GLORIA

GEOMAR Library Ocean Research Information Access

Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 468-468
    Abstract: Antioxidants are electron donors that can neutralize and inhibit reactive oxygen species (ROS). Some evidence has shown that not only normal cells but cancer cells also require antioxidants, as cancer cells produce even more ROS as a result of their rapid growth. N-acetylcysteine (NAC) and glutathione (GSH) are popular antioxidant supplements for humans. A recent study done by Martin O' team has revealed that NAC could accelerate lung cancer via reducing the ROS-related DNA damage in mice. Tak Mak's group has reported that the GSH is essential for breast cancer initiation in mice. Liver cancer (hepatocellular carcinoma) is the fifth leading cause of cancer-related death worldwide. Whether NAC and GSH would aggravate HCC remains unknown. In this study, we aimed to investigate the effects of popular antioxidant supplements including NAC and GSH on HCC both in vitro and in vivo. We first screened the cell viability and colony formation effects of NAC and GSH on 5 HCC cell lines, MHCC97L cells showed the most promoting effects upon treatments. To further evaluate the effects of NAC and GSH on HCC initiation, growth and progression in vitro, we performed functional tests such as sphere formation, cell proliferation, migration and invasion assays in MHCC97L cells. Subcutaneous implantation and orthotopic liver injection mouse models were employed to further investigate the effects of NAC on tumorigenicity and tumor progression in vivo. To reveal the underlying mechanism, we also tested the expression levels of proteins involved in the ROS defence system. From the in vitro experiments, NAC-treated HCC cells promoted colony formation, suggesting that NAC might facilitate tumor cell growth. GSH increased the sphere formation, which is a test for self-renewal ability in vitro, and cell proliferation in MHCC97L cells. Significant enhancement in cell migratory and invasive abilities was observed in both NAC- and GSH-treated HCC cells. For the in vivo tumorigenicity assay, 120 mg/kg/day of NAC (based on conversion from the human equivalent dosage) was given to mice immediately after the tumor cell injection subcutaneously. Toxic side effects were observed in NAC-treated mice such as distended abdomen and weight loss despite a reduction in tumor incidence. In the orthotopic liver injection model, mice were treated with 60 mg/kg/day of NAC after tumor onset. NAC administration significantly increased the tumor masses and incidence of lung metastases when compared to the control group. Furthermore, NAC reduced the expression of p21 by western blotting while GSH showed no significant effect on protein markers involved in ROS defence pathways, such as AMT, P53 and H2AX. To conclude, our data show that antioxidants supplements could exacerbate HCC growth and progression in HCC cells in vitro and mice in vivo. The role of antioxidant supplements such as NAC and GSH in cancer therapy deserves more extensive studies. Citation Format: Xin Zhang, Eva Lee, Yu-Man Tsui, Man-Fong Sze, Yung-Tuen Chiu, Lo-Kong Chan, Daniel Wai-Hung Ho, Lu-Qing Zhao, Macrina Wai-Ling Lam, Lu Tian, Joyce Man-Fong Lee, Derek Lee, Carmen Chak-Lui Wong, Irene Oi-Lin Ng. Antioxidant supplements may accelerate the formation and progression of hepatocellular carcinoma in vitro and in vivo [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 468.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 91-91
    Abstract: RalA is a Ras-related small GTP binding protein A; however, its functional roles and regulatory mechanisms in hepatocellular carcinoma (HCC) are unclear. In this study, using the RNA-Seq data from TCGA database and our in-house HKU database, we observed that RalA expression was significantly up-regulated in human HCCs (P=0.001). This RalA over-expression was validated in a separate cohort of our HCC patients. Upon clinicopathological correlation of RalA in HCC, we found that over-expression of RalA was associated with more aggressive features of HCC patients, with more frequent tumor microsatellite formation (P=0.001), venous invasion (P=0.005) and absence of tumor encapsulation (P=0.005). The over-expression also correlated with poorer overall survival of HCC patients (P & lt;0.001). Functionally, we established RalA stable knockdown (KD) in three HCC cell lines (Hep3B, BEL7402 and MHCC-97L-Luc) and demonstrated that KD of RalA significantly inhibited cell proliferation, colony formation, migration and invasion in vitro. Conversely, ectopic