LIVIVO - The Search Portal for Life Sciences

zur deutschen Oberfläche wechseln
Advanced search

Search results

Result 1 - 10 of total 294

Search options

  1. Article ; Online: Advances and opportunities in targeted protein degradation.

    Nomura, Daniel K / Dey, Mishtu

    Cell chemical biology

    2021  Volume 28, Issue 7, Page(s) 887–888

    MeSH term(s) Humans ; Proteolysis ; Ubiquitin-Protein Ligases/metabolism
    Chemical Substances Ubiquitin-Protein Ligases (EC 2.3.2.27)
    Language English
    Publishing date 2021-07-16
    Publishing country United States
    Document type Editorial ; Introductory Journal Article
    ISSN 2451-9448
    ISSN (online) 2451-9448
    DOI 10.1016/j.chembiol.2021.06.011
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  2. Article ; Online: Virtual Issue on the Work of John Casida.

    Nomura, Daniel K

    Chemical research in toxicology

    2018  Volume 31, Issue 8, Page(s) 637–638

    MeSH term(s) California ; History, 20th Century ; History, 21st Century ; Toxicology
    Language English
    Publishing date 2018-08-06
    Publishing country United States
    Document type Biography ; Editorial ; Historical Article ; Introductory Journal Article
    ZDB-ID 639353-6
    ISSN 1520-5010 ; 0893-228X
    ISSN (online) 1520-5010
    ISSN 0893-228X
    DOI 10.1021/acs.chemrestox.8b00195
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  3. Article ; Online: Exploiting the Cullin E3 Ligase Adaptor Protein SKP1 for Targeted Protein Degradation.

    Hong, Seong Ho / Divakaran, Anand / Osa, Akane / Huang, Oscar W / Wertz, Ingrid E / Nomura, Daniel K

    ACS chemical biology

    2024  Volume 19, Issue 2, Page(s) 442–450

    Abstract: Targeted protein degradation with proteolysis targeting chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited ... ...

    Abstract Targeted protein degradation with proteolysis targeting chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited substrate receptors of Cullin-RING E3 ubiquitin ligases such as cereblon and VHL. Whether core, shared, and essential components of the Cullin-RING E3 ubiquitin ligase complex can be used for PROTAC applications remains less explored. Here, we discovered a cysteine-reactive covalent recruiter EN884 against the SKP1 adapter protein of the SKP1-CUL1-F-box containing the SCF complex. We further showed that this recruiter can be used in PROTAC applications to degrade neo-substrate proteins such as BRD4 and the androgen receptor in a SKP1- and proteasome-dependent manner. Our studies demonstrate that core and essential adapter proteins within the Cullin-RING E3 ubiquitin ligase complex can be exploited for targeted protein degradation applications and that covalent chemoproteomic strategies can enable recruiter discovery against these targets.
    MeSH term(s) Ubiquitin-Protein Ligases/metabolism ; Cullin Proteins/metabolism ; Proteolysis ; Proteasome Endopeptidase Complex/metabolism ; Nuclear Proteins/metabolism ; Transcription Factors/metabolism ; S-Phase Kinase-Associated Proteins/metabolism ; Adaptor Proteins, Signal Transducing/metabolism
    Chemical Substances Ubiquitin-Protein Ligases (EC 2.3.2.27) ; Cullin Proteins ; Proteasome Endopeptidase Complex (EC 3.4.25.1) ; Nuclear Proteins ; Transcription Factors ; S-Phase Kinase-Associated Proteins ; Adaptor Proteins, Signal Transducing
    Language English
    Publishing date 2024-02-02
    Publishing country United States
    Document type Journal Article
    ISSN 1554-8937
    ISSN (online) 1554-8937
    DOI 10.1021/acschembio.3c00642
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  4. Article ; Online: Targeted Protein Degradation through Recruitment of the CUL4 Complex Adaptor Protein DDB1.

    Meyers, Margot / Cismoski, Sabine / Panidapu, Anoohya / Chie-Leon, Barbara / Nomura, Daniel K

    ACS chemical biology

    2024  Volume 19, Issue 1, Page(s) 58–68

    Abstract: Targeted protein degradation has arisen as a powerful therapeutic modality for eliminating proteins. Thus far, most heterobifunctional proteolysis targeting chimeras (PROTACs) have utilized recruiters against substrate receptors of Cullin RING E3 ... ...

