1
|
Zhao MM, Ren TT, Wang JK, Yao L, Liu TT, Zhang JC, Liu Y, Yuan L, Liu D, Xu JH, Tu PF, Tang XD, Zeng KW. Endoplasmic reticulum membrane remodeling by targeting reticulon-4 induces pyroptosis to facilitate antitumor immune. Protein Cell 2025; 16:121-135. [PMID: 39252612 PMCID: PMC11786723 DOI: 10.1093/procel/pwae049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 08/12/2024] [Indexed: 09/11/2024] Open
Abstract
Pyroptosis is an identified programmed cell death that has been highly linked to endoplasmic reticulum (ER) dynamics. However, the crucial proteins for modulating dynamic ER membrane curvature change that trigger pyroptosis are currently not well understood. In this study, a biotin-labeled chemical probe of potent pyroptosis inducer α-mangostin (α-MG) was synthesized. Through protein microarray analysis, reticulon-4 (RTN4/Nogo), a crucial regulator of ER membrane curvature, was identified as a target of α-MG. We observed that chemically induced proteasome degradation of RTN4 by α-MG through recruiting E3 ligase UBR5 significantly enhances the pyroptosis phenotype in cancer cells. Interestingly, the downregulation of RTN4 expression significantly facilitated a dynamic remodeling of ER membrane curvature through a transition from tubules to sheets, consequently leading to rapid fusion of the ER with the cell plasma membrane. In particular, the ER-to-plasma membrane fusion process is supported by the observed translocation of several crucial ER markers to the "bubble" structures of pyroptotic cells. Furthermore, α-MG-induced RTN4 knockdown leads to pyruvate kinase M2 (PKM2)-dependent conventional caspase-3/gasdermin E (GSDME) cleavages for pyroptosis progression. In vivo, we observed that chemical or genetic RTN4 knockdown significantly inhibited cancer cells growth, which further exhibited an antitumor immune response with anti-programmed death-1 (anti-PD-1). In translational research, RTN4 high expression was closely correlated with the tumor metastasis and death of patients. Taken together, RTN4 plays a fundamental role in inducing pyroptosis through the modulation of ER membrane curvature remodeling, thus representing a prospective druggable target for anticancer immunotherapy.
Collapse
Affiliation(s)
- Mei-Mei Zhao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ting-Ting Ren
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing 100044, China
| | - Jing-Kang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Lu Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ting-Ting Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Ji-Chao Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Yang Liu
- Center of Basic Medical Research, Institute of Medical Innovation and Research, Peking University Third Hospital, Beijing 100191, China
| | - Lan Yuan
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing 100191, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing 100191, China
| | - Jiu-Hui Xu
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing 100044, China
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| | - Xiao-Dong Tang
- Beijing Key Laboratory of Musculoskeletal Tumor, Peking University People’s Hospital, Beijing 100044, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing 100191, China
| |
Collapse
|
2
|
Zou JY, Yang YY, Gu J, Liu F, Ye Z, Yi W, He Y. Asymmetric Allylic Substitution-Isomerization for the Modular Synthesis of Axially Chiral N-Vinylquinazolinones. Angew Chem Int Ed Engl 2023; 62:e202310320. [PMID: 37582683 DOI: 10.1002/anie.202310320] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/12/2023] [Accepted: 08/15/2023] [Indexed: 08/17/2023]
Abstract
Axially chiral N-substituted quinazolinones are important bioactive molecules, which are presented in many synthetic drugs. However, most strategies toward their atroposelective synthesis are mainly limited to the axially chiral arylquinazolinone frameworks. The development of modular synthetic methods to access diverse quinazolinone-based atropisomers remains scarce and challenging. Herein, we report the regio- and atroposelective synthesis of axially chiral N-vinylquinazolinones via the strategy of asymmetric allylic substitution-isomerization. The catalysis system utilized both asymmetric transition-metal catalysis and organocatalysis to efficiently afford trisubstituted and tetrasubstituted N-vinylquinazolinone atropisomers, respectively. With the meticulous design of β-substituted allylic substrates, both Z- and E-tetrasubstituted axially chiral N-vinylquinazolinones were obtained in good yields and high enantioselectivities.
