced by UVB as compared with traditional ISR activators. The rationale for this discordant response is that ATF4 mRNA is reduced by UVB, and despite its ability to be preferentially translated, there are diminished levels of available transcript. Forced expression of ATF4 and CHOP protein before UVB irradiation significantly enhanced apoptosis, suggesting that this portion of the ISR is deleterious in keratinocytes following UVB. Inhibition of eIF2-P and translational control reduced viability following UVB that was alleviated by cycloheximide (CHX), indicating that translation repression through eIF2-P is central to keratinocyte survival.


Subject(s)
Activating Transcription Factor 4/metabolism , Apoptosis/radiation effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , Stress, Physiological/physiology , Transcription Factor CHOP/metabolism , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/radiation effects , Apoptosis/physiology , Cells, Cultured , Humans , Immunoblotting , Keratinocytes/cytology , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Transcription Factor CHOP/radiation effects , Ultraviolet Rays/adverse effects , Unfolded Protein Response/physiology , Unfolded Protein Response/radiation effects , eIF-2 Kinase/radiation effects
15.
J Dermatol Sci ; 75(1): 24-35, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24794973

ABSTRACT

BACKGROUND: Exposure of skin to ultraviolet (UV) radiation, an environmental stressor induces number of adverse biological effects (photodamage), including cancer. The damage induced by UV-irradiation in skin cells is initiated by the photochemical generation of reactive oxygen species (ROS) and induction of endoplasmic reticulum (ER) stress and consequent activation of unfolded protein response (UPR). OBJECTIVE: To decipher cellular and molecular events responsible for UV-B mediated ER stress and UPR activation in skin cells. METHODS: The study was performed on human skin fibroblast (Hs68) and keratinocyte (HaCaT) cells exposed to UV-B radiations in lab conditions. Different parameters of UVB induced cellular and molecular changes were analyzed using Western-blotting, microscopic studies and flow cytometry. RESULTS: Our results depicted that UV-B induces an immediate ROS generation that resulted in emptying of ER Ca(2+) stores inducing ER stress and activation of PERK-peIF2α-CHOP pathway. Quenching ROS generation by anti-oxidants prevented Ca(2+) release and subsequent induction of ER stress and UPR activation. UV-B irradiation induced PERK dependent G2/M phase cell cycle arrest in Hs68 and G1/S phase cell cycle arrest in HaCaT. Also our study reflects that UV-B exposure leads to loss of mitochondrial membrane potential, activation of apoptotic cascade as evident by AnnexinV/PI staining, decreased expression of Bcl-2 and increased cleavage of PARP-1 protein. CONCLUSION: UV-B induced Ca(2+) deficit within ER lumen was mediated by immediate ROS generation. Insufficient Ca(2+) concentration within ER lumen developed ER stress leading to UPR activation. These changes were reversed by use of anti-oxidants which quench ROS.


Subject(s)
Calcium Signaling/radiation effects , Endoplasmic Reticulum Stress/radiation effects , Endoplasmic Reticulum/radiation effects , Fibroblasts/radiation effects , Keratinocytes/radiation effects , Oxidative Stress/radiation effects , Ultraviolet Rays , Unfolded Protein Response/radiation effects , Antioxidants/pharmacology , Apoptosis/radiation effects , Calcium Signaling/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Line , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress/drug effects , Eukaryotic Initiation Factor-2/metabolism , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/pathology , Membrane Potential, Mitochondrial/radiation effects , Oxidative Stress/drug effects , Phosphorylation , Reactive Oxygen Species/metabolism , Transcription Factor CHOP/metabolism , Unfolded Protein Response/drug effects , eIF-2 Kinase/metabolism
16.
Int J Radiat Biol ; 90(4): 306-12, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24456547

