223 results on '"K Nomura"'
Search Results
2. Correction to 'Rational Chemical Design of Molecular Glue Degraders'
- Author
-
Ethan S. Toriki, James W. Papatzimas, Kaila Nishikawa, Dustin Dovala, Andreas O. Frank, Matthew J. Hesse, Daniela Dankova, Jae-Geun Song, Megan Bruce-Smythe, Heidi Struble, Francisco J. Garcia, Scott M. Brittain, Andrew C. Kile, Lynn M. McGregor, Jeffrey M. McKenna, John A. Tallarico, Markus Schirle, and Daniel K. Nomura
- Subjects
Chemistry ,QD1-999 - Published
- 2023
- Full Text
- View/download PDF
3. Ligandability of E3 Ligases for Targeted Protein Degradation Applications
- Author
-
Daniel K. Nomura, Bridget P. Belcher, and Carl C. Ward
- Subjects
biology ,medicine.diagnostic_test ,Chemistry ,Proteolysis ,Druggability ,Computational biology ,Protein degradation ,Biochemistry ,Small molecule ,Article ,Ubiquitin ligase ,Ubiquitin ,Proteasome ,Proteome ,biology.protein ,medicine - 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. While 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 which may represent potential ligandable sites that can be pharmacologically interrogated to uncover additional E3 ligase recruiters.
- Published
- 2021
- Full Text
- View/download PDF
4. Manumycin Polyketides Act as Molecular Glues Between UBR7 and P53
- Author
-
Scott M. Brittain, Xiaoyou Liang, Mikiko Okumura, Lynn M. McGregor, Yosuke Isobe, Daniel K. Nomura, Thomas J. Maimone, Ross White, Markus Schirle, William C. Forrester, John A. Tallarico, Michael D. Jones, and Jeffrey Mckenna
- Subjects
natural products ,Molecular Conformation ,chemistry.chemical_compound ,molecular glues ,Drug Discovery ,0303 health sciences ,Tumor ,biology ,Molecular Structure ,Chemistry ,030302 biochemistry & molecular biology ,Limiting ,Small molecule ,Ubiquitin ligase ,Gene Expression Regulation, Neoplastic ,Cross-Linking Reagents ,Gene Knockdown Techniques ,Female ,Biochemistry & Molecular Biology ,Polyunsaturated Alkamides ,Ubiquitin-Protein Ligases ,Static Electricity ,Breast Neoplasms ,Antineoplastic Agents ,covalent ligands ,Computational biology ,Polyenes ,manumycin ,Article ,Cell Line ,03 medical and health sciences ,Structure-Activity Relationship ,Medicinal and Biomolecular Chemistry ,E3 ligases ,Cell Line, Tumor ,Humans ,Chemoproteomics ,asukamycin ,Molecular Biology ,protein-protein interaction (PPI) ,undruggable ,030304 developmental biology ,activity-based protein profiling ,Neoplastic ,Natural product ,Cell Biology ,Anticancer mechanism ,chemoproteomics ,Gene Expression Regulation ,Polyketides ,biology.protein ,Breast cancer cells ,Biochemistry and Cell Biology ,Tumor Suppressor Protein p53 - Abstract
Molecular glues are an intriguing therapeutic modality that harness small-molecules to induce interactions between proteins that typically do not interact. However, such molecules are rare and have been discovered fortuitously, thus limiting their potential as a general strategy for therapeutic intervention. We postulated that natural products bearing one or more electrophilic sites may be an unexplored source of new molecular glues, potentially acting through multi-covalent attachment. Using chemoproteomic platforms, we show that members of the manumycin family of polyketides, which bear multiple potentially reactive sites, target C374 of the putative E3 ligase UBR7 in breast cancer cells and engage in molecular glue interactions with the neo-substrate tumor-suppressor TP53, leading to p53 transcriptional activation and cell death. Our results reveal a novel anti-cancer mechanism of this natural product family and highlight the potential for combining chemoproteomics and multi-covalent natural products for the discovery of new molecular glues.
- Published
- 2020
5. Chemical investigations into the biosynthesis of the gymnastatin and dankastatin alkaloids
- Author
-
Bingqi Tong, Thomas J. Maimone, Bridget P. Belcher, and Daniel K. Nomura
- Subjects
Natural product ,Gymnascella ,biology ,010405 organic chemistry ,Stereochemistry ,Growth inhibitory ,General Chemistry ,010402 general chemistry ,biology.organism_classification ,01 natural sciences ,0104 chemical sciences ,chemistry.chemical_compound ,Chemistry ,chemistry ,Biosynthesis ,Covalent bond ,Clinical Research ,Electrophile ,Breast Cancer ,Chemical Sciences ,Tyrosine ,Cytotoxicity ,Cancer - Abstract
Electrophilic natural products have provided fertile ground for understanding how nature inhibits protein function using covalent bond formation. The fungal strain Gymnascella dankaliensis has provided an especially interesting collection of halogenated cytotoxic agents derived from tyrosine which feature an array of reactive functional groups. Herein we explore chemical and potentially biosynthetic relationships between architecturally complex gymnastatin and dankastatin members, finding conditions that favor formation of a given scaffold from a common intermediate. Additionally, we find that multiple natural products can also be formed from aranorosin, a non-halogenated natural product also produced by Gymnascella sp. fungi, using simple chloride salts thus offering an alternative hypothesis for the origins of these compounds in nature. Finally, growth inhibitory activity of multiple members against human triple negative breast cancer cells is reported., Total synthesis sheds light on biosynthetic relationships among the chlorinated gymnastatin and dankastatin alkaloids.
- Published
- 2021
6. ER–lysosome contacts enable cholesterol sensing by mTORC1 and drive aberrant growth signalling in Niemann–Pick type C
- Author
-
Daniel K. Nomura, Roberto Zoncu, Hijai R. Shin, Xuntian Jiang, Justin Zhang, Charles A. Berdan, Daniel S. Ory, Oliver B. Davis, Chun Yan Lim, and Jessica L. Counihan
- Subjects
Receptors, Steroid ,Vesicular Transport Proteins ,GTPase ,Mechanistic Target of Rapamycin Complex 1 ,Endoplasmic Reticulum ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Niemann-Pick C1 Protein ,Lysosome ,medicine ,Animals ,Humans ,OSBP ,030304 developmental biology ,Niemann-Pick Diseases ,0303 health sciences ,Membrane Glycoproteins ,Chemistry ,Endoplasmic reticulum ,Intracellular Signaling Peptides and Proteins ,Cell Biology ,VAPB ,Cell biology ,Cholesterol ,HEK293 Cells ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,lipids (amino acids, peptides, and proteins) ,biological phenomena, cell phenomena, and immunity ,NPC1 ,Signal transduction ,Carrier Proteins ,Lysosomes ,Oxysterol-binding protein ,Signal Transduction - Abstract
Cholesterol activates the master growth regulator, mTORC1 kinase, by promoting its recruitment to the surface of lysosomes by the Rag guanosine triphosphatases (GTPases). The mechanisms that regulate lysosomal cholesterol content to enable mTORC1 signalling are unknown. Here, we show that oxysterol binding protein (OSBP) and its anchors at the endoplasmic reticulum (ER), VAPA and VAPB, deliver cholesterol across ER-lysosome contacts to activate mTORC1. In cells lacking OSBP, but not other VAP-interacting cholesterol carriers, the recruitment of mTORC1 by the Rag GTPases is inhibited owing to impaired transport of cholesterol to lysosomes. By contrast, OSBP-mediated cholesterol trafficking drives constitutive mTORC1 activation in a disease model caused by the loss of the lysosomal cholesterol transporter, Niemann-Pick C1 (NPC1). Chemical and genetic inactivation of OSBP suppresses aberrant mTORC1 signalling and restores autophagic function in cellular models of Niemann-Pick type C (NPC). Thus, ER-lysosome contacts are signalling hubs that enable cholesterol sensing by mTORC1, and targeting the sterol-transfer activity of these signalling hubs could be beneficial in patients with NPC.
- Published
- 2019
- Full Text
- View/download PDF
7. Covalent targeting of the vacuolar H+-ATPase activates autophagy via mTORC1 inhibition
- Author
-
Carl C. Ward, James A. Olzmann, Roberto Zoncu, Charles A. Berdan, Hijai R. Shin, Clive Yik-Sham Chung, Daniel K. Nomura, and Breanna Ford
- Subjects
autophagy ,Vacuolar Proton-Translocating ATPases ,Biochemistry & Molecular Biology ,Proto-Oncogene Proteins c-akt ,Protein subunit ,Cellular homeostasis ,Guanosine ,mTORC1 ,Neurodegenerative ,Mechanistic Target of Rapamycin Complex 1 ,Article ,Cell Line ,Mice ,Medicinal and Biomolecular Chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Autophagy ,Animals ,Humans ,cysteine ,Molecular Biology ,activity-based protein profiling ,030304 developmental biology ,0303 health sciences ,Molecular Structure ,Activator (genetics) ,Kinase ,030302 biochemistry & molecular biology ,Cell Biology ,chemoproteomics ,3. Good health ,Cell biology ,v-ATPase ,Gene Expression Regulation ,chemistry ,Gene Knockdown Techniques ,covalent ligand ,lysosome ,Pyrazoles ,Biochemistry and Cell Biology ,ATP6V1A ,biological phenomena, cell phenomena, and immunity - Abstract
Autophagy is a lysosomal degradation pathway that eliminates aggregated proteins and damaged organelles to maintain cellular homeostasis. A major route for activating autophagy involves inhibition of the mTORC1 kinase, but current mTORC1-targeting compounds do not allow complete and selective mTORC1 blockade. Here, we have coupled screening of a covalent ligand library with activity-based protein profiling to discover EN6, a small-molecule in vivo activator of autophagy that covalently targets cysteine 277 in the ATP6V1A subunit of the lysosomal v-ATPase, which activates mTORC1 via the Rag guanosine triphosphatases. EN6-mediated ATP6V1A modification decouples the v-ATPase from the Rags, leading to inhibition of mTORC1 signaling, increased lysosomal acidification, and activation of autophagy. Consistently, EN6 clears TDP-43 aggregates, a causative agent in frontotemporal dementia, in a lysosome-dependent manner. Our results provide insight into how the v-ATPase regulates mTORC1, and reveal a unique approach for enhancing cellular clearance based on covalent inhibition of lysosomal mTORC1 signaling., Editorial Summary We report here a covalent ligand that targets C277 of ATP6V1A leading to enhanced v-ATPase activity, inhibition of mTORC1 signaling, increasesd lysosomal acidification, activation of autophagy, and clearance of toxic protein aggregates.
- Published
- 2019
- Full Text
- View/download PDF
8. Oncogene Regulated Release of Extracellular Vesicles
- Author
-
Rebekka Paisner, Suprit Gupta, Seda Kilinc, Rushika M. Perera, Rebecca A. Kohnz, Noelle D. L'Etoile, Daniel K. Nomura, Roman Camarda, Olga Momcilovic, Andrei Goga, and Baris Avsaroglu
- Subjects
oncogenes ,MYC ,Medical and Health Sciences ,chemistry.chemical_compound ,0302 clinical medicine ,2.1 Biological and endogenous factors ,Aurora Kinase B ,Aetiology ,ras ,Cancer ,0303 health sciences ,AURKB ,Extracellular vesicle ,Biological Sciences ,MAP Kinase Kinase Kinases ,Cell biology ,Gene Expression Regulation, Neoplastic ,medicine.anatomical_structure ,HRAS ,miRNAs ,lysosome ,Signal Transduction ,Ceramide ,MAP Kinase Signaling System ,Context (language use) ,Biology ,General Biochemistry, Genetics and Molecular Biology ,ESCRT ,Article ,Proto-Oncogene Proteins c-myc ,03 medical and health sciences ,Extracellular Vesicles ,Affordable and Clean Energy ,Downregulation and upregulation ,Lysosome ,Genetics ,medicine ,Humans ,ceramide ,EVs ,Molecular Biology ,030304 developmental biology ,Neoplastic ,Oncogene ,Cell Biology ,Oncogenes ,Genes, ras ,Metabolism ,Gene Expression Regulation ,Genes ,chemistry ,Cancer cell ,Generic health relevance ,Energy Metabolism ,Lysosomes ,030217 neurology & neurosurgery ,Developmental Biology - Abstract
Oncogenes can alter metabolism by changing the balance between anabolic and catabolic processes. However, how oncogenes regulate tumor cell biomass remains poorly understood. Using isogenic MCF10A cells transformed with nine different oncogenes, we show that specific oncogenes reduce the biomass of cancer cells by promoting extracellular vesicle (EV) release. While MYC and AURKB elicited the highest number of EVs, each oncogene selectively altered the protein composition of released EVs. Likewise, oncogenes alter secreted miRNAs. MYC-overexpressing cells require ceramide, whereas AURKB requires ESCRT to release high levels of EVs. We identify an inverse relationship between MYC upregulation and activation of the RAS/MEK/ERK signaling pathway for regulating EV release in some tumor cells. Finally, lysosome genes and activity are downregulated in the context of MYC and AURKB, suggesting that cellular contents, instead of being degraded, were released via EVs. Thus, oncogene-mediated biomass regulation via differential EV release is a new metabolic phenotype.