expression of RalA, by transfecting the RalA dominant active form G23V construct into HCC cells, promoted HCC metastasis using transwell invasion assays. Also, with immunofluorescence assay, RalA mainly located in the cytoplasm and cell membrane. Over-expression of RalA changed the HCC cell morphology from polygonal to spindled shape, suggestive of epithelial-mesenchymal transition. We conducted in vivo study using the orthotopic liver injection model of RalA stable-KD MHCC-97L cells in nude mice, and observed that KD of RalA suppressed HCC tumorigenicity and reduced lung metastasis (P & lt;0.001). Moreover, KD of RalA also reduced sphere formation ability of HCC cells, as well as suppressed the expression of stemness markers, including CD24, NANOG, NOTCH1, NESTIN, and EpCAM, using qPCR. For the liver cancer stem cell surface markers, the expression level of CD24 was significantly decreased in RalA KD Hep3B and BEL7402 cells by flow cytometry. Additionally, with RNA-Seq analysis of Hep3B and BEL7402 shRalA clones, 12 genes were found to be significantly altered (among them 5 were up-regulated and 7 were down-regulated) for both cell lines, when compared with in-house cohort as well as TCGA cohort. Further analysis showed that RalA expression level was positively and negatively correlated with a small group of genes. Taken together, our findings have shown that RalA is significantly up-regulated in human HCCs and its over-expression enhances HCC metastasis and cancer stemness. Further investigation of the interplay between RalA and its downstream targets will derive novel mechanistic insight regarding the oncogenic role of RalA in HCC. Citation Format: Luqing Zhao, Lo-Kong Chan, Daniel Wai-Hung Ho, Goofy Yu-Man Tsui, Macrina Wai-Ling Lam, Charles Shing Kam, Karen Man-Fong Sze, Vanilla Xin Zhang, Abdullah Husain, Joyce Man-Fong Lee, Irene Oi-Lin Ng. RalA is frequently up-regulated in human hepatocellular carcinoma and promotes metastasis and cancer stemness [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2018; 2018 Apr 14-18; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2018;78(13 Suppl):Abstract nr 91.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2018
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Journal of Hepatology, Elsevier BV, Vol. 77 ( 2022-07), p. S38-
    Type of Medium: Online Resource
    ISSN: 0168-8278
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2027112-8
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    Frontiers Media SA ; 2022
    In:  Frontiers in Medicine Vol. 9 ( 2022-7-5)
    In: Frontiers in Medicine, Frontiers Media SA, Vol. 9 ( 2022-7-5)
    Abstract: Lines of evidence implicate CENPF and FOXM1 may have novel co-operative roles in driving hepatocellular carcinoma (HCC). Objective We investigated the clinicopathological correlation, functional characterization, molecular mechanism and translational significance of CENPF and FOXM1. Methods We carried out integrative studies investigating functional synergism of CENPF and FOXM1 in HCC and its metastasis. Human HCC samples, HCC cell lines and mouse model were used in the studies. Stable knockdown, q-PCR, Western blotting, whole-transcriptomic sequencing (RNA-seq), as well as cell and mouse assays were performed. Results Upon clinicopathological correlation, we found that co-overexpression of CENPF and FOXM1 in human HCCs was associated with more aggressive tumor behavior including presence of venous invasion, tumor microsatellite formation, and absence of tumor encapsulation. Moreover, co-silencing FOXM1 and CENPF using shRNA approach in HCC cell lines resulted in significantly reduced cell proliferation. Furthermore, our RNA-seq and differential gene expression analysis delineated that CENPF and FOXM1 co-regulated a specific set of target genes in various metabolic processes and oncogenic signaling pathways. Among them, POLD1, which encodes the catalytic subunit of DNA polymerase δ, was ranked as the top downstream target co-regulated by CENPF and FOXM1. POLD1 expression was positively correlated with that of FOXM1 and CENPF in HCCs. In addition, POLD1 expression was significantly upregulated in HCC tumors. Functionally, in vivo orthotopic injection model showed that stable knockdown of POLD1 in HCC cells suppressed tumor incidence and tumorigenicity and had a trend of diminished lung metastasis. Conclusion Taken together, our data suggest that CENPF and FOXM1 could synergistically support hepatocarcinogenesis via the regulation of POLD1. CENPF and FOXM1 may represent new vulnerabilities to novel drug-based therapy in HCC.
