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: Biomedicines, MDPI AG, Vol. 10, No. 2 ( 2022-01-23), p. 238-
    Abstract: Platelet-rich plasma is a promising regenerative therapeutic with controversial efficacy. We and others have previously demonstrated regenerative functions of human platelet lysate (HPL) as an alternative platelet-derived product. Here we separated extracellular vesicles (EVs) from soluble factors of HPL to understand the mode of action during skin-organoid formation and immune modulation as model systems for tissue regeneration. HPL-EVs were isolated by tangential-flow filtration (TFF) and further purified by size-exclusion chromatography (SEC) separating EVs from (lipo)protein-enriched soluble fractions. We characterized samples by tunable resistive pulse sensing, western blot, tandem mass-tag proteomics and super-resolution microscopy. We evaluated EV function during angiogenesis, wound healing, organoid formation and immune modulation. We characterized EV enrichment by TFF and SEC according to MISEV2018 guidelines. Proteomics showed three major clusters of protein composition separating TSEC-EVs from HPL clustering with TFF soluble fractions and TFF-EVs clustering with TSEC soluble fractions, respectively. HPL-derived TFF-EVs promoted skin-organoid formation and inhibited T-cell proliferation more efficiently than TSEC-EVs or TSEC-soluble fractions. Recombining TSEC-EVs with TSEC soluble fractions re-capitulated TFF-EV effects. Zeta potential and super-resolution imaging further evidenced protein corona formation on TFF-EVs. Corona depletion on SEC-EVs could be artificially reconstituted by TSEC late fraction add-back. In contrast to synthetic nanoparticles, which commonly experience reduced function after corona formation, the corona-bearing EVs displayed improved functionality. We conclude that permissive isolation technology, such as TFF, and better understanding of the mechanism of EV corona function are required to realize the complete potential of platelet-based regenerative therapies.
    Type of Medium: Online Resource
    ISSN: 2227-9059
    Language: English
    Publisher: MDPI AG
    Publication Date: 2022
    detail.hit.zdb_id: 2720867-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2013
    In:  Cancer Prevention Research Vol. 6, No. 11_Supplement ( 2013-11-01), p. C38-C38
    In: Cancer Prevention Research, American Association for Cancer Research (AACR), Vol. 6, No. 11_Supplement ( 2013-11-01), p. C38-C38
    Abstract: Background: Colorectal cancer (CRC) is the third most common cancer among American adults and a leading cause of cancer-related mortality. The initiation and progression of colorectal tumorigenesis is characterized by genetic alterations that promote expression of tumor-promoting genes such as COX-2. In non-transformed cells, the mRNAs of these genes are often targeted for rapid decay through AU-rich elements (ARE) present in their 3'UTR. However, ARE-mediated mRNA decay is compromised during colorectal tumorigenesis, allowing for pathogenic gene overexpression. Our prior work identified the RNA-binding protein HuR to be overexpressed during colon tumorigenesis. In this capacity, HuR promotes COX-2 expression by interfering with rapid mRNA degradation and is associated with poor clinical prognosis. In the present study, we examined the therapeutic potential of inhibiting HuR using the small molecule MS-444. Methods: HCT116, HCA-7, SW480, RKO, and HT-29 colon cancer cells along with non-transformed intestinal epithelial (RIE-1) and colonocytes (YAMC) were used. MS-444 was obtained from Novartis. Cell proliferation and apoptosis were assayed by MTT assay and flow cytometry. HuR trafficking was visualized by immunofluorescence. qPCR, western blot, and immunohistochemistry were performed to assess changes in gene expression. In vivo studies were performed to xenograft tumor models of colon cancer cells using IVIS-based imaging for COX-2. Results: Treatment of the CRC cells lines HCT116, HCA-7, SW480, RKO, and HT-29 with MS-444 resulted in growth inhibition with IC50 values ranging between 5.6-12.8 μM, and