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
Filter
  • American Association for Cancer Research (AACR)  (24)
  • 2015-2019  (24)
Material
Publisher
  • American Association for Cancer Research (AACR)  (24)
Language
Years
  • 2015-2019  (24)
Year
Subjects(RVK)
  • 1
    In: Cancer Discovery, American Association for Cancer Research (AACR), Vol. 7, No. 1 ( 2017-01-01), p. 38-53
    Abstract: Somatic mutations in CREBBP occur frequently in B-cell lymphoma. Here, we show that loss of CREBBP facilitates the development of germinal center (GC)–derived lymphomas in mice. In both human and murine lymphomas, CREBBP loss-of-function resulted in focal depletion of enhancer H3K27 acetylation and aberrant transcriptional silencing of genes that regulate B-cell signaling and immune responses, including class II MHC. Mechanistically, CREBBP-regulated enhancers are counter-regulated by the BCL6 transcriptional repressor in a complex with SMRT and HDAC3, which we found to bind extensively to MHC class II loci. HDAC3 loss-of-function rescued repression of these enhancers and corresponding genes, including MHC class II, and more profoundly suppressed CREBBP-mutant lymphomas in vitro and in vivo. Hence, CREBBP loss-of-function contributes to lymphomagenesis by enabling unopposed suppression of enhancers by BCL6/SMRT/HDAC3 complexes, suggesting HDAC3-targeted therapy as a precision approach for CREBBP-mutant lymphomas. Significance: Our findings establish the tumor suppressor function of CREBBP in GC lymphomas in which CREBBP mutations disable acetylation and result in unopposed deacetylation by BCL6/SMRT/HDAC3 complexes at enhancers of B-cell signaling and immune response genes. Hence, inhibition of HDAC3 can restore the enhancer histone acetylation and may serve as a targeted therapy for CREBBP-mutant lymphomas. Cancer Discov; 7(1); 38–53. ©2016 AACR. See related commentary by Höpken, p. 14. This article is highlighted in the In This Issue feature, p. 1
    Type of Medium: Online Resource
    ISSN: 2159-8274 , 2159-8290
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    detail.hit.zdb_id: 2607892-2
    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. 1071-1071
    Abstract: Background: The prognosis of gastric adenocarcinoma (GAC) patients with metastases is very poor. Understanding of molecular biology is limited. Hedgehog (Hh) signaling plays an important role in many tumor types and expression of Shh/Gli-1, two major molecules in Hh pathway has been documented in GAC. However, their clinical impact on GAC patients particularly in peritoneal metastasis remains elusive. Methods: Expression of Gli1 and Shh were examined using IHC in tissue microarrays containing more than 500 cases of GAC tissues with clinical annotation. The prognostic variables were determined using univariate and multivariate Cox regression analyses. GAC cell lines, patient-derived peritoneal metastatic cells and novel PDX metastatic model were used to determine the functional role of Shh/Gli-1 in vitro and in vivo. Genetic knockout Gli-1 using LentiCRISPR/Cas9 and Hh inhibitor GDC0449 as well as BET inhibitor were used to test their antitumor activities in GAC cell line and patient-derived cells. Cell proliferation, colony formation, invasion, tumor sphere assays and immunofluorescence were performed to evaluate their functionality and effects of targeted therapy. Results: Both Gli1 and Shh expression are significantly overexpressed in GAC tissue. Among 519 GAC cases, 80.76% and 87.02% were positive for nuclear Gli-1 and cytoplasmic Shh expression respectively, while the strong nuclear expression rate for Gli-1 is 69.56% and 50.10% for Shh. In the univariate Cox analysis, the overall survival was shorter for patients with high Gli-1 (p=0.018) or high Shh expression (p=0.038). In the multivariate cox analysis for both markers, only Gli-1 remained as an independently prognostic for short survival. We also observed high Gli-1 nuclear expression correlated with the presence of lymph node metastasis (p=0.032). Gli1 was highly expressed in most human malignant ascites cells. Interestingly, Gli-1 was significantly upregulated in mouse PDX-ascites cells