GLORIA

GEOMAR Library Ocean Research Information Access

Ihre E-Mail wurde erfolgreich gesendet. Bitte prüfen Sie Ihren Maileingang.

Leider ist ein Fehler beim E-Mail-Versand aufgetreten. Bitte versuchen Sie es erneut.

Vorgang fortführen?

Exportieren
Filter
  • American Association for Cancer Research (AACR)  (3)
  • Nimmakayala, Rama Krishna  (3)
Materialart
Verlag/Herausgeber
  • American Association for Cancer Research (AACR)  (3)
Sprache
Erscheinungszeitraum
Fachgebiete(RVK)
  • 1
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 4438-4438
    Kurzfassung: Background: RNA polymerase associated factor 1 (PAF1)/Pancreatic differentiation 2 (PD2) is one of the core subunit of the human PAF1 complex (PAF1C), which regulates various cellular functions such as transcriptional elongation and histone modification. We have previously demonstrated its unique role in oncogenesis and stem cell maintenance. Studies have demonstrated that PAF1/PD2 gene yields a protein of 59.9 Kda (531 amino acids), however it has been found that it always gives a band at 80 Kda. Further, previous studies suggests that the 60 Kda protein represents the precursor, which rapidly process into an 80 Kda mature protein. SUMOylation is a process of reversible posttranslational modification that adds a small ubiquitin-related modifier (SUMO)-1 protein to the target protein. SUMOylation plays various molecular biology function such as transcriptional regulation, protein-protein interaction, and DNA damage repair and in cell cycle. DNA damage happen due to several physiological processes, but can also be caused by genotoxic agents. Promyelocytic Leukemia (PML) is protein that forms nuclear bodies and may be modified by SUMO1 and act as a DNA-damage sensor. Hypothesis: PAF1/PD2 is interacting with SUMO-1 and PML and, thus, plays an important function in providing gemcitabine resistance to pancreatic cancer cells Methods: SW1990, F9 (mouse embryonic cells) and CD18/HPAF cells were treated with 2-D08, a potent SUMOylation inhibitor for 24 hours and with siRNA against SUMO1 for 48 hours followed by protein isolation and western blotting. Immunoprecipitation and immunofluorescence were done to show the interaction between PAF1/PD2 and SUMO1 and with PML. To study the effect of PAF1/PD2 on gemcitabine resistance, SW1990 and Capan-1 cells were treated with different concentration of gemcitabine and then the expression of PAF1/PD2 along with SUMO1 was checked through immunoblotting and confocal imaging. Results: Results shows that inhibiting SUMOylation with both 2D08 and siRNA resulted in a decrease expression of PAF1/PD2 80 Kda protein. Immunoprecipitation and immunofluorescence analysis showed that endogenous PAF1/PD2 interacts with SUMO1. This finding was further verified using ectopically overexpressed Flag- tagged PAF1/PD2 and HA-tagged SUMO1, which showed a physical interaction between PAF1/PD2 and SUMO1. Interestingly, we observed that gemcitabine treatment significantly increased the SUMOylated status of PAF1/PD2 in pancreatic cancer cells. Further our results proved that SUMOylated PAF1/PD2 form nuclear bodies along with PML in pancreatic cancer cells. Conclusions: Our observation suggest that PAF1/PD2 undergoes SUMOylation and covalently interacts with SUMO1. Treatment with gemcitabine results in enhanced expression of PAF1/PD2 and increased co-localization with SUMO1 and PML, indicates a role of SUMOylated PAF1/PD2 in gemcitabine resistance. Citation Format: Sanchita Rauth, Saswati Karmakar, Ashu Shah, Rama Krishna Nimmakayala, Rakesh Bhatia, Sakthivel Muniyan, Sushil Kumar, Samikshan Dutta, Kaustubh Datta, Surinder K. Batra, Moorthy Palanimuthu Ponnusamy. Role of post translational modification of PAF1/PD2 in gemcitabine resistance of pancreatic cancer [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 4438.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 2
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 78, No. 13_Supplement ( 2018-07-01), p. 5897-5897
    Kurzfassung: Background: Docetaxel remains the first-line therapeutic intervention for metastatic and castration-resistant (CR) prostate cancer (PCa). However, the therapeutic efficacy is limited due to unresponsiveness, toxicity, and drug-resistance. The availability of additional therapies increases the PCa patient survival modestly, but the development of cross-resistance limit the therapeutic efficacy. Hence, there is a need to understand the mechanisms of resistance and identify a novel target for the better management of CR PCa patients. Methods: Docetaxel resistant 22Rv1, LNCaP C-81, and PC3 PCa cells were established, and its resistance phenotype was determined by cell growth inhibition (MTT assay), apoptosis markers (western blot), tumor sphere assay, drug efflux property (SP analysis by FACS), auto fluorescence (FACS). qRT-PCR, western blot, and confocal microscopic analysis were also performed to confirm the drug-resistant marker phenotype. Additional experiments were performed to determine the underlying molecular mechanisms of docetaxel resistance. Results: The docetaxel resistant PCa cells grew well at the higher concentration of docetaxel (120, 15 and 50 nM of docetaxel, respectively) and did not result in apoptosis as measured by cPARP and caspase-3 cleavage. Docetaxel resistant PCa cells confer cross-resistance to second-generation chemotherapeutic agent cabazitaxel and show