Your email was sent successfully. Check your inbox.

An error occurred while sending the email. Please try again.

Proceed reservation?

Export
  • 1
    In: Oncotarget, Impact Journals, LLC, Vol. 10, No. 32 ( 2019-04-30), p. 3051-3065
    Type of Medium: Online Resource
    ISSN: 1949-2553
    URL: Issue
    Language: English
    Publisher: Impact Journals, LLC
    Publication Date: 2019
    detail.hit.zdb_id: 2560162-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 2
    In: Endocrine-Related Cancer, Bioscientifica, Vol. 28, No. 7 ( 2021-07-01), p. 403-418
    Abstract: The patient’s hormonal context plays a crucial role in the outcome of cancer. However, the association between thyroid disease and breast cancer risk remains unclear. We evaluated the effect of thyroid status on breast cancer growth and dissemination in an immunocompetent mouse model. For this, hyperthyroid and hypothyroid Balb/c mice were orthotopically inoculated with triple-negative breast cancer 4T1 cells. Tumors from hyperthyroid mice showed an increased growth rate and an immunosuppressive tumor microenvironment, characterized by increased IL-10 levels and decreased percentage of activated cytotoxic T cells. On the other hand, delayed tumor growth in hypothyroid animals was associated with increased tumor infiltration of activated CD8 + cells and a high IFNγ/IL-10 ratio. Paradoxically, hypothyroid mice developed a higher number of lung metastasis than hyperthyroid animals. This was related to an increased secretion of tumor CCL2 and an immunosuppressive systemic environment, with increased proportion of regulatory T cells and IL-10 levels in spleens. A lower number of lung metastasis in hyperthyroid mice was related to the reduced presence of mesenchymal stem cells in tumors and metastatic sites. These animals also exhibited decreased percentages of regulatory T lymphocytes and myeloid-derived suppressor cells in spleens but increased activated CD8 + cells and the IFNγ/IL-10 ratio. Therefore, thyroid hormones modulate the cellular and cytokine content of the breast tumor microenvironment. A better understanding of the mechanisms involved in these effects could be a starting point for the discovery of new therapeutic targets for breast cancer.
    Type of Medium: Online Resource
    ISSN: 1351-0088 , 1479-6821
    Language: Unknown
    Publisher: Bioscientifica
    Publication Date: 2021
    detail.hit.zdb_id: 2010895-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 3
    In: Blood, American Society of Hematology, Vol. 130, No. Suppl_1 ( 2017-12-07), p. 732-732
    Abstract: Bexarotene (Bex) is an oral RXR agonist that is effective for the treatment of early and advanced-stage CTCL. However, Bex is associated with the unavoidable side effect of hypothyroidism in about 95% of pts, which is prophylactically managed with the administration of thyroid hormone (TH). Paradoxically, we found that physiological levels of TH increase the proliferation of CTCL by activating both the nuclear TRA receptor and the membrane integrin αVβ3 in these cells. Here, we determined the influence of TH replacement therapy on the anti-lymphoma activity of Bex, an unknown topic with clinical implications. Results: In standard culture conditions, Bex decreases the proliferation and viability of CTCL cell lines HuT78 and MJ. We conducted RNA-sequencing in HuT78 cells treated with Bex (vs. vehicle) to understand the mechanism/s underpinning these effects. We found that Bex regulates pathways related to "cell proliferation and differentiation" (e.g. REL, SCD, CCND1) and "immune system" (e.g. TBX21, IFNG, MX1) , which we independently validated. This suggests that Bex treatment could also impact anti-neoplastic immunity by increasing INF-gamma release from CTCL cells. We then conducted the same experiment culturing HuT78 and MJ in TH-depleted culture conditions and observed a higher effect of Bex in decreasing proliferation and viability; supporting the notion that Bex should not be administered with TH replacement. However, hypothyroidism is associated with marked immunosuppression that could favour CTCL progression and potentially affect the immune modulator effect of Bex that we