compared to primary mice tumors. Genetic knockdown Gli-1 or pharmacologically inhibition of Gli-1 by GDC0449 Hh inhibitor or BET inhibitor JQ1 decreased Gli-1 and restored E-cadherin expression and significantly suppressed malignant cell properties and reduced population of cancer stem cells (ALDH1+ or CD133+) in patients' derived metastatic cells. Conclusions: These findings indicate that overexpression of Gli1 and Shh plays an important role in progression of peritoneal metastases of GAC. Targeting Gli1/Hh signaling may provide novel therapeutic strategies for GAC patients with peritoneal metastases. Keywords:Hh pathway, Gli1, Shh, Gastric cancer, metastasis Citation Format: Yan Xu, Yuan Li, Bovey Liu, Melissa Pool Pizzi, Yongxi Song, Kazuto Harada, Ailing Scott, Lang Ma, Jiankang Jin, Xiaochuan Dong, Ying Wang, Brian D. Badgwell, Jeannelyn S. Estrella, Roy-Chowdhuri Sinchita, Fatemeh G. Amlashi, Zhenning Wang, Shumei Song, Jaffer A. Ajani. Overexpression of SHH and GLI1 contributes to poor prognosis and peritoneal metastases in gastric adenocarcinoma [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 1071.
    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: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 7, No. 9 ( 2019-09-01), p. 1497-1510
    Abstract: Immune-checkpoint protein V-domain immunoglobulin suppressor of T-cell activation (VISTA) controls antitumor immunity and is a valuable target for cancer immunotherapy. This study identified a role of VISTA in regulating Toll-like receptor (TLR) signaling in myeloid cells and controlling myeloid cell–mediated inflammation and immunosuppression. VISTA modulated the polyubiquitination and protein expression of TRAF6. Consequently, VISTA dampened TLR-mediated activation of MAPK/AP-1 and IKK/NF-κB signaling cascades. At cellular levels, VISTA regulated the effector functions of myeloid-derived suppressor cells and tolerogenic dendritic cell (DC) subsets. Blocking VISTA augmented their ability to produce proinflammatory mediators and diminished their T cell–suppressive functions. These myeloid cell–dependent effects resulted in a stimulatory tumor microenvironment that promoted T-cell infiltration and activation. We conclude that VISTA is a critical myeloid cell–intrinsic immune-checkpoint protein and that the reprogramming of tolerogenic myeloid cells following VISTA blockade promotes the development of T cell–mediated antitumor immunity.
    Type of Medium: Online Resource
    ISSN: 2326-6066 , 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 20 ( 2017-10-15), p. 5491-5502
    Abstract: Lymphangioleiomyomatosis (LAM) is a progressive destructive neoplasm of the lung associated with inactivating mutations in the TSC1 or TSC2 tumor suppressor genes. Cell or animal models that accurately reflect the pathology of LAM have been challenging to develop. Here, we generated a robust human cell model of LAM by reprogramming TSC2 mutation–bearing fibroblasts from a patient with both tuberous sclerosis complex (TSC) and LAM (TSC-LAM) into induced pluripotent stem cells (iPSC), followed by selection of cells that resemble those found in LAM tumors by unbiased in vivo differentiation. We established expandable cell lines under smooth muscle cell (SMC) growth conditions that retained a patient-specific genomic TSC2+/− mutation and recapitulated the molecular and functional characteristics of pulmonary LAM cells. These include multiple indicators of hyperactive mTORC1 signaling, presence of specific neural crest and SMC markers, expression of VEGF-D and female sex hormone receptors, reduced autophagy, and metabolic reprogramming. Intriguingly, the LAM-like features of these cells suggest that haploinsufficiency at the TSC2 locus contributes to LAM pathology, and demonstrated that iPSC reprogramming and SMC lineage differentiation of somatic patient cells with germline mutations was a viable approach to generate LAM-like cells. The patient-derived SMC lines we have developed thus represent a novel cellular model of LAM that can advance our understanding of disease pathogenesis and develop therapeutic strategies against LAM. Cancer Res; 77(20); 5491–502. ©2017 AACR.