this observed growth inhibition resulted from increased cell apoptosis. Importantly, treatment of non-transformed intestinal and colonic epithelial cells with MS-444 exhibited IC50 values of 40.7μM and 28.1 μM, respectively, and were resistant to MS-444-induced apoptosis. The expression of HuR was not impacted by MS-444, whereas MS-444 altered HuR's presence in the cytoplasm. HuR performs its mRNA stabilization function when present in the cytoplasm, and CRC cells treated with MS-444 displayed nuclear HuR as a result inhibition of HuR cytoplasmic trafficking. Consistent with growth-inhibitory IC50 values, MS-444 inhibited COX-2 mRNA and protein expression in CRC cells. This inhibition required the COX-2 ARE-containing 3'UTR and occurred on a post-transcriptional level. In vivo, delivery of MS-444 resulted in xenograft CRC tumor growth inhibition and prevented HuR cytoplasmic localization. Furthermore, treatment of MS-444 inhibited tumor COX-2 overexpression. Conclusions: These findings indicate that small molecule-based inhibition of HuR results in selective CRC cell death in part by promoting COX-2 mRNA degradation, while also indicating that HuR is a necessary component for colorectal tumor cell survival. Citation Format: Fernando F. Blanco, Yuan Li, Nicole Meisner-Kober, Shrikant Anant, Dan A. Dixon. Small molecule targeting of the mRNA stability factor HuR impacts COX-2 expression in colon cancer. [abstract] . In: Proceedings of the Twelfth Annual AACR International Conference on Frontiers in Cancer Prevention Research; 2013 Oct 27-30; National Harbor, MD. Philadelphia (PA): AACR; Can Prev Res 2013;6(11 Suppl): Abstract nr C38.
    Type of Medium: Online Resource
    ISSN: 1940-6207 , 1940-6215
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2013
    detail.hit.zdb_id: 2422346-3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    MDPI AG ; 2020
    In:  Microorganisms Vol. 8, No. 9 ( 2020-08-31), p. 1328-
    In: Microorganisms, MDPI AG, Vol. 8, No. 9 ( 2020-08-31), p. 1328-
    Abstract: Persistent infections with the human pathogen Helicobacter pylori (H. pylori) have been closely associated with the induction and progression of a wide range of gastric disorders, including acute and chronic gastritis, ulceration in the stomach and duodenum, mucosa-associated lymphoid tissue (MALT) lymphoma, and gastric adenocarcinoma. The pathogenesis of H. pylori is determined by a complicated network of manifold mechanisms of pathogen–host interactions, which involves a coordinated interplay of H. pylori pathogenicity and virulence factors with host cells. While these molecular and cellular mechanisms have been intensively investigated to date, the knowledge about outer membrane vesicles (OMVs) derived from H. pylori and their implication in bacterial pathogenesis is not well developed. In this review, we summarize the current knowledge on H. pylori-derived OMVs.
    Type of Medium: Online Resource
    ISSN: 2076-2607
    Language: English
    Publisher: MDPI AG
    Publication Date: 2020
    detail.hit.zdb_id: 2720891-6
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Journal of Extracellular Biology, Wiley, Vol. 1, No. 10 ( 2022-10)
    Abstract: Extracellular vesicles (EVs) large‐scale production is a crucial point for the translation of EVs from discovery to application of EV‐based products. In October 2021, the International Society for Extracellular Vesicles (ISEV), along with support by the FET‐OPEN projects, “The Extracellular Vesicle Foundry” (evFOUNDRY) and “Extracellular vesicles from a natural source for tailor‐made nanomaterials” (VES4US), organized a workshop entitled “massivEVs” to discuss the potential challenges for translation of EV‐based products. This report gives an overview of the topics discussed during “massivEVs”, the most important points raised, and the points of consensus reached after discussion among academia and industry representatives. Overall, the review of the existing EV manufacturing, upscaling challenges and directions for their resolution highlighted in the workshop painted an optimistic future for the expanding EV field.
