GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Association for Cancer Research (AACR)  (30)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (30)
Sprache
Erscheinungszeitraum
Fachgebiete(RVK)
  • 1
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Research Vol. 83, No. 7_Supplement ( 2023-04-04), p. 1451-1451
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 1451-1451
    Kurzfassung: Androgen deprivation therapy (ADT) is the mainstay treatment for non-metastatic and metastatic hormone-sensitive prostate cancer. Long-term ADT treatment results in adverse side-effects in patients including depression, memory loss, cognitive decline, and dementia. We aimed to identify critical molecules that could provide link between brain-related changes and pathological mechanisms responsible for cognitive decline in prostate cancer patients on ADT protocol. For these studies, gene expression data (GSE150807 and GSE151083) of human prostate cancer cells exposed to enzalutamide emulating ADT protocol was used to identify differential expression of genes at the transcript level. RNA sequencing analysis identified 9409 and 7757 differentially expressed genes (DEGs) in LNCaP and C4-2B enzalutamide exposed cells at p-value & lt; 0.0005 and FDR & lt; 0.05. In both cell lines, neuronal receptor signaling pathway was significantly overrepresented at 0.05 FDR-adjusted p-value (q-value). The genes associated with neuronal receptor signaling pathway belong to the ligand-gated ion channel and G-protein coupled receptors. Validation studies using brain glial cells treated with enzalutamide showed significant upregulation (p-value 0.001) of GABRB1, GABRA3, GABBR2 (GABA receptors/ligand-gated ion channel), BDNF (brain-derived neurotrophic factor), IFNB1 (interferon beta 1), NR3C1 (nuclear receptor subfamily 3 group c member/glucocorticoid receptor), ATGR1 (type-1 angiotensin II receptor), GRIN1 (glutamate ionotropic receptor NMDA type submit 1/ligated-gated ion channel), GRIN2D (glutamate ionotropic receptor NMDA type submit 2D/ligated-gated ion channel). In brain neuronal cells, enzalutamide exposure resulted in increased expression of NR3C1 whereas GABA/ligand-gated ion channel and others were downregulated and/or statistically non-significant. Further analysis using DisGenET (cytoscape) highlighted ATGR1, GABRB1, GRIN1, GRIN2D, and NR3C1 association with neuronal diseases including cognition disorder, forgetfulness, depressive mood, and others. The results of our study provide the first proof of evidence linking cognitive decline, depression, memory loss, and other neurological disorder more likely to develop in prostate cancer patients undergoing long-term androgen deprivation therapy. Citation Format: Shiv Shankar Verma, Eswar Shankar, Vaibhav Singh, Vijay Krishna, Sanjay Gupta. Cognition-mediated transcriptomic signature associated with androgen deprivation therapy in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 1451.
    Materialart: Online-Ressource
    ISSN: 1538-7445
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2023
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5168-5168
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5168-5168
    Kurzfassung: The majority of bladder cancers present with non-muscle invasive disease (NIMBC) confined to the superficial layers of the bladder wall, however, 50-70% of patients will subsequently progress to muscle-invasive bladder cancer (MIBC) that will require more aggressive treatment. For patient stratification, the current prognostication relies on tumor stage and grade, which remains challenging due to the lack of appropriate biomarker(s) to monitor disease progress. Therefore, there is an urgent need for predictive biomarkers that can distinguish between progressive versus non-progressive disease for the identification of patients to better stratify their risk of progression. Here we aim to identify novel prognostic biomarker(s) associated with disease progression by exploring bladder cancer patient gene expression database. We explored and analyzed the transcriptomic gene expression of bladder cancer patient datasets consisting of NIMBC and MIBC subtype from the NCBI GEO (GSE154261, GSE57813, and GSE37317) database. These datasets were analyzed using GEO2R (an R-based web application) and Limma R packages. Pathway enrichment analysis of differentially expressed genes (DEGs) between the NMIBC and MIBC group was analyzed using the ingenuity pathway analysis (IPA), metascape web-based portal, and Cytoscape. The above finding was functionally validated in human bladder cancer cell lines; RT4 (transitional cell papilloma), J82 (transitional cell carcinoma), HT1197 (bladder carcinoma), and 253JB-V (metastatic phenotype) and compared with the relative expression ofUROtsa (benign) urothelial cell line. A total of 1516 DEGs were identified between non-muscle-invasive and muscle-invasive bladder cancer specimens. To identify genes of prognostic value, we performed Gene Ontology (GO) and Kyoto Gene and Genomic encyclopedia (KEGG)analysis. A total of seven genes including CDKN2A, CDC20, CTSV, FOXM1, MAGEA6, KRT23, and S100A9 were confirmed with strong prognostic values in bladder cancer and validated by qRT-PCR conducted in various human bladder cancer cells representing stage-specific disease progression. ULCAN, human protein atlas, and The Cancer Genome Atlas datasets were used to confirm the predictive value of these genes in bladder cancer progression. Moreover, Kaplan-Meier analysis and Cox-hazard ratio analysis were performed to determine the prognostic role of these genes. Univariate analysis performed on a validation set identified a 3-panel gene set viz. CDKN2A, CTSV and FOXM1 with 95.5% sensitivity and 100% specificity in predicting bladder cancer progression. Our study screened and confirmed a panel of a 3-genes biomarkers that could precisely predict the progression and prognosis of bladder cancer. Citation Format: Shiv Shankar Verma, Eswar Shankar, Spencer Lin, Vaibhav Singh, E. Ricky Chan, Shufen Cao, Gregory T MacLennan, Lee E. Ponsky, Sanjay Gupta. Assessment and prognostic implication of 3-gene signature identified as potential biomarker in bladder cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5168.
    Materialart: Online-Ressource
    ISSN: 1538-7445
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2022
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1126-1126
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 1126-1126
    Kurzfassung: Prostate cancer is the second-leading cause of cancer-related mortality among men in the United States. Androgen receptor (AR) signaling is dominant survival pathway for prostate cancer cells, and androgen-deprivation therapy (ADT) through medical or surgical castration is the mainstay treatment for prostate cancer patients. Enzalutamide (Xtandi®), a second generation antiandrogen, is frequently prescribed for use in this clinical setting. Patient's response to this medication is usually temporary, and prolong treatment results in the emergence of drug resistance. A better understanding of molecular mechanism and its association with enzalutamide resistance will facilitate circumventing this problem. We employed next generation sequencing (NGS) and compared the transcriptomic profile of paired enzalutamide-sensitive and resistant human prostate cancer LNCaP and C4-2B cells for identification of genes involved in drug resistance along with in silico analysis. NGS data identified 9409 and 7757 genes differentially expressed in LNCaP and C4-2B cells compared to their parental counterparts. A subset of differentially expressed genes in both cell lines were validated at transcript level by qRT-PCR. Data analysis performed by IPA/i-pathway revealed metabolic signaling as predominantly overrepresented pathway and membrane transporters including solute carrier proteins, ATP-binding cassette transporters and other metabolizing enzymes as the most prominent genes dysregulated during metabolic reprogramming in both resistant cell lines. RNA-Seq data of resistant LNCaP cells demonstrate predominance of solute carrier genes, in particular, SLC12A5, SLC25A17 and SLC27A6, during metabolic reprogramming and in the development of drug resistance. Upregulation of these genes were associated with an increase in stemness; higher uptake of lactic/citric acid and lower glucose intake by the resistant cells. Our data suggest predominance of solute carrier genes (SLCs) during metabolic reprogramming of prostate cancer cells in an androgen-deprived environment. SLCs are important in cellular uptake of nutrients and drug absorption, thus could play a significant role in the emergence of drug-resistant phenotype, designating SLC genes as potentially attractive therapeutic targets. Citation Format: Shiv Shankar Verma, Eswar Shankar, Ricky Chan, Sanjay Gupta. Solute carrier (SLC) transporter-mediated metabolic reprogramming during enzalutamide resistance in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1126.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 4