- Published
- 2021
9. Adhesion-mediated mechanosignaling forces mitohormesis
- Author
-
Valerie M. Weaver, Andrew S. Moore, Danielle L. Swaney, Jonathon M. Muncie, Andrew Dillin, Alexander R. Dunn, Carlos Garzon-Coral, Kuei-Ho Chen, Breanna Ford, Catherine Schneider, Joseph R. Daniele, Hao Shao, Marc Hellerstein, Sagar S. Manoli, Phillip A. Frankino, Daniel K. Nomura, Jason E. Gestwicki, Kevin M. Tharp, Gregory M. Ku, Greg A. Timblin, Nevan J. Krogan, Karou Saijo, Connor Stashko, Alicia L. Richards, and Ryo Higuchi-Sanabria
- Subjects
0301 basic medicine ,Integrins ,Aging ,Mechanotransduction ,Physiology ,mechanotabolism ,Mitochondrion ,Medical Biochemistry and Metabolomics ,medicine.disease_cause ,Mitochondrial Dynamics ,Mechanotransduction, Cellular ,Fight-or-flight response ,Extracellular matrix ,Animals, Genetically Modified ,Mice ,0302 clinical medicine ,Transcription (biology) ,UPRmt ,oxidative stress ,Cells, Cultured ,0303 health sciences ,Microscopy ,Microscopy, Confocal ,Cultured ,Sodium-Hydrogen Exchanger 1 ,Chemistry ,Middle Aged ,tension ,Phenotype ,Cell biology ,Extracellular Matrix ,Mitochondria ,Ion Exchange ,adhesion ,030220 oncology & carcinogenesis ,Confocal ,Female ,Signal Transduction ,Adult ,Programmed cell death ,Cells ,extracellular matrix ,1.1 Normal biological development and functioning ,Cell Respiration ,Genetically Modified ,Time-Lapse Imaging ,Article ,03 medical and health sciences ,Endocrinology & Metabolism ,Underpinning research ,medicine ,Cell Adhesion ,Animals ,Humans ,cancer ,Caenorhabditis elegans ,Molecular Biology ,Eukaryotic cell ,030304 developmental biology ,mechanical stress ,Cell Biology ,Solute carrier family ,Heat shock factor ,Oxidative Stress ,030104 developmental biology ,HEK293 Cells ,Ageing ,Hyperglycemia ,Cellular ,Generic health relevance ,Biochemistry and Cell Biology ,Reactive Oxygen Species ,metabolism ,030217 neurology & neurosurgery ,Oxidative stress ,Function (biology) - Abstract
Mitochondria control eukaryotic cell fate by producing the energy needed to support life and the signals required to execute programmed cell death. The biochemical milieu is known to affect mitochondrial function and contribute to the dysfunctional mitochondrial phenotypes implicated in cancer and the morbidities of ageing. However, the physical characteristics of the extracellular matrix are also altered in cancer and in aging tissues. We demonstrate that cells sense the physical properties of the extracellular matrix and activate a mitochondrial stress response that adaptively tunes mitochondrial function via SLC9A1-dependent ion exchange and HSF1-dependent transcription. Overall, our data indicate that adhesion-mediated mechanosignaling may play an unappreciated role in the altered mitochondrial functions observed in aging and cancer.Graphical Abstract
- Published
- 2021
10. Deubiquitinase-Targeting Chimeras for Targeted Protein Stabilization
- Author
-
Nathaniel J. Henning, Scott M. Brittain, Lynn M. McGregor, John A. Tallarico, Hesse M, Carl C. Ward, Bridget P. Belcher, Lydia Boike, Jessica N. Spradlin, Markus Schirle, Jeffery M. McKenna, Dustin Dovala, and Daniel K. Nomura
- Subjects
chemistry.chemical_compound ,Ubiquitin ,biology ,Chemistry ,OTUB1 ,Allosteric regulation ,Lumacaftor ,biology.protein ,Protein degradation ,Protein stabilization ,Ligand (biochemistry) ,Cell biology ,Deubiquitinating enzyme - Abstract
Targeted protein degradation is a powerful therapeutic modality that uses heterobifunctional small-molecules to induce proximity between E3 ubiquitin ligases and target proteins to ubiquitinate and degrade specific proteins of interest. However, many proteins are ubiquitinated and degraded to drive disease pathology; in these cases targeted protein stabilization (TPS), rather than degradation, of the actively degraded target using a small-molecule would be therapeutically beneficial. Here, we present the Deubiquitinase-Targeting Chimera (DUBTAC) platform for TPS of specific proteins. Using chemoproteomic approaches, we discovered the covalent ligand EN523 that targets a non-catalytic allosteric cysteine C23 in the K48 ubiquitin-specific deubiquitinase OTUB1. We then developed a heterobifunctional DUBTAC consisting of our EN523 OTUB1 recruiter linked to lumacaftor, a drug used to treat cystic fibrosis that binds ΔF508-CFTR. We demonstrated proof-of-concept of TPS by showing that this DUBTAC robustly stabilized ΔF508-CFTR in human cystic fibrosis bronchial epithelial cells in an OTUB1-dependent manner. Our study underscores the utility of chemoproteomics-enabled covalent ligand discovery approaches to develop new induced proximity-based therapeutic modalities and introduces the DUBTAC platform for TPS.Editorial summaryWe have developed the Deubiquitinase Targeting Chimera (DUBTAC) platform for targeted protein stabilization. We have discovered a covalent recruiter against the deubiquitinase OTUB1 that we have linked to the mutant ΔF508-CFTR targeting cystic fibrosis drug Lumacaftor to stabilize mutant CFTR protein in cells.
- Published
- 2021
- Full Text
- View/download PDF
11. Discovery of a Covalent FEM1B Recruiter for Targeted Protein Degradation Applications
- Author
-
John A. Tallarico, Jessica N. Spradlin, Scott M. Brittain, Michael Rape, Markus Schirle, Daniel K. Nomura, Jeffery M. McKenna, Andrew G. Manford, and Nathaniel J. Henning
- Subjects
BRD4 ,biology ,Covalent bond ,Drug discovery ,Chemistry ,Ligand ,biology.protein ,Binding site ,Protein degradation ,Combinatorial chemistry ,Ubiquitin ligase ,Bromodomain - Abstract
Proteolysis Targeting Chimeras (PROTACs), heterobifunctional compounds that consist of protein-targeting ligands linked to an E3 ligase recruiter, have arisen as a powerful therapeutic modality for targeted protein degradation (TPD). Despite the popularity of TPD approaches in drug discovery, only a small number of E3 ligase recruiters are available for the >600 E3 ligases that exist in human cells. Here, we have discovered a cysteine-reactive covalent ligand, EN106, that targets FEM1B, an E3 ligase recently discovered as the critical component of the cellular response to reductive stress. By targeting Cys186 in FEM1B, EN106 disrupts recognition of the key reductive stress substrate of FEM1B, FNIP1. We further establish that EN106 can be used as a covalent recruiter for FEM1B in TPD applications, in which we demonstrate that a PROTAC linking EN106 to the BET Bromodomain inhibitor JQ1 leads to specific FEM1B- and proteasome-dependent degradation of BRD4 in cells. Our study showcases a covalent ligand that targets a natural E3 ligase-substrate binding site and highlights the utility of covalent ligand screening in expanding the arsenal of E3 ligase recruiters that can be deployed for TPD applications.
- Published
- 2021
- Full Text
- View/download PDF
12. Lipidome-based Targeting of STAT3-driven Breast Cancer Cells Using Poly-l-glutamic Acid–coated Layer-by-Layer Nanoparticles
- Author
-
Isidora Tošić, Lisa N. Heppler, Susana P. Egusquiaguirre, Douglas S. Richardson, Sharmistha Pal, Paula T. Hammond, Daniel F. Costa, Daphne A. Haas-Kogan, Elizabeth A. Grossman Moore, David A. Frank, Natalie Boehnke, Daniel K. Nomura, Alexander R. Ivanov, and Santiago Correa
- Subjects
0301 basic medicine ,STAT3 Transcription Factor ,Cancer Research ,Glutamic Acid ,Triple Negative Breast Neoplasms ,Article ,Cell membrane ,03 medical and health sciences ,0302 clinical medicine ,Breast cancer ,Drug Delivery Systems ,medicine ,Humans ,Cytotoxicity ,STAT3 ,Transcription factor ,Cisplatin ,biology ,Chemistry ,Lipidome ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,Oncology ,Apoptosis ,030220 oncology & carcinogenesis ,Lipidomics ,Cancer research ,biology.protein ,Nanoparticles ,medicine.drug - Abstract
The oncogenic transcription factor STAT3 is aberrantly activated in 70% of breast cancers, including nearly all triple-negative breast cancers (TNBCs). Because STAT3 is difficult to target directly, we considered whether metabolic changes driven by activated STAT3 could provide a therapeutic opportunity. We found that STAT3 prominently modulated several lipid classes, with most profound effects on N-acyl taurine and arachidonic acid, both of which are involved in plasma membrane remodeling. To exploit these metabolic changes therapeutically, we screened a library of layer-by-layer (LbL) nanoparticles (NPs) differing in the surface layer that modulates interactivity with the cell membrane. We found that poly-l-glutamic acid (PLE)–coated NPs bind to STAT3-transformed breast cancer cells with 50% greater efficiency than to nontransformed cells, and the heightened PLE-NP binding to TNBC cells was attenuated by STAT3 inhibition. This effect was also observed in densely packed three-dimensional breast cancer organoids. As STAT3-transformed cells show greater resistance to cytotoxic agents, we evaluated whether enhanced targeted delivery via PLE-NPs would provide a therapeutic advantage. We found that cisplatin-loaded PLE-NPs induced apoptosis of STAT3-driven cells at lower doses compared with both unencapsulated cisplatin and cisplatin-loaded nontargeted NPs. In addition, because radiation is commonly used in breast cancer treatment, and may alter cellular lipid distribution, we analyzed its effect on PLE-NP–cell binding. Irradiation of cells enhanced the STAT3-targeting properties of PLE-NPs in a dose-dependent manner, suggesting potential synergies between these therapeutic modalities. These findings suggest that cellular lipid changes driven by activated STAT3 may be exploited therapeutically using unique LbL NPs.
- Published
- 2021
13. Mitohormesis reprograms macrophage metabolism to enforce tolerance
- Author
-
Andreas Stahl, Janet M. Winchenster, Stella Zhu, Jerome Wang, Johanna ten Hoeve, Kevin M. Tharp, Greg A. Timblin, Shannon K. Louie, Rida I. Khan, Daniel K. Nomura, Anthony T. Iavarone, Kaoru Saijo, and Breanna Ford
- Subjects
Cell signaling ,biology ,Lipopolysaccharide ,Inflammation ,Mitochondrion ,Proinflammatory cytokine ,Cell biology ,chemistry.chemical_compound ,Histone ,chemistry ,medicine ,biology.protein ,Macrophage ,medicine.symptom ,Proto-oncogene tyrosine-protein kinase Src - Abstract
Macrophages generate mitochondrial reactive oxygen and electrophilic species (mtROS, mtRES) as antimicrobials during Toll-like receptor (TLR)-dependent inflammatory responses. Whether mitochondrial stress caused by these molecules impacts macrophage function is unknown. Here we demonstrate that both pharmacologically- and lipopolysaccharide (LPS)-driven mitochondrial stress in macrophages triggers a stress response called mitohormesis. LPS-driven mitohormetic stress adaptations occur as macrophages transition from an LPS-responsive to LPS-tolerant state where stimulus-induced proinflammatory gene transcription is impaired, suggesting tolerance is a product of mitohormesis. Indeed, like LPS, pharmacologically-triggered mitohormesis suppresses mitochondrial oxidative metabolism and acetyl-CoA production needed for histone acetylation and proinflammatory gene transcription, and is sufficient to enforce an LPS-tolerant state. Thus, mtROS and mtRES are TLR-dependent signaling molecules that trigger mitohormesis as a negative feedback mechanism to restrain inflammation via tolerance. Moreover, bypassing TLR signaling and pharmacologically triggering mitohormesis represents a novel anti-inflammatory strategy that co-opts this stress response to impair epigenetic support of proinflammatory gene transcription by mitochondria.Abstract Figure
- Published
- 2020
- Full Text
- View/download PDF
14. Bardoxolone conjugation enables targeted protein degradation of BRD4
- Author
-
Thomas J. Maimone, Jeffrey Mckenna, Jessica N. Spradlin, Yi Xie, John A. Tallarico, Mai Luo, Daniel K. Nomura, Markus Schirle, and Bingqi Tong
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Ubiquitin-Protein Ligases ,Chemical biology ,lcsh:Medicine ,Organic chemistry ,Medicinal chemistry ,Cell Cycle Proteins ,Plasma protein binding ,Protein degradation ,01 natural sciences ,Article ,Cell Line ,03 medical and health sciences ,chemistry.chemical_compound ,Cell Line, Tumor ,Drug Discovery ,Humans ,Chemical synthesis ,Oleanolic Acid ,Bifunctional ,lcsh:Science ,Natural products ,Multidisciplinary ,Tumor ,biology ,010405 organic chemistry ,Drug discovery ,lcsh:R ,Small molecules ,Ubiquitination ,Azepines ,Triazoles ,Combinatorial chemistry ,Small molecule ,0104 chemical sciences ,Ubiquitin ligase ,030104 developmental biology ,chemistry ,Proteolysis ,biology.protein ,lcsh:Q ,Bardoxolone ,Chemical tools ,Transcription Factors - Abstract
Targeted protein degradation (TPD) has emerged as a powerful tool in drug discovery for the perturbation of protein levels using heterobifunctional small molecules. E3 ligase recruiters remain central to this process yet relatively few have been identified relative to the ~ 600 predicted human E3 ligases. While, initial recruiters have utilized non-covalent chemistry for protein binding, very recently covalent engagement to novel E3’s has proven fruitful in TPD application. Herein we demonstrate efficient proteasome-mediated degradation of BRD4 by a bifunctional small molecule linking the KEAP1-Nrf2 activator bardoxolone to a BRD4 inhibitor JQ1.
