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: Fertility and Sterility, Elsevier BV, Vol. 60, No. 2 ( 1993-08), p. 242-246
    Type of Medium: Online Resource
    ISSN: 0015-0282
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1993
    detail.hit.zdb_id: 1500469-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Fertility and Sterility, Elsevier BV, Vol. 61, No. 5 ( 1994-05), p. 872-879
    Type of Medium: Online Resource
    ISSN: 0015-0282
    Language: English
    Publisher: Elsevier BV
    Publication Date: 1994
    detail.hit.zdb_id: 1500469-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 3408-3408
    Abstract: Background: Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer (BC) that accounts for a disproportionate amount of BC mortality. The relationship(s) between obesity and the TNBC is crucial because the obesity prevalence in the USA is on the rise. Obese patients are generally diagnosed with large primary tumors, more lymph node metastases, and obesity effects on tumor microenvironment (TME) are suspected to accelerate the development of TNBC, but the exact mechanism(s) by which this occurs remain unknown. The goal of this study is to investigate the hypothesis that obesity enhances leukemia inhibitory factor receptor (LIFR) oncogenic signaling in TNBC and to test the utility of LIFR inhibitor in blocking obesity driven progression of TNBC. Methods: Established TNBC cell lines were co-cultured with human primary adipocytes, incubated with adipocyte conditioned media, or exposed to high glucose (HG), then treated with the LIFR inhibitor EC359. Cell viability, colony formation, and invasion assays were used to analyze the impact of obesity on TNBC cells and to test utility of EC359. RT-qPCR, Western blotting, reporter gene assays, and RNA-seq analyses were used in the mechanistic studies. Xenografts and patient-derived organoid (PDO) models were used to evaluate the effectiveness of the EC359. Results: The cell proliferation and invasion of TNBC cells were accelerated by adipocyte conditioned media or when exposed to HG. RNA-seq and RT-qPCR analysis revealed a correlation between elevated LIFR expression and downstream LIFR signaling, including STAT3, as well as the subsequent activation of STAT3 target genes. The cell viability, colony formation, and invasion of TNBC cells under HG and adipose conditions were all markedly decreased after treatment with LIFR inhibitor EC359. Results from Western blotting demonstrated that co-culture with adipocytes or incubation with HG dramatically increased LIFR downstream signaling in TNBC model cells, and that this signaling is effectively suppressed by EC359 therapy. In addition, administration of EC359 prevented organoid proliferation that was mediated by the adipose conditioned media. Importantly, co-implantation of adipocytes greatly increased the growth of the TNBC xenograft tumor; however, therapy with EC359 significantly reduced the growth of TNBC caused by adipocyte co-implantation. Conclusions: Collectively, these findings suggest that obesity conditions promote the activation of LIF/LIFR pathway, which in turn enhances TNBC cell proliferation. The LIFR inhibitor EC359 may be employed as a new therapeutic drug to treat obesity driven TNBC and LIF/LIFR axis represents a potential therapeutic target for obesity driven TNBC. Citation Format: Lois Randolph, Alondra Rodriguez Sanchez, Logan Blankenship, Uday P. Pratap, Xue Yang, Durga Meenakshi Panneerdoss, Swapna Konda, Bindu Santhamma, Gangadhara R. Sareddy, Manjeet K. Rao, Edward R. Kost, Rajeshwar R. Tekmal, Hareesh B. Nair, Ratna K. Vadlamudi, Suryavathi Viswanadhapalli. The role of obesity in promoting the LIF/LIFR signaling in triple negative breast cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 3408.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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: Gynecologic Oncology, Elsevier BV, Vol. 89, No. 2 ( 2003-5), p. 218-226