described above. We thus determined the impact of TH replacement on the anti-lymphoma effect of Bex using a murine CTCL model. We implanted murine EL4 TCL cells in the hypodermis of immunocompetent C57BL/6 mice. Once tumors developed, mice were randomized into treatment with vehicle (Veh), Bex with no TH replacement (Bex) and Bex with TH replacement (BexT4+). TH replacement was done with oral administration of T4. We measured T4 in these mice plasma by RIA to assure that mice with T4 replacement reach physiological levels and those with no T4 develop hypothyroidism. Compared to Veh mice, the administration of Bex significantly decreased lymphoma growth in both conditions although slightly better in mice without T4 replacement (p & lt;0.001 and p & lt;0.01) for Bex and BexT4+, respectively). However, mice with Bex alone that developed hypothyroidism showed a significant decrease of CD8+CD44+ T-cells (p & lt;0.05 vs. BexT4+) and increase of myeloid-derived suppressor cells in the tumor microenvironment and draining lymph nodes. Since infiltration of CD8+ cells decreases with CTCL progression, our data indicates that Bex treatment should be administered with T4 replacement to avoid a negative immune microenvironment. Considering the cellular effects of TH are exerted through TRA and integrin αVβ3 that can be independently modulated pharmacologically, we investigated whether integrin αVβ3 inhibition would be sufficient to blunt the TH-induced decrease on the antineoplastic efficacy of Bex. The lack of expression of integrin αVβ3 in normal T-cells offers a rationale for a selective effect on CTCL cells while sparing immune cells. In HuT78 and MJ cell lines, either αVβ3 silencing by siRNAs or pharmacologic inhibition with the clinical drug cilengitide avoided the decrease in the anti-CTCL effect of Bex upon TH supplementation. Moreover, gene expression analysis (RNA-seq and PCR) demonstrated that αVβ3 silencing induced higher (vs. Bex) up-regulation of "immune " genes like TBX21 and INFG, or down-regulation of "proliferation" genes like REL and CCND1 . We thus tested if this mechanism can be therapeutically capitalized to improve Bex treatment in our CTCL murine model. We implanted 24 C57BL/6 mice with murine EL4 TCL cells and randomized them to vehicle (Veh), Bex with TH replacement in combination with cilengitide 40mg/kg (BexT4+Cil) or without cilengitide (BexT4+). At end of treatment, BexT4+Cil mice showed significantly smaller tumors compared with Veh and BexT4+ mice (p & lt;0.001 and p & lt;0.05, respectively). Importantly, the anti-tumoral immune response of CD8+CD44+ T-cells in the CTCL microenvironment and draining lymph nodes remained similar in BexT4+Cil vs. BexT4+ mice. Our data indicates that inhibition of the integrin αVβ3 is an effective strategy to improve Bex-based treatments in CTCL. Disclosures Cerchietti: Celgene: Research Funding; Lymphoma Research Foundation: Research Funding; Leukemia and Lymphoma Society: Research Funding; Weill Cornell Medicine - New York Presbyterian Hospital: Employment.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2017
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 4
    In: Pharmacological Research, Elsevier BV, Vol. 109 ( 2016-07), p. 55-63
    Type of Medium: Online Resource
    ISSN: 1043-6618
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2016
    detail.hit.zdb_id: 1471456-5
    SSG: 15,3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 5
    In: Frontiers in Endocrinology, Frontiers Media SA, Vol. 10 ( 2019-2-13)
    Type of Medium: Online Resource
    ISSN: 1664-2392
    Language: Unknown
    Publisher: Frontiers Media SA
    Publication Date: 2019
    detail.hit.zdb_id: 2592084-4
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 71, No. 8_Supplement ( 2011-04-15), p. 2285-2285