Collapse
Affiliation(s)
- Jia-Yu Zou
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Yu-Ying Yang
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Jun Gu
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Fei Liu
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Zhiwen Ye
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Wenbin Yi
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| | - Ying He
- School of Chemistry and Chemical Engineering, Nanjing University of Science and Technology, Nanjing, 210094, China
| |
Collapse
|
3
|
Jiang Y, Cui J, Cui M, Jing R. SLC7A11 promotes the progression of gastric cancer and regulates ferroptosis through PI3K/AKT pathway. Pathol Res Pract 2023; 248:154646. [PMID: 37399587 DOI: 10.1016/j.prp.2023.154646] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 05/09/2023] [Accepted: 06/25/2023] [Indexed: 07/05/2023]
Abstract
OBJECTIVE Ferroptosis is a form of regulated cell death that occurs depending on iron and reactive oxygen species (ROS), but the underlying molecular mechanisms remain poorly understood. The aim of our study was to investigate the role of solute carrier family 7 member 11(SLC7A11) in the progression of gastric cancer (GC) and its molecular mechanism. METHOD The expression of SLC7A11 in GC was detected by real-time fluorescence quantitative polymerase chain reaction (RT-PCR), immunohistochemistry (IHC) and western blot. SLC7A11 interference and overexpression vector was constructed in vitro, transfected into GC cells, and the high efficiency plasmid vector fragment was screened.CCK-8 assay was used to detect the effect of cell proliferation. The migration ability of cells was detected by transwell assay. The mitochondrial structure was observed by transmission electron microscopy.CCK-8 assay was also used to detect the effect of SLC7A11 on the growth inhibition rate of ferroptosis in GC cells. The level of malondialdehyde (MDA), the ultimate product of lipid peroxidation, was detected by micro-method. The effect of SLC7A11 on PI3K/AKT signaling pathway was detected by Western blot. RESULTS SLC7A11 was significantly overexpressed in GC tissues than that in adjacent tissues. Knockdown of SLC7A11 inhibits cell proliferation, cell migration and invasion of GC, and increases the sensitivity of ferroptosis via moderating ROS and lipid peroxidation. Besides, overexpression of the SLC7A11 in GC cells reverses erastin-induced ferroptosis partially. Mechanistically, we reveal that suppression of SCL7A11 leads to inactivity of PI3K/AKT signaling pathway and further enhancing ferroptosis related lipid peroxidation, and thereby inhibiting GC progression. CONCLUSION SLC7A11 plays an oncogene role in malignant progression of GC. SLC7A11 reversely regulates ferroptosis of GC cells by activating PI3K/AKT signaling pathway. Silencing SLC7A11 expression can inhibit the progression of GC.
Collapse
Affiliation(s)
- Yun Jiang
- Department of Clinical Laboratory, Wuxi 9th People's Hospital Affiliated to Soochow University, China; Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Medical school of Nantong University, China
| | - Jingyi Cui
- Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Medical school of Nantong University, China
| | - Ming Cui
- Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Medical school of Nantong University, China.
| | - Rongrong Jing
- Department of Clinical Laboratory, Affiliated Hospital of Nantong University, Medical school of Nantong University, China.
| |
Collapse
|
4
|
Stockwell BR, Jiang X. The Chemistry and Biology of Ferroptosis. Cell Chem Biol 2021; 27:365-375. [PMID: 32294465 DOI: 10.1016/j.chembiol.2020.03.013] [Citation(s) in RCA: 257] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 03/07/2020] [Accepted: 03/17/2020] [Indexed: 01/09/2023]
Abstract
Ferroptosis is a recently described form of cell death driven by iron-dependent lipid peroxidation. This type of cell death was first observed in response to treatment of tumor cells with a small-molecule chemical probe named erastin. Most subsequent advances in understanding the mechanisms governing ferroptosis involved the use of genetic screens and small-molecule probes. We describe herein the utility and limitations of chemical probes that have been used to analyze and perturb ferroptosis, as well as mechanistic studies of ferroptosis that benefitted from the use of these probes and genetic screens. We also suggest probes for ferroptosis and highlight mechanistic questions surrounding this form of cell death that will be a high priority for exploration in the future.
Collapse
Affiliation(s)
- Brent R Stockwell
- Department of Chemistry and Department of Biological Sciences, Columbia University, New York, NY 10027, USA.
| | - Xuejun Jiang
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
5
|
Gao Z, Qian J, Yang H, Zhang J, Jiang G. Enantioselective Construction of C-C Axially Chiral Quinazolinones via Chirality Exchange and Phase-Transfer Catalysis. Org Lett 2021; 23:1731-1737. [PMID: 33586979 DOI: 10.1021/acs.orglett.1c00156] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
A family of axially chiral quinazolinone-based heterobiaryls were constructed with high levels of enantiocontrol (up to 94% ee). Convergently, three different synthetic methods have been realized to prepare these valuable compounds including central-to-axial chirality transfer, dynamic kinetic resolution, and phase-transfer catalysis. Importantly, novel P,N-ligands with a π-π stacking can be derived from heterobiaryls by chirality exchange strategy or synthesized directly from complementary phase-transfer catalysis by using the inexpensive chiral quaternary ammonium salt.
