401
|
Hu Z, Yang Y, Fang L, Zhou J, Zhang H. Insight into the dichotomous regulation of STING activation in immunotherapy. Immunopharmacol Immunotoxicol 2021; 43:126-137. [PMID: 33618600 DOI: 10.1080/08923973.2021.1890118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Cyclic GMP-AMP synthase (cGAS)-stimulator of interferon gene (STING) signaling pathway (cGAS-STING) is a hub linking innate immunity and adaptive immunity against pathogen infection by inducing the production of type I interferon (IFN-I). It also plays pivotal roles in modulating tumorigenesis by ensuring the antigen presentation, T cell priming, activation, and tumor regression. Given its antitumor immune properties, cGAS-STING has attracted intense focus and several STING agonists have entered into clinical trials. However, some problems still exist when activating STING for use in oncological indications. It is remarkable that multiple downstream cytokines such as TNF-α, IL-6 may lead to inflammatory disease and even tumor metastasis in practical trials. Besides, there is a synergistic effect when STING agonists are combined with other immunotherapies. In this review, we discussed the advanced understanding between STING and anti-tumor immunity, as well as a variety of promising clinical treatment strategies.
Collapse
Affiliation(s)
- Zhaoxue Hu
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Yifei Yang
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Lincheng Fang
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Jinpei Zhou
- Department of Medicinal Chemistry, China Pharmaceutical University, Nanjing, China
| | - Huibin Zhang
- Center of Drug Discovery, Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
402
|
Quarleri J, Cevallos C, Delpino MV. Apoptosis in infectious diseases as a mechanism of immune evasion and survival. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 125:1-24. [PMID: 33931136 DOI: 10.1016/bs.apcsb.2021.01.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
In pluricellular organisms, apoptosis is indispensable for the development and homeostasis. During infection, apoptosis plays the main role in the elimination of infected cells. Infectious diseases control apoptosis, and this contributes to disease pathogenesis. Increased apoptosis may participate in two different ways. It can assist the dissemination of intracellular pathogens or induce immunosuppression to favor pathogen dissemination. In other conditions, apoptosis can benefit eradicate infectious agents from the host. Accordingly, bacteria, viruses, fungi, and parasites have developed strategies to inhibit host cell death by apoptosis to allow intracellular survival and persistence of the pathogen. The clarification of the intracellular signaling pathways, the receptors involved and the pathogen factors that interfere with apoptosis could disclose new therapeutic targets for blocking microbial actions on apoptotic pathways. In this review, we summarize the current knowledge on pathogen anti-apoptotic and apoptotic approaches and the mechanisms involving in disease.
Collapse
Affiliation(s)
- Jorge Quarleri
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - Cintia Cevallos
- Instituto de Investigaciones Biomédicas en Retrovirus y Sida (INBIRS), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina
| | - María Victoria Delpino
- Instituto de Inmunología, Genética y Metabolismo (INIGEM), Universidad de Buenos Aires, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
403
|
Wang L, Ning S. TRIMming Type I Interferon-Mediated Innate Immune Response in Antiviral and Antitumor Defense. Viruses 2021; 13:279. [PMID: 33670221 PMCID: PMC7916971 DOI: 10.3390/v13020279] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/04/2021] [Accepted: 02/09/2021] [Indexed: 12/17/2022] Open
Abstract
The tripartite motif (TRIM) family comprises at least 80 members in humans, with most having ubiquitin or SUMO E3 ligase activity conferred by their N-terminal RING domain. TRIMs regulate a wide range of processes in ubiquitination- or sumoylation-dependent manners in most cases, and fewer as adaptors. Their roles in the regulation of viral infections, autophagy, cell cycle progression, DNA damage and other stress responses, and carcinogenesis are being increasingly appreciated, and their E3 ligase activities are attractive targets for developing specific immunotherapeutic strategies for immune diseases and cancers. Given their importance in antiviral immune response, viruses have evolved sophisticated immune escape strategies to subvert TRIM-mediated mechanisms. In this review, we focus on their regulation of IFN-I-mediated innate immune response, which plays key roles in antiviral and antitumor defense.
Collapse
Affiliation(s)
- Ling Wang
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA;
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| | - Shunbin Ning
- Department of Internal Medicine, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA;
- Center of Excellence for Inflammation, Infectious Diseases and Immunity, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA
| |
Collapse
|
404
|
Paul BD, Snyder SH, Bohr VA. Signaling by cGAS-STING in Neurodegeneration, Neuroinflammation, and Aging. Trends Neurosci 2021; 44:83-96. [PMID: 33187730 PMCID: PMC8662531 DOI: 10.1016/j.tins.2020.10.008] [Citation(s) in RCA: 180] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/22/2020] [Accepted: 10/15/2020] [Indexed: 12/11/2022]
Abstract
Recognition of foreign or misplaced nucleic acids is one of the principal modes by which the immune system detects pathogenic entities. When cytosolic DNA is sensed, a signal is relayed via the cGAS-STING pathway: this involves the activation of cyclic GMP-AMP (cGMP-AMP) synthase (cGAS) and generation of the cyclic dinucleotide cGAMP, followed by the induction of stimulator of interferon genes (STING). The cGAS-STING pathway responds to viral, bacterial, and self-DNA. Whereas it generally mediates immune surveillance and is often neuroprotective, excessive engagement of the system can be deleterious. This is relevant in aging and age-related neurological diseases, where neuroinflammation contributes to disease progression. This review focuses on cGAS-STING signaling in aging, neurodegeneration, and neuroinflammation, and on therapeutic implications.
Collapse
Affiliation(s)
- Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Vilhelm A Bohr
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
405
|
Hu M, Zhou M, Bao X, Pan D, Jiao M, Liu X, Li F, Li CY. ATM inhibition enhances cancer immunotherapy by promoting mtDNA leakage and cGAS/STING activation. J Clin Invest 2021; 131:139333. [PMID: 33290271 PMCID: PMC7843232 DOI: 10.1172/jci139333] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Accepted: 11/25/2020] [Indexed: 01/10/2023] Open
Abstract
Novel approaches are needed to boost the efficacy of immune checkpoint blockade (ICB) therapy. Ataxia telangiectasia mutated (ATM) protein plays a central role in sensing DNA double-stranded breaks (DSBs) and coordinating their repair. Recent data indicated that ATM might be a promising target to enhance ICB therapy. However, the molecular mechanism involved has not been clearly elucidated. Here, we show that ATM inhibition could potentiate ICB therapy by promoting cytoplasmic leakage of mitochondrial DNA (mtDNA) and activation of the cGAS/STING pathway. We show that genetic depletion of ATM in murine cancer cells delayed tumor growth in syngeneic mouse hosts in a T cell-dependent manner. Furthermore, chemical inhibition of ATM potentiated anti-PD-1 therapy of mouse tumors. ATM inhibition potently activated the cGAS/STING pathway and enhanced lymphocyte infiltration into the tumor microenvironment by downregulating mitochondrial transcription factor A (TFAM), which led to mtDNA leakage into the cytoplasm. Moreover, our analysis of data from a large patient cohort indicated that ATM mutations, especially nonsense mutations, predicted for clinical benefits of ICB therapy. Our study therefore provides strong evidence that ATM may serve as both a therapeutic target and a biomarker to enable ICB therapy.
Collapse
Affiliation(s)
- Mengjie Hu
- Department of Dermatology, Duke University Medical Center, Durham, North Caronina, USA
| | - Min Zhou
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Shanghai Jiaotong University, Shanghai, China
| | - Xuhui Bao
- Department of Dermatology, Duke University Medical Center, Durham, North Caronina, USA
| | - Dong Pan
- Department of Dermatology, Duke University Medical Center, Durham, North Caronina, USA
| | - Meng Jiao
- Department of Dermatology, Duke University Medical Center, Durham, North Caronina, USA
| | - Xinjian Liu
- Department of Biochemistry, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Fang Li
- Department of Dermatology, Duke University Medical Center, Durham, North Caronina, USA
| | - Chuan-Yuan Li
- Department of Dermatology, Duke University Medical Center, Durham, North Caronina, USA
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, North Carolina, USA
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
406
|
Sharma J, Parsai K, Raghuwanshi P, Ali SA, Tiwari V, Bhargava A, Mishra PK. Emerging role of mitochondria in airborne particulate matter-induced immunotoxicity. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 270:116242. [PMID: 33321436 DOI: 10.1016/j.envpol.2020.116242] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/23/2020] [Accepted: 12/06/2020] [Indexed: 05/05/2023]
Abstract
The immune system is one of the primary targets of airborne particulate matter. Recent evidence suggests that mitochondria lie at the center of particulate matter-induced immunotoxicity. Particulate matter can directly interact with mitochondrial components (proteins, lipids, and nucleic acids) and impairs the vital mitochondrial processes including redox mechanisms, fusion-fission, autophagy, and metabolic pathways. These disturbances impede different mitochondrial functions including ATP production, which acts as an important platform to regulate immunity and inflammatory responses. Moreover, the mitochondrial DNA released into the cytosol or in the extracellular milieu acts as a danger-associated molecular pattern and triggers the signaling pathways, involving cGAS-STING, TLR9, and NLRP3. In the present review, we discuss the emerging role of mitochondria in airborne particulate matter-induced immunotoxicity and its myriad biological consequences in health and disease.
Collapse
Affiliation(s)
- Jahnavi Sharma
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Kamakshi Parsai
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Pragati Raghuwanshi
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Sophiya Anjum Ali
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Vineeta Tiwari
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Arpit Bhargava
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India
| | - Pradyumna Kumar Mishra
- Department of Molecular Biology, ICMR-National Institute for Research in Environmental Health, Bhopal, India.
| |
Collapse
|
407
|
Okude H, Ori D, Kawai T. Signaling Through Nucleic Acid Sensors and Their Roles in Inflammatory Diseases. Front Immunol 2021; 11:625833. [PMID: 33633744 PMCID: PMC7902034 DOI: 10.3389/fimmu.2020.625833] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/14/2020] [Indexed: 12/14/2022] Open
Abstract
Recognition of pathogen-derived nucleic acids by pattern-recognition receptors (PRRs) is essential for eliciting antiviral immune responses by inducing the production of type I interferons (IFNs) and proinflammatory cytokines. Such responses are a prerequisite for mounting innate and pathogen-specific adaptive immune responses. However, host cells also use nucleic acids as carriers of genetic information, and the aberrant recognition of self-nucleic acids by PRRs is associated with the onset of autoimmune or autoinflammatory diseases. In this review, we describe the mechanisms of nucleic acid sensing by PRRs, including Toll-like receptors, RIG-I-like receptors, and DNA sensor molecules, and their signaling pathways as well as the disorders caused by uncontrolled or unnecessary activation of these PRRs.
