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)  (34)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 12 ( 2006-06-15), p. 6327-6335
    Abstract: Transforming growth factor βs (TGF-β) play a dual role in carcinogenesis, functioning as tumor suppressors early in the process, and then switching to act as prometastatic factors in late-stage disease. We have previously shown that high molecular weight TGF-β antagonists can suppress metastasis without the predicted toxicities. To address the underlying mechanisms, we have used the 4T1 syngeneic mouse model of metastatic breast cancer. Treatment of mice with a monoclonal anti-TGF-β antibody (1D11) significantly suppressed metastasis of 4T1 cells to the lungs. When metastatic 4T1 cells were recovered from lungs of 1D11-treated and control mice, the most differentially expressed gene was found to be bone sialoprotein (Bsp). Immunostaining confirmed the loss of Bsp protein in 1D11-treated lung metastases, and TGF-β was shown to regulate and correlate with Bsp expression in vitro. Functionally, knockdown of Bsp in 4T1 cells reduced the ability of TGF-β to induce local collagen degradation and invasion in vitro, and treatment with recombinant Bsp protected 4T1 cells from complement-mediated lysis. Finally, suppression of Bsp in 4T1 cells reduced metastasis in vivo. We conclude that Bsp is a plausible mediator of at least some of the tumor cell–targeted prometastatic activity of TGF-β in this model and that Bsp expression in metastases can be successfully suppressed by systemic treatment with anti-TGF-β antibodies. (Cancer Res 2006; 66(12): 6327-35)
    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 ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 66, No. 14 ( 2006-07-15), p. 7176-7184
    Abstract: Dysadherin, a cancer-associated membrane glycoprotein, down-regulates E-cadherin and promotes cancer metastasis. This study examined the role of dysadherin in breast cancer progression. Expression of dysadherin was found to be highest in breast cancer cell lines and tumors that lacked the estrogen receptor (ER). Knockdown of dysadherin caused increased association of E-cadherin with the actin cytoskeleton in breast cancer cell lines that expressed E-cadherin. However, knockdown of dysadherin could still suppress cell invasiveness in cells that had no functional E-cadherin, suggesting the existence of a novel mechanism of action. Global gene expression analysis identified chemokine (C-C motif) ligand 2 (CCL2) as the transcript most affected by dysadherin knockdown in MDA-MB-231 cells, and dysadherin was shown to regulate CCL2 expression in part through activation of the nuclear factor-κB pathway. The ability of dysadherin to promote tumor cell invasion in vitro was dependent on the establishment of a CCL2 autocrine loop, and CCL2 secreted by dysadherin-positive tumor cells also promoted endothelial cell migration in a paracrine fashion. Finally, experimental suppression of CCL2 in MDA-MB-231 cells reduced their ability to metastasize in vivo. This study shows that dysadherin has prometastatic effects that are independent of E-cadherin expression and that CCL2 could play an important role in mediating the prometastatic effect of dysadherin in ER-negative breast cancer. (Cancer Res 2006; 66(14): 7176-84)
    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. 5484-5484
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 5484-5484
    Abstract: TGF-βs are pleiotropic growth factors with complex roles in tumorigenesis. Overexpression of TGF-β1 in many advanced human tumors correlates with metastasis and poor prognosis, and TGF-β antagonists are being developed for cancer therapy. Using a panel of syngeneic mouse transplant models of metastatic breast cancer, we found that an anti-TGF-β neutralizing antibody suppresses metastasis in several models, including the widely-used 4T1 model. However, as expected from the complexity of TGF-β action, responses were heterogeneous and anti-TGF-β antibody treatment had no effect or even stimulated metastasis in other models. This panel of models provides a powerful platform to identify candidate biomarkers for their ability predict therapeutic response to TGF-β antibodies. Previously we have shown that therapeutic outcome does not correlate with the expression of TGF-β ligands, the growth inhibitory response of the tumor model to TGF-β in vitro, the level of Six-1 expression, or the activation of non-canonical TGF-β signaling through formation of mixed Smad complexes. We next performed extensive histopathologic and immunohistochemical characterization of the tumor models to look for features that might correlate with therapeutic response. We found no obvious correlations between therapeutic response and a variety of markers, including estrogen receptor status, cytokeratin expression profile (luminal vs basal differentiation), p63 expression (basal marker), p53 expression or mutation status, smooth muscle actin expression or desmoplasia. There was also no correlation of response with tumor cell proliferation as assessed by quantitation of mitotic figures and Ki67, tumor cell apoptosis, extent of necrosis or histomorphology of the tumor cells (spindled vs polygonal). We then focused on mechanistic analysis of the undesirable stimulatory effect of anti-TGF-β antibodies in the Mvt-1 