Collapse
Affiliation(s)
- Zeng Gao
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, China.,University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jinlong Qian
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, China
| | - Huameng Yang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, China
| | - Jinlong Zhang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, China
| | - Gaoxi Jiang
- State Key Laboratory for Oxo Synthesis and Selective Oxidation, Center for Excellence in Molecular Synthesis, Suzhou Research Institute of LICP, Lanzhou Institute of Chemical Physics (LICP), Chinese Academy of Sciences, Lanzhou 730000, China
| |
Collapse
|
6
|
Teng F, Yu T, Peng Y, Hu W, Hu H, He Y, Luo S, Zhu Q. Palladium-Catalyzed Atroposelective Coupling–Cyclization of 2-Isocyanobenzamides to Construct Axially Chiral 2-Aryl- and 2,3-Diarylquinazolinones. J Am Chem Soc 2021; 143:2722-2728. [DOI: 10.1021/jacs.1c00640] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Fan Teng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
- Key Laboratory of Jiangxi Province for Persistent Pollutants Control and Resources Recycle, Nanchang Hangkong University, Nanchang 330063, China
| | - Ting Yu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Yan Peng
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Weiming Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
| | - Huaanzi Hu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Yimiao He
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, College of Chemistry and Materials, Nanning Normal University, Nanning 530001, China
| | - Shuang Luo
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| | - Qiang Zhu
- State Key Laboratory of Respiratory Disease, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, 190 Kaiyuan Avenue, Guangzhou 510530, China
- University of Chinese Academy of Sciences, No. 19(A) Yuquan Road, Shijingshan District, Beijing 100049, China
- Guangxi Key Laboratory of Natural Polymer Chemistry and Physics, College of Chemistry and Materials, Nanning Normal University, Nanning 530001, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510005, China
| |
Collapse
|
7
|
Shen L, Lin D, Li X, Wu H, Lenahan C, Pan Y, Xu W, Chen Y, Shao A, Zhang J. Ferroptosis in Acute Central Nervous System Injuries: The Future Direction? Front Cell Dev Biol 2020; 8:594. [PMID: 32760721 PMCID: PMC7373735 DOI: 10.3389/fcell.2020.00594] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 06/18/2020] [Indexed: 12/15/2022] Open
Abstract
Acute central nervous system (CNS) injuries, such as stroke, traumatic brain injury (TBI), and spinal cord injury (SCI) present a grave health care challenge worldwide due to high morbidity and mortality, as well as limited clinical therapeutic strategies. Established literature has shown that oxidative stress (OS), inflammation, excitotoxicity, and apoptosis play important roles in the pathophysiological processes of acute CNS injuries. Recently, there have been many studies on the topic of ferroptosis, a form of regulated cell death characterized by the accumulation of iron-dependent lipid peroxidation. Some studies have revealed an emerging connection between acute CNS injuries and ferroptosis. Ferroptosis, induced by the abnormal metabolism of lipids, glutathione (GSH), and iron, can accelerate acute CNS injuries. However, pharmaceutical agents, such as iron chelators, ferrostatin-1 (Fer-1), and liproxstatin-1 (Lip-1), can inhibit ferroptosis and may have neuroprotective effects after acute CNS injuries. However, the specific mechanisms underlying this connection has not yet been clearly elucidated. In this paper, we discuss the general mechanisms of ferroptosis and its role in stroke, TBI, and SCI. We also summarize ferroptosis-related drugs and highlight the potential therapeutic strategies in treating various acute CNS injuries. Additionally, this paper suggests a testable hypothesis that ferroptosis may be a novel direction for further research of acute CNS injuries by providing corresponding evidence.
Collapse
Affiliation(s)
- Lesang Shen
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Danfeng Lin
- Department of Surgical Oncology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyi Li
- Department of Nuclear Medicine and PET-CT Center, The Second Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haijian Wu
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cameron Lenahan
- Burrell College of Osteopathic Medicine, Las Cruces, NM, United States.,Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Yuanbo Pan
- Burrell College of Osteopathic Medicine, Las Cruces, NM, United States.,Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Weilin Xu
- Burrell College of Osteopathic Medicine, Las Cruces, NM, United States.,Center for Neuroscience Research, School of Medicine, Loma Linda University, Loma Linda, CA, United States
| | - Yiding Chen
- Department of Breast Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Anwen Shao
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
8
|
Bateman LA, Nguyen TB, Roberts AM, Miyamoto DK, Ku WM, Huffman TR, Petri Y, Heslin MJ, Contreras CM, Skibola CF, Olzmann JA, Nomura DK. Chemoproteomics-enabled covalent ligand screen reveals a cysteine hotspot in reticulon 4 that impairs ER morphology and cancer pathogenicity. Chem Commun (Camb) 2018; 53:7234-7237. [PMID: 28352901 DOI: 10.1039/c7cc01480e] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Chemical genetics has arisen as a powerful approach for identifying novel anti-cancer agents. However, a major bottleneck of this approach is identifying the targets of lead compounds that arise from screens. Here, we coupled the synthesis and screening of fragment-based cysteine-reactive covalent ligands with activity-based protein profiling (ABPP) chemoproteomic approaches to identify compounds that impair colorectal cancer pathogenicity and map the druggable hotspots targeted by these hits. Through this coupled approach, we discovered a cysteine-reactive acrylamide DKM 3-30 that significantly impaired colorectal cancer cell pathogenicity through targeting C1101 on reticulon 4 (RTN4). While little is known about the role of RTN4 in colorectal cancer, this protein has been established as a critical mediator of endoplasmic reticulum tubular network formation. We show here that covalent modification of C1101 on RTN4 by DKM 3-30 or genetic knockdown of RTN4 impairs endoplasmic reticulum and nuclear envelope morphology as well as colorectal cancer pathogenicity. We thus put forth RTN4 as a potential novel colorectal cancer therapeutic target and reveal a unique druggable hotspot within RTN4 that can be targeted by covalent ligands to impair colorectal cancer pathogenicity. Our results underscore the utility of coupling the screening of fragment-based covalent ligands with isoTOP-ABPP platforms for mining the proteome for novel druggable nodes that can be targeted for cancer therapy.