- Published
- 2020
15. 4β-hydroxycholesterol is a pro-lipogenic factor that promotes SREBP1c expression and activity through Liver X-receptor
- Author
-
Daniel S. Ory, Peter-James H. Zushin, Xuntian Jiang, Mingxing Qian, Daniel K. Nomura, Ofer Moldavski, Ethan J. Weiss, Charles A. Berdan, Robert J. van Eijkeren, Douglas F. Covey, Andreas Stahl, and Roberto Zoncu
- Subjects
Oxysterol ,Chemistry ,Lipid biosynthesis ,Lipogenesis ,polycyclic compounds ,Transcriptional regulation ,Regulator ,lipids (amino acids, peptides, and proteins) ,Liver X receptor ,Beta (finance) ,Transcription factor ,Cell biology - Abstract
Oxysterols are oxidized derivatives of cholesterol that play signaling roles in lipid biosynthesis and homeostasis. Here we show that 4β-hydroxycholesterol (4β-HC), a liver and serum abundant oxysterol of poorly defined function, is a potent and selective inducer of the master lipogenic transcription factor, Sterol Regulatory Element Binding Protein 1c (SREBP1c), but not the related steroidogenic transcription factor SREBP2. Mechanistically, 4β-HC acts as a putative agonist for Liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c, and triggeredde novolipogenesis both in primary hepatocytes and in mouse liver. 4β-HC-acted in parallel to insulin-PI3K-dependent signaling to stimulate triglyceride synthesis and lipid droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
- Published
- 2020
- Full Text
- View/download PDF
16. Mitohormesis reprogrammes macrophage metabolism to enforce tolerance
- Author
-
Stella Zhu, Kevin M. Tharp, Kaoru Saijo, Breanna Ford, Rida I. Khan, Daniel K. Nomura, Johanna ten Hoeve, Shannon K. Louie, Anthony T. Iavarone, Janet M. Winchester, Greg A. Timblin, Andreas Stahl, and Jerome Wang
- Subjects
Lipopolysaccharides ,Lipopolysaccharide ,Endocrinology, Diabetes and Metabolism ,Anti-Inflammatory Agents ,Inflammation ,Mitochondrion ,Models, Biological ,Article ,chemistry.chemical_compound ,Acetyl Coenzyme A ,Stress, Physiological ,Physiology (medical) ,Internal Medicine ,medicine ,Immune Tolerance ,Macrophage ,Humans ,Epigenetics ,chemistry.chemical_classification ,Reactive oxygen species ,biology ,Chemistry ,Macrophages ,Estrogens ,Cell Biology ,Macrophage Activation ,Cellular Reprogramming ,Cell biology ,Mitochondria ,Histone ,Gene Expression Regulation ,Acetylation ,biology.protein ,medicine.symptom ,Energy Metabolism ,Reactive Oxygen Species - Abstract
Macrophages generate mitochondrial reactive oxygen species and mitochondrial reactive electrophilic species as antimicrobials during Toll-like receptor (TLR)-dependent inflammatory responses. Whether mitochondrial stress caused by these molecules impacts macrophage function is unknown. Here, we demonstrate that both pharmacologically driven and lipopolysaccharide (LPS)-driven mitochondrial stress in macrophages triggers a stress response called mitohormesis. LPS-driven mitohormetic stress adaptations occur as macrophages transition from an LPS-responsive to LPS-tolerant state wherein stimulus-induced pro-inflammatory gene transcription is impaired, suggesting tolerance is a product of mitohormesis. Indeed, like LPS, hydroxyoestrogen-triggered mitohormesis suppresses mitochondrial oxidative metabolism and acetyl-CoA production needed for histone acetylation and pro-inflammatory gene transcription, and is sufficient to enforce an LPS-tolerant state. Thus, mitochondrial reactive oxygen species and mitochondrial reactive electrophilic species are TLR-dependent signalling molecules that trigger mitohormesis as a negative feedback mechanism to restrain inflammation via tolerance. Moreover, bypassing TLR signalling and pharmacologically triggering mitohormesis represents a new anti-inflammatory strategy that co-opts this stress response to impair epigenetic support of pro-inflammatory gene transcription by mitochondria.
- Published
- 2020
17. Unbiased Proteomic Profiling Uncovers a Targetable GNAS/PKA/PP2A Axis in Small Cell Lung Cancer Stem Cells
- Author
-
Jaya Sangodkar, Nevan J. Krogan, Myung Chang Lee, Julia Arand, Janos Demeter, Christina S. Kong, Yeonjoo C. Hwang, Michael Ohlmeyer, Rebecca S. Levin, Julie H. Ko, Kevan M. Shokat, Garry L. Coles, John D. Gordan, James T. Webber, Julien Sage, Peter K. Jackson, Brandon Mauch, Steven M. Moss, Yan Ting Shue, Danielle L. Swaney, Nancie Mooney, Vicky Le, Sandra Cristea, Jessica N. Spradlin, Daniel K. Nomura, Goutham Narla, Andy He, and Alexandros P. Drainas
- Subjects
0301 basic medicine ,Proteomics ,cancer stem cells ,Cancer Research ,Lung Neoplasms ,Mice, SCID ,Regenerative Medicine ,Gs ,Mice ,0302 clinical medicine ,Mice, Inbred NOD ,GTP-Binding Protein alpha Subunits, Gs ,2.1 Biological and endogenous factors ,PKA ,Protein Phosphatase 2 ,Aetiology ,Mice, Knockout ,Tumor ,biology ,Kinase ,Chemistry ,Lung Cancer ,SCLC ,humanities ,GTP-Binding Protein alpha Subunits ,PP2A ,Oncology ,030220 oncology & carcinogenesis ,Neoplastic Stem Cells ,Stem cell ,Signal Transduction ,Biotechnology ,kinase ,Knockout ,Oncology and Carcinogenesis ,Antineoplastic Agents ,SCID ,Article ,Cell Line ,lung ,phosphatase ,03 medical and health sciences ,GNAS ,Rare Diseases ,Cancer stem cell ,Cell Line, Tumor ,GNAS complex locus ,Chromogranins ,Animals ,Humans ,cancer ,neuroendocrine ,Oncology & Carcinogenesis ,Protein kinase A ,neoplasms ,Proteomic Profiling ,Neurosciences ,Cell Biology ,Protein phosphatase 2 ,Stem Cell Research ,Cyclic AMP-Dependent Protein Kinases ,Xenograft Model Antitumor Assays ,Small Cell Lung Carcinoma ,respiratory tract diseases ,Transplantation ,030104 developmental biology ,A549 Cells ,Cancer research ,biology.protein ,Inbred NOD ,Cisplatin - Abstract
Summary Using unbiased kinase profiling, we identified protein kinase A (PKA) as an active kinase in small cell lung cancer (SCLC). Inhibition of PKA activity genetically, or pharmacologically by activation of the PP2A phosphatase, suppresses SCLC expansion in culture and in vivo. Conversely, GNAS (G-protein α subunit), a PKA activator that is genetically activated in a small subset of human SCLC, promotes SCLC development. Phosphoproteomic analyses identified many PKA substrates and mechanisms of action. In particular, PKA activity is required for the propagation of SCLC stem cells in transplantation studies. Broad proteomic analysis of recalcitrant cancers has the potential to uncover targetable signaling networks, such as the GNAS/PKA/PP2A axis in SCLC.
- Published
- 2020
18. A Nimbolide-Based Kinase Degrader Preferentially Degrades Oncogenic BCR-ABL
- Author
-
Luiz F. T. Novaes, Xirui Hu, Malte Moeller, Jessica N. Spradlin, Jeffrey Mckenna, Scott M. Brittain, Erika Zhang, Markus Schirle, Lynn M. McGregor, Bingqi Tong, Thomas J. Maimone, Daniel K. Nomura, and John A. Tallarico
- Subjects
0301 basic medicine ,Limonins ,Ubiquitin-Protein Ligases ,Fusion Proteins, bcr-abl ,Protein degradation ,01 natural sciences ,Biochemistry ,Article ,03 medical and health sciences ,chemistry.chemical_compound ,hemic and lymphatic diseases ,medicine ,Humans ,Protein Kinase Inhibitors ,Natural product ,biology ,Oncogene ,010405 organic chemistry ,Chemistry ,Kinase ,Cereblon ,General Medicine ,0104 chemical sciences ,Ubiquitin ligase ,Cell biology ,Dasatinib ,Thiazoles ,030104 developmental biology ,Cancer cell ,Proteolysis ,biology.protein ,Molecular Medicine ,K562 Cells ,medicine.drug - Abstract
Targeted protein degradation (TPD) and proteolysis-targeting chimeras (PROTACs) have arisen as powerful therapeutic modalities for degrading specific proteins in a proteasome-dependent manner. However, a major limitation of TPD is the lack of E3 ligase recruiters. Recently, we discovered the natural product nimbolide as a covalent recruiter for the E3 ligase RNF114. Here, we show the broader utility of nimbolide as an E3 ligase recruiter for TPD applications. We demonstrate that a PROTAC linking nimbolide to the kinase and BCR-ABL fusion oncogene inhibitor dasatinib, BT1, selectively degrades BCR-ABL over c-ABL in leukemia cancer cells, compared to previously reported cereblon or VHL-recruiting BCR-ABL degraders that show opposite selectivity or, in some cases, inactivity. Thus, we further establish nimbolide as an additional general E3 ligase recruiter for PROTACs, and we demonstrate the importance of expanding upon the arsenal of E3 ligase recruiters, as such molecules confer differing selectivity for the degradation of neo-substrate proteins.
- Published
- 2020
19. Oncogene Regulated Release of Extracellular Vesicles
- Author
-
Daniel K. Nomura, Andrei Goga, Seda Kilinc, Rushika M. Perera, Rebecca A. Kohnz, Olga Momcilovic, Rebekka Paisner, Noelle D. L'Etoile, and Roman Camarda
- Subjects
0303 health sciences ,Ceramide ,Cell signaling ,Oncogene ,Context (language use) ,Extracellular vesicle ,ESCRT ,Cell biology ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,chemistry ,030220 oncology & carcinogenesis ,microRNA ,Cancer cell ,030304 developmental biology - Abstract
Oncogenes can alter cellular structure, function, development and metabolism including changing the balance between anabolic and catabolic processes. However, how oncogenes regulate tumor cell biomass remains poorly understood. Using isogenic mammary breast epithelial cells transformed with a panel of ten oncogenes found commonly mutated, amplified or overexpressed in multiple cancers, we show that specific oncogenes reduce the biomass of cancer cells by promoting extracellular vesicle release. While MYC and AURKB elicited the highest number of EVs, each oncogene tested selectively altered the protein composition of released EVs. Likewise, miRNAs were differentially sorted into EVs in an oncogene-specific manner. MYC overexpressing cells require ceramide, while AURKB require ESCRT to release high levels of EVs. Finally, lysosome-associated genes are broadly downregulated in the context of MYC and AURKB, suggesting that cellular contents instead of being degraded, were released via EVs. Thus, oncogene mediated biomass regulation via differential EV release is a new metabolic phenotype which may have implications for cellular signaling and homeostasis.
- Published
- 2020
- Full Text
- View/download PDF
20. A Nimbolide-Based Kinase Degrader Preferentially Degrades Oncogenic BCR-ABL
- Author
-
Bingqi Tong, Daniel K. Nomura, Thomas J. Maimone, Erika Zhang, Lynn M. McGregor, Luiz F. T. Novaes, John A. Tallarico, Xirui Hu, Jessica N. Spradlin, Markus Schirle, Scott M. Brittain, Jeffrey Mckenna, and Malte Moeller
- Subjects
BRD4 ,biology ,Chemistry ,Kinase ,Cereblon ,Protein degradation ,Ubiquitin ligase ,law.invention ,Cell biology ,Dasatinib ,law ,hemic and lymphatic diseases ,Cancer cell ,biology.protein ,medicine ,Suppressor ,medicine.drug - Abstract
Targeted protein degradation (TPD) and proteolysis-targeting chimeras (PROTACs) have arisen as powerful therapeutic modalities for degrading specific protein targets in a proteasome-dependent manner. However, a major limitation to broader TPD applications is the lack of E3 ligase recruiters. Recently, we discovered the natural product nimbolide as a covalent ligand for the E3 ligase RNF114. When linked to the BET family inhibitor JQ1, the resulting heterobifunctional PROTAC molecule was capable of selectively degrading BRD4 in cancer cells. Here, we show the broader utility of nimbolide as an E3 ligase recruiter for TPD applications. We demonstrate that a PROTAC linking nimbolide to the kinase and BCR-ABL fusion oncogene inhibitor dasatinib, BT1, selectively degrades BCR-ABL over c-ABL in leukemia cancer cells, compared to previously reported cereblon or VHL-recruiting BCR-ABL degraders that show opposite selectivity or in some cases inactivity. Further contrasting from cereblon or VHL-recruiting degradation, we show that BT1 treatment not only leads to BCR-ABL degradation, but also stabilizes the endogenous RNF114 substrate and tumor suppressor substrate p21. This leads to additional anti-proliferative effects in leukemia cancer cells beyond those observed with cereblon or VHL-recruiting BCR-ABL PROTACs. Thus, we further establish nimbolide as an additional general E3 ligase recruiter for PROTACs with unique additional benefits for oncology applications. We also further demonstrate the importance of expanding upon the arsenal of E3 ligase recruiters, as such molecules confer differing and unpredictable selectivity for the degradation of neo-substrate proteins.