    Abstract: The signal transducer and activator of transcription (Stat) family of proteins was found to be involved in crosstalks with both steroid hormones and type I receptor tyrosine kinases (RTKs) signaling pathways. We have previously demonstrated that HRG, a ligand of RTKs, transactivates PR both in C4HD cells from an experimental model of hormonal carcinogenesis in which MPA induced mammary adenocarcinomas in Balb/c mice, and in the human breast cancer cell line T47D (Mol Cell Biol. 2003 Feb;23(3):1095-111). We have also shown that HRG induces Stat3 tyrosine phosphorylation and transcriptional activation by a mechanism that requires PR signaling (Mol Cell Biol. 2009 Mar;29(5):1249-65). In the present work we explored whether Stat3 acting as a coactivator, could modulate ligand-independent activation of PR by HRG. Assessment of the expression of the progestin-regulated gene bcl-X showed that HRG treatment of C4HD cells resulted in an increase in bcl-X protein levels. This effect was completely abolished when Stat3 expression was silenced using siRNAs. HRG treatment of cells transfected with a luciferase reporter plasmid under the control of the murine bcl-X promoter which contains two progesterone response elements (PREs), resulted in an increase in luciferase activity. HRG had no effect on PR transcriptional activation when cells were transfected with a mutant vector containing a deletion spanning both PREs in bcl-X promoter. We assessed the specific association of Stat3 and PR to the PRE region of bcl-X promoter in the context of living cells by performing Chromatin Immunoprecipitation (chIP) Assays. We found that HRG treatment of primary cultures of C4HD cells induced PR and Stat3 recruitment to the bcl-X promoter. HRG also induced PR and Stat3 occupancy of the stably integrated MMTV promoter in T47D-Cat0 breast cancer cells. By performing sequential chIP assays we showed that HRG induced simultaneous PR and Stat3 occupancy of the bcl-X promoter region. We explored Stat3 role in the non-classical mechanism of action of PR, where PR is recruited to p21 promoter indirectly through interaction with Sp1 transcription factor. Interestingly, when cells were treated with HRG, we detected Stat3 binding to the Sp1 binding sites of the PR-regulated p21 promoter, together with Sp1 and PR. These results provide the first evidence that Stat3 modulates ligand-independent activation of PR by HRG in breast cancer cells. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 102nd Annual Meeting of the American Association for Cancer Research; 2011 Apr 2-6; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2011;71(8 Suppl):Abstract nr 2285. doi:10.1158/1538-7445.AM2011-2285
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2011
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 7
    In: Blood, American Society of Hematology, Vol. 122, No. 21 ( 2013-11-15), p. 3810-3810
    Abstract: Cutaneous T-cell lymphomas (CTCL) are a heterogeneous group of T-cell non-Hodgkin lymphomas (T-NHL) derived from skin-homing mature T cells. Thyroid hormones (THs) are crucial regulators of cellular differentiation, growth and metabolism. We recently found that THs stimulate the proliferation and metabolism of T cells and that this pathway is co-opted by T-NHL cells to maintain their malignant phenotype. Accordingly, a decrease in TH levels affects the proliferation of T-NHL cells. We also found that THs modulate these functions by activating both canonical nuclear (TR) and membrane receptors (mTR, that for most cells is represented by integrin αvβ3). We now hypothesized that the characterization of the relative contribution of these receptors to the malignant phenotype of T-NHLs will allow for a more precise therapeutic intervention. The study presented here was conducted to determine (i) whether THs are pro-survival factors in CTCL and (ii) the relative contribution of TR and mTR to the effect of THs in CTCLs. To test this hypothesis, we first analyzed the expression of TR and mTR in CTLC cells (HuT 78, MJ and HuT102) vs. normal T cells (from peripheral blood) by qRT-PCR and immunoblotting. We found that CTCL expressed 2 to 10 fold more THRA, ITGAV and ITGB3 mRNA and protein than their normal counterpart (p 〈 0.05, T test). Moreover, in a tissue microarray of 52 CTCLs, the integrin avβ3 was overexpressed in 92% of the cases. To differentiate between nuclear vs. membrane-initiated effects, CTCL cells were treated with physiological concentrations of free and cell impermeable agarose-bound T3/T4 (TH-ag). After 24h of treatment, cell