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: 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 ...
  • 2
    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 ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 83, No. 7_Supplement ( 2023-04-04), p. 440-440
    Abstract: Background: Endometrial cancer (ECa) is the fourth most prevalent type of cancer in women. Although ECa mostly affects older women, both the mortality and incidence rates are rapidly rising in women under the age of 40. Response rates and progression free survival are still low for recurrent ECa treatments such as hormone therapy, chemotherapy, and targeted therapies like Bevacizumab and Everolimus. There is an unmet need for the development of novel targeted therapeutics to support current ECa-directed treatments. The progression of many cancers, including ECa, is linked to proto-oncogenic PELP1 (proline-, glutamic acid-, and leucine-rich protein 1). In this investigation, we evaluated the efficacy of the PELP1 inhibitor, SMIP34, both alone and in combination with mTOR inhibitors in ECa. Methods: Using 13 distinct ECa cells, the effect of SMIP34 alone and in conjunction with mTOR inhibitors was assessed using the cell viability and colony formation assays. Using flow cytometry and Annexin-V/PI labeling, the ability of SMIP34 to promote apoptosis was evaluated. The effect of SMIP34 on cell cycle progression was evaluated using FACS analyses. RT-qPCR, Western blotting, and global RNA-seq were used for mechanistic studies. ECa patient-derived organoids (PDO), explants (PDEX) and xenograft model were used to assess the impact of SMIP34. TNM and Dependency Map (DepMap) databases were used to profile the status of PELP1 in ECa. The status of PELP1 in ECa tissues was analyzed using IHC analyses of ECa tumor tissue Microarrays. Results: Analysis of TNM plots showed that PELP1 expression was higher in ECa than in normal endometrium. Data from DepMap confirmed that PELP1 is required for the survival and expansion of ECa cells. SMIP34 treatment of established and primary patient-derived ECa cells significantly reduced cell viability and colony formation with an IC50 of 2-10µM. PELP1 knockdown significantly reduced the effect of SMIP34. Further, SMIP34 treatment induced apoptosis and promoted S-phase arrest in ECa cells. RNA-seq analyses showed that SMIP34 regulated genes positively correlated with p53 and apoptosis pathways, while negatively correlated with ribosome and eukaryotic translation elongation pathways. Western blot analyses confirmed that SMIP34 treatment reduced the levels of PELP1 and its complex of proteins including TEX10, LAS1L, SENP3. Mechanistic studies demonstrated that SMIP34 treatment attenuated the activation of mTOR signaling in ECa cells. Accordingly, SMIP34 enhanced the effect of mTOR inhibitors on ECa cells. SMIP34 is effective in reducing cell viability in PDO and limited the cells proliferation of PDEX. Importantly, SMIP34 treatment significantly reduced of the growth of ECa cell line-derived xenografts. Conclusions: Collectively, our results suggest that SMIP34 inhibits ECa growth in vitro, in vivo, and ex vivo and combination of SMIP34 with mTOR inhibitor represents a novel therapeutic strategy for ECa. Citation Format: Xue Yang, Zexuan Liu, Weiwei Tang, Uday P. Pratap, Alexia B. Collier, Kristin A. Alwegg, Nicole Spencer, Xiaonan Li, Philip T. Valente, Edward R. Kost, Suryavathi Viswanadhapalli, Ratna K. Vadlamudi. The role of PELP1 inhibitor SMIP34 in the treatment of endometrial 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 440.
    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: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 4_Supplement ( 2022-02-15), p. P5-10-01-P5-10-01
    Abstract: Background: The obesity epidemic is rapidly increasing in the USA and obese women are at a higher likelihood of developing triple negative breast cancer (TNBC). Several studies implicated the importance of the breast microenvironment on the aggressive cancer biology especially obese microenvironment. However, the underlying mechanism(s) by which obesity contributes to the progression of TNBC remains unclear. The objective of this study is to test a novel concept that obesity upregulates leukemia inhibitory factor receptor (LIFR) oncogenic signaling in TNBC and test whether LIFR inhibition blocks TNBC progression. Methods: Established TNBC cell lines were co-cultured with human primary adipocytes or incubated with adipocyte conditioned medium or with high glucose (HG) followed by treatment with LIFR inhibitor EC359. The effect of adiposity on TNBC cells was determined using cell viability, colony formation, and invasion assays. Mechanistic studies were performed using CRISPR/Cas9 KO of LIFR, Western blotting, RT-qPCR, and reporter gene assays. Utility of LIFR inhibitor EC359 was tested using xenografts, and patient derived organoid (PDO) models. Results: Treatment of TNBC cells with adipose conditions or HG increased the proliferation and invasion of TNBC cells. Western blot and RT-qPCR analyses confirmed that increased expression of LIFR correlated with enhanced downstream LIFR signaling such as STAT3 and subsequent activation of STAT3 target genes. CRISPR KO of LIFR or treatment of TNBC cells with EC359 significantly reduced the cell viability, colony formation and invasion under adipose conditions. Western blotting results showed that co-culture with adipocytes significantly enhanced LIFR downstream signaling in TNBC model cells and is effectively blocked by LIFR KO or EC359 treatment. Further, EC359 treatment blocked the adipose environment mediated growth of organoids. Importantly, co-implantation of adipocytes significantly enhanced TNBC xenograft tumor growth, however treatment with EC359 significantly attenuated adipocyte induced TNBC progression. Conclusions: Collectively, these results suggest that adiposity contributes to increased TNBC cell growth via upregulation of the LIF/LIFR pathway. The LIF/LIFR axis represents a potential therapeutic target for adiposity driven TNBC and the LIFR inhibitor EC359 could be used as a new therapeutic agent to treat obesity associated TNBC. Citation Format: Suryavathi Viswanadhapalli, Uday P Pratap, Behnam Ebrahimi, Logan Blankenship, Jaitri Joshi, Zexuan Liu, Kristin A Altwegg, Xiaonan Li, Gangadhara R Sareddy, Bindu Santhamma, Swapna Konda, Manjeet Rao, Edward Kost, Rajeshwar R Tekmal, Hareesh B Nair, Ratna K Vadlamudi. Leukemia inhibitory factor receptor inhibition reduces obesity driven progression of triple negative breast cancer [abstract]. In: Proceedings of the 2021 San Antonio Breast Cancer Symposium; 2021 Dec 7-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2022;82(4 Suppl):Abstract nr P5-10-01.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2022