Collapse
Affiliation(s)
- Haruna Okude
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Daisuke Ori
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and Technology, Nara Institute of Science and Technology (NAIST), Ikoma, Japan
| |
Collapse
|
408
|
Smith JA. STING, the Endoplasmic Reticulum, and Mitochondria: Is Three a Crowd or a Conversation? Front Immunol 2021; 11:611347. [PMID: 33552072 PMCID: PMC7858662 DOI: 10.3389/fimmu.2020.611347] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 12/04/2020] [Indexed: 12/20/2022] Open
Abstract
The anti-viral pattern recognition receptor STING and its partnering cytosolic DNA sensor cGAS have been increasingly recognized to respond to self DNA in multiple pathologic settings including cancer and autoimmune disease. Endogenous DNA sources that trigger STING include damaged nuclear DNA in micronuclei and mitochondrial DNA (mtDNA). STING resides in the endoplasmic reticulum (ER), and particularly in the ER-mitochondria associated membranes. This unique location renders STING well poised to respond to intracellular organelle stress. Whereas the pathways linking mtDNA and STING have been addressed recently, the mechanisms governing ER stress and STING interaction remain more opaque. The ER and mitochondria share a close anatomic and functional relationship, with mutual production of, and inter-organelle communication via calcium and reactive oxygen species (ROS). This interdependent relationship has potential to both generate the essential ligands for STING activation and to regulate its activity. Herein, we review the interactions between STING and mitochondria, STING and ER, ER and mitochondria (vis-à-vis calcium and ROS), and the evidence for 3-way communication.
Collapse
Affiliation(s)
- Judith A Smith
- Department of Pediatrics and Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
409
|
Choi I, Son H, Baek JH. Tricarboxylic Acid (TCA) Cycle Intermediates: Regulators of Immune Responses. Life (Basel) 2021; 11:69. [PMID: 33477822 PMCID: PMC7832849 DOI: 10.3390/life11010069] [Citation(s) in RCA: 111] [Impact Index Per Article: 27.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/06/2023] Open
Abstract
The tricarboxylic acid cycle (TCA) is a series of chemical reactions used in aerobic organisms to generate energy via the oxidation of acetylcoenzyme A (CoA) derived from carbohydrates, fatty acids and proteins. In the eukaryotic system, the TCA cycle occurs completely in mitochondria, while the intermediates of the TCA cycle are retained inside mitochondria due to their polarity and hydrophilicity. Under cell stress conditions, mitochondria can become disrupted and release their contents, which act as danger signals in the cytosol. Of note, the TCA cycle intermediates may also leak from dysfunctioning mitochondria and regulate cellular processes. Increasing evidence shows that the metabolites of the TCA cycle are substantially involved in the regulation of immune responses. In this review, we aimed to provide a comprehensive systematic overview of the molecular mechanisms of each TCA cycle intermediate that may play key roles in regulating cellular immunity in cell stress and discuss its implication for immune activation and suppression.
Collapse
Affiliation(s)
| | | | - Jea-Hyun Baek
- School of Life Science, Handong Global University, Pohang, Gyeongbuk 37554, Korea; (I.C.); (H.S.)
| |
Collapse
|
410
|
Koren E, Fuchs Y. Modes of Regulated Cell Death in Cancer. Cancer Discov 2021; 11:245-265. [PMID: 33462123 DOI: 10.1158/2159-8290.cd-20-0789] [Citation(s) in RCA: 240] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 09/15/2020] [Accepted: 10/29/2020] [Indexed: 11/16/2022]
Abstract
Cell suicide pathways, termed regulated cell death (RCD), play a critical role in organismal development, homeostasis, and pathogenesis. Here, we provide an overview of key RCD modalities, namely apoptosis, entosis, necroptosis, pyroptosis, and ferroptosis. We explore how various RCD modules serve as a defense mechanism against the emergence of cancer as well as the manner in which they can be exploited to drive oncogenesis. Furthermore, we outline current therapeutic agents that activate RCD and consider novel RCD-based strategies for tumor elimination. SIGNIFICANCE: A variety of antitumor therapeutics eliminate cancer cells by harnessing the devastating potential of cellular suicide pathways, emphasizing the critical importance of RCD in battling cancer. This review supplies a mechanistic perspective of distinct RCD modalities and explores the important role they play in tumorigenesis. We discuss how RCD modules serve as a double-edged sword as well as novel approaches aimed at selectively manipulating RCD for tumor eradication.
Collapse
Affiliation(s)
- Elle Koren
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel. Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, Israel
| | - Yaron Fuchs
- Laboratory of Stem Cell Biology and Regenerative Medicine, Department of Biology, Technion Israel Institute of Technology, Haifa, Israel. Lorry Lokey Interdisciplinary Center for Life Sciences and Engineering, Technion Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
411
|
Vashi N, Bakhoum SF. The Evolution of STING Signaling and Its Involvement in Cancer. Trends Biochem Sci 2021; 46:446-460. [PMID: 33461879 DOI: 10.1016/j.tibs.2020.12.010] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 12/04/2020] [Accepted: 12/17/2020] [Indexed: 12/14/2022]
Abstract
The cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway has been primarily characterized as an inflammatory mechanism in higher eukaryotes in response to cytosolic double-stranded DNA (dsDNA). Since its initial discovery, detailed mechanisms delineating the dynamic subcellular localization of its different components and downstream signaling have been uncovered, leading to attempts to harness its proinflammatory properties for therapeutic benefit in cancer. Emerging evidence, however, indicates that a crucial primordial function of STING is to promote autophagy, and that downstream interferon (IFN) signaling emerged recently in its evolutionary history. Furthermore, studies suggest that this pathway is a crucial regulator of cellular metabolism that potentially couples inflammation to nutrient availability. We focus on the evolutionarily conserved functions of STING, and we discuss how a broader understanding of this pathway can help us to better appreciate its potential role in cancer and harness it for therapeutic benefit.
Collapse
Affiliation(s)
- Nimi Vashi
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Samuel F Bakhoum
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
412
|
Lu RO, Ho WS. Mitochondrial Dysfunction, Macrophage, and Microglia in Brain Cancer. Front Cell Dev Biol 2021; 8:620788. [PMID: 33520994 PMCID: PMC7843701 DOI: 10.3389/fcell.2020.620788] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/10/2020] [Indexed: 12/02/2022] Open
Abstract
Glioblastoma (GBM) is the most common malignant brain cancer. Increasing evidence suggests that mitochondrial dysfunction plays a key role in GBM progression as mitochondria is essential in regulating cell metabolism, oxidative stress, and cell death. Meanwhile, the immune microenvironment in GBM is predominated by tumor-associated macrophages and microglia (TAM), which is a heterogenous population of myeloid cells that, in general, create an immunosuppressive milieu to support tumor growth. However, subsets of TAMs can be pro-inflammatory and thereby antitumor. Therapeutic strategies targeting TAMs are increasingly explored as novel treatment strategies for GBM. The connection between mitochondrial dysfunction and TAMs phenotype in the tumor microenvironment is unclear. This review aims to provide perspectives and discuss possible molecular mechanisms mediating the interplay between glioma mitochondrial dysfunction and TAMs phenotype in shaping tumor immune microenvironment.
Collapse
Affiliation(s)
- Rongze Olivia Lu
- Department of Neurosurgery, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Mulva Clinic for the Neurosciences, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| | - Winson S. Ho
- Department of Neurosurgery, Dell Medical School, University of Texas at Austin, Austin, TX, United States
- Mulva Clinic for the Neurosciences, Dell Medical School, University of Texas at Austin, Austin, TX, United States
| |
Collapse
|
413
|
Rosenbaum SR, Wilski NA, Aplin AE. Fueling the Fire: Inflammatory Forms of Cell Death and Implications for Cancer Immunotherapy. Cancer Discov 2021; 11:266-281. [PMID: 33451983 DOI: 10.1158/2159-8290.cd-20-0805] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 09/04/2020] [Accepted: 10/01/2020] [Indexed: 11/16/2022]
Abstract
Unleashing the immune system with immune checkpoint inhibitors (ICI) has significantly improved overall survival for subsets of patients with stage III/IV cancer. However, many tumors are nonresponsive to ICIs, in part due to a lack of tumor-infiltrating lymphocytes (TIL). Converting these immune "cold" tumors to "hot" tumors that are thus more likely to respond to ICIs is a major obstacle for cancer treatment. Triggering inflammatory forms of cell death, such as necroptosis and pyroptosis, may alter the tumor immune microenvironment and the influx of TILs. We present an emerging view that promoting tumor-localized necroptosis and pyroptosis may ultimately enhance responses to ICI. SIGNIFICANCE: Many tumor types respond poorly to ICIs or respond but subsequently acquire resistance. Effective therapies for ICI-nonresponsive tumors are lacking and should be guided by evidence from preclinical studies. Promoting inflammatory cell death mechanisms within the tumor may alter the local immune microenvironment toward an ICI-responsive state.
Collapse
Affiliation(s)
- Sheera R Rosenbaum
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Nicole A Wilski
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, Pennsylvania. .,Sidney Kimmel Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
414
|
Vitale I, Yamazaki T, Wennerberg E, Sveinbjørnsson B, Rekdal Ø, Demaria S, Galluzzi L. Targeting Cancer Heterogeneity with Immune Responses Driven by Oncolytic Peptides. Trends Cancer 2021; 7:557-572. [PMID: 33446447 DOI: 10.1016/j.trecan.2020.12.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/09/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
Accumulating preclinical and clinical evidence indicates that high degrees of heterogeneity among malignant cells constitute a considerable obstacle to the success of cancer therapy. This calls for the development of approaches that operate - or enable established treatments to operate - despite such intratumoral heterogeneity (ITH). In this context, oncolytic peptides stand out as promising therapeutic tools based on their ability to drive immunogenic cell death associated with robust anticancer immune responses independently of ITH. We review the main molecular and immunological pathways engaged by oncolytic peptides, and discuss potential approaches to combine these agents with modern immunotherapeutics in support of superior tumor-targeting immunity and efficacy in patients with cancer.