    Type of Medium: Online Resource
    ISSN: 2296-858X
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2022
    detail.hit.zdb_id: 2775999-4
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    Online Resource
    Online Resource
    Springer Science and Business Media LLC ; 2015
    In:  Cell Research Vol. 25, No. 12 ( 2015-12), p. 1299-1313
    In: Cell Research, Springer Science and Business Media LLC, Vol. 25, No. 12 ( 2015-12), p. 1299-1313
    Type of Medium: Online Resource
    ISSN: 1001-0602 , 1748-7838
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2015
    detail.hit.zdb_id: 2082402-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4848-4848
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 4848-4848
    Abstract: Hepatocellular carcinoma (HCC) is the second leading cause of cancer death worldwide and metastasis is regarded as the major cause of HCC-associated lethality. In this study, we have analysed the TCGA whole-transcriptome sequencing data of paired human HCC samples (n=50), and identified Forkhead Box M1 (FOXM1) and Centromere Protein F (CENPF) to be the top-listing upregulated genes. Interestingly, both of them are the essential components in cell-cycle progression. FOXM1 encodes for the cell-cycle-dependent transcription factor that regulates genes for DNA replication and mitosis, while CENPF encodes for the centromere protein that is required for kinetochore function and chromosome segregation in mitosis. We hypothesized that the upregulation of FOXM1-CENPF signaling axis may drive hepatocarcinogenesis. In our human HCC cohort (n=34), FOXM1 and CENPF were shown to be upregulated compared with the non-tumorous liver tissues, and their mRNA expressions were positively correlated (p & lt;0.0001). Co-upregulation of FOXM1 and CENPF in patient samples was demonstrated to be positively correlated with the absence of tumor encapsulation (p=0.035) in our clinico-pathological correlation analysis, whereas the samples without their co-upregulation was less likely to be correlated with venous invasion (p =0.045). This indicated that FOXM1 and CENPF are likely to be associated with HCC metastasis synergistically. Stable single and co-knockdown clones of FOXM1 and CENPF in four HCC cell lines (BEL7402, SMMC-7721, MHCC-97L, Huh7) were established using short-hairpin (sh) RNA approach for subsequent functional characterization. Both single and co-knockdown of FOXM1 and CENPF cells have shown reduced proliferation rate (p & lt;0.05) in cell proliferation assay. Moreover, all single and co-knockdown of FOXM1 and CENPF cell lines were found to have a at least 2-fold reduction in migration and invasion rate (p & lt;0.0001) in transwell cell migration and invasion assay. These suggested that FOXM1 and CENPF are critical for HCC cell proliferation, migration and invasion. To assess the translational significance of targeting FOXM1 and CENPF, seven HCC cell lines (BEL7402, SMMC-7721, MHCC-97L, Huh7, Hep3B, HepG2, PLC/PRF/5) were challenged with thiostrepton and zoledronic acid, which are inhibitors against FOXM1 and CENPF, respectively. Intriguingly, both individual and combined treatments of these inhibitors effectively inhibited HCC cell growth. It was also demonstrated that such inhibition acts via suppressing the respective endogenous transcript and protein expression of FOXM1 and CENPF. Taken together, our study has demonstrated that FOXM1 and CENPF play a critical oncogenic role in HCC and they may function as an attractive molecular therapeutic target. This study is supported in part by Health and Medical Research Fund (03142836). Citation Format: Wai Ling Macrina Lam, Lo Kong Chan, Daniel Wai-Hung Ho, Charles Shing Kam, Irene Oi-Lin Ng. The functional synergism and pro-metastatic role of FOXM1 and CENPF in hepatocellular carcinoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2017; 2017 Apr 1-5; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2017;77(13 Suppl):Abstract nr 4848. doi:10.1158/1538-7445.AM2017-4848
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5728-5728
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 5728-5728
    Abstract: Liver fibrosis is characterized by hepatic stellate cell activation and extracellular matrix deposition upon persistent injury and inflammation, which can impair hepatic function and its ability to regenerate. The origin and physiological role of facultative liver progenitor cells (LPCs) have been a controversial issue as it was found to be a major player to regenerate the damaged liver, but it is also associated with fibrosis, disease progression or tumor initiation. The receptor for advanced glycation end products (RAGE) signaling axis is often associated with chronic inflammation-associated tissue damage and plays an essential role in modulating the tumor microenvironment. Our previous data suggested that RAGE mediates LPC expansion, onset of liver fibrosis and HCC formation (Pusterla et al. Hepatology. 