Collapse
Affiliation(s)
- L A Bateman
- Departments of Chemistry and Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Anderson KE, To M, Olzmann JA, Nomura DK. Chemoproteomics-Enabled Covalent Ligand Screening Reveals a Thioredoxin-Caspase 3 Interaction Disruptor That Impairs Breast Cancer Pathogenicity. ACS Chem Biol 2017; 12:2522-2528. [PMID: 28892616 PMCID: PMC6205226 DOI: 10.1021/acschembio.7b00711] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Covalent ligand discovery is a promising strategy to develop small-molecule effectors against therapeutic targets. Recent studies have shown that dichlorotriazines are promising reactive scaffolds that preferentially react with lysines. Here, we have synthesized a series of dichlorotriazine-based covalent ligands and have screened this library to reveal small molecules that impair triple-negative breast cancer cell survival. Upon identifying a lead hit from this screen KEA1-97, we used activity-based protein profiling (ABPP)-based chemoproteomic platforms to identify that this compound targets lysine 72 of thioredoxin-a site previously shown to be important in protein interactions with caspase 3 to inhibit caspase 3 activity and suppress apoptosis. We show that KEA1-97 disrupts the interaction of thioredoxin with caspase 3, activates caspases, and induces apoptosis without affecting thioredoxin activity. Moreover, KEA1-97 impairs in vivo breast tumor xenograft growth. Our study showcases how the screening of covalent ligands can be coupled with ABPP platforms to identify unique anticancer lead and target pairs.
Collapse
Affiliation(s)
- Kimberly E Anderson
- Departments of Chemistry, Molecular and Cell Biology, and ‡Nutritional Sciences and Toxicology, University of California, Berkeley , Berkeley, California 94720, United States
| | - Milton To
- Departments of Chemistry, Molecular and Cell Biology, and ‡Nutritional Sciences and Toxicology, University of California, Berkeley , Berkeley, California 94720, United States
| | - James A Olzmann
- Departments of Chemistry, Molecular and Cell Biology, and ‡Nutritional Sciences and Toxicology, University of California, Berkeley , Berkeley, California 94720, United States
| | - Daniel K Nomura
- Departments of Chemistry, Molecular and Cell Biology, and ‡Nutritional Sciences and Toxicology, University of California, Berkeley , Berkeley, California 94720, United States
| |
Collapse
|
10
|
Brønsted acid-catalysed enantioselective construction of axially chiral arylquinazolinones. Nat Commun 2017; 8:15489. [PMID: 28524863 PMCID: PMC5454535 DOI: 10.1038/ncomms15489] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2017] [Accepted: 04/03/2017] [Indexed: 11/21/2022] Open
Abstract
The axially chiral arylquinazolinone acts as a privileged structural scaffold, which is present in a large number of natural products and biologically active compounds as well as in chiral ligands. However, a direct catalytic enantioselective approach to access optically pure arylquinazolinones has been underexplored. Here we show a general and efficient approach to access enantiomerically pure arylquinazolinones in one-pot fashion catalysed by chiral phosphoric acids. A variety of axially chiral arylquinazolinones were obtained in high yields with good to excellent enantioselectivities under mild condition. Furthermore, we disclosed a method for atroposelective synthesis of alkyl-substituted arylquinazolinones involving Brønsted acid-catalysed carbon–carbon bond cleavage strategy. Finally, the asymmetric total synthesis of eupolyphagin bearing a cyclic arylquinazolinone skeleton was accomplished with an overall yield of 32% in six steps by utilizing the aforementioned methodology. Axially chiral arylquinazolinones are structural motifs in several natural products and can also act as chiral ligands. Here, the authors show a chiral phosphoric acid-catalysed strategy to access enantiomerically pure arylquinazolinones by efficient transfer of central chirality into axial chirality.
Collapse
|
11
|
Roberts AM, Miyamoto DK, Huffman TR, Bateman LA, Ives AN, Akopian D, Heslin MJ, Contreras CM, Rape M, Skibola CF, Nomura DK. Chemoproteomic Screening of Covalent Ligands Reveals UBA5 As a Novel Pancreatic Cancer Target. ACS Chem Biol 2017; 12:899-904. [PMID: 28186401 DOI: 10.1021/acschembio.7b00020] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
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.
Collapse
Affiliation(s)
- Allison M. Roberts
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - David K. Miyamoto
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Tucker R. Huffman
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Leslie A. Bateman
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - Ashley N. Ives
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| | - David Akopian
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Martin J. Heslin
- The University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Carlo M. Contreras
- The University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Michael Rape
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California 94720, United States
| | - Christine F. Skibola
- The University of Alabama at Birmingham, Birmingham, Alabama 35233, United States
| | - Daniel K. Nomura
- Departments of Chemistry,
Molecular and Cell Biology, and Nutritional Sciences and Toxicology, 127 Morgan Hall, University of California, Berkeley, Berkeley, California 94720, United States
| |
Collapse
|
12
|
Collina S, Bignardi E, Rui M, Rossi D, Gaggeri R, Zamagni A, Cortesi M, Tesei A. Are sigma modulators an effective opportunity for cancer treatment? A patent overview (1996-2016). Expert Opin Ther Pat 2017; 27:565-578. [PMID: 28051882 DOI: 10.1080/13543776.2017.1276569] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
INTRODUCTION Although several molecular targets against cancer have been identified, there is a continuous need for new therapeutic strategies. Sigma Receptors (SRs) overexpression has been recently associated with different cancer conditions. Therefore, novel anticancer agents targeting SRs may increase the specificity of therapies, overcoming some of the common drawbacks of conventional chemotherapy. Areas covered: The present review focuses on patent documents disclosing SR modulators with possible application in cancer therapy and diagnosis. The analysis reviews patents of the last two decades (1996-2016); patents were grouped according to target subtypes (S1R, S2R, pan-SRs) and relevant Applicants. The literature was searched through Espacenet, ISI Web, PatentScope and PubMed databases. Expert opinion: The number of patents related to SRs and cancer has increased in the last twenty years, confirming the importance of this receptor family as valuable target against neoplasias. Despite their short history in the cancer scenario, many SR modulators are at pre-clinical stage and one is undergoing a phase II clinical trial. SRs ligands may represent a powerful source of innovative antitumor therapeutics. Further investigation is needed for validating SR modulators as anti-cancer drugs. We strongly hope that this review could stimulate the interest of both Academia and pharmaceutical companies.
