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: Journal of Cellular Biochemistry, Wiley, Vol. 53, No. 1 ( 1993-09), p. 51-60
    Type of Medium: Online Resource
    ISSN: 0730-2312 , 1097-4644
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 1993
    detail.hit.zdb_id: 1479976-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    Wiley ; 1999
    In:  Journal of Cellular Biochemistry Vol. 75, No. 4 ( 1999-12-15), p. 566-577
    In: Journal of Cellular Biochemistry, Wiley, Vol. 75, No. 4 ( 1999-12-15), p. 566-577
    Type of Medium: Online Resource
    ISSN: 0730-2312 , 1097-4644
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 1999
    detail.hit.zdb_id: 1479976-5
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Stem Cells, Oxford University Press (OUP), Vol. 34, No. 2 ( 2016-02-01), p. 504-509
    Abstract: Twist-1 encodes a basic helix-loop-helix transcription factor, known to contribute to mesodermal and skeletal tissue development. We have reported previously that Twist-1 maintains multipotent human bone marrow-derived mesenchymal stem/stromal cells (BMSC) in an immature state, enhances their life-span, and influences cell fate determination. In this study, human BMSC engineered to express high levels of Twist-1 were found to express elevated levels of the chemokine, CXCL12. Analysis of the CXCL12 proximal promoter using chromatin immunoprecipitation analysis identified several E-box DNA sites bound by Twist-1. Functional studies using a luciferase reporter construct showed that Twist-1 increased CXCL12 promoter activity in a dose dependent manner. Notably, Twist-1 over-expressing BMSC exhibited an enhanced capacity to maintain human CD34 + hematopoietic stem cells (HSC) in long-term culture-initiating cell (LTC-IC) assays. Moreover, the observed increase in HSC maintenance by Twist-1 over-expressing BMSC was blocked in the presence of the CXCL12 inhibitor, AMD3100. Supportive studies, using Twist-1 deficient heterozygous mice demonstrated a significant decrease in the frequency of stromal progenitors and increased numbers of osteoblasts within the bone. These observations correlated to a decreased incidence in the number of clonogenic stromal progenitors (colony forming unit–fibroblasts) and lower levels of CXCL12 in Twist-1 mutant mice. Furthermore, Twist-1 deficient murine stromal feeder layers, exhibited a significant decrease in CXCL12 levels and lower numbers of hematopoietic colonies in LTC-IC assays, compared with wild type controls. These findings demonstrate that Twist-1, which maintains BMSC at an immature state, endows them with an increased capacity for supporting hematopoiesis via direct activation of CXCL12 gene expression.
    Type of Medium: Online Resource
    ISSN: 1066-5099 , 1549-4918
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2016
    detail.hit.zdb_id: 2030643-X
    detail.hit.zdb_id: 1143556-2
    detail.hit.zdb_id: 605570-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Stem Cells, Oxford University Press (OUP), Vol. 30, No. 2 ( 2012-02-01), p. 314-325
    Abstract: Metastasis to multiple organs is the primary cause of mortality in breast cancer patients. The poor prognosis for patients with metastatic breast cancer and toxic side effects of currently available treatments necessitate the development of effective tumor-selective therapies. Neural stem cells (NSCs) possess inherent tumor tropic properties that enable them to overcome many obstacles of drug delivery that limit effective chemotherapy strategies for breast cancer. We report that increased NSC tropism to breast tumor cell lines is strongly correlated with the invasiveness of cancer cells. Interleukin 6 (IL-6) was identified as a major cytokine mediating NSC tropism to invasive breast cancer cells. We show for the first time in a preclinical mouse model of metastatic human breast cancer that NSCs preferentially target tumor metastases in multiple organs, including liver, lung, lymph nodes, and femur, versus the primary intramammary fat pad tumor. For proof-of-concept of stem cell-mediated breast cancer therapy, NSCs were genetically modified to secrete rabbit carboxylesterase (rCE), an enzyme that activates the CPT-11 prodrug to SN-38, a potent topoisomerase I inhibitor, to effect tumor-localized chemotherapy. In vitro data demonstrate that exposure of breast cancer cells to conditioned media from rCE-secreting NSCs (NSC.rCE) increased their sensitivity to CPT-11 by 200-fold. In vivo, treatment of tumor-bearing mice with NSC.rCE cells in combination with CPT-11 resulted in reduction of metastatic tumor burden in lung and lymph nodes. These data suggest that NSC-mediated enzyme/prodrug therapy may be more effective and less toxic than currently available chemotherapy strategies for breast cancer metastases. Disclosure of potential conflicts of interest is found at the end of this article.
