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)  (6)
  • 1
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2010
    In:  Cancer Research Vol. 70, No. 8_Supplement ( 2010-04-15), p. 293-293
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 70, No. 8_Supplement ( 2010-04-15), p. 293-293
    Abstract: Increasing evidence supports a role for protein kinase C (PKC) signaling in regulation of intestinal epithelial self-renewal, and alterations in PKC isozyme expression/activity have been broadly linked to intestinal carcinogenesis. PKCα, which exhibits growth inhibitory and tumor suppressor properties in the intestine, is lost early during intestinal tumor development. To gain further insight into the tumor suppressive effects of PKCα signaling, we explored the consequences of restoration of PKCα expression in DLD-1 human colon cancer cells by microarray analysis. The most downregulated known gene in PKCα-expressing DLD-1 cells was inhibitor of differentiation 1 (Id1), a member of the Id family of dominant negative antagonists of bHLH transcription factors that has cell cycle stimulatory functions and has been associated with tumorigenesis in a number of systems. Suppression of Id1 protein by PKCα in colon cancer cells was confirmed by Western blot analysis in a panel of cell lines including DLD-1, HCT116, FET, FET/DNR, and GEO. Notably, activation of PKC/PKCα in non-transformed IEC-18 intestinal crypt cells also resulted in rapid and profound downregulation of Id1 mRNA and protein, pointing to a role for PKCα in Id1 regulation in the normal intestinal epithelium. PKC-induced suppression of Id1 was found to occur via an ERK/MAPK-dependent, PI3K-independent mechanism. Manipulation of Id1 levels in normal and transformed intestinal cells demonstrated its ability to affect two key cell cycle targets of PKCα, the potent mitogen cyclin D1 and the cdk inhibitor p21Waf1/Cip1. Knockdown of Id1 consistently decreased steady-state levels of cyclin D1, while overexpression of the factor resulted in reduced levels of p21Waf1/Cip1. Thus, Id1 appears to control the expression of important cell cycle regulators in intestinal cells. Immunohistochemical analysis of normal intestinal mucosa further demonstrated that PKCα activation, which occurs precisely at the point of growth arrest in the mid/upper crypt, coincides with marked downregulation of Id1 in vivo. Conversely, loss of PKCα in intestinal tumors is associated with Id1 overexpression. Ongoing studies are focused on delineating the link between the tumor suppressor functions of PKCα and its inhibitory effects on Id1 expression in intestinal cells. Supported by NIH grants DK54909, DK60632, CA16506, and CA113048. Citation Format: {Authors}. {Abstract title} [abstract]. In: Proceedings of the 101st Annual Meeting of the American Association for Cancer Research; 2010 Apr 17-21; Washington, DC. Philadelphia (PA): AACR; Cancer Res 2010;70(8 Suppl):Abstract nr 293.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2010
    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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2016
    In:  Cancer Research Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1884-1884
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1884-1884
    Abstract: Members of the Id family (Id1-Id4) are emerging as attractive therapeutic targets in multiple tumor types. Id proteins are dominant negative antagonists of basic helix-loop-helix transcription factors, with recognized functions in development, stem cell maintenance/self-renewal, and cell fate determination. Recent studies have identified Id proteins as master regulators of cancer-initiating cells and tumor aggressiveness, with critical roles in regulation of central hallmarks of cancer, including cell proliferation, survival, senescence, angiogenesis, migration, metastasis, and chemoresistance. Deregulated Id1 and/or Id3 expression has been observed in more than twenty human cancers, including colorectal cancer (CRC). Based on their important roles in tumors, Id proteins are being actively explored as therapeutic targets, with promising results in mouse models and human tumor cell lines. Id1 and Id3 appear to have redundant functions in CRC; thus Id-based therapy would ideally involve a strategy that targets Id1 and Id3 simultaneously. We have identified a novel pathway of Id1 and Id3 repression involving the signal transduction molecule PKCα. Activation of PKCα in non-transformed intestinal cells (IEC-18 cells) and human CRC cells that retain PKCα potently suppresses Id1/Id3 mRNA and protein. Restoration of the enzyme in PKCα-deficient CRC cells also results in Id1/Id3 downregulation. Suppression occurs at the transcriptional level and is mediated by the proximal 932 bp of the promoter for Id1. Notably, the effects of PKCα were observed in CRC cell lines with diverse genetic backgrounds, differing in the status of APC, β-catenin, K-RAS, the PI3K/AKT pathway, and/or TP53. PKCα activates a growth inhibitory MEK/ERK signaling pathway in intestinal cells that is required for Id1/Id3 downregulation. Our demonstration that PKCα can activate ERK and suppress Id1/Id3 in KRAS-mutant CRC cells suggested that the effects of PKCα occur downstream of RAS activation. Notably, PKCα was unable to downregulate Id1/Id3 in CRC cells harboring V600E mutations in BRAF, indicating that RAF activation is a key mediator of PKCα-induced Id downregulation. This conclusion was further supported by the ability of the RAF inhibitor, sorafenib, to block the effects of PKCα on Id1/Id3 in BRAF-wild type cells. KSR1 is an important regulator of RAF function; however, PKCα retained its ability to downregulate Id1 in KSR1 knockdown CRC cells, excluding a role for altered KSR1-RAF interactions in the effects of PKCα. Collectively, these data indicate that PKCα regulates a novel signaling pathway for downregulation of Id1/Id3 that intersects the MEK/ERK pathway at or upstream of RAF, but downstream of RAS. Manipulation of this pathway for coincident downregulation of Id1 and Id3 offers a promising therapeutic strategy for treatment of CRC and other cancer types. Supported by NIH grants CA036727, CA016056, CA191894 and DK60632. Citation Format: Michelle A. Lum, Robert E. Lewis, Adrian R. Black, Jennifer D. Black. A PKCα-RAF-ERK signaling axis for downregulation of Id1 and Id3 in colorectal cancer cells. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 1884.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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) ; 2020
    In:  Cancer Research Vol. 80, No. 16_Supplement ( 2020-08-15), p. 307-307
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 80, No. 16_Supplement ( 2020-08-15), p. 307-307
    Abstract: KRAS is mutated and activated in an estimated 40% of colon cancers but has proven difficult to target directly due to the lack of drug-binding pockets on the surface of Ras. Thus, identifying effectors that transmit signals from oncogenic Ras is a critical step in targeting vulnerabilities in the tumors of patients with colorectal cancer (CRC). Kinase Suppressor of Ras 1 (KSR1) is a molecular scaffold that coordinates the interaction of effectors in the Raf/MEK/ERK signaling cascade downstream of Ras. KSR1 is necessary for oncogenic transformation of cells expressing mutated Ras but is dispensable for normal cell growth making KSR1 an attractive therapeutic target for Ras-driven colon cancers. In CRC cells, KSR1 and ERK mediate Ras-dependent effects on protein translation, notably driving cap-dependent and cap-independent translation of the Myc oncogene. Depletion of KSR1 or treatment with ERK inhibitor (SCH772984) leads to a decrease in phophorylated 4EBP1 and a decrease in PDCD4, which collectively impair protein translation, notably leading to a decrease in Myc translation. Patient-derived CRC organoids with deletion of APC, p53, and SMAD4, and an activating G12D mutation in KRAS treated with ERK inhibitor show a marked decrease in Myc protein expression, coincident with changes in PDCD4 and 4EBP1 that impair protein translation. These observations indicate that the KSR1-dependent ERK activation observed in CRC cell lines is present in pre-clinical colon tumoroid models. Genome-wide polysome profiling in CRC cell lines HCT116 and HCT15 depleted of KSR1 identified mRNAs that were preferentially translated in a KSR1 and ERK-dependent manner. Several of these mRNAs were previously predicted by our Functional Signature Ontology (FUSION) screen to be critical to CRC viability but dispensable for normal cell growth. GSEA identified functional classes of mRNAs whose translation is KSR1-dependent, including mediators of oncogenic signaling in pathways that regulate mTOR, MAPK, WNT and JAK-STAT. Our data suggest that an essential component of oncogenic Ras-induced and KSR1-dependent signaling is the preferential translation of mRNAs supporting the transformed phenotype of CRC cells. Citation Format: Heidi Vieira, Chaitra Rao, Adrian R. Black, Siddesh Southekal, Tomohiro Mizutani, Babu Guda, Hans Clevers, Jennifer D. Black, Robert E. Lewis. KSR1-dependent modulation of the translational landscape in Ras-driven colorectal cancer [abstract]. In: Proceedings of the Annual Meeting of the American Association for Cancer Research 2020; 2020 Apr 27-28 and Jun 22-24. Philadelphia (PA): AACR; Cancer Res 2020;80(16 Suppl):Abstract nr 307.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2020
    detail.hit.zdb_id: 2036785-5
    detail.hit.zdb_id: 1432-1
    detail.hit.zdb_id: 410466-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2014
    In:  Cancer Research Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4211-4211
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 74, No. 19_Supplement ( 2014-10-01), p. 4211-4211
    Abstract: Endometrial cancer is the most common gynecological malignancy in the US and the fourth most common cancer in women. Despite its high incidence, there is limited knowledge of the molecular etiology of this disease. With obesity being a major risk factor, the incidence of endometrial cancer has been on a steady incline and is expected to become an increasing cause of cancer mortality in future years. Thus, improved understanding of endometrial tumorigenesis is of critical importance. Our analysis of human endometrial tumors identified loss of the signaling molecule PKCα in approximately 60% of endometrioid (Type I) tumors. Loss of PKCα trends with increasing tumor grade and is associated with aggressive disease features such as lymphovascular involvement and myometrial invasion, pointing to an important role for this molecule in regulating endometrial cancer progression. Inactivation of the tumor suppressor PTEN and deregulation of the PI3K/AKT pathway are key drivers of Type I endometrial cancer. Thus, mice with allele-specific knock-in of cancer-related Pten mutants (i.e., PtenΔ4-5, PtenC124R, and PtenG120E) offer unique models for the disease. Precancerous endometrial hyperplasias arising in these mice show loss of PTEN and increased AKT activity. While PKCα is expressed in the normal murine endometrial epithelium at all phases of the estrus