Collapse
Affiliation(s)
- Ilio Vitale
- Italian Institute for Genomic Medicine (IIGM), Istituto Di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, Fondazione del Piemonte per l'Oncologia (FPO)-IRCCS, Candiolo, Italy
| | - Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Erik Wennerberg
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, London, UK
| | - Baldur Sveinbjørnsson
- Lytix Biopharma, Oslo, Norway; Department of Medical Biology, University of Tromsø, Tromsø, Norway; Childhood Cancer Research Unit, Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Øystein Rekdal
- Lytix Biopharma, Oslo, Norway; Department of Medical Biology, University of Tromsø, Tromsø, Norway
| | - Sandra Demaria
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA; Department of Dermatology, Yale School of Medicine, New Haven, CT, USA; Université de Paris, Paris, France.
| |
Collapse
|
415
|
Schmidt MF, Gan ZY, Komander D, Dewson G. Ubiquitin signalling in neurodegeneration: mechanisms and therapeutic opportunities. Cell Death Differ 2021; 28:570-590. [PMID: 33414510 PMCID: PMC7862249 DOI: 10.1038/s41418-020-00706-7] [Citation(s) in RCA: 221] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Revised: 12/01/2020] [Accepted: 12/01/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases are characterised by progressive damage to the nervous system including the selective loss of vulnerable populations of neurons leading to motor symptoms and cognitive decline. Despite millions of people being affected worldwide, there are still no drugs that block the neurodegenerative process to stop or slow disease progression. Neuronal death in these diseases is often linked to the misfolded proteins that aggregate within the brain (proteinopathies) as a result of disease-related gene mutations or abnormal protein homoeostasis. There are two major degradation pathways to rid a cell of unwanted or misfolded proteins to prevent their accumulation and to maintain the health of a cell: the ubiquitin–proteasome system and the autophagy–lysosomal pathway. Both of these degradative pathways depend on the modification of targets with ubiquitin. Aging is the primary risk factor of most neurodegenerative diseases including Alzheimer’s disease, Parkinson’s disease and amyotrophic lateral sclerosis. With aging there is a general reduction in proteasomal degradation and autophagy, and a consequent increase of potentially neurotoxic protein aggregates of β-amyloid, tau, α-synuclein, SOD1 and TDP-43. An often over-looked yet major component of these aggregates is ubiquitin, implicating these protein aggregates as either an adaptive response to toxic misfolded proteins or as evidence of dysregulated ubiquitin-mediated degradation driving toxic aggregation. In addition, non-degradative ubiquitin signalling is critical for homoeostatic mechanisms fundamental for neuronal function and survival, including mitochondrial homoeostasis, receptor trafficking and DNA damage responses, whilst also playing a role in inflammatory processes. This review will discuss the current understanding of the role of ubiquitin-dependent processes in the progressive loss of neurons and the emergence of ubiquitin signalling as a target for the development of much needed new drugs to treat neurodegenerative disease. ![]()
Collapse
Affiliation(s)
- Marlene F Schmidt
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, 3052, Australia
| | - Zhong Yan Gan
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, 3052, Australia
| | - David Komander
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, VIC, 3052, Australia.,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, 3052, Australia
| | - Grant Dewson
- The Walter and Eliza Hall Institute of Medical Research, 1G Royal Parade, Melbourne, VIC, 3052, Australia. .,Department of Medical Biology, University of Melbourne, Royal Parade, Melbourne, VIC, 3052, Australia.
| |
Collapse
|
416
|
Bahat A, MacVicar T, Langer T. Metabolism and Innate Immunity Meet at the Mitochondria. Front Cell Dev Biol 2021; 9:720490. [PMID: 34386501 PMCID: PMC8353256 DOI: 10.3389/fcell.2021.720490] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/05/2021] [Indexed: 12/14/2022] Open
Abstract
Mitochondria are master regulators of metabolism and have emerged as key signalling organelles of the innate immune system. Each mitochondrion harbours potent agonists of inflammation, including mitochondrial DNA (mtDNA), which are normally shielded from the rest of the cell and extracellular environment and therefore do not elicit detrimental inflammatory cascades. Mitochondrial damage and dysfunction can lead to the cytosolic and extracellular exposure of mtDNA, which triggers inflammation in a number of diseases including autoimmune neurodegenerative disorders. However, recent research has revealed that the extra-mitochondrial exposure of mtDNA is not solely a negative consequence of mitochondrial damage and pointed to an active role of mitochondria in innate immunity. Metabolic cues including nucleotide imbalance can stimulate the release of mtDNA from mitochondria in order to drive a type I interferon response. Moreover, important effectors of the innate immune response to pathogen infection, such as the mitochondrial antiviral signalling protein (MAVS), are located at the mitochondrial surface and modulated by the cellular metabolic status and mitochondrial dynamics. In this review, we explore how and why metabolism and innate immunity converge at the mitochondria and describe how mitochondria orchestrate innate immune signalling pathways in different metabolic scenarios. Understanding how cellular metabolism and metabolic programming of mitochondria are translated into innate immune responses bears relevance to a broad range of human diseases including cancer.
Collapse
Affiliation(s)
- Amir Bahat
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Thomas MacVicar
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Thomas Langer
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- *Correspondence: Thomas Langer,
| |
Collapse
|
417
|
Abstract
The cGAS-STING signalling pathway has emerged as a key mediator of inflammation in the settings of infection, cellular stress and tissue damage. Underlying this broad involvement of the cGAS-STING pathway is its capacity to sense and regulate the cellular response towards microbial and host-derived DNAs, which serve as ubiquitous danger-associated molecules. Insights into the structural and molecular biology of the cGAS-STING pathway have enabled the development of selective small-molecule inhibitors with the potential to target the cGAS-STING axis in a number of inflammatory diseases in humans. Here, we outline the principal elements of the cGAS-STING signalling cascade and discuss the general mechanisms underlying the association of cGAS-STING activity with various autoinflammatory, autoimmune and degenerative diseases. Finally, we outline the chemical nature of recently developed cGAS and STING antagonists and summarize their potential clinical applications.
Collapse
|
418
|
Cheng A, Kawahata I, Fukunaga K. Fatty Acid Binding Protein 5 Mediates Cell Death by Psychosine Exposure through Mitochondrial Macropores Formation in Oligodendrocytes. Biomedicines 2020; 8:biomedicines8120635. [PMID: 33419250 PMCID: PMC7766880 DOI: 10.3390/biomedicines8120635] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/08/2020] [Accepted: 12/17/2020] [Indexed: 12/21/2022] Open
Abstract
Oligodendrocytes, the myelinating cells in the central nervous system (CNS), are critical for producing myelin throughout the CNS. The loss of oligodendrocytes is associated with multiple neurodegenerative disorders mediated by psychosine. However, the involvement of psychosine in the critical biochemical pathogenetic mechanism of the loss of oligodendrocytes and myelin in krabbe disease (KD) remains unclear. Here, we addressed how oligodendrocytes are induced by psychosine treatment in both KG-1C human oligodendroglial cells and mouse oligodendrocyte precursor cells. We found that fatty acid binding protein 5 (FABP5) expressed in oligodendrocytes accelerates mitochondria-induced glial death by inducing mitochondrial macropore formation through voltage-dependent anion channels (VDAC-1) and BAX. These two proteins mediate mitochondrial outer membrane permeabilization, thereby leading to the release of mitochondrial DNA and cytochrome C into the cytosol, and the activation of apoptotic caspases. Furthermore, we confirmed that the inhibition of FABP5 functions by shRNA and FABP5-specific ligands blocking mitochondrial macropore formation, thereby rescuing psychosine-induced oligodendrocyte death. Taken together, we identified FABP5 as a critical factor in mitochondrial injury associated with psychosine-induced apoptosis in oligodendrocytes.
Collapse
|
419
|
Liu Y, Lu X, Qin N, Qiao Y, Xing S, Liu W, Feng F, Liu Z, Sun H. STING, a promising target for small molecular immune modulator: A review. Eur J Med Chem 2020; 211:113113. [PMID: 33360799 DOI: 10.1016/j.ejmech.2020.113113] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 12/19/2022]
Abstract
Stimulator of interferon genes (STING) plays a crucial role in human innate immune system, which is gradually concerned following the emerging immunotherapy. Activated STING induces the production of type I interferons (IFNs) and proinflammatory cytokines through STING-TBK1-IRF3/NF-κB pathway, which could be applied into the treatment of infection, inflammation, and tumorigenesis. Here, we provided a detailed summary of STING from its structure, function and regulation. Especially, we illustrated the canonical or noncanonical cyclic dinucleotides (CDNs) and synthetic small molecules for STING activation or inhibition and their efficacy in related diseases. Importantly, we particularly emphasized the discovery, development and modification of STING agonist or antagonist, attempting to enlighten reader's mind for enriching small molecular modulator of STING. In addition, we summarized biological evaluation methods for the assessment of small molecules activity.
Collapse
Affiliation(s)
- Yijun Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Xin Lu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Nan Qin
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Yuting Qiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Shuaishuai Xing
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Wenyuan Liu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China
| | - Feng Feng
- Department of Natural Medicinal Chemistry, China Pharmaceutical University, Nanjing 211198, People's Republic of China; Jiangsu Food and Pharmaceuticals Science College, Institute of Food and Pharmaceuticals Research, 223005, People's Republic of China
| | - Zongliang Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, PR China.
| | - Haopeng Sun
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, People's Republic of China.
| |
Collapse
|
420
|
Abstract
Immunogenic cell death (ICD) is a type of cancer cell death triggered by certain chemotherapeutic drugs, oncolytic viruses, physicochemical therapies, photodynamic therapy, and radiotherapy. It involves the activation of the immune system against cancer in immunocompetent hosts. ICD comprises the release of damage-associated molecular patterns (DAMPs) from dying tumor cells that result in the activation of tumor-specific immune responses, thus eliciting long-term efficacy of anticancer drugs by combining direct cancer cell killing and antitumor immunity. Remarkably, subcutaneous injection of dying tumor cells undergoing ICD has been shown to provoke anticancer vaccine effects in vivo. DAMPs include the cell surface exposure of calreticulin (CRT) and heat-shock proteins (HSP70 and HSP90), extracellular release of adenosine triphosphate (ATP), high-mobility group box-1 (HMGB1), type I IFNs and members of the IL-1 cytokine family. In this review, we discuss the cell death modalities connected to ICD, the DAMPs exposed during ICD, and the mechanism by which they activate the immune system. Finally, we discuss the therapeutic potential and challenges of harnessing ICD in cancer immunotherapy.
