1
|
Garland KM, Sheehy TL, Wilson JT. Chemical and Biomolecular Strategies for STING Pathway Activation in Cancer Immunotherapy. Chem Rev 2022; 122:5977-6039. [PMID: 35107989 PMCID: PMC8994686 DOI: 10.1021/acs.chemrev.1c00750] [Citation(s) in RCA: 165] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The stimulator of interferon genes (STING) cellular signaling pathway is a promising target for cancer immunotherapy. Activation of the intracellular STING protein triggers the production of a multifaceted array of immunostimulatory molecules, which, in the proper context, can drive dendritic cell maturation, antitumor macrophage polarization, T cell priming and activation, natural killer cell activation, vascular reprogramming, and/or cancer cell death, resulting in immune-mediated tumor elimination and generation of antitumor immune memory. Accordingly, there is a significant amount of ongoing preclinical and clinical research toward further understanding the role of the STING pathway in cancer immune surveillance as well as the development of modulators of the pathway as a strategy to stimulate antitumor immunity. Yet, the efficacy of STING pathway agonists is limited by many drug delivery and pharmacological challenges. Depending on the class of STING agonist and the desired administration route, these may include poor drug stability, immunocellular toxicity, immune-related adverse events, limited tumor or lymph node targeting and/or retention, low cellular uptake and intracellular delivery, and a complex dependence on the magnitude and kinetics of STING signaling. This review provides a concise summary of the STING pathway, highlighting recent biological developments, immunological consequences, and implications for drug delivery. This review also offers a critical analysis of an expanding arsenal of chemical strategies that are being employed to enhance the efficacy, safety, and/or clinical utility of STING pathway agonists and lastly draws attention to several opportunities for therapeutic advancements.
Collapse
Affiliation(s)
- Kyle M Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - Taylor L Sheehy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
| | - John T Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, 37235 United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee, 37232 United States
| |
Collapse
|
2
|
Garland KM, Rosch JC, Carson CS, Wang-Bishop L, Hanna A, Sevimli S, Van Kaer C, Balko JM, Ascano M, Wilson JT. Pharmacological Activation of cGAS for Cancer Immunotherapy. Front Immunol 2021; 12:753472. [PMID: 34899704 PMCID: PMC8662543 DOI: 10.3389/fimmu.2021.753472] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 10/29/2021] [Indexed: 01/23/2023] Open
Abstract
When compartmentally mislocalized within cells, nucleic acids can be exceptionally immunostimulatory and can even trigger the immune-mediated elimination of cancer. Specifically, the accumulation of double-stranded DNA in the cytosol can efficiently promote antitumor immunity by activating the cGAMP synthase (cGAS) / stimulator of interferon genes (STING) cellular signaling pathway. Targeting this cytosolic DNA sensing pathway with interferon stimulatory DNA (ISD) is therefore an attractive immunotherapeutic strategy for the treatment of cancer. However, the therapeutic activity of ISD is limited by several drug delivery barriers, including susceptibility to deoxyribonuclease degradation, poor cellular uptake, and inefficient cytosolic delivery. Here, we describe the development of a nucleic acid immunotherapeutic, NanoISD, which overcomes critical delivery barriers that limit the activity of ISD and thereby promotes antitumor immunity through the pharmacological activation of cGAS at the forefront of the STING pathway. NanoISD is a nanoparticle formulation that has been engineered to confer deoxyribonuclease resistance, enhance cellular uptake, and promote endosomal escape of ISD into the cytosol, resulting in potent activation of the STING pathway via cGAS. NanoISD mediates the local production of proinflammatory cytokines via STING signaling. Accordingly, the intratumoral administration of NanoISD induces the infiltration of natural killer cells and T lymphocytes into murine tumors. The therapeutic efficacy of NanoISD is demonstrated in preclinical tumor models by attenuated tumor growth, prolonged survival, and an improved response to immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Kyle M. Garland
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Jonah C. Rosch
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Carcia S. Carson
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
| | - Lihong Wang-Bishop
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Ann Hanna
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Sema Sevimli
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
| | - Casey Van Kaer
- Department of Bioengineering, Northeastern University, Boston, MA, United States
| | - Justin M. Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Manuel Ascano
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, United States
| | - John T. Wilson
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, TN, United States
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, United States
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, United States
- Vanderbilt Institute of Chemical Biology, Vanderbilt University Medical Center, Nashville, TN, United States
| |
Collapse
|
3
|
Cheng P, Shen P, Shan Y, Yang Y, Deng R, Chen W, Lu Y, Wei Z. Gut Microbiota-Mediated Modulation of Cancer Progression and Therapy Efficacy. Front Cell Dev Biol 2021; 9:626045. [PMID: 34568308 PMCID: PMC8455814 DOI: 10.3389/fcell.2021.626045] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 08/18/2021] [Indexed: 12/18/2022] Open
Abstract
The role of gut microbiota in the development of various tumors has been a rising topic of public interest, and in recent years, many studies have reported a close relationship between microbial groups and tumor development. Gut microbiota play a role in host metabolism, and the positive and negative alterations of these microbiota have an effect on tumor treatment. The microbiota directly promote, eliminate, and coordinate the efficacy of chemotherapy drugs and the toxicity of adjuvant drugs, and enhance the ability of patients to respond to tumors in adjuvant immunotherapy. In this review, we outline the significance of gut microbiota in tumor development, reveal its impacts on chemotherapy and immunotherapy, and discover various potential mechanisms whereby they influence tumor treatment. This review demonstrates the importance of intestinal microbiota-related research for clinical tumor treatment and provides additional strategy for clinical assistance in cancer treatment.
