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) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. LB025-LB025
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. LB025-LB025
    Abstract: Metastatic relapse is the major causes of mortality in patients with cancer and occur due to metastatic reactivation of dormant tumor cells. Early dissemination of tumor cells undergoing a protected period of dormancy in the target organs potentially explains this prevalent clinical behavior. Long non-coding RNAs (lncRNAs) are involved in various biological processes and diseases. Malat1 is one of the most abundant and highly conserved nuclear lncRNAs and have shown the associated with metastasis and serving as a predictive marker for various tumor progression. However, the correlation of tumor intrinsic lncRNAs in regulation of tumor dormancy and immune evasion are largely unknown. Using an in vivo screening platform for the isolation of genetic entities involved in either dormancy or reactivation of breast cancer tumor cells, we have identified Malat1 as a positive mediator of metastatic reactivation. To dissect the functional role of Malat1 in metastatic reactivation, we developed a clean Malat1 knockout (KO) model using paired gRNA CRISPR-Cas9 in metastatic murine syngeneic breast cancer. As a proof of concept, we also used inducible knockdown system under in vivo models. To delineate the immune microenvironment, we used single cell RNA-seq, ChIRP-seq, multicolor flowcytometry, RNA-FISH, and coculture experiments. Our data revealed that deletion of Malat1 induces dormancy and attenuated the metastatic colonization resulting in long-term survival of syngeneic mice model. In contrast, overexpression of Malat1 leads to metastatic reactivation of dormant breast cancer cells. Interestingly, 4T1-Malat1 KO dormant breast cancer cells exhibit metastatic outgrowth in T cells defective mice. Our single-cell RNA-seq and multicolor flowcytometry evaluation reveal enhanced T cells and reduced neutrophils proportions in mice with Malat1 KO cells. This indicates a critical role of immune microenvironment via Malat1-dependent immune evasion. Additionally, Malat1 KO inhibits cancer stemness properties. Similarly, RNA-seq and ChIRP-seq data suggest that KO of Malat1 hampers immune evasion and downregulates metastasis associated genes including Serpins and Wnts. Additionally, our data strongly suggests that Malat1 KO cells persists as non-proliferative dormant cells in lung due to CD8+ T cell-umpired immune activity. Our mechanistic studies showed that Malat1 regulated Sepinb6b blocks the CstG enzymatic activity and suppress cleavage of GsdmD and T cells induced pyroptosis. Interestingly, rescue experiments suggest that Malat1 or Serpinb6b protects T cell-induced cell death and induces dormancy re-awakening thereby rescue the metastatic potential of 4T1 Malat1 KO cells. Combination of Malat1 ASO with double immune checkpoint inhibitors greatly affects the metastatic outgrowth in breast cancer. Taken together, our studies demonstrate that tumor intrinsic Malat1 regulates Serpinb6b that suppresses CstG-GsdmD mediated pyroptosis which eventually promotes immune evasion and breast cancer dormancy metastatic reactivation. Citation Format: Dhiraj Kumar, Filippo Giancotti. Malat1-Serpinb6b signaling blocks T cells-mediated pyroptosis and governs cancer dormancy metastatic reactivation [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr LB025.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    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 ...
  • 2
    In: Leukemia, Springer Science and Business Media LLC
    Type of Medium: Online Resource
    ISSN: 0887-6924 , 1476-5551
    RVK:
    Language: English
    Publisher: Springer Science and Business Media LLC
    Publication Date: 2024
    detail.hit.zdb_id: 2008023-2
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 3
    In: Cell Reports, Elsevier BV, Vol. 39, No. 1 ( 2022-04), p. 110595-
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2022
    detail.hit.zdb_id: 2649101-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 4
    In: Cell Reports, Elsevier BV, Vol. 42, No. 12 ( 2023-12), p. 113470-
    Type of Medium: Online Resource
    ISSN: 2211-1247
    Language: English
    Publisher: Elsevier BV
    Publication Date: 2023
    detail.hit.zdb_id: 2649101-1
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 5
    In: Science Translational Medicine, American Association for the Advancement of Science (AAAS), Vol. 11, No. 497 ( 2019-06-19)
    Abstract: The activated B cell (ABC-like) subtype of diffuse large B cell lymphoma (DLBCL) is characterized by chronic activation of signaling initiated by immunoglobulin μ (IgM). By analyzing the DNA copy number profiles of 1000 DLBCL tumors, we identified gains of 18q21.2 as the most frequent genetic alteration in ABC-like DLBCL. Using integrative analysis of matched gene expression profiling data, we found that the TCF4 ( E2-2 ) transcription factor gene was the target of these alterations. Overexpression of TCF4 in ABC-like DLBCL cell lines led to its occupancy on immunoglobulin ( IGHM ) and MYC gene enhancers and increased expression of these genes at the transcript and protein levels. Inhibition of TCF4 activity with dominant-negative constructs was synthetically lethal to ABC-like DLBCL cell lines harboring TCF4 DNA copy gains, highlighting these gains as an attractive potential therapeutic target. Furthermore, the TCF4 gene was one of the top BRD4-regulated genes in DLBCL cell lines. BET proteolysis-targeting chimera (PROTAC) ARV771 extinguished TCF4, MYC, and IgM expression and killed ABC-like DLBCL cells in vitro. In DLBCL xenograft models, ARV771 treatment reduced tumor growth and prolonged survival. This work highlights a genetic mechanism for promoting immunoglobulin signaling in ABC-like DLBCL and provides a functional rationale for the use of BET inhibitors in this disease.