ABSTRACT

PURPOSE: Perturbations in protein folding induce endoplasmic reticulum (ER) stress, which elicits coordinated response, namely the unfolded protein response (UPR), to cope with the accumulation of misfolded proteins in ER. In this study, we characterized mechanisms underlying ionizing radiation (IR)-induced UPR signaling pathways. MATERIALS AND METHODS: We analyzed alterations in UPR signaling pathways in human umbilical vein endothelial cells (HUVEC) and human coronary artery endothelial cells (HCAEC) irradiated with 15 Gy IR. RESULTS: IR selectively activated the eIF2α/ATF4 branch of the UPR signaling pathway, with no alterations in the IRE1 and ATF6 branches in HUVEC and HCAEC. Phosphorylation of PERK was enhanced in response to IR, and the IR-induced activation of the eIF2α/ATF4 signaling pathway was completely inhibited by PERK knockdown with siRNA. Surprisingly, chemical chaperones, which inhibit the formation of misfolded proteins and sequential protein aggregates to reduce ER stress, failed to prevent the IR-induced phosphorylation of PERK and the subsequent activation of the eIF2α/ATF4 signaling pathway. CONCLUSIONS: PERK mediates the IR-induced selective activation of the eIF2α/ATF4 signaling pathway, and the IR-induced activation of PERK/eIF2α/ATF4 signaling in human vascular endothelial cells is independent of alterations in protein-folding homeostasis in the ER.


Subject(s)
Activating Transcription Factor 4/physiology , Endoplasmic Reticulum Stress/physiology , Endothelial Cells/radiation effects , Eukaryotic Initiation Factor-2/physiology , Signal Transduction/radiation effects , eIF-2 Kinase/physiology , Aspartate-Ammonia Ligase/genetics , Cell Cycle Proteins/genetics , Cells, Cultured , Endothelial Cells/metabolism , Humans , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Repressor Proteins/genetics , Unfolded Protein Response/radiation effects
17.
J Biol Chem ; 285(43): 33165-33174, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20732869

ABSTRACT

In response to different environmental stresses, phosphorylation of eIF2 (eIF2∼P) represses global translation coincident with preferential translation of ATF4. ATF4 is a transcriptional activator of the integrated stress response, a program of gene expression involved in metabolism, nutrient uptake, anti-oxidation, and the activation of additional transcription factors, such as CHOP/GADD153, that can induce apoptosis. Although eIF2-P elicits translational control in response to many different stress arrangements, there are selected stresses, such as exposure to UV irradiation, that do not increase ATF4 expression despite robust eIF2∼P. In this study we addressed the underlying mechanism for variable expression of ATF4 in response to eIF2∼P during different stress conditions and the biological significance of omission of enhanced ATF4 function. We show that in addition to translational control, ATF4 expression is subject to transcriptional regulation. Stress conditions such as endoplasmic reticulum stress induce both transcription and translation of ATF4, which together enhance expression of ATF4 and its target genes in response to eIF2∼P. By contrast, UV irradiation represses ATF4 transcription, which diminishes ATF4 mRNA available for translation during eIF2∼P. eIF2∼P enhances cell survival in response to UV irradiation. However, forced expression of ATF4 and its target gene CHOP leads to increased sensitivity to UV irradiation. This combination of transcriptional regulation and translational control allows the eIF2 kinase pathway to selectively repress or activate key regulatory genes subject to preferential translation, providing the integrated stress response versatility to direct the transcriptome that is essential for maintaining the balance between stress remediation and apoptosis.


Subject(s)
Activating Transcription Factor 4/biosynthesis , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation/physiology , Protein Biosynthesis/physiology , Stress, Physiological/physiology , Transcription, Genetic/physiology , Activating Transcription Factor 4/genetics , Animals , Apoptosis/physiology , Apoptosis/radiation effects , Cell Line, Tumor , Cell Survival , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Eukaryotic Initiation Factor-2/genetics , Gene Expression Regulation/radiation effects , Humans , Mice , Mice, Knockout , Protein Biosynthesis/radiation effects , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Stress, Physiological/radiation effects , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism , Transcription, Genetic/radiation effects , Ultraviolet Rays , Unfolded Protein Response/physiology , Unfolded Protein Response/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...