    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 ...
  • 5
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4064-4064
    Abstract: Background: NRG Oncology/NSABP B-52 neoadjuvant clinical trial was conducted to test if the addition of estrogen deprivation (ED) would improve the pCR rate in HER2+/ER+ breast cancer pts treated with docetaxel, carboplatin, trastuzumab, and pertuzumab (TCHP). A numerical increase in the pCR rate was observed with ED (46.1% v 40.9%), but the difference was not statistically significant. The purpose of this study was to determine the utility of using stromal tumor infiltrating lymphocytes (sTILs), mutations, established, and novel signatures, to assess their value for predicting pCR, particularly in the TCHP + ED arm. Methods: Whole transcriptome RNA-Seq and Ampli-Seq libraries were sequenced on the Ion Torrent platform. Mutations were assessed with a custom Ampli-Seq panel of 117 genes, including HER2-activated pathways and/or trastuzumab (T)-resistance markers. The 8-gene T-benefit signature was prospectively tested for association with pCR, and was previously validated in the adjuvant setting in B-31 and NCCTG N9831. Hot-spot mutations, sTILs, and signatures for immune cells, intrinsic subtypes, risk of recurrence proliferation (RORP), and MammaPrint, were also tested for associations with pCR. RNA-Seq data was also used to identify ED-predictive genes. Differential expression was assessed in normalized RNA-Seq data using DESeq2 and each sample was subtyped with the AIMS classifier. Wilcoxon two-sided test, chi-square, or Fisher's exact tests were used to assess associations with pCR. Results: Subtypes were determined with RNA-Seq data from pretreatment biopsies (N=230). The 8-gene T-benefit signature was associated with pCR. The high-, medium-, and low- T-benefit groups had pCR rates of 55%, 44%, and 6.8%, respectively (p=1.3e-13). Intrinsic subtypes were associated with pCR by comparing HER2E to all other subtypes combined (69% v 28%, p=3.9e-08). Hot spot mutations in PIK3CA alone (p=0.014), or combined with hot spot mutations in ERBB2, ERBB3, AKT1, PTEN, and MAP3K1, were associated with no pCR (p=0.0007) and may be useful as resistance markers. sTILs, MammaPrint, RORP, and some immune cell signatures also showed statistically significant associations with pCR but did not identify a subset of pts with an increased pCR rate in the TCHP + ED arm. In contrast, expression of the SH3BP2 gene was associated with pCR only in the TCHP + ED arm (interaction p=0.03). Conclusion: The previously validated 8-gene T-benefit signature identifies a subset of pts with very low pCR rate (6.8%) with TCHP. PIK3CA and other activating mutations were associated with no pCR. These findings may identify pts with resistant disease who may require a different treatment. Exploratory analyses suggest that SH3BP2 expression may identify pts who may benefit from TCHP + ED, but validation is required for clinically utility. Support: BCRF, U10CA180868, -180822; UG1CA189867, Genentech, PUMA Biother Citation Format: Katherine L. Pogue-Geile, Ying Wang, Huichen Feng, Corey Lipchik, Rim S. Kim, Reena S. Cecchini, Samuel A. Jacobs, Ashok Srinivasan, Joseph P. Costantino, Eleftherios P. Mamounas, Charles E. Geyer, Priya Rastogi, Peter C. Lucas, Soonmyung Paik, C. Kent Osborne, Norman Wolmark, Mothaffar F. Rimawi. Association of molecular signatures, mutations, and sTILs, with pCR in breast cancer patients in NRG Oncology/NSABP B-52 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 4064.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3048-3048
    Abstract: As an enzyme that removes N-glycans from glycopeptides, N-glycanase 1 (NGLY1) deglycosylates denatured glycoproteins and allows proteasome-mediated protein degradation to efficiently occur. Although NGLY1 is known for this pivotal function, information regarding the responses of human cancer and normal cells to NGLY1 suppression is limited. Here, we examined how NGLY1 expression affects viability, tumor growth, and responses to therapeutic agents in melanoma cells and an animal model. Molecular mechanisms contributing to NGLY1 suppression-induced anticancer responses were revealed by systems biology and chemical biology studies. Using computational and medicinal chemistry-assisted