1
|
Islam S, Mukherjee C. Hypoxia inducible factor HIF1α elevates expression of mRNA capping enzyme during cobalt chloride-induced hypoxia. BIOCHIMICA ET BIOPHYSICA ACTA. GENE REGULATORY MECHANISMS 2025; 1868:195087. [PMID: 40189045 DOI: 10.1016/j.bbagrm.2025.195087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 02/19/2025] [Accepted: 04/01/2025] [Indexed: 04/29/2025]
Abstract
In response to hypoxia, hypoxia-inducible factors (HIFs) control the transcriptomic output to mitigate the hypoxic stress. Long noncoding RNAs (lncRNA) are found to be very crucial in regulating hypoxia. Like mRNAs, lncRNAs are protected by 5' caps that are added by mRNA capping enzyme (CE) in the nucleus. The previous concept that capping takes place in the nucleus was changed by the recognition of a cytoplasmic pool of capping enzyme (cCE). cCE has been shown to recap its substrate uncapped mRNAs or long noncoding RNAs (lncRNAs) present in the cytoplasm, preventing their degradation, even during arsenite-induced oxidative stress. In this study, we examined the effect of CoCl2 induced hypoxia on cCE and its function in regulating the substrate lncRNAs. Here, we show that CoCl2 induced hypoxia elevates the expressions of nuclear and cytoplasmic CE in HIF1α dependent manner as evidenced by Chromatin immunoprecipitation and HIF1α inhibitor experiments. Furthermore, we found cCE post-transcriptionally controls the stability of its target lncRNAs amidst CoCl2 induced hypoxia. These results suggest that cCE, upregulated by HIF1α, may act as a posttranscriptional modulator for a few cCE-targeted lncRNAs.
Collapse
Affiliation(s)
- Safirul Islam
- Institute of Health Sciences, Presidency University, Plot No. DG/02/02, Premises No. 14-0358, Action Area 1D, Kolkata 700156, India.
| | - Chandrama Mukherjee
- Institute of Health Sciences, Presidency University, Plot No. DG/02/02, Premises No. 14-0358, Action Area 1D, Kolkata 700156, India.
| |
Collapse
|
2
|
Khakshour E, Bahreyni-Toossi MT, Anvari K, Shahram MA, Vaziri-Nezamdoust F, Azimian H. Evaluation of the effects of simulated hypoxia by CoCl 2 on radioresistance and change of hypoxia-inducible factors in human glioblastoma U87 tumor cell line. Mutat Res 2024; 828:111848. [PMID: 38154290 DOI: 10.1016/j.mrfmmm.2023.111848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 11/24/2023] [Accepted: 11/29/2023] [Indexed: 12/30/2023]
Abstract
PURPOSE Glioblastoma (GBM) is considered the most common and lethal type of brain tumor with a poor prognosis. GBM treatment has challenges due to its aggressive nature, which often causes treatment failure and recurrence. Hypoxia is one of the characteristics of glioblastoma tumors that contribute to radioresistance and malignant phenotypes of GBM. In this study, we aimed to determine the effects of hypoxia on the radiosensitivity of U87 GBM cells by the hypoxia-mimicking model. METHODS Following the treatment of cells with different concentrations of CoCl2, an MTT assay was used to evaluate the cytotoxicity of CoCl2. To understand the effects of Ionizing radiation on CoCl2-treated groups, cells were exposed to irradiation after pretreating with 100 μM CoCl2, and a clonogenic survival assay was performed to determine the radiosensitivity of U87 cells. Also, the intracellular Reactive oxygen level was measured by 2',7'-dichlorofluorescein diacetate (DCFDA) probe staining. Additionally, the expression of hypoxia-associated genes, including HIF-1α, HIF-2α, and their target genes (GLUT-1), was monitored by reverse transcription polymerase chain reaction (RT-PCR). RESULTS Our study revealed that the cell viability of CoCl2-treated cells was decreased in a concentration-dependent manner. Also, CoCl2 did not cause any cytotoxicity on U87 cells at a concentration of 100 μM after treatment for 24 h. Colony formation assay showed that CoCl2 pretreatment induced radioresistance of tumor cells compared to non-treated cells. Also, CoCl2 can protect cells against irradiation by the clearance of ROS. Moreover, Real-time results showed that the mRNA expression of HIF-1α and GLUT-1 were significantly upregulated following hypoxia induction and/or irradiation condition. However, the level of HIF-2α mRNA did not change significantly in hypoxia or irradiation alone conditions, but it increased significantly only in hypoxia + irradiation conditions. CONCLUSION Taken together, our results indicated that simulating hypoxia by CoCl2 can effectively increase hypoxia-associated genes, specially HIF-1α and GLUT-1, but did not affect HIF-2α gene expression. Also, it can increase the clearance of ROS, respectively, and it leads to inducing radioresistance of U87 cells.
Collapse
Affiliation(s)
- Elham Khakshour
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Taghi Bahreyni-Toossi
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Kazem Anvari
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammad Amin Shahram
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Hosein Azimian
- Department of Medical Physics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Physics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Correia AS, Marques L, Cardoso A, Vale N. Exploring the Role of Drug Repurposing in Bridging the Hypoxia-Depression Connection. MEMBRANES 2023; 13:800. [PMID: 37755222 PMCID: PMC10537732 DOI: 10.3390/membranes13090800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 09/05/2023] [Accepted: 09/15/2023] [Indexed: 09/28/2023]
Abstract
High levels of oxidative stress are implicated in hypoxia, a physiological response to low levels of oxygen. Evidence supports a connection between this response and depression. Previous studies indicate that tryptophan hydroxylase can be negatively affected in hypoxia, impairing serotonin synthesis and downstream pathways. Some studies also hypothesize that increasing hypoxia-inducible factor-1 (HIF-1) levels may be a new therapeutic modality for depression. Hence, this study delved into the influence of hypoxia on the cellular response to drugs designed to act in depression. By the induction of hypoxia in SH-SY5Y cells through a hypoxia incubator chamber or Cobalt Chloride treatment, the effect of Mirtazapine, an antidepressant, and other drugs that interact with serotonin receptors (TCB-2, Dextromethorphan, Ketamine, Quetiapine, Scopolamine, Celecoxib, and Lamotrigine) on SH-SY5Y cellular viability and morphology was explored. The selection of drugs was initially conducted by literature search, focusing on compounds with established potential for employment in depression therapy. Subsequently, we employed in silico approaches to forecast their ability to traverse the blood-brain barrier (BBB). This step was particularly pertinent as we aimed to assess their viability for inducing potential antidepressant effects. The effect of these drugs in hypoxia under the inhibition of HIF-1 by Echinomycin was also tested. Our results revealed that all the potential repurposed drugs promoted cell viability, especially when hypoxia was chemically induced. When combined with Echinomycin, all drugs decreased cellular viability, possibly by the inability to interact with HIF-1.