model in an attempt to generate new leads for biomarker analysis. We showed that the stimulatory effect of anti-TGF-β antibody treatment in the Mvt-1 model is still seen in SCID mice, suggesting that it does not involve T-cell mediated immune responses. Comparison of treated and untreated Mvt-1 tumors showed no effect of anti-TGF-β antibodies on tumor cell apoptosis, or infiltration by F480+ or CD11b+ immune cells. Tumor cell proliferation was actually decreased by the antibody treatment. In vitro assays showed that TGF-β had similar stimulatory effects on migration and invasion in both the 4T1 and Mvt-1 models, suggesting that the stimulatory effect of antibody therapy in the Mvt-1 model could not be explained by an anomalous invasion response to TGF-β. Thus the mechanism by which anti-TGF-β antibodies can stimulate metastasis in this model are unclear. Transcriptomic analyses of treated and untreated tumors are ongoing and results will be presented. Citation Format: Yu-an Yang, Jia Weng, Michael Welsh, Nancy Guan, Josh Webster, Kathy Flanders, Scott M. Lonning, John McPherson, Lalage M. Wakefield. Heterogeneity of response to anti-TGF-β antibody therapy in preclinical models. [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 5484. doi:10.1158/1538-7445.AM2013-5484
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 67, No. 18 ( 2007-09-15), p. 8643-8652
    Abstract: The transforming growth factor-β (TGF-β) pathway has tumor-suppressor activity in many epithelial tissues. Because TGF-β is a potent inhibitor of epithelial cell proliferation, it has been widely assumed that this property underlies the tumor-suppressor effect. Here, we have used a xenograft model of breast cancer to show that endogenous TGF-β has the potential to suppress tumorigenesis through a novel mechanism, involving effects at two distinct levels in the hierarchy of cellular progeny that make up the epithelial component of the tumor. First, TGF-β reduces the size of the putative cancer stem or early progenitor cell population, and second it promotes differentiation of a more committed, but highly proliferative, progenitor cell population to an intrinsically less proliferative state. We further show that reduced expression of the type II TGF-β receptor correlates with loss of luminal differentiation in a clinical breast cancer cohort, suggesting that this mechanism may be clinically relevant. At a molecular level, the induction of differentiation by TGF-β involves down-regulation of Id1, and forced overexpression of Id1 can promote tumorigenesis despite persistence of the antiproliferative effect of TGF-β. These data suggest new roles for the TGF-β pathway in regulating tumor cell dynamics that are independent of direct effects on proliferation. [Cancer Res 2007;67(18):8643–52]
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2018
    In:  Cancer Research Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1768-1768
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 1768-1768
    Abstract: TGF-β family members are overexpressed in many advanced cancers and correlate with metastasis and poor prognosis. Based on encouraging preclinical data, therapeutics that target the TGF-β pathway are now in early phase clinical trials in oncology. While the three isoforms of TGF-β have essentially identical activities in vitro, there is relatively little known about how they might differ in vivo. Using a panel of twelve mouse allograft models of metastatic breast cancer, we showed that while TGF-β1 protein was consistently up-regulated in mammary tumors compared with normal mammary gland, the opposite was true for TGF-β3. Furthermore, in human breast cancer high TGF-β3 mRNA or protein expression was associated with better outcome, particularly in estrogen-receptor positive breast cancers. Collectively the data suggest that TGF-β1 and TGF-β3 may have opposing effects on breast cancer progression. Using an antibody that selectively neutralizes only TGF-β1 and TGF-β2, we explored the effect of sparing TGF-β3 on therapeutic outcome in the 4T1 and TSAE1 models of metastatic breast cancer. While the TGFβ1,2 antibody and two pan-TGF-β antibodies had similar efficacy against the metastasis endpoint in these very aggressive models, transcriptomic analysis of primary tumors after two weeks of antibody therapy suggested that sparing TGF-β3 might have positive effects on the metabolic profile of treated animals. To address this issue directly, we showed that mice without tumors had a significantly improved glucose tolerance following treatment with anti-TGF-β1/2 antibodies for 2-3 weeks when compared with mice treated with pan-TGF-β antibodies. Addressing potential human relevance, we showed that high expression of transcripts that were selectively upregulated in the primary tumors when TGF-β3 was spared correlated with good outcome in human breast cancer. The data suggest that use of isoform-selective TGF-β antagonists may offer advantages over the use of pan-TGF-β blocking agents for the treatment of breast cancer. Citation Format: Yu-an Yang, Srividya Vasu, Howard Yang, Maxwell P. Lee, Sushil Rane, Amer M. Mirza, Lalage M. Wakefield. An anti-TGF-β1/2 antibody that spares TGF-β3 retains full anti-tumor efficacy and generates an improved metabolic profile [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 1768.