approaches, we established novel NGLY1-inhibitory small molecules. Compared with normal cells, NGLY1 was upregulated in melanoma cell lines and patient tumors. NGLY1 knockdown caused melanoma cell death and tumor growth retardation. Targeting NGLY1 induced pleiotropic responses, predominantly stress signaling-associated apoptosis and cytokine surges, which synergize with the anti-melanoma activity of chemotherapy and targeted therapy agents. Pharmacological and molecular biology tools that inactivate NGLY1 elicited highly similar responses in melanoma cells. Unlike normal cells, melanoma cells presented distinct responses and high vulnerability to NGLY1 suppression. Our work demonstrated the significance of NGLY1 in melanoma cells, provided mechanistic insights into how NGLY1 inactivation leads to eradication of melanoma with limited impact on normal cells, and suggested that targeting NGLY1 represents a novel anti-melanoma strategy with the opportunity for a broad therapeutic window. Citation Format: Victor J.T. Lin, Ashwini Zolekar, Nigam M. Mishra, Yin Ying Ho, Hamed S. Hayatshahi, Abhishek Parab, Rohit Sampat, Xiaoyan Liao, Peter Hoffmann, Jin Liu, Kyle A. Emmitte, Yu-Chieh Wang. Targeting protein deglycosylation as a newly identified vulnerable point in melanoma [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2019; 2019 Mar 29-Apr 3; Atlanta, GA. Philadelphia (PA): AACR; Cancer Res 2019;79(13 Suppl):Abstract nr 3048.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2019
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 4234-4234
    Abstract: Background: Higher educational levels have been linked to reduced risk of death in previous studies. Most of these studies, however, were conducted in the United States and Europe. Methods: We performed a pooled analysis to evaluate the association between educational levels and risk of death among 694,434 Asian individuals from 15 prospective cohort studies that participated in the Asia Cohort Consortium. The analysis included 103,023 deaths during a mean follow-up period of 12.5 years, among which 33,945 were due to cancer and 34,645 were due to cardiovascular diseases (CVD). We employed Cox proportional hazards regression models to estimate hazard ratios (HRs) and 95% confidence intervals (95% CIs) for the association between educational level and the risk of death after adjusting for potential confounders. Results: Higher educational levels were significantly associated with lower risk of death from all causes; compared to a low educational level (≤ primary school), HRs and 95% CIs for secondary education, trade/technical education, and ≥ university degree were 0.86 (0.83-0.90), 0.80 (0.71-0.89), and 0.68 (0.61-0.75), respectively (Ptrend & lt;0.0001). Similar associations were observed for cancer or CVD-specific mortality, with HRs of 0.93 (0.89-0.97), 0.87 (0.78-0.97), and 0.79 (0.71-0.87) for risk of cancer deaths with increasing levels of education (Ptrend = 0.003) and HRs of 0.86 (0.81-0.91), 0.75 (0.64-0.87), and 0.64 (0.57-0.73) for risk of CVD deaths with increasing levels of education (Ptrend & lt;0.0001). The pattern of the association among East Asians and South Asians was similar; compared to ≤ primary school, HR (95% CI) for all-cause mortality associated with ≥ university degree was 0.68 (0.61-0.76) among 539,724 East Asians (Chinese, Japanese, and Korean) and 0.61 (0.54-0.69) among 154,710 South Asians (Indians and Bangladeshis) included in this analysis. Conclusions: Higher educational level was associated with substantially lower risk of death in Asian populations including both East and South Asians. Citation Format: Keming Yang, Ying Zhang, Eiko Saito, Yu Chen, Prakash C. Gupta, Yu-Tang Gao, Akiko Tamakoshi, Yong-Bing Xiang, Woon-Puay Koh, Norie Sawada, Kemmyo Sugiyama, Yasutake Tomata, Atsuko Sadakane, Chisato Nagata, San-Lin You, Renwei Wang, Myung-Hee Shin, Habibul Ahsan, Wen-Harn Pan, Mangesh S. Pednekar, Jian-Min Yuan, Xiao-Ou Shu, Hui Cai, Shoichiro Tsugane, Ichiro Tsuji, Seiki Kanemura, Keiko Wada, Yoon-Ok Ahn, Kotaro Ozasa, John D. Potter, Wei Zheng, Hongmei Nan. Association between educational level and mortality: A pooled analysis of over 694,000 individuals in the Asia Cohort Consortium [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 4234.
