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)  (11)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2007
    In:  Cancer Research Vol. 67, No. 18 ( 2007-09-15), p. 8708-8715
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 67, No. 18 ( 2007-09-15), p. 8708-8715
    Abstract: Loss of SEMA3F occurs frequently in lung cancer and correlates with advanced stage of disease. We previously reported that SEMA3F blocked tumor formation by H157 lung cancer cells in a rat orthotopic model. This was associated with loss of activated αVβ3 integrin, impaired cell adhesion to extracellular matrix components, and down-regulation of phospho-extracellular signal-regulated kinase 1/2 (ERK1/2). These results suggested that SEMA3F might interfere with integrin outside-in signaling. In the present report, we found that SEMA3F decreased adhesion to vitronectin, whereas integrin-linked kinase (ILK) kinase activity was down-regulated in SEMA3F-expressing H157 cells. Exposure to SEMA3F-conditioned medium led to diminution of phospho-ERK1/2 in four of eight lung cancer cell lines, and ILK silencing by small interfering RNA led to similar loss of phospho-ERK1/2 in H157 cells. Moreover, SEMA3F expression (with constitutive and inducible systems) also reduced AKT and signal transducer and activator of transcription 3 (STAT3) phosphorylation independently of ILK-ERK1/2. These signaling changes extended downstream to hypoxia-inducible factor-1α (HIF-1α) protein and vascular endothelial growth factor (VEGF) mRNA levels, which were both reduced in three of four SEMA3F-transfected cell lines. Mechanistically, the effects on HIF-1α were consistent with inhibition of its AKT-driven protein translation initiation, with no effect on HIF-1α mRNA level or protein degradation. Furthermore, when H157 cells were injected s.c. in nude mice, tumors derived from SEMA3F-expressing cells showed lower microvessel density and tumor growth. These results show that SEMA3F negatively affects ILK-ERK1/2 and AKT-STAT3 signaling, along with inhibition of HIF-1α and VEGF. These changes would be anticipated to contribute significantly to the observed antitumor activity of SEMA3F. [Cancer Res 2007;67(18):8708–15]
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2007
    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. 66, No. 2 ( 2006-01-15), p. 944-950
    Abstract: The epidermal growth factor receptor (EGFR) is overexpressed in the majority of non–small cell lung cancers (NSCLC). EGFR tyrosine kinase inhibitors, such as gefitinib and erlotinib, produce 9% to 27% response rates in NSCLC patients. E-Cadherin, a calcium-dependent adhesion molecule, plays an important role in NSCLC prognosis and progression, and interacts with EGFR. The zinc finger transcriptional repressor, ZEB1, inhibits E-cadherin expression by recruiting histone deacetylases (HDAC). We identified a significant correlation between sensitivity to gefitinib and expression of E-cadherin, and ZEB1, suggesting their predictive value for responsiveness to EGFR-tyrosine kinase inhibitors. E-Cadherin transfection into a gefitinib-resistant line increased its sensitivity to gefitinib. Pretreating resistant cell lines with the HDAC inhibitor, MS-275, induced E-cadherin along with EGFR and led to a growth-inhibitory and apoptotic effect of gefitinib similar to that in gefitinib-sensitive NSCLC cell lines including those harboring EGFR mutations. Thus, combined HDAC inhibitor and gefitinib treatment represents a novel pharmacologic strategy for overcoming resistance to EGFR inhibitors in patients with lung cancer. (Cancer Res 2006; 66(2); 944-50)
    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 ...