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 4094-4094
    Abstract: Overexpression of transforming growth factor-βs (TGF-βs) correlates with metastasis and poor prognosis in many advanced cancers, and TGF-βs have pro-oncogenic effects on nearly every cell type in the ecosystem of advanced tumors. Based on these observations and encouraging results in preclinical models, strategies to block TGF-β signaling are in early phase clinical oncology trials. To date however, preclinical studies supporting the development of anti-TGF-β therapeutics in cancer have focused around a few well-characterized mouse models which do not capture the heterogeneity of the human disease. To assess the generalizability of these findings within a given tumor type, we have assembled a panel of transplantable mouse models of metastatic breast cancer. Using this panel of models, we tested the efficacy of a pan-TGF-β neutralizing antibody (1D11), against the metastatic endpoint using fully immunocompetent mouse hosts to capture the contribution of anti-tumor immune responses. We observed therapeutic efficacy or trend to efficacy in 5 models (InhibMet), no effect in 2 models (NoEff) and an undesirable stimulation or trend to stimulation of metastasis in 4 models (StimMet). This heterogeneity in therapeutic responses suggests that it will be critical to develop good predictive biomarkers for patient selection in clinical trials using TGF-β antagonists. Plausible candidate biomarkers suggested by the existing literature, such as p53 mutation status, claudin-low status, or TGF-β protein expression, did not correlate with therapeutic response, so we applied integrated genomic discovery approaches to the panel. We find significant differences in patterns of gene expression, gene polymorphism/mutation and copy number variation between the StimMet and InhibMet models. Notably, transcriptomic analyses of the untreated primary tumors show that these segregate by response to therapy in principal component analysis, after removal of mouse strain and tumor origin (spontaneous vs genetically-engineered) as factors. Analysis of differentially-expressed genes suggests that InhibMet models are characterized by higher TGF-β pathway activation, higher angiogenesis, poor immune cell infiltration/activation, and other markers of tumor aggressiveness such as higher tumor cell proliferation and survival. The data point to fundamental differences in tumor biology between the two classes of model and suggest that it will be possible to generate biomarkers that predict therapeutic response to TGF-β pathway antagonists. Citation Format: Yu-an Yang, Kathleen C. Flanders, Binwu Tang, Miriam R. Anver, Anand Merchant, Howard Yang, Maxwell Lee, Scott Lonning, John M. McPherson, Lalage M. Wakefield. Neutralizing anti-TGF-β antibodies elicit heterogeneous therapeutic responses in a panel of murine metastatic breast cancer models. [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 4094. doi:10.1158/1538-7445.AM2015-4094
    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. 68, No. 10 ( 2008-05-15), p. 3835-3843
    Abstract: Overexpression of transforming growth factor β (TGF-β) is frequently associated with metastasis and poor prognosis, and TGF-β antagonism has been shown to prevent metastasis in preclinical models with surprisingly little toxicity. Here, we have used the transplantable 4T1 model of metastatic breast cancer to address underlying mechanisms. We showed that efficacy of the anti–TGF-β antibody 1D11 in suppressing metastasis was dependent on a synergistic combination of effects on both the tumor parenchyma and microenvironment. The main outcome was a highly significant enhancement of the CD8+ T-cell–mediated antitumor immune response, but effects on the innate immune response and on angiogenesis also contributed to efficacy. Treatment with 1D11 increased infiltration of natural killer cells and T cells at the metastatic site, and enhanced expression of coactivators (NKG2D) and cytotoxic effectors (perforin and granzyme B) on CD8+ T cells. On the tumor cells, increased expression of an NKG2D ligand (Rae1γ) and of a death receptor (TNFRSF1A) contributed to enhanced immune cell-mediated recognition and lysis. The data suggest that elevated TGF-β expression in the tumor microenvironment modulates a complex web of intercellular interactions that aggregately promote metastasis and progression. TGF-β antibodies reverse this effect, and the absence of a major effect of TGF-β antagonism on any one cell compartment may be critical for a good therapeutic window and the avoidance of autoimmune complications. [Cancer Res 2008;68(10):3835–43]
    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 ...
