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
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2023
    In:  Cancer Immunology Research Vol. 11, No. 12_Supplement ( 2023-12-01), p. PR03-PR03
    In: Cancer Immunology Research, American Association for Cancer Research (AACR), Vol. 11, No. 12_Supplement ( 2023-12-01), p. PR03-PR03
    Abstract: Pancreatic ductal adenocarcinoma (PDAC) is a deadly malignancy characterized by poor response to all existing therapies. Although immunotherapy has shown great promise against multiple deadly cancers, it has been largely ineffective in PDAC. This lack of response is in part attributed to its extensive, fibroinflammatory stroma and hypoxic microenvironment. Cancer-associated fibroblasts (CAFs) are a predominant and heterogeneous stromal cell type in PDAC, and single-cell transcriptomics of human and mouse PDAC has recently revealed a distinct CAF subpopulation, inflammatory CAFs (iCAFs). These inflammatory fibroblasts produce high levels of cytokines and chemokines in PDAC and have the potential to contribute to its immunosuppressive microenvironment and tumorigenesis. By injecting a hypoxia probe into PDAC mouse models, we recently found that iCAFs predominantly reside in hypoxic tumor regions. We also observed that the hypoxia-related gene signature is positively enriched in iCAFs in human PDAC samples. Importantly, by exposing three-dimensional (3D) co-cultures of pancreatic cancer cells and fibroblasts to either hypoxia or normoxia, we showed that hypoxia induces IL1α from cancer cells and that IL1α is required for hypoxia-mediated iCAF formation. Our data implicate hypoxia as a critical regulator of tumor stroma. Efforts are ongoing to understand the role of hypoxia in the crosstalk between cancer cells, fibroblasts, and immune cells in driving an immunosuppressive tumor microenvironment. Citation Format: Ashley M. Mello, Tenzin Ngodup, Yusoo Lee, Katelyn L. Donahue, Jinju Li, Arvind Rao, Eileen S. Carpenter, Howard C. Crawford, Marina Pasca Di Magliano, Kyoung Eun Lee. Hypoxia promotes inflammatory fibroblast formation in pancreatic cancer [abstract]. In: Proceedings of the AACR Special Conference in Cancer Research: Tumor Immunology and Immunotherapy; 2023 Oct 1-4; Toronto, Ontario, Canada. Philadelphia (PA): AACR; Cancer Immunol Res 2023;11(12 Suppl):Abstract nr PR03.
    Type of Medium: Online Resource
    ISSN: 2326-6074
    Language: English
    Publisher: American Association for Cancer Research (AACR)
    Publication Date: 2023
    detail.hit.zdb_id: 2732517-9
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 2
    In: Genes & Development, Cold Spring Harbor Laboratory, Vol. 35, No. 3-4 ( 2021-02-01), p. 218-233
    Abstract: Pancreatic ductal adenocarcinoma is a lethal disease characterized by late diagnosis, propensity for early metastasis and resistance to chemotherapy. Little is known about the mechanisms that drive innate therapeutic resistance in pancreatic cancer. The ataxia-telangiectasia group D-associated gene (ATDC) is overexpressed in pancreatic cancer and promotes tumor growth and metastasis. Our study reveals that increased ATDC levels protect cancer cells from reactive oxygen species (ROS) via stabilization of nuclear factor erythroid 2-related factor 2 (NRF2). Mechanistically, ATDC binds to Kelch-like ECH-associated protein 1 (KEAP1), the principal regulator of NRF2 degradation, and thereby prevents degradation of NRF2 resulting in activation of a NRF2-dependent transcriptional program, reduced intracellular ROS and enhanced chemoresistance. Our findings define a novel role of ATDC in regulating redox balance and chemotherapeutic resistance by modulating NRF2 activity.