  • 3
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Research Vol. 73, No. 8_Supplement ( 2013-04-15), p. 274-274
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 274-274
    Abstract: Neuropilins (NRP1 and NRP2) are high affinity receptors for the class-3 semaphorins, cell guidance molecules involved in tissue development, immune responses, angiogenesis and cancer. Semaphorins can inhibit proliferation, affect cell protrusion, spreading and adhesion, and trigger migratory responses often in a repulsive manner depending on the cellular context.However, neuropilins can also serve as receptors for galectin-1 and growth promoting factors including VEGF, PlGF, EGF and TGFβ. We previously cloned the SEMA3F gene from chromosome 3p21.3, which undergoes homozygous deletion and frequent loss of heterozygosity in lung cancer. We found that SEMA3F levels were inversely correlated with tumor aggressiveness in human lung cancer and confirmed its tumor suppressor activity in experimental xenograft models. We subsequently discovered that SEMA3F is directly downregulated by ZEB1, a transcription factor involved in the epithelial to mesenchymal transition (EMT). In the present study, we asked whether NRP2, the SEMA3F-specific receptor, was also regulated during the EMT process.TGFβ, a physiologic EMT inducer, rapidly increased NRP2 expression in NSCLC cell lines. Mechanistically, this increase resulted primarily from enhanced translation with some contribution from increased mRNA levels. However, TGFβ did not stabilize NRP2 mRNA or protein. Furthermore, we found out that a substantial amount of protein at the cell surface directly depended on the presence of TGFβ in the tumor microenvironment. The pathways regulating NRP2 induction include ERK1/2 and Akt but are Smad independent. In addition, forced expression of ZEB1 also induced NRP2. Conversely, ZEB1 inhibition reduced NRP2 upregulation by TGFβ. Importantly, inhibiting NRP2 affected TGFβ-induced morphologic changes, migration and ERK1/2 activation. Interestingly, some EMT target genes were also affected by NRP2 knockdown. Preliminary data in xenograft models indicate that NRP2 inhibition counteracts tumor growth-promoting effects of TGFβ .In a lung tumor tissue microarray, we observed that higher tumor grades were characterized by positive staining for NRP2 and negative staining for E-cadherin while lower tumor grades often presented the opposite profile. Since NRP2 is the highest affinity receptor for SEMA3F, our results suggest that loss of SEMA3F coupled with increased NRP2 would facilitate the binding of growth factors to NRP2 to further promote EMT and metastasis. Therefore, targeting NRP2 could be an important therapeutic approach against EMT in lung cancer. Citation Format: Patrick Nasarre, Joelle Roche, Vincent A. Potiron, Joyce Nair-Menon, Robert M. Gemmill, Harry A. Drabkin. Neuropilin-2 is upregulated during EMT in lung cancer. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 274. doi:10.1158/1538-7445.AM2013-274
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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 ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2012
    In:  Cancer Research Vol. 72, No. 8_Supplement ( 2012-04-15), p. 2416-2416
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 72, No. 8_Supplement ( 2012-04-15), p. 2416-2416
    Abstract: Neuropilins (NRP1 and NRP2) are high affinity receptors for the class-3 semaphorins, cell guidance molecules involved in tissue development, immune responses, angiogenesis and cancer. Semaphorins affect cell protrusion, spreading and adhesion, and can trigger migratory responses often in a repulsive manner depending on the cellular context. Interestingly, neuropilins are also receptors for galectin-1 and growth factors including VEGF, PlGF, EGF and TGFβ. We previously cloned the SEMA3F gene from chromosome 3p21.3, which undergoes homozygous deletion and frequent loss of heterozygosity in lung cancer. We found that SEMA3F levels were inversely correlated with tumor aggressiveness in human lung cancer and confirmed its tumor suppressor activity in experimental xenograft models. We subsequently discovered that SEMA3F is directly downregulated by ZEB1, a transcription factor involved in the epithelial to mesenchymal transition (EMT). In the present study, we asked whether SEMA3F specific receptor, NRP2, was also regulated during the EMT process. TGFβ, a physiologic EMT inducer, stimulated NRP2 expression in two NSCLC cell lines. Forced expression of ZEB1, but not Snail, also induced NRP2. Conversely, ZEB1 and Snail inhibition blocked NRP2 upregulation by TGFβ. Importantly, inhibiting NRP2 or ZEB1 expression reduced TGFβ-induced migration in an equivalent manner. In lung cancer tissue microarrays, NRP1 and NRP2 were preferentially expressed in the tumor compartment, whereas the EMT-related transcription factors, ZEB1 and Snail, were predominantly expressed in the stroma. Of note, NRP2 was also expressed in stromal cells and was significantly associated with both ZEB1 and a higher (worse) tumor grade. Since NRP2 is the highest affinity receptor for SEMA3F, our results suggest that loss of SEMA3F coupled with increased NRP2 would facilitate the binding of growth factors to NRP2 to further promote EMT and metastasis. Therefore, targeting NRP2 could be an important therapeutic approach against EMT in lung cancer. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 103rd Annual Meeting of the American Association for Cancer Research; 2012 Mar 31-Apr 4; Chicago, IL. Philadelphia (PA): AACR; Cancer Res 2012;72(8 Suppl):Abstract nr 2416. doi:1538-7445.AM2012-2416