    Abstract Targeted protein degradation has arisen as a powerful therapeutic modality for eliminating proteins. Thus far, most heterobifunctional proteolysis targeting chimeras (PROTACs) have utilized recruiters against substrate receptors of Cullin RING E3 ubiquitin ligases, such as cereblon and VHL. However, previous studies have surprisingly uncovered molecular glue degraders that exploit a CUL4 adaptor protein DDB1 to degrade neosubstrate proteins. Here, we sought to investigate whether DDB1 recruiters can be discovered that can be exploited for PROTAC applications. We utilized activity-based protein profiling and cysteine chemoproteomic screening to identify a covalent recruiter that targets C173 on DDB1 and exploited this recruiter to develop PROTACs against BRD4 and androgen receptor (AR). We demonstrated that the BRD4 PROTAC results in selective degradation of the short BRD4 isoform over the long isoform in a proteasome, NEDDylation, and DDB1-dependent manner. We also demonstrated degradation of AR with the AR PROTAC in prostate cancer cells. Our study demonstrated that covalent chemoproteomic approaches can be used to discover recruiters against Cullin RING adapter proteins and that these recruiters can be used for PROTAC applications to degrade neo-substrates.
    MeSH term(s) Proteolysis ; Cullin Proteins/metabolism ; Transcription Factors/metabolism ; Nuclear Proteins/metabolism ; Ubiquitin-Protein Ligases/metabolism ; Adaptor Proteins, Signal Transducing/metabolism ; Proteasome Endopeptidase Complex/metabolism ; Protein Isoforms/metabolism
    Chemical Substances Cullin Proteins ; Transcription Factors ; Nuclear Proteins ; Ubiquitin-Protein Ligases (EC 2.3.2.27) ; Adaptor Proteins, Signal Transducing ; Proteasome Endopeptidase Complex (EC 3.4.25.1) ; Protein Isoforms
    Language English
    Publishing date 2024-01-08
    Publishing country United States
    Document type Journal Article
    ISSN 1554-8937
    ISSN (online) 1554-8937
    DOI 10.1021/acschembio.3c00487
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  5. Article: Covalent 14-3-3 Molecular Glues and Heterobifunctional Molecules Against Nuclear Transcription Factors and Regulators.

    Shao, Qian / Duong, Tuong Nghi / Park, Inji / Nomura, Daniel K

    bioRxiv : the preprint server for biology

    2023  

    Abstract: 14-3-3 proteins have the unique ability to bind and sequester a multitude of diverse phosphorylated signaling proteins and transcription factors. Many previous studies have shown that 14-3-3 interactions with specific phosphorylated substrate proteins ... ...

    Abstract 14-3-3 proteins have the unique ability to bind and sequester a multitude of diverse phosphorylated signaling proteins and transcription factors. Many previous studies have shown that 14-3-3 interactions with specific phosphorylated substrate proteins can be enhanced through small-molecule natural product or fully synthetic molecular glue interactions. However, enhancing 14-3-3 interactions with both therapeutically intractable transcription factor substrates as well as potential neo-substrates to sequester and inhibit their function has remained elusive. One of the 14-3-3 proteins, 14-3-3σ or SFN, has a cysteine C38 at the substrate binding interface near sites where previous 14-3-3 molecular glues have been found to bind. In this study, we screened a fully synthetic cysteine-reactive covalent ligand library to identify molecular glues that enhance interaction of 14-3-3σ with not only druggable transcription factors such as estrogen receptor (ERα), but also challenging oncogenic transcription factors such as YAP and TAZ that are part of the Hippo transducer pathway. We identified a hit EN171 that covalently targets 14-3-3 to enhance 14-3-3 interactions with ERα, YAP, and TAZ leading to impaired estrogen receptor and Hippo pathway transcriptional activity. We further demonstrate that EN171 could not only be used as a molecular glue to enhance native protein interactions, but also could be used as a covalent 14-3-3 recruiter in heterobifunctional molecules to sequester nuclear neo-substrates such as BRD4 into the cytosol. Overall, our study reveals a covalent ligand that acts as a novel 14-3-3 molecular glue for challenging transcription factors such as YAP and TAZ and also demonstrates that these glues can be potentially utilized in heterobifunctional molecules to sequester nuclear neo-substrates out of the nucleus and into the cytosol to enable targeted protein localization.
    Language English
    Publishing date 2023-11-06
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.11.06.565850
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  6. Article ; Online: N-Acryloylindole-alkyne (NAIA) enables imaging and profiling new ligandable cysteines and oxidized thiols by chemoproteomics.

    Koo, Tin-Yan / Lai, Hinyuk / Nomura, Daniel K / Chung, Clive Yik-Sham

    Nature communications

    2023  Volume 14, Issue 1, Page(s) 3564

    Abstract: Cysteine has been exploited as the binding site of covalent drugs. Its high sensitivity to oxidation is also important for regulating cellular processes. To identify new ligandable cysteines which can be hotspots for therapy and to better study cysteine ... ...