Collapse
Affiliation(s)
- Asma Ahmed
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowUK
| | - Stephen W.G. Tait
- Cancer Research UK Beatson InstituteGlasgowUK
- Institute of Cancer SciencesUniversity of GlasgowUK
| |
Collapse
|
421
|
Imre G. Cell death signalling in virus infection. Cell Signal 2020; 76:109772. [PMID: 32931899 PMCID: PMC7486881 DOI: 10.1016/j.cellsig.2020.109772] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 09/01/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023]
Abstract
Apoptosis, necroptosis and pyroptosis represent three major regulated cell death modalities. Apoptosis features cell shrinkage, nuclear fragmentation and cytoplasm-blebbing. Necroptosis and pyroptosis exhibit osmotic imbalances in the cell accompanied by early membrane ruptures, which morphologically resembles necrosis. Importantly, these two lytic cell death forms facilitate the release of damage associated molecular patterns into the extracellular space leading to inflammatory response. Whereas, during apoptosis, the membrane integrity is preserved and the apoptotic cell is removed by neighbouring cells ensuring the avoidance of immune-stimulation. Viruses comprise a versatile group of intracellular pathogens, which elicit various strategies to infect and to propagate. Viruses are recognized by a myriad of pathogen recognition receptors in the human cells, which consequently lead to activation of the immune system and in certain circumstances cell-autonomous cell death. Importantly, the long-standing view that a cell death inducing capacity of a virus is equal to its pathogenic potential seems to be only partially valid. The altruistic cell death of an infected cell may serve the whole organism by ultimately curbing the way of virus manufacturing. In fact, several viruses express "anti-cell death" proteins to avoid this viral-defence mechanism. Conversely, some viruses hijack cell death pathways to selectively destroy cell populations in order to compromise the immune system of the host. This review discusses the pros and cons of virus induced cell death from the perspective of the host cells and attempts to provide a comprehensive overview of the complex network of cell death signalling in virus infection.
Collapse
Affiliation(s)
- Gergely Imre
- Institute of General Pharmacology and Toxicology, University Hospital Frankfurt, Goethe University Frankfurt, Frankfurt am Main 60590, Germany.
| |
Collapse
|
422
|
Liao W, Du C, Wang J. The cGAS-STING Pathway in Hematopoiesis and Its Physiopathological Significance. Front Immunol 2020; 11:573915. [PMID: 33329537 PMCID: PMC7734179 DOI: 10.3389/fimmu.2020.573915] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 10/29/2020] [Indexed: 01/09/2023] Open
Abstract
Cytosolic DNA sensing is a fundamental mechanism by which organisms handle various stresses, including infection and genotoxicity. The hematopoietic system is sensitive to stresses, and hematopoietic changes are often rapid and the first response to stresses. Based on the transcriptome database, cytosolic DNA sensing pathways are widely expressed in the hematopoietic system, and components of these pathways may be expressed at even higher levels in hematopoietic stem and progenitor cells (HSPCs) than in their certain progeny immune cells. Recent studies have described a previously unrecognized role for cytosolic DNA sensing pathways in the regulation of hematopoiesis under both homeostatic and stress conditions. In particular, the recently discovered cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway is a critical modulator of hematopoiesis. Perturbation of the cGAS-STING pathway in HSPCs may be involved in the pathogenesis of hematopoietic disorders, autoimmune diseases, and inflammation-related diseases and may be candidate therapeutic targets. In this review, we focus on the recent findings of the cGAS-STING pathway in the regulation of hematopoiesis, and its physiopathological significance including its implications in diseases and therapeutic potential.
Collapse
Affiliation(s)
- Weinian Liao
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Changhong Du
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| | - Junping Wang
- State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
423
|
Song Y, Zhou Y, Zhou X. The role of mitophagy in innate immune responses triggered by mitochondrial stress. Cell Commun Signal 2020; 18:186. [PMID: 33239048 PMCID: PMC7687798 DOI: 10.1186/s12964-020-00659-x] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 09/06/2020] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are important cellular organelles involved in many different functions, from energy generation and fatty acid oxidation to cell death regulation and immune responses. Accumulating evidence indicates that mitochondrial stress acts as a key trigger of innate immune responses. Critically, the dysfunctional mitochondria can be selectively eliminated by mitophagy. The elimination of dysfunctional mitochondria may function as an effective way employed by mitophagy to keep the immune system in check. In addition, mitophagy can be utilized by pathogens for immune evasion. In this review, we summarize how mitochondrial stress triggers innate immune responses and the roles of mitophagy in innate immunity and in infection, as well as the molecular mechanisms of mitophagy. Video Abstract.
Collapse
Affiliation(s)
- Yinjuan Song
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yang Zhou
- College of Animal Science, Southwest University, Chongqing, 402460, China.,Immunology Research Center, Medical Research Institute, Southwest University, Chongqing, China
| | - Xiangmei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
424
|
Storozynsky Q, Hitt MM. The Impact of Radiation-Induced DNA Damage on cGAS-STING-Mediated Immune Responses to Cancer. Int J Mol Sci 2020; 21:E8877. [PMID: 33238631 PMCID: PMC7700321 DOI: 10.3390/ijms21228877] [Citation(s) in RCA: 128] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/19/2022] Open
Abstract
Radiotherapy is a major modality used to combat a wide range of cancers. Classical radiobiology principles categorize ionizing radiation (IR) as a direct cytocidal therapeutic agent against cancer; however, there is an emerging appreciation for additional antitumor immune responses generated by this modality. A more nuanced understanding of the immunological pathways induced by radiation could inform optimal therapeutic combinations to harness radiation-induced antitumor immunity and improve treatment outcomes of cancers refractory to current radiotherapy regimens. Here, we summarize how radiation-induced DNA damage leads to the activation of a cytosolic DNA sensing pathway mediated by cyclic GMP-AMP (cGAMP) synthase (cGAS) and stimulator of interferon genes (STING). The activation of cGAS-STING initiates innate immune signaling that facilitates adaptive immune responses to destroy cancer. In this way, cGAS-STING signaling bridges the DNA damaging capacity of IR with the activation of CD8+ cytotoxic T cell-mediated destruction of cancer-highlighting a molecular pathway radiotherapy can exploit to induce antitumor immune responses. In the context of radiotherapy, we further report on factors that enhance or inhibit cGAS-STING signaling, deleterious effects associated with cGAS-STING activation, and promising therapeutic candidates being investigated in combination with IR to bolster immune activation through engaging STING-signaling. A clearer understanding of how IR activates cGAS-STING signaling will inform immune-based treatment strategies to maximize the antitumor efficacy of radiotherapy, improving therapeutic outcomes.
Collapse
Affiliation(s)
| | - Mary M. Hitt
- Department of Oncology, University of Alberta, Edmonton, AB T6G 2E1, Canada;
| |
Collapse
|
425
|
Adjemian S, Oltean T, Martens S, Wiernicki B, Goossens V, Vanden Berghe T, Cappe B, Ladik M, Riquet FB, Heyndrickx L, Bridelance J, Vuylsteke M, Vandecasteele K, Vandenabeele P. Ionizing radiation results in a mixture of cellular outcomes including mitotic catastrophe, senescence, methuosis, and iron-dependent cell death. Cell Death Dis 2020; 11:1003. [PMID: 33230108 PMCID: PMC7684309 DOI: 10.1038/s41419-020-03209-y] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 11/02/2020] [Accepted: 11/03/2020] [Indexed: 12/21/2022]
Abstract
Radiotherapy is commonly used as a cytotoxic treatment of a wide variety of tumors. Interestingly, few case reports underlined its potential to induce immune-mediated abscopal effects, resulting in regression of metastases, distant from the irradiated site. These observations are rare, and apparently depend on the dose used, suggesting that dose-related cellular responses may be involved in the distant immunogenic responses. Ionizing radiation (IR) has been reported to elicit immunogenic apoptosis, necroptosis, mitotic catastrophe, and senescence. In order to link a cellular outcome with a particular dose of irradiation, we performed a systematic study in a panel of cell lines on the cellular responses at different doses of X-rays. Remarkably, we observed that all cell lines tested responded in a similar fashion to IR with characteristics of mitotic catastrophe, senescence, lipid peroxidation, and caspase activity. Iron chelators (but not Ferrostatin-1 or vitamin E) could prevent the formation of lipid peroxides and cell death induced by IR, suggesting a crucial role of iron-dependent cell death during high-dose irradiation. We also show that in K-Ras-mutated cells, IR can induce morphological features reminiscent of methuosis, a cell death modality that has been recently described following H-Ras or K-Ras mutation overexpression.
Collapse
Affiliation(s)
- Sandy Adjemian
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - Teodora Oltean
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Sofie Martens
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Bartosz Wiernicki
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Vera Goossens
- VIB Screening Core & UGhent Expertise Centre for Bioassay Development and Screening (C-BIOS), VIB, UGhent, Ghent, Belgium
| | - Tom Vanden Berghe
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Benjamin Cappe
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Maria Ladik
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Franck B Riquet
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium.,Université de Lille, Lille, France
| | - Liesbeth Heyndrickx
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | - Jolien Bridelance
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium.,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium
| | | | - Katrien Vandecasteele
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium.,Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium.,Radiation Oncology, Ghent University Hospital, Ghent, Belgium
| | - Peter Vandenabeele
- Unit of Molecular Signaling and Cell Death, VIB Center for Inflammation Research, Ghent, Belgium. .,Department of Biomedical Molecular Biology, Cancer Research Institute Ghent (CRIG), Ghent University, Ghent, Belgium. .,Cancer Research Institute Ghent (CRIG), Ghent, Belgium. .,Methusalem program, Ghent University, Ghent, Belgium.
| |
Collapse
|
426
|
Picca A, Guerra F, Calvani R, Coelho-Júnior HJ, Landi F, Bernabei R, Romano R, Bucci C, Marzetti E. Extracellular Vesicles and Damage-Associated Molecular Patterns: A Pandora's Box in Health and Disease. Front Immunol 2020; 11:601740. [PMID: 33304353 PMCID: PMC7701251 DOI: 10.3389/fimmu.2020.601740] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 10/21/2020] [Indexed: 12/11/2022] Open
Abstract
Sterile inflammation develops as part of an innate immunity response to molecules released upon tissue injury and collectively indicated as damage-associated molecular patterns (DAMPs). While coordinating the clearance of potential harmful stimuli, promotion of tissue repair, and restoration of tissue homeostasis, a hyper-activation of such an inflammatory response may be detrimental. The complex regulatory pathways modulating DAMPs generation and trafficking are actively investigated for their potential to provide relevant insights into physiological and pathological conditions. Abnormal circulating extracellular vesicles (EVs) stemming from altered endosomal-lysosomal system have also been reported in several age-related conditions, including cancer and neurodegeneration, and indicated as a promising route for therapeutic purposes. Along this pathway, mitochondria may dispose altered components to preserve organelle homeostasis. However, whether a common thread exists between DAMPs and EVs generation is yet to be clarified. A deeper understanding of the highly complex, dynamic, and variable intracellular and extracellular trafficking of DAMPs and EVs, including those of mitochondrial origin, is needed to unveil relevant pathogenic pathways and novel targets for drug development. Herein, we describe the mechanisms of generation of EVs and mitochondrial-derived vesicles along the endocytic pathway and discuss the involvement of the endosomal-lysosomal in cancer and neurodegeneration (i.e., Alzheimer's and Parkinson's disease).