    Type of Medium: Online Resource
    ISSN: 1946-6234 , 1946-6242
    Language: English
    Publisher: American Association for the Advancement of Science (AAAS)
    Publication Date: 2019
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 6
    Online Resource
    Online Resource
    BMJ ; 2021
    In:  Journal for ImmunoTherapy of Cancer Vol. 9, No. Suppl 2 ( 2021-11), p. A977-A977
    In: Journal for ImmunoTherapy of Cancer, BMJ, Vol. 9, No. Suppl 2 ( 2021-11), p. A977-A977
    Abstract: Metastatic relapse is the major causes of mortality in patients with cancer and occur due to metastatic reactivation of dormant tumor cells. Early dissemination of tumor cells undergoing a protected period of dormancy in the target organs potentially explains this prevalent clinical behavior. 1–4 Long non-coding RNAs (lncRNAs) are involved in various biological processes and diseases. Malat1 is one of the most abundant and highly conserved nuclear lncRNAs and have shown the associated with metastasis and serving as a predictive marker for various tumor progression. 5 However, the correlation of tumor intrinsic lncRNAs in regulation of tumor dormancy and immune evasion is largely unknown. Methods Using an in vivo screening platform for the isolation of genetic entities involved in either dormancy or reactivation of breast cancer tumor cells, we have identified Malat1 as a positive mediator of metastatic reactivation. 4 To dissect the functional role of Malat1 in metastatic reactivation, we developed a clean Malat1 knockout (KO) model using paired gRNA CRISPR-Cas9 in metastatic murine syngeneic breast cancer. As proof of concept we also used inducible knockdown system under in vivo models. To delineate the immune microenvironment, we used single cell RNA-seq, ChIRP-seq, multicolor flowcytometry, RNA-FISH, and coculture experiments. Results Our data revealed that deletion of Malat1 induces dormancy and attenuated the metastatic colonization resulting in long-term survival of syngeneic mice model. In contrast, overexpression of Malat1 leads to metastatic reactivation of dormant breast cancer cells. Interestingly, 4T1-Malat1 KO dormant breast cancer cells exhibit metastatic outgrowth in T cells defective mice. Our single-cell RNA-seq and multicolor flowcytometry evaluation reveal enhanced T cells and reduced neutrophils proportions in mice with Malat1 KO cells. This indicates a critical role of immune microenvironment via Malat1-dependent immune evasion. Additionally, Malat1 KO inhibits cancer stemness properties. Similarly, RNA-seq and ChIRP-seq data suggest that KO of Malat1 hampers immune evasion and downregulates metastasis associated genes including Serpins and Wnts. Additionally, our data strongly suggests that Malat1 KO cells persists as non-proliferative dormant cells in lung due to CD8+ T cell-umpired immune activity. Interestingly, rescue experiments suggest that Malat1 or Serpinb6b protects T cell-induced cell death and induces dormancy re-awakening thereby rescue the metastatic potential of 4T1 Malat1 KO cells. Combination of Malat1 ASO with double immune checkpoint inhibitors greatly affects the metastatic outgrowth in breast cancer. Conclusions Taken together, our studies demonstrate that tumor intrinsic Malat1 regulates Serpinb6b that eventually controls immune evasion and promote dormancy metastatic reactivation. Acknowledgements NGS data generated was supported by Core grant CA016672(ATGC) and NIH 1S10OD024977-01 award to the ATGC. Single cell RNA sequencing data was supported by the CPRIT Single Core grant RP180684. The Advanced Cytometry & Sorting Core Facility is supported by NCI P30CA016672. References Arun G, Diermeier S, Akerman M, et al . Differentiation of mammary tumors and reduction in metastasis upon Malat1 lncRNA loss. Genes Dev 2016 January 1; 30 (1):34–51. Filippo G Giancotti. Mechanisms governing metastatic dormancy and reactivation. Cell 2013 November 7; 155 (4):750–764. Gao H, Chakraborty G, Lee-Lim AP, et al . The BMP inhibitor Coco reactivates breast cancer cells at lung metastatic sites. Cell 2012b; 150 :764–779. Gao H, Chakraborty G, Lee-Lim AP, et al . Forward genetic screens in mice uncover mediators and suppressors of metastatic reactivation. Proc Natl Acad Sci U S A 2014 November 18; 111 (46):16532–16537. Huang D, Chen J, Yang L, et al . NKILA lncRNA promotes tumor immune evasion by sensitizing T cells to activation-induced cell death. Nat Immunol 2018; 19 :1112–1125.