Collapse
Affiliation(s)
- Ana Salomé Correia
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Institute of Biomedical Sciences Abel Salazar (ICBAS), University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
| | - Lara Marques
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| | - Armando Cardoso
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- NeuroGen Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
| | - Nuno Vale
- OncoPharma Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, 4200-450 Porto, Portugal
| |
Collapse
|
4
|
Toragall V, Muzaffar JC, Baskaran V. Lutein loaded double-layered polymer nanocarrier modulate H 2O 2 and CoCl 2 induced oxidative and hypoxia damage and angiogenic markers in ARPE-19 cells. Int J Biol Macromol 2023; 240:124378. [PMID: 37030468 DOI: 10.1016/j.ijbiomac.2023.124378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 04/03/2023] [Accepted: 04/04/2023] [Indexed: 04/10/2023]
Abstract
Lutein plays a crucial role in the protection of retina by diminishing oxidative stress in diabetic retinopathy (DR). However, its poor aqueous solubility, chemical instability and low bioavailability edge its application. Also, beneficial effects of lutein supplementation and lower lutein levels in the serum and retina of DR patients created an interest in nanopreparation. Hence, lutein-loaded chitosan‑sodium alginate nanocarrier comprising oleic acid core (LNCs) was developed and examined its protective effect on hyperglycemia-mediated changes in oxidative stress and angiogenesis in ARPE-19 cells. Results showed that the LNCs have smaller size and a smooth spherical morphology and did not affect the ARPE-19 cell viability (up to 20 μM) and showed higher cellular uptake in both normal and H2O2-induced stress conditions. LNCs pre-treatment suppressed the H2O2-induced oxidative stress and CoCl2-induced hypoxia-mediated elevation of intracellular reactive oxygen species, protein carbonyl and malondialdehyde levels by restoring antioxidant enzymes in ARPE-19 cells. Further, LNCs protected H2O2-mediated down-regulation of Nrf2 and its downstream antioxidant enzymes. LNCs also restored the H2O2-altered angiogenic (Vascular endothelial growth factor (VEGF), X-box binding protein 1 (XBP-1) and Hypoxia-inducible factor 1-alpha (HIF-1α)), endoplasmic reticulum stress (activating transcription factor-4 (ATF4)) and tight junction (Zona occludens 1 (ZO-1)) markers. To conclude, we could successfully develop biodegradable LNCs to improve the cellular uptake of lutein to treat DR by curtailing oxidative stress in retina.
Collapse
Affiliation(s)
- Veeresh Toragall
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - J C Muzaffar
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India
| | - Vallikanan Baskaran
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore 570020, Karnataka, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
5
|
Zhao SC, Qiao FX, Sun MX, Liu YC, Wang HL, Xu ZR, Liu Y. Cobalt chloride exposure disturbs spindle assembly and decreases mouse oocyte development potential. Toxicology 2023; 486:153450. [PMID: 36739938 DOI: 10.1016/j.tox.2023.153450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Cobalt is a kind of heavy metal which is widely used in petrochemical and biomedical industries. Animal studies have reported that cobalt would exert systemic toxicity, but its effects on the ovarian function in mammals, especially for oocyte quality remains unknown. In the present study, we report that cobalt chloride treatment affects ovary coefficient and follicular growth. Oocytes in cobalt chloride exposed mice exhibited a decreased development potential, with the evidence of decreased occurrence rate of germ vesicle breakdown and polar body extrusion. Besides, cobalt chloride disorganized meiotic spindle formation and movement, mechanically associated with affecting TACC3 and Ac-a-tubulin levels, and disturbing actin reorganization. In addition, cobalt chloride exposure result in mitochondrial cristae structures disappear, cluster distribution and potential depolarization. Altogether, these findings suggest that cobalt chloride impairs the ovarian follicle growth and affects oocyte development by disrupted spindle assembly and mitochondrial function.
Collapse
Affiliation(s)
- Si-Cheng Zhao
- Medical College, Guangxi University, Nanning, Guangxi 530004, China; Key Laboratory of Biomedical Information Engineering of the Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, Shaanxi 710049, China
| | - Feng-Xin Qiao
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Ming-Xin Sun
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Yue-Cen Liu
- Medical College, Guangxi University, Nanning, Guangxi 530004, China
| | - Hai-Long Wang
- Department of Basic Medicine, School of Medicine, Xiamen University, Xiamen, Fujian 361102, China
| | - Zhi-Ran Xu
- Translational Medicine Research Center, Ruikang Hospital Affiliated to Guangxi University of Chinese Medicine, Nanning, Guangxi 530011, China.
| | - Yu Liu
- Medical College, Guangxi University, Nanning, Guangxi 530004, China.
| |
Collapse
|
6
|
Chen P, Wang T, Chen Q. Ginkgo biloba Golden Leaf Extract (GGLE) Inhibits Melanoma Cell Invasion and Angiogenesis Through Inhibition of Angiogenin. Integr Cancer Ther 2023; 22:15347354221134513. [PMID: 36859800 PMCID: PMC9983097 DOI: 10.1177/15347354221134513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2023] Open
Abstract
The popular dietary supplements of Ginkgo biloba (Ginkgo) products have been reported to have anti-cancer activities in multiple cellular and animal studies, with the benefits yet to be proven with clinical trials. The mechanisms of action are not clear, forming a barrier to investigation in Gingko-specific benefits to cancer patients, especially when combined with other therapies. Here we reported on the discovery of a novel mechanism by which a Ginkgo golden leaf extract (GGLE) inhibited melanoma cell invasion and angiogenesis. GGLE did not inhibit melanoma cells via direct cytotoxicity. Instead, GGLE significantly inhibited total RNase activities in melanoma cells under both normoxia and hypoxia conditions. The RNase angiogenin was induced twofolds by hypoxia, and the induction was significantly suppressed by GGLE treatment in a dose dependent manner. As a result of angiogenin inhibition, GGLE inhibited melanoma cell migration and invasion in a dose dependent manner. Conditioned media from melanoma cell culture sufficiently induced in vitro angiogenesis in human endothelial cells, whereas the conditioned media of GGLE-treated melanoma cells significantly inhibited this angiogenetic activity. This was accompanied with markedly reduced angiogenin concentrations in the GGLE-treated melanoma cell conditioned media. We concluded that, instead of direct cytotoxicity, GGLE inhibited angiogenin synthesis and secretion by melanoma cells, resulting in inhibition of tumor cell invasion and tumor-induced angiogenesis. This new mechanism opens the door for investigation in GGLE influencing tumor microenvironment, and warrants further investigation and validation in vivo.
Collapse
Affiliation(s)
- Ping Chen
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Tao Wang
- University of Kansas Medical Center, Kansas City, KS, USA
| | - Qi Chen
- University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
7
|
Vazifehmand R, Ali DS, Othman Z, Chau DM, Stanslas J, Shafa M, Sekawi Z. The evaluation expression of non-coding RNAs in response to HSV-G47∆ oncolytic virus infection in glioblastoma multiforme cancer stem cells. J Neurovirol 2022; 28:566-582. [PMID: 35951174 DOI: 10.1007/s13365-022-01089-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 07/11/2022] [Accepted: 07/25/2022] [Indexed: 01/13/2023]
Abstract
Glioblastoma multiforme is the most aggressive astrocytes brain tumor. Glioblastoma cancer stem cells and hypoxia conditions are well-known major obstacles in treatment. Studies have revealed that non-coding RNAs serve a critical role in glioblastoma progression, invasion, and resistance to chemo-radiotherapy. The present study examined the expression levels of microRNAs (in normoxic condition) and long non-coding RNAs (in normoxic and hypoxic conditions) in glioblastoma stem cells treated with the HSV-G47∆. The expression levels of 43 miRNAs and 8 lncRNAs isolated from U251-GBM-CSCs were analyzed using a miRCURY LNA custom PCR array and a quantitative PCR assay, respectively. The data revealed that out of 43 miRNAs that only were checked in normoxic condition, the only 8 miRNAs, including miR-7-1, miR-let-7b, miR-130a, miR-137, miR-200b, miR-221, miR-222, and miR-874, were markedly upregulated. The expression levels of lncRNAs, including LEF1 antisense RNA 1 (LEF1-AS1), metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), long intergenic non-protein coding RNA 470 (LINC00470), tumor suppressor candidate 7 (TUSC7), HOX transcript antisense RNA (HOTAIR), nuclear paraspeckle assembly transcript 1 (NEAT1), and X inactive specific transcript (XIST), were markedly downregulated in the hypoxic microenvironment, and H19-imprinted maternally expressed transcript (H19) was not observed to be dysregulated in this environment. Under normoxic conditions, LEF1-AS1, MALAT1, LINC00470, H19, HOTAIR, NEAT1, and XIST were downregulated and TUSC7 was not targeted by HSV-G47∆. Overall, the present data shows HSVG47Δ treatment deregulates non-coding RNA expression in GBM-CSC tumor microenvironments.