Collapse
Affiliation(s)
- Simona Collina
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Emanuele Bignardi
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Marta Rui
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Daniela Rossi
- a Drug Sciences Department, Medicinal Chemistry and Pharmaceutical Technology Section , University of Pavia , Pavia , Italy
| | - Raffaella Gaggeri
- b Pharmacy Unit , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| | - Alice Zamagni
- c Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| | - Michela Cortesi
- c Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| | - Anna Tesei
- d MBiochem, Biosciences Laboratory , Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST), IRCCS , Meldola , Italy
| |
Collapse
|
13
|
Diener ME, Metrano AJ, Kusano S, Miller SJ. Enantioselective synthesis of 3-arylquinazolin-4(3H)-ones via peptide-catalyzed atroposelective bromination. J Am Chem Soc 2015; 137:12369-77. [PMID: 26343278 PMCID: PMC5134330 DOI: 10.1021/jacs.5b07726] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
We report the development of a tertiary amine-containing β-turn peptide that catalyzes the atroposelective bromination of pharmaceutically relevant 3-arylquinazolin-4(3H)-ones (quinazolinones) with high levels of enantioinduction over a broad substrate scope. The structure of the free catalyst and the peptide-substrate complex were explored using X-ray crystallography and 2D-NOESY experiments. Quinazolinone rotational barriers about the chiral anilide axis were also studied using density functional theory calculations and are discussed in light of the high enantioselectivities observed. Mechanistic studies also suggest that the initial bromination event is stereodetermining, and the major monobromide intermediate is an atropisomerically stable, mono-ortho-substituted isomer. The observation of stereoisomerically stable monobromides stimulated the conversion of the tribromide products to other atropisomerically defined products of interest. For example, (1) a dehalogenation Suzuki-Miyaura cross-coupling sequence delivers ortho-arylated derivatives, and (2) a regioselective Buchwald-Hartwig amination procedure installs para-amine functionality. Stereochemical information was retained during these subsequent transformations.
Collapse
Affiliation(s)
| | | | | | - Scott J. Miller
- Department of Chemistry, Yale University, New Haven, CT 06520-8107, United States
| |
Collapse
|
14
|
Meesters C, Kombrink E. Screening for bioactive small molecules by in vivo monitoring of luciferase-based reporter gene expression in Arabidopsis thaliana. Methods Mol Biol 2014; 1056:19-31. [PMID: 24306859 DOI: 10.1007/978-1-62703-592-7_3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Chemical genetics is a scientific strategy that utilizes bioactive small molecules as experimental tools to dissect biological processes. Bioactive compounds occurring in nature represent an enormous diversity of structures that potentially can be used as activators or inhibitors of biochemical pathways, transport processes, regulatory networks, or developmental programs. Screening methods to identify bioactive small molecules can vary greatly, ranging from visual evaluation of phenotypic alterations to quantifying biometric traits such as enzyme activities. Here, we describe a general methodology that permits identification of compounds modulating the expression of reporter genes in Arabidopsis thaliana seedlings. The selection of luciferase-based reporter systems has the advantage that it allows in vivo imaging of reporter gene activity in a semiquantitative manner without affecting plant viability. We chose an Arabidopsis line harboring the luciferase reporter under the control of the jasmonate-inducible LOX2 promoter to screen for either activators or inhibitors of gene expression. The outlined assay conditions can readily be applied to Arabidopsis lines containing other reporter genes. Thereby screening for small molecules affecting different signaling pathways and/or phenotypic responses is possible.
Collapse
Affiliation(s)
- Christian Meesters
- Department of Plant-Microbe Interactions, Max Planck Institute for Plant Breeding Research, Köln, Germany
| | | |
Collapse
|
15
|
Audenaert D, Nguyen L, De Rybel B, Beeckman T. Fully automated compound screening in Arabidopsis thaliana seedlings. Methods Mol Biol 2014; 1056:3-9. [PMID: 24306857 DOI: 10.1007/978-1-62703-592-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
High-throughput small molecule screenings in model plants are of great value to identify compounds that interfere with plant developmental processes. In academic research, the plant Arabidopsis thaliana is the most commonly used model organism for this purpose. However, compared to plant cellular systems, A. thaliana plants are less amenable to develop high-throughput screening assays. In this chapter, we describe a screening procedure that is compatible with liquid handling systems and increases the throughput of compound screenings in A. thaliana seedlings.