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2814-2814
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2814-2814
    Abstract: Tocopherols (T), which exist as α-, β-, γ-, and δ-T are important dietary antioxidants. Our previous studies have demonstrated that a mixture of tocopherols (γ-TmT), δ-T, and γ-T effectively inhibited colon carcinogenesis in AOM/DSS-treated mice or AOM-treated rats. However, the effects of different forms of tocopherols on carcinogenesis in animal models and in humans remain controversial. In the present study, we investigated the inhibitory effects of α-T, γ-T, and δ-T on colon carcinogenesis induced by a dietary carcinogen, 2-amino-1-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), and promoted by dextran sodium sulfate (DSS) in CYP1A-humanized (hCYP1A) mice. At 5-6 weeks age, male hCYP1A mice were fed the AIN93M diet or diet supplemented with 0.2% α-T, γ-T, or δ-T for 1 week before PhIP treatment (2x 100mg/kg b.w., 3 days apart), which was followed by DSS (1.5% in drinking water for 4 days) treatment. The mice were sacrificed 10 weeks after the first dose of PhIP. Significant inhibition of colon tumor incidences was caused by dietary δ-T and γ-T, but not by α-T. In addition, oxidative and nitrosative stress markers (i.e., 8-oxo-dG and nitrotyrosine) as well as pro-inflammatory stress markers (i.e., PGE2, LTB4, NFκB and p-STAT3) were significantly reduced by δ-T. Dietary treatment with δ-T and γ-T significantly increased the δ-T and γ-T levels, respectively, in the blood and tissues, but their levels were still lower than the levels of α-T. However, the levels of their side-chain degradation metabolites (δ-T and γ-T forms of carboxyethyl hydroxychroman and carboxymethylbutyl hydroxychroman) were much higher than their parent tocopherols in blood and tissues. Altogether, we demonstrated the inhibitory effects of δ-T and γ-T in a dietary carcinogen-induced colon carcinogenesis model. The inhibition is possibly due to the antioxidative, reactive nitrogen species-trapping, and anti-inflammatory activities δ-T and γ-T. (Supported by NIH grants F31CA168333, RO1CA120915 and RO1AT007036 as well as shared facilities funded by CA72720 and ES05022) Citation Format: Chung S. Yang, Jayson X. Chen, Anna B. Liu, Hong Wang, Marlon Lee, Siyuan Yu, Chunfang Zhao, Ying-Yi Kuo, Eric Chi, Nanjoo Suh. Inhibition of PhIP/DSS-induced colon carcinogenesis by different forms of tocopherols in CYP1A-humanized mice. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 2814. doi:10.1158/1538-7445.AM2015-2814
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4718-4718
    Abstract: Inositol polyphosphate 4-phosphatase type II (INPP4B) inhibits phosphatidylinositol 3-kinase (PI3K)/Akt signaling and is emerging as a tumor suppressor in a variety of tissues. Here we report that, conversely, it functions as an oncogenic regulator in human melanoma through activating PI3K/SGK3 signaling. While it was upregulated in a subset of melanomas, knockdown of INPP4B inhibited melanoma cell proliferation in vitro, and retarded melanoma growth in a xenograft model. In contrast, overexpression of INPP4B resulted in increased proliferation and anchorage-independent growth of melanocytes. Strikingly, INPP4B did not impinge on