    Type of Medium: Online Resource
    ISSN: 1066-5099 , 1549-4918
    Language: English
    Publisher: Oxford University Press (OUP)
    Publication Date: 2012
    detail.hit.zdb_id: 2030643-X
    detail.hit.zdb_id: 1143556-2
    detail.hit.zdb_id: 605570-9
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 13, No. 4 ( 2015-04-01), p. 636-650
    Abstract: Jumonji domain-containing protein 3 (JMJD3/KDM6B) demethylates lysine 27 on histone H3 (H3K27me3), a repressive epigenetic mark controlling chromatin organization and cellular senescence. To better understand the functional consequences of JMJD3 its expression was investigated in brain tumor cells. Querying patient expression profile databases confirmed JMJD3 overexpression in high-grade glioma. Immunochemical staining of two glioma cell lines, U251 and U87, indicated intrinsic differences in JMJD3 expression levels that were reflected in changes in cell phenotype and variations associated with cellular senescence, including senescence-associated β-galactosidase (SA-β-gal) activity and the senescence-associated secretory phenotype (SASP). Overexpressing wild-type JMJD3 (JMJD3wt) activated SASP-associated genes, enhanced SA-β-gal activity, and induced nuclear blebbing. Conversely, overexpression of a catalytically inactive dominant negative mutant JMJD3 (JMJD3mut) increased proliferation. In addition, a large number of transcripts were identified by RNA-seq as altered in JMJD3 overexpressing cells, including cancer- and inflammation-related transcripts as defined by Ingenuity Pathway Analysis. These results suggest that expression of the SASP in the context of cancer undermines normal tissue homeostasis and contributes to tumorigenesis and tumor progression. These studies are therapeutically relevant because inflammatory cytokines have been linked to homing of neural stem cells and other stem cells to tumor loci. Implications: This glioma study brings together actions of a normal epigenetic mechanism (JMJD3 activity) with dysfunctional activation of senescence-related processes, including secretion of SASP proinflammatory cytokines and stem cell tropism toward tumors. Mol Cancer Res; 13(4); 636–50. ©2015 AACR.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 2024-2024
    Abstract: Purpose: TWIST is a transcription factor critical to embryonic development, and aberrantly activated in many cancers. It regulates epithelial-to-mesenchymal transition (EMT), the process underlying metastatic spread and drug resistance. The majority of epithelial ovarian cancer (EOC) patients respond well to chemotherapy, but relapse with metastatic and drug-resistant disease. We are investigating the role of TWIST-mediated relapse, with the goal of developing treatments that sensitize chemoresistant tumors and improve survival. Experimental Procedures: We employ siRNA to target TWIST mRNA and have created a mesoporous silica nanoparticle with hyaluronic acid (siTWIST-MSN-HA) as a delivery vehicle. Microscopy was conducted to ensure cancer stem cell (CSC) targeting via CD44, and efficient uptake of MSN-HAs. We tested TWIST knockdown combined with cisplatin in vitro and in vivo. At necropsy, total tumor, metastasis, and ascites were evaluated to determine effects of siTWIST-MSN-HA compared to cisplatin alone. qPCR was conducted on tumors to examine effect of siRNA against TWIST, and its target genes. Mice were imaged via bioluminescence to observe CSC localization of siTWIST-MSN-HA compared to siTWIST-MSN (No HA). Tissue sections were stained via IHC to determine MSN-HA tumor targeting. CD44 staining was conducted to reveal HA co-localization in tumor. Results: siTWIST-MSN-HAs significantly knocked down TWIST, and sensitized cells to cisplatin compared to control in vitro. Following necropsy, tumor quantification revealed mice treated with siTWIST-MSN-HAs plus cisplatin exhibited a startling 75% loss of overall tumor burden (p=0.0012), 88% loss of total metastasis (p=0.001), and further 86% loss of total ascites (p=0.002) compared to cisplatin-alone treatment groups. EMT target expression by qPCR analysis revealed loss of TWIST1, vimentin, N-cadherin, and gain of E-cadherin in tumors treated with siTWIST-MSN-HA compared to controls. Bioluminescent images of mice at necropsy revealed highly specific tumor localization of MSN-HA Dylight (680nm) compared