proliferation increased 20-38% in HuT-78 cells and 18-32% in MJ cells with both forms of THs (p 〈 0.05 for both comparisons vs. control). Long-term exposure (96h) to free TH and TH-ag accelerated HuT-78 doubling time by 17-25% and 16-22%, respectively (p 〈 0.05 for both comparisons). By time course analysis of mRNA abundance (1, 2, 3, 6, 12 and 24 h post TH), we found that TH-induced phenotypic changes were associated with 1.5 to 3 fold up-regulation of proliferation markers such as PCNA, CCND1, CCND2, CCND3, CCNE2 and CCNB1 in addition to a concomitant decrease of the tumor suppressor genes TP53 and CDKN1A. To determine the specific transcriptional programs regulated by mTR and TR, T-NHLs were treated with free T3/T4 (for total TR activity) and agarose-coupled T3/T4 (for mTR activity) for 24h and analyzed by mRNA-sequencing. Differentially regulated genes were further analyzed using Ingenuity Pathway Analysis. We found the TNFR and interleukin-2 pathways (e.g. TNFRSF19, TNFRSF9, IL2RA) among the top programs regulated by free THs through canonical nuclear TR activation whereas mTR activation upregulated genes involved in angiogenesis induction (e.g. PDGFR, VEGFA, VEFGB), lymphocyte proliferation/differentiation and DNA replication (e.g. IL4, DOK2). We independently validated these genes in HuT-78 and MJ cell lines by qRT-PCR. Taken together, these data suggest that TR and mTR regulate distinct transcriptional programs that are complimentary towards CTCL proliferation. Considering that (i) mTR are over-expressed in CTCL vs. normal T cells (in contrast to canonical nuclear TR that are broadly expressed) and (ii) mTR regulate pro-survival pathways in CTLC, we wished to determine whether mTR inhibition will be sufficient to decrease CTCL proliferation. We therefore tested the effect of mTR abrogation by using siRNA to ITGAV and ITGB3 as well as the effect of pharmacological mTR inhibitors (i.e: RGD peptide and Tetrac) in CTCL proliferation. We found that mTR inhibition with siRNA or compounds affected mTR-induced transcriptional programs. In fact, the TH-ag mediated upregulation of VEGFB, VEGFA, DOK2 and IL4 mRNA levels were totally abrogated in CTCL cells transfected with the siRNA to ITGAV and ITGB3. Moreover, almost 100% of the TH-ag-induced and 80% of the free-TH-induced CTCL proliferative effect was abrogated by ITGAV- and/or ITGB3-targeting siRNA. Taken together these data suggest that THs are important inducers of CTCL proliferation by transcriptionally regulating major survival pathways, and inhibition of mTR could constitute a selective chemotherapy-free treatment for CTCLs. Disclosures: No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2013
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 8
    In: Blood, American Society of Hematology, Vol. 124, No. 21 ( 2014-12-06), p. 510-510
    Abstract: Malignant T cell proliferation, survival and drug resistant are dependent on a combination of external stimuli delivered by the microenvironment. Previous studies have shown that the transmembrane receptor integrin αVβ3 plays a critical role mediating the interaction of T cell lymphoma (TCL) cells with external signals. Integrin αVβ3 ligands include extracellular matrix-associated signaling proteins and soluble factors such as thyroid hormones (TH). We have also shown that THs stimulate the proliferation of TCLs through complimentary intracellular pathways involving the αVβ3 integrin. We therefore hypothesized that targeting integrin αVβ3 could represent a novel strategy to treat TCL patients. To determine survival pathways induced by TH on αVβ3 integrins, we evaluated the transcriptional changes (RNA-seq) induced by physiological concentrations of cell impermeable T3/T4 coupled to agarose (TH-AG) vs. control in the TCL cell line CUTLL1. We identified 123 up- and 5 down-regulated transcripts (p 〈 0.01), belonging to “angiogenesis” (e.g. VEFGB), “lymphocyte proliferation/differentiation” and “DNA replication/transcription” (e.g. DBP, IL4, EDF1, DOK2) pathways. Target-gene promoter analysis suggested that TH acting on αVβ3 integrin activated NFkB (that was later confirmed by EMSA-like assays). We then focused on the angiogenesis pathway