    Type of Medium: Online Resource
    ISSN: 0090-8258
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2003
    detail.hit.zdb_id: 1467974-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: International Journal of Molecular Sciences, MDPI AG, Vol. 23, No. 13 ( 2022-06-28), p. 7159-
    Abstract: Ovarian cancer (OCa) is the deadliest gynecologic cancer. Emerging studies suggest ovarian cancer stem cells (OCSCs) contribute to chemotherapy resistance and tumor relapse. Recent studies demonstrated estrogen receptor beta (ERβ) exerts tumor suppressor functions in OCa. However, the status of ERβ expression in OCSCs and the therapeutic utility of the ERβ agonist LY500307 for targeting OCSCs remain unknown. OCSCs were enriched from ES2, OV90, SKOV3, OVSAHO, and A2780 cells using ALDEFLUOR kit. RT-qPCR results showed ERβ, particularly ERβ isoform 1, is highly expressed in OCSCs and that ERβ agonist LY500307 significantly reduced the viability of OCSCs. Treatment of OCSCs with LY500307 significantly reduced sphere formation, self-renewal, and invasion, while also promoting apoptosis and G2/M cell cycle arrest. Mechanistic studies using RNA-seq analysis demonstrated that LY500307 treatment resulted in modulation of pathways related to cell cycle and apoptosis. Western blot and RT-qPCR assays demonstrated the upregulation of apoptosis and cell cycle arrest genes such as FDXR, p21/CDKN1A, cleaved PARP, and caspase 3, and the downregulation of stemness markers SOX2, Oct4, and Nanog. Importantly, treatment of LY500307 significantly attenuated the tumor-initiating capacity of OCSCs in orthotopic OCa murine xenograft models. Our results demonstrate that ERβ agonist LY500307 is highly efficacious in reducing the stemness and promoting apoptosis of OCSCs and shows significant promise as a novel therapeutic agent in treating OCa.
    Type of Medium: Online Resource
    ISSN: 1422-0067
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2019364-6
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Cancers, MDPI AG, Vol. 14, No. 21 ( 2022-11-02), p. 5400-
    Abstract: Endometrial cancer (EC) is the fourth most common cancer in women, and half of the endometrioid EC (EEC) cases are attributable to obesity. However, the underlying mechanism(s) of obesity-driven EEC remain(s) unclear. In this study, we examined whether LIF signaling plays a role in the obesity-driven progression of EEC. RNA-seq analysis of EEC cells stimulated by adipose conditioned medium (ADP-CM) showed upregulation of LIF/LIFR-mediated signaling pathways including JAK/STAT and interleukin pathways. Immunohistochemistry analysis of normal and EEC tissues collected from obese patients revealed that LIF expression is upregulated in EEC tissues compared to the normal endometrium. Treatment of both primary and established EEC cells with ADP-CM increased the expression of LIF and its receptor LIFR and enhanced proliferation of EEC cells. Treatment of EEC cells with the LIFR inhibitor EC359 abolished ADP-CM induced colony formation andcell viability and decreased growth of EEC organoids. Mechanistic studies using Western blotting, RT-qPCR and reporter assays confirmed that ADP-CM activated LIF/LIFR downstream signaling, which can be effectively attenuated by the addition of EC359. In xenograft assays, co-implantation of adipocytes significantly enhanced EEC xenograft tumor growth. Further, treatment with EC359 significantly attenuated adipocyte-induced EEC progression in vivo. Collectively, our data support the premise that LIF/LIFR signaling plays an important role in obesity-driven EEC progression and the LIFR inhibitor EC359 has the potential to suppress adipocyte-driven tumor progression.