to MSN without HA. MSN-HA exhibited CSC targeting at metastatic sites, where most of the signal was emitted from the primary tumor; and negligible quantities of MSN-HAs were detected elsewhere. MSN-HA RITC (576nm) reveals highly specific tumor targeting via IHC; MSN-HA nanoparticles localized primarily in tumor cells and not in control organs. CD44 staining of these tissues reveals MSN-HAs co-localized with CD44 in CSCs. Conclusions: These studies demonstrate TWIST as a promising target for metastasis and acquired drug resistance in EOC. MSN-HAs provide CSC specific CD44 targeting with high efficacy and reduce tumor burden via siTWIST therapy. Thus, siTWIST-MSN-HAs provide a promising platform to improve survival from metastatic, drug-resistant ovarian cancer. Citation Format: Carlotta A. Glackin, Sophia A. Shahin, Shirleen Simargi, Ruining Wang, Altagracia Contreras, Liliana Parra, Loiuse Qu, Wei Wen, Thanh Dellinger, Julia Unternaehrer, Fuyujiko Tamanoi, Jeffrey Zink. Hyaluronic acid conjugated nanoparticle delivery of siTWIST reduces tumor burden and enhances chemosensitivity in ovarian cancer [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 2024.
    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 ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Clinical Cancer Research Vol. 22, No. 2_Supplement ( 2016-01-15), p. A42-A42
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 22, No. 2_Supplement ( 2016-01-15), p. A42-A42
    Abstract: Introduction: Epithelial ovarian cancer (EOC) is particularly deadly due to its fast rate of growth, dissemination, and chemoresistant recurrences. Novel therapies are urgently needed to limit metastasis and restore platinum sensitivity. We have investigated the role of TWIST1, a basic helix-loop-helix transcription factor, in these processes. TWIST1 is a well-documented regulator of metastasis from solid tumors, which has been linked to cancer stem cell phenotypes, drug resistance, and poor prognosis in several tumor types. Here we present in vitro and in vivo data supporting TWIST1's role in EOC growth and drug resistance, and propose a therapeutic modality to target TWIST1. Methodology: In vitro and in vivo studies: We created two cell lines, derived from the EOC line OVCAR8, that differ only in their TWIST1 expression. The cells were stably transfected with a G418-selectible vector containing either TWIST1 or an shRNA targeting TWIST1, under the control of the CMV viral promoter. Cells from each line were injected IP (intraperitoneally) into NSG mice, allowed to grow for 7 weeks, and tumor burden was evaluated at necropsy. In addition, RNA from each of these OVCAR8 cell lines (TWIST1 overexpressing versus knockdown) was isolated and used for RNA-seq analysis. Raw data was processed using the Galaxy online platform, and differentially expressed genes were further analyzed using Ingenuity Pathway Analysis. To determine the effect of TWIST1 on drug resistance, OVCAR8 cells with and without TWIST1 were treated with varying doses of cisplatin, and survival was quantified using colorimetric cytotoxicity assays. Therapeutic Application: Since we and others have shown that TWIST1's transactivation/protein binding (WR) domain is required for TWIST1 function, we produced a fusion protein comprised of GFP and the WR domain, and used CoIP to evaluate its ability to competitively inhibit TWIST1 binding in HEK 293 cells. Results: TWIST1 knockdown resulted in reduced tumor size and overall tumor burden in an NSG mouse model of EOC. Specifically, all mice receiving TWIST1-overexpressing cells developed primary ovarian tumors which were scored +4 on H & E by a veterinary pathologist, whereas only 50% of animals with TWIST1 knockdown developed ovarian tumors, and only 1 of 4 mice had a tumor that scored +4. In addition, fewer metastases were present in mice receiving TWIST knockdown cells, and no mice with TWIST1 knockdown tumors produced ascites. RNA-seq analysis of the TWIST overexpressing versus knockdown cell lines revealed marked differences in genes and pathways responsible for cell migration and cell survival, including upregulation of genes implicated in metastasis, DNA repair, and resistance to apoptosis in TWIST1 overexpressing cells. TWIST1 overexpressing cells also showed less cell death following cisplatin treatment. Finally, in