Collapse
|
16
|
Affiliation(s)
- Marco Colombini
- Department of Biology,
University of Maryland, College
Park, Maryland 20742, United States
| |
Collapse
|
17
|
Cottier S, Mönig T, Wang Z, Svoboda J, Boland W, Kaiser M, Kombrink E. The yeast three-hybrid system as an experimental platform to identify proteins interacting with small signaling molecules in plant cells: potential and limitations. FRONTIERS IN PLANT SCIENCE 2011; 2:101. [PMID: 22639623 PMCID: PMC3355722 DOI: 10.3389/fpls.2011.00101] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2011] [Accepted: 12/07/2011] [Indexed: 05/18/2023]
Abstract
Chemical genetics is a powerful scientific strategy that utilizes small bioactive molecules as experimental tools to unravel biological processes. Bioactive compounds occurring in nature represent an enormous diversity of structures that can be used to dissect functions of biological systems. Once the bioactivity of a natural or synthetic compound has been critically evaluated the challenge remains to identify its molecular target and mode of action, which usually is a time-consuming and labor-intensive process. To facilitate this task, we decided to implement the yeast three-hybrid (Y3H) technology as a general experimental platform to scan the whole Arabidopsis proteome for targets of small signaling molecules. The Y3H technology is based on the yeast two-hybrid system and allows direct cloning of proteins that interact in vivo with a synthetic hybrid ligand, which comprises the biologically active molecule of interest covalently linked to methotrexate (Mtx). In yeast nucleus the hybrid ligand connects two fusion proteins: the Mtx part binding to dihydrofolate reductase fused to a DNA-binding domain (encoded in the yeast strain), and the bioactive molecule part binding to its potential protein target fused to a DNA-activating domain (encoded on a cDNA expression vector). During cDNA library screening, the formation of this ternary, transcriptional activator complex leads to reporter gene activation in yeast cells, and thereby allows selection of the putative targets of small bioactive molecules of interest. Here we present the strategy and experimental details for construction and application of a Y3H platform, including chemical synthesis of different hybrid ligands, construction of suitable cDNA libraries, the choice of yeast strains, and appropriate screening conditions. Based on the results obtained and the current literature we discuss the perspectives and limitations of the Y3H approach for identifying targets of small bioactive molecules.
Collapse
Affiliation(s)
- Stéphanie Cottier
- Department of Plant-Microbe Interactions, Max Planck Institute for Plant Breeding ResearchKöln, Germany
| | - Timon Mönig
- Center for Medical Biotechnology, University of Duisburg–EssenEssen, Germany
| | - Zheming Wang
- Center for Medical Biotechnology, University of Duisburg–EssenEssen, Germany
| | - Jiří Svoboda
- Department of Bioorganic Chemistry, Max Planck Institute for Chemical EcologyJena, Germany
| | - Wilhelm Boland
- Department of Bioorganic Chemistry, Max Planck Institute for Chemical EcologyJena, Germany
| | - Markus Kaiser
- Center for Medical Biotechnology, University of Duisburg–EssenEssen, Germany
| | - Erich Kombrink
- Department of Plant-Microbe Interactions, Max Planck Institute for Plant Breeding ResearchKöln, Germany
| |
Collapse
|
18
|
Sekhar KR, Reddy YT, Reddy PN, Crooks PA, Venkateswaran A, McDonald WH, Geng L, Sasi S, Van Der Waal RP, Roti JLR, Salleng KJ, Rachakonda G, Freeman ML. The novel chemical entity YTR107 inhibits recruitment of nucleophosmin to sites of DNA damage, suppressing repair of DNA double-strand breaks and enhancing radiosensitization. Clin Cancer Res 2011; 17:6490-9. [PMID: 21878537 DOI: 10.1158/1078-0432.ccr-11-1054] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
PURPOSE Radiation therapy continues to be an important therapeutic strategy for providing definitive local/regional control of human cancer. However, oncogenes that harbor driver mutations and/or amplifications can compromise therapeutic efficacy. Thus, there is a need for novel approaches that enhance the DNA damage produced by ionizing radiation. EXPERIMENTAL DESIGN A forward chemical genetic approach coupled with cell-based phenotypic screening of several tumor cell lines was used to identify a novel chemical entity (NCE) that functioned as a radiation sensitizer. Proteomics, comet assays, confocal microscopy, and immunoblotting were used to identify the biological target. RESULTS The screening process identified a 5-((N-benzyl-1H-indol-3-yl)-methylene)pyrimidine-2,4,6(1H,3H,5H)trione as an NCE that radiosensitized cancer cells expressing amplified and/or mutated RAS, ErbB, PIK3CA, and/or BRAF oncogenes. Affinity-based solid-phase resin capture followed by liquid chromatography/tandem mass spectrometry identified the chaperone nucleophosmin (NPM) as the NCE target. SiRNA suppression of NPM abrogated radiosensitization by the NCE. Confocal microscopy showed that the NCE inhibited NPM shuttling to radiation-induced DNA damage repair foci, and the analysis of comet assays indicated a diminished rate of DNA double-strand break repair. CONCLUSION These data support the hypothesis that inhibition of DNA repair due to inhibition of NPM shuttling increases the efficacy of DNA-damaging therapeutic strategies.