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2012
    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. 73, No. 23 ( 2013-12-01), p. 7111-7121
    Abstract: The epithelial–mesenchymal transition (EMT) and its reversal, mesenchymal–epithelial transition (MET), are fundamental processes involved in tumor cell invasion and metastasis. SEMA3F is a secreted semaphorin and tumor suppressor downregulated by TGF-β1 and ZEB1-induced EMT. Here, we report that neuropilin (NRP)-2, the high-affinity receptor for SEMA3F and a coreceptor for certain growth factors, is upregulated during TGF-β1–driven EMT in lung cancer cells. Mechanistically, NRP2 upregulation was TβRI dependent and SMAD independent, occurring mainly at a posttranscriptional level involving increased association of mRNA with polyribosomes. Extracellular signal—regulated kinase (ERK) and AKT inhibition blocked NRP2 upregulation, whereas RNA interference-mediated attenuation of ZEB1 reduced steady-state NRP2 levels. In addition, NRP2 attenuation inhibited TGF-β1–driven morphologic transformation, migration/invasion, ERK activation, growth suppression, and changes in gene expression. In a mouse xenograft model of lung cancer, NRP2 attenuation also inhibited locally invasive features of the tumor and reversed TGF-β1–mediated growth inhibition. In support of these results, human lung cancer specimens with the highest NRP2 expression were predominantly E-cadherin negative. Furthermore, the presence of NRP2 staining strengthened the association of E-cadherin loss with high-grade tumors. Together, our results demonstrate that NRP2 contributes significantly to TGF-β1–induced EMT in lung cancer. Cancer Res; 73(23); 7111–21. ©2013 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5192-5192
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5192-5192
    Abstract: The epithelial to mesenchymal transition (EMT), which converts epithelial cells into an elongated, motile, and invasive phenotype, is considered to be a critical step in the dissemination of tumor cells. During EMT, the loss of E-cadherin (ECad) and upregulation of N-cadherin (NCad) or fibronectin (FN) have been most frequently described. FN binds integrin Alpha5 and is known for its upregulation in aggressive tumor cells. In addition, FN binds tissue transglutaminase (TGM-2) with high affinity (∼in nM) and plays a role in TGM-2 secretion. TGM-2, a multifunctional enzyme with transamidation or crosslinking, and GTP/ATP binding activities, is up-regulated in various malignant diseases, including lung cancer. In breast, cervical and ovarian cancers, TGM-2 has been shown to play a role in chemotherapy resistance and tumor metastasis during induction of EMT. TGM-2 overexpression results in the loss of epithelial features, up-regulation of mesenchymal markers, increased migration and invasion, and up-regulation of transcriptional repressors (e.g., ZEB1/2, Snail) known to mediate the EMT process. However, most of these studies were performed in aggressive tumor cells. In this study, we investigated the role of TGM-2 in TGFbeta-induced EMT process using NSCLC H358 cells. H358 cells express high levels of epithelial genes and are relatively resistant to TGFbeta induced EMT when compared to A549 cells. Particularly, H358 expresses barely detectable TGM-2 and FN. To investigate the role of TGM-2 during TGFbeta induced EMT, we constructed three isogenic cell lines with doxycycline (Dox)-inducible TGM-2 or C277A (an inactive mutant of TGM-2 with no transamidating activity) using InVitrogen's Flp-in TRex system. We found continuing treatment of 10 ng/ml TGFbeta for 6 days inducing H358 flp-in (vector control) cells to mescenchymal phenotype as demonstrated by the disappearance of Ecad, and the increased expression of NCad and FN. Under the same condition, Dox inducible TGM-2 and C277A facilitate the disappearance of Ecad, and Occludin and the appearance of Ncad. However, the expression of FN was undetectable in H358/TGM-2 and H358/C277A cells. As a result, H358/TGM-2 and H358/C277A remained localized and failed to migrate as demonstrated by a scratch wound healing assay. Seahorse mitochondria stress assay also demonstrated H358/TGM-2 and H358/C277A utilize more oxidative phosphorylation pathway than glycolytic pathway when compared with H358/flp-in, an indication of shifting cells to more normal phenotype. In summary, overexpression of TGM-2 (or inactive C277A mutant) inhibited the expression of FN and resulted in a less migratory H358 cells. Our results demonstrated that the role of TGM-2 during EMT in less aggressive epithelial cells such as H358 is different from its role in promoting EMT in other aggressive tumor cell; the suppression of FN might be a critical factor. Note: This abstract was not presented at the meeting. Citation Format: Thung S. Lai, Pin-Tsen Lin, Charles S. Greenberg, Harry A. Drabkin, Robert Gemmill. TGM-2 mediated downregulation of fibronectin during TGFb-induced epithelial-mesenchymal transition in lung cancer NCI-H358 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 5192. doi:10.1158/1538-7445.AM2015-5192
    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 ...