Collapse
Affiliation(s)
- Peng Cheng
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Peiliang Shen
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Yu Yang
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wenxing Chen
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
4
|
Tian M, Liu W, Zhang Q, Huang Y, Li W, Wang W, Zhao P, Huang S, Song Y, Shereen MA, Qin M, Liu Y, Wu K, Wu J. MYSM1 Represses Innate Immunity and Autoimmunity through Suppressing the cGAS-STING Pathway. Cell Rep 2020; 33:108297. [PMID: 33086059 DOI: 10.1016/j.celrep.2020.108297] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/25/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
The immune system is not only required for preventing threats exerted by pathogens but also essential for developing immune tolerance to avoid tissue damage. This study identifies a distinct mechanism by which MYSM1 suppresses innate immunity and autoimmunity. The expression of MYSM1 is induced upon DNA virus infection and by intracellular DNA stimulation. MYSM1 subsequently interacts with STING and cleaves STING K63-linked ubiquitination to suppress cGAS-STING signaling. Notably, Mysm1-deficient mice exhibit a hyper-inflammatory response, acute tissue damage, and high mortality upon virus infection. Moreover, in the PBMCs of patients with systemic lupus erythematosus (SLE), MYSM1 production decreases, while type I interferons and pro-inflammatory cytokine expressions increase. Importantly, MYSM1 treatment represses the production of IFNs and pro-inflammatory cytokines in the PBMCs of SLE patients. Thus, MYSM1 is a critical repressor of innate immunity and autoimmunity and is thus a potential therapeutic agent for infectious, inflammatory, and autoimmune diseases.
Collapse
Affiliation(s)
- Mingfu Tian
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Weiyong Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Qi Zhang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yuqing Huang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Wen Li
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Wenbiao Wang
- Guangzhou Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China
| | - Peiyi Zhao
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Shanyu Huang
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yunting Song
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Muhammad Adnan Shereen
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Mengying Qin
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Yingle Liu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China
| | - Kailang Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China.
| | - Jianguo Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan 430072, China; Guangzhou Provincial Key Laboratory of Virology, Institute of Medical Microbiology, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
5
|
Kötzner L, Huck B, Garg S, Urbahns K. Small molecules-Giant leaps for immuno-oncology. PROGRESS IN MEDICINAL CHEMISTRY 2020; 59:1-62. [PMID: 32362326 DOI: 10.1016/bs.pmch.2019.11.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Immuno-oncology therapies are revolutionizing the oncology landscape with checkpoint blockade becoming the treatment backbone for many indications. While inspiring, much work remains to increase the number of cancer patients that can benefit from these treatments. Thus, a new era of immuno-oncology research has begun which is focused on identifying novel combination regimes that lead to improved response rates. This review highlights the significance of small molecules in this approach and illustrates the huge progress that has been made to date.
Collapse
Affiliation(s)
- Lisa Kötzner
- Healthcare R&D, Discovery and Development Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Bayard Huck
- Healthcare R&D, Discovery and Development Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Sakshi Garg
- Healthcare R&D, Discovery and Development Technologies, Merck Healthcare KGaA, Darmstadt, Germany
| | - Klaus Urbahns
- Healthcare R&D, Discovery and Development Technologies, Merck Healthcare KGaA, Darmstadt, Germany.