- Published
- 2020
- Full Text
- View/download PDF
21. Targeted Protein Degradation via a Covalent Reversible Degrader Based on Bardoxolone
- Author
-
Xie Y, Tong B, Luo M, Jessica N. Spradlin, Markus Schirle, Jeffery M. McKenna, Thomas J. Maimone, John A. Tallarico, and Daniel K. Nomura
- Subjects
chemistry.chemical_compound ,biology ,Drug discovery ,Chemistry ,Covalent bond ,biology.protein ,Protein degradation ,Bardoxolone ,Bifunctional ,Combinatorial chemistry ,KEAP1 ,Small molecule ,Ubiquitin ligase - Abstract
Targeted protein degradation (TPD) has emerged as a powerful tool in drug discovery for the perturbation of protein levels using heterobifunctional small molecules (i.e. PROTACs). E3 ligase recruiters remain central to this process yet relatively few have been identified relative to the >500 predicted human E3 ligases. While, initial recruiters have utilized non-covalent chemistry for protein binding, very recently covalent engagement to novel E3’s has proven fruitful in TPD application. Herein we demonstrate efficient proteasome-mediated degradation of BRD4 by a bifunctional small molecule linking the KEAP1-NRF2 activator bardoxolone to a BRD4 inhibitor JQ1. Notably, this work reports the first covalent, reversible E3 ligase recruiter for TPD applications.
- Published
- 2020
- Full Text
- View/download PDF
22. Unexpected transformation of dissolved phenols to toxic dicarbonyls by hydroxyl radicals and UV light
- Author
-
Breanna Ford, David L. Sedlak, Daniel K. Nomura, and Carsten Prasse
- Subjects
0301 basic medicine ,Proteome ,Ultraviolet Rays ,Radical ,Chemical ,010501 environmental sciences ,exposome ,Photochemistry ,01 natural sciences ,reactive transformation products ,Water Purification ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,Phenols ,Animals ,Water Pollutants ,Hydrogen peroxide ,0105 earth and related environmental sciences ,chemistry.chemical_classification ,Reactive oxygen species ,Aldehydes ,Multidisciplinary ,Hydroxyl Radical ,advanced oxidation processes ,Proteins ,water treatment ,chemoproteomics ,6. Clean water ,3. Good health ,Amino acid ,030104 developmental biology ,chemistry ,Liver ,13. Climate action ,Electrophile ,Physical Sciences ,Tyrosine ,Water treatment ,Oxidation-Reduction ,Environmental Sciences ,Water Pollutants, Chemical ,Cysteine - Abstract
Significance Phenols are common anthropogenic and natural chemicals that contaminate drinking water sources. To reduce exposure to these compounds, hydroxyl radicals are often used as chemical oxidants during water treatment. Although this treatment process removes phenols, we have found that it unexpectedly produces toxic transformation products. We identify these products and simultaneously assess their toxicity with a technique that detects products formed when the transformation products react with amino acids and peptides. Our results highlight the potential risks of using oxidative treatment on alternative drinking water sources, such as contaminated groundwater and recycled municipal wastewater. They also suggest that these reactions produce these toxic transformation products in other situations, including in clouds and sunlit surface waters and within living cells., Water treatment systems frequently use strong oxidants or UV light to degrade chemicals that pose human health risks. Unfortunately, these treatments can result in the unintended transformation of organic contaminants into toxic products. We report an unexpected reaction through which exposure of phenolic compounds to hydroxyl radicals (•OH) or UV light results in the formation of toxic α,β-unsaturated enedials and oxoenals. We show that these transformation products damage proteins by reacting with lysine and cysteine moieties. We demonstrate that phenolic compounds react with •OH produced by the increasingly popular UV/hydrogen peroxide (H2O2) water treatment process or UV light to form toxic enedials and oxoenals. In addition to raising concerns about potential health risks of oxidative water treatment, our findings suggest the potential for formation of these toxic compounds in sunlit surface waters, atmospheric water, and living cells. For the latter, our findings may be particularly relevant to efforts to understand cellular damage caused by in vivo production of reactive oxygen species. In particular, we demonstrate that exposure of the amino acid tyrosine to •OH yields an electrophilic enedial product that undergoes cross-linking reaction with both lysine and cysteine residues.
- Published
- 2018
23. Lysosomal cholesterol activates mTORC1 via an SLC38A9–Niemann-Pick C1 signaling complex
- Author
-
Xuntian Jiang, Roberto Zoncu, Daniel K. Nomura, Ofer Moldavski, Laurel Mydock-McGrane, Daniel S. Ory, McKenna Feltes, Oliver B. Davis, Robert J. van Eijkeren, Rushika M. Perera, Brian M. Castellano, Sharon M. Louie, Douglas F. Covey, Ashley M. Thelen, and Reini E. N. van der Welle
- Subjects
0301 basic medicine ,Amino Acid Transport Systems ,HDL ,General Science & Technology ,1.1 Normal biological development and functioning ,Amino Acid Motifs ,CHO Cells ,mTORC1 ,Mechanistic Target of Rapamycin Complex 1 ,Biology ,Article ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,Cricetulus ,Underpinning research ,Lysosome ,medicine ,Animals ,Humans ,Protein kinase A ,Multidisciplinary ,Cell growth ,Cholesterol ,TOR Serine-Threonine Kinases ,Cholesterol, HDL ,Nuclear Proteins ,Biological Transport ,Fibroblasts ,Sterol transport ,Transmembrane protein ,Enzyme Activation ,HEK293 Cells ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Biochemistry ,Multiprotein Complexes ,Mutation ,lipids (amino acids, peptides, and proteins) ,biological phenomena, cell phenomena, and immunity ,NPC1 ,Lysosomes ,Carrier Proteins ,Signal Transduction - Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) protein kinase is a master growth regulator that becomes activated at the lysosome in response to nutrient cues. Here, we identify cholesterol, an essential building block for cellular growth, as a nutrient input that drives mTORC1 recruitment and activation at the lysosomal surface. The lysosomal transmembrane protein, SLC38A9, is required for mTORC1 activation by cholesterol through conserved cholesterol-responsive motifs. Moreover, SLC38A9 enables mTORC1 activation by cholesterol independently from its arginine-sensing function. Conversely, the Niemann-Pick C1 (NPC1) protein, which regulates cholesterol export from the lysosome, binds to SLC38A9 and inhibits mTORC1 signaling through its sterol transport function. Thus, lysosomal cholesterol drives mTORC1 activation and growth signaling through the SLC38A9-NPC1 complex.
- Published
- 2017
- Full Text
- View/download PDF
24. Argininosuccinate Synthase 1 is a Metabolic Regulator of Colorectal Cancer Pathogenicity
- Author
-
Daniel K. Nomura, Leslie A. Bateman, Carlo M. Contreras, Wan-min Ku, Christine F. Skibola, and Martin J. Heslin
- Subjects
0301 basic medicine ,Colorectal cancer ,Metabolite ,Cell ,Argininosuccinate Synthase ,Mouse model of colorectal and intestinal cancer ,Biology ,Bioinformatics ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Metabolomics ,Downregulation and upregulation ,medicine ,Humans ,Enzyme Inhibitors ,Cancer ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Metabolome ,Cancer research ,Molecular Medicine ,Signal transduction ,Colorectal Neoplasms ,Signal Transduction - Abstract
Like many cancer types, colorectal cancers have dysregulated metabolism that promotes their pathogenic features. In this study, we used the activity-based protein profiling chemoproteomic platform to profile cysteine-reactive metabolic enzymes that are upregulated in primary human colorectal tumors. We identified argininosuccinate synthase 1 (ASS1) as an upregulated target in primary human colorectal tumors and show that pharmacological inhibition or genetic ablation of ASS1 impairs colorectal cancer pathogenicity. Using metabolomic profiling, we show that ASS1 inhibition leads to reductions in the levels of oncogenic metabolite fumarate, leading to impairments in glycolytic metabolism that supports colorectal cancer cell pathogenicity. We show here that ASS1 inhibitors may represent a novel therapeutic approach for attenuating colorectal cancer through compromising critical metabolic and metabolite signaling pathways and demonstrate the utility of coupling chemoproteomic and metabolomic strategies to map novel metabolic regulators of cancer.
- Published
- 2017
- Full Text
- View/download PDF
25. Chemoproteomic Screening of Covalent Ligands Reveals UBA5 As a Novel Pancreatic Cancer Target
- Author
-
Carlo M. Contreras, Tucker R. Huffman, Ashley N. Ives, David Akopian, David K. Miyamoto, Allison M. Roberts, Daniel K. Nomura, Christine F. Skibola, Leslie A. Bateman, Martin J. Heslin, and Michael Rape
- Subjects
Proteomics ,0301 basic medicine ,Ubiquitin-activating enzyme ,Druggability ,Antineoplastic Agents ,Ubiquitin-Activating Enzymes ,Biology ,Ligands ,Polymerase Chain Reaction ,01 natural sciences ,Biochemistry ,03 medical and health sciences ,Pancreatic cancer ,medicine ,Humans ,chemistry.chemical_classification ,010405 organic chemistry ,Ligand ,General Medicine ,medicine.disease ,Small molecule ,0104 chemical sciences ,Pancreatic Neoplasms ,030104 developmental biology ,Enzyme ,Mechanism of action ,chemistry ,Gene Knockdown Techniques ,Molecular Medicine ,medicine.symptom - Abstract
Chemical genetic screening of small-molecule libraries has been a promising strategy for discovering unique and novel therapeutic compounds. However, identifying the targets of lead molecules that arise from these screens has remained a major bottleneck in understanding the mechanism of action of these compounds. Here, we have coupled the screening of a cysteine-reactive fragment-based covalent ligand library with an isotopic tandem orthogonal proteolysis-enabled activity-based protein profiling (isoTOP-ABPP) chemoproteomic platform to rapidly couple the discovery of lead small molecules that impair pancreatic cancer pathogenicity with the identification of druggable hotspots for potential cancer therapy. Through this coupled approach, we have discovered a covalent ligand DKM 2-93 that impairs pancreatic cancer cell survival and in vivo tumor growth through covalently modifying the catalytic cysteine of the ubiquitin-like modifier activating enzyme 5 (UBA5), thereby inhibiting its activity as a protein that activates the ubiquitin-like protein UFM1 to UFMylate proteins. We show that UBA5 is a novel pancreatic cancer therapeutic target and show DKM 2-93 as a relatively selective lead inhibitor of UBA5. Our results underscore the utility of coupling the screening of covalent ligand libraries with isoTOP-ABPP platforms for mining the proteome for druggable hotspots for cancer therapy.
- Published
- 2017
- Full Text
- View/download PDF
26. The regulation of glucose and lipid homeostasis via PLTP as a mediator of BAT-liver communication
- Author
-
Daniel K. Nomura, Zachary Brown, Mark P. Jedrychowski, Makoto Takahashi, Yoko Yokoyama, Rachana N. Pradhan, Hiroshi Karasawa, Carlos H.G. Sponton, Qiang Wang, Mitsuhiro Watanabe, Yumi Matsui, Yong Chen, Hiroki Taoka, Kenji Ikeda, Kazuki Tajima, Kosaku Shinoda, Carl C. Ward, Takeshi Yoneshiro, Shingo Kajimura, Lindsay S. Roberts, Junki Taura, Yasuo Oguri, and Takashi Hosono
- Subjects
medicine.medical_specialty ,Glucose uptake ,Biochemistry ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Mediator ,Adipose Tissue, Brown ,Phospholipid transfer protein ,Internal medicine ,Brown adipose tissue ,Genetics ,medicine ,Homeostasis ,Humans ,Molecular Biology ,030304 developmental biology ,0303 health sciences ,Cholesterol ,Thermogenesis ,Articles ,Sphingolipid ,Lipids ,Endocrinology ,medicine.anatomical_structure ,Glucose ,chemistry ,Liver ,Energy Metabolism ,030217 neurology & neurosurgery - Abstract
While brown adipose tissue (BAT) is well-recognized for its ability to dissipate energy in the form of heat, recent studies suggest multifaced roles of BAT in the regulation of glucose and lipid homeostasis beyond stimulating thermogenesis. One of the functions involves interorgan communication with metabolic organs, such as the liver, through BAT-derived secretory factors, a.k.a., batokine. However, the identity and the roles of such mediators remain insufficiently understood. Here, we employed proteomics and transcriptomics in human thermogenic adipocytes and identified previously unappreciated batokines, including phospholipid transfer protein (PLTP). We found that increased circulating levels of PLTP, via systemic or BAT-specific overexpression, significantly improve glucose tolerance and insulin sensitivity, increased energy expenditure, and decrease the circulating levels of cholesterol, phospholipids, and sphingolipids. Such changes were accompanied by increased bile acids in the circulation, which in turn enhances glucose uptake and thermogenesis in BAT. Our data suggest that PLTP is a batokine that contributes to the regulation of systemic glucose and lipid homeostasis as a mediator of BAT-liver interorgan communication.