our in vitro inhibitor tests, a GFP-WR domain fusion protein successfully reduced TWIST1 binding to its partner protein NFkB-p65 in HEK 293 cells. Conclusions: The importance of TWIST1 for tumor growth has been demonstrated for multiple tumor types – including ovarian cancer, as presented here. Our RNA-seq analysis reveals a complex network of TWIST1-mediated factors implicated in cell movement, survival, and resistance to DNA damaging agents, and we are working to determine the most important of these pathways in EOC. Most importantly, we have shown that TWIST1 expression in EOC cells leads to greater tumor burden in a mouse model, and by knocking down TWIST1, we were able to prevent peritoneal metastases and reduce tumor size. Therefore, we believe that TWIST1 is an attractive therapeutic target in EOC, owing to its roles at the crossroads of metastasis, drug resistance, and tumor growth. Citation Format: Cai M. Roberts, Michelle A. Tran, James Finlay, Sophia Allaf Shahin, Joana Loeza, Leslie Cisneros, Emily Ye, Thanh Dellinger, Carlotta A. Glackin. TWIST1 as a driver of accelerated tumor growth and drug resistance: Potential novel pathways and therapeutics. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Ovarian Cancer Research: Exploiting Vulnerabilities; Oct 17-20, 2015; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2016;22(2 Suppl):Abstract nr A42.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. LB-225-LB-225
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. LB-225-LB-225
    Abstract: Twist-1, a basic helix-loop-helix transcription factor, regulates embryogenesis and epithelial-mesenchymal transition (EMT). Twist-1 is essential for the differentiation and specification of mesoderm-derived tissues and hematopoietic stem cells. In hematopoiesis, Twist-1 plays an important role in maintaining the bone marrow niche to support normal hematopoiesis. In human tumorigenesis, Twist-1 is overexpressed and promotes EMT, a key process in the metastases and drug resistance. In addition, recent evidence indicates that Twist-1-Bmi1 axis confers acute myeloid leukemia (AML), cell self-renewal and apoptosis resistance which highlights the importance of the Twist1-Bmi1 axis in the regulation of self-renewal in both normal hematopoietic and leukemia stem cells (LSCs). The hypothesis of the current study states that dysregulation of Twist-1 is implicated in the pathogenesis of various hematological malignancies, AML. This study demonstrates the efficacy of nanoparticle delivery using hyaluronic-acid conjugated mesoporous silica nanoparticles (MSN-HA) as an intracellular delivery mechanism for siRNA designed to knock down Twist-1 (siTwist-1). The nanoparticle is designed for tumor stem cell specificity as its HA component exclusively targets LSCs through their native ligand, CD44. In addition, the nanoparticles' electrostatically bonded polyethylene (PEI) coat protects the siRNA from nuclease degradation. Using siRNA nanotechnology, previous studies demonstrated the ability to target and silence cytoplasmic Twist-1 mRNA and re-sensitize cancer cells to conventional chemotherapeutic agents in solid tumor models. In this study, we demonstrate the efficacy of this nanoparticle siRNA technology on the knockdown of Twist-1 in the AML cell line, KG1a, and the consequent cascading effect of upregulating the tumor suppressor genes and downregulating stemness genes. Our data indicate that Twist-1 promotes leukemia cell growth and colony-forming units (CFU) through the Twist/c-MPL axis, and knockdown of Twist1 inhibits tumor growth and reduce the number of CFUs. Our Results also showed that Twist-1 interacts with Runt domain of Runx-1, and c-MPL, reported to be negatively regulated by RUNx-1. Thus, nanotechnology which combines CD44 targeting and Twist Knockdown, has shown promising results for LSCs targeting, mitigating metastasis in AML. Further investigations of the role of Twist HSC and bone marrow niche will provide more insight into strategic management and treatment for leukemic stem cells. Citation Format: Ebtesam Nafie, Angelina Flores, Idoroenyi Amanam, Ruining Wang, Jeffrey Zink, Lucy Ghoda, Guido Marcucci, Carlotta Glackin. Twisting stemness in AML using nanoparticle as a delivery system for siRNA targeting Twist [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 LB-225.