| |
Collapse
|
6
|
Li S, Hong Z, Wang Z, Li F, Mei J, Huang L, Lou X, Zhao S, Song L, Chen W, Wang Q, Liu H, Cai Y, Yu H, Xu H, Zeng G, Wang Q, Zhu J, Liu X, Tan N, Wang C. The Cyclopeptide Astin C Specifically Inhibits the Innate Immune CDN Sensor STING. Cell Rep 2019; 25:3405-3421.e7. [PMID: 30566866 DOI: 10.1016/j.celrep.2018.11.097] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 09/04/2018] [Accepted: 11/28/2018] [Indexed: 12/22/2022] Open
Abstract
cGAS-STING signaling is essential for innate immunity. Its misregulation promotes cancer or autoimmune and autoinflammatory diseases, and it is imperative to identify effective lead compounds that specifically downregulate the pathway. We report here that astin C, a cyclopeptide isolated from the medicinal plant Aster tataricus, inhibits cGAS-STING signaling and the innate inflammatory responses triggered by cytosolic DNAs. Moreover, mice treated with astin C are more susceptible to HSV-1 infection. Consistently, astin C markedly attenuates the autoinflammatory responses in Trex1-/- BMDM cells and in Trex1-/- mouse autoimmune disease model. Mechanistically, astin C specifically blocks the recruitment of IRF3 onto the STING signalosome. Collectively, this study characterizes a STING-specific small-molecular inhibitor that may be applied for potentially manipulating the STING-mediated clinical diseases.
Collapse
Affiliation(s)
- Senlin Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China; State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Ze Hong
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China
| | - Zhe Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Fei Li
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Jiahao Mei
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China
| | - Lulu Huang
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xiwen Lou
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Simeng Zhao
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Lihua Song
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China
| | - Wei Chen
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Qiang Wang
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Heng Liu
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yanni Cai
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huansha Yu
- State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Huimin Xu
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Guangzhi Zeng
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China
| | - Xing Liu
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Ninghua Tan
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China; State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming, 650201, China.
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, School of Traditional Chinese Pharmacy, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, 211198, China; State Key Laboratory of Cell Biology, Innovation Center for Cell Signaling Network, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
7
|
Sharma A, Johnson A. Exosome DNA: Critical regulator of tumor immunity and a diagnostic biomarker. J Cell Physiol 2019; 235:1921-1932. [DOI: 10.1002/jcp.29153] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/26/2019] [Indexed: 12/14/2022]
Affiliation(s)
- Aman Sharma
- ExoCan Healthcare Technologies Ltd, Lab 4 400 NCL Innovation Park Pune India
| | - Abiya Johnson
- ExoCan Healthcare Technologies Ltd, Lab 4 400 NCL Innovation Park Pune India
| |
Collapse
|
8
|
Ralph EC, Hall J. An SPR-based analysis of cGAS substrate K D and steady-state K M values. Methods Enzymol 2019; 625:61-76. [PMID: 31455537 DOI: 10.1016/bs.mie.2019.04.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Surface plasmon resonance (SPR) is a standard method for evaluating direct protein-small molecule binding. While studying the catalytic mechanism of cyclic GMP-AMP synthase (cGAS), we developed an SPR-based method to measure steady-state KM values that complements traditional SPR affinity measurements. The method relies on refractive changes to detect protein interaction with substrates and products, and takes advantage of stimulator of type 1 interferon genes (STING) binding to the cGAS product, 2',3'-cGAMP. The specific method described here uses co-immobilization of cGAS and double-stranded DNA through a biotin tag; it should be generally applicable to other proteins and protein complexes.
Collapse
Affiliation(s)
- Erik C Ralph
- Worldwide Medicinal Chemistry, Pfizer, Groton, CT, United States.
| | - Justin Hall
- Worldwide Medicinal Chemistry, Pfizer, Groton, CT, United States
| |
Collapse
|
9
|
Zhang L, Wei N, Cui Y, Hong Z, Liu X, Wang Q, Li S, Liu H, Yu H, Cai Y, Wang Q, Zhu J, Meng W, Chen Z, Wang C. The deubiquitinase CYLD is a specific checkpoint of the STING antiviral signaling pathway. PLoS Pathog 2018; 14:e1007435. [PMID: 30388174 PMCID: PMC6235404 DOI: 10.1371/journal.ppat.1007435] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 11/14/2018] [Accepted: 10/24/2018] [Indexed: 01/05/2023] Open
Abstract
Stimulator of interferon genes (STING) is critical for cytosolic DNA-triggered innate immunity. STING is modified by several types of polyubiquitin chains. Here, we report that the deubiquitinase CYLD sustains STING signaling by stabilizing the STING protein. CYLD deficiency promoted the K48-linked polyubiquitination and degradation of STING, attenuating the induction of IRF3-responsive genes after HSV-1 infection or the transfection of DNA ligands. Additionally, CYLD knockout mice were more susceptible to HSV-1 infection than their wild-type (WT) littermates. Mechanistically, STING translocated from the ER to the Golgi upon HSV-1 stimulation; CYLD partially accumulated with STING and interacted selectively with K48-linked polyubiquitin chains on STING, specifically removing the K48-linked polyubiquitin chains from STING and ultimately boosting the innate antiviral response. Our study reveals that CYLD is a novel checkpoint in the cGAS-STING signaling pathway and sheds new light on the dynamic regulation of STING activity by ubiquitination.