- Published
- 2019
27. The CoQ oxidoreductase FSP1 acts parallel to GPX4 to inhibit ferroptosis
- Author
-
Joseph M. Hendricks, Breanna Ford, James A. Olzmann, Thomas J. Maimone, Roberto Zoncu, Zhipeng Li, Peter H. Tang, Bingqi Tong, Melissa A. Roberts, Leslie Magtanong, Daniel K. Nomura, Michael C. Bassik, Scott J. Dixon, and Kirill Bersuker
- Subjects
0301 basic medicine ,Enzymologic ,Male ,Programmed cell death ,Lipid Peroxides ,Ubiquinone ,General Science & Technology ,GPX4 ,SCID ,law.invention ,Cell Line ,Mitochondrial Proteins ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,law ,2.1 Biological and endogenous factors ,Animals ,Humans ,Ferroptosis ,Aetiology ,AIFM2 ,Cancer ,Coenzyme Q10 ,Multidisciplinary ,Tumor ,Chemistry ,Cell Membrane ,Glutathione ,Phospholipid Hydroperoxide Glutathione Peroxidase ,Cell biology ,Good Health and Well Being ,030104 developmental biology ,Gene Expression Regulation ,5.1 Pharmaceuticals ,Cell culture ,030220 oncology & carcinogenesis ,Cancer cell ,Suppressor ,Heterografts ,Development of treatments and therapeutic interventions ,Apoptosis Regulatory Proteins - Abstract
Ferroptosis is a form of regulated cell death that is caused by the iron-dependent peroxidation of lipids1,2. The glutathione-dependent lipid hydroperoxidase glutathione peroxidase 4 (GPX4) prevents ferroptosis by converting lipid hydroperoxides into non-toxic lipid alcohols3,4. Ferroptosis has previously been implicated in the cell death that underlies several degenerative conditions2, and induction of ferroptosis by the inhibition of GPX4 has emerged as a therapeutic strategy to trigger cancer cell death5. However, sensitivity to GPX4 inhibitors varies greatly across cancer cell lines6, which suggests that additional factors govern resistance to ferroptosis. Here, using a synthetic lethal CRISPR–Cas9 screen, we identify ferroptosis suppressor protein 1 (FSP1) (previously known as apoptosis-inducing factor mitochondrial 2 (AIFM2)) as a potent ferroptosis-resistance factor. Our data indicate that myristoylation recruits FSP1 to the plasma membrane where it functions as an oxidoreductase that reduces coenzyme Q10 (CoQ) (also known as ubiquinone-10), which acts as a lipophilic radical-trapping antioxidant that halts the propagation of lipid peroxides. We further find that FSP1 expression positively correlates with ferroptosis resistance across hundreds of cancer cell lines, and that FSP1 mediates resistance to ferroptosis in lung cancer cells in culture and in mouse tumour xenografts. Thus, our data identify FSP1 as a key component of a non-mitochondrial CoQ antioxidant system that acts in parallel to the canonical glutathione-based GPX4 pathway. These findings define a ferroptosis suppression pathway and indicate that pharmacological inhibition of FSP1 may provide an effective strategy to sensitize cancer cells to ferroptosis-inducing chemotherapeutic agents. A synthetic lethal CRISPR–Cas9 screen identifies ferroptosis suppressor protein 1 as a key ferroptosis-resistance factor, the expression of which correlates with ferroptosis resistance in hundreds of cancer cell lines.
- Published
- 2019
28. Manumycin Polyketides Act as Molecular Glues Between UBR7 and P53 to Impair Breast Cancer Pathogenicity
- Author
-
Daniel K. Nomura, John A. Tallarico, Lynn M. McGregor, Ross White, Markus Schirle, Mikiko Okumura, Jeffrey Mckenna, Yosuke Isobe, and Thomas J. Maimone
- Subjects
Natural product ,Mechanism (biology) ,Drug discovery ,Computational biology ,Biology ,Pathogenicity ,medicine.disease ,Ubiquitin ligase ,Protein profiling ,chemistry.chemical_compound ,Breast cancer ,chemistry ,biology.protein ,medicine ,Chemoproteomics - Abstract
Molecular glues are an intriguing therapeutic modality that harness small-molecules to induce interactions between proteins that typically do not interact, thus enabling the creation of novel protein functions not naturally encoded in biology. While molecular glues such as thalidomide and rapamycin have catalyzed drug discovery efforts, such molecules are rare and have often been discovered fortuitously, thus limiting their potential as a general strategy for therapeutic intervention of disease. Historically, natural products have proven to be important sources of molecular glues and we postulated that natural products bearing multiple electrophilic sites may be an unexplored source of such molecules, potentially through multi-covalent attachment. Using activity-based protein profiling (ABPP)-based chemoproteomic platforms, we show that members of the manumycin family of polyketides, which bear multiple potentially reactive sites, target C374 of the putative E3 ligase UBR7 in breast cancer cells to impair breast cancer pathogenicity through engaging in molecular glue interactions with the neo-substrate tumor-suppressor TP53, leading to the activation of p53 transcriptional activity and cell death. Our results reveal a previously undiscovered anti-cancer mechanism of this natural product family and highlight the potential for combining chemoproteomics and multi-covalent natural products for the discovery and characterization of new molecular glues.
- Published
- 2019
- Full Text
- View/download PDF
29. Parthenolide Covalently Targets and Inhibits Focal Adhesion Kinase in Breast Cancer Cells
- Author
-
Milton To, James A. Olzmann, Xirui Hu, Haley S. Lehtola, Thomas J. Maimone, Tucker R. Huffman, Yana Petri, Daniel K. Nomura, Chad R. Altobelli, Sasha G. Demeulenaere, Charles A. Berdan, and Raymond Ho
- Subjects
natural products ,Clinical Biochemistry ,Tanacetum parthenium ,01 natural sciences ,Biochemistry ,chemistry.chemical_compound ,Lactones ,0302 clinical medicine ,Cell Movement ,Drug Discovery ,2.1 Biological and endogenous factors ,Aetiology ,Cancer ,0303 health sciences ,Tumor ,PTK2 ,Activity-based proteomics ,3. Good health ,5.1 Pharmaceuticals ,030220 oncology & carcinogenesis ,Molecular Medicine ,FAK1 ,Female ,medicine.symptom ,Signal transduction ,Development of treatments and therapeutic interventions ,Sesquiterpenes ,Signal Transduction ,Cell Survival ,parthenolide ,Motility ,Breast Neoplasms ,covalent ligands ,Biology ,Article ,Cell Line ,Focal adhesion ,03 medical and health sciences ,Cell Line, Tumor ,Breast Cancer ,medicine ,Humans ,Parthenolide ,ABPP ,Molecular Biology ,030304 developmental biology ,Cell Proliferation ,activity-based protein profiling ,Pharmacology ,Biological Products ,Natural product ,010405 organic chemistry ,chemoproteomics ,0104 chemical sciences ,chemistry ,Mechanism of action ,Focal Adhesion Protein-Tyrosine Kinases ,Focal Adhesion Kinase 1 ,Cancer cell ,Cancer research ,Cysteine - Abstract
Parthenolide, a natural product from the feverfew plant and member of the large family of sesquiterpene lactones, exerts multiple biological and therapeutic activities including anti-inflammatory and anti-cancer effects. Herein, we further study parthenolide mechanism of action using activity-based protein profiling (ABPP)-based chemoproteomic platforms to map additional covalent targets engaged by parthenolide in human breast cancer cells. We find that parthenolide, as well as other related exocyclic methylene lactone-containing sesquiterpenes, covalently modify cysteine 427 (C427) of focal adhesion kinase 1 (FAK1) leading to impairment of FAK1-dependent signaling pathways and breast cancer cell proliferation, survival, and motility. These studies reveal a novel functional target exploited by members of a large family of anticancer natural products.
- Published
- 2019
30. Covalent Ligand Screening Uncovers a RNF4 E3 Ligase Recruiter for Targeted Protein Degradation Applications
- Author
-
Jason R. Thomas, Patrick Lee, Markus Schirle, Carl C. Ward, Scott M. Brittain, Jordan I. Kleinman, Daniel K. Nomura, Clive Yik-Sham Chung, Jeffrey Mckenna, Yana Petri, Kenneth Kim, and John A. Tallarico
- Subjects
0301 basic medicine ,Proteasome Endopeptidase Complex ,Ubiquitin-Protein Ligases ,Protein domain ,Cell Cycle Proteins ,Computational biology ,Protein degradation ,Ligands ,01 natural sciences ,Biochemistry ,Article ,Small Molecule Libraries ,03 medical and health sciences ,Structure-Activity Relationship ,Ubiquitin ,Protein Domains ,Coordination Complexes ,Humans ,Cysteine ,biology ,010405 organic chemistry ,RNF4 ,Drug discovery ,Chemistry ,Ubiquitination ,Nuclear Proteins ,General Medicine ,0104 chemical sciences ,Ubiquitin ligase ,Bromodomain ,Molecular Docking Simulation ,Zinc ,030104 developmental biology ,Proteasome ,Proteolysis ,biology.protein ,Molecular Medicine ,Protein Binding ,Transcription Factors - Abstract
Targeted protein degradation has arisen as a powerful strategy for drug discovery allowing the targeting of undruggable proteins for proteasomal degradation. This approach most often employs heterobifunctional degraders consisting of a protein-targeting ligand linked to an E3 ligase recruiter to ubiquitinate and mark proteins of interest for proteasomal degradation. One challenge with this approach, however, is that only a few E3 ligase recruiters currently exist for targeted protein degradation applications, despite the hundreds of known E3 ligases in the human genome. Here, we utilized activity-based protein profiling (ABPP)-based covalent ligand screening approaches to identify cysteine-reactive small-molecules that react with the E3 ubiquitin ligase RNF4 and provide chemical starting points for the design of RNF4-based degraders. The hit covalent ligand from this screen reacted with either of two zinc-coordinating cysteines in the RING domain, C132 and C135, with no effect on RNF4 activity. We further optimized the potency of this hit and incorporated this potential RNF4 recruiter into a bifunctional degrader linked to JQ1, an inhibitor of the BET family of bromodomain proteins. We demonstrate that the resulting compound CCW 28-3 is capable of degrading BRD4 in a proteasome- and RNF4-dependent manner. In this study, we have shown the feasibility of using chemoproteomics-enabled covalent ligand screening platforms to expand the scope of E3 ligase recruiters that can be exploited for targeted protein degradation applications.
- Published
- 2019
31. Chemoproteomics-enabled discovery of covalent RNF114-based degraders that mimic natural product function
- Author
-
John A. Tallarico, Thomas J. Maimone, Markus Schirle, Daniel K. Nomura, Bingqi Tong, Mai Luo, Jeffrey Mckenna, Jessica N. Spradlin, Lydia Boike, and Scott M. Brittain
- Subjects
Proteomics ,Ubiquitin-Protein Ligases ,Clinical Biochemistry ,Chemical biology ,Computational biology ,01 natural sciences ,Biochemistry ,Article ,chemistry.chemical_compound ,Drug Discovery ,Humans ,Chemoproteomics ,Molecular Biology ,Pharmacology ,Biological Products ,Natural product ,Molecular Structure ,biology ,010405 organic chemistry ,Drug discovery ,Ubiquitination ,0104 chemical sciences ,Ubiquitin ligase ,chemistry ,Covalent bond ,biology.protein ,Molecular Medicine ,Identification (biology) ,Function (biology) - Abstract
The translation of functionally active natural products into fully synthetic small molecule mimetics has remained an important process in medicinal chemistry. We recently discovered that the terpene natural product nimbolide can be utilized as a covalent recruiter of the E3 ubiquitin ligase RNF114 for use in targeted protein degradation (TPD) – a powerful therapeutic modality within modern day drug discovery. Using activity-based protein profiling-enabled covalent ligand screening approaches, we herein report the discovery of fully synthetic RNF114-based recruiter molecules that can also be exploited for PROTAC applications, and demonstrate their utility in degrading therapeutically relevant targets such as BRD4 and BCR-ABL in cells. The identification of simple and easily manipulated drug-like scaffolds that can mimic the function of a complex natural product is beneficial in further expanding the toolbox of E3 ligase recruiters, an area of great importance in drug discovery and chemical biology.
- Published
- 2021
- Full Text
- View/download PDF
32. Discovery of a Functional Covalent Ligand Targeting an Intrinsically Disordered Cysteine within MYC
- Author
-
Nathaniel J. Henning, Alexander G. Cioffi, Markus Schirle, John A. Tallarico, Lydia Boike, Jeffrey Mckenna, Jennifer Co, Daniel K. Nomura, Michael D. Jones, Felix C. Majewski, and Gang Liu
- Subjects
Clinical Biochemistry ,Biology ,Ligands ,medicine.disease_cause ,01 natural sciences ,Biochemistry ,Proto-Oncogene Proteins c-myc ,chemistry.chemical_compound ,Drug Discovery ,Consensus sequence ,medicine ,Humans ,Chemoproteomics ,Cysteine ,Molecular Biology ,Transcription factor ,Cells, Cultured ,Pharmacology ,Dose-Response Relationship, Drug ,Molecular Structure ,010405 organic chemistry ,Activity-based proteomics ,Ligand (biochemistry) ,0104 chemical sciences ,Cell biology ,chemistry ,Molecular Medicine ,Carcinogenesis ,DNA - Abstract
Summary MYC is a major oncogenic transcriptional driver of most human cancers that has remained intractable to direct targeting because much of MYC is intrinsically disordered. Here, we have performed a cysteine-reactive covalent ligand screen to identify compounds that could disrupt the binding of MYC to its DNA consensus sequence in vitro and also impair MYC transcriptional activity in situ in cells. We have identified a covalent ligand, EN4, that targets cysteine 171 of MYC within a predicted intrinsically disordered region of the protein. We show that EN4 directly targets MYC in cells, reduces MYC and MAX thermal stability, inhibits MYC transcriptional activity, downregulates multiple MYC transcriptional targets, and impairs tumorigenesis. We also show initial structure-activity relationships of EN4 and identify compounds that show improved potency. Overall, we identify a unique ligandable site within an intrinsically disordered region of MYC that leads to inhibition of MYC transcriptional activity.