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4097-4097
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 4097-4097
    Kurzfassung: Androgen deprivation therapy (ADT) continues to be the mainstay therapy for advance-stage prostate cancer. Although malignant cells respond to ADT initially, subsequently colonize and re-emerge as castration-resistant prostate cancer (CRPC). Enzalutamide (ENZU), a second-generation AR antagonist exhibits survival advantage in CRPC patients, however, ~30% of patients develop resistance after ENZU treatment, activating AR in these tumors. These ENZU-resistant tumors poorly respond to chemotherapy and other treatment modalities. Therefore, identification of an effective low-cost therapeutic alternative with fewer side effects may lead to increased survival and greatly benefit the quality-of-life of these patients. Activation of aerobic glycolysis and biomass production is implicated, in part, to be responsible for CRPC cells to acquire androgen resistance. Previous studies from our laboratory have demonstrated that synergistic combination of simvastatin (SIM), a drug for the treatment of hypercholesterolemia and metformin (MET), a glucose-lowering drug inhibits CRPC growth, invasiveness and migration with minimal effect on normal prostate epithelial cells (Mol Cancer Ther. 13(10):2288-2302, 2014). Here we examine whether the combination of SIM and MET could be effective in the treatment of ENZU-resistant prostate cancer cells. Human CRPC cells viz. C4-2B and its variant C4-2B-ENZU (C4-2B enzalutamide resistant) and 22Rv1 were used for the experiments. C4-2B-ENZU cells were generated by growing C4-2B cells in 20µM ENZU over 6 months and maintained in 5µM ENZU in the cell culture medium. 22Rv1 cells were inherently resistant to ENZU treatment. Both cells were treated with SIM (4µM) and MET (2mM) individually and in combination, followed by the assessment of cell viability, crystal violet assay, cell cycle analysis, migration, invasion, and expression of various target genes by Western blotting. C4-2B-ENZU and 22Rv1 cells were treated with a combination of SIM and MET at pharmacological dose range (500nM-4µM SIM and 250µM-2mM MET). Combination treatment with 4μM SIM and 2mM MET (SIM+MET) led to significant and synergistic inhibition of cell viability, migration, invasion and cell cycle blockade in both cancer cell lines. The individual treatments of SIM and MET exhibited little or no effect on these cells. Furthermore, SIM+MET combination decreased the expression of AR, AR-V7, p-Akt (Ser473), p-AMPKα1/α2 (Ser-485/491) and simultaneously increased p-AMPKα1 (Thr-172) and AMPKα kinase activity in these cells. We demonstrate, for the first time, that synergistic combination of SIM and MET may be an effective regimen for treatment ENZU-resistant prostate cancer cells. Citation Format: Eswar Shankar, Pingfu Fu, Gregory T. MacLennan, Sanjay Gupta. Enhanced synergistic efficacy of simvastatin and metformin on enzalutamide resistant prostate cancer cells [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 4097.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2020
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 5
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1914-1914
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 1914-1914
    Kurzfassung: Glioblastoma (GBM) is the most common and lethal form of intracranial tumor. Using a Cre-inducible lentiviral GBM mouse model we recently showed that gliomas can originate from dedifferentiation of mature neurons and astrocytes. Expression analysis of the known markers (of the differentiated cell-types) showed diminished expression while the expression of the undifferentiated state markers were significantly up-regulated in the dedifferentiated transformed neurons and astrocytes. In this study, we performed whole transcriptome analysis of these cells along with mouse pluripotent embryonic stem cells (mESC), neural stem cells (NSC), neurons and astrocytes to characterize the undifferentiated state of these cells. Our analysis revealed that dedifferentiated cell-types shared traits with neurons and NSCs at the global transcriptome level suggesting that they have retained memory of their cell-of-origin and share markers of undifferentiated cells with NSCs. Functional analysis of the transcriptomics data revealed involvement