LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Your last searches

  1. AU="Florian, Andrea C"
  2. AU=Hamzeh Nabeel

Search results

Result 1 - 6 of total 6

Search options

  1. Article ; Online: Mitotic gene regulation by the N-MYC-WDR5-PDPK1 nexus.

    Streeter, Sarah A / Williams, Alexandria G / Evans, James R / Wang, Jing / Guarnaccia, Alissa D / Florian, Andrea C / Al-Tobasei, Rafet / Liu, Qi / Tansey, William P / Weissmiller, April M

    BMC genomics

    2024  Volume 25, Issue 1, Page(s) 360

    Abstract: Background: During mitosis the cell depends on proper attachment and segregation of replicated chromosomes to generate two identical progeny. In cancers defined by overexpression or dysregulation of the MYC oncogene this process becomes impaired, ... ...

    Abstract Background: During mitosis the cell depends on proper attachment and segregation of replicated chromosomes to generate two identical progeny. In cancers defined by overexpression or dysregulation of the MYC oncogene this process becomes impaired, leading to genomic instability and tumor evolution. Recently it was discovered that the chromatin regulator WDR5-a critical MYC cofactor-regulates expression of genes needed in mitosis through a direct interaction with the master kinase PDPK1. However, whether PDPK1 and WDR5 contribute to similar mitotic gene regulation in MYC-overexpressing cancers remains unclear. Therefore, to characterize the influence of WDR5 and PDPK1 on mitotic gene expression in cells with high MYC levels, we performed a comparative transcriptomic analysis in neuroblastoma cell lines defined by MYCN-amplification, which results in high cellular levels of the N-MYC protein.
    Results: Using RNA-seq analysis, we identify the genes regulated by N-MYC and PDPK1 in multiple engineered CHP-134 neuroblastoma cell lines and compare them to previously published gene expression data collected in CHP-134 cells following inhibition of WDR5. We find that as expected N-MYC regulates a multitude of genes, including those related to mitosis, but that PDPK1 regulates specific sets of genes involved in development, signaling, and mitosis. Analysis of N-MYC- and PDPK1-regulated genes reveals a small group of commonly controlled genes associated with spindle pole formation and chromosome segregation, which overlap with genes that are also regulated by WDR5. We also find that N-MYC physically interacts with PDPK1 through the WDR5-PDPK1 interaction suggesting regulation of mitotic gene expression may be achieved through a N-MYC-WDR5-PDPK1 nexus.
    Conclusions: Overall, we identify a small group of genes highly enriched within functional gene categories related to mitotic processes that are commonly regulated by N-MYC, WDR5, and PDPK1 and suggest that a tripartite interaction between the three regulators may be responsible for setting the level of mitotic gene regulation in N-MYC amplified cell lines. This study provides a foundation for future studies to determine the exact mechanism by which N-MYC, WDR5, and PDPK1 converge on cell cycle related processes.
    MeSH term(s) Humans ; 3-Phosphoinositide-Dependent Protein Kinases/genetics ; 3-Phosphoinositide-Dependent Protein Kinases/metabolism ; Cell Line, Tumor ; Chromosome Segregation ; Gene Expression Regulation, Neoplastic ; Genes, myc ; Intracellular Signaling Peptides and Proteins/genetics ; Neuroblastoma/metabolism
    Chemical Substances 3-Phosphoinositide-Dependent Protein Kinases (EC 2.7.11.1) ; Intracellular Signaling Peptides and Proteins ; PDPK1 protein, human (EC 2.7.11.1) ; WDR5 protein, human
    Language English
    Publishing date 2024-04-11
    Publishing country England
    Document type Journal Article
    ZDB-ID 2041499-7
    ISSN 1471-2164 ; 1471-2164
    ISSN (online) 1471-2164
    ISSN 1471-2164
    DOI 10.1186/s12864-024-10282-6
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article: Ribosome subunit attrition and activation of the p53-MDM4 axis dominate the response of MLL-rearranged cancer cells to WDR5 WIN site inhibition.