Collapse
Affiliation(s)
- Lele Zhang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ning Wei
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, China
| | - Ye Cui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Ze Hong
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, China
| | - Xing Liu
- Key Laboratory of Molecular Virology and Immunology, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Senlin Li
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Heng Liu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Huansha Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yanni Cai
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, China
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, China
| | - Wei Meng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, China
| | - Zhengjun Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, Shanghai Tech University, Shanghai, China
- * E-mail: (ZC); (CW)
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, China
- * E-mail: (ZC); (CW)
| |
Collapse
|
10
|
Huck BR, Kötzner L, Urbahns K. Kleine Moleküle, ganz groß: niedermolekulare immunonkologische Kombinationstherapien. Angew Chem Int Ed Engl 2018. [DOI: 10.1002/ange.201707816] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Bayard R. Huck
- Healthcare R&D, Discovery Technologies, Merck KGaA; Frankfurter Straße 250 64293 Darmstadt Deutschland
| | - Lisa Kötzner
- Healthcare R&D, Discovery Technologies, Merck KGaA; Frankfurter Straße 250 64293 Darmstadt Deutschland
| | - Klaus Urbahns
- Healthcare R&D, Discovery Technologies, Merck KGaA; Frankfurter Straße 250 64293 Darmstadt Deutschland
| |
Collapse
|
11
|
Huck BR, Kötzner L, Urbahns K. Small Molecules Drive Big Improvements in Immuno-Oncology Therapies. Angew Chem Int Ed Engl 2018; 57:4412-4428. [PMID: 28971564 PMCID: PMC5900885 DOI: 10.1002/anie.201707816] [Citation(s) in RCA: 108] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Indexed: 12/16/2022]
Abstract
Immuno-oncology therapies have the potential to revolutionize the armamentarium of available cancer treatments. To further improve clinical response rates, researchers are looking for novel combination regimens, with checkpoint blockade being used as a backbone of the treatment. This Review highlights the significance of small molecules in this approach, which holds promise to provide increased benefit to cancer patients.
Collapse
Affiliation(s)
- Bayard R. Huck
- Healthcare R&D, Discovery Technologies, Merck KGaAFrankfurter Strasse 25064293DarmstadtGermany
| | - Lisa Kötzner
- Healthcare R&D, Discovery Technologies, Merck KGaAFrankfurter Strasse 25064293DarmstadtGermany
| | - Klaus Urbahns
- Healthcare R&D, Discovery Technologies, Merck KGaAFrankfurter Strasse 25064293DarmstadtGermany
| |
Collapse
|
12
|
Gaston J, Cheradame L, Yvonnet V, Deas O, Poupon MF, Judde JG, Cairo S, Goffin V. Intracellular STING inactivation sensitizes breast cancer cells to genotoxic agents. Oncotarget 2018; 7:77205-77224. [PMID: 27791205 PMCID: PMC5363581 DOI: 10.18632/oncotarget.12858] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 10/17/2016] [Indexed: 11/25/2022] Open
Abstract
Activation of the IFN/STAT1 pathway is closely associated with drug response and recurrence of breast cancer treated by chemotherapy. The aim of the current study was to elucidate the molecular mechanisms involved upstream and downstream of this pathway in order to identify distinct entities that might be manipulated to improve treatment efficacy. Four breast cancer cell lines (T-47D, MCF7, MDA-MB-231 and HBCx-19 established from the eponymous PDX) were treated in vitro with mafosfamide, a DNA damage inducer. In two of these cell lines (MCF7 and HBCx-19), genotoxic treatment upregulated type I IFN expression leading to paracrine activation of IFN/STAT1 signaling pathway after 6–8 days. We show that STING, a well-characterized inducer of IFN in immune cells, is rapidly triggered in MCF7 cells under genotoxic stress and forms nuclear foci that co-localize with phosphorylated IRF-3 and γH2AX. STING silencing abrogated chemotherapy-induced type I IFN production and signaling and potentiated genotoxic treatment efficacy as it promoted cell death extent and delayed cell colony regrowth. Similar results were obtained after silencing PARP12, one selected gene of the IFN/STAT1 pathway fingerprint. In summary, this study provides the first demonstration of STING activation in breast cancer cells. Our data suggest that genotoxic-induced, STING-mediated type I IFN signaling is a cell-intrinsic mechanism of breast cancer cell survival and regrowth.