2013.) Hence, in this study, we seek to delineate the functional role and underlying mechanism of RAGE activity in LPC activation in response to inflammation-associated liver injury. R26TomHnf1β-CreER transgenic mice were crossed with Rage flox/flox (Ragefl/fl) mice to generate tamoxifen-inducible LPC-specific RAGE knockout mice (RAGEΔLPC). They were exposed to a choline-deficient ethionine-supplemented (CDE) diet for three weeks to induce liver damage. Ablation of RAGE in LPCs strongly impairs LPC expanding capacities in CDE diet-treated mice. Strikingly, this is accompanied by reduction of activated hepatic stellate cells and bridging fibrosis. This demonstrated that RAGE signaling in LPCs is a mediator of liver fibrosis in vivo. Necroinflammation is known to be associated with liver fibrosis. To investigate the role of RAGE in LPCs in the context of necroinflammation in vitro, primary LPCs were isolated from CDE-treated Ragefl/fl C57BL/6 mouse. Wildtype and knockout Rage cell lines were established. LPCs were stimulated with supernatants from necrotic hepatocytes followed by whole transcriptome sequencing to identify downstream targets of RAGE-dependent pathways. Stress response, inflammatory and pro-fibrotic pathways were enriched in LPCs upon treatment with necrotic medium. Most interestingly, signaling pathways that regulate organ size, tissue homeostasis and cell survival were found to be RAGE-dependent. Moreover, clusters of stem cell renewal-related genes were deregulated upon ablation of RAGE. In line with the whole transcriptome profile, we demonstrated that ablation of RAGE attenuates LPCs organoid-forming ability, implying that RAGE regulates stemness properties of LPCs. Our recent results demonstrated that RAGE is required for LPCs activation and proliferation, as well as the crosstalk with stellate cells in supporting fibrogenesis. Taken together, our data uncovers a potential mechanistic insight on the role of RAGE in LPCs in association with fibrosis upon chronic liver injury. Citation Format: Wai Ling Macrina Lam, Gisela Gabernet, Tanja Poth, Aurora De Ponti, Anna Saborowski, Sven Nahnsen, Doris Schneller, Peter Angel. Liver progenitor cells induces fibrosis via RAGE signaling upon liver injury [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 5728.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Journal of the American College of Cardiology, Elsevier BV, Vol. 41, No. 6 ( 2003-03), p. 210-
    Type of Medium: Online Resource
    ISSN: 0735-1097
    RVK:
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2003
    detail.hit.zdb_id: 1468327-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Scientific Reports, Springer Science and Business Media LLC, Vol. 6, No. 1 ( 2016-02-15)
    Abstract: Hydrogen sulfide is a highly toxic gas—second only to carbon monoxide as a cause of inhalational deaths. Its mechanism of toxicity is only partially known and no specific therapy exists for sulfide poisoning. We show in several cell types, including human inducible pluripotent stem cell (hiPSC)-derived neurons, that sulfide inhibited complex IV of the mitochondrial respiratory chain and induced apoptosis. Sulfide increased hydroxyl radical production in isolated mouse heart mitochondria and F 2 -isoprostanes in brains and hearts of mice. The vitamin B 12 analog cobinamide reversed the cellular toxicity of sulfide and rescued Drosophila melanogaster and mice from lethal exposures of hydrogen sulfide gas. Cobinamide worked through two distinct mechanisms: direct reversal of complex IV inhibition and neutralization of sulfide-generated reactive oxygen species. We conclude that sulfide produces a high degree of oxidative stress in cells and tissues and that cobinamide has promise as a first specific treatment for sulfide poisoning.
    Type of Medium: Online Resource
    ISSN: 2045-2322
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2016
    detail.hit.zdb_id: 2615211-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 10
    In: eLife, eLife Sciences Publications, Ltd, Vol. 8 ( 2019-05-21)
    Abstract: The mammalian target of rapamycin complex 1 (mTORC1) regulates cell growth, metabolism, and autophagy. Extensive research has focused on pathways that activate mTORC1 like growth factors and amino acids; however, much less is known about signaling cues that directly inhibit mTORC1 activity. Here, we report that G-protein coupled receptors (GPCRs) paired to Gαs proteins increase cyclic adenosine 3’5’ monophosphate (cAMP) to activate protein kinase A (PKA) and inhibit mTORC1. Mechanistically, PKA phosphorylates the mTORC1 component Raptor on Ser 791, leading to decreased mTORC1 activity. Consistently, in cells where Raptor Ser 791 is mutated to Ala, mTORC1 activity is partially rescued even after PKA activation. Gαs-coupled GPCRs stimulation leads to inhibition of mTORC1 in multiple cell lines and mouse tissues. Our results uncover a signaling pathway that directly inhibits mTORC1, and suggest that GPCRs paired to Gαs proteins may be potential therapeutic targets for human diseases with hyperactivated mTORC1.
    Type of Medium: Online Resource
    ISSN: 2050-084X
    Language: English
    Publisher: eLife Sciences Publications, Ltd
    Publication Date: 2019
    detail.hit.zdb_id: 2687154-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...