Collapse
Affiliation(s)
- Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Aging Research Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet and Stockholm University, Stockholm, Sweden
| | | | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberto Bernabei
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| | - Roberta Romano
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Cecilia Bucci
- Department of Biological and Environmental Sciences and Technologies, University of Salento, Lecce, Italy
| | - Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, Rome, Italy
- Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
427
|
Wang Y, Luo M, Chen Y, Wang Y, Zhang B, Ren Z, Bao L, Wang Y, Wang JE, Fu YX, Luo W, Wang Y. ZMYND8 Expression in Breast Cancer Cells Blocks T-Lymphocyte Surveillance to Promote Tumor Growth. Cancer Res 2020; 81:174-186. [PMID: 33148660 DOI: 10.1158/0008-5472.can-20-1710] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 10/05/2020] [Accepted: 10/30/2020] [Indexed: 11/16/2022]
Abstract
Emerging studies indicate that DNA damage in cancer cells triggers antitumor immunity, but its intrinsic regulatory mechanism in breast cancer cells remains poorly understood. Here, we show that ZMYND8 is upregulated and inhibits micronucleus formation and DNA damage in breast cancer cells. Loss of ZMYND8 triggered activation of the DNA sensor cyclic guanosine monophosphate-adenosine monophosphate synthase in micronuclei, leading to further activation of the downstream signaling effectors stimulator of IFN genes and NF-κB, but not TANK-binding kinase 1 and IFN regulatory factor 3, thereby inducing the expression of IFNβ and IFN-stimulated genes (ISG) in breast cancer cells in vitro and tumors in vivo. ZMYND8 knockout (KO) in breast cancer cells promoted infiltration of CD4+ and CD8+ T cells, leading to tumor inhibition in syngeneic mouse models, which was significantly attenuated by treatment of anti-CD4/CD8-depleting antibodies or anti-IFNAR1 antibody and in immunodeficient Rag1 KO mice. In human breast tumors, ZMYND8 was negatively correlated with ISGs, CD4, CD8A, CD8B, and the tumor-lymphocyte infiltration phenotype. Collectively, these findings demonstrate that maintenance of genome stability by ZMYND8 causes breast cancer cells to evade cytotoxic T-lymphocyte surveillance, which leads to tumor growth. SIGNIFICANCE: These findings show that ZMYND8 is a new negative and intrinsic regulator of the innate immune response in breast tumor cells, and ZMYND8 may be a possible target for antitumor immunotherapy.
Collapse
Affiliation(s)
- Yong Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Maowu Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yan Chen
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yijie Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Bo Zhang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Zhenhua Ren
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Lei Bao
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yanan Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Jennifer E Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Yang-Xin Fu
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - Weibo Luo
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas. .,Department of Pharmacology, UT Southwestern Medical Center, Dallas, Texas
| | - Yingfei Wang
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas. .,Department of Neurology, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
428
|
Putative Origins of Cell-Free DNA in Humans: A Review of Active and Passive Nucleic Acid Release Mechanisms. Int J Mol Sci 2020; 21:ijms21218062. [PMID: 33137955 PMCID: PMC7662960 DOI: 10.3390/ijms21218062] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 10/27/2020] [Accepted: 10/27/2020] [Indexed: 12/14/2022] Open
Abstract
Through various pathways of cell death, degradation, and regulated extrusion, partial or complete genomes of various origins (e.g., host cells, fetal cells, and infiltrating viruses and microbes) are continuously shed into human body fluids in the form of segmented cell-free DNA (cfDNA) molecules. While the genetic complexity of total cfDNA is vast, the development of progressively efficient extraction, high-throughput sequencing, characterization via bioinformatics procedures, and detection have resulted in increasingly accurate partitioning and profiling of cfDNA subtypes. Not surprisingly, cfDNA analysis is emerging as a powerful clinical tool in many branches of medicine. In addition, the low invasiveness of longitudinal cfDNA sampling provides unprecedented access to study temporal genomic changes in a variety of contexts. However, the genetic diversity of cfDNA is also a great source of ambiguity and poses significant experimental and analytical challenges. For example, the cfDNA population in the bloodstream is heterogeneous and also fluctuates dynamically, differs between individuals, and exhibits numerous overlapping features despite often originating from different sources and processes. Therefore, a deeper understanding of the determining variables that impact the properties of cfDNA is crucial, however, thus far, is largely lacking. In this work we review recent and historical research on active vs. passive release mechanisms and estimate the significance and extent of their contribution to the composition of cfDNA.
Collapse
|
429
|
Wu Y, Li S. Role of Post-Translational Modifications of cGAS in Innate Immunity. Int J Mol Sci 2020; 21:ijms21217842. [PMID: 33105828 PMCID: PMC7660100 DOI: 10.3390/ijms21217842] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/18/2020] [Accepted: 10/20/2020] [Indexed: 12/23/2022] Open
Abstract
Cyclic GMP–AMP synthase (cGAS) is the synthase that generates the second messenger cyclic GMP–AMP (cGAMP) upon DNA binding. cGAS was first discovered as the cytosolic DNA sensor that detects DNA exposed in the cytoplasm either from pathogens or cellular damage. Activated cGAS instigates the signaling cascades to activate type I interferon (IFN) expression, critical for host defense and autoimmune diseases. In addition, cGAS plays a role in senescence, DNA repair, apoptosis, and tumorigenesis. Recently, various post-translational modifications (PTMs) of cGAS have been reported, such as phosphorylation, ubiquitination, acetylation, glutamylation, and sumoylation. These PTMs profoundly affect cGAS functions. Thus, here we review the recent reported PTMs of cGAS and how these PTMs regulate cGAS enzymatic activity, DNA binding, and protein stability, and discuss the potential future directions.
Collapse
Affiliation(s)
| | - Shitao Li
- Correspondence: ; Tel.: +1-504-988-2203
| |
Collapse
|
430
|
Selim NM, El-Hawary SS, El Zalabani SM, Shamma RN, Mahdy NES, Sherif NH, Fahmy HA, Mekkawy MH, Yasri A, Sobeh M. Impact of Washingtonia robusta Leaves on Gamma Irradiation-Induced Hepatotoxicity in Rats and Correlation with STING Pathway and Phenolic Composition. Pharmaceuticals (Basel) 2020; 13:E320. [PMID: 33086669 PMCID: PMC7603372 DOI: 10.3390/ph13100320] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/23/2022] Open
Abstract
Exposure to ionizing radiation usually results in cellular oxidative damage and may induce liver toxicity. The efficiency of the ethanol extracts of Washingtonia filifera (EWF) and Washingtonia robusta (EWR) leaves in alleviating γ-radiation-induced oxidative hepatotoxicity was herein explored. Proximate and macronutrient composition of the leaves was determined to establish reliable quality control criteria. Colorimetric estimation of total phenolic (TPC) and flavonoid (TFC) contents revealed their occurrence in larger amounts in EWR. In vitro evaluation of the antioxidant capacity by 2,2-azinobis (3-ethylbenzothiazoline-6-sulfonic acid (ABTS) and ferric reducing antioxidant power (FRAP) assays confirmed higher efficiency of EWR designating a close correlation with phenolic composition. Four phenolics, viz., naringenin, kaempferol, quercetin, and gallic acid, were isolated from EWR. In vivo assessment of the extracts' antioxidant potential was performed on γ-irradiated (7.5 Gy) female rats. EWR was found more efficient in restoring the elevated liver index, ALT, albumin, cholesterol, and reactive oxygen species (ROS) levels. Both extracts ameliorated the increase in the stimulator of interferon gene (STING) expression. Bioactivity was confirmed by immuno-histochemical examination of inflammatory and apoptotic biomarkers (TNF-α, IL-6 and caspase-3) and histopathological architecture. In addition, the interactions of the isolated compounds with STING were assessed in silico by molecular docking. Therefore, Washingtonia robusta leaves might be suggested as a valuable nutritional supplement to alleviate radiotherapy-induced hepatotoxicity.