    Type of Medium: Online Resource
    ISSN: 2768-2811 , 2768-2811
    URL: Issue
    Language: English
    Publisher: Wiley
    Publication Date: 2022
    detail.hit.zdb_id: 3120403-X
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Journal of Controlled Release, Elsevier BV, Vol. 361 ( 2023-09), p. 694-716
    Type of Medium: Online Resource
    ISSN: 0168-3659
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 1482453-X
    SSG: 15,3
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    In: Clinical Cancer Research, American Association for Cancer Research (AACR), Vol. 21, No. 4_Supplement ( 2015-02-15), p. A05-A05
    Abstract: Pancreatic ductal adenocarcinoma (PDA) is expected to become the 2nd leading cause of cancer-related mortality in the United States by 2020. One of the reasons PDA remains one of the most lethal malignancies is due to drug resistance mechanisms to both conventional and targeted chemotherapeutic regimens. Previously, it has been shown that PDA tumors exhibit high levels of hypoxia, characterized by low oxygen pressure (pO2) and decreased O2 intracellular perfusion. Importantly, chronic hypoxia is strongly associated with resistance to cytotoxic chemotherapy and chemoradiation in a widely understudied phenomenon known as hypoxia-induced chemoresistance. Recently, the hypoxia-inducible, serine-threonine, pro-oncogenic kinase PIM1 has emerged as a key regulator of hypoxia-induced chemoresistance in PDA and other cancers. While its role in therapeutic resistance has been previously described, the molecular mechanism(s) behind PIM1 overexpression in PDA is unknown. Here, we demonstrate that cis-acting AU-rich elements (ARE) present in the PIM1 mRNA 3′UTR mediate a regulatory interaction with the mRNA-stability factor HuR in the context of tumor hypoxia. We have previously demonstrated that HuR, a predominantly nuclear protein, translocates to the cytoplasm upon acute cancer-associated stress and selectively stabilizes key survival-associated transcripts. Here, we demonstrate by immunofluorescence and western blot analyses of fractionated lysates that PDA cells in hypoxic conditions mobilize HuR to the cytoplasm, where it binds and stabilizes the PIM1 transcript. The functional consequence of this interaction is observed with a 5-fold increase in PIM1 protein expression (p≤0.001). PIM1 imparts its oncogenic role by phosphorylating and inactivating key apoptotic proteins (e.g., BAD) and proteins involved in the DNA repair pathway. In this regard, siRNA-mediated knockdown of HuR abrogates hypoxia-induced PIM1 expression and its downstream phosphorylation of the pro-apoptotic protein BAD. As a result, cells undergo growth arrest and succumb to apoptosis. Most importantly, we demonstrate that HuR's regulation of PIM1 modulates a potent chemoresistance phenotype. HuR-mediated stabilization of PIM1 promoted dramatic resistance to 5-Fluorouracil (5-FU) and Oxaliplatin, two critical components of the promising combination therapy FOLFIRINOX. We employed confocal microscopy to analyze DNA damage foci formation by tracking and quantifying γH2AX foci in response to 5-FU and oxaliplatin. As a result of PIM1 up-regulation in hypoxia, or forced PIM1 overexpression in normoxia, PDA cells exhibit fewer DNA damage foci (p≤0.01) due to PIM1's ability to regulate DNA repair. From a translational standpoint, we demonstrate that disruption of the HuR:PIM1 axis by HuR knockdown results in PDA cell growth inhibition and enhances sensitivity to cytotoxic chemotherapeutic agents (5-FU and oxaliplatin). Similarly, pharmacological inhibition of HuR by MS-444 (Novartis), a small molecule that inhibits HuR homodimerization and its cytoplasmic translocation, abrogates hypoxia-induced PIM1 overexpression and dramatically enhances PDA cell sensitivity to oxaliplatin and 5-FU under physiologic low oxygen conditions. Taken together, these results support the notion that HuR has pro-oncogenic properties in PDA cells by enabling them with selective growth advantages in stressful tumor microenvironment niches. Accordingly, our studies provide evidence that therapeutic disruption of HuR's regulation of PIM1 may be a key strategy in breaking an elusive chemotherapeutic resistance mechanism acquired by PDA cells that reside in a hypoxic tumor microenvironment. Citation Format: Fernando F. Blanco, Nicole C. Meisner-Kober, Eric Londin, Isidore Rigoutsos, Makarand V. Risbud, Charles J. Yeo, Jordan M. Winter, Jonathan R. Brody. The mRNA-binding protein HuR promotes hypoxia-induced chemoresistance through post-transcriptional regulation of the serine-threonine kinase PIM1. [abstract]. In: Proceedings of the AACR Precision Medicine Series: Drug Sensitivity and Resistance: Improving Cancer Therapy; Jun 18-21, 2014; Orlando, FL. Philadelphia (PA): AACR; Clin Cancer Res 2015;21(4 Suppl): Abstract nr A05.