    Howard, Gregory C / Wang, Jing / Rose, Kristie Lindsey / Jones, Camden / Patel, Purvi / Tsui, Tina / Florian, Andrea C / Vlach, Logan / Lorey, Shelly L / Grieb, Brian C / Smith, Brianna N / Slota, Macey J / Reynolds, Elizabeth M / Goswami, Soumita / Savona, Michael R / Mason, Frank M / Lee, Taekyu / Fesik, Stephen W / Liu, Qi /
    Tansey, William P

    bioRxiv : the preprint server for biology

    2024  

    Abstract: The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the "WIN" site that tethers WDR5 to chromatin. WIN site inhibitors ( ... ...

    Abstract The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the "WIN" site that tethers WDR5 to chromatin. WIN site inhibitors (WINi) are active against multiple cancer cell types in vitro, the most notable of which are those derived from MLL-rearranged (MLLr) leukemias. Peptidomimetic WINi were originally proposed to inhibit MLLr cells via dysregulation of genes connected to hematopoietic stem cell expansion. Our discovery and interrogation of small molecule WIN site inhibitors, however, revealed that they act in MLLr cell lines to suppress ribosome protein gene (RPG) transcription, induce nucleolar stress, and activate p53. Because there is no precedent for an anti-cancer strategy that specifically targets RPG expression, we took an integrated multi-omics approach to further interrogate the mechanism of action of WINi in MLLr cancer cells. We show that WINi induce depletion of the stock of ribosomes, accompanied by a broad yet modest translational choke and changes in alternative mRNA splicing that inactivate the p53 antagonist MDM4. We also show that WINi are synergistic with agents including venetoclax and BET-bromodomain inhibitors. Together, these studies reinforce the concept that WINi are a novel type of ribosome-directed anti-cancer therapy and provide a resource to support their clinical implementation in MLLr leukemias and other malignancies.
    Language English
    Publishing date 2024-01-10
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.07.26.550648
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Ribosome subunit attrition and activation of the p53-MDM4 axis dominate the response of MLL-rearranged cancer cells to WDR5 WIN site inhibition.

    Howard, Gregory Caleb / Wang, Jing / Rose, Kristie L / Jones, Camden / Patel, Purvi / Tsui, Tina / Florian, Andrea C / Vlach, Logan / Lorey, Shelly L / Grieb, Brian C / Smith, Brianna N / Slota, Macey J / Reynolds, Elizabeth M / Goswami, Soumita / Savona, Michael R / Mason, Frank M / Lee, Taekyu / Fesik, Stephen / Liu, Qi /
    Tansey, William P

    eLife

    2024  Volume 12

    Abstract: The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the 'WIN' site that tethers WDR5 to chromatin. WIN site inhibitors ( ... ...

    Abstract The chromatin-associated protein WD Repeat Domain 5 (WDR5) is a promising target for cancer drug discovery, with most efforts blocking an arginine-binding cavity on the protein called the 'WIN' site that tethers WDR5 to chromatin. WIN site inhibitors (WINi) are active against multiple cancer cell types in vitro, the most notable of which are those derived from MLL-rearranged (MLLr) leukemias. Peptidomimetic WINi were originally proposed to inhibit MLLr cells via dysregulation of genes connected to hematopoietic stem cell expansion. Our discovery and interrogation of small-molecule WINi, however, revealed that they act in MLLr cell lines to suppress ribosome protein gene (RPG) transcription, induce nucleolar stress, and activate p53. Because there is no precedent for an anticancer strategy that specifically targets RPG expression, we took an integrated multi-omics approach to further interrogate the mechanism of action of WINi in human MLLr cancer cells. We show that WINi induce depletion of the stock of ribosomes, accompanied by a broad yet modest translational choke and changes in alternative mRNA splicing that inactivate the p53 antagonist MDM4. We also show that WINi are synergistic with agents including venetoclax and BET-bromodomain inhibitors. Together, these studies reinforce the concept that WINi are a novel type of ribosome-directed anticancer therapy and provide a resource to support their clinical implementation in MLLr leukemias and other malignancies.
    MeSH term(s) Humans ; Antineoplastic Agents/pharmacology ; Cell Cycle Proteins/metabolism ; Cell Cycle Proteins/genetics ; Cell Line, Tumor ; Histone-Lysine N-Methyltransferase/metabolism ; Histone-Lysine N-Methyltransferase/genetics ; Intracellular Signaling Peptides and Proteins/antagonists & inhibitors ; Intracellular Signaling Peptides and Proteins/genetics ; Intracellular Signaling Peptides and Proteins/metabolism ; Myeloid-Lymphoid Leukemia Protein/metabolism ; Myeloid-Lymphoid Leukemia Protein/genetics ; Nuclear Proteins/metabolism ; Nuclear Proteins/genetics ; Proto-Oncogene Proteins/genetics ; Proto-Oncogene Proteins/metabolism ; Ribosomes/drug effects ; Ribosomes/metabolism ; Tumor Suppressor Protein p53/metabolism ; Tumor Suppressor Protein p53/genetics ; Peptidomimetics/pharmacology
    Chemical Substances Antineoplastic Agents ; Cell Cycle Proteins ; Histone-Lysine N-Methyltransferase (EC 2.1.1.43) ; Intracellular Signaling Peptides and Proteins ; KMT2A protein, human ; MDM4 protein, human ; Myeloid-Lymphoid Leukemia Protein (149025-06-9) ; Nuclear Proteins ; Proto-Oncogene Proteins ; TP53 protein, human ; Tumor Suppressor Protein p53 ; WDR5 protein, human ; Peptidomimetics
    Language English
    Publishing date 2024-04-29
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't ; Research Support, N.I.H., Extramural
    ZDB-ID 2687154-3
    ISSN 2050-084X ; 2050-084X
    ISSN (online) 2050-084X
    ISSN 2050-084X
    DOI 10.7554/eLife.90683
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: WIN site inhibition disrupts a subset of WDR5 function.