- Published
- 2021
- Full Text
- View/download PDF
33. 4β-Hydroxycholesterol is a prolipogenic factor that promotes SREBP1c expression and activity through the liver X receptor
- Author
-
Douglas F. Covey, Daniel K. Nomura, Ofer Moldavski, Xuntian Jiang, Mingxing Qian, Ethan J. Weiss, Robert J. van Eijkeren, Andreas Stahl, Peter-James H. Zushin, Charles A. Berdan, Daniel S. Ory, and Roberto Zoncu
- Subjects
0301 basic medicine ,insulin ,Biochemistry & Molecular Biology ,4β-HC, 4β-hydroxycholesterol ,LD, lipid droplet ,mTOR, mechanistic Target of Rapamycin ,Oxysterol ,lipid droplets ,LDS, lipid-depleted serum ,QD415-436 ,DNL, de novo lipogenesis ,Medical Biochemistry and Metabolomics ,030204 cardiovascular system & hematology ,HC, hydroxycholesterol ,Biochemistry ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,EtOAc, ethyl acetate ,ent-4HC, enantiomer of 4β-HC ,Lipid droplet ,Lipid biosynthesis ,Genetics ,polycyclic compounds ,Transcriptional regulation ,Liver X receptor ,Transcription factor ,Chemistry ,Liver Disease ,de-novo-lipogenesis ,liver-X-Receptor ,PI3K, phosphatidylinositol 3-kinase ,Lipid metabolism ,DPBS, dulbecco’s phosphate buffered saline ,Cell Biology ,SREBP1c ,Cell biology ,030104 developmental biology ,Lipogenesis ,lipids (amino acids, peptides, and proteins) ,NAFLD, nonalcoholic fatty liver disease ,Biochemistry and Cell Biology ,Digestive Diseases ,Sterol Regulatory Element Binding Protein 1 ,oxysterol ,THF, tetrahydrofuran ,Research Article - Abstract
Oxysterols are oxidized derivatives of cholesterol that play regulatory roles in lipid biosynthesis and homeostasis. How oxysterol signaling coordinates different lipid classes such as sterols and triglycerides remains incompletely understood. Here, we show that 4β-hydroxycholesterol (HC) (4β-HC), a liver and serum abundant oxysterol of poorly defined functions, is a potent and selective inducer of the master lipogenic transcription factor, SREBP1c, but not the related steroidogenic transcription factor SREBP2. By correlating tracing of lipid synthesis with lipogenic gene expression profiling, we found that 4β-HC acts as a putative agonist for the liver X receptor (LXR), a sterol sensor and transcriptional regulator previously linked to SREBP1c activation. Unique among the oxysterol agonists of the LXR, 4β-HC induced expression of the lipogenic program downstream of SREBP1c and triggered de novo lipogenesis both in primary hepatocytes and in the mouse liver. In addition, 4β-HC acted in parallel to insulin-PI3K–dependent signaling to stimulate triglyceride synthesis and lipid-droplet accumulation. Thus, 4β-HC is an endogenous regulator of de novo lipogenesis through the LXR-SREBP1c axis.
- Published
- 2021
- Full Text
- View/download PDF
34. The Secreted Enzyme PM20D1 Regulates Lipidated Amino Acid Uncouplers of Mitochondria
- Author
-
Daniel K. Nomura, Theodore M. Kamenecka, Patrick R. Griffin, Leslie A. Bateman, Bruce M. Spiegelman, Hua Lin, Katrin J. Svensson, Jonathan Z. Long, Mi Ra Chang, Rajesh R. Rao, Joao A. Paulo, Steven P. Gygi, Isha A. Lokurkar, Mark P. Jedrychowski, and Jesse Lou
- Subjects
Male ,0301 basic medicine ,Cellular respiration ,1.1 Normal biological development and functioning ,Cell Respiration ,Mitochondrion ,Carbohydrate metabolism ,Biology ,Inbred C57BL ,Skin Diseases ,Medical and Health Sciences ,Article ,General Biochemistry, Genetics and Molecular Biology ,Amidohydrolases ,Mitochondrial Proteins ,Mice ,03 medical and health sciences ,Underpinning research ,Adipocytes ,Humans ,Animals ,Homeostasis ,Glucose homeostasis ,Obesity ,Amino Acids ,Nutrition ,chemistry.chemical_classification ,Fatty Acids ,Diabetes ,Thermogenesis ,Biological Sciences ,Thermogenin ,Mitochondria ,Amino acid ,Mice, Inbred C57BL ,Glucose ,030104 developmental biology ,Enzyme ,chemistry ,Biochemistry ,Burns ,Energy Metabolism ,Metabolic Networks and Pathways ,Biotechnology ,Developmental Biology - Abstract
Summary Brown and beige adipocytes are specialized cells that express uncoupling protein 1 (UCP1) and dissipate chemical energy as heat. These cells likely possess alternative UCP1-independent thermogenic mechanisms. Here, we identify a secreted enzyme, peptidase M20 domain containing 1 (PM20D1), that is enriched in UCP1 + versus UCP1 − adipocytes. We demonstrate that PM20D1 is a bidirectional enzyme in vitro, catalyzing both the condensation of fatty acids and amino acids to generate N -acyl amino acids and also the reverse hydrolytic reaction. N -acyl amino acids directly bind mitochondria and function as endogenous uncouplers of UCP1-independent respiration. Mice with increased circulating PM20D1 have augmented respiration and increased N- acyl amino acids in blood. Lastly, administration of N- acyl amino acids to mice improves glucose homeostasis and increases energy expenditure. These data identify an enzymatic node and a family of metabolites that regulate energy homeostasis. This pathway might be useful for treating obesity and associated disorders.
- Published
- 2016
- Full Text
- View/download PDF
35. Properties of fine Sr2+-substituted hydroxyapatite synthesized using ultrasonication
- Author
-
K Nomura, Ayumu Onda, Yoshiyuki Kojima, Hirogo Minamisawa, and Tetsuo Umegaki
- Subjects
Strontium ,Materials science ,chemistry.chemical_element ,Apatite ,chemistry.chemical_compound ,chemistry ,Strontium hydroxide ,Specific surface area ,visual_art ,visual_art.visual_art_medium ,Atomic ratio ,Spectroscopy ,Phosphoric acid ,Nuclear chemistry ,Solid solution - Abstract
This study investigated the properties of fine Sr2+-substituted hydroxyapatite (SrHAp) synthesized using ultrasonication. SrHAp was synthesized by the addition of phosphoric acid solution to an ultrasonicated calcium hydroxide suspension containing strontium hydroxide. The X-ray diffraction peaks for SrHAp were shifted to low angle with increasing Sr/(Ca + Sr) atomic ratio. X-ray diffraction, energy dispersive X-ray spectroscopy and inductively coupled plasma spectroscopy measurements confirmed that HAp and strontium apatite (Sr10(PO4)6(OH)2;SrAp) formed a complete solid solution. When the Sr/(Ca + Sr) atomic ratio was 0.3 or less, the (Ca + Sr)/P atomic ratio was 1.67 or less, and the specific surface area was also large. The bending strength of a SrHAp sintered body was increased and the bending strength of the SrAp sintered body became 40 MPa when the Sr/(Ca + Sr) atomic ratio was above 0.3. The ethanol conversion rate using SrAp as catalyst was 7% for 1 hour and was 40% of HAp reagent. However, selectivity of SrAp from ethanol to butanol was 70%.
- Published
- 2020
- Full Text
- View/download PDF
36. Acyl-CoA synthetase 6 enriches the neuroprotective omega-3 fatty acid DHA in the brain
- Author
-
Jason R. Cannon, Regina F. Fernandez, Sora Q. Kim, Rachel M. Foguth, Jessica L. Counihan, Daniel K. Nomura, Yingwei Zhao, Marcus M. Weera, Julia A. Chester, and Jessica M. Ellis
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Multidisciplinary ,Lipopolysaccharide ,Fatty acid metabolism ,Microglia ,Fatty acid ,Brain ,Biological Sciences ,medicine.disease ,Neuroprotection ,Astrogliosis ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,chemistry ,Biochemistry ,Docosahexaenoic acid ,Fatty Acids, Omega-3 ,medicine ,lipids (amino acids, peptides, and proteins) ,Omega 3 fatty acid ,030217 neurology & neurosurgery - Abstract
Docosahexaenoic acid (DHA) is an omega-3 fatty acid that is highly abundant in the brain and confers protection against numerous neurological diseases, yet the fundamental mechanisms regulating the enrichment of DHA in the brain remain unknown. Here, we have discovered that a member of the long-chain acyl-CoA synthetase family, Acsl6, is required for the enrichment of DHA in the brain by generating an Acsl6-deficient mouse (Acsl6(−/−)). Acsl6 is highly enriched in the brain and lipid profiling of Acsl6(−/−) tissues reveals consistent reductions in DHA-containing lipids in tissues highly abundant with Acsl6. Acsl6(−/−) mice demonstrate motor impairments, altered glutamate metabolism, and increased astrogliosis and microglia activation. In response to a neuroinflammatory lipopolysaccharide injection, Acsl6(−/−) brains show similar increases in molecular and pathological indices of astrogliosis compared with controls. These data demonstrate that Acsl6 is a key mediator of neuroprotective DHA enrichment in the brain.
- Published
- 2018
37. Enzyme promiscuity drives branched-chain fatty acid synthesis in adipose tissues
- Author
-
Joan Sanchez-Gurmaches, Theodore P. Ciaraldi, Courtney R. Green, Jivani M. Gengatharan, Christian M. Metallo, Lindsay S. Roberts, Pedro Cabrales, Yujung Michelle Lee, Janelle S. Ayres, Justin L. McCarville, Rohit Loomba, Martina Wallace, Daniel K. Nomura, David A. Guertin, Justin D Hover, Susan A. Phillips, and Noah Meurs
- Subjects
0301 basic medicine ,brown adipocytes ,obesity ,Adipose tissue ,Mice, Obese ,odd-chain fatty acids ,Inbred C57BL ,Obese ,Mice ,Cytosol ,metabolic flux analysis ,Adipocytes ,RNA, Small Interfering ,Amino Acids ,Hypoxia ,chemistry.chemical_classification ,Mice, Knockout ,fatty acid synthase ,biology ,Chemistry ,Fatty Acids ,carnitine acetyl transferase ,3T3 Cells ,Lipidome ,Branched-chain amino acids ,Amino acid ,adipose tissue ,Fatty acid synthase ,Biochemistry ,Adipose Tissue ,Lipogenesis ,branched-chain fatty acids ,Female ,Biochemistry & Molecular Biology ,white adipocytes ,Knockout ,short-chain fatty acids ,liver ,Small Interfering ,Article ,03 medical and health sciences ,Medicinal and Biomolecular Chemistry ,Animals ,Obesity ,Molecular Biology ,Metabolic and endocrine ,Nutrition ,Carnitine O-Acetyltransferase ,Catabolism ,Lentivirus ,Cell Biology ,Metabolism ,Branched-Chain ,Mice, Inbred C57BL ,030104 developmental biology ,de novo lipogenesis ,biology.protein ,RNA ,Biochemistry and Cell Biology ,Fatty Acid Synthases ,CRISPR-Cas Systems ,Digestive Diseases ,Amino Acids, Branched-Chain - Abstract
Summary Fatty acid synthase (FASN) predominantly generates straight-chain fatty acids using acetyl-CoA as the initiating substrate. However, monomethyl branched-chain fatty acids (mmBCFAs) are also present in mammals but thought to be primarily diet-derived. Here we demonstrate that mmBCFAs are de novo synthesized via mitochondrial BCAA catabolism, exported to the cytosol by adipose-specific expression of carnitine acetyltransferase (CrAT), and elongated by FASN. Brown fat exhibits the highest BCAA catabolic and mmBCFA synthesis fluxes, whereas these lipids are largely absent from liver and brain. mmBCFA synthesis is also sustained in the absence of microbiota. We identify hypoxia as a potent suppressor of BCAA catabolism that decreases mmBCFA synthesis in obese adipose tissue, such that mmBCFAs are significantly decreased in obese animals. These results identify adipose tissue mmBCFA synthesis as a novel link between BCAA metabolism and lipogenesis, highlighting roles for CrAT and FASN promiscuity that influence acyl-chain diversity in the lipidome.
- Published
- 2018
- Full Text
- View/download PDF
38. Metabolomic Markers of Phthalate Exposure in Plasma and Urine of Pregnant Women
- Author
-
Vy Tran, Asa Bradman, Karen Huen, Brenda Eskenazi, Breanna Ford, Robert B. Gunier, Michael Zhou, Nina Holland, Gwen Tindula, Douglas P. Lee, and Daniel K. Nomura
- Subjects
0301 basic medicine ,Metabolite ,Physiology ,Context (language use) ,Urine ,010501 environmental sciences ,01 natural sciences ,03 medical and health sciences ,chemistry.chemical_compound ,Metabolomics ,Clinical Research ,Biomonitoring ,targeted metabolomics ,Metabolome ,2.1 Biological and endogenous factors ,Medicine ,Aetiology ,Original Research ,0105 earth and related environmental sciences ,phthalates ,screening and diagnosis ,business.industry ,lcsh:Public aspects of medicine ,Prevention ,Public Health, Environmental and Occupational Health ,Phthalate ,lcsh:RA1-1270 ,Metabolism ,Estrogen ,in utero exposure ,4.1 Discovery and preclinical testing of markers and technologies ,3. Good health ,Detection ,endocrine disruptors ,030104 developmental biology ,chemistry ,inflammation ,Public Health and Health Services ,Public Health ,pregnancy ,business - Abstract
Phthalates are known endocrine disruptors and found in almost all people with several associated adverse health outcomes reported in humans and animal models. Limited data are available on the relationship between exposure to endocrine disrupting chemicals and the human metabolome. We examined the relationship of metabolomic profiles in plasma and urine of 115 pregnant women with eleven urine phthalate metabolites measured at 26 weeks of gestation to identify potential biomarkers and relevant pathways. Targeted metabolomics was performed by selected reaction monitoring liquid chromatography and triple quadrupole mass spectrometry to measure 415 metabolites in plasma and 151 metabolites in urine samples. We have chosen metabolites with the best defined peaks for more detailed analysis (138 in plasma and 40 in urine). Relationship between urine phthalate metabolites and concurrent metabolomic markers in plasma and urine suggested potential involvement of diverse pathways including lipid, steroid, and nucleic acid metabolism and enhanced inflammatory response. Most of the correlations were positive for both urine and plasma, and further confirmed by regression and PCA analysis. However, after the FDR adjustment for multiple comparisons, only 9 urine associations remained statistically significant (q-values 0.0001-0.0451), including Nicotinamide mononucleotide, Cysteine T2, Cystine, and L-Aspartic acid. Additionally, we found negative associations of maternal pre-pregnancy body mass index (BMI) with more than 20 metabolomic markers related to lipid and amino-acid metabolism and inflammation pathways in plasma (p = 0.01-0.0004), while Mevalonic acid was positively associated (p = 0.009). Nicotinic acid, the only significant metabolite in urine, had a positive association with maternal BMI (p = 0.002). In summary, when evaluated in the context of metabolic pathways, the findings suggest enhanced lipid biogenesis, inflammation and altered nucleic acid metabolism in association with higher phthalate levels. These results provide new insights into the relationship between phthalates, common in most human populations, and metabolomics, a novel approach to exposure and health biomonitoring.