    Type of Medium: Online Resource
    ISSN: 1078-0432 , 1557-3265
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2015
    detail.hit.zdb_id: 1225457-5
    detail.hit.zdb_id: 2036787-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2015
    In:  Cancer Research Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5476-5476
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 75, No. 15_Supplement ( 2015-08-01), p. 5476-5476
    Abstract: Introduction: Pancreatic ductal adenocarcinoma (PDA) is the 4th leading cause of cancer-related deaths in the United States, and the 3rd most common cancer associated with BRCA mutations. Frontline therapies have significant toxicities and only minimally extend overall survival, highlighting the need to optimize targeted therapies. Poly-ADP ribose polymerase (PARP) inhibitors (PARPi), a ‘poster child’ for personalized medicine, depend on the concept of synthetic lethality where the combined perturbation of DNA repair genes, via genetic mutations within the tumor cells, and pharmacological PARP inhibition effectively targets BRCA-deficient tumors. Although PARPi have delivered promising preclinical and clinical results, initially- responsive patients ultimately develop resistance. A unique mechanism elucidated by our lab demonstrates that the mRNA-binding protein HuR mediates resistance to DNA damaging agents through post-transcriptional regulation of select mRNA cargo. Predominantly expressed in the nucleus, HuR translocates to the cytoplasm upon tumor-associated stress. Cytoplasmic HuR binds and stabilizes unique pro-survival transcripts, resulting in resistance to a harsh tumor microenvironment. Here, we sought to evaluate the role of HuR in regulating PARPi response. Methods and Results: Through immunofluorescence and western blot of fractionated lysates, we demonstrate that the PARP inhibitors Veliparib, Olaparib, and Rucaparib induced cytoplasmic HuR localization. Conversely, pre-treatment with MS-444 (Novartis), an established small molecule inhibitor of HuR, abrogated its nuclear export induced by PARPi treatment. Consistent with these findings, the growth-inhibitory effects of PARPi treatment were significantly potentiated upon HuR silencing whereas ectopic HuR overexpression promoted resistance, as observed in short term cell survival and long-term anchorage-independent growth assays. Additionally, silencing of HuR enhanced PARPi-induced cytotoxicity, assessed by increased accumulation of DNA damage (γH2Ax) foci and Poly ADP-ribose (PAR) polymers. Ribonucleotide protein immunoprecipitation (RNP-IP) assays demonstrated that HuR binds and upregulates Poly-ADP Ribose Glycohydrolase (PARG) mRNA, the major enzyme responsible for catabolism of PAR. Taken together, when PDA cells are exposed to PARPi, HuR mediates upregulation of PARG, thereby decreasing PARylation and facilitating DNA repair. Conversely, HuR inhibition results in detrimental accumulation of PAR and enhanced DNA damage, which ultimately leads to increased PARPi-conferred cytotoxicity. Discussion: These results demonstrate that HuR imposes a significant barrier to PARPi therapy by orchestrating a strong chemoresistance mechanism. Thus, we provide evidence that HuR (and/or its target) inhibition via an HuR inhibitor (MS-444) can optimize PARPi-based therapies for better patient outcomes. Citation Format: Saswati N. Chand, Akshay R. Kamath, Nicole Meisner-Kober, Charles J. Yeo, Jordan M. Winter, Jonathan R. Brody. A novel PARP inhibitor resistance mechanism mediated by the RNA-binding protein HuR. [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 5476. doi:10.1158/1538-7445.AM2015-5476
    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 ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 77, No. 18 ( 2017-09-15), p. 5011-5025