Collapse
Affiliation(s)
- Reza Vazifehmand
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang, Selangor Darul Ehsan, 43400, Malaysia
| | - Dhuha Saeed Ali
- Halal Products Research Institute, Universiti Putra Malaysia UPM, Serdang, Selangor, 43400, Malaysia
| | - Zulkefley Othman
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang, Selangor Darul Ehsan, 43400, Malaysia
| | - De-Ming Chau
- Department of Biomedical Science, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang, Selangor Darul Ehsan, 43400, Malaysia
| | - Johnson Stanslas
- Pharmacotherapeutics Unit, Department of Medicine, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia UPM, Serdang, Selangor, 43400, Malaysia
| | - Mehdi Shafa
- Cell Therapy process development, Lonza Houston Inc, Houston, TX, USA
| | - Zamberi Sekawi
- Department of Medical Microbiology & Parasitology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia (UPM), Serdang, Selangor Darul Ehsan, 43400, Malaysia.
| |
Collapse
|
8
|
Liyanage DS, Omeka WKM, Sandamalika WMG, Udayantha HMV, Jeong T, Lee S, Lee J. PDI family thioredoxin from disk abalone (Haliotis discus discus): Responses to stimulants (PAMPs, bacteria, and viral) and functional characterization. FISH & SHELLFISH IMMUNOLOGY 2022; 120:261-270. [PMID: 34848304 DOI: 10.1016/j.fsi.2021.11.037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 11/23/2021] [Accepted: 11/25/2021] [Indexed: 06/13/2023]
Abstract
Thioredoxin, a highly conserved class of proteins involved in redox signaling, is found in a range of organisms from bacteria to higher-level eukaryotes. Thioredoxin acts as an active regulatory enzyme to eliminate excessive reactive oxygen species, thereby preventing cellular damage. In this study, the cDNA sequence of thioredoxin domain-containing 5 (AbTXNDC5) from the disk abalone transcriptomic database was characterized. An in silico analysis of AbTXNDC5 was performed, and its spatial and temporal expression patterns in hemocytes and gills in response to bacteria (Vibrio parahaemolyticus, Listeria monocytogenes), viral hemorrhagic septicemia virus, and pathogen-associated molecular pattern molecules were observed. Furthermore, AbTXNDC5 expression was examined in different developmental stages. Functional assays to explore insulin disulfide reduction, anti-apoptotic activity, and protection against hypoxic cell death of AbTXNDC5 were conducted through recombinant proteins or overexpression in cells. AbTXNDC5 contains a 1179-bp open reading frame coding for 392 amino acids. Conserved thiol-disulfide cysteine residues within two Cys-X-X-Cys motifs were found in AbTXNDC5. Quantitative real-time polymerase chain reaction indicated that healthy digestive tract and hemocyte tissues expressed high levels of AbTXNDC5 mRNA, which may protect the host from invading pathogens. Immune-challenged abalone hemocytes and gills exhibited upregulated expression of AbTXNDC5 at different time points. rAbTXNDC5 also exhibited a functional insulin disulfide reductase activity. AbTXNDC5 conferred protection to cultured cells from apoptosis and hypoxia-induced stress, compared to the pcDNA3.1(+) transfected control cells. Therefore, AbTXNDC5 can be considered an important gene in abalones in relation to the primary immune system and regulation of redox homeostasis and confers protection from stress.
Collapse
Affiliation(s)
- D S Liyanage
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - W K M Omeka
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - W M Gayashani Sandamalika
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - H M V Udayantha
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea
| | - Taehyug Jeong
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea
| | - Sukkyoung Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| | - Jehee Lee
- Department of Marine Life Sciences & Fish Vaccine Research Center, Jeju National University, Jeju, 63243, Republic of Korea; Marine Science Institute, Jeju National University, Jeju, 63333, Republic of Korea.
| |
Collapse
|
9
|
Choi JY, Seok HJ, Kim RK, Choi MY, Lee SJ, Bae IH. miR-519d-3p suppresses tumorigenicity and metastasis by inhibiting Bcl-w and HIF-1α in NSCLC. Mol Ther Oncolytics 2021; 22:368-379. [PMID: 34553025 PMCID: PMC8430049 DOI: 10.1016/j.omto.2021.06.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Bcl-w, a member of the Bcl-2 family, is highly expressed in various solid tumor, including lung cancer, suggesting that it is involved in cancer cell survival and carcinogenesis. Solid cancer-induced hypoxia has been reported to increase angiogenesis, growth factor, gene instability, invasion, and metastasis. Despite many studies on the treatment of non-small cell lung cancer (NSCLC) with a high incidence rate, the survival rate of patients has not improved because the cancer cells acquired resistance to treatment. This study investigated the correlation between Bcl-w expression and hypoxia in tumor malignancy of NSCLC. Meanwhile, microRNAs (miRNAs) are involved in a variety of key signaling mechanisms associated with hypoxia. Therefore, we discovered miR-519d-3p, which inhibits the expression of Bcl-w and hypoxia-inducing factor (HIF)-1α, and found that it reduces hypoxia-induced tumorigenesis. Spearman's correlation analysis showed that the expression levels of miR-519d-3p and Bcl-w/HIF-1α were negatively correlated, respectively. This showed that miR-519d-3p can be used as a diagnostic biomarker and target therapy for NSCLC.
Collapse
Affiliation(s)
- Jae Yeon Choi
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Hyun Jeong Seok
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| | - Rae-Kwon Kim
- Department of Life Science, Hanyang University, Seoul, Republic of Korea
| | - Mi Young Choi
- Department of Life Science, Hanyang University, Seoul, Republic of Korea
| | - Su-Jae Lee
- Department of Life Science, Hanyang University, Seoul, Republic of Korea
| | - In Hwa Bae
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul 01812, Republic of Korea
| |
Collapse
|
10
|
Kim A, Jung HG, Kim SC, Choi M, Park JY, Lee SG, Hwang EM. Astrocytic AEG-1 regulates expression of TREK-1 under acute hypoxia. Cell Biochem Funct 2019; 38:167-175. [PMID: 31782179 DOI: 10.1002/cbf.3469] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/29/2019] [Accepted: 10/13/2019] [Indexed: 12/16/2022]
Abstract
TREK-1 (TWIK-related K+ channel), a member of the two-pore domain K+ (K2P) channel family, is highly expressed in astrocytes, where it plays a key role in glutamate release and passive conductance. In addition, TREK-1 is induced to protect neurons under pathological conditions such as hypoxia. However, the upstream regulation of TREK-1 remains poorly understood. In this study, we found that AEG-1 (astrocyte elevated gene-1) regulates the expression of astrocytic TREK-1 under hypoxic conditions. Upregulation of AEG-1 increased expression of TREK-1 in astrocytes, and knockdown of AEG-1 dramatically decreased the mRNA and protein levels of TREK-1, which were restored by expression of shRNA-insensitive AEG-1. In addition, expression of TREK-1 was not regulated in the absence of AEG-1, even when HIF1α was present. Together, these results suggest that AEG-1 acts as a major upstream regulator of TREK-1 channels in astrocytes under hypoxia. SIGNIFICANCE OF THE STUDY: Previous studies have reported that hypoxia increases the expression of astrocytic TREK-1 and that increased TREK-1 expression protects neuronal cells from apoptosis. However, its cellular mechanism is not clear. In this study we first showed that AEG-1 is a major mediator of hypoxic-regulated TREK-1 expression in normal astrocytes independently of HIF-1α.