- Published
- 2018
- Full Text
- View/download PDF
39. Covalent Ligand Screening Uncovers a RNF4 E3 Ligase Recruiter for Targeted Protein Degradation Applications
- Author
-
Clive Yik-Sham Chung, Jason R. Thomas, Daniel K. Nomura, Jeffrey Mckenna, Yana Petri, Jordan I. Kleinman, John A. Tallarico, Patrick Lee, Markus Schirle, Carl C. Ward, and Kenneth Kim
- Subjects
0303 health sciences ,BRD4 ,biology ,010405 organic chemistry ,Chemistry ,RNF4 ,Drug discovery ,Computational biology ,Protein degradation ,Ligand (biochemistry) ,01 natural sciences ,0104 chemical sciences ,Bromodomain ,Ubiquitin ligase ,03 medical and health sciences ,Proteasome ,biology.protein ,030304 developmental biology - Abstract
Targeted protein degradation has arisen as a powerful strategy for drug discovery allowing the targeting of undruggable proteins for proteasomal degradation. This approach most often employs heterobifunctional degraders consisting of a protein-targeting ligand linked to an E3 ligase recruiter to ubiquitinate and mark proteins of interest for proteasomal degradation. One challenge with this approach, however, is that only few E3 ligase recruiters currently exist for targeted protein degradation applications, despite the hundreds of known E3 ligases in the human genome. Here, we utilized activity-based protein profiling (ABPP)-based covalent ligand screening approaches to identify cysteine-reactive small-molecules that react with the E3 ubiquitin ligase RNF4 and provide chemical starting points for the design of RNF4-based degraders. The hit covalent ligand from this screen reacted with either of two zinc-coordinating cysteines in the RING domain, C132 and C135, with no effect on RNF4 activity. We further optimized the potency of this hit and incorporated this potential RNF4 recruiter into a bifunctional degrader linked to JQ1, an inhibitor of the BET family of bromodomain proteins. We demonstrate that the resulting compound CCW 28-3 is capable of degrading BRD4 in a proteasome- and RNF4-dependent manner. In this study, we have shown the feasibility of using chemoproteomics-enabled covalent ligand screening platforms to expand the scope of E3 ligase recruiters that can be exploited for targeted protein degradation applications.
- Published
- 2018
- Full Text
- View/download PDF
40. Harnessing the Anti-Cancer Natural Product Nimbolide for Targeted Protein Degradation
- Author
-
Jessica N. Spradlin, Carl C. Ward, Daniel K. Nomura, Dirksen E. Bussiere, Lisha Ou, Thomas J. Maimone, Andrew Proudfoot, James A. Olzmann, Jason R. Thomas, Mikias Woldegiorgis, Markus Schirle, Xirui Hu, John A. Tallarico, Jeffrey Mckenna, Michael D. Jones, Elizabeth Ornelas, Scott M. Brittain, and Milton To
- Subjects
Druggability ,Drug Screening Assays ,01 natural sciences ,law.invention ,chemistry.chemical_compound ,Ubiquitin ,law ,Phytogenic ,ZNF313 ,Cancer ,0303 health sciences ,RNF114 ,biology ,030302 biochemistry & molecular biology ,Cell biology ,Ubiquitin ligase ,5.1 Pharmaceuticals ,Female ,Development of treatments and therapeutic interventions ,Limonins ,Biochemistry & Molecular Biology ,Ubiquitin-Protein Ligases ,Cancer therapy ,Antineoplastic Agents ,Breast Neoplasms ,Protein degradation ,Article ,Medicinal and Biomolecular Chemistry ,03 medical and health sciences ,Breast Cancer ,medicine ,Humans ,ABPP ,Molecular Biology ,activity-based protein profiling ,Cell Proliferation ,030304 developmental biology ,Biological Products ,Natural product ,010405 organic chemistry ,Antitumor ,Cell Biology ,medicine.disease ,chemoproteomics ,Antineoplastic Agents, Phytogenic ,0104 chemical sciences ,Protein profiling ,nimbolide ,chemistry ,Proteolysis ,biology.protein ,Suppressor ,Biochemistry and Cell Biology ,degraders ,Drug Screening Assays, Antitumor ,Carrier Proteins ,targeted protein degradation - Abstract
Nimbolide, a terpenoid natural product derived from the Neem tree, impairs cancer pathogenicity across many types of human cancers; however, the direct targets and mechanisms by which nimbolide exerts its effects are poorly understood. Here, we used activity-based protein profiling (ABPP) chemoproteomic platforms to discover that nimbolide reacts with a novel functional cysteine crucial for substrate recognition in the E3 ubiquitin ligase RNF114. Nimbolide impairs breast cancer cell proliferation in-part by disrupting RNF114 substrate recognition, leading to inhibition of ubiquitination and degradation of the tumor-suppressors such as p21, resulting in their rapid stabilization. We further demonstrate that nimbolide can be harnessed to recruit RNF114 as an E3 ligase in targeted protein degradation applications and show that synthetically simpler scaffolds are also capable of accessing this unique reactive site. Our study highlights the utility of ABPP platforms in uncovering unique druggable modalities accessed by natural products for cancer therapy and targeted protein degradation applications.
- Published
- 2018
- Full Text
- View/download PDF
41. Development of alkyl glycerone phosphate synthase inhibitors: Structure-activity relationship and effects on ether lipids and epithelial-mesenchymal transition in cancer cells
- Author
-
Giulia Stazi, Sharon M. Louie, Sara Marchese, Giancarlo Fabrizi, Andrea Mattevi, Roberto Cirilli, Valentina Piano, Monica Viviano, Rino Ragno, Alexandros Patsilinakos, Cecilia Battistelli, Alessia Ciogli, Raffaele Strippoli, Antonello Mai, Lorenzo Antonini, Marco Tripodi, Alessandra Marchetti, Roberta Mazzone, Daniel K. Nomura, Biagina Marrocco, Gianluca Sbardella, Clemens Zwergel, and Sergio Valente
- Subjects
Epithelial-Mesenchymal Transition ,Motility ,01 natural sciences ,Cell Line ,03 medical and health sciences ,Structure-Activity Relationship ,Cell Movement ,Cell Line, Tumor ,Neoplasms ,Drug Discovery ,medicine ,AGPS inhibitors ,Cancer ,E-cadherin ,Ether lipids ,Snail ,Humans ,Epithelial–mesenchymal transition ,030304 developmental biology ,Cell Proliferation ,Pharmacology ,0303 health sciences ,Alkyl and Aryl Transferases ,010405 organic chemistry ,Cell growth ,Chemistry ,Organic Chemistry ,Lipid metabolism ,Cell migration ,General Medicine ,medicine.disease ,Cadherins ,Lipid Metabolism ,0104 chemical sciences ,Cell biology ,Cell culture ,Cancer cell ,Matrix Metalloproteinase 2 ,Snail Family Transcription Factors - Abstract
In aggressive tumors, alkylglyceronephosphate synthase (AGPS) controls cellular ether phospholipid utilization and metabolism to promote cancer cell proliferation and motility. SAR studies on the first-in-class AGPS inhibitor 1, discovered by our group, led to the 2,6-difluoro analog 2i which showed higher binding affinity than 1 in vitro. In 231MFP cancer cells, 2i reduced ether lipids levels and cell migration rate. When tested in PC-3 and MDA-MB-231 cancer cells, 2i specifically impaired epithelial to mesenchymal transition (EMT) by modulating E-cadherin, Snail and MMP2 expression levels. Moreover, the combination of siRNAs against AGPS and 2i provided no additive effect, confirming that the modulation of 2i on EMT specifically relies on AGPS inhibition. Finally, this compound also affected cancer cell proliferation especially in MDA-MB-231 cells expressing higher AGPS level, whereas it provided negligible effects on MeT5A, a non-tumorigenic cell line, thus showing cancer specificity.
- Published
- 2018
42. Chemoproteomics-Enabled Covalent Ligand Screening Reveals ALDH3A1 as a Lung Cancer Therapy Target
- Author
-
Kimberly E. Anderson, Jessica L. Counihan, Daniel K. Nomura, and Amanda L Wiggenhorn
- Subjects
0301 basic medicine ,Proteomics ,Cell Survival ,Cell ,Antineoplastic Agents ,Mice, SCID ,Ligands ,Biochemistry ,Article ,Small Molecule Libraries ,03 medical and health sciences ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Chemoproteomics ,Cysteine ,Enzyme Inhibitors ,Lung cancer ,Cell Proliferation ,Base Sequence ,Ligand ,Chemistry ,HEK 293 cells ,Epithelial Cells ,General Medicine ,Aldehyde Dehydrogenase ,medicine.disease ,Small molecule ,Xenograft Model Antitumor Assays ,030104 developmental biology ,medicine.anatomical_structure ,HEK293 Cells ,Covalent bond ,Cancer research ,Molecular Medicine ,Chemical genetics - Abstract
Chemical genetics is a powerful approach for identifying therapeutically active small-molecules, but identifying the mechanisms of action underlying hit compounds remains challenging. Chemoproteomic platforms have arisen to tackle this challenge and enable rapid mechanistic deconvolution of small-molecule screening hits. Here, we have screened a cysteine-reactive covalent ligand library to identify hit compounds that impair cell survival and proliferation in non-small cell lung carcinoma cells, but not in primary human bronchial epithelial cells. Through this screen, we identified a covalent ligand hit, DKM 3–42 which impaired both in situ and in vivo lung cancer pathogenicity. We used activity-based protein profiling to discover that the primary target of DKM 3–42 was the catalytic cysteine in aldehyde dehydrogenase 3A1 (ALDH3A1). We performed further chemoproteomics-enabled covalent ligand screening directly against ALDH3A1, and identified a more potent and selective lead covalent ligand, EN40, which inhibits ALDH3A1 activity and impairs lung cancer pathogenicity. We show here that ALDH3A1 represents a potentially novel therapeutic target for lung cancers that express ALDH3A1 and put forth two selective ALDH3A1 inhibitors. Overall, we show the utility of combining chemical genetics screening of covalent ligand libraries with chemoproteomic approaches to rapidly identify anti-cancer leads and targets.
- Published
- 2018
43. Ablation of PM20D1 reveals N -acyl amino acid control of metabolism and nociception
- Author
-
Mark P. Jedrychowski, Charles A. Berdan, Zhaoming Deng, Katrin J. Svensson, Amanda J. Roberts, Amir I. Mina, Jonathan Z. Long, John M. Asara, Bruce M. Spiegelman, Sharon M. Louie, Theodore M. Kamenecka, Hua Lin, Daniel K. Nomura, Patrick R. Griffin, Leslie A. Bateman, Florence Y. Dou, Alexander S. Banks, and Alexander M. Roche
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,Multidisciplinary ,Voltage-dependent calcium channel ,ATP synthase ,biology ,Chemistry ,TRPV1 ,Endogeny ,Metabolism ,nervous system diseases ,3. Good health ,Cell biology ,Amino acid ,03 medical and health sciences ,Transient receptor potential channel ,030104 developmental biology ,Enzyme ,nervous system ,mental disorders ,biology.protein ,lipids (amino acids, peptides, and proteins) - Abstract
N-acyl amino acids (NAAs) are a structurally diverse class of bioactive signaling lipids whose endogenous functions have largely remained uncharacterized. To clarify the physiologic roles of NAAs, we generated mice deficient in the circulating enzyme peptidase M20 domain-containing 1 (PM20D1). Global PM20D1-KO mice have dramatically reduced NAA hydrolase/synthase activities in tissues and blood with concomitant bidirectional dysregulation of endogenous NAAs. Compared with control animals, PM20D1-KO mice exhibit a variety of metabolic and pain phenotypes, including insulin resistance, altered body temperature in cold, and antinociceptive behaviors. Guided by these phenotypes, we identify N-oleoyl-glutamine (C18:1-Gln) as a key PM20D1-regulated NAA. In addition to its mitochondrial uncoupling bioactivity, C18:1-Gln also antagonizes certain members of the transient receptor potential (TRP) calcium channels including TRPV1. Direct administration of C18:1-Gln to mice is sufficient to recapitulate a subset of phenotypes observed in PM20D1-KO animals. These data demonstrate that PM20D1 is a dominant enzymatic regulator of NAA levels in vivo and elucidate physiologic functions for NAA signaling in metabolism and nociception.