of the Wnt signaling, cell cycle and the focal adhesion pathways in defining the state of the dedifferentiated cell-types. Our analysis further revealed conservation of a gene interaction network in both dedifferentiated cell-types. This network exhibited a modular architecture, connecting components of the cell cycle pathway to Wnt signaling and the focal adhesion pathways. One of these components, Osteopontin (OPN), also known as Spp1, is highly expressed and secreted by the tumors and dedifferentiated transformed neurons and astrocytes. Attempts to inhibit the action of OPN, either by using a neutralizing antibody or silencing the OPN gene (shRNA), blocked the formation of tumorspheres and diminished their proliferating capacity. We are currently assessing the effects of the inhibition of OPN function in vivo. Further genetic perturbation of additional interacting partners and/or the abolishment of the interactions can help elucidate the regulatory mechanism of this network in maintaining the dedifferentiated state of the transformed neurons and astrocytes. Citation Format: Dinorah Friedmann-Morvinski, Vipul Bhargava, Shakti Gupta, Inder M. Verma, Shankar Subramaniam. Functional characterization of oncogenic-induced dedifferentiation in neurons and astrocytes using DP-seq. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 1914. doi:10.1158/1538-7445.AM2014-1914
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2014
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 5_Supplement ( 2023-03-01), p. P2-16-05-P2-16-05
    Kurzfassung: Objective: Triple Negative Breast Cancers (TNBC) are highly invasive and 46% of patients develop distant metastases, leading to higher mortality. Chemotherapy remains the most efficacious option, however, there is still a largely unmet need to identify novel therapeutic targets in TNBC to increase treatment options and improve patient outcomes, survival. Enhancer of zeste homologue 2 (EZH2), a member of the catalytic subunit of the polycomb repressive complex 2, is a histone methyltransferase and its canonical function is to methylate lysine 27 of histone 3. EZH2 is a potential driver of TNBC metastasis, and its high expression strongly associates with the TNBC phenotype as compared with other molecular subtypes of breast cancer. Despite the advancement in the discovery of inhibitors for EZH2 that attenuate its catalytic activity. Currently, several EZH2 inhibitors are under development and undergoing clinical trials and these compounds have been proved to be effective in the treatment of hematological malignancies, sarcomas and malignant rhabdoid tumors. However these inhibitors do not affect the intrinsic protein stability of EZH2, but typically competes with the cofactor S-adenosylmethionine (SAM) and binds to the SET domain of EZH2. Hence, the EZH2 inhibitors are only effective for some malignant blood tumors, and have poor efficacy for solid tumors, such as TNBC. Several studies have shown the involvement of neurotransmitter dopamine in proliferation, apoptosis, tumor angiogenesis, and drug resistance among different cancers, including the breast1. Dopamine D1 receptor activation in TNBC cell line induces apoptosis, autophagy and phosphorylation of eukaryotic translation initiation factor 2-alpha (eIF2a)2. Also, D1R agonists inhibits the invasion of breast cancer cell lines MDA-MB-231 and BT-20 and regress mammary tumors3. The oncogenic nature of EZH2 in driving aggressiveness in TNBC cells led us to simultaneously target dopamine D1 receptor and EZH2 to completely ablate tumor growth and metastasis. Methods: Schrodinger protein modeling software was employed for docking studies. TNBC cells MDA-MB-231 cells were treated with the EZH2 inhibitor GSK126 and/or D1R agonists A77636 and SKF38393 for assessing cell viability, migration. Immuno precipitation to investigate if the combination could disrupt the PRC2 complex in TNBC cells. To model tumor burden and the effects of combination therapy on tumor growth in vitro, we tested the combinatory effect of GSK126 and D1R agonists in a 3D culture system of MDA-MB-231 cells encapsulated in calcium-alginate microgels seeded from a microfluidic droplet generator. Cell death was determined by Yo-pro-Propidium Iodide staining. Result: In silico analysis confirmed that D1R agonists exhibited strong stabilization of protein structures upon binding to the EZH2 catalytic active site, albeit with slightly weaker affinity than GSK126. Combination