    Abstract: The majority of pancreatic ductal adenocarcinomas (PDAC) rely on the mRNA stability factor HuR (ELAV-L1) to drive cancer growth and progression. Here, we show that CRISPR-Cas9–mediated silencing of the HuR locus increases the relative sensitivity of PDAC cells to PARP inhibitors (PARPi). PDAC cells treated with PARPi stimulated translocation of HuR from the nucleus to the cytoplasm, specifically promoting stabilization of a new target, poly (ADP-ribose) glycohydrolase (PARG) mRNA, by binding a unique sequence embedded in its 3′ untranslated region. HuR-dependent upregulation of PARG expression facilitated DNA repair via hydrolysis of polyADP-ribose on related repair proteins. Accordingly, strategies to inhibit HuR directly promoted DNA damage accumulation, inefficient PAR removal, and persistent PARP-1 residency on chromatin (PARP-1 trapping). Immunoprecipitation assays demonstrated that the PARP-1 protein binds and posttranslationally modifies HuR in PARPi-treated PDAC cells. In a mouse xenograft model of human PDAC, PARPi monotherapy combined with targeted silencing of HuR significantly reduced tumor growth compared with PARPi therapy alone. Our results highlight the HuR–PARG axis as an opportunity to enhance PARPi-based therapies. Cancer Res; 77(18); 5011–25. ©2017 AACR.
    Type of Medium: Online Resource
    ISSN: 0008-5472 , 1538-7445
    RVK:
    RVK:
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2017
    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. 75, No. 15_Supplement ( 2015-08-01), p. 2113-2113
    Abstract: Background: PDA will become the 2nd leading cause of cancer-related mortality in the US by 2020. A recent Phase III randomized controlled trial revealed a 4 month overall survival benefit in metastatic PDA with FOLFIRINOX (Folinic acid, 5-Fluorouracil (5FU), Irinotecan, and Oxaliplatin) compared to gemcitabine, the standard of care. However, the long-term clinical efficacy of FOLFIRINOX and other chemotherapy regimens are limited by tumor-associated drug resistance driven by factors in the tumor microenvironment (e.g., hypoxia). We identified a novel drug resistance mechanism driven by the hypoxia-inducible pro-oncogenic kinase PIM1 and regulated by the RNA binding protein HuR. Herein, we launched into developing a strategy to target this tractable mechanism in an effort to optimize current therapeutic treatments for PDA. Methods: To model hypoxia, PDA cells were incubated in 1% O2 and responses to 5FU or oxaliplatin assessed to obtain IC50 doses. Stabilizing interactions between the RNA-binding protein HuR and PIM1 mRNA were quantified in vitro through binding assays, and confirmed in patients by immunohistochemistry in resected PDAs (n = 44). The contribution of HuR-mediated regulation of PIM1 to resistance to 5FU or oxaliplatin in hypoxia was examined using MS-444 (Novartis), a low-molecular-weight HuR inhibitor. Results: In response to hypoxia, HuR translocates from the nucleus to the cytoplasm where it binds and stabilizes the PIM1 mRNA transcript, thus amplifying PIM1 translation and protein expression. Clinically, we identified a positive correlation (p = 0.011) between cytoplasmic HuR and PIM1 protein expression in a cohort of PDA patients from our institution. In vitro mechanistic studies demonstrated that hypoxia-mediated induction of PIM1 overexpression enhanced DNA repair and evaded the apoptotic response elicited by hypoxic stress. Targeted inhibition of HuR by the HuR inhibitor MS-444 abrogated hypoxia-induced PIM1 overexpression, enhancing PDA cell sensitivity to oxaliplatin and 5FU (P & lt;0.001). Conclusion: The mRNA-stability factor HuR post-transcriptionally induces PIM1 expression under hypoxic conditions, and thereby promotes hypoxia-induced chemoresistance. Ongoing pre-clinical studies will evaluate pharmacologic inhibition of HuR's regulation of PIM1 (e.g., MS-444) as a novel modality to enhance the therapeutic value of FOLFIRINOX for the treatment of metastatic PDA. Citation Format: Fernando F. Blanco, Masaya Jimbo, Liz Enyenihi, Nicole Meisner-Kober, Eric Londin, Isidore Rigoutsos, Makarand Risbud, Peter McCue, Charles Yeo, Jordan Winter, Jonathan R. Brody. Targeting tumor-associated hypoxia to overcome chemoresistance in pancreatic ductal adenocarcinoma (PDA). [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 2113. doi:10.1158/1538-7445.AM2015-2113
    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 ...