since (i) VEGF expression and angiogenesis correlate with survival and prognosis in TCL patients, and (ii) we found a positive correlation between integrin αVβ3 and VEGFA and VEGFB expression in 169 cases of TCLs. We analyzed T3/T4-αVβ3-dependent increase of VEGFB and VEGFA in an extended panel of cell lines (n=5) representing the spectrum of immature and peripheral TCL. Similarly to CUTLL1, treatment with TH-AG increased VEGFB and VEGFA mRNA levels in Jurkat (TCL/L), HuT-78 (CTCL), OCI-Ly12 (PTCL-NOS) and Karpas299 (ALCL-ALK+) cells. Increase in VEGF production was completely abrogated by knocking-down either αV or β3components with specific si-RNAs (vs. siRNA control) in CUTLL1, HuT-78 and OCI-Ly12 cells. Moreover, exposing HMEC1 endothelial cells to conditioned medium from CUTLL1 cells treated with TH-AG vs control increased they proliferation and migration (to 481 ± 118 cells from 206 ± 82 cells, respectively, p= 0.01). Importantly, this higher migration was completely abrogated when conditioned medium was obtained from CUTLL1 cells knocked-down for either αV or β3 and treated with TH-AG. To determine whether targeting αVβ3integrin could be of therapeutic benefit for TCL, we developed TCL xenografts in SCID mice using CUTLL1 cells transfected with si-control, si-αV and si-β3, and monitored tumor growth and angiogenesis. We found that CUTLL1 transfected with si-αV and si-β3 developed significant smaller tumors than si-control. Also, tumors from integrin knocked-down cells, showed decreased tumor vascularization (by CD31) and VEGF expression. To determine the translational impact of this strategy, we assessed the effect of cilengitide, a selective αVβ3 integrin inhibitor in phase 3 for glioma, in pre-clinical models of PTCL-NOS and ALCL-ALK+. For PTCL-NOS we xenografted OCI-Ly12 cells in NOD/SCID mice (n=12) and for ALCL-ALK+ we developed a patient-derived tumorgraft (PDT) in NSG mice (n=8). Cilentide treatment for 10 days (vs. vehicle), at human equivalent dose, induced tumor remission in both models (p 〈 0.001, T-test), without toxicity to normal tissues. Similarly to si-αV and si-β3 treated mice, the anti-lymphoma effect of cilengitide correlated with lower levels of angiogenesis and NFkB activation. In sum, we elucidated the mechanism by which integrin αVβ3 activation increases TCL proliferation by activating pro-survival pathways in malignant T cells while promoting angiogenesis. We also showed that genetic and pharmacological targeting of integrin αVβ3 induces anti-lymphoma effect in TCLs, including an ALCL-ALK+ PDT model obtained from an ALK inhibitor refractory patient. Disclosures No relevant conflicts of interest to declare.
    Type of Medium: Online Resource
    ISSN: 0006-4971 , 1528-0020
    RVK:
    RVK:
    Language: English
    Publisher: American Society of Hematology
    Publication Date: 2014
    detail.hit.zdb_id: 1468538-3
    detail.hit.zdb_id: 80069-7
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 73, No. 8_Supplement ( 2013-04-15), p. 1802-1802
    Abstract: T cell non-Hodgkin lymphomas (T-NHL) are a heterogeneous group of lymphoproliferative disorders with aggressive clinical course and no specific treatments. Thyroid Hormones (TH) are crucial regulators of differentiation, growth and metabolism. TH modulate these functions by activating canonical nuclear (TR) and membrane receptors (mTR, for most cells represented by RGD integrin dimers). Recent studies of us showed that TH stimulate the proliferation of T-NHLs through complimentary intracellular pathways involving both TH receptors, thus suggesting that both pathways are co-opted by malignant T cells to maintain proliferation and survival. This could represent a specific way to target these lymphoma subtypes. To characterize their participation on the T-NHL malignant phenotype, we analyzed the effect of TH through TR and mTR in 8 human cell lines representing the spectrum of immature and peripheral T-NHLs (CUTLL-1, Jurkat, Hut78, Mac-2A, OCI-Ly12, OCI-Ly13.2, SUDHL-1, Karpas299). Compared to normal T cells, T-NHL expressed higher levels of TRα and TRβ, and mTR (integrins with RGD site). To discriminate between nuclear vs. membrane-initiated