- Published
- 2018
- Full Text
- View/download PDF
44. Cancer Metabolism: Current Understanding and Therapies
- Author
-
Daniel K. Nomura, Elizabeth A. Grossman, and Jessica L. Counihan
- Subjects
0301 basic medicine ,Abnormal cell ,Bioinformatics ,Malignancy ,Cancer pathogenesis ,Pentose Phosphate Pathway ,03 medical and health sciences ,0302 clinical medicine ,Neoplasms ,medicine ,Humans ,Amino Acids ,End point ,Extramural ,Chemistry ,Tumor Suppressor Proteins ,Fatty Acids ,Cancer ,General Chemistry ,Oncogenes ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Cancer metabolism ,Cancer cell ,Carbohydrate Metabolism - Abstract
Dysregulation of cancer cell metabolism contributes to abnormal cell growth, the biological end point of cancer. We review here numerous affected oncogenes and metabolic pathways common in cancer and how they contribute to cancer pathogenesis and malignancy. This review also discusses various pharmacological manipulations that take advantage of these metabolic abnormalities and the current targeted therapies that have arisen from this research.
- Published
- 2018
45. Suppressing fatty acid uptake has therapeutic effects in preclinical models of prostate cancer
- Author
-
Matthew J. Watt, Magdalene K. Montgomery, Poornima R. Wijayaratne, Gail P. Risbridger, Richard J. Rebello, Vanessa R. Haynes, Mark Frydenberg, Maria Matzaris, Ashlee K. Clark, Cheng Huang, Daniel K. Nomura, Laura H Porter, Luke A. Selth, Melissa Papargiris, Jennifer Chi Yi Lo, Renea A. Taylor, Maria Febbraio, Ralf B. Schittenhelm, Sam Norden, Sarah T. Whitby, Kimberly E. Anderson, Luc Furic, Birunthi Niranjan, and Natalie Lister
- Subjects
CD36 Antigens ,Male ,CD36 ,Metastasis ,Prostate cancer ,Mice ,Prostate ,Cell Line, Tumor ,medicine ,Animals ,Humans ,Neoplasm Invasiveness ,Biomass ,Gene Silencing ,RNA, Small Interfering ,chemistry.chemical_classification ,biology ,Chemistry ,Fatty Acids ,PTEN Phosphohydrolase ,Cancer ,Fatty acid ,Antibodies, Monoclonal ,Prostatic Neoplasms ,Lipid metabolism ,Epithelial Cells ,General Medicine ,medicine.disease ,Lipid Metabolism ,Tumor Burden ,Disease Models, Animal ,medicine.anatomical_structure ,Lipogenesis ,biology.protein ,Cancer research ,Disease Progression ,Gene Deletion - Abstract
Metabolism alterations are hallmarks of cancer, but the involvement of lipid metabolism in disease progression is unclear. We investigated the role of lipid metabolism in prostate cancer using tissue from patients with prostate cancer and patient-derived xenograft mouse models. We showed that fatty acid uptake was increased in human prostate cancer and that these fatty acids were directed toward biomass production. These changes were mediated, at least partly, by the fatty acid transporter CD36, which was associated with aggressive disease. Deleting Cd36 in the prostate of cancer-susceptible Pten-/- mice reduced fatty acid uptake and the abundance of oncogenic signaling lipids and slowed cancer progression. Moreover, CD36 antibody therapy reduced cancer severity in patient-derived xenografts. We further demonstrated cross-talk between fatty acid uptake and de novo lipogenesis and found that dual targeting of these pathways more potently inhibited proliferation of human cancer-derived organoids compared to the single treatments. These findings identify a critical role for CD36-mediated fatty acid uptake in prostate cancer and suggest that targeting fatty acid uptake might be an effective strategy for treating prostate cancer.
- Published
- 2018
46. Exogenous Monounsaturated Fatty Acids Promote a Ferroptosis-Resistant Cell State
- Author
-
Leslie Magtanong, Gary J. Patti, Daniel K. Nomura, Kevin Cho, James A. Olzmann, Milton To, Amy Tarangelo, Pin-Joe Ko, Jennifer Yinuo Cao, Scott J. Dixon, Carl C. Ward, and Giovanni C. Forcina
- Subjects
Membrane lipids ,1.1 Normal biological development and functioning ,Clinical Biochemistry ,lipid droplet ,Oxidative phosphorylation ,Biology ,GPX4 ,01 natural sciences ,Biochemistry ,Cell Line ,Fatty Acids, Monounsaturated ,oleate ,Mice ,iron ,Underpinning research ,Lipid droplet ,MUFAs ,Drug Discovery ,Coenzyme A Ligases ,Animals ,Ferroptosis ,Molecular Biology ,Pharmacology ,chemistry.chemical_classification ,Reactive oxygen species ,Arachidonic Acid ,010405 organic chemistry ,Fatty Acids ,Cell Membrane ,food and beverages ,Lipid Droplets ,lipotoxicity ,Phospholipid Hydroperoxide Glutathione Peroxidase ,Lipids ,ferroptosis ,0104 chemical sciences ,Monounsaturated ,cell death ,chemistry ,Lipotoxicity ,Apoptosis ,Molecular Medicine ,lipids (amino acids, peptides, and proteins) ,Reactive Oxygen Species ,Oxidation-Reduction ,lipid ROS ,Polyunsaturated fatty acid - Abstract
Summary The initiation and execution of cell death can be regulated by various lipids. How the levels of environmental (exogenous) lipids impact cell death sensitivity is not well understood. We find that exogenous monounsaturated fatty acids (MUFAs) potently inhibit the non-apoptotic, iron-dependent, oxidative cell death process of ferroptosis. This protective effect is associated with the suppression of lipid reactive oxygen species (ROS) accumulation at the plasma membrane and decreased levels of phospholipids containing oxidizable polyunsaturated fatty acids. Treatment with exogenous MUFAs reduces the sensitivity of plasma membrane lipids to oxidation over several hours. This effect requires MUFA activation by acyl-coenzyme A synthetase long-chain family member 3 (ACSL3) and is independent of lipid droplet formation. Exogenous MUFAs also protect cells from apoptotic lipotoxicity caused by the accumulation of saturated fatty acids, but in an ACSL3-independent manner. Our work demonstrates that ACSL3-dependent MUFA activation promotes a ferroptosis-resistant cell state.
- Published
- 2018
47. Long‐Chain Acyl‐CoA synthetase 6 deficiency reduces the omega‐3 fatty acid DHA in the brain and disrupts motor control
- Author
-
Yingwei Zhao, Jessica M. Ellis, Jessica L. Counihan, Daniel K. Nomura, Julia A. Chester, and Regina F. Fernandez
- Subjects
Biochemistry ,Chemistry ,Genetics ,Motor control ,Omega 3 fatty acid ,Molecular Biology ,LONG CHAIN ACYL-CoA SYNTHETASE ,Biotechnology - Published
- 2018
- Full Text
- View/download PDF
48. Discovery of hydrolysis-resistant isoindoline N-acyl amino acid analogs that stimulate mitochondrial respiration
- Author
-
Hua Lin, Alexander M. Roche, Mi Ra Chang, Katrin J. Svensson, Charles A. Berdan, Claudia Ruiz, Michael D. Cameron, Sharon M. Louie, Daniel K. Nomura, Florence Y. Dou, Patrick R. Griffin, Bruce M. Spiegelman, Jonathan Z. Long, Timothy S. Strutzenberg, Theodore M. Kamenecka, and Scott J. Novick
- Subjects
0301 basic medicine ,Indoles ,Mitochondrion ,Carbohydrate metabolism ,Article ,Amidohydrolases ,Cell Line ,03 medical and health sciences ,Hydrolysis ,chemistry.chemical_compound ,Mice ,Structure-Activity Relationship ,0302 clinical medicine ,Oxygen Consumption ,Drug Discovery ,Glucose homeostasis ,Structure–activity relationship ,Animals ,Homeostasis ,Amino Acids ,Unsaturated fatty acid ,chemistry.chemical_classification ,Chemistry ,Isoindoline ,Stimulation, Chemical ,Amino acid ,Mitochondria ,030104 developmental biology ,Glucose ,Biochemistry ,Fatty Acids, Unsaturated ,Molecular Medicine ,Energy Metabolism ,030217 neurology & neurosurgery - Abstract
N-Acyl amino acids directly bind mitochondria and function as endogenous uncouplers of UCP1-independent respiration. We found that administration of N-acyl amino acids to mice improves glucose homeostasis and increases energy expenditure, indicating that this pathway might be useful for treating obesity and associated disorders. We report the full account of the synthesis and mitochondrial uncoupling bioactivity of lipidated N-acyl amino acids and their unnatural analogues. Unsaturated fatty acid chains of medium length and neutral amino acid head groups are required for optimal uncoupling activity on mammalian cells. A class of unnatural N-acyl amino acid analogues, characterized by isoindoline-1-carboxylate head groups (37), were resistant to enzymatic degradation by PM20D1 and maintained uncoupling bioactivity in cells and in mice.
- Published
- 2018
49. Tumor cell-adipocyte gap junctions activate lipolysis in breast cancer
- Author
-
Roman Camarda, Jeremy Williams, Serghei Malkov, Lisa J. Zimmerman, Suzanne Manning, Dvir Aran, Andrew Beardsley, Daniel Van de Mark, Yong Chen, Charles Berdan, Sharon M. Louie, Celine Mahieu, Daphne Superville, Matthew Gruner, Juliane Winkler, Elizabeth Willey, John D. Gagnon, Kosaku Shinoda, K. Mark Ansel, Zena Werb, Daniel K. Nomura, Shingo Kajimura, Atul J. Butte, Melinda E. Sanders, Daniel C. Liebler, Hope Rugo, Gregor Krings, John A. Shepherd, and Andrei Goga
- Subjects
0303 health sciences ,Stromal cell ,Chemistry ,Gap junction ,Connexin ,Cancer ,medicine.disease ,medicine.disease_cause ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Breast cancer ,030220 oncology & carcinogenesis ,Adipocyte ,medicine ,Cancer research ,Lipolysis ,Carcinogenesis ,030304 developmental biology - Abstract
During tumorigenesis, a heterotypic interface exists between cancer and stromal cells that can both support and repress tumor growth. In the breast, studies have demonstrated a pro-tumorigenic role for adipocytes. However, the molecular mechanisms by which breast cancer cells coopt adipocytes remain elusive. Studying breast tumors and normal adjacent tissue (NAT) from several patient cohorts, patient-derived xenografts and mouse models, we show that lipolysis and lipolytic signaling are activated in NAT. We investigated the tumor-adipocyte interface and find that functional gap junctions form between breast cancer cells and adipocytes. As a result, cAMP, a critical lipolysis-inducing signaling molecule, is transferred from breast cancer cells to adipocytes and activates lipolysis in a gap junction-dependent manner. We found that gap junction formation depends upon connexin 31 (Cx31), and that Cx31 is essential for breast tumor growth and activation of lipolysisin vivo. Thus, direct tumor cell-adipocyte interaction is critical for tumorigenesis, and may serve as a new therapeutic target in breast cancer.One sentence summaryGap junctions between breast cancer cells and adipocytes transfer cAMP and activate lipolysis in the breast tumor microenvironment.
- Published
- 2018
- Full Text
- View/download PDF
50. Deletion of Adipose Triglyceride Lipase Links Triacylglycerol Accumulation to a More-Aggressive Phenotype in A549 Lung Carcinoma Cells
- Author
-
Ruth Birner-Gruenberger, Juergen Gindlhuber, Gerhard G. Thallinger, Linda Waldherr, Bettina Pucher, Matthias Schittmayer, Daniel K. Nomura, Katarina Fritz, and Tamara Tomin
- Subjects
0301 basic medicine ,Proteomics ,Lung Neoplasms ,Phospholipid ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Western blot ,Cell Movement ,Lipid droplet ,medicine ,Humans ,Triglycerides ,medicine.diagnostic_test ,Cancer ,General Chemistry ,Lipase ,medicine.disease ,Lipid Metabolism ,Phenotype ,Cell biology ,030104 developmental biology ,src-Family Kinases ,chemistry ,A549 Cells ,030220 oncology & carcinogenesis ,Adipose triglyceride lipase ,Phosphorylation ,lipids (amino acids, peptides, and proteins) ,Gene Deletion ,Proto-oncogene tyrosine-protein kinase Src ,Signal Transduction - Abstract
Adipose triglyceride lipase (ATGL) catalyzes the rate limiting step in triacylglycerol breakdown in adipocytes but is expressed in most tissues. The enzyme was shown to be lost in many human tumors, and its loss may play a role in early stages of cancer development. Here, we report that loss of ATGL supports a more-aggressive cancer phenotype in a model system in which ATGL was deleted in A549 lung cancer cells by CRISPR/Cas9. We observed that loss of ATGL led to triacylglycerol accumulation in lipid droplets and higher levels of cellular phospholipid and bioactive lipid species (lyso- and ether-phospholipids). Label-free quantitative proteomics revealed elevated expression of the pro-oncogene SRC kinase in ATGL depleted cells, which was also found on mRNA level and confirmed on protein level by Western blot. Consistently, higher expression of phosphorylated (active) SRC (Y416 phospho-SRC) was observed in ATGL-KO cells. Cells depleted of ATGL migrated faster, which was dependent on SRC kinase activity. We propose that loss of ATGL may thus increase cancer aggressiveness by activation of pro-oncogenic signaling via SRC kinase and increased levels of bioactive lipids.
- Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.