Collapse
Affiliation(s)
- Konjeti R Sekhar
- Department of Radiation Oncology, Division of Animal Care, Vanderbilt University School of Medicine, Nashville, Tennessee, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
A cyclopalladated complex interacts with mitochondrial membrane thiol-groups and induces the apoptotic intrinsic pathway in murine and cisplatin-resistant human tumor cells. BMC Cancer 2011; 11:296. [PMID: 21756336 PMCID: PMC3156809 DOI: 10.1186/1471-2407-11-296] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2010] [Accepted: 07/14/2011] [Indexed: 12/11/2022] Open
Abstract
Background Systemic therapy for cancer metastatic lesions is difficult and generally renders a poor clinical response. Structural analogs of cisplatin, the most widely used synthetic metal complexes, show toxic side-effects and tumor cell resistance. Recently, palladium complexes with increased stability are being investigated to circumvent these limitations, and a biphosphinic cyclopalladated complex {Pd2 [S(-)C2, N-dmpa]2 (μ-dppe)Cl2} named C7a efficiently controls the subcutaneous development of B16F10-Nex2 murine melanoma in syngeneic mice. Presently, we investigated the melanoma cell killing mechanism induced by C7a, and extended preclinical studies. Methods B16F10-Nex2 cells were treated in vitro with C7a in the presence/absence of DTT, and several parameters related to apoptosis induction were evaluated. Preclinical studies were performed, and mice were endovenously inoculated with B16F10-Nex2 cells, intraperitoneally treated with C7a, and lung metastatic nodules were counted. The cytotoxic effects and the respiratory metabolism were also determined in human tumor cell lines treated in vitro with C7a. Results Cyclopalladated complex interacts with thiol groups on the mitochondrial membrane proteins, causes dissipation of the mitochondrial membrane potential, and induces Bax translocation from the cytosol to mitochondria, colocalizing with a mitochondrial tracker. C7a also induced an increase in cytosolic calcium concentration, mainly from intracellular compartments, and a significant decrease in the ATP levels. Activation of effector caspases, chromatin condensation and DNA degradation, suggested that C7a activates the apoptotic intrinsic pathway in murine melanoma cells. In the preclinical studies, the C7a complex protected against murine metastatic melanoma and induced death in several human tumor cell lineages in vitro, including cisplatin-resistant ones. The mitochondria-dependent cell death was also induced by C7a in human tumor cells. Conclusions The cyclopalladated C7a complex is an effective chemotherapeutic anticancer compound against primary and metastatic murine and human tumors, including cisplatin-resistant cells, inducing apoptotic cell death via the intrinsic pathway.
Collapse
|
20
|
Portt L, Norman G, Clapp C, Greenwood M, Greenwood MT. Anti-apoptosis and cell survival: a review. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1813:238-59. [PMID: 20969895 DOI: 10.1016/j.bbamcr.2010.10.010] [Citation(s) in RCA: 447] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Revised: 10/04/2010] [Accepted: 10/11/2010] [Indexed: 02/08/2023]
Abstract
Type I programmed cell death (PCD) or apoptosis is critical for cellular self-destruction for a variety of processes such as development or the prevention of oncogenic transformation. Alternative forms, including type II (autophagy) and type III (necrotic) represent the other major types of PCD that also serve to trigger cell death. PCD must be tightly controlled since disregulated cell death is involved in the development of a large number of different pathologies. To counter the multitude of processes that are capable of triggering death, cells have devised a large number of cellular processes that serve to prevent inappropriate or premature PCD. These cell survival strategies involve a myriad of coordinated and systematic physiological and genetic changes that serve to ward off death. Here we will discuss the different strategies that are used to prevent cell death and focus on illustrating that although anti-apoptosis and cellular survival serve to counteract PCD, they are nevertheless mechanistically distinct from the processes that regulate cell death.
Collapse
Affiliation(s)
- Liam Portt
- Department of Chemistry and Chemical Engineering, Royal Military College, Ontario, Canada
| | | | | | | | | |
Collapse
|
21
|
Geng L, Rachakonda G, Morré DJ, Morré DM, Crooks PA, Sonar VN, Roti JLR, Rogers BE, Greco S, Ye F, Salleng KJ, Sasi S, Freeman ML, Sekhar KR. Indolyl-quinuclidinols inhibit ENOX activity and endothelial cell morphogenesis while enhancing radiation-mediated control of tumor vasculature. FASEB J 2009; 23:2986-95. [PMID: 19395476 DOI: 10.1096/fj.09-130005] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a need for novel strategies that target tumor vasculature, specifically those that synergize with cytotoxic therapy, in order to overcome resistance that can develop with current therapeutics. A chemistry-driven drug discovery screen was employed to identify novel compounds that inhibit endothelial cell tubule formation. Cell-based phenotypic screening revealed that noncytotoxic concentrations of (Z)-(+/-)-2-(1-benzenesulfonylindol-3-ylmethylene)-1-azabicyclo[2. 2.2]octan-3-ol (analog I) and (Z)-(+/-)-2-(1-benzylindol-3-ylmethylene)-1-azabicyclo[2.2.2]octan-3-ol (analog II) inhibited endothelial cell migration and the ability to form capillary-like structures in Matrigel by > or =70%. The ability to undergo neoangiogenesis, as measured in a window-chamber model, was also inhibited by 70%. Screening of biochemical pathways revealed that analog II inhibited the enzyme ENOX1 (EC(50) = 10 microM). Retroviral-mediated shRNA suppression of endothelial ENOX1 expression inhibited cell migration and tubule formation, recapitulating the effects observed with the small-molecule analogs. Genetic or chemical suppression of ENOX1 significantly increased radiation-mediated Caspase3-activated apoptosis, coincident with suppression of p70S6K1 phosphorylation. Administration of analog II prior to fractionated X-irradiation significantly diminished the number and density of tumor microvessels, as well as delayed syngeneic and xenograft tumor growth compared to results obtained with radiation alone. Analysis of necropsies suggests that the analog was well tolerated. These results suggest that targeting ENOX1 activity represents a novel therapeutic strategy for enhancing the radiation response of tumors.