    Type of Medium: Online Resource
    ISSN: 2072-6694
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2527080-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Cancer Letters, Elsevier BV, Vol. 575 ( 2023-10), p. 216383-
    Type of Medium: Online Resource
    ISSN: 0304-3835
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 195674-7
    detail.hit.zdb_id: 2004212-7
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 4966-4966
    Abstract: Background: Of all gynecologic cancers, ovarian cancer (OCa) has the highest mortality rates. Nearly 90% of patients who receive standard surgical and cytotoxic treatment experience disease recurrence. Leukemia inhibitory factor (LIF) and its receptor LIFR are implicated in the progression of several cancers. A knowledge gap exists on whether LIF/LIFR plays a role in the evolution of OCa. We recently developed EC359, a first-in-class LIFR inhibitor. Here, we examined whether autocrine loops of LIF/LIFR contribute to OCa progression and tested the utility of EC359 as a potential targeted therapy. Methods: Eighteen different OCa model cells, both established and primary, were used to profile the expression of LIF and LIFR. Cell viability, colony formation, apoptosis, and reporter assays were used to assess EC359 impact on OCa cells. Mechanistic studies were carried out using RNA-seq and RT-qPCR analysis. Using cell-based xenografts, syngeneic xenografts, patient derived organoids (PDO), and patient derived xenograft (PDX) models, the effectiveness of LIFR inhibitor EC359 as a targeted therapy was examined. Results: Kaplan-Meier survival analysis (KMplot) revealed increased expression of LIF and LIFR was linked to poor progression-free survival in OCa patients. The levels of LIF and LIFR were considerably greater in OCa chemotherapy non-responders than responders. We validated the existence of LIF/LIFR autocrine signaling using 18 distinct OCa cells. Treatment with the LIFR inhibitor EC359 dramatically decreased OCa cell viability, cell survival and increased apoptosis, with an IC50 of 5 to 50 nM. The activation of STAT3, mTOR, AKT, and p42/44 MAPKs as well as other downstream LIFR signaling was markedly decreased by EC359 treatment. Treatment with EC359 also decreased the stemness of OCa cells, slowed PDO development, and sensitized chemotherapy-resistant OCa cells to chemotherapy. One of the significant pathways elevated by EC359, according to RNA-seq data, is the regulation of apoptosis. In six different cell-based xenografts and PDX tumors, we demonstrated that the EC359 at 5mg/kg dose significantly reduced the OCa xenograft growth. In comparison to the vehicle control, the tumor volume was significantly reduced by EC359 treatment of murine ID8 xenografts in C57BL6 mice. Our findings indicated that EC359 had both intrinsic and extrinsic effects on tumors. Tumor-associated macrophages (TAMs) with a significant M1 polarity (CD11b+Gr1-CD68high/phosphoSTAT1+/cMAF-) and robust tumor infiltration by (CD45+) leukocytes were enhanced with EC359 therapy of ID8 xenograft tumors. Importantly, normal T, B, and other immune cells in the blood demonstrated that EC359 had no effect on immune cell homeostasis. Conclusions: Together, our findings support the existence of LIF/LIFR autocrine loops, and EC359 is a viable treatment option for OCa. Citation Format: Behnam Ebrahimi, Suryavathi Viswanadhapalli, Uday P. Pratap, Rahul Gopalam, Xue Yang, Bindhu Santhamma, Swapna Konda, Xiaonan Li, Hui Yan, Gangadhara R. Sareddy, Zhenming Xu, Edward R. Kost, Rajeshwar R. Tekmal, Hareesh B. Nair, Ratna K. Vadlamudi. Targeting LIF/LIFR autocrine loops with EC359 in ovarian cancer: A novel LIFR targeted therapy. [abstract]. In: Proc eedings of the American Association for Cancer Research Annual Meeting 2023; Part 1 (Regular and Invited Abstracts); 2023 Apr 14-19; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2023;83(7_Suppl):Abstract nr 4966.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    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
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5875-5875