  • 8
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Molecular Cancer Research Vol. 14, No. 2_Supplement ( 2016-02-01), p. B13-B13
    In: Molecular Cancer Research, American Association for Cancer Research (AACR), Vol. 14, No. 2_Supplement ( 2016-02-01), p. B13-B13
    Abstract: Overexpression of transforming growth factor-βs (TGF-βs) correlates with metastasis and poor prognosis in many advanced cancers, and TGF-βs have pro-oncogenic effects on nearly every cell type in the ecosystem of advanced tumors. Based on these observations and encouraging results in preclinical models, strategies to block TGF-β signaling are in early phase clinical oncology trials. To date however, preclinical studies supporting the development of anti-TGF-β therapeutics in cancer have focused around a few well-characterized mouse models which do not capture the heterogeneity of the human disease. To address this issue for breast cancer, we have assembled a panel of transplantable mouse models of metastatic mammary cancer that can be used in fully immunocompetent hosts, so as to preserve any immune contributions to therapeutic efficacy. Hormone receptor status, histopathological characteristics and intrinsic subtype of the models were assessed. Using this panel of models, we tested the efficacy of a pan-TGF-β neutralizing antibody (1D11) in inhibiting lung metastasis. We observed therapeutic efficacy or trend to efficacy in 5 models (InhibMet), no effect in 2 models (NoEff) and an undesirable stimulation or trend to stimulation of metastasis in 4 models (StimMet). This heterogeneity in therapeutic responses suggests that it will be critical to develop good predictive biomarkers for patient selection in clinical trials using TGF-β antagonists. Since the model panel provides an excellent platform for biomarker discovery, we have performed transcriptomics on untreated primary tumors across the panel, as well as completing full exome gDNA sequencing and copy number variant analysis on the tumor cell lines. The relationship between response-to-therapy and mutation load will be presented. Analyses of the untreated primary tumors show that the tumor transcriptomes segregate by response to anti-TGF-β therapy in principal component analysis, after removal of mouse strain as a confounding factor. Encouragingly for biomarker development, this observation suggests that there are major pre-existing differences in the biology of InhibMet and StimMet primary tumors before they are treated. A gene signature generated from the differentially expressed gene list was strongly associated with outcome in a metaanalysis of human breast cancer datasets, suggesting human relevance. Ingenuity Pathway Analysis of differentially-expressed genes indicates that InhibMet models are characterized by higher TGF-β pathway activation, higher angiogenesis, poor immune cell infiltration/activation, and other markers of tumor aggressiveness such as higher tumor cell proliferation and survival. Interestingly, the higher TGF-β pathway activation that was strongly predicted in the InhibMet tumors by the transcriptomic analyses (p=5.8e-25) was not evident from a multipronged quantitative proteomics assessment of activation of canonical Smad signaling, or non-canonical signaling through Akt, Erk, Jnk or p38MAPK pathways. Thus steady-state signaling by TGF-β in tumors in vivo may represent the integrated sum of the activities of more downstream signaling pathways than were captured in this analysis, or additional information about the intracellular localization of the phospho-forms of the signaling proteins and/or the identity of interacting proteins may be necessary to fully assess the activation state of the pathway. Despite these caveats, the data suggest that TGF-β antagonists will have therapeutic efficacy in more aggressive, poor prognosis breast cancers, and suggest that it will be possible to generate molecular signatures that predict the therapeutic response. Citation Format: Yu-an Yang, Kathleen Flanders, Jin-qui Chen, Anand S. Merchant, Howard Yang, Maxwell P. Lee, Lalage M. Wakefield. Targeting the TGF-β pathway in breast cancer: Insights from preclinical studies. [abstract]. In: Proceedings of the AACR Special Conference on Advances in Breast Cancer Research; Oct 17-20, 2015; Bellevue, WA. Philadelphia (PA): AACR; Mol Cancer Res 2016;14(2_Suppl):Abstract nr B13.