Collapse
Affiliation(s)
- Ling Geng
- Department of Radiation Oncology, Vanderbilt University School of Medicine, Nashville, TN 37232, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Nandy JP, Prakesch M, Khadem S, Reddy PT, Sharma U, Arya P. Advances in Solution- and Solid-Phase Synthesis toward the Generation of Natural Product-like Libraries. Chem Rev 2009; 109:1999-2060. [DOI: 10.1021/cr800188v] [Citation(s) in RCA: 139] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Jyoti P. Nandy
- Ontario Institute for Cancer Research, MaRS Centre, South Tower, 101 College Street, Toronto, Ontario M5G 1L7, Canada, Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada, and Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Michael Prakesch
- Ontario Institute for Cancer Research, MaRS Centre, South Tower, 101 College Street, Toronto, Ontario M5G 1L7, Canada, Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada, and Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Shahriar Khadem
- Ontario Institute for Cancer Research, MaRS Centre, South Tower, 101 College Street, Toronto, Ontario M5G 1L7, Canada, Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada, and Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - P. Thirupathi Reddy
- Ontario Institute for Cancer Research, MaRS Centre, South Tower, 101 College Street, Toronto, Ontario M5G 1L7, Canada, Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada, and Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Utpal Sharma
- Ontario Institute for Cancer Research, MaRS Centre, South Tower, 101 College Street, Toronto, Ontario M5G 1L7, Canada, Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada, and Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Prabhat Arya
- Ontario Institute for Cancer Research, MaRS Centre, South Tower, 101 College Street, Toronto, Ontario M5G 1L7, Canada, Steacie Institute for Molecular Sciences, National Research Council of Canada, 100 Sussex Drive, Ottawa, Ontario K1A 0R6, Canada, and Department of Chemistry, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| |
Collapse
|
23
|
Connecting chemosensitivity, gene expression and disease. Trends Pharmacol Sci 2007; 29:1-5. [PMID: 18055024 DOI: 10.1016/j.tips.2007.10.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2007] [Revised: 10/03/2007] [Accepted: 10/05/2007] [Indexed: 01/21/2023]
Abstract
Omics-based investigations offer potentially powerful readouts that might be useful for probing the underlying biology of normal and diseased states, identifying novel therapeutic targets and proposing relevant markers for designing treatment strategies. A vital component of these investigations involves a systematic analysis of gene expression and chemosensitivity data in the context of disease states and small molecule probes into the function of targets responsible for a disease phenotype. Systematic analysis of chemical and pharmacogenetics data offers a possible means to identify novel, small-molecule, potentially therapeutic, agents that affect the phenotype of a particular target. Elegantly simple in concept, the covariation of genetic and chemosensitivity readouts provide a hypothetical link for relating compounds through genomic expression profiles to underlying biology.
Collapse
|
24
|
Walsh TA. The emerging field of chemical genetics: potential applications for pesticide discovery. PEST MANAGEMENT SCIENCE 2007; 63:1165-71. [PMID: 17912687 DOI: 10.1002/ps.1452] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The use of small molecules to probe biological systems, generally described as 'chemical genetics', has grown considerably in the past 7 years, especially in areas related to human biology and therapeutics. This review describes some aspects of chemical genetics technologies that can be usefully applied to pesticide target discovery and lead generation. The chemical genetics approach (consisting of a phenotype screen, a chemical library and a robust target identification methodology) is compared with conventional and target-based screening. The outcomes of a chemical genetics approach are novel protein targets coupled with in vivo-active chemical ligands. The 'chemistry-first' paradigm of the chemical genetics approach can circumvent some of the obstacles that have emerged for the exploitation of novel but chemically unvalidated targets identified from genetic or genomic screens. Some of the advantages and challenges in using chemical genetics approaches are reviewed.
Collapse
Affiliation(s)
- Terence A Walsh
- Dow AgroSciences, 9330 Zionsville Road, Indianapolis, IN 46268, USA.
| |
Collapse
|
25
|
Abstract
Recognition of some of the limitations of target-based drug discovery has recently led to the renaissance of a more holistic approach in which complex biological systems are investigated for phenotypic changes upon exposure to small molecules. The subsequent identification of the molecular targets that underlie an observed phenotypic response--termed target deconvolution--is an important aspect of current drug discovery, as knowledge of the molecular targets will greatly aid drug development. Here, the broad panel of experimental strategies that can be applied to target deconvolution is critically reviewed.
Collapse
|