Collapse
Affiliation(s)
- Julie Gaston
- Inserm, U1151, Institut Necker Enfants Malades (INEM), University Paris Descartes, Faculty of Medicine, Paris, France.,XenTech, 91000 Evry, France
| | - Laura Cheradame
- Inserm, U1151, Institut Necker Enfants Malades (INEM), University Paris Descartes, Faculty of Medicine, Paris, France.,XenTech, 91000 Evry, France
| | | | | | | | | | - Stefano Cairo
- XenTech, 91000 Evry, France.,LTTA Center, Department of Morphology, Surgery and Experimental Medicine, University of Ferrara, Italy
| | - Vincent Goffin
- Inserm, U1151, Institut Necker Enfants Malades (INEM), University Paris Descartes, Faculty of Medicine, Paris, France
| |
Collapse
|
13
|
Hall J, Ralph EC, Shanker S, Wang H, Byrnes LJ, Horst R, Wong J, Brault A, Dumlao D, Smith JF, Dakin LA, Schmitt DC, Trujillo J, Vincent F, Griffor M, Aulabaugh AE. The catalytic mechanism of cyclic GMP-AMP synthase (cGAS) and implications for innate immunity and inhibition. Protein Sci 2017; 26:2367-2380. [PMID: 28940468 PMCID: PMC5699495 DOI: 10.1002/pro.3304] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 09/12/2017] [Accepted: 09/18/2017] [Indexed: 01/05/2023]
Abstract
Cyclic GMP-AMP synthase (cGAS) is activated by ds-DNA binding to produce the secondary messenger 2',3'-cGAMP. cGAS is an important control point in the innate immune response; dysregulation of the cGAS pathway is linked to autoimmune diseases while targeted stimulation may be of benefit in immunoncology. We report here the structure of cGAS with dinucleotides and small molecule inhibitors, and kinetic studies of the cGAS mechanism. Our structural work supports the understanding of how ds-DNA activates cGAS, suggesting a site for small molecule binders that may cause cGAS activation at physiological ATP concentrations, and an apparent hotspot for inhibitor binding. Mechanistic studies of cGAS provide the first kinetic constants for 2',3'-cGAMP formation, and interestingly, describe a catalytic mechanism where 2',3'-cGAMP may be a minor product of cGAS compared with linear nucleotides.
Collapse
Affiliation(s)
- Justin Hall
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Erik C Ralph
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Suman Shanker
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Hong Wang
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Laura J Byrnes
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Reto Horst
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Jimson Wong
- Hit Discovery and Lead Profiling, Pfizer Centers for Therapeutic Innovation (CTI), Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Amy Brault
- Hit Discovery and Lead Profiling, Pfizer Centers for Therapeutic Innovation (CTI), Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Darren Dumlao
- Hit Discovery and Lead Profiling, Pfizer Centers for Therapeutic Innovation (CTI), Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - James F Smith
- Hit Discovery and Lead Profiling, Pfizer Centers for Therapeutic Innovation (CTI), Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Leslie A Dakin
- Worldwide Medicinal Chemistry, Pfizer, 610 Main St, Cambridge, Massachusetts, 02139
| | - Daniel C Schmitt
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - John Trujillo
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Fabien Vincent
- Hit Discovery and Lead Profiling, Pfizer Centers for Therapeutic Innovation (CTI), Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Matt Griffor
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| | - Ann E Aulabaugh
- Worldwide Medicinal Chemistry, Pfizer, Eastern Point Road, Groton, Connecticut, 06340
| |
Collapse
|
14
|
Hall J, Brault A, Vincent F, Weng S, Wang H, Dumlao D, Aulabaugh A, Aivazian D, Castro D, Chen M, Culp J, Dower K, Gardner J, Hawrylik S, Golenbock D, Hepworth D, Horn M, Jones L, Jones P, Latz E, Li J, Lin LL, Lin W, Lin D, Lovering F, Niljanskul N, Nistler R, Pierce B, Plotnikova O, Schmitt D, Shanker S, Smith J, Snyder W, Subashi T, Trujillo J, Tyminski E, Wang G, Wong J, Lefker B, Dakin L, Leach K. Discovery of PF-06928215 as a high affinity inhibitor of cGAS enabled by a novel fluorescence polarization assay. PLoS One 2017; 12:e0184843. [PMID: 28934246 PMCID: PMC5608272 DOI: 10.1371/journal.pone.0184843] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Accepted: 08/31/2017] [Indexed: 12/30/2022] Open
Abstract