    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 ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2006
    In:  Cancer Research Vol. 66, No. 7 ( 2006-04-01), p. 3365-3369
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 7 ( 2006-04-01), p. 3365-3369
    Abstract: The gain of N-cadherin expression in carcinomas has been shown to be important in the regulation of cell migration, invasion, and survival. Here, we show that N-cadherin mRNA expression in PC-3 prostate carcinoma cells is dependent on β1 integrin–mediated cell adhesion to fibronectin and the basic helix-loop-helix transcription factor Twist1. Depletion of Twist1 mRNA by small interfering RNA resulted in decreased expression of both Twist1 and N-cadherin and the inhibition of cell migration. Whereas Twist1 gene expression was independent of β1 integrin–mediated adhesion, Twist1 protein failed to accumulate in the nuclei of cells cultured in anchorage-independent conditions. The increased nuclear accumulation of Twist1 following cell attachment was suppressed by treatment with an inhibitor of Rho kinase or a β1 integrin neutralizing antibody. The effect of Twist1 on induction of N-cadherin mRNA required an E-box cis-element located within the first intron (+2,627) of the N-cadherin gene. These data raise the possibility that integrin-mediated adhesion to interstitial matrix proteins during metastasis differentially regulates the nuclear/cytoplasmic translocation and DNA binding of Twist1, activating N-cadherin transcription. (Cancer Res 2006; 66(7): 3365-9)
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2006
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2008
    In:  Cancer Research Vol. 68, No. 11 ( 2008-06-01), p. 4455-4464
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 68, No. 11 ( 2008-06-01), p. 4455-4464
    Abstract: Aromatase is the enzyme responsible for the last step of estrogen synthesis. The female hormone, estrogen, is known to stimulate breast cancer cell growth. Because the expression of aromatase in breast cancer tissues is driven by unique promoters I.3 and II, a more complete understanding of the regulatory mechanism of aromatase expression through promoters I.3/II in breast tumors should be valuable in developing targeted therapies, which selectively suppress estrogen production in breast tumor tissue. Results from in vivo footprinting analyses revealed several protein binding sites, numbered 1 to 5. When site 2 (−124/−112 bp, exon I.3 start site as +1) was mutated, promoters I.3/II activity was dramatically reduced, suggesting that site 2 is a positive regulatory element. Yeast one-hybrid screening revealed that a potential protein binding to site 2 was CCAAT/enhancer binding protein δ (C/EBPδ). C/EBPδ was shown to bind to site 2 of aromatase promoters I.3/II in vitro and in vivo. C/EBPδ up-regulated promoters I.3/II activity through this site and, as a result, it also up-regulated aromatase transcription and enzymatic activity. p65, a subunit of nuclear factor-κB (NF-κB) transcription factor, inhibited C/EBPδ–up-regulated aromatase promoters I.3/II and enzymatic activity. This inhibitory effect of p65 was mediated, in part, through prevention of the C/EBPδ binding to site 2. This C/EBPδ binding site in aromatase promoters I.3/II seems to act as a positive regulatory element in non–p65-overexpressing breast cancer epithelial cells, whereas it is possibly inactive in p65 overexpressing cancer epithelial cells, such as estrogen receptor–negative breast cancer cells. [Cancer Res 2008;68(11):4455–64]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2008
    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...