    Type of Medium: Online Resource
    ISSN: 2051-1426
    Language: English
    Publisher: BMJ
    Publication Date: 2021
    detail.hit.zdb_id: 2719863-7
    Location Call Number Limitation Availability
    BibTip Others were also interested in ...
  • 7
    Online Resource
    Online Resource
    American Association for Cancer Research (AACR) ; 2022
    In:  Cancer Research Vol. 82, No. 12_Supplement ( 2022-06-15), p. LB023-LB023
    In: Cancer Research, American Association for Cancer Research (AACR), Vol. 82, No. 12_Supplement ( 2022-06-15), p. LB023-LB023
    Abstract: Background: Systemic factors from the tumor microenvironment (TME) promote prostate cancer (PCa) progression, metastasis, and drug resistance. Therefore, the identification of the systemic factors that can modulate the tumor cells and promote metastases will be critical to improve the efficacy of clinical treatment. Both mechanistic and functional studies revealed that acquisition of an open chromatin state on master regulators or cis-regulatory elements allow them to respond readily to microenvironment signals, thereby increasing cancer cell plasticity, tumorigenicity and malignant potential. Accordingly, analysis of open chromatin regions can help prioritize and identify risk of functionally noncoding variants. Given the power of open chromatin profiling on environmental risk prediction, we hypothesized that analysis of open chromatin regions may enable us to recognize functional trans-regulatory elements with their relevant systemic factors. Methods and Results: In human prostate cancer cells, AR blockage by enzalutamide can induces cell reprogramming, as well as transcriptional dysregulation of the heterochromatin compaction pathway. Hence, we proposed that the cells were undergoing chromatin decondensation followed by epigenetic reprogramming. To detect the genome-wide open chromatin changes in this process, we performed time course ATAC-seq and H3K27ac ChIP-seq experiments on LNCaP cells treated with enzalutamide over a 2 weeks period. Bioinformatic analysis was performed to identify the “open” enhancers and the transcriptional factors potentially bound to these enhancers. Integrated transcription factor binding motif analysis of metastatic castration-resistant prostate cancer (mCRPC) associated enhancer regions (GSE130408) and multiplexed siRNA screening revealed Thyroid Hormone Receptor Beta (THRB) as the top candidate. THRB is regulated by the thyroid hormone (T3), a systemic nuclear hormone that, in enzalutamide pretreated LNCaP, VCaP and 3PDXs, is able to induce subsequent reprogramming to a fully enzalutamide-resistant and metastatic state. Interestingly, we observed that THRB regulated genes are upregulated in T3-induced resistant prostate cancer cells. Consistently, the THRB protein, TRβ, is abundant in several aggressive PCa cell lines. Notably, knockdown of THRB reverted the growth and the bone metastatic potential of highly aggressive PCa models, such as Ptenpc-/-Smad4pc-/- and PC3M cells. Furthermore, both transcriptomic and functional analyses identified RUNX2 as a transcriptional target of THRB. In summary, we identify a novel mechanism of THRB-mediated tumor progression and drug resistance in PCa cells. Conclusion: Epidemiologic evidence indicates a higher incidence of PCa in men with elevated thyroid hormone levels. Accordingly, our results suggest a new mechanism for thyroid hormone signaling regulating metastasis potential and drug resistance. This study will provide a new perspective to identify the signals from the TME that may increase the risk of PCa progression. Citation Format: Yan Wang, Gurrapu Harsha, Dhiraj Kumar, Josue Curto, Xiaobo Wang, Filippo Giancotti. Thyroid hormone receptor signaling promotes antiandrogen resistance and bone metastasis in prostate cancer [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2022; 2022 Apr 8-13. Philadelphia (PA): AACR; Cancer Res 2022;82(12_Suppl):Abstract nr LB023.
    Type of Medium: Online Resource
    ISSN: 1538-7445
    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 ...
Close ⊗
This website uses cookies and the analysis tool Matomo. More information can be found here...