activation of Akt in melanocytic cells. Instead, it promoted PI3K/SGK3 signaling, in that INPP4B knockdown inhibited, whereas overexpression of INPP4B enhanced, activation of SGK3. Indeed, the effect of INPP4B on melanocytic cell proliferation was due to enhanced activation of SGK3, as co-introduction of an active form of SGK3 rescued melanocytes and melanoma cells from inhibition of proliferation triggered by INPP4B knockdown, and knockdown of SGK3 abolished enhancement in cell proliferation resulting from INPP4B overexpression. Upregulation of INPP4B appeared largely due to downregulation of microRNA-494 (miR-494) and/or miR-599 as a result of gene copy number reduction in melanoma cells. Collectively, these results reveal that INPP4B upregulation mediated by loss of miR-494 and/or miR-599 promotes melanoma cell proliferation through activation of PI3K/SGK3 signaling, and suggest that the role of INPP4B in the pathogenesis of cancers of different origins needs to be defined discretely. Citation Format: Chen Chen Jiang, Meng Na Chi, Su Tang Guo, James S. Wilmott, Xiang Yun Guo, Xu Guang Yan, Chun Yan Wang, Xiao Ying Liu, Lei Jin, Hsin-Yi Tseng, Amanda Croft, Hubert Hondermarck, Tao Liu, Richard A. Scolyer, Xu Dong Zhang. Inositol polyphosphate 4-phosphatase II activates PI3K/SGK3 signaling to promote proliferation of human melanoma cells. [abstract]. In: Proceedings of the 106th Annual Meeting of the American Association for Cancer Research; 2015 Apr 18-22; Philadelphia, PA. Philadelphia (PA): AACR; Cancer Res 2015;75(15 Suppl):Abstract nr 4718. doi:10.1158/1538-7445.AM2015-4718
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13 ( 2017-07-01), p. 3479-3490
    Abstract: Glioblastoma (GBM) stem-like cells (GSC) promote tumor initiation, progression, and therapeutic resistance. Here, we show how GSCs can be targeted by the FDA-approved drug mibefradil, which inhibits the T-type calcium channel Cav3.2. This calcium channel was highly expressed in human GBM specimens and enriched in GSCs. Analyses of the The Cancer Genome Atlas and REMBRANDT databases confirmed upregulation of Cav3.2 in a subset of tumors and showed that overexpression associated with worse prognosis. Mibefradil treatment or RNAi-mediated attenuation of Cav3.2 was sufficient to inhibit the growth, survival, and stemness of GSCs and also sensitized them to temozolomide chemotherapy. Proteomic and transcriptomic analyses revealed that Cav3.2 inhibition altered cancer signaling pathways and gene transcription. Cav3.2 inhibition suppressed GSC growth in part by inhibiting prosurvival AKT/mTOR pathways and stimulating proapoptotic survivin and BAX pathways. Furthermore, Cav3.2 inhibition decreased expression of oncogenes (PDGFA, PDGFB, and TGFB1) and increased expression of tumor suppressor genes (TNFRSF14 and HSD17B14). Oral administration of mibefradil inhibited growth of GSC-derived GBM murine xenografts, prolonged host survival, and sensitized tumors to temozolomide treatment. Our results offer a comprehensive characterization of Cav3.2 in GBM tumors and GSCs and provide a preclinical proof of concept for repurposing mibefradil as a mechanism-based treatment strategy for GBM. Cancer Res; 77(13); 3479–90. ©2017 AACR.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...