    Type of Medium: Online Resource
    ISSN: 0890-9369 , 1549-5477
    RVK:
    Language: English
    Publisher: Cold Spring Harbor Laboratory
    Publication Date: 2021
    detail.hit.zdb_id: 1467414-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    Online Resource
    Online Resource
    Elsevier BV ; 2019
    In:  Cellular and Molecular Gastroenterology and Hepatology Vol. 8, No. 2 ( 2019), p. 173-192
    In: Cellular and Molecular Gastroenterology and Hepatology, Elsevier BV, Vol. 8, No. 2 ( 2019), p. 173-192
    Type of Medium: Online Resource
    ISSN: 2352-345X
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2019
    detail.hit.zdb_id: 2819778-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 76, No. 24_Supplement ( 2016-12-15), p. A62-A62
    Abstract: We have recently demonstrated that ATDC, a novel oncogenic protein, serves as an invasive switch in pancreatic cancer (PDA) by activation of beta–catenin signaling and upregulation of CD44, resulting in EMT and an invasive phenotype during PanIN progression. To further explore the tumorigenic function of ATDC, we generated a floxed ATDC mouse (A F/F) to evaluate the impact of conditional knockout of ATDC on oncogenic Kras-induced PDA initiation and progression. Pancreas-specific ATDC knockout did not cause any histologic abnormalities in pancreas, up to 1 year of age (n=8). Through a series of crosses of LSL-KrasG12D (K), p53F/+ (P), RosaYFP (Y), Pdx1-Cre (C) and AF/F mice, KrasG12D; CY (KCY); KrasG12D; p53+/-; CY (KPCY), KCYA-/- KPCYA-/- mice were generated. Knockout of ATDC in KPCY mice completely prevented the development of ADM and PanIN lesions in 3 month old mice (n= 8), and resulted in the formation of very rare ADM and PanIN1 lesions (2 out of 8) in KPCYA-/- mice at 12 months of age (n=8). In contrast, all KPCY mice developed extensive PanIN (low and high grade) at 3 months of age (n= 8), with the subsequent development of invasive and metastatic cancer at frequencies similar to that reported in the literature. To determine the possible mechanisms by which ATDC inhibited KrasG12D-induced acinar-ductal metaplasia (ADM), we isolated acini from 1.5 month old KCY and KCYA-/- pancreata and performed in vitro 3D cultures and ADM assays. ADM lesions readily formed in 3-D cultures of acini from KCY mice at 5 days, and this was significantly inhibited in acini isolated from KCYA-/- mice (duct-like structures: 95.1±3.5% to 28.0±2.2%*, KCY vs KCYA-/-, n=3, *p & lt;0.05). Expression of ATDC specific shRNA in acini from KCY mice also effectively decreased ADM formation in 3D culture, an effect that was completely reversed by ATDC overexpression using an ATDC-shRNA-resistant expression vector. To further evaluate the role of ATDC in ADM and PanIN formation, we induced caerulein-mediated acute pancreatitis in 1.5 month old WT, CYA-/-, KCY, KCYA-/- mice and analyzed pancreatic tissue 1 and 7 days following cerulein treatment. 1 day post-caerulein treatment, KCY, KCYA-/-, CYA-/- and WT mice exhibited widespread ADM, which was replaced by normal acini by 7 days in WT, CYA-/- and KCYA-/- mice. However, in KCY mice 7 days post-cerulein treatment, extensive ADM and PanIN lesions were present, suggesting that ATDC is required for oncogenic KRAS to promote ADM and PanIN formation. Conclusions: Knockout of ATDC markedly reduces KrasG12D-induced ADM and PanIN formation, highlighting a key biologic function for ATDC in this process and its role in driving progression of KRAS-induced tumorigenesis in the pancreas. Citation Format: Lidong Wang, Huibin Yang, Ethan V. Abel, Phillip L. Palmbos, Christophe Halbrook, Kenneth Takeuchi, Jiaqi Shi, Yaqing Zhang, Sumithra Urs, Meghna Waghray, Marina Pasca di Magliano, Andrew D. Rhim, Howard C. Crawford, Diane M. Simeone.{Authors}. ATDC is required for KRAS-induced pancreatic tumorigenesis. [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2016 May 12-15; Orlando, FL. Philadelphia (PA): AACR; Cancer Res 2016;76(24 Suppl):Abstract nr A62.
    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 ...