Cyclic GMP-AMP synthase (cGAS) initiates the innate immune system in response to cytosolic dsDNA. After binding and activation from dsDNA, cGAS uses ATP and GTP to synthesize 2', 3' -cGAMP (cGAMP), a cyclic dinucleotide second messenger with mixed 2'-5' and 3'-5' phosphodiester bonds. Inappropriate stimulation of cGAS has been implicated in autoimmune disease such as systemic lupus erythematosus, thus inhibition of cGAS may be of therapeutic benefit in some diseases; however, the size and polarity of the cGAS active site makes it a challenging target for the development of conventional substrate-competitive inhibitors. We report here the development of a high affinity (KD = 200 nM) inhibitor from a low affinity fragment hit with supporting biochemical and structural data showing these molecules bind to the cGAS active site. We also report a new high throughput cGAS fluorescence polarization (FP)-based assay to enable the rapid identification and optimization of cGAS inhibitors. This FP assay uses Cy5-labelled cGAMP in combination with a novel high affinity monoclonal antibody that specifically recognizes cGAMP with no cross reactivity to cAMP, cGMP, ATP, or GTP. Given its role in the innate immune response, cGAS is a promising therapeutic target for autoinflammatory disease. Our results demonstrate its druggability, provide a high affinity tool compound, and establish a high throughput assay for the identification of next generation cGAS inhibitors.
Collapse
Affiliation(s)
- Justin Hall
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Amy Brault
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Fabien Vincent
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Shawn Weng
- Pfizer Centers for Therapeutic Innovation (CTI), Boston, Massachusetts, United States of America
| | - Hong Wang
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Darren Dumlao
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Ann Aulabaugh
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Dikran Aivazian
- Pfizer Centers for Therapeutic Innovation (CTI), San Diego, California, United States of America
| | - Dana Castro
- Pfizer Centers for Therapeutic Innovation (CTI), San Diego, California, United States of America
| | - Ming Chen
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Jeffrey Culp
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Ken Dower
- Inflammation and Immunology, Pfizer, Cambridge, Massachusetts, United States of America
| | - Joseph Gardner
- External Research Solutions, Pfizer, Groton, Connecticut, United States of America
| | - Steven Hawrylik
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Douglas Golenbock
- University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
| | - David Hepworth
- Medicine Design, Pfizer, Cambridge, Massachusetts, United States of America
| | - Mark Horn
- Pfizer Centers for Therapeutic Innovation (CTI), San Diego, California, United States of America
| | - Lyn Jones
- Medicine Design, Pfizer, Cambridge, Massachusetts, United States of America
| | - Peter Jones
- Medicine Design, Pfizer, Cambridge, Massachusetts, United States of America
| | - Eicke Latz
- University of Massachusetts Medical School, Worcester, Massachusetts, United States of America
- Institute of Innate Immunity, University Hospitals Bonn, Bonn, Germany
| | - Jing Li
- Medicine Design, Pfizer, Cambridge, Massachusetts, United States of America
| | - Lih-Ling Lin
- Inflammation and Immunology, Pfizer, Cambridge, Massachusetts, United States of America
| | - Wen Lin
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - David Lin
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Frank Lovering
- Medicine Design, Pfizer, Cambridge, Massachusetts, United States of America
| | | | - Ryan Nistler
- Pfizer Centers for Therapeutic Innovation (CTI), Boston, Massachusetts, United States of America
| | - Betsy Pierce
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Olga Plotnikova
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Daniel Schmitt
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Suman Shanker
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - James Smith
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - William Snyder
- Pfizer Centers for Therapeutic Innovation (CTI), San Diego, California, United States of America
| | - Timothy Subashi
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - John Trujillo
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Edyta Tyminski
- Pfizer Centers for Therapeutic Innovation (CTI), Boston, Massachusetts, United States of America
| | - Guoxing Wang
- Pfizer Centers for Therapeutic Innovation (CTI), Boston, Massachusetts, United States of America
| | - Jimson Wong
- Medicine Design, Pfizer, Groton, Connecticut, United States of America
| | - Bruce Lefker
- Medicine Design, Pfizer, Cambridge, Massachusetts, United States of America
| | - Leslie Dakin
- Medicine Design, Pfizer, Cambridge, Massachusetts, United States of America
| | - Karen Leach
- Pfizer Centers for Therapeutic Innovation (CTI), Boston, Massachusetts, United States of America
| |
Collapse
|
15
|
Wang Q, Huang L, Hong Z, Lv Z, Mao Z, Tang Y, Kong X, Li S, Cui Y, Liu H, Zhang L, Zhang X, Jiang L, Wang C, Zhou Q. The E3 ubiquitin ligase RNF185 facilitates the cGAS-mediated innate immune response. PLoS Pathog 2017; 13:e1006264. [PMID: 28273161 PMCID: PMC5358892 DOI: 10.1371/journal.ppat.1006264] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 03/20/2017] [Accepted: 02/27/2017] [Indexed: 12/22/2022] Open
Abstract