Collapse
Affiliation(s)
- Ajung Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hyun-Gug Jung
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Seung-Chan Kim
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Minji Choi
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, Republic of Korea
| | - Seok-Geun Lee
- KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea.,Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Eun Mi Hwang
- Center for Functional Connectomics, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,KHU-KIST Department of Converging Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
11
|
The Attitudes Towards the Use of Restraint and Restrictive Intervention Amongst Healthcare Staff on Acute Medical and Frailty Wards-A Brief Literature Review. Geriatrics (Basel) 2019; 4:geriatrics4030050. [PMID: 31487923 PMCID: PMC6787583 DOI: 10.3390/geriatrics4030050] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Revised: 08/28/2019] [Accepted: 08/30/2019] [Indexed: 01/12/2023] Open
Abstract
Restraint in modern non-psychiatric-based healthcare is often regarded as a rare occurrence. It is deemed to be used as a last resort to prevent patients from directly harming themselves. However, techniques are used in modern day practice which are considered direct and indirect restraints with the justification of maintaining patient safety, but they are often not classified as “restraints”. Examples of these include the use of bed rails or tables to prevent patients from “wandering” and to reduce the risk of falls and injuries. More indirect techniques would involve passive interactions with patients or leaving mobility aids out of reach. Staff subconsciously restrain patients and reduce their liberties despite agreeing that patient autonomy should be upheld—a necessary evil to maintain a duty of care. Whilst the use of restraints is often justified to ensure patient care and prevent injury, it is not without consequence. There are physical and psychological health risks such as pressure sores from the inability to mobilise, or the brewing of anger and frustration when denied access to everyday actions. The reasons why restraints are used, whilst stemming from maintaining patient safety, are often due to low staffing levels and the inability to constantly watch at-risk patients due to a large workload. Inadequate training is another factor; by improving education in direct and indirect restraint and providing alternative methods, more ethical decisions and positive outcomes can be implemented. Healthcare professionals are reluctant to use restraint but often conduct it without realising it; assessing their understanding of restraint and providing education to raise awareness of the consequences of direct and indirect methods would result in positive steps toward reducing their use at the same time as looking to provide alternatives to uphold patient care whilst maintaining their dignity and liberty.
Collapse
|
12
|
Makgoo L, Laka K, Mbita Z. Downregulation of RBBP6 variant 1 during arsenic trioxide-mediated cell cycle arrest and curcumin-induced apoptosis in MCF-7 breast cancer cells. Future Sci OA 2019; 5:FSO409. [PMID: 31534777 PMCID: PMC6745614 DOI: 10.2144/fsoa-2019-0047] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Aim To determine the expression patterns of the RBBP6 spliced variants during arsenic trioxide-mediated cell cycle arrest and curcumin-induced apoptosis in MCF-7 cells. Materials & methods As2O3 and curcumin were used to study cytotoxicity, cell cycle arrest, apoptosis and the expression of RBBP6 variants. The MUSE Cell Analyser was used to analyze cell cycle arrest, apoptosis and multicaspase activity while apoptosis was further confirmed using microscopy. Semi-quantitative RT-PCR was employed to quantitate the expression of the RBBP6 variants. Results This study showed that the MCF-7 cells expressed RBBP6 variant 1 but lacked both variant 2 and variant 3. Both As2O3 and curcumin significantly downregulated RBBP6 variant 1 (p < 0.001). Conclusion RBBP6 variants are promising therapeutic targets.
Collapse
Affiliation(s)
- Lilian Makgoo
- Department of Biochemistry, Microbiology, & Biotechnology, University of Limpopo, Private Bag x1106, Sovenga, 0727, Polokwane, South Africa
| | - Kagiso Laka
- Department of Biochemistry, Microbiology, & Biotechnology, University of Limpopo, Private Bag x1106, Sovenga, 0727, Polokwane, South Africa
| | - Zukile Mbita
- Department of Biochemistry, Microbiology, & Biotechnology, University of Limpopo, Private Bag x1106, Sovenga, 0727, Polokwane, South Africa
| |
Collapse
|
13
|
CoCl 2 simulated hypoxia induce cell proliferation and alter the expression pattern of hypoxia associated genes involved in angiogenesis and apoptosis. Biol Res 2019; 52:12. [PMID: 30876462 PMCID: PMC6419504 DOI: 10.1186/s40659-019-0221-z] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Accepted: 03/04/2019] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND/AIMS Hypoxia microenvironment plays a crucial role during tumor progression and it tends to exhibit poor prognosis and make resistant to various conventional therapies. HIF-1α acts as an important transcriptional regulator directly or indirectly associated with genes involved in cell proliferation, angiogenesis, apoptosis and energy metabolism during tumor progression in hypoxic microenvironment. This study was aimed to investigate the expression pattern of the hypoxia associated genes and their association during breast cancer progression under hypoxic microenvironment in breast cancer cells. METHODS Cell proliferation in MCF-7 and MDA-MB-231 cell lines treated with different concentration of CoCl2 was analyzed by MTT assay. Flow cytometry was performed to check cell cycle distribution, whereas cell morphology was examined by phase contrast microscopy in both the cells during hypoxia induction. Expression of hypoxia associated genes HIF-1α, VEGF, p53 and BAX were determined by semiquantitative RT-PCR and real-time PCR. Western blotting was performed to detect the expression at protein level. RESULTS Our study revealed that cell proliferation in CoCl2 treated breast cancer cells were concentration dependent and varies with different cell types, further increase in CoCl2 concentration leads to apoptotic cell death. Further, accumulation of p53 protein in response to hypoxia as compare to normoxia showed that induction of p53 in breast cancer cells is HIF-1α dependent. HIF-1α dependent BAX expression during hypoxia revealed that after certain extent of hypoxia induction, over expression of BAX conquers the effect of anti-apoptotic proteins and ultimately leads to apoptosis in breast cancer cells. CONCLUSION In conclusion our results clearly indicate that CoCl2 simulated hypoxia induce the accumulation of HIF-1α protein and alter the expression of hypoxia associated genes involved in angiogenesis and apoptosis.
Collapse
|
14
|
Niu N, Li Z, Zhu M, Sun H, Yang J, Xu S, Zhao W, Song R. Effects of nuclear respiratory factor‑1 on apoptosis and mitochondrial dysfunction induced by cobalt chloride in H9C2 cells. Mol Med Rep 2019; 19:2153-2163. [PMID: 30628711 PMCID: PMC6390059 DOI: 10.3892/mmr.2019.9839] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2017] [Accepted: 09/28/2018] [Indexed: 01/31/2023] Open
Abstract
Hypoxia-induced apoptosis occurs in various diseases. Cobalt chloride (CoCl2) is a hypoxia mimic agent that is frequently used in studies investigating the mechanisms of hypoxia. Nuclear respiratory factor-1 (NRF-1) is a transcription factor with an important role in the expression of mitochondrial respiratory and mitochondria-associated genes. However, few studies have evaluated the effects of NRF-1 on apoptosis, particularly with regard to damage caused by CoCl2. In the present study, the role of NRF-1 in mediating CoCl2-induced apoptosis was investigated using cell viability analysis, flow cytometry, fluorescence imaging, western blotting analysis, energy metabolism analysis and reverse transcription-quantitative polymerase chain reaction. The present results revealed that the apoptosis caused by CoCl2 could be alleviated by NRF-1. Furthermore, overexpression of NRF-1 increased the expression of B-cell lymphoma-2, hypoxia inducible factor-1α and NRF-2. Also, cell damage induced by CoCl2 may be associated with depolarization of mitochondrial membrane potential, and NRF-1 suppressed this effect. Notably, the oxygen consumption rate (OCR) was reduced in CoCl2-treated cells, whereas overexpression of NRF-1 enhanced the OCR, suggesting that NRF-1 had protective effects. In summary, the present study demonstrated that NRF-1 protected against CoCl2-induced apoptosis, potentially by strengthening mitochondrial function to resist CoCl2-induced damage to H9C2 cells. The results of the present study provide a possible way for the investigation of myocardial diseases.