    Siladi, Andrew J / Wang, Jing / Florian, Andrea C / Thomas, Lance R / Creighton, Joy H / Matlock, Brittany K / Flaherty, David K / Lorey, Shelly L / Howard, Gregory C / Fesik, Stephen W / Weissmiller, April M / Liu, Qi / Tansey, William P

    Scientific reports

    2022  Volume 12, Issue 1, Page(s) 1848

    Abstract: WDR5 nucleates the assembly of histone-modifying complexes and acts outside this context in a range of chromatin-centric processes. WDR5 is also a prominent target for pharmacological inhibition in cancer. Small-molecule degraders of WDR5 have been ... ...

    Abstract WDR5 nucleates the assembly of histone-modifying complexes and acts outside this context in a range of chromatin-centric processes. WDR5 is also a prominent target for pharmacological inhibition in cancer. Small-molecule degraders of WDR5 have been described, but most drug discovery efforts center on blocking the WIN site of WDR5, an arginine binding cavity that engages MLL/SET enzymes that deposit histone H3 lysine 4 methylation (H3K4me). Therapeutic application of WIN site inhibitors is complicated by the disparate functions of WDR5, but is generally guided by two assumptions-that WIN site inhibitors disable all functions of WDR5, and that changes in H3K4me drive the transcriptional response of cancer cells to WIN site blockade. Here, we test these assumptions by comparing the impact of WIN site inhibition versus WDR5 degradation on H3K4me and transcriptional processes. We show that WIN site inhibition disables only a specific subset of WDR5 activity, and that H3K4me changes induced by WDR5 depletion do not explain accompanying transcriptional responses. These data recast WIN site inhibitors as selective loss-of-function agents, contradict H3K4me as a relevant mechanism of action for WDR5 inhibitors, and indicate distinct clinical applications of WIN site inhibitors and WDR5 degraders.
    MeSH term(s) Antineoplastic Agents/pharmacology ; Binding Sites ; Bridged Bicyclo Compounds, Heterocyclic/pharmacology ; Cell Cycle/drug effects ; Cell Line, Tumor ; Cell Proliferation/drug effects ; Chromatin Assembly and Disassembly ; Gene Expression Regulation, Neoplastic ; Histones/metabolism ; Humans ; Intracellular Signaling Peptides and Proteins/antagonists & inhibitors ; Intracellular Signaling Peptides and Proteins/genetics ; Intracellular Signaling Peptides and Proteins/metabolism ; Lymphoma, B-Cell/drug therapy ; Lymphoma, B-Cell/genetics ; Lymphoma, B-Cell/metabolism ; Methylation ; Protein Binding ; Protein Interaction Domains and Motifs ; Proteolysis ; Signal Transduction ; Transcription, Genetic
    Chemical Substances Antineoplastic Agents ; Bridged Bicyclo Compounds, Heterocyclic ; Histones ; Intracellular Signaling Peptides and Proteins ; WDR5 protein, human
    Language English
    Publishing date 2022-02-03
    Publishing country England
    Document type Comparative Study ; Journal Article ; Research Support, N.I.H., Extramural ; Research Support, Non-U.S. Gov't
    ZDB-ID 2615211-3
    ISSN 2045-2322 ; 2045-2322
    ISSN (online) 2045-2322
    ISSN 2045-2322
    DOI 10.1038/s41598-022-05947-9
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article: The SWI/SNF ATPase BRG1 facilitates multiple pro-tumorigenic gene expression programs in SMARCB1-deficient cancer cells.