Collapse
Affiliation(s)
- Nabil M. Selim
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt; (S.S.E.-H.); (S.M.E.Z.); (N.E.S.M.)
| | - Seham S. El-Hawary
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt; (S.S.E.-H.); (S.M.E.Z.); (N.E.S.M.)
| | - Soheir M. El Zalabani
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt; (S.S.E.-H.); (S.M.E.Z.); (N.E.S.M.)
| | - Rehab Nabil Shamma
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt;
| | - Nariman El Sayed Mahdy
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo 12613, Egypt; (S.S.E.-H.); (S.M.E.Z.); (N.E.S.M.)
| | - Noheir H. Sherif
- Pharmacognosy Department, Faculty of Pharmacy, Nahda University, Beni-Suef 62513, Egypt;
- Drug Radiation Research Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, P.O. Box: 29 Nasr City, Cairo 11865, Egypt; (H.A.F.); (M.H.M.)
| | - Hanan A. Fahmy
- Drug Radiation Research Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, P.O. Box: 29 Nasr City, Cairo 11865, Egypt; (H.A.F.); (M.H.M.)
| | - Mai H. Mekkawy
- Drug Radiation Research Department, National Centre for Radiation Research and Technology, Egyptian Atomic Energy Authority, P.O. Box: 29 Nasr City, Cairo 11865, Egypt; (H.A.F.); (M.H.M.)
| | - Abdelaziz Yasri
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660–Hay MoulayRachid, 43150 Ben-Guerir, Morocco;
| | - Mansour Sobeh
- AgroBioSciences Research Division, Mohammed VI Polytechnic University, Lot 660–Hay MoulayRachid, 43150 Ben-Guerir, Morocco;
| |
Collapse
|
431
|
Genovese I, Vezzani B, Danese A, Modesti L, Vitto VAM, Corazzi V, Pelucchi S, Pinton P, Giorgi C. Mitochondria as the decision makers for cancer cell fate: from signaling pathways to therapeutic strategies. Cell Calcium 2020; 92:102308. [PMID: 33096320 DOI: 10.1016/j.ceca.2020.102308] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 10/05/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023]
Abstract
As pivotal players in cellular metabolism, mitochondria have a double-faceted role in the final decision of cell fate. This is true for all cell types, but it is even more important and intriguing in the cancer setting. Mitochondria regulate cell fate in many diverse ways: through metabolism, by producing ATP and other metabolites deemed vital or detrimental for cancer cells; through the regulation of Ca2+ homeostasis, especially by the joint participation of the endoplasmic reticulum in a membranous tethering system for Ca2+ signaling called mitochondria-ER associated membranes (MAMs); and by regulating signaling pathways involved in the survival of cancer cells such as mitophagy. Recent studies have shown that mitochondria can also play a role in the regulation of inflammatory pathways in cancer cells, for example, through the release of mitochondrial DNA (mtDNA) involved in the activation of the cGAS-cGAMP-STING pathway. In this review, we aim to explore the role of mitochondria as decision makers in fostering cancer cell death or survival depending on the tumor cell stage and describe novel anticancer therapeutic strategies targeting mitochondria.
Collapse
Affiliation(s)
- Ilaria Genovese
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Bianca Vezzani
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Alberto Danese
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Lorenzo Modesti
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Veronica Angela Maria Vitto
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Virginia Corazzi
- ENT & Audiology Department, University Hospital of Ferrara, Ferrara, Italy
| | - Stefano Pelucchi
- ENT & Audiology Department, University Hospital of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|
432
|
Wanderoy S, Hees JT, Klesse R, Edlich F, Harbauer AB. Kill one or kill the many: interplay between mitophagy and apoptosis. Biol Chem 2020; 402:73-88. [PMID: 33544491 DOI: 10.1515/hsz-2020-0231] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Mitochondria are key players of cellular metabolism, Ca2+ homeostasis, and apoptosis. The functionality of mitochondria is tightly regulated, and dysfunctional mitochondria are removed via mitophagy, a specialized form of autophagy that is compromised in hereditary forms of Parkinson's disease. Through mitophagy, cells are able to cope with mitochondrial stress until the damage becomes too great, which leads to the activation of pro-apoptotic BCL-2 family proteins located on the outer mitochondrial membrane. Active pro-apoptotic BCL-2 proteins facilitate the release of cytochrome c from the mitochondrial intermembrane space (IMS) into the cytosol, committing the cell to apoptosis by activating a cascade of cysteinyl-aspartate specific proteases (caspases). We are only beginning to understand how the choice between mitophagy and the activation of caspases is determined on the mitochondrial surface. Intriguingly in neurons, caspase activation also plays a non-apoptotic role in synaptic plasticity. Here we review the current knowledge on the interplay between mitophagy and caspase activation with a special focus on the central nervous system.
Collapse
Affiliation(s)
- Simone Wanderoy
- Max Planck Institute for Neurobiology, Am Klopferspitz 18, D-82152Martinsried, Germany
| | - J Tabitha Hees
- Max Planck Institute for Neurobiology, Am Klopferspitz 18, D-82152Martinsried, Germany
| | - Ramona Klesse
- Institute for Biochemistry and Molecular Biology, University of Freiburg, D-79104Freiburg, Germany.,Faculty of Biology, University of Freiburg, D-79104Freiburg, Germany
| | - Frank Edlich
- Institute for Biochemistry and Molecular Biology, University of Freiburg, D-79104Freiburg, Germany
| | - Angelika B Harbauer
- Max Planck Institute for Neurobiology, Am Klopferspitz 18, D-82152Martinsried, Germany.,Technical University of Munich, Institute of Neuronal Cell Biology, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
433
|
McArthur K, Kile BT. Apoptotic mitochondria prime anti-tumour immunity. Cell Death Discov 2020; 6:98. [PMID: 33083019 PMCID: PMC7542170 DOI: 10.1038/s41420-020-00335-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/06/2020] [Accepted: 09/10/2020] [Indexed: 11/19/2022] Open
Affiliation(s)
- Kate McArthur
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800 Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005 Australia
| | - Benjamin T. Kile
- Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Melbourne, VIC 3800 Australia
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, 5005 Australia
| |
Collapse
|
434
|
Yu CH, Davidson S, Harapas CR, Hilton JB, Mlodzianoski MJ, Laohamonthonkul P, Louis C, Low RRJ, Moecking J, De Nardo D, Balka KR, Calleja DJ, Moghaddas F, Ni E, McLean CA, Samson AL, Tyebji S, Tonkin CJ, Bye CR, Turner BJ, Pepin G, Gantier MP, Rogers KL, McArthur K, Crouch PJ, Masters SL. TDP-43 Triggers Mitochondrial DNA Release via mPTP to Activate cGAS/STING in ALS. Cell 2020; 183:636-649.e18. [PMID: 33031745 PMCID: PMC7599077 DOI: 10.1016/j.cell.2020.09.020] [Citation(s) in RCA: 617] [Impact Index Per Article: 123.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 07/21/2020] [Accepted: 09/08/2020] [Indexed: 02/07/2023]
Abstract
Cytoplasmic accumulation of TDP-43 is a disease hallmark for many cases of amyotrophic lateral sclerosis (ALS), associated with a neuroinflammatory cytokine profile related to upregulation of nuclear factor κB (NF-κB) and type I interferon (IFN) pathways. Here we show that this inflammation is driven by the cytoplasmic DNA sensor cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS) when TDP-43 invades mitochondria and releases DNA via the permeability transition pore. Pharmacologic inhibition or genetic deletion of cGAS and its downstream signaling partner STING prevents upregulation of NF-κB and type I IFN induced by TDP-43 in induced pluripotent stem cell (iPSC)-derived motor neurons and in TDP-43 mutant mice. Finally, we document elevated levels of the specific cGAS signaling metabolite cGAMP in spinal cord samples from patients, which may be a biomarker of mtDNA release and cGAS/STING activation in ALS. Our results identify mtDNA release and cGAS/STING activation as critical determinants of TDP-43-associated pathology and demonstrate the potential for targeting this pathway in ALS. TDP-43 enters mitochondria, triggers mtDNA release via the mPTP TDP-43-induced cytosolic mtDNA accumulation activates the cGAS/STING pathway Evidence of cytoplasmic mtDNA was found in ALS patient cells and disease models Blocking STING prevents inflammation and neurodegeneration in vitro and in vivo
Collapse
Affiliation(s)
- Chien-Hsiung Yu
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Sophia Davidson
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Cassandra R Harapas
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - James B Hilton
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC 3010, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Michael J Mlodzianoski
- Centre for Dynamic Imaging, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Pawat Laohamonthonkul
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Cynthia Louis
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Ronnie Ren Jie Low
- Personalised Oncology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Jonas Moecking
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Institute of Structural Biology, University of Bonn, 53127 Bonn, Germany
| | - Dominic De Nardo
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia
| | - Katherine R Balka
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia
| | - Dale J Calleja
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Fiona Moghaddas
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Department of Immunology and Allergy, The Royal Melbourne Hospital, Parkville, VIC 3052, Australia
| | - Erya Ni
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Catriona A McLean
- Anatomical Pathology, The Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Andre L Samson
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Shiraz Tyebji
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christopher J Tonkin
- Infection and Immunity Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Christopher R Bye
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Bradley J Turner
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Genevieve Pepin
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Michael P Gantier
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, VIC, Australia; Department of Molecular and Translational Science, Monash University, Clayton, VIC 3168, Australia
| | - Kelly L Rogers
- Centre for Dynamic Imaging, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia
| | - Kate McArthur
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC 3168, Australia
| | - Peter J Crouch
- Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, VIC 3010, Australia; Florey Institute of Neuroscience and Mental Health, University of Melbourne, Parkville, VIC 3010, Australia
| | - Seth L Masters
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3010, Australia; Immunology Laboratory, Guangzhou Institute of Paediatrics, Guangzhou Women and Children's Medical Centre, Guangzhou, Guangdong 510623, China.
| |
Collapse
|
435
|
Yamazaki T, Kirchmair A, Sato A, Buqué A, Rybstein M, Petroni G, Bloy N, Finotello F, Stafford L, Navarro Manzano E, Ayala de la Peña F, García-Martínez E, Formenti SC, Trajanoski Z, Galluzzi L. Mitochondrial DNA drives abscopal responses to radiation that are inhibited by autophagy. Nat Immunol 2020; 21:1160-1171. [PMID: 32747819 DOI: 10.1038/s41590-020-0751-0] [Citation(s) in RCA: 257] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 06/23/2020] [Indexed: 12/18/2022]
Abstract
Autophagy supports both cellular and organismal homeostasis. However, whether autophagy should be inhibited or activated for cancer therapy remains unclear. Deletion of essential autophagy genes increased the sensitivity of mouse mammary carcinoma cells to radiation therapy in vitro and in vivo (in immunocompetent syngeneic hosts). Autophagy-deficient cells secreted increased amounts of type I interferon (IFN), which could be limited by CGAS or STING knockdown, mitochondrial DNA depletion or mitochondrial outer membrane permeabilization blockage via BCL2 overexpression or BAX deletion. In vivo, irradiated autophagy-incompetent mammary tumors elicited robust immunity, leading to improved control of distant nonirradiated lesions via systemic type I IFN signaling. Finally, a genetic signature of autophagy had negative prognostic value in patients with breast cancer, inversely correlating with mitochondrial abundance, type I IFN signaling and effector immunity. As clinically useful autophagy inhibitors are elusive, our findings suggest that mitochondrial outer membrane permeabilization may represent a valid target for boosting radiation therapy immunogenicity in patients with breast cancer.