Collapse
Affiliation(s)
- Nan Niu
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Zihua Li
- School of Pharmacy, Tsinghua University, Beijing 100084, P.R. China
| | - Mingxing Zhu
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Hongli Sun
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Jihui Yang
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Shimei Xu
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Wei Zhao
- College of Basic Medicine, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| | - Rong Song
- Department of Critical Care Medicine, The Fifth Hospital of the Chinese People's Liberation Army, Yinchuan, Ningxia Hui Autonomous Region 750001, P.R. China
| |
Collapse
|
15
|
Hsu HL, Liao PL, Cheng YW, Huang SH, Wu CH, Li CH, Kang JJ. Chloramphenicol Induces Autophagy and Inhibits the Hypoxia Inducible Factor-1 Alpha Pathway in Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2019; 20:ijms20010157. [PMID: 30609861 PMCID: PMC6337541 DOI: 10.3390/ijms20010157] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 12/16/2018] [Accepted: 12/22/2018] [Indexed: 12/31/2022] Open
Abstract
Chloramphenicol is an inexpensive and excellent bactericidal antibiotic. It is used to combat anaerobic infections in the Third World countries, whereas its systemic application has been abandoned in developed countries. However, in recent years, clinicians have reintroduced chloramphenicol in clinical practice. In this study, chloramphenicol was found to repress the oxygen-labile transcription factor, hypoxia inducible factor-1 alpha (HIF-1α), in hypoxic A549 and H1299 cells. Furthermore, it suppressed the mRNA levels of vascular endothelial growth factor (VEGF) and glucose transporter 1, eventually decreasing VEGF release. Chloramphenicol initiated the autophagy pathway in treated cells, as observed by the increase in formation of Atg12-Atg5 conjugates, and in beclin-1 and LC3-II levels. The chloramphenicol-mediated HIF-1α degradation was completely reverted by autophagic flux blockage. In HIF-1α-overexpressing cells, the formation of HIF-1α/SENP-1 (Sentrin/SUMO-specific protease 1) protein complex seemed to facilitate the escape of HIF-1α from degradation. Chloramphenicol inhibited HIF-1α/SENP-1 protein interaction, thereby destabilizing HIF-1α protein. The enhancement in HIF-1α degradation due to chloramphenicol was evident during the incubation of the antibiotic before hypoxia and after HIF-1α accumulation. Since HIF-1α plays multiple roles in infections, inflammation, and cancer cell stemness, our findings suggest a potential clinical value of chloramphenicol in the treatment of these conditions.
Collapse
Affiliation(s)
- Han-Lin Hsu
- Division of Pulmonary Medicine, Department of Internal Medicine, Taipei Medical University-Wan Fang Hospital, Taipei 116, Taiwan.
| | - Po-Lin Liao
- Institute of Food Safety and Health Risk Assessment, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 110, Taiwan.
| | - Yu-Wen Cheng
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan.
| | - Shih-Hsuan Huang
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110, Taiwan.
| | - Chien-Hua Wu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan.
| | - Ching-Hao Li
- Department of Physiology, School of Medicine; Graduate Institute of Medical Sciences, College of Medicine; Taipei Medical University, Taipei 110, Taiwan.
| | - Jaw-Jou Kang
- Institute of Food Safety and Health Risk Assessment, School of Pharmaceutical Sciences, National Yang-Ming University, Taipei 112, Taiwan.
| |
Collapse
|
16
|
Toro-Urrego N, Vesga-Jiménez DJ, Herrera MI, Luaces JP, Capani F. Neuroprotective Role of Hypothermia in Hypoxic-ischemic Brain Injury: Combined Therapies using Estrogen. Curr Neuropharmacol 2019; 17:874-890. [PMID: 30520375 PMCID: PMC7052835 DOI: 10.2174/1570159x17666181206101314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 10/26/2018] [Accepted: 11/28/2018] [Indexed: 12/15/2022] Open
Abstract
Hypoxic-ischemic brain injury is a complex network of factors, which is mainly characterized by a decrease in levels of oxygen concentration and blood flow, which lead to an inefficient supply of nutrients to the brain. Hypoxic-ischemic brain injury can be found in perinatal asphyxia and ischemic-stroke, which represent one of the main causes of mortality and morbidity in children and adults worldwide. Therefore, knowledge of underlying mechanisms triggering these insults may help establish neuroprotective treatments. Selective Estrogen Receptor Modulators and Selective Tissue Estrogenic Activity Regulators exert several neuroprotective effects, including a decrease of reactive oxygen species, maintenance of cell viability, mitochondrial survival, among others. However, these strategies represent a traditional approach of targeting a single factor of pathology without satisfactory results. Hence, combined therapies, such as the administration of therapeutic hypothermia with a complementary neuroprotective agent, constitute a promising alternative. In this sense, the present review summarizes the underlying mechanisms of hypoxic-ischemic brain injury and compiles several neuroprotective strategies, including Selective Estrogen Receptor Modulators and Selective Tissue Estrogenic Activity Regulators, which represent putative agents for combined therapies with therapeutic hypothermia.
Collapse
Affiliation(s)
- Nicolás Toro-Urrego
- Address correspondence to this author at the Laboratorio de Citoarquitectura y Plasticidad Neuronal, Instituto de Investigaciones Cardiológicas, Universidad de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas, Buenos Aires, Argentina; E-mail:
| | | | | | | | | |
Collapse
|
17
|
Heirani-Tabasi A, Naderi-Meshkin H, Matin MM, Mirahmadi M, Shahriyari M, Ahmadiankia N, Sanjar Moussavi N, Bidkhori HR, Raeesolmohaddeseen M, Bahrami AR. Augmented migration of mesenchymal stem cells correlates with the subsidiary CXCR4 variant. Cell Adh Migr 2018; 12:118-126. [PMID: 29466916 DOI: 10.1080/19336918.2016.1243643] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Use of mesenchymal stem cells (MSCs) has been introduced as a promising tool, for structural and functional recovery of damaged tissues/organs. Studies have indicated that interactions between chemokine receptors and their ligands have a critical role in homing of MSCs to the site of injury. Although CXCR4 variants have been characterized, the exact role of each transcript in homing has remained unclear. In this study, cells were pretreated with various hypoxia-mimicking compounds (valproic acid, cobalt-chloride, and deferoxamine mesylate). Results indicated that both variants of CXCR4 were overexpressed after 24 hours of treatments and their expression could cooperatively induce and promote the cell migration. Moreover, deferoxamine mesylate was more effective in overexpression of variant A (lo), which resulted in higher level of CXCR4 protein and the highest rate of migration of the cells. In conclusion, our findings may have important potential implications in clinical applications, reinforcing the concept that manipulating the expression of specific CXCR4 variants may increase migration of MSCs.