treatment with GSK126 and dopamine agonists A77636 or SKF 38393 led to significant and synergistic inhibition of cell viability (p & lt; 0.01), migration, invasion, and EZH2 activity (p & lt; 0.01) in TNBC cells. In addition, MDA-MB-231 cells formed tumor spheroids that were subjected to the EZH2 inhibitor alongside dopamine agonists, indicating tumor reduction relative to single agent treatment in 3D culture (p & lt; 0.0001). The combination also led to increased necrotic cell death (p & lt; 0.05). Immunoprecipitation of EZH2 in the presence of GSK126 and SKF38393 and A77636 dissociated the physical interaction between EZH2, EED, and SUZ12 in MDA-MB-231 cells. Conclusion: Our data suggest that the combination of GSK126 and Dopamine D1 agonists synergistically inhibits TNBC proliferation by disrupting EZH2 functions leading to necrotic cell death. (This work is supported by DOD: W81XWH2010065, for Eswar Shankar). 1) PMID: 21531818 2) PMID: 29773888 3) PMID: 26477316 Citation Format: Eswar Shankar, Sridhar SNC, Xilal Y. Rima, Prem Kushwaha, Shiv Verma, Gautam K. Sarathy, Anagh kulkarni, Dharmaraja Allimuthu, Eduardo Reátegui, Sanjay Gupta, Bhuvaneswari Ramaswamy. Targeting EZH2 to overcome chemoresistance in Triple Negative Breast Cancers Employing Combinatorial Approach. [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr P2-16-05.
    Materialart: Online-Ressource
    ISSN: 1538-7445
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2023
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 7
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2021
    In:  Cancer Research Vol. 81, No. 13_Supplement ( 2021-07-01), p. 2594-2594
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 81, No. 13_Supplement ( 2021-07-01), p. 2594-2594
    Kurzfassung: Loss of tumor suppressors leads to acquisition of metastatic capability during prostate cancer progression leading to higher mortality. Maspin (SERPINB5) is a unique member of the serpin (serine protease inhibitor) family shown to regulate cell motility, migration and invasiveness. Loss of maspin is frequently identified in clinical prostate cancer specimens and prostate cancer cell lines, therefore novel therapeutic approaches to restore its expression are needed. We recently demonstrated that knockdown of class I HDACs increase maspin expression in prostate cancer cells [Mol Carcinog. 59(8):955-966, 2020] . Apigenin (4', 5, 7-trihydroxyflavone), a plant flavone has shown to possess anticancer properties and alters pathways that regulate tumor cell invasion and metastasis, however, the molecular basis of these effects remains unclear. We investigated whether apigenin has ability to restore maspin expression and contribute to the inhibition of metastasis. Treatment of human prostate cancer LNCaP and 22Rv1 cells, both harboring wild type p53, with 1-40 µM apigenin for 72 h resulted in a dose- and time- dependent decrease in cell invasion and migration with concurrent increase in maspin expression in the nuclear compartment. Since, p53 activates maspin promoter by binding directly to p53 consensus-binding site, we studied the effect of apigenin in potentially restoring p53-mediated maspin levels. Exposure of LNCaP and 22Rv1 cells with 5-20 μM of apigenin resulted in dose-dependent increase in maspin expression and p53 activation through acetylation at the Lys305 residue. These effects were associated with the inhibition of class I HDAC levels. Furthermore, apigenin withdrawal resulted in the loss of maspin expression and p53 acetylation in LNCaP cells. The increased apigenin-mediated p53 acetylation enhanced its binding on maspin promoter, which was associated with decrease in cell invasion and migration. Apigenin treatment also caused accumulation of acetylated histone H3 in total cellular chromatin, increasing accessibility to bind with the promoter sequences of maspin, consistent with the effects elicited by HDAC inhibitor, Tricostatin A. Similar observations of inhibition of class I HDACs and increase in p53 transcriptional activity were noted after feeding apigenin at 20- and 50- µg/day to 22Rv1 tumor xenograft implanted in athymic nude mice. Our results demonstrate that apigenin-mediated increase in maspin expression together with downregulation of class I HDACs, increases p53 activation resulting in decreased invasiveness and migration capabilities in prostate cancer. Citation Format: Eswar Shankar, Albert Lee, Rajnee Kanwal, Sanjay Gupta. Reactivation of maspin by plant flavone apigenin through inhibition of class I HDACs and increase in p53 transcriptional activity in prostate cancer cells [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 2594.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2021