    Type of Medium: Online Resource
    ISSN: 1541-7786 , 1557-3125
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    detail.hit.zdb_id: 2097884-4
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 9
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3676-3676
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3676-3676
    Abstract: Breast cancer accounts for & gt;500,000 deaths annually worldwide. Despite therapeutic advances, treated patients often relapse with metastases. Emerging research indicates that relapse may be driven by a subpopulation of tumor cells termed cancer stem cells (CSCs), that are uniquely capable of self-renewal and have enhanced drug resistance. Understanding the properties of these CSCs is critical for development of more effective therapies. Peptidyl arginine deiminase 4 (PADI4) is an enzyme that catalyzes deimination of arginine to citrulline. Citrullination of histones modulates expression of key stem cell transcription factors in embryonic stem cells (Christophorou, et al., 2014, Nature 507:104), so we hypothesized that PADI4 could be involved in CSC regulation. We showed that PADI4 mRNA and protein are expressed at varying levels across a panel of breast cancer cell lines, and we knocked down PADI4 in two high expressing lines, MCF10Ca1h and MDAMB231-LM2. PADI4 knockdown did not alter tumor cell proliferation in vitro, but it enhanced migration and invasion and increased cell survival. Importantly, PADI4 knockdown increased clonogenicity and enhanced tumorsphere formation, suggesting that it specifically increases the CSC population. In vivo studies showed higher tumor initiation efficiency following PADI4 knockdown, and increased lung metastasis. PADI4 is predicted to have multiple isoforms with unknown biological activity that complicate PADI4 expression-based analysis in patient data sets. We circumvented this issue by pharmacologically inhibiting PADI4 in MCF10Ca1h to generate a transcriptomic signature reflecting PADI4 gene activity. This PADI4 gene activity signature was shown to correlate with lower tumor grade and better disease outcome in transcriptomic datasets from human ER+ breast cancers. In conclusion, our findings suggest that PADI4 functions as a tumor suppressor in breast cancer, in part through effects on the CSC. Citation Format: Humberto J. Ochoa, Nellie Moshkovich, Binwu Tang, Howard H. Yang, Maxwell P. Lee, Lalage M. Wakefield. Novel tumor suppressive role of Peptidylarginine deiminase IV involving cancer stem cell regulation in breast cancer models [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 3676.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 2244-2244
    Abstract: Breast cancer is a worldwide problem that accounts for almost a quarter of all cancers in women; however, better therapeutic approaches are required since it is estimated that one to three deaths from overtreatment occur for every one death avoided. Our lab is interested in transforming growth factor beta (TGF-β) signaling and its dual role as tumor suppressor/promoter in breast cancer and its therapeutic applications. We previously performed genome-wide ChIP/Chip analysis to identify TGF-β-activated Smad3 target genes in a model of breast cancer progression. We discovered that the estrogen-related-receptor β (ESRRB) transcription factor binding motif was significantly enriched in Smad3 binding regions in breast cancer cell lines. The data suggested that functional interactions between ESRRB and the TGF-β pathway may influence breast cancer progression. ESRRs (α, β, γ) are members of the nuclear orphan receptor family that share significant homology with the estrogen receptors but are not activated by natural estrogens. Additionally, ESRRB maintains pluripotency in embryonic stem cells (ESCs) and is activated by Wnt signaling to promote self-renewal in ESCs. Thus we hypothesized that mechanistically ESRRB/TGF-β may affect breast cancer progression through effects on cancer stem cell (CSC) dynamics and cancer cell differentiation. We showed that ESRRB protein is overexpressed in human breast cancer compared with matched normal breast tissue, and that high expression of ESRRB colocalizes with the stem cell master regulator OCT4 in human breast cancer xenografts. We performed ESRRB knockdown in the MCF10Ca1h, MCF10Ca1a and MDAMB231 breast cancer cell lines and our data demonstrate that ESRRB opposes the inhibitory effects of TGF-β on CSCs as measured by tumorsphere formation assay, while having little or no effect on proliferation of the bulk tumor cell culture in vitro. More importantly, knockdown of ESRRB reduced the in vivo tumorigenicity of all three breast cancer lines and enhanced their histologic differentiation. Extreme limiting dilution assays in vivo showed that knockdown of ESRRB in MDAMB231 cells caused a 78-fold decrease in the relative number of CSCs. In conclusion, our preliminary findings suggest that ESRRB antagonizes the inhibitory effects of TGF-β on CSCs and breast cancer progression, making ESRRB an attractive therapeutic target whose inhibition can restore the tumor suppressive effects of TGF-β and reduce the tumorigenic breast CSC population. Citation Format: Nellie Moshkovich, Misako Sato, Binwu Tang, Yu-an Yang, Kathleen C. Flanders, Mitsutaka Kadota, Howard Yang, Maxwell P. Lee, Lalage M. Wakefield. Functional interactions between estrogen-related-receptor β (ESRRB) and transforming growth factor-beta (TGF-β) in the regulation of breast cancer stem cell dynamics. [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 2244. doi:10.1158/1538-7445.AM2015-2244
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...