Collapse
Affiliation(s)
- Asieh Heirani-Tabasi
- a Department of Biology , Faculty of Science, Ferdowsi University of Mashhad , Mashhad , Iran.,b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Hojjat Naderi-Meshkin
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Maryam M Matin
- a Department of Biology , Faculty of Science, Ferdowsi University of Mashhad , Mashhad , Iran.,b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran.,c Cell and Molecular Biotechnology Research Group , Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad , Iran
| | - Mahdi Mirahmadi
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Mina Shahriyari
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | | | - Nasser Sanjar Moussavi
- e Department of Surgery , Faculty of Medicine, Islamic Azad University-Mashhad Branch , Iran
| | - Hamid Reza Bidkhori
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Mahmood Raeesolmohaddeseen
- b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran
| | - Ahmad Reza Bahrami
- a Department of Biology , Faculty of Science, Ferdowsi University of Mashhad , Mashhad , Iran.,b Stem Cells and Regenerative Medicine Research Group , Academic Center for Education, Culture and Research (ACECR), Khorasan Razavi Branch , Mashhad , Iran.,c Cell and Molecular Biotechnology Research Group , Institute of Biotechnology, Ferdowsi University of Mashhad , Mashhad , Iran
| |
Collapse
|
18
|
Zeeshan M, Murugadas A, Ghaskadbi S, Ramaswamy BR, Akbarsha MA. Ecotoxicological assessment of cobalt using Hydra model: ROS, oxidative stress, DNA damage, cell cycle arrest, and apoptosis as mechanisms of toxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2017; 224:54-69. [PMID: 28222982 DOI: 10.1016/j.envpol.2016.12.042] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 12/14/2016] [Accepted: 12/17/2016] [Indexed: 06/06/2023]
Abstract
The mechanisms underlying cobalt toxicity in aquatic species in general and cnidarians in particular remain poorly understood. Herein we investigated cobalt toxicity in a Hydra model from morphological, histological, developmental, and molecular biological perspectives. Hydra, exposed to cobalt (0-60 mg/L), were altered in morphology, histology, and regeneration. Exposure to standardized sublethal doses of cobalt impaired feeding by affecting nematocytes, which in turn affected reproduction. At the cellular level, excessive ROS generation, as the principal mechanism of action, primarily occurred in the lysosomes, which was accompanied by the upregulation of expression of the antioxidant genes SOD, GST, GPx, and G6PD. The number of Hsp70 and FoxO transcripts also increased. Interestingly, the upregulations were higher in the 24-h than in the 48-h time-point group, indicating that ROS overwhelmed the cellular defense mechanisms at the latter time-point. Comet assay revealed DNA damage. Cell cycle analysis indicated the induction of apoptosis accompanied or not by cell cycle arrest. Immunoblot analyses revealed that cobalt treatment triggered mitochondria-mediated apoptosis as inferred from the modulation of the key proteins Bax, Bcl-2, and caspase-3. From this data, we suggest the use of Hydra as a model organism for the risk assessment of heavy metal pollution in aquatic ecosystems.
Collapse
Affiliation(s)
- Mohammed Zeeshan
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India; Dept. of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli 620024, India
| | - Anbazhagan Murugadas
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India; Dept. of Environmental Biotechnology, Bharathidasan University, Tiruchirappalli 620024, India
| | - Surendra Ghaskadbi
- Developmental Biology Group, MACS-Agharkar Research Institute, Pune 411004, India
| | | | - Mohammad Abdulkader Akbarsha
- Mahatma Gandhi-Doerenkamp Center, Bharathidasan University, Tiruchirappalli 620024, India; Department of Food Science and Nutrition, College of Food and Agriculture, King Saud University, Riyadh, Saudi Arabia.
| |
Collapse
|
19
|
Xiong H, Nie X, Zou Y, Gong C, Li Y, Wu H, Qiu H, Yang L, Zhuang L, Zhang P, Zhang J, Wang Y, Xiong H. Twist1 Enhances Hypoxia Induced Radioresistance in Cervical Cancer Cells by Promoting Nuclear EGFR Localization. J Cancer 2017; 8:345-353. [PMID: 28261334 PMCID: PMC5332884 DOI: 10.7150/jca.16607] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 10/29/2016] [Indexed: 01/02/2023] Open
Abstract
Twist1 is a crucial transcription factor that regulates epithelial mesenchymal transition and involves in metastasis. Recent evidence suggests that Twist1 plays important role in hypoxia-induced radioresistance, but the underlying mechanism remains elusive. Here we investigated the change of Twist1 expression in human cervical squamous cancer cell line SiHa after hypoxia treatment. We also explored the role of Twist1 in radioresistance by manipulating the expression level of Twist1. We observed that hypoxia treatment elevated the expression of Twist1 in SiHa cells. Knockdown of Twist1 with siRNA increased the radiosensitivity of SiHa cells under hypoxia condition, accompanied by reduced levels of nuclear Epidermal Growth Factor Receptor (EGFR) and DNA-dependent protein kinase (DNA-PK). Conversely, overexpression of Twist1 led to increased radioresistance of SiHa cells, which in turn increased nuclear EGFR localization and expression levels of nuclear DNA-PK. Moreover, concomitant high expression of hypoxia-inducible factor-1α (HIF-1α) and Twist1 in primary tumors of cervical cancer patients correlated with the worse prognosis after irradiation treatment. Taken together, these data provide new insights into molecular mechanism underlying hypoxia-induced radioresistance in cervical cancer cells, and suggest that Twist1 is a promising molecular target to improve the efficacy of cancer radiotherapy.
Collapse
Affiliation(s)
- Hua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Xin Nie
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yanmei Zou
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Chen Gong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yang Li
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hua Wu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Hong Qiu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Lin Yang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Liang Zhuang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Peng Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Jing Zhang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yihua Wang
- Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | - Huihua Xiong
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
20
|
Rath S, Das L, Kokate SB, Ghosh N, Dixit P, Rout N, Singh SP, Chattopadhyay S, Ashktorab H, Smoot DT, Swamy MM, Kundu TK, Crowe SE, Bhattacharyya A. Inhibition of histone/lysine acetyltransferase activity kills CoCl 2-treated and hypoxia-exposed gastric cancer cells and reduces their invasiveness. Int J Biochem Cell Biol 2016; 82:28-40. [PMID: 27890795 DOI: 10.1016/j.biocel.2016.11.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 10/23/2016] [Accepted: 11/21/2016] [Indexed: 12/17/2022]
Abstract
Hypoxia enhances immortality and metastatic properties of solid tumors. Deregulation of histone acetylation has been associated with several metastatic cancers but its effect on hypoxic responses of cancer cells is not known. This study aimed at understanding the effectiveness of the hydrazinocurcumin, CTK7A, an inhibitor of p300 lysine/histone acetyltransferase (KAT/HAT) activity, in inducing apoptosis of gastric cancer cells (GCCs) exposed to cobalt chloride (CoCl2), a hypoxia-mimetic chemical, or 1% O2. Here, we show that CTK7A-induced hydrogen peroxide (H2O2) generation in CoCl2-exposed and invasive gastric cancer cells (GCCs) leads to p38 MAPK-mediated Noxa expression and thereafter, mitochondrial apoptotic events. Noxa induction in normal immortalized gastric epithelial cells after CTK7A and hypoxia-exposure is remarkably less in comparison to similarly-treated GCCs. Moreover, hypoxia-exposed GCCs, which have acquired invasive properties, become apoptotic after CTK7A treatment to a significantly higher extent than normoxic cells. Thus, we show the potential of CTK7A in sensitizing hypoxic and metastatic GCCs to apoptosis induction.
Collapse
Affiliation(s)
- Suvasmita Rath
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, 752050, Odisha, India
| | - Lopamudra Das
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, 752050, Odisha, India
| | - Shrikant Babanrao Kokate
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, 752050, Odisha, India
| | - Nilabh Ghosh
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, 752050, Odisha, India
| | - Pragyesh Dixit
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, 752050, Odisha, India
| | - Niranjan Rout
- Oncopathology, Acharya Harihar Regional Cancer Centre, Cuttack, 753007, Odisha, India
| | - Shivaram P Singh
- Department of Gastroenterology, SCB Medical College, Cuttack, 753007, Odisha, India
| | - Subhasis Chattopadhyay
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, 752050, Odisha, India
| | - Hassan Ashktorab
- Department of Medicine, Howard University, Washington, DC, 20059, USA
| | - Duane T Smoot
- Department of Medicine, Meharry Medical Center, Nashville, TN, 37208, USA
| | - Mahadeva M Swamy
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Jakkur PO, Bangalore 560064, Karnataka, India
| | - Tapas K Kundu
- Transcription and Disease Laboratory, Molecular Biology and Genetics Unit, JNCASR, Jakkur PO, Bangalore 560064, Karnataka, India
| | - Sheila E Crowe
- School of Medicine, University of California, San Diego, CA, 92093, USA
| | - Asima Bhattacharyya
- School of Biological Sciences, National Institute of Science Education and Research (NISER) Bhubaneswar, HBNI, P.O. Bhimpur-Padanpur, Via Jatni, Dist. Khurda, 752050, Odisha, India.