The cyclic GMP-AMP synthase (cGAS), upon cytosolic DNA stimulation, catalyzes the formation of the second messenger 2'3'-cGAMP, which then binds to stimulator of interferon genes (STING) and activates downstream signaling. It remains to be elucidated how the cGAS enzymatic activity is modulated dynamically. Here, we reported that the ER ubiquitin ligase RNF185 interacted with cGAS during HSV-1 infection. Ectopic-expression or knockdown of RNF185 respectively enhanced or impaired the IRF3-responsive gene expression. Mechanistically, RNF185 specifically catalyzed the K27-linked poly-ubiquitination of cGAS, which promoted its enzymatic activity. Additionally, Systemic Lupus Erythematosus (SLE) patients displayed elevated expression of RNF185 mRNA. Collectively, this study uncovers RNF185 as the first E3 ubiquitin ligase of cGAS, shedding light on the regulation of cGAS activity in innate immune responses.
Collapse
Affiliation(s)
- Qiang Wang
- Division of Molecular Nephrology and the Creative Training Center for Undergraduates, the Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Liyuan Huang
- Division of Molecular Nephrology and the Creative Training Center for Undergraduates, the Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Ze Hong
- School of Life Science and Technology, China Pharmaceutical University, Jiangning District, Nanjing, China
| | - Zhongshi Lv
- Division of Molecular Nephrology and the Creative Training Center for Undergraduates, the Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Zhaomin Mao
- Division of Molecular Nephrology and the Creative Training Center for Undergraduates, the Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yijun Tang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiufang Kong
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Senlin Li
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Ye Cui
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Heng Liu
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Lele Zhang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xiaojie Zhang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lindi Jiang
- Department of Rheumatology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Wang
- State Key Laboratory of Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- School of Life Science and Technology, China Pharmaceutical University, Jiangning District, Nanjing, China
| | - Qin Zhou
- Division of Molecular Nephrology and the Creative Training Center for Undergraduates, the Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
16
|
Cui Y, Yu H, Zheng X, Peng R, Wang Q, Zhou Y, Wang R, Wang J, Qu B, Shen N, Guo Q, Liu X, Wang C. SENP7 Potentiates cGAS Activation by Relieving SUMO-Mediated Inhibition of Cytosolic DNA Sensing. PLoS Pathog 2017; 13:e1006156. [PMID: 28095500 PMCID: PMC5271409 DOI: 10.1371/journal.ppat.1006156] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Revised: 01/27/2017] [Accepted: 12/28/2016] [Indexed: 12/13/2022] Open
Abstract
Cyclic GMP-AMP (cGAMP) synthase (cGAS, a.k.a. MB21D1), a cytosolic DNA sensor, catalyzes formation of the second messenger 2’3’-cGAMP that activates the stimulator of interferon genes (STING) signaling. How the cGAS activity is modulated remains largely unknown. Here, we demonstrate that sentrin/SUMO-specific protease 7 (SENP7) interacted with and potentiated cGAS activation. The small ubiquitin-like modifier (SUMO) was conjugated onto the lysine residues 335, 372 and 382 of cGAS, which suppressed its DNA-binding, oligomerization and nucleotidyl-transferase activities. SENP7 reversed this inhibition via catalyzing the cGAS de-SUMOylation. Consistently, silencing of SENP7 markedly impaired the IRF3-responsive gene expression induced by cGAS-STING axis. SENP7-knockdown mice were more susceptible to herpes simplex virus 1 (HSV-1) infection. SENP7 was significantly up-regulated in patients with SLE. Our study highlights the temporal modulation of the cGAS activity via dynamic SUMOylation, uncovering a novel mechanism for fine-tuning the STING signaling in innate immunity. The Cyclic GMP-AMP (cGAMP) synthase (cGAS, a.k.a. MB21D1) is critical for monitoring the pathogen-derived DNA upon microbial infection. Its activity should be dynamically modulated in case the inadvertent recognition of the aberrant self nucleic acids in cytosol leads to severe autoimmune diseases. Protein posttranslational modifications dynamically shape the strength and duration of the immune signaling pathways. It is intriguing to explore whether SUMOylation could modulate the cGAS-initiated signaling. In this study, we characterized sentrin/SUMO-specific protease 7 (SENP7) to specifically potentiate the cGAS activation. Upon microbial DNA challenge, the small ubiquitin-like modifier (SUMO) was conjugated onto cGAS, which suppressed its DNA-binding, oligomerization and nucleotidyl-transferase activities. SENP7 reversed this inhibition via catalyzing the de-SUMOylation of cGAS. Our study sheds new light on the dynamic function of the SUMOylation in cytosolic DNAs-triggered innate immunity response.