effects, T-NHL cells were treated with physiological concentrations of free and cell impermeable agarose-bound T3/T4 (TH-AG), for 1 to 5 days. In these conditions, both free and TH-AG increased the proliferation of T-NHL (doubling time increased between 24 to 43%), PCNA and CCNDs proliferation markers and ERK phosphorylation. To further characterize TR and mTR initiated transcriptional programs, CUTLL1 cells were exposed to Free TH and TH-AG for 24 h and 96 h and mRNA extracted for RNA-sequencing in triplicates. We found 16 and 174 transcripts were significantly (p & lt;0.05) regulated at 24 h in free TH vs. TH-AG, respectively; and 27 and 31 at 96 h in free TH vs TH-AG, respectively. Among the top programs regulated by free TH we found activation of TR, TNFR and IL2 pathways. While activation of the mTR caused activation of genes involved in mitochondrial respiration, fatty-acid synthesis, angiogenesis and DNA replication. Common pathways activated by TR and mTR included the TNFR/ASK1/ERK and WNT/CCND signaling pathways. Transcripts of interest were validated by RT-qPCR. These data suggest that both TR and mTR regulate T-NHL proliferation to distinct yet complimentary programs. To determine whether abrogation of the mTR causes decrease in cell proliferation, we treated our panel of T-NHLs with siRNA for ITGAV, ITGB1, ITGB3 and mTR inhibitors. We found that mTR inhibition affected the mTR-induced transcriptional program and abrogated TH-induced proliferative effect. All these data suggest that (i) TH are important inducers of T-NHLs proliferation by transcriptionally regulating major survival pathways, and (ii) inhibition of mTR could constitute a selective chemotherapy-free treatment for T-NHLs. Citation Format: Florencia Cayrol, Tharu Fernando, Maria Celeste Diaz Flaqué, Ana Maria Genaro, Ricardo Farias, Leandro Cerchietti, Graciela Cremaschi. Thyroid hormones maintain the proliferative phenotype in T cell lymphomas through nuclear and membrane-initiated transcriptional programs. [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 1802. doi:10.1158/1538-7445.AM2013-1802
    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
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
  • 10
    In: Molecular Endocrinology, The Endocrine Society, Vol. 29, No. 10 ( 2015-10-01), p. 1468-1485
    Abstract: Accumulated findings have demonstrated the presence of bidirectional interactions between progesterone receptor (PR) and the ErbB family of receptor tyrosine kinases signaling pathways in breast cancer. We previously revealed signal transducer and activator of transcription 3 (Stat3) as a nodal convergence point between said signaling pathways proving that Stat3 is activated by one of the ErbBs' ligands, heregulin (HRG)β1 via ErbB2 and through the co-option of PR as a signaling molecule. Here, we found that HRGβ1 induced Stat3 recruitment to the promoters of the progestin-regulated cell cycle modulators Bcl-XL and p21CIP1 and also stimulated Stat3 binding to the mouse mammary tumor virus promoter, which carries consensus progesterone response elements. Interestingly, HRGβ1-activated Stat3 displayed differential functions on PR activity depending on the promoter bound. Indeed, Stat3 was required for PR binding in bcl-X, p21CIP1, and c-myc promoters while exerting a PR coactivator function on the mouse mammary tumor virus promoter. Stat3 also proved to be necessary for HRGβ1-induced in vivo tumor growth. Our results endow Stat3 a novel function as a coregulator of HRGβ1-activated PR to promote breast cancer growth. These findings underscore the importance of understanding the complex interactions between PR and other regulatory factors, such as Stat3, that contribute to determine the context-dependent transcriptional actions of PR.
    Type of Medium: Online Resource
    ISSN: 0888-8809 , 1944-9917
    Language: English
    Publisher: The Endocrine Society
    Publication Date: 2015
    detail.hit.zdb_id: 1492112-1
    Library Location Call Number Volume/Issue/Year Availability
    BibTip Others were also interested in ...
Close ⊗
This website uses cookies and the analysis tool Matomo. Further information can be found on the KOBV privacy pages