    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. 81, No. 13_Supplement ( 2021-07-01), p. 1253-1253
    Abstract: Background: Endometrial cancer (EC) is the fourth most common cancer in women. Approximately 80% of EC belong to the endometroid-EC subtype and are driven by estrogen signaling. Advanced-stage EC has limited treatment options with poor prognosis. There is an urgent need for the identification of actionable drivers as new targets for treating advance stage EC. Leukemia inhibitory factor receptor (LIFR) and its ligand LIF plays a major role in cancer progression, metastasis, stemness, and therapy resistance. Published and our preliminary data suggest a critical role of the LIF-LIFR signaling axis in EC progression. The objective of this study is to test the utility of targeting the LIF/LIFR axis using a novel LIFR inhibitor, EC359. Methods: We used multiple established and primary EC cells to test the utility LIFR inhibitor, EC359 in treating EC. CRISPR/Cas9 system was used to generate LIFR KO EC cells. In vitro activity was tested using Cell-Titer Glo, MTT, invasion, and apoptosis assays. Mechanistic studies were conducted using Western blot, reporter gene assays, and RNA-seq analysis. EC cell-derived xenograft (CDX) and patient-derived explant (PDEX) models were used for preclinical evaluation and toxicity. Results: EC359 treatment of seven EC cells showed anti-proliferative effects in MTT cell viability assays with an IC50 of 25-100 nM. Further, EC359 treatment reduced invasiveness, stemness, and promoted apoptosis of EC cells. The activity of EC359 is dependent on LIF/LIFR expression in EC cells. CRISPR mediated knockout of LIFR significantly abolished EC359 activity. In vivo xenograft studies using Ishikawa-vector or LIFR-KO cells demonstrated that LIFR-KO significantly reduced EC tumor growth, and tumor weights. Further, EC359 treatment attenuated the activation of LIF/LIFR driven pathways, including STAT3, AKT-mTOR signaling. Mechanistic studies using RNA-seq revealed that EC359 significantly upregulated 213 genes and down regulated 126 genes. Pathway analyses of differential genes revealed enrichment in the apoptotic pathways upon EC359 treatment. EC359 (5mg/kg body weight) treatment significantly reduced CDX tumor progression and reduced proliferation in PDEX models. Conclusions: Collectively, these data support EC359 as a novel targeted therapy for EC by inhibiting LIF/LIFR oncogenic signaling pathway. Citation Format: Weiwei Tang, Kumaraguruparan Ramasamy, Sureshkumar M. Pillai, Bindu Santhamma, Swapna Konda, Prabhakar P. Vekata, Logan Blankenship, Junhao Liu, Zexuan Liu, Kristin A. Altwegg, Behnam Ebrahimi, Uday P. Pratap, Xiaonan Li, Edward Kost, Gangadhara R. Sareddy, Ratna K. Vadlamudi, Hareesh B. Nair, Rajeshwar R. Tekmal, Suryavathi Viswanadhapalli. Therapeutic targeting of endometrial cancer with novel LIFR inhibitor EC359 [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2021; 2021 Apr 10-15 and May 17-21. Philadelphia (PA): AACR; Cancer Res 2021;81(13_Suppl):Abstract nr 1253.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2021
    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: Cell Death Discovery, Springer Science and Business Media LLC, Vol. 7, No. 1 ( 2021-08-16)
    Abstract: Endometrial cancer (EC) is the fourth most common cancer in women. Advanced-stage EC has limited treatment options with a poor prognosis. There is an unmet need for the identification of actionable drivers for the development of targeted therapies in EC. Leukemia inhibitory factor receptor (LIFR) and its ligand LIF play a major role in cancer progression, metastasis, stemness, and therapy resistance. However, little is known about the functional significance of the LIF/LIFR axis in EC progression. In this study using endometrial tumor tissue arrays, we identified that expression of LIF, LIFR is upregulated in EC. Knockout of LIFR using CRISPR/Cas9 in two different EC cells resulted in a significant reduction of their cell viability and cell survival. In vivo studies demonstrated that LIFR-KO significantly reduced EC xenograft tumor growth. Treatment of established and primary patient-derived EC cells with a novel LIFR inhibitor, EC359 resulted in the reduction of cell viability with an IC 50 in the range of 20–100 nM and induction of apoptosis. Further, treatment with EC359 reduced the spheroid formation of EC cancer stem cells and reduced the levels of cancer stem cell markers SOX2, OCT4, NANOG, and Axin2. Mechanistic studies demonstrated that EC359 treatment attenuated the activation of LIF-LIFR driven pathways, including STAT3 and AKT/mTOR signaling in EC cells. Importantly, EC359 treatment resulted in a significant reduction of the growth of EC patient-derived explants ex vivo, EC cell line-derived xenografts, and patient-derived xenografts in vivo. Collectively, our work revealed the oncogenic potential of the LIF/LIFR axis in EC and support the utility of LIFR inhibitor, EC359, as a novel targeted therapy for EC via the inhibition of LIF/LIFR oncogenic signaling.
    Type of Medium: Online Resource
    ISSN: 2058-7716
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2842546-7
    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...