| |
Collapse
|
21
|
Song B, Zhou T, Yang WL, Liu J, Shao LQ. Programmed cell death in periodontitis: recent advances and future perspectives. Oral Dis 2016; 23:609-619. [PMID: 27576069 DOI: 10.1111/odi.12574] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/31/2016] [Accepted: 08/19/2016] [Indexed: 12/18/2022]
Abstract
Periodontitis is a highly prevalent infectious disease, characterized by destruction of the periodontium, and is the main cause of tooth loss. Periodontitis is initiated by periodontal pathogens, while other risk factors including smoking, stress, and systemic diseases aggravate its progression. Periodontitis affects many people worldwide, but the molecular mechanisms by which pathogens and risk factors destroy the periodontium are unclear. Programmed cell death (PCD), different from necrosis, is an active cell death mediated by a cascade of gene expression events and can be mainly classified into apoptosis, autophagy, necroptosis, and pyroptosis. Although PCD is involved in many inflammatory diseases, its correlation with periodontitis is unclear. After reviewing the relevant published articles, we found that apoptosis has indeed been reported to play a role in periodontitis. However, the role of autophagy in periodontitis needs further verification. Additionally, implication of necroptosis or pyroptosis in periodontitis remains unknown. Therefore, we recommend future studies, which will unravel the pivotal role of PCD in periodontitis, allowing us to prevent, diagnose, and treat the disease, as well as predict its outcomes.
Collapse
Affiliation(s)
- B Song
- Guizhou Provincial People's Hospital, Guiyang, China.,Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - T Zhou
- Guizhou Provincial People's Hospital, Guiyang, China
| | - W L Yang
- Guizhou Provincial People's Hospital, Guiyang, China
| | - J Liu
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - L Q Shao
- Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
22
|
Lualdi M, Pedrini E, Rea K, Monti L, Scaldaferri D, Gariboldi M, Camporeale A, Ghia P, Monti E, Tomassetti A, Acquati F, Taramelli R. Pleiotropic modes of action in tumor cells of RNASET2, an evolutionary highly conserved extracellular RNase. Oncotarget 2016; 6:7851-65. [PMID: 25797262 PMCID: PMC4480721 DOI: 10.18632/oncotarget.3490] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 02/02/2015] [Indexed: 02/03/2023] Open
Abstract
As widely recognized, tumor growth entails a close and complex cross-talk among cancer cells and the surrounding tumor microenvironment. We recently described the human RNASET2 gene as one key player of such microenvironmental cross-talk. Indeed, the protein encoded by this gene is an extracellular RNase which is able to control cancer growth in a non-cell autonomous mode by inducing a sustained recruitment of immune-competent cells belonging to the monocyte/macrophage lineage within a growing tumor mass. Here, we asked whether this oncosuppressor gene is sensitive to stress challenges and whether it can trigger cell-intrinsic processes as well. Indeed, RNASET2 expression levels were consistently found to increase following stress induction. Moreover, changes in RNASET2 expression levels turned out to affect several cancer-related parameters in vitro in an ovarian cancer cell line model. Of note, a remarkable rearrangement of the actin cytoskeleton organization, together with changes in cell adhesion and motility, emerged as putative mechanisms by which such cell-autonomous role could occur. Altogether, these biological features allow to put forward the hypothesis that the RNASET2 protein can act as a molecular barrier for limiting the damages and tissue remodeling events occurring during the earlier step of cell transformation.
Collapse
Affiliation(s)
- Marta Lualdi
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Varese, Italy
| | - Edoardo Pedrini
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Varese, Italy
| | - Katia Rea
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Laura Monti
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Varese, Italy
| | - Debora Scaldaferri
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Varese, Italy
| | - Marzia Gariboldi
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Busto Arsizio, Italy
| | - Annalisa Camporeale
- Division of Molecular Oncology and Department of Onco-Hematology, IRCCS Ospedale San Raffaele, Milan, Italy.,Present address: Molecular Biotechnology Center and Department of Molecular Biotechnology and Life Sciences, University of Turin, Turin, Italy
| | - Paolo Ghia
- Division of Molecular Oncology and Department of Onco-Hematology, IRCCS Ospedale San Raffaele, Milan, Italy.,Università Vita-Salute San Raffaele, Milan, Italy
| | - Elena Monti
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Busto Arsizio, Italy
| | - Antonella Tomassetti
- Unit of Molecular Therapies, Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Francesco Acquati
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Varese, Italy
| | - Roberto Taramelli
- Department of Theoretical and Applied Sciences, Università degli Studi dell'Insubria, Varese, Italy
| |
Collapse
|
23
|
Hypoxia induced epithelial-mesenchymal transition and vasculogenic mimicry formation by promoting Bcl-2/Twist1 cooperation. Exp Mol Pathol 2015; 99:383-91. [PMID: 26318343 DOI: 10.1016/j.yexmp.2015.08.009] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/20/2015] [Indexed: 10/23/2022]
Abstract
Hypoxia plays a pivotal role in tumor progression. The functions of hypoxia and subsequent Bcl-2/Twist1 activation in epithelial-mesenchymal transition (EMT) and vasculogenic mimicry (VM) formation are currently unclear. This study aimed to investigate the role of Bcl-2/Twist1 cooperation in hypoxia-induced EMT and VM formation. In in vitro experiments, we found that hypoxia resulted in co-overexpression of Bcl-2 and Twist1, facilitated Twist1 nuclear translocation and promoted EMT and VM formation. Co-overexpression of Bcl-2 and Twist1 under normoxia could also induce EMT and promote VM formation. Furthermore, blocking Bcl-2 or Twist1 attenuated the effects of hypoxia on EMT progress and VM formation in hepatocellular carcinoma cells. In in vivo experiments, the mechanism by which hypoxia promoted Bcl-2 and Twist1 co-overexpression and induced EMT process and VM formation was demonstrated using murine xenograft models. These results above suggest that hypoxia could activate the cooperation of Bcl-2 and Twist1, Bcl-2 plays an important role in assisting Twist1 nuclear translocation which could change the expression of a wide range of genes and lead to the induction of EMT and VM formation.
Collapse
|
24
|
LIU KUN, SUN BAOCUN, ZHAO XIULAN, WANG XUDONG, LI YANLEI, QIU ZHIQIANG, LIU TIEJU, GU QIANG, DONG XUEYI, ZHANG YANHUI, WANG YONG, ZHAO NAN. Hypoxia promotes vasculogenic mimicry formation by the Twist1-Bmi1 connection in hepatocellular carcinoma. Int J Mol Med 2015. [DOI: 10.3892/ijmm.2015.2293] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
25
|
Zhang YB, Wang X, Meister EA, Gong KR, Yan SC, Lu GW, Ji XM, Shao G. The effects of CoCl2 on HIF-1α protein under experimental conditions of autoprogressive hypoxia using mouse models. Int J Mol Sci 2014; 15:10999-1012. [PMID: 24945310 PMCID: PMC4100194 DOI: 10.3390/ijms150610999] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2014] [Revised: 06/10/2014] [Accepted: 06/11/2014] [Indexed: 12/22/2022] Open
Abstract
It is well known that cobalt chloride (CoCl2) can enhance the stability of hypoxia-inducible factor (HIF)-1α. The aim of this study is to detect the effect of CoCl2 on the hypoxia tolerance of mice which were repeatedly exposed to autoprogressive hypoxia. Balb/c mice were randomly divided into groups of chemical pretreatment and normal saline (NS), respectively injected with CoCl2 and NS 3 h before exposure to hypoxia for 0 run (H0), 1 run (H1), and 4 runs (H4). Western Blot, electrophoretic mobility shift assay (EMSA), extracellular recordings population spikes in area cornus ammonis I (CA 1) of mouse hippocampal slices and real-time were used in this study. Our results demonstrated that the tolerance of mice to hypoxia, the changes of HIF-1α protein level and HIF-1 DNA binding activity in mice hippocampus, the mRNA level of erythropoietin (EPO) and vascular endothelial growth factor (VEGF), and the disappearance time of population spikes of hippocampal slices were substantially different between the control group and the CoCl2 group. Over-induction of HIF-1α by pretreatment with CoCl2 before hypoxia did not increase the hypoxia tolerance.