    Abstract Cysteine has been exploited as the binding site of covalent drugs. Its high sensitivity to oxidation is also important for regulating cellular processes. To identify new ligandable cysteines which can be hotspots for therapy and to better study cysteine oxidations, we develop cysteine-reactive probes, N-acryloylindole-alkynes (NAIAs), which have superior cysteine reactivity owing to delocalization of π electrons of the acrylamide warhead over the whole indole scaffold. This allows NAIAs to probe functional cysteines more effectively than conventional iodoacetamide-alkyne, and to image oxidized thiols by confocal fluorescence microscopy. In mass spectrometry experiments, NAIAs successfully capture new oxidized cysteines, as well as a new pool of ligandable cysteines and proteins. Competitive activity-based protein profiling experiments further demonstrate the ability of NAIA to discover lead compounds targeting these cysteines and proteins. We show the development of NAIAs with activated acrylamide for advancing proteome-wide profiling and imaging ligandable cysteines and oxidized thiols.
    MeSH term(s) Cysteine/metabolism ; Sulfhydryl Compounds/chemistry ; Alkynes/chemistry ; Proteins/chemistry ; Acrylamides
    Chemical Substances Cysteine (K848JZ4886) ; Sulfhydryl Compounds ; Alkynes ; Proteins ; Acrylamides
    Language English
    Publishing date 2023-06-15
    Publishing country England
    Document type Journal Article ; Research Support, Non-U.S. Gov't
    ZDB-ID 2553671-0
    ISSN 2041-1723 ; 2041-1723
    ISSN (online) 2041-1723
    ISSN 2041-1723
    DOI 10.1038/s41467-023-39268-w
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  7. Article ; Online: Advances in covalent drug discovery.

    Boike, Lydia / Henning, Nathaniel J / Nomura, Daniel K

    Nature reviews. Drug discovery

    2022  Volume 21, Issue 12, Page(s) 881–898

    Abstract: Covalent drugs have been used to treat diseases for more than a century, but tools that facilitate the rational design of covalent drugs have emerged more recently. The purposeful addition of reactive functional groups to existing ligands can enable ... ...

    Abstract Covalent drugs have been used to treat diseases for more than a century, but tools that facilitate the rational design of covalent drugs have emerged more recently. The purposeful addition of reactive functional groups to existing ligands can enable potent and selective inhibition of target proteins, as demonstrated by the covalent epidermal growth factor receptor (EGFR) and Bruton's tyrosine kinase (BTK) inhibitors used to treat various cancers. Moreover, the identification of covalent ligands through 'electrophile-first' approaches has also led to the discovery of covalent drugs, such as covalent inhibitors for KRAS(G12C) and SARS-CoV-2 main protease. In particular, the discovery of KRAS(G12C) inhibitors validates the use of covalent screening technologies, which have become more powerful and widespread over the past decade. Chemoproteomics platforms have emerged to complement covalent ligand screening and assist in ligand discovery, selectivity profiling and target identification. This Review showcases covalent drug discovery milestones with emphasis on the lessons learned from these programmes and how an evolving toolbox of covalent drug discovery techniques facilitates success in this field.
    MeSH term(s) Humans ; Drug Discovery/methods ; Ligands ; Protein Kinase Inhibitors/pharmacology ; Protein Kinase Inhibitors/therapeutic use ; Proto-Oncogene Proteins p21(ras)/metabolism ; SARS-CoV-2 ; Structure-Activity Relationship ; COVID-19 Drug Treatment
    Chemical Substances Ligands ; Protein Kinase Inhibitors ; Proto-Oncogene Proteins p21(ras) (EC 3.6.5.2) ; BTK protein, human (EC 2.7.10.2) ; EGFR protein, human (EC 2.7.10.1)
    Language English
    Publishing date 2022-08-25
    Publishing country England
    Document type Journal Article ; Review
    ZDB-ID 2062954-0
    ISSN 1474-1784 ; 1474-1776
    ISSN (online) 1474-1784
    ISSN 1474-1776
    DOI 10.1038/s41573-022-00542-z
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  8. Article: Exploiting the Cullin E3 Ligase Adaptor Protein SKP1 for Targeted Protein Degradation.

    Hong, Seong Ho / Osa, Akane / Huang, Oscar W / Wertz, Ingrid E / Nomura, Daniel K

    bioRxiv : the preprint server for biology

    2023  

    Abstract: Targeted protein degradation with Proteolysis Targeting Chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited ... ...