Collapse
Affiliation(s)
- Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Alexander Kirchmair
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Ai Sato
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Marissa Rybstein
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Norma Bloy
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Francesca Finotello
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Lena Stafford
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Esther Navarro Manzano
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
- Universidad de Murcia, Murcia, Spain
| | - Francisco Ayala de la Peña
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
- Universidad de Murcia, Murcia, Spain
| | - Elena García-Martínez
- Hematology and Oncology Department, Hospital Universitario Morales Meseguer, Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB-Arrixaca), Murcia, Spain
- Universidad Católica San Antonio de Murcia, Guadalupe, Spain
| | - Silvia C Formenti
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, New York, NY, USA
| | - Zlatko Trajanoski
- Biocenter, Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, New York, NY, USA.
- Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA.
- Université Paris Descartes/Paris V, Paris, France.
| |
Collapse
|
436
|
Gao M, He Y, Tang H, Chen X, Liu S, Tao Y. cGAS/STING: novel perspectives of the classic pathway. MOLECULAR BIOMEDICINE 2020; 1:7. [PMID: 35006429 PMCID: PMC8603984 DOI: 10.1186/s43556-020-00006-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 08/09/2020] [Indexed: 02/06/2023] Open
Abstract
Cyclic GMP-AMP (cGAMP) synthase (cGAS) is a cytosolic DNA sensor and innate immune response initiator. Binding with exogenous or endogenous nucleic acids, cGAS activates its downstream adaptor, stimulator of interferon genes (STING). STING then triggers protective immune to enable the elimination of the pathogens and the clearance of cancerous cells. Apparently, aberrantly activated by self-DNA, cGAS/STING pathway is threatening to cause autoimmune and inflammatory diseases. The effects of cGAS/STING in defenses against infection and autoimmune diseases have been well studied, still it is worthwhile to discuss the roles of cGAS/STING pathway beyond the “classical” realm of innate immunity. Recent studies have revealed its involvement in non-canonical inflammasome formation, calcium hemostasis regulation, endoplasmic reticulum (ER) stress response, perception of leaking mitochondrial DNA (mtDNA), autophagy induction, cellular senescence and senescence-associated secretory phenotype (SASP) production, providing an exciting area for future exploration. Previous studies generally focused on the function of cGAS/STING pathway in cytoplasm and immune response. In this review, we summarize the latest research of this pathway on the regulation of other physiological process and STING independent reactions to DNA in micronuclei and nuclei. Together, these studies provide a new perspective of cGAS/STING pathway in human diseases.
Collapse
Affiliation(s)
- Menghui Gao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Yuchen He
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China
| | - Haosheng Tang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Xiangyu Chen
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China.,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China
| | - Shuang Liu
- Department of Oncology, Institute of Medical Sciences, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Hunan, 410078, China. .,NHC Key Laboratory of Carcinogenesis (Central South University), Cancer Research Institute and School of Basic Medicine, Central South University, Changsha, 410078, Hunan, China. .,Hunan Key Laboratory of Tumor Models and Individualized Medicine, Department of Thoracic Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, China.
| |
Collapse
|
437
|
Murthy AMV, Robinson N, Kumar S. Crosstalk between cGAS-STING signaling and cell death. Cell Death Differ 2020; 27:2989-3003. [PMID: 32948836 DOI: 10.1038/s41418-020-00624-8] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/07/2020] [Indexed: 02/06/2023] Open
Abstract
Cytosolic nucleic acid sensors have a critical role in detecting endogenous nucleic acids to initiate innate immune responses during microbial infections and/or cell death. Several seminal studies over the past decade have delineated the conserved mechanism of cytosolic DNA sensor cyclic GMP-AMP synthase (cGAS) and the downstream signaling adapter stimulator of interferon genes (STING) in mediating innate immune signaling pathways as a host defense mechanism. Besides the predominant role in microbial infections and inflammatory diseases, there is an increased attention on alternative functional responses of cGAS-STING-mediated signaling. Here we review the complexity of interactions between the cGAS-STING signaling and cell death pathways. A better understanding of molecular mechanisms of this interplay is important with regard to the development of new therapeutics targeting cGAS-STING signaling in cancer, infectious, and chronic inflammatory diseases.
Collapse
Affiliation(s)
- Ambika M V Murthy
- Centre for Cancer Biology, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia.
| | - Nirmal Robinson
- Centre for Cancer Biology, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia.
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia, GPO Box 2471, Adelaide, SA, 5001, Australia.
| |
Collapse
|
438
|
Galluzzi L, Petroni G, Kroemer G. Immunogenicity of cell death driven by immune effectors. J Immunother Cancer 2020; 8:jitc-2020-000802. [PMID: 32253221 PMCID: PMC7174067 DOI: 10.1136/jitc-2020-000802] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2020] [Indexed: 02/01/2023] Open
Abstract
Whether cell death caused by T lymphocytes and natural killer (NK) cells would be immunogenic per se has been a matter of intense debate. Two back-to-back papers from the Melero’s and Pardo’s groups have now resolved this conundrum, demonstrating that T and NK cell–mediated cytotoxicity represents indeed a bona fide variant of immunogenic cell death.
Collapse
Affiliation(s)
- Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA .,Sandra and Edward Meyer Cancer Center, New York, NY, United States.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.,Department of Dermatology, Yale School of Medicine, New Haven, CT, United States.,Université de Paris, Paris, France
| | - Giulia Petroni
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Guido Kroemer
- Université de Paris, Paris, France .,Equipe labellisée par la Ligue contre le cancer, INSERM U1138, Centre de Recherche des Cordeliers, Paris, France.,Metabolomics and Cell Biology Platforms, Gustave Roussy Comprehensive Cancer Institute, Villejuif, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Sciences, Suzhou, China.,Department of Women's and Children's Health, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
439
|
Tao L, Lemoff A, Wang G, Zarek C, Lowe A, Yan N, Reese TA. Reactive oxygen species oxidize STING and suppress interferon production. eLife 2020; 9:e57837. [PMID: 32886065 PMCID: PMC7473769 DOI: 10.7554/elife.57837] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
Reactive oxygen species (ROS) are by-products of cellular respiration that can promote oxidative stress and damage cellular proteins and lipids. One canonical role of ROS is to defend the cell against invading bacterial and viral pathogens. Curiously, some viruses, including herpesviruses, thrive despite the induction of ROS, suggesting that ROS are beneficial for the virus. However, the underlying mechanisms remain unclear. Here, we found that ROS impaired interferon response during murine herpesvirus infection and that the inhibition occurred downstream of cytoplasmic DNA sensing. We further demonstrated that ROS suppressed the type I interferon response by oxidizing Cysteine 147 on murine stimulator of interferon genes (STING), an ER-associated protein that mediates interferon response after cytoplasmic DNA sensing. This inhibited STING polymerization and activation of downstream signaling events. These data indicate that redox regulation of Cysteine 147 of mouse STING, which is equivalent to Cysteine 148 of human STING, controls interferon production. Together, our findings reveal that ROS orchestrates anti-viral immune responses, which can be exploited by viruses to evade cellular defenses.
Collapse
Affiliation(s)
- Lili Tao
- Department of Immunology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical CenterDallasUnited States
| | - Guoxun Wang
- Department of Immunology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Christina Zarek
- Department of Immunology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Alexandria Lowe
- Department of Immunology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Nan Yan
- Department of Immunology, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Microbiology, University of Texas Southwestern Medical CenterDallasUnited States
| | - Tiffany A Reese
- Department of Immunology, University of Texas Southwestern Medical CenterDallasUnited States
- Department of Microbiology, University of Texas Southwestern Medical CenterDallasUnited States
| |
Collapse
|
440
|
Jing R, Hu ZK, Lin F, He S, Zhang SS, Ge WY, Dai HJ, Du XK, Lin JY, Pan LH. Mitophagy-Mediated mtDNA Release Aggravates Stretching-Induced Inflammation and Lung Epithelial Cell Injury via the TLR9/MyD88/NF-κB Pathway. Front Cell Dev Biol 2020; 8:819. [PMID: 33015037 PMCID: PMC7504878 DOI: 10.3389/fcell.2020.00819] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 08/03/2020] [Indexed: 01/04/2023] Open
Abstract
Background In animal models of ventilation-induced lung injury, mitophagy triggers mitochondria damage and the release of mitochondrial (mt) DNA, which activates inflammation. However, the mechanism of this process is unclear. Methods A model of cyclic stretching (CS)-induced lung epithelial cell injury was established. The genetic intervention of phosphatase and tensin homolog-induced kinase 1 (PINK1) expression via lentivirus transfection was used to identify the relationship between PINK1-mediated mitophagy and mtDNA release in stretching-induced inflammatory response and injury. Pharmacological inhabitation of Toll-like receptor 9 (TLR9) and myeloid differentiation factor 88 (MyD88) expression was performed via their related inhibitors, while pre-treatment of exogenous mtDNA was used to verify the role of mtDNA in stretching-induced inflammatory response and injury. Results Using a cell culture model of CS, we found that knocking down PINK1 in lung epithelial cells reduced mitophagy activation and mtDNA release, leading to milder inflammatory response and injury; conversely, up-regulating PINK1 exacerbated stretching-induced inflammation and injury, and similar effects were observed by upregulating TLR9 to induce expression of MyD88 and nuclear factor-κB (NF-κB)/p65. Down-regulating MyD88 protected lung epithelial cells from stretching injury and decreased NF-κB/p65 expression. Conclusion These findings suggest that PINK1-dependent mitophagy and associated TLR9 activation is indeed a major factor in stretch-induced cell injury via a mechanism in which released mtDNA activates TLR9 and thereby the MyD88/NF-κB pathway. Inhibiting this process may be a therapeutic approach to prevent inflammation and cell injury in patients on mechanical ventilation.