    Abstract: Background: Ovarian cancer (OCa) is the deadliest of all gynecologic cancers. OCa patients initially respond to standard combinations of surgical and cytotoxic therapy; however, ~80% will develop recurrence and inevitably succumb to chemotherapy-resistant disease. OCa stem cells are implicated in the tumor initiation and therapy resistance. LIFR signaling plays a critical role in OCa progression and stemness. Further, high circulating LIF levels correlate with tumor recurrence and chemoresistance. The autocrine loop involving LIF, LIFR and STAT3 axis drives sustained fibroblast production of inflammatory mediators. This represents a significant problem and a critical need exists for development of novel therapies targeting the LIFR axis for treating OCa. Methods: We have rationally designed and synthesized a small organic molecule (EC359) that emulates the LIF-LIFR binding site and functions as a LIFR inhibitor from a library of compounds. In silico docking studies were used to identify the putative interaction of the EC359 and LIF/LIFR complex. Binding of EC359 to LIFR was confirmed using surface plasmon resonance (SPR) and IP assays. Mechanistic studies were conducted using Western, RT-qPCR, and RNA-Seq analysis. Xenograft models were used for preclinical evaluation and toxicity. The efficacy of EC359 was tested using Patient-Derived eXplants (PDeX). Results: Global analysis of online databases revealed negative correlation of OCa survival with LIFR expression. Molecular docking studies showed EC359 interacts at the LIF-LIFR binding interface. SPR studies confirmed interaction of EC359 to LIFR. Western analysis of eight cells that represent four subtypes of OCa confirmed higher expression of LIF and LIFR. EC359 reduced the growth of eight OCa cells with high potency (IC50 10-50 nM) and promoted apoptosis. EC359 treatment reduced stemness of OCa cells. EC359 activity is dependent on the level of expression of LIFR with little activity on cells that do not express LIFR. EC359 significantly reduced the viability of carboplatin- and taxol-resistant OCa cells. Mechanistic studies showed EC359 interacts with LIFR and block its interaction with LIF. EC359 treatment reduced the STAT3 phosphorylation, mTOR and downstream survival signaling cascades. RNA sequencing revealed unique pathways blocked by EC359. Treatment of xenograft tumors with EC359 significantly reduced the tumor volume compared to control. Further, using PDeX of OCa, we demonstrated that EC359 has potential to reduce the proliferation. Pharmacologically, EC359 exhibited high oral bioavailability and long half-life with a wide therapeutic window. Conclusions: EC359 is a novel agent that targets LIF-LIFR axis and has activity against chemotherapy-resistant and primary OCa tumors. EC359 has the distinct pharmacologic advantages of oral bioavailability, in vivo stability, and is associated with minimal systemic side effects. Citation Format: Suryavathi Viswanadhapalli, Hareesh B Nair, Bindu Santhamma, Gangadhara R Sareddy, Yiliao Luo, Xinlei Pan, Edward R Kost, Ramachandran Murali, Rajeshwar Rao Tekmal, Klaus J Nickisch, Ratna K Vadlamudi. Development of LIFR inhibitor EC359 as a novel therapeutic for 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 5875.
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 1 ( 2019-01-01), p. 196-208
    Abstract: Emerging evidence indicates that adipose stromal cells (ASC) are recruited to enhance cancer development. In this study, we examined the role these adipocyte progenitors play relating to intercellular communication in obesity-associated endometrial cancer. This is particularly relevant given that gap junctions have been implicated in tumor suppression. Examining the effects of ASCs on the transcriptome of endometrial epithelial cells (EEC) in an in vitro coculture system revealed transcriptional repression of GJA1 (encoding the gap junction protein Cx43) and other genes related to intercellular communication. This repression was recapitulated in an obesity mouse model of endometrial cancer. Furthermore, inhibition of plasminogen activator inhibitor 1 (PAI-1), which was the most abundant ASC adipokine, led to reversal of cellular distribution associated with the GJA1 repression profile, suggesting that PAI-1 may mediate actions of ASC on transcriptional regulation in EEC. In an endometrial cancer cohort (n = 141), DNA hypermethylation of GJA1 and related loci TJP2 and PRKCA was observed in primary endometrial endometrioid tumors and was associated with obesity. Pharmacologic reversal of DNA methylation enhanced gap-junction intercellular communication and cell–cell interactions in vitro. Restoring Cx43 expression in endometrial cancer cells reduced cellular migration; conversely, depletion of Cx43 increased cell migration in immortalized normal EEC. Our data suggest that persistent repression by ASC adipokines leads to promoter hypermethylation of GJA1 and related genes in the endometrium, triggering long-term silencing of these loci in endometrial tumors of obese patients. Significance: Studies reveal that adipose-derived stem cells in endometrial cancer pathogenesis influence epigenetic repression of gap junction loci, which suggests targeting of gap junction activity as a preventive strategy for obesity-associated endometrial cancer.
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...