    Moe, Kylie C / Maxwell, Jack N / Wang, Jing / Jones, Cheyenne A / Csaki, Grace T / Florian, Andrea C / Romer, Alexander S / Bryant, Daniel L / Farone, Anthony L / Liu, Qi / Tansey, William P / Weissmiller, April M

    Oncogenesis

    2022  Volume 11, Issue 1, Page(s) 30

    Abstract: Malignant rhabdoid tumor (MRT) is driven by the loss of the SNF5 subunit of the SWI/SNF chromatin remodeling complex and then thought to be maintained by residual SWI/SNF (rSWI/SNF) complexes that remain present in the absence of SNF5. rSWI/SNF subunits ... ...

    Abstract Malignant rhabdoid tumor (MRT) is driven by the loss of the SNF5 subunit of the SWI/SNF chromatin remodeling complex and then thought to be maintained by residual SWI/SNF (rSWI/SNF) complexes that remain present in the absence of SNF5. rSWI/SNF subunits colocalize extensively on chromatin with the transcription factor MYC, an oncogene identified as a novel driver of MRT. Currently, the role of rSWI/SNF in modulating MYC activity has neither been delineated nor has a direct link between rSWI/SNF and other oncogenes been uncovered. Here, we expose the connection between rSWI/SNF and oncogenic processes using a well-characterized chemical degrader to deplete the SWI/SNF ATPase, BRG1. Using a combination of gene expression and chromatin accessibility assays we show that rSWI/SNF complexes facilitate MYC target gene expression. We also find that rSWI/SNF maintains open chromatin at sites associated with hallmark cancer genes linked to the AP-1 transcription factor, suggesting that AP-1 may drive oncogenesis in MRT. Interestingly, changes in MYC target gene expression are not overtly connected to the chromatin remodeling function of rSWI/SNF, revealing multiple mechanisms used by rSWI/SNF to control transcription. This work provides an understanding of how residual SWI/SNF complexes may converge on multiple oncogenic processes when normal SWI/SNF function is impaired.
    Language English
    Publishing date 2022-06-01
    Publishing country United States
    Document type Journal Article
    ZDB-ID 2674437-5
    ISSN 2157-9024
    ISSN 2157-9024
    DOI 10.1038/s41389-022-00406-6
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: Synergistic action of WDR5 and HDM2 inhibitors in SMARCB1-deficient cancer cells.

    Florian, Andrea C / Woodley, Chase M / Wang, Jing / Grieb, Brian C / Slota, Macey J / Guerrazzi, Kiana / Hsu, Chih-Yuan / Matlock, Brittany K / Flaherty, David K / Lorey, Shelly L / Fesik, Stephen W / Howard, Gregory C / Liu, Qi / Weissmiller, April M / Tansey, William P

    NAR cancer

    2022  Volume 4, Issue 1, Page(s) zcac007

    Abstract: Rhabdoid tumors (RT) are rare and deadly pediatric cancers driven by loss ... ...

    Abstract Rhabdoid tumors (RT) are rare and deadly pediatric cancers driven by loss of
    Language English
    Publishing date 2022-03-03
    Publishing country England
    Document type Journal Article
    ISSN 2632-8674
    ISSN (online) 2632-8674
    DOI 10.1093/narcan/zcac007
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top