  • 5
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2019
    In:  Cancer Research Vol. 79, No. 24_Supplement ( 2019-12-15), p. C56-C56
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. C56-C56
    Abstract: Rare acinar cells expressing Bmi1, a component of the Polycomb Repressor Complex 1 (PRC1), represent a reservoir of cells contributing to pancreatic repair during injury and stress. This is marked by acinar-ductal metaplasia (ADM), a transition of acinar cells to a more duct-like phenotype, which is seen in pancreatitis and early stages of Kras-driven pancreatic neoplasia. The mechanism behind Kras-driven ADM at the genomic level is not completely understood. Developmental transcription factor (TF) networks regulate and maintain the fate of cells and rely on appropriate chromatin context for their activity. Deletion of Bmi1 represses Kras-driven ADM and neoplasia in genetically engineered mouse models (GEMMs) of PDA. Thus, we hypothesized that Kras reprograms the acinar fate TF network to promote PDA in a Bmi1-dependent epigenetic context. We used GEMMs to study the expression of Bmi1 in the acinar compartment and its role in early pancreatic neoplasia. We combined an acinar cell-specific tamoxifen-inducible Cre-recombinase under the elastase promoter (Ela-CreER) with the Kras G12D oncogenic allele and the conditional Bmi1 knockout allele and the Rosa26 tdTomato reporter to generate Ela-CreER, KrasLSL-G12D/+, Bmi1, R26 tdTomato mice and their littermate controls. We also generated Ela-CreER, Bmi1GFP, R26tdTomato and Bmi1-CreER, R26 tdTomato mice to quantify the fraction of acinar cells expressing Bmi1. We activated the Cre-recombinase in 6- to 8-week-old Ela-CreER mice with 5 tamoxifen gavages (4mg/day) prior to induction of acute pancreatitis by intraperitoneal caerulein administration (8 hourly injections x 2 days) one week after the first tamoxifen dose. We FACS sorted tdTomato+ cells from untreated mice (0 hr) and one week (168 hr) after pancreatitis induction and analyzed RNA levels of 8 TFs, reported to play a role in pancreatic development, homeostasis, and neoplasia. Expression levels of two lineage markers (amylase and elastase) were also measured by RT-qPCR. Lineage tracing in Bmi1-CreER and Ela-CreER, Bmi1GFP mice revealed expression of Bmi1 in vast majority of acinar cells. RT-qPCR of tdTomato+ cells in the Ela-CreER models indicated a loss of acinar-specific TFs in mice expressing mutant Kras one week after induction of pancreatitis consistent with cells undergoing ADM. Genetic ablation of Bmi1 in the presence of mutant Kras restored elastase and amylase mRNA levels. At the morphologic level, loss of Bmi1 also led to recovery of acinar cells from ADM and correlated with an increase in RNA expression of three key acinar-specific fate TFs—Mist1, Hnf1a, and Nr5a2. The mRNA levels of other key acinar TFs—Pdx1 and Ptf1a/p48—did not recover with the loss of Bmi1 in the presence of KrasG12D. Together, our data suggest that Bmi1 is expressed in the majority of acinar cells and regulates oncogenic Kras-driven ADM by altering master TF gene regulatory networks. Its deletion leads to partial reprogramming of these networks to allow acinar cells to resist Kras-driven oncogenesis. Citation Format: Joyce K. Thompson, Emily Wu, Osama Alkhalili, Howard C. Crawford, Marina Pasca di Magliano, Filip Bednar. Bmi1 is widely expressed in acini and regulates Kras-driven transcription factor networks in early pancreatic neoplasia [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr C56.
    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 ...
  • 6
    In: Nature Communications, Springer Science and Business Media LLC, Vol. 12, No. 1 ( 2021-08-11)
    Abstract: Cancer metabolism is rewired to support cell survival in response to intrinsic and environmental stressors. Identification of strategies to target these adaptions is an area of active research. We previously described a cytosolic aspartate aminotransaminase (GOT1)-driven pathway in pancreatic cancer used to maintain redox balance. Here, we sought to identify metabolic dependencies following GOT1 inhibition to exploit this feature of pancreatic cancer and to provide additional insight into regulation of redox metabolism. Using pharmacological methods, we identify cysteine, glutathione, and lipid antioxidant function as metabolic vulnerabilities following GOT1 withdrawal. We demonstrate that targeting any of these pathways triggers ferroptosis, an oxidative, iron-dependent form of cell death, in GOT1 knockdown cells. Mechanistically, we reveal that GOT1 inhibition represses mitochondrial metabolism and promotes a catabolic state. Consequently, we find that this enhances labile iron availability through autophagy, which potentiates the activity of ferroptotic stimuli. Overall, our study identifies a biochemical connection between GOT1, iron regulation, and ferroptosis.