  • 7
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 2994-2994
    Abstract: Introduction: Neuropilin-2b (NRP2b) expression is associated with pro-tumorigenic properties of NSCLCs. We previously reported that NRP2b promotes metastasis and drug resistance, while the canonical NRP2a isoform is inhibitory. Our recent work indicates that the NRP2b cytoplasmic domain recruits GSK3β to phosphorylate and promote degradation of co-recruited PTEN, thereby enhancing AKT activity leading to enhanced survival, migration and drug resistance. We identified a 15-amino acid motif near the NRP2b C-terminus required for interaction with GSK3β. Herein, we describe the anticancer effect of peptides and drug-like macrocycles designed to disrupt this interaction. Methods: We used the cytoplasmic sequence of NRP2b for decoy peptides designed to disrupt the interaction with GSK3β. Control peptides were synthesized with alanine replacements for three amino acids suggested as crucial for GSK3β recruitment. All peptides were N-terminally myristoylated to promote association with and flipping to the inner leaflet of the plasma membrane. A library of over 42,000 drug-like macrocycles was screened in silico for compounds predicted to fit and compete with the NRP2b:GSK3β binding interface. Decoy peptides and macrocycles were assessed for anti-NRP2b activity using assays for migration and drug-tolerant persister cells. Transwell migration assays were performed using a Neuro Probe Reusable Multiwell Chemotaxis Chamber. Persister cell assays were performed with EGFR mutant PC9 and HCC827 cells in the presence of EGFR inhibitors (gefitinib or osimertinib), followed by drug withdrawal and colony formation assays. Results: As previously reported, knockdown of NRP2b inhibited, while knockdown of NRP2a enhanced, migration of lung cancer cell lines. Importantly, emergence of drug-tolerant persister cells was similarly inhibited by knockdown of NRP2b and enhanced by knockdown of NRP2a. Wild type, but not control, peptides abrogated the pro-tumorigenic effects of NRP2a knockdown on both migration and persister cell formation with IC50s of ~250 nM. The most effective macrocycles, M3 and M7, inhibited NRP2b-dependent migration with IC50s of 1-3 µM. Both macrocycles also inhibited generation of persister cell colonies from shNRP2a-PC-9 cells. However, only M3 was effective at blocking persisters from shNRP2a-HCC827 cells. Conclusions: NRP2b:GSK3β interaction is a therapeutic target in NSCLC affecting migration/invasion and persister colony formation associated with the emergence of resistance to EGFR inhibitors. This work justifies further development of these peptides and drug-like macrocycles as therapeutic modalities in lung cancer. Citation Format: Cecile Nasarre, Yuri K. Peterson, Patrick Nasarre, Anastasios Dimou, Kent E. Armeson, Harry A. Drabkin, Nancy Demore, Chadrick E. Denlinger, Robert M. Gemmill. Inhibition of the NRP2b:GSK3β binding interaction with peptides and macrocycles exerts anticancer effects in lung cancer [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 2994.
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Molecular Cancer Research Vol. 8, No. 1 ( 2010-01-01), p. 93-106
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 8, No. 1 ( 2010-01-01), p. 93-106
    Abstract: TRC8/RNF139 encodes an endoplasmic reticulum–resident E3 ubiquitin ligase that inhibits growth in a RING- and ubiquitylation-dependent manner. TRC8 also contains a predicted sterol-sensing domain. Here, we report that TRC8 protein levels are sterol responsive and that it binds and stimulates ubiquitylation of the endoplasmic reticulum anchor protein INSIG. Induction of TRC8 destabilized the precursor forms of the transcription factors SREBP-1 and SREBP-2. Loss of SREBP precursors was proteasome dependent, required a functional RING domain, occurred without generating processed nuclear forms, and suppressed SREBP target genes. TRC8 knockdown had opposite effects in sterol-deprived cells. In Drosophila, growth inhibition by DTrc8 was genetically suppressed by loss of specific Mprlp, Padlp N-terminal domain–containing proteins found in the COP9 signalosome and eIF3. DTrc8 genetically and physically interacted with two eIF3 subunits: eIF3f and eIF3h. Coimmunoprecipitation experiments confirmed these interactions in mammalian cells, and TRC8 overexpression suppressed polysome profiles. Moreover, high–molecular weight ubiquitylated proteins were observed in eIF3 immunoprecipitations from TRC8-overexpressing cells. Thus, TRC8 function may provide a regulatory link between the lipid and protein biosynthetic pathways. Mol Cancer Res; 8(1); 93–106
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 10 ( 2006-05-15), p. 5338-5345