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 8
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5358-5358
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. 5358-5358
    Kurzfassung: Enzalutamide (XTANDI®), an antiandrogen, is used for the treatment of advance-stage prostatecancer. Approximately, 60% of patients receiving enzalutamide show initial remission followedby disease relapse with the emergence of highly aggressive castration-resistant prostate cancer.Solute carrier (SLC) transporters play a critical role in cancer drug resistance by altering cellularmetabolism. We have recently reported the predominance of SLC25A17 and SLC27A6 duringenzalutamide resistance (Cells 9(12):2535, 2020) by generating enzalutamide resistant (C4-2B-Enzu) cells exposing human prostate cancer C4-2B cells in the presence of increasingconcentration of enzalutamide (1-20 µM) and maintained in the same condition for six months.However, the role of these molecules during metabolic reprogramming have not beenelucidated. Knockdown of SLC25A17 and SLC27A6 gene by sgRNA transfection suppressedcell proliferation, and migration in C4-2B-Enzu cells. Analysis of cell cycle showed that in theabsence of SLC25A17 and SLC27A6 there is induction of G1/S cell cycle arrest and reductionin the protein expression of checkpoint factors viz. Cyclin D1, CDK6, CDK4, and CDK2.SLC25A17 and SLC27A6 knockdown resulted in downregulation of acetyl-CoA carboxylase(ACC), fatty acid synthase (FASN) and Acyl-CoA synthetase long chain family member 5(ACSL5) with simultaneous decrease in the levels of lactic acid, triglyceride and citric acid in C4-2B-Enzu cells. In addition, SLC25A17 knockdown resulted in marked increase in apoptosis asevident by increase in protein expression of RIPK3 and BAX, compared to SLC27A6 knockdownin C4-2B-Enzu cells. Taken together, our results demonstrate that solute carriers SLC25A17and SLC27A6 play an important role in the development of enzalutamide resistance throughmetabolic reprogramming and loss of these molecules results in cell death. Thus, SLC25A17and SLC27A6 may serve as a therapeutic target for enzalutamide resistant prostate cancer. Citation Format: Prem P. Kushwaha, Shiv S. Verma, Eswar Shankar, Spencer Lin, Sanjay Gupta. Involvement of solute carrier transporters SLC25A17 and SLC27A6 in metabolic reprograming of castration-resistant prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr 5358.
    Materialart: Online-Ressource
    ISSN: 1538-7445
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2022
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 9
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2017
    In:  Cancer Research Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2230-2230
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 13_Supplement ( 2017-07-01), p. 2230-2230
    Kurzfassung: The histone methyltransferase enhancer of zeste homolog 2 (EZH2) is the catalytic subunit of the polycomb repressive complex (PRC2) and is involved in chromatin remodeling and gene silencing through its methylation of histone H3 on lysine 27 (H3K27). Specifically, trimethylation of H3K27 is associated with gene silencing and plays critical role in cancer initiation and progression. Therefore, blocking EZH2 catalytic activity may present a valid preventive and/or therapeutic strategy for the treatment of cancers with EZH2 overexpression including prostate cancer. Our multistep approach of molecular modeling and direct binding has led to the identification of plant flavone luteolin (3’, 4’, 5, 7-tetrahydroxyflavone) as a specific inhibitor of EZH2 with preferential blocking of its catalytic site. Here we report luteolin, in micro molar range, inhibits EZH2 catalytic activity, demonstrates anti-proliferative and anti-invasive activities in functional cell based assays. Treatment of human prostate cancer DU145 and 22Rv1 cells, which possess high constitutive EZH2 expression, with 5-20 μM luteolin significantly inhibits EZH2 and SUZ12 protein expression in dose and time dependent manner, although luteolin treatment did not affect protein expression of EED and RbAp46/48 protein, other members of PRC2 complex. Treatment of both cell lines with luteolin also reduced H3K27me3 and