  • 10
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 14_Supplement ( 2016-07-15), p. 1995-1995
    Abstract: Introduction: Despite our deep understanding of genetic drivers of the disease, pancreatic ductal adenocarcinoma (PDA) continues to be associated with dismal survival rates. Targeting the DNA repair machinery has emerged as a promising therapeutic strategy to treat pancreatic cancer patients carrying DNA damage repair (DDR) mutations. Such mutations promote tumorigenesis, but also paradoxically render tumor cells particularly susceptible to platinum-based agents and PARP inhibitors (PARPi). However, despite promising preclinical and clinical results, early data demonstrate that eventually most tumors, regardless of DDR status, become resistant to PARPi-therapies. The mRNA-binding protein HuR, predominantly expressed in the nucleus, translocates to the cytoplasm upon tumor-associated stress where it post-transcriptionally regulates select mRNA cargo, resulting in resistance to DNA damaging agents in a harsh tumor microenvironment. Here, we sought to evaluate the role of HuR in regulating PARPi efficacy. Results: In response to cellular stress induced by IC50 dosages of a panel of PARPis such as Veliparib, Olaparib, Rucaparib, Talazoparib and Niraparib, nuclear localized HuR undergoes cytoplasmic translocation. Silencing of HuR via siRNA, CRISPR and a DOX-inducible system resulted in significant decrease in long and short- term PDA cell survival, irrespective of DDR status. To complement and validate in vitro findings, we employed a heterotropic mouse xenograft model using Mia.sh290 cells wherein DOX induction significantly reduced HuR expression. Olaparib mediated PARP inhibition (50mg/kg, 5 days a week) combined with DOX-induced HuR silencing resulted in significant reduction in tumor volumes, compared to Olaparib alone or DOX alone. Mechanistically, we demonstrate that the pro-survival protein HuR facilitates PDA cells to recover from PARPi insult by, in part, regulating poly ADP ribose glycohydrolase (PARG), the major enzyme responsible for hydrolyzing poly-ADP ribose (PAR) polymers, on chromatin and associated proteins. HuR binds to two 41- 43bp long sites in the 3’ untranslated region (3’UTR) of PARG, increasing its mRNA stability and protein expression. Increased PARG activity, further validated via exogenous overexpression, promotes DNA repair efficiency and increases PDA cell survival. Functional analysis indicate that such inhibition of HuR and/or PARG significantly enhances PARPi sensitivity in PDA cells, via increased accumulation of DNA damage γH2AX foci, preventing efficient removal of PAR polymers, and enhancing detrimental trapping of PARP1 on chromatin. Conclusions: Taken together, our results indicate that HuR- mediated upregulation of PARG acts as a universal pro-survival mechanism and HuR inhibition could significantly potentiate PARPi therapy in PDA, irrespective of DNA repair status. Citation Format: Saswati N. Chand, Mahsa Zarei, Akshay R. Kamath, Matthew J. Schiewer, Carmella Romeo, Joseph A. Cozzitorto, Nicole Meisner- Kober, Eric Londin, Isidore Rigoutsos, Karen Knudsen, John M. Pascal, Charles J. Yeo, Jordan M. Winter, Jonathan R. Brody. HuR dependent inhibition of PARG enhances PARP inhibitor therapy for DNA repair proficient and deficient pancreatic 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 1995.
    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...