    Type of Medium: Online Resource
    ISSN: 2041-1723
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2021
    detail.hit.zdb_id: 2553671-0
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    In: Genes & Development, Cold Spring Harbor Laboratory, Vol. 33, No. 11-12 ( 2019-06-01), p. 641-655
    Abstract: Pancreatic adenocarcinoma (PDA) is an aggressive disease driven by oncogenic KRAS and characterized by late diagnosis and therapeutic resistance. Here we show that deletion of the ataxia-telangiectasia group D-complementing ( Atdc ) gene, whose human homolog is up-regulated in the majority of pancreatic adenocarcinoma, completely prevents PDA development in the context of oncogenic KRAS. ATDC is required for KRAS-driven acinar–ductal metaplasia (ADM) and its progression to pancreatic intraepithelial neoplasia (PanIN). As a result, mice lacking ATDC are protected from developing PDA. Mechanistically, we show ATDC promotes ADM progression to PanIN through activation of β-catenin signaling and subsequent SOX9 up-regulation. These results provide new insight into PDA initiation and reveal ATDC as a potential target for preventing early tumor-initiating events.
    Type of Medium: Online Resource
    ISSN: 0890-9369 , 1549-5477
    RVK:
    Language: English
    Publisher: Cold Spring Harbor Laboratory
    Publication Date: 2019
    detail.hit.zdb_id: 1467414-2
    SSG: 12
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 8
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. I04-I04
    Abstract: In human patients and in mouse models, immunosuppressive regulatory T cells (Tregs) accumulate in pancreatic cancer. Tregs are considered a potential therapeutic target with the goal to reverse immunosuppression in a number of malignancies, including pancreatic cancer. Using a genetically engineered mouse model of pancreatic cancer, we determined that—unlike in other malignancies—Treg depletion failed to relieve immunosuppression. Contradicting the current paradigm, depletion of Tregs led to accelerated tumor progression. We show that Tregs are a key source of TGFb ligands both in human samples and in mouse models. Accordingly, their depletion reprograms the fibroblast population, exemplified by loss of smooth muscle actin expression and an increase in the myeloid-recruiting chemokines Ccl3, Ccl6, and Ccl8. Our findings point to a dual role of Tregs in pancreatic cancer, with their immune-suppressive function being balanced by their induction of a SMA-high (tumor-restraining) fibroblast phenotype. Further, our data point to a compensatory immunosuppression mechanism that confers resistance to Treg depletion in pancreatic cancer. Finally, blockade of the common CCL3/6/8 receptor CCR1 combined with Treg-depletion successfully inhibited tumorigenesis, a finding with potential therapeutic implications. Citation Format: Yaqing Zhang, Jenny Lazarus, Nina Steele, Ho-Joon Lee, Wei Yan, Cristopher J. Halbrook, Rosa Menjivar, Samantha B. Kemp, Veerin Sirihoracai, Eileen S. Carpenter, Anna C. Nevison, Alekya Vinta, Michelle A. Anderson, Howard C. Crawford, Costas A. Lyssiotis, Timothy L. Frankel, Filip Bednar, Marina Pasca di Magliano. Regulatory T-cell depletion causes compensatory immune suppression and accelerates pancreatic carcinogenesis [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr I04.
    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 ...