altered cell proliferation and invasion. Blocking by the ABCB1 specific inhibitor enhances docetaxel-induced cell death on par with parental cells. Side population analysis by flow cytometry confirms the acquired drug efflux property. Side population, autofluorescence and tumor sphere analyses confirmed the drug-resistance and stem-like cell phenotype. qRT-PCR, western blot and confocal microscopy show the abundant expression levels of the drug transporters, ABCB1 and ABCG2. Further, the docetaxel resistant PCa cells show higher stem cell network proteins such as PAF1, POU5F1, NANOG and SOX9 expression levels compared to age-matched control cells. Conversely, Tet-inducible PAF1 knockdown reduces embryonic stem cell network proteins and reverses docetaxel-resistance phenotype. Conclusions: Collectively, our study suggests that the stem cell factors such as PAF1 play a major role in docetaxel resistance and aggressiveness to PCa cells. Understanding the associated mechanisms and targeting these factors could lead a better management approach for CRPCa patients. Citation Format: Sakthivel Muniyan, Rama Krishna Nimmakayala, Saswati Karmakar, Satyanarayana Rachagani, Jawed A. Siddiqui, Parthasarathy Seshacharyulu, Ming-Fong Lin, Kaustubh Datta, Moorthy P. Ponnusamy, Surinder K. Batra. Role of polymerase II associated factor 1, PAF1, in docetaxel resistant prostate cancer cells [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 5897.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2018
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
  • 3
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 13_Supplement ( 2019-07-01), p. 3679-3679
    Kurzfassung: Background: Cancer stem cells (CSCs), a minor subset of cancer cells, mediate aggressiveness, metastasis, and drug resistance of many cancers including the deadly pancreatic cancer (PC). Molecular governance of pancreatic CSCs remains elusive, mandating the need for their better molecular characterization to improve management of PC. PD2 (Pancreatic Differentiation 2) or RNA Polymerase II-Associated Factor 1 (Paf1) is a core component of human PAF1 complex (PAF1C), which regulates transcription elongation and mRNA processing. Human PAF1C consists of 5 members: PAF1, LEO1, CDC73, CTR9 and SKI8. Recently, PAF1 has emerged as a novel pancreatic CSC marker that enhances tumorigenic and metastatic potential of PC. Paf1 also maintains the self-renewal of mouse embryonic stem cells and ovarian CSCs via its interaction with OCT3/4, a major regulator of pluripotency. However, the mechanistic role of PAF1 in CSC maintenance and CSC-mediated PC pathogenesis is poorly understood. Hypothesis: We hypothesize that “PAF1 forms a sub-complex exclusive of PAF1C, wherein PAF1 functions as the master-regulator for maintaining pancreatic CSCs by regulating stem cell gene signature”. Experimental Design: PAF1 knockdown (KD) was used to identify its targets and role in tumorigenesis. PAF1 sub-complex in CSCs was identified using immunoprecipitation (IP) and mass spectrometry (MS). ChIP-Seq was performed to confirm binding of PAF1 on its target genes. Results: Inducible KD of PAF1 led to a significant reduction in tumor burden from orthotopically implanted human PC cells in athymic nude mice, indicating its role in pancreatic tumorigenesis. CSCs from different PC cell lines demonstrated higher expression of PAF1 along with CSC markers and PAF1 KD caused a significant decrease in CSC and self-renewal markers analyzed through Western blotting and immunofluorescence. Reciprocal co-IP and MS revealed that PAF1 interacted with Phf5a, DDX3, and hnRNP-K in CSCs. PAF1 KD in CSCs showed significant down-regulation of tumorigenic and stemness maintenance genes by RNA-Seq and PCR array analysis. PAF1 along with its binding partners occupied Nanog promoter in pancreatic CSCs. Further, KD of PAF1 in CSCs did not affect the expression of other PAF1C components, whereas individual KD of single PAF1C components decreased other remaining PAF1C members including PAF1. Moreover, other PAF1C components did not interact with Phf5a, a PHD-fold harboring nuclear protein in CSCs, suggesting that PAF1 forms a sub-complex with Phf5a, independent of PAF1C that functions in CSC maintenance. Depletion of Paf1 from mouse pancreas since birth using a CRISPR/Cas9-based conditional Paf1 knockout mouse model severely affected exocrine pancreas development with peri-ductal sclerosis and inflammation. Conclusion: Altogether, PAF1 functions as the master-regulator for CSC maintenance by forming a sub-complex, which regulates CSC-network genes in PC. Citation Format: Saswati Karmakar, Sanchita Rauth, Rama Krishna Nimmakayala, Srikanth Barkeer, Mohd W. Nasser, Satyanarayana Rachagani, Dario Ghersi, Moorthy P. Ponnusamy, Surinder K. Batra. A novel mechanism for pancreatic cancer stem cell maintenance: PAF1C-independent role of PAF1 [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 3679.
    Materialart: Online-Ressource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Sprache: Englisch
    Verlag: American Association for Cancer Research (AACR)
    Publikationsdatum: 2019
    ZDB Id: 2036785-5
    ZDB Id: 1432-1
    ZDB Id: 410466-3
    Standort Signatur Einschränkungen Verfügbarkeit
    BibTip Andere fanden auch interessant ...
Schließen ⊗
Diese Webseite nutzt Cookies und das Analyse-Tool Matomo. Weitere Informationen finden Sie hier...