H3K27me2 protein and its enzymatic activity in dose and time dependent manner without affecting total H3 protein. In addition, luteolin was also effective in suppressing in vitro methylation performed using recombinant PCR2 complex. These events led to increase in the expression of downstream tumor suppressor genes including E-cadherin, SLIT2 and TIMP3, respectively. Interestingly, treatment of cells with proteasome inhibitor, MG132 together with luteolin did not prevent EZH2 degradation indicating that proteasomal degradation might not contribute to EZH2 inhibition. Taken together, our study suggest that luteolin acts on the catalytic binding site of EZH2 to exhibit downregulation of histone H3 methylation and could be developed as a potential promising agent for the prevention and/or treatment of various human cancers including prostate cancer with EZH2 overexpression. Citation Format: Rajnee Kanwal, Xiaoping Yang, Eswar Shankar, Sanjay Gupta. Luteolin selectively inhibits EZH2 and blocks H3K27 methylation in prostate cancer cells [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 2230. doi:10.1158/1538-7445.AM2017-2230
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2017
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 10
    Online-Ressource
    Online-Ressource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 5084-5084
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 5084-5084
    Kurzfassung: Metastasis is responsible for more than 90% of prostate cancer-associated mortality in the United States. One of the distinctive reason for metastasis has been the imbalance of matrix metalloproteinases (MMPs) as a result of reduced expression of RECK (reversion-inducing-cysteine-rich protein with Kazal motifs) a novel tumor suppressor. RECK has been shown to be a potent inhibitor of MMP-2 and MMP-9. Loss of RECK has been frequently identified in clinical prostate cancer specimens and prostate cancer cell lines, therefore mechanistic understanding of its loss and approaches to restore its expression would facilitate inhibition of metastasis. Green tea polyphenols (GTP) and its major constituent, epigallocatechin-3-gallate (EGCG) has been shown to suppress prostate cancer metastasis, however the mechanism has not been fully elucidated. We determined whether treatment with GTP has ability to restore induction of RECK and play a key role in suppressing invasiveness in prostate cancer. Human prostate cancer LNCaP tumor were implanted in the ventral prostate of athymic nude mice for 2 weeks followed by per-oral intake of GTP at 7.5 and 15.0 mg/kg body weight freshly prepared in 100µl PBS. Other groups were treated with DNA methyltransferase inhibitor-5-aza-2'-deoxycytidine (AZA), histone deacetylase inhibitor-trichostatin A (TSA) and histone methyltransferase inhibitor-3-Deazaneplanocin A (DZNep) individually at 0.1 mg/kg body weight intraperitoneally at alternate days/week; and combination of AZA+TSA and DZNep+TSA at similar doses and times. Treatment of mice with GTP resulted in marked decrease in tumor growth and its local invasion in dose dependent manner, compared to treatment with epigenetic inhibitors and their combination after 8 weeks of intervention. GTP treatment significantly reduced serum levels of MMP-2, MMP-9 and VEGF, compared to treatment with epigenetic inhibitors alone. Combination of AZA+TSA exhibited similar effect which was equivalent to the lower dose of GTP treatment. Furthermore, GTP treatment significantly reduced EZH2 and H3K27me3 and class I HDAC protein levels in tumors. GTP partially reversed RECK hypermethylation and significantly enhanced its expression in the tumor tissue. Similar findings were noted in cell culture where treatment of PC-3 and LNCaP cells with 20µM EGCG and 10µg/mL GTP for 72 h significantly induced RECK expression at mRNA and protein levels along with decrease invasiveness in these cells. Our findings suggest that induction of RECK is a key epigenetic event reactivated by GTP/EGCG that results in suppression of MMP-2/MMP-9, matrix degradation and angiogenesis to delay prostate cancer invasion and its subsequent progression. Citation Format: Eswar Shankar, Omair Iqbal, Natarajan Bhaskaran, Gauri Deb, Gregory T. MacLennan, Pingfu Fu, Sanjay Gupta. Epigenetic modifications involving reactivation of RECK inhibiting MMP-9 and MMP-2 in prostate cancer [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 5084.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...