Collapse
Affiliation(s)
- Yan-Bo Zhang
- Department of Neurology, Affiliated Hospital of Tai Shan Medical University, Taishan 271000, China.
| | - Xiulian Wang
- Department of Intensive Care Unit , 2nd Affiliated Hospital of Baotou Medical College, Baotou 014030, China.
| | - Edward A Meister
- Department of Medicine, University of Arizona, Tucson, AZ 85721, USA.
| | - Ke-Rui Gong
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| | - Shao-Chun Yan
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| | - Guo-Wei Lu
- Institute for Hypoxia Medicine, Xuanwu Hospital of Capital Medical University, Beijing 10054, China.
| | - Xun-Ming Ji
- Institute for Hypoxia Medicine, Xuanwu Hospital of Capital Medical University, Beijing 10054, China.
| | - Guo Shao
- Biomedicine Research Center and Basic Medical College, Baotou Medical College, Baotou 014060, China.
| |
Collapse
|
26
|
Angelé-Martínez C, Goodman C, Brumaghim J. Metal-mediated DNA damage and cell death: mechanisms, detection methods, and cellular consequences. Metallomics 2014; 6:1358-81. [DOI: 10.1039/c4mt00057a] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Metal ions cause various types of DNA damage by multiple mechanisms, and this damage is a primary cause of cell death and disease.
Collapse
Affiliation(s)
| | - Craig Goodman
- Department of Chemistry
- Clemson University
- Clemson, USA
| | | |
Collapse
|
27
|
Lee EJ, Cha HJ, Ahn KJ, An IS, An S, Bae S. Oridonin exerts protective effects against hydrogen peroxide‑induced damage by altering microRNA expression profiles in human dermal fibroblasts. Int J Mol Med 2013; 32:1345-54. [PMID: 24145963 DOI: 10.3892/ijmm.2013.1533] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2013] [Accepted: 10/10/2013] [Indexed: 11/05/2022] Open
Abstract
The aim of the present study was to evaluate the protective effects of oridonin on hydrogen peroxide-induced cytotoxicity in normal human dermal fibroblasts (NHDFs) using microRNA (miRNA) expression profile analysis. Oridonin was not cytotoxic at low doses (≤5 µM) in the NHDFs, and pre-treatment of the cells with oridonin significantly reduced hydrogen dioxide (H2O2)-mediated cytotoxicity and cell death. Whereas oridonin showed no free radical scavenging activity in in vitro and in vivo antioxidant assays, treatment of the NHDFs with oridonin was associated with intracellular scavenging of reactive oxygen species. High-density miRNA microarray analysis revealed alterations in the expression profiles of specific miRNAs (5 upregulated and 22 downregulated) following treatment with oridonin in the H2O2-treated NHDFs. Moreover, the use of a miRNA target-gene prediction tool and Gene Ontology analysis demonstrated that these miRNAs are functionally related to the inhibition of apoptosis and cell growth. These data provide valuable insight into the cellular responses to oridonin in H2O2-induced damage in NHDFs.
Collapse
Affiliation(s)
- Eun-Jin Lee
- Molecular-Targeted Drug Research Center, Konkuk University, Seoul 143-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
28
|
Kewitz S, Staege MS. Expression and Regulation of the Endogenous Retrovirus 3 in Hodgkin's Lymphoma Cells. Front Oncol 2013; 3:179. [PMID: 23847767 PMCID: PMC3706881 DOI: 10.3389/fonc.2013.00179] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2013] [Accepted: 06/25/2013] [Indexed: 11/13/2022] Open
Abstract
Human endogenous retroviruses (ERV) are an integral part of our genome. Expression of ERV is usually switched off but reactivation of ERV has been observed in varying human diseases including cancer. Recently, reactivation of ERV associated promoters in Hodgkin's lymphoma (HL) cells has been described. Despite relatively good prognosis, not all patients with HL can be cured with the established therapy and this therapy is associated with severe late side effects. Therefore, new targets are required for the development of future treatment strategies. Reactivated ERV might represent such target structures. Therefore, we asked which ERV loci are expressed in HL cells. Using DNA microarray analysis, we found no evidence for a general activation of ERV transcription in HL cells. In contrast, we observed down-regulation of ERV3, an ERV with potential tumor suppressor function, in HL cells in comparison to normal blood cells. Interestingly, ERV3 was also differentially expressed in published DNA microarray data from resting versus cycling B cells. Treatment of HL cells with the histone deacetylase inhibitor vorinostat strongly up-regulated ERV3 expression. In addition, we observed up-regulation in HL cells after treatment with hypoxia-mimetic cobalt(II) chloride. Like vorinostat, cobalt(II) chloride inhibited cell growth of HL cells. Our results suggest that cell cycle inhibition of HL cells is accompanied by up-regulation of ERV3.
Collapse
Affiliation(s)
- Stefanie Kewitz
- Department of Pediatrics, Martin-Luther-University Halle-Wittenberg , Halle , Germany
| | | |
Collapse
|
29
|
Cheng JC, Klausen C, Leung PCK. Hypoxia-inducible factor 1 alpha mediates epidermal growth factor-induced down-regulation of E-cadherin expression and cell invasion in human ovarian cancer cells. Cancer Lett 2012; 329:197-206. [PMID: 23142284 DOI: 10.1016/j.canlet.2012.10.029] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 10/23/2012] [Accepted: 10/29/2012] [Indexed: 01/11/2023]
Abstract
Hypoxia-inducible factor 1α (HIF-1α) regulates the transcription of a number of genes under hypoxia and other extracellular stimulations. It has been shown that E-cadherin is down-regulated by epidermal growth factor receptor (EGF) stimulation, and that cells with low E-cadherin expression are more invasive. Our recent study demonstrated a novel mechanism by which EGF down-regulates E-cadherin expression through production of hydrogen peroxide (H(2)O(2)) and the activation of p38 MAPK in human ovarian cancer cells. In this study, we were interested in examining the potential role of HIF-1α in cell invasion under normoxic conditions, specifically when cells are treated with EGF, which is known to down-regulate E-cadherin and increase invasiveness. We show that EGF treatment induces HIF-1α expression in two human ovarian cancer cell lines (SKOV3 and OVCAR5), and that this effect is diminished by treatment with a membrane-permeable H(2)O(2) scavenger, PEG-catalase. However, the induction of HIF-1α by EGF did not require the activation of p38 MAPK. Treatment with siRNA targeting HIF-1α reduces both basal and EGF-induced HIF-1α levels. Importantly, treatment with HIF-1α siRNA diminishes the up-regulation of Snail and Slug as well as the down-regulation of E-cadherin by EGF. The involvement of HIF-1α in the down-regulation of E-cadherin was confirmed with cobalt chloride (CoCl(2)), a hypoxia-mimetic reagent. Finally, we also show that EGF-induced cell invasion is attenuated by treatment with HIF-1α siRNA. This study demonstrates an important role for HIF-1α in mediating the effects of EGF on Snail, Slug and E-cadherin expression as well as invasiveness in human ovarian cancer cells.
Collapse
Affiliation(s)
- Jung-Chien Cheng
- Department of Obstetrics and Gynaecology, Child & Family Research Institute, University of British Columbia, Vancouver, British Columbia, Canada V5Z 4H4
| | | | | |
Collapse
|