Collapse
Affiliation(s)
- Ye Cui
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Huansha Yu
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Xin Zheng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Peng
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Qiang Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Yi Zhou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rui Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Jiehua Wang
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Qu
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Nan Shen
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Qiang Guo
- Department of Rheumatology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xing Liu
- Program in Cellular and Molecular Medicine, Boston Children’s Hospital and Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail: (XL); (CW)
| | - Chen Wang
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Science, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- State Key Laboratory of Natural Medicines and school of Life Science and Technology, China Pharmaceutical University, 639 Longmian Avenue, Jiangning District, Nanjing, China
- * E-mail: (XL); (CW)
| |
Collapse
|
17
|
Gwiazda KS, Grier AE, Sahni J, Burleigh SM, Martin U, Yang JG, Popp NA, Krutein MC, Khan IF, Jacoby K, Jensen MC, Rawlings DJ, Scharenberg AM. High Efficiency CRISPR/Cas9-mediated Gene Editing in Primary Human T-cells Using Mutant Adenoviral E4orf6/E1b55k "Helper" Proteins. Mol Ther 2016; 24:1570-80. [PMID: 27203437 PMCID: PMC5113096 DOI: 10.1038/mt.2016.105] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 04/20/2016] [Indexed: 12/12/2022] Open
Abstract
Many future therapeutic applications of Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)/Cas9 and related RNA-guided nucleases are likely to require their use to promote gene targeting, thus necessitating development of methods that provide for delivery of three components-Cas9, guide RNAs and recombination templates-to primary cells rendered proficient for homology-directed repair. Here, we demonstrate an electroporation/transduction codelivery method that utilizes mRNA to express both Cas9 and mutant adenoviral E4orf6 and E1b55k helper proteins in association with adeno-associated virus (AAV) vectors expressing guide RNAs and recombination templates. By transiently enhancing target cell permissiveness to AAV transduction and gene editing efficiency, this novel approach promotes efficient gene disruption and/or gene targeting at multiple loci in primary human T-cells, illustrating its broad potential for application in translational gene editing.
Collapse
Affiliation(s)
- Kamila S Gwiazda
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Alexandra E Grier
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
| | - Jaya Sahni
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Stephen M Burleigh
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Unja Martin
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Julia G Yang
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Nicholas A Popp
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Michelle C Krutein
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Iram F Khan
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Kyle Jacoby
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
| | - Michael C Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington, USA
| | - David J Rawlings
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Washington, USA
| | - Andrew M Scharenberg
- Program for Cell and Gene Therapy, Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Immunology, University of Washington, Seattle, Washington, USA
- Department of Pediatrics, University of Washington, Washington, USA
| |
Collapse
|
18
|
Weinmann H. Cancer Immunotherapy: Selected Targets and Small-Molecule Modulators. ChemMedChem 2016; 11:450-66. [PMID: 26836578 DOI: 10.1002/cmdc.201500566] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 01/13/2016] [Indexed: 01/01/2023]
Abstract
There is a significant amount of excitement in the scientific community around cancer immunotherapy, as this approach has renewed hope for many cancer patients owing to some recent successes in the clinic. Currently available immuno-oncology therapeutics under clinical development and on the market are mostly biologics (antibodies, proteins, engineered cells, and oncolytic viruses). However, modulation of the immune system with small molecules offers several advantages that may be complementary and potentially synergistic to the use of large biologicals. Therefore, the discovery and development of novel small-molecule modulators is a rapidly growing research area for medicinal chemists working in cancer immunotherapy. This review provides a brief introduction into recent trends related to selected targets and pathways for cancer immunotherapy and their small-molecule pharmacological modulators.
Collapse
Affiliation(s)
- Hilmar Weinmann
- Bayer Pharma AG, Drug Discovery, Medicinal Chemistry Berlin, Muellerstrasse 178, 13353, Berlin, Germany.
| |
Collapse
|