    Abstract Targeted protein degradation with Proteolysis Targeting Chimeras (PROTACs) is a powerful therapeutic modality for eliminating disease-causing proteins through targeted ubiquitination and proteasome-mediated degradation. Most PROTACs have exploited substrate receptors of Cullin-RING E3 ubiquitin ligases such as cereblon and VHL. Whether core, shared, and essential components of the Cullin-RING E3 ubiquitin ligase complex can be used for PROTAC applications remains less explored. Here, we discovered a cysteine-reactive covalent recruiter EN884 against the SKP1 adapter protein of the SKP1-CUL1-F-box containing SCF complex. We further showed that this recruiter can be used in PROTAC applications to degrade neo-substrate proteins such as BRD4 and the androgen receptor in a SKP1- and proteasome-dependent manner. Our studies demonstrate that core and essential adapter proteins within the Cullin-RING E3 ubiquitin ligase complex can be exploited for targeted protein degradation applications and that covalent chemoproteomic strategies can enable recruiter discovery against these targets.
    Language English
    Publishing date 2023-11-02
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.10.20.563371
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  9. Article: Targeted Protein Degradation through Recruitment of the CUL4A Complex Adaptor Protein DDB1.

    Meyers, Margot / Cismoski, Sabine / Panidapu, Anoohya / Chie-Leon, Barbara / Nomura, Daniel K

    bioRxiv : the preprint server for biology

    2023  

    Abstract: Targeted protein degradation has arisen as a powerful therapeutic modality for eliminating proteins. Thus far, most heterobifunctional Proteolysis Targeting Chimeras (PROTACs) have utilized recruiters against substrate receptors of Cullin RING E3 ... ...

    Abstract Targeted protein degradation has arisen as a powerful therapeutic modality for eliminating proteins. Thus far, most heterobifunctional Proteolysis Targeting Chimeras (PROTACs) have utilized recruiters against substrate receptors of Cullin RING E3 ubiquitin ligases, such as cereblon and VHL. However, previous studies have surprisingly uncovered molecular glue degraders that exploit a CUL4A adaptor protein DDB1 to degrade neosubstrate proteins. Here, we sought to investigate whether DDB1 recruiters can be discovered that can be exploited for PROTAC applications. We utilized activity-based protein profiling and cysteine chemoproteomic screening to identify a covalent recruiter that targets C173 on DDB1 and exploited this recruiter to develop PROTACs against BRD4 and androgen receptor (AR). We demonstrated that the BRD4 PROTAC results in selective degradation of the short BRD4 isoform over the long isoform in a proteasome, NEDDylation, and DDB1-dependent manner. We also demonstrated degradation of AR with the AR PROTAC in prostate cancer cells. Our study demonstrated that covalent chemoproteomic approaches can be used to discover recruiters against Cullin RING adapter proteins and that these recruiters can be used for PROTAC applications to degrade neo-substrates.
    Language English
    Publishing date 2023-08-12
    Publishing country United States
    Document type Preprint
    DOI 10.1101/2023.08.11.553046
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

  10. Article ; Online: Ligandability of E3 Ligases for Targeted Protein Degradation Applications.

    Belcher, Bridget P / Ward, Carl C / Nomura, Daniel K

    Biochemistry

    2021  Volume 62, Issue 3, Page(s) 588–600

    Abstract: Targeted protein degradation (TPD) using proteolysis targeting chimeras (PROTACs) and molecular glue degraders has arisen as a powerful therapeutic modality for eliminating disease-causing proteins from cells. PROTACs and molecular glue degraders employ ... ...

    Abstract Targeted protein degradation (TPD) using proteolysis targeting chimeras (PROTACs) and molecular glue degraders has arisen as a powerful therapeutic modality for eliminating disease-causing proteins from cells. PROTACs and molecular glue degraders employ heterobifunctional or monovalent small molecules, respectively, to chemically induce the proximity of target proteins with E3 ubiquitin ligases to ubiquitinate and degrade specific proteins via the proteasome. Whereas TPD is an attractive therapeutic strategy for expanding the druggable proteome, only a relatively small number of E3 ligases out of the >600 E3 ligases encoded by the human genome have been exploited by small molecules for TPD applications. Here we review the existing E3 ligases that have thus far been successfully exploited for TPD and discuss chemoproteomics-enabled covalent screening strategies for discovering new E3 ligase recruiters. We also provide a chemoproteomic map of reactive cysteines within hundreds of E3 ligases that may represent potential ligandable sites that can be pharmacologically interrogated to uncover additional E3 ligase recruiters.
    MeSH term(s) Humans ; Ubiquitin-Protein Ligases/metabolism ; Proteolysis ; Proteins/metabolism ; Proteasome Endopeptidase Complex/metabolism ; Ubiquitination
    Chemical Substances Ubiquitin-Protein Ligases (EC 2.3.2.27) ; Proteins ; Proteasome Endopeptidase Complex (EC 3.4.25.1)
    Language English
    Publishing date 2021-09-02
    Publishing country United States
    Document type Journal Article ; Review ; Research Support, Non-U.S. Gov't
    ZDB-ID 1108-3
    ISSN 1520-4995 ; 0006-2960
    ISSN (online) 1520-4995
    ISSN 0006-2960
    DOI 10.1021/acs.biochem.1c00464
    Database MEDical Literature Analysis and Retrieval System OnLINE

    More links

    Kategorien

To top