  • 9
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 79, No. 24_Supplement ( 2019-12-15), p. A62-A62
    Abstract: Objective: Pancreatic cancer is a deadly malignancy, and little progress has been made in the survival of patients during the past 40 years. Harnessing the immune system to elicit anticancer response is an attractive therapeutic option, alone or in combination with antitumor agents. Targeting regulatory T cell (Treg), which acts to suppress immune responses, holds promise as one potential approach to relieve the immune suppression of pancreatic cancer, but functional studies investigating whether Treg depletion at different stages of pancreatic cancer has therapeutic benefit are needed. Methods: We are using KC; Foxp3DTR mice generated by crossing KC (p48-Cre; LSL-KrasG12D) with Foxp3DTR (B6.129(Cg)-Foxp3tm3(DTR/GFP)Ayr/J, Jackson Laboratory) to determine the requirement of Tregs during oncogenic Kras-induced pancreatic cancer precursor lesion formation and progression. We depleted Foxp3-expressing Tregs by Diphtheria Toxin (DT) injection at different time points—either before or after PanIN formation. Multiplex immunohistochemistry and Cytometry by Time-of-Flight (CyTOF) were performed for tumor immunophenotyping; RNA sequencing was used for transcriptomic analysis of the tumor epithelial cells, fibroblasts, and myeloid cells. Results: Surprisingly, instead of blocking tumorigenesis, depletion of Tregs during these early stages led to accelerated tumor progression. We show that Tregs are a key source of TGFb, and accordingly their depletion alters fibroblast population, exemplified by loss of Smooth Muscle Actin expression. Treg depletion further resulted in a compensatory influx of suppressive myeloid cells, thereby preventing an antitumor immune response. Mechanistically, we show that this myeloid recruitment was driven through the CCR1 axis, with the receptor expressed on tumor-associated myeloid cells and several ligands upregulated in epithelial cells and fibroblasts upon Treg depletion. Finally, blockade of CCR1 signaling successfully inhibits tumor progression upon Treg-depletion. Conclusion: Our study revealed an immunosuppressive network of interactions between different components within the pancreatic tumor microenvironment, including Tregs, fibroblasts, tumor cells, and macrophages. Depleting one component (Tregs) can lead to the compensation of immunosuppression from the rest of components within the network. Thus, a thorough understanding of the immunosuppressive network in pancreatic neoplasia is necessary to optimize our approaches to treatment and identify opportunities for combination therapy and prevent potential feedback mechanisms resulting in tumor resistance and relapse. Citation Format: Yaqing Zhang, Jenny Lazarus, Nina Steele, Wei Yan, Ho-Joon Lee, Christopher J. Halbrook, Rosa Menjivar, Samantha B. Kemp, Veerin Sirihorachai, Eileen S. Carpenter, Anna C. Nevison, Alekya Vinta, Michelle A. Anderson, Howard C. Crawford, Costas A. Lyssiotis, Timothy L. Frankel, Filip Bednar, Marina Pasca di Magliano. Regulatory T-cell depletion promotes oncogenic Kras-driven pancreatic carcinogenesis [abstract]. In: Proceedings of the AACR Special Conference on Pancreatic Cancer: Advances in Science and Clinical Care; 2019 Sept 6-9; Boston, MA. Philadelphia (PA): AACR; Cancer Res 2019;79(24 Suppl):Abstract nr A62.
    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 ...
  • 10
    Online Resource
    Online Resource
    Annual Reviews ; 2019
    In:  Annual Review of Physiology Vol. 81, No. 1 ( 2019-02-10), p. 211-233
    In: Annual Review of Physiology, Annual Reviews, Vol. 81, No. 1 ( 2019-02-10), p. 211-233
    Abstract: Pancreatic cancer is characterized by an extensive fibroinflammatory reaction that includes immune cells, fibroblasts, extracellular matrix, vascular and lymphatic vessels, and nerves. Overwhelming evidence indicates that the pancreatic cancer microenvironment regulates cancer initiation, progression, and maintenance. Pancreatic cancer treatment has progressed little over the past several decades, and the prognosis remains one of the worst for any cancer. The contribution of the microenvironment to carcinogenesis is a key area of research, offering new potential targets for treating the disease. Here, we explore the composition of the pancreatic cancer stroma, discuss the network of interactions between different components, and describe recent attempts to target the stroma therapeutically. We also discuss current areas of active research related to the tumor microenvironment.
    Type of Medium: Online Resource
    ISSN: 0066-4278 , 1545-1585
    URL: Issue
    RVK:
    Language: English
    Publisher: Annual Reviews
    Publication Date: 2019
    detail.hit.zdb_id: 1474465-X
    SSG: 12
    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...