    Abstract: Elevated tumor cyclooxygenase-2 (COX-2) expression is associated with tumor invasion, metastasis, and poor prognosis in non–small cell lung cancer (NSCLC). Here, we report that COX-2-dependent pathways contribute to the modulation of E-cadherin expression in NSCLC. First, whereas genetically modified COX-2-sense (COX-2-S) NSCLC cells expressed low E-cadherin and showed diminished capacity for cellular aggregation, genetic or pharmacologic inhibition of tumor COX-2 led to increased E-cadherin expression and resulted in augmented homotypic cellular aggregation among NSCLC cells in vitro. An inverse relationship between COX-2 and E-cadherin was shown in situ by double immunohistochemical staining of human lung adenocarcinoma tissue sections. Second, treatment of NSCLC cells with exogenous prostaglandin E2 (PGE2) significantly decreased the expression of E-cadherin, whereas treatment of COX-2-S cells with celecoxib (1 μmol/L) led to increased E-cadherin expression. Third, the transcriptional suppressors of E-cadherin, ZEB1 and Snail, were up-regulated in COX-2-S cells or PGE2-treated NSCLC cells but decreased in COX-2-antisense cells. PGE2 exposure led to enhanced ZEB1 and Snail binding at the chromatin level as determined by chromatin immunoprecipitation assays. Small interfering RNA–mediated knockdown of ZEB1 or Snail interrupted the capacity of PGE2 to down-regulate E-cadherin. Fourth, an inverse relationship between E-cadherin and ZEB1 and a direct relationship between COX-2 and ZEB1 were shown by immunohistochemical staining of human lung adenocarcinoma tissue sections. These findings indicate that PGE2, in autocrine or paracrine fashion, modulates transcriptional repressors of E-cadherin and thereby regulates COX-2-dependent E-cadherin expression in NSCLC. Thus, blocking PGE2 production or activity may contribute to both prevention and treatment of NSCLC. (Cancer Res 2006; 66(10): 5338-45)
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 735-735
    Abstract: We experimentally demonstrate the ability of Photonic Crystal microcavity-based biosensors to specifically detect the EMT transcription factor, ZEB1, in minute volumes of sample. Two-dimensional Photonic Crystals manufactured from silicon-on-insulator (SOI) have recently demonstrated the ability to confine and guide slow light on length scales of the wavelength of light. This has led to high sensitivity as well as miniaturization into compact sensors for chemical and bio-sensing. The precise wavelength of infrared light trapped by the resonance cavity of a Photonic Crystal is influenced in a very sensitive fashion by binding interactions between biomolecules attached to the silicon surface. In essence, the binding interaction alters the refractive index of the resonance cavity and this is detected as a shift in the resonance wavelength trapped by the device. When the sensor surface is derivatized with a specific antibody, the binding of the corresponding antigen will generate a resonance shift, revealing the presence of the antigen even when the antigen is present as a minor component in a complex whole-cell lysate. The resonance shift is detected directly and does not require any secondary labeling. The sensor cavity used here is dimensionally comparable to a mitochondrion, having a surface area of ∼5-10 μm2 which contacts approximately 13 nL of sample. The device was able to reliably detect ZEB1 binding in diluted samples of NCI-H358 non-small cell lung cancer cells containing ∼10 cells/μL. Specificity was demonstrated using a sandwich assay in which a second antibody was introduced following the initial binding and washing steps. The resonance wavelength was then super-shifted, but only when the second reagent applied was a different ZEB1-specific antibody that recognized a distinct epitope. These Photonic Crystal waveguides can be multiplexed on a single chip yielding a useful platform for highly sensitive and rapid detection of several biomolecules simultaneously in minute samples. Moreover, the sandwich assay principle can be applied to interrogate post-translational modifications of the captured proteins, so long as site-specific antibodies are available. The Photonic Crystal biosensor should be applicable to biomarker detection in minute biopsies, monitoring of pathway activation states in small samples, and detection of fusion or mutant proteins in hematological malignancies and solid tumors. Citation Format: Swapnajit Chakravarty, Wei-Cheng Lai, Yi Zou, Harry A. Drabkin, George R. Simon, Steve H. Chin, Ray T. Chen, Robert M. Gemmill. Photonic crystal microarray sensing of biomolecules: Detection of ZEB1 associated with the epithelial-mesenchymal transition in lung cancer cells. [abstract]. In: Proceedings of the 104th Annual Meeting of the American Association for Cancer Research; 2013 Apr 6-10; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2013;73(8 Suppl):Abstract nr 735. doi:10.1158/1538-7445.AM2013-735
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    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...