cycle, the enzyme is uniformly lost in these precancerous lesions. Thus, disruption of PKCα signaling can occur early in endometrial tumorigenesis. In human endometrial cancer cell lines, low PKCα levels also correlate with PTEN loss; however, shRNA-mediated knockdown of PTEN in PTEN expressing cells did not affect PKCα expression indicating that downregulation of the kinase is not the direct result of changes in PTEN activity. Notably, PKC agonists suppress AKT activity in cells expressing high levels PKCα (HEC-1-A, HEC-50) but not in PKCα-low cells (Ishikawa, RL95-2). Collectively, these data suggest that, in addition to PTEN inactivation, loss of PKCα is required for full activation of the PI3K/AKT signaling pathway during endometrial carcinogenesis. Analysis of the molecular basis for loss of PKCα expression in endometrial tumors identified multiple regulatory mechanisms. PKCα protein expression in tumors generally parallels that of its mRNA and promoter activity assays point to transcriptional mechanisms for PKCα downregulation. Differential mRNA and protein stability were identified as additional levels of regulation of PKCα expression in endometrial cancer. Taken together, our findings indicate that 1) PKCα signaling regulates the PI3K/AKT pathway in the endometrial epithelium; 2) concomitant loss of PKCα and PTEN may act in a co-operative manner in regulation of endometrial turmorigenesis; and 3) loss of PKCα can occur by multiple mechanisms, as often seen with tumor suppressive molecules during neoplastic progression. Supported by NIH grants CA036727, CA016056, and DK60632. Citation Format: Alice H. Hsu, Kathryn J. Curry, Kang-Sup Shim, Peter Frederick, Carl D. Morrison, Baojing Chen, Subodh M. Lele, Gustavo Leone, Adrian R. Black, Jennifer D. Black. Protein kinase C alpha (PKCα) signaling in endometrial cancer. [abstract]. In: Proceedings of the 105th Annual Meeting of the American Association for Cancer Research; 2014 Apr 5-9; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2014;74(19 Suppl):Abstract nr 4211. doi:10.1158/1538-7445.AM2014-4211
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2014
    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. 75, No. 15_Supplement ( 2015-08-01), p. 4720-4720
    Abstract: Endometrial cancer is the most common gynecological malignancy and the fourth most common cancer in women in the United States. Although loss of PTEN and deregulation of PI3K/AKT signaling has been implicated in approximately 90% of endometrial cancer cases, understanding of the molecular etiology of the disease remains limited. Our analysis of tissue from 448 endometrial cancer patients has determined that ∼60% of human tumors show reduced expression or loss of the signal transduction molecule PKCα. Loss of this enzyme is also seen in endometrial hyperplasias arising in mouse models carrying germline mutations of PTEN (PtenΔ4-5, PtenC124R and PtenG129E) or endometrial specific deletion of PTEN (Ptenpr-/−), indicating that loss of PKCα can be an early event in development of the disease. In patients, reduced expression of PKCα correlates with markers of disease aggressiveness, such as increased myometrial invasion and lymph node involvement, supporting a tumor suppressive role for the enzyme in the endometrium. This correlation is particularly marked in endometrioid, PTEN-mutant disease, pointing to a potential link between PKCα and PI3K/AKT signaling. The role of PKCα in endometrial cancer was further examined using a panel of 17 human endometrial cancer cell lines with varying expression of the kinase. Restoration of PKCα in cell lines lacking the enzyme blocked their ability to grow in soft agarose, further supporting a tumor suppressive role of the kinase in this tissue. Stimulation of cells that express PKCα with PKC agonists reduced phosphorylation/activation of AKT and promoted loss of downstream PI3K/AKT targets such as cyclin D1 and inhibitor of DNA binding 1 (Id1). Analysis of the PKC isozyme expression profile in these cells combined with the use of selective inhibitors pointed to PKCα as the mediator of the effect. The demonstration that PKC agonists failed to inhibit AKT signaling in cell lines that lack expression of PKCα, and that expression of exogenous PKCα restored the effect, confirmed the involvement of PKCα. The inhibition of AKT by PKCα is dependent on PP2A since okadaic acid and calyculin A, but not the PHLPP1/2 inhibitors NSC117079 and NSC45586, blocked the effect, a finding consistent with the ability of PKCα to inhibit AKT activity in PTEN mutant cell lines. Taken together, our study provides evidence that 1) loss of PKCα is a common and early event in endometrial cancer; 2) PKCα plays a crucial role in regulating AKT activation and growth promoting signaling molecules in endometrial cancer, and 3) effects of PKCα on AKT in endometrial cancer are mediated by PP2A. Given the importance of PI3K/AKT in the disease, these findings highlight the potential of PKCα signaling as a potential biomarker for disease risk and as a potential therapeutic target in endometrial cancer. Supported by NIH grants CA036727, CA016056, and DK60632. Citation Format: Alice H. Hsu, Kathryn J. Curry, Kang-Sup Shim, Peter Frederick, Carl Morrison, Baojing Chen, Subodh M. Lele, Takiko Daikoku, Sudhansu K. Dey, Gustavo Leone, Adrian R. Black, Jennifer D. Black. Protein kinase C alpha (PKCα) regulates PI3K/AKT signaling in endometrial cancer. [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 4720. doi:10.1158/1538-7445.AM2015-4720
    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 ...
  • 6
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 189-189
    Abstract: Endometrial cancer (EC) is the most common gynecological malignancy and the fourth leading cancer in women in the United States. Hyperactivation of the PI3K/AKT pathway is observed in ∼90% of EC cases and this pathway is the most prominent driver of endometrial carcinogenesis. Mutations in multiple PI3K/AKT pathway components (including PTEN, PIK3CA, PIK3R1, AKT) often coexist in EC, indicating that EC development requires full, unopposed activation of PI3K/AKT signaling. In this study, we identified PKCα as a negative regulator of AKT activation that suppresses endometrial carcinogenesis. Our analysis revealed that PKCα protein and mRNA are reduced or lost in 50% of EC cell lines and ∼60% of human ECs. PKCα deficiency was also observed in hyperplastic endometrial lesions in murine models with allelic knock-in of mutant Pten (ptenΔ4-5/+, ptenC124R/+ and ptenC129E/+) or endometrial-specific Pten deletion (ptenpr-/-, ptenltf-/-), indicating that loss of PKCα is an early event in endometrial carcinogenesis. Mechanistic analysis has further revealed that loss of PKCα is independent of changes in PI3K/AKT signaling; however, the enzyme acts as a potent negative regulator of the PI3K/AKT pathway in EC cells. PKC agonists reduced AKT phosphorylation/activity in EC cells that retain PKCα, via a PP2A-dependent mechanism. Conversely, PKCα knockdown led to increased AKT phosphorylation. Notably, while PKC agonists did not affect AKT in EC cells with loss of PKCα, exogenous expression of the enzyme in these cells restored the inhibitory effect of the agonists on PI3K/AKT signaling. Collectively these data indicate that PKCα has a restraining effect on PI3K/AKT signaling in EC cells, providing a basis for its loss during endometrial tumor progression. Physiological relevance of these findings was established using cell lines, patient samples and mouse models. PKCα restoration in human EC cells decreased expression of oncogenes such as cyclin D1 and Id1 and abolished their anchorage-independent growth, supporting a tumor suppressive role for the isozyme in this tissue. Analysis of PKCα expression in patient specimens determined that loss of PKCα expression correlates with high grade disease. Finally, in mice heterozygous for mutant Pten, PKCα knockout led to a 3-fold increase in endometrial tumor burden at three months, confirming a role of PKCα as a tumor suppressor in the endometrium. Taken together, our results provide evidence that PKCα 1) negatively regulates AKT signaling and the expression of important oncogenes in EC cells; and 2) functions as tumor suppressor in the endometrium whose loss is associated with disease progression. Thus, PKCα signaling represents a promising biomarker for risk stratification in early stage disease and may provide insight into therapeutic strategies for late stage disease. Supported by NIH grants CA036727, CA016056, and DK60632. Citation Format: Alice H. Hsu, Kathryn J. Curry, Kang-Sup Shim, Peter Frederick, Carl D. Morrison, Baojiang Chen, Subodh M. Lele, Takiko Daikoku, Sudhansu K. Dey, Gustavo Leone, Adrian R. Black, Jennifer D. Black. Protein kinase C alpha suppresses AKT activation in endometrial cancer. [abstract]. In: Proceedings of the 107th Annual Meeting of the American Association for Cancer Research; 2016 Apr 16-20; New Orleans, LA. Philadelphia (PA): AACR; Cancer Res 2016;76(14 Suppl):Abstract nr 189.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2016
    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...