Collapse
Affiliation(s)
- Ren Jing
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Zhao-Kun Hu
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Fei Lin
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Sheng He
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Sui-Sui Zhang
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Wan-Yun Ge
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Hui-Jun Dai
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Xue-Ke Du
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Jin-Yuan Lin
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| | - Ling-Hui Pan
- Department of Anesthesiology, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China.,The Laboratory of Perioperative Medicine Research Center, Guangxi Medical University Affiliated Tumor Hospital & Oncology Medical College, Nanning, China
| |
Collapse
|
441
|
Bu L, Wang H, Hou P, Guo S, He M, Xiao J, Li P, Zhong Y, Jia P, Cao Y, Liang G, Yang C, Chen L, Guo D, Li CM. The Ubiquitin E3 Ligase Parkin Inhibits Innate Antiviral Immunity Through K48-Linked Polyubiquitination of RIG-I and MDA5. Front Immunol 2020; 11:1926. [PMID: 32983119 PMCID: PMC7492610 DOI: 10.3389/fimmu.2020.01926] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 07/17/2020] [Indexed: 01/02/2023] Open
Abstract
Innate immunity is the first-line defense against antiviral or antimicrobial infection. RIG-I and MDA5, which mediate the recognition of pathogen-derived nucleic acids, are essential for production of type I interferons (IFN). Here, we identified mitochondrion depolarization inducer carbonyl cyanide 3-chlorophenylhydrazone (CCCP) inhibited the response and antiviral activity of type I IFN during viral infection. Furthermore, we found that the PTEN-induced putative kinase 1 (PINK1) and the E3 ubiquitin-protein ligase Parkin mediated mitophagy, thus negatively regulating the activation of RIG-I and MDA5. Parkin directly interacted with and catalyzed the K48-linked polyubiquitination and subsequent degradation of RIG-I and MDA5. Thus, we demonstrate that Parkin limits RLR-triggered innate immunity activation, suggesting Parkin as a potential therapeutic target for the control of viral infection.
Collapse
Affiliation(s)
- Lang Bu
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Huan Wang
- Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Panpan Hou
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Shuting Guo
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Miao He
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Jingshu Xiao
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Ping Li
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Yongheng Zhong
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Penghui Jia
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Yuanyuan Cao
- School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Guanzhan Liang
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Chenwei Yang
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Lang Chen
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Deyin Guo
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Chun-Mei Li
- MOE Key Laboratory of Tropical Disease Control, the Infection and Immunity Center (TIIC), School of Medicine, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
442
|
Ma R, Ortiz Serrano TP, Davis J, Prigge AD, Ridge KM. The cGAS-STING pathway: The role of self-DNA sensing in inflammatory lung disease. FASEB J 2020; 34:13156-13170. [PMID: 32860267 PMCID: PMC8121456 DOI: 10.1096/fj.202001607r] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
The presence of DNA in the cytosol is usually a sign of microbial infections, which alerts the host innate immune system to mount a defense response. Cyclic GMP-AMP synthase (cGAS) is a critical cytosolic DNA sensor that elicits robust innate immune responses through the production of the second messenger, cyclic GMP-AMP (cGAMP), which binds and activates stimulator of interferon genes (STING). However, cGAS binds to DNA irrespective of DNA sequence, therefore, self-DNA leaked from the nucleus or mitochondria can also serve as a cGAS ligand to activate this pathway and trigger extensive inflammatory responses. Dysregulation of the cGAS-STING pathway is responsible for a broad array of inflammatory and autoimmune diseases. Recently, evidence has shown that self-DNA release and cGAS-STING pathway over-activation can drive lung disease, making this pathway a promising therapeutic target for inflammatory lung disease. Here, we review recent advances on the cGAS-STING pathway governing self-DNA sensing, highlighting its role in pulmonary disease.
Collapse
Affiliation(s)
- Ruihua Ma
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tatiana P Ortiz Serrano
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer Davis
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew D Prigge
- Division of Critical Care Medicine, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
443
|
Tang R, Xu J, Zhang B, Liu J, Liang C, Hua J, Meng Q, Yu X, Shi S. Ferroptosis, necroptosis, and pyroptosis in anticancer immunity. J Hematol Oncol 2020; 13:110. [PMID: 32778143 PMCID: PMC7418434 DOI: 10.1186/s13045-020-00946-7] [Citation(s) in RCA: 830] [Impact Index Per Article: 166.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
In recent years, cancer immunotherapy based on immune checkpoint inhibitors (ICIs) has achieved considerable success in the clinic. However, ICIs are significantly limited by the fact that only one third of patients with most types of cancer respond to these agents. The induction of cell death mechanisms other than apoptosis has gradually emerged as a new cancer treatment strategy because most tumors harbor innate resistance to apoptosis. However, to date, the possibility of combining these two modalities has not been discussed systematically. Recently, a few studies revealed crosstalk between distinct cell death mechanisms and antitumor immunity. The induction of pyroptosis, ferroptosis, and necroptosis combined with ICIs showed synergistically enhanced antitumor activity, even in ICI-resistant tumors. Immunotherapy-activated CD8+ T cells are traditionally believed to induce tumor cell death via the following two main pathways: (i) perforin-granzyme and (ii) Fas-FasL. However, recent studies identified a new mechanism by which CD8+ T cells suppress tumor growth by inducing ferroptosis and pyroptosis, which provoked a review of the relationship between tumor cell death mechanisms and immune system activation. Hence, in this review, we summarize knowledge of the reciprocal interaction between antitumor immunity and distinct cell death mechanisms, particularly necroptosis, ferroptosis, and pyroptosis, which are the three potentially novel mechanisms of immunogenic cell death. Because most evidence is derived from studies using animal and cell models, we also reviewed related bioinformatics data available for human tissues in public databases, which partially confirmed the presence of interactions between tumor cell death and the activation of antitumor immunity.
Collapse
Affiliation(s)
- Rong Tang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Bo Zhang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jiang Liu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Chen Liang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jie Hua
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Qingcai Meng
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| | - Si Shi
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, No. 270 Dong'An Road, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Pancreatic Cancer Institute, No. 270 Dong'An Road, Shanghai, 200032, China.
- Pancreatic Cancer Institute, Fudan University, Shanghai, China.
| |
Collapse
|
444
|
Ni G, Ma Z, Wong JP, Zhang Z, Cousins E, Major MB, Damania B. PPP6C Negatively Regulates STING-Dependent Innate Immune Responses. mBio 2020; 11:e01728-20. [PMID: 32753499 PMCID: PMC7407089 DOI: 10.1128/mbio.01728-20] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 06/30/2020] [Indexed: 12/17/2022] Open
Abstract
Stimulator of interferon genes (STING) is an essential adaptor protein of the innate DNA-sensing signaling pathway, which recognizes genomic DNA from invading pathogens to establish antiviral responses in host cells. STING activity is tightly regulated by several posttranslational modifications, including phosphorylation. However, specifically how the phosphorylation status of STING is modulated by kinases and phosphatases remains to be fully elucidated. In this study, we identified protein phosphatase 6 catalytic subunit (PPP6C) as a binding partner of Kaposi's sarcoma-associated herpesvirus (KSHV) open reading frame 48 (ORF48), which is a negative regulator of the cyclic GMP-AMP synthase (cGAS)-STING pathway. PPP6C depletion enhances double-stranded DNA (dsDNA)-induced and 5'ppp double-stranded RNA (dsRNA)-induced but not poly(I:C)-induced innate immune responses. PPP6C negatively regulates dsDNA-induced IRF3 activation but not NF-κB activation. Deficiency of PPP6C greatly inhibits the replication of herpes simplex virus 1 (HSV-1) and vesicular stomatitis virus (VSV) as well as the reactivation of KSHV, due to increased type I interferon production. We further demonstrated that PPP6C interacts with STING and that loss of PPP6C enhances STING phosphorylation. These data demonstrate the important role of PPP6C in regulating STING phosphorylation and activation, which provides an additional mechanism by which the host responds to viral infection.IMPORTANCE Cytosolic DNA, which usually comes from invading microbes, is a dangerous signal to the host. The cGAS-STING pathway is the major player that detects cytosolic DNA and then evokes the innate immune response. As an adaptor protein, STING plays a central role in controlling activation of the cGAS-STING pathway. Although transient activation of STING is essential to trigger the host defense during pathogen invasion, chronic STING activation has been shown to be associated with several autoinflammatory diseases. Here, we report that PPP6C negatively regulates the cGAS-STING pathway by removing STING phosphorylation, which is required for its activation. Dephosphorylation of STING by PPP6C helps prevent the sustained production of STING-dependent cytokines, which would otherwise lead to severe autoimmune disorders. This work provides additional mechanisms on the regulation of STING activity and might facilitate the development of novel therapeutics designed to prevent a variety of autoinflammatory disorders.
Collapse
Affiliation(s)
- Guoxin Ni
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Zhe Ma
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Jason P Wong
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Zhigang Zhang
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Emily Cousins
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - M Ben Major
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Blossom Damania
- Department of Microbiology and Immunology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
445
|
Zhong W, Rao Z, Rao J, Han G, Wang P, Jiang T, Pan X, Zhou S, Zhou H, Wang X. Aging aggravated liver ischemia and reperfusion injury by promoting STING-mediated NLRP3 activation in macrophages. Aging Cell 2020; 19:e13186. [PMID: 32666684 PMCID: PMC7431827 DOI: 10.1111/acel.13186] [Citation(s) in RCA: 102] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 04/29/2020] [Accepted: 06/06/2020] [Indexed: 12/19/2022] Open
Abstract
Although aggravated liver injury has been reported in aged livers post‐ischemia and reperfusion (IR), the underlying mechanism of innate immune activation of aged macrophages is not well understood. Here, we investigated whether and how Stimulator of interferon genes (STING) signaling regulated macrophage proinflammatory activation and liver IR injury. Mice were subjected to hepatic IR in vivo. Macrophages isolated from IR‐stressed livers and bone marrow‐derived macrophages (BMDMs) from young and aged mice were used for in vitro studies. Enhanced nucleotide‐binding domain and leucine‐rich repeat containing protein 3 (NLRP3) activation was found in both livers and macrophages of aged mice post‐IR. NLRP3 knockdown in macrophages inhibited intrahepatic inflammation and liver injury in both young and aged mice. Interestingly, enhanced activation of the STING/ TANK‐binding kinase 1 (TBK1) signaling pathway was observed in aged macrophages post‐IR and mitochondria DNA (mtDNA) stimulation. STING suppression blocked over‐activation of NLRP3 signaling and excessive secretion of proinflammatory cytokines/chemokines in the mtDNA‐stimulated BMDMs from aged mice. More importantly, STING knockdown in macrophages abrogated the detrimental role of aging in aggravating liver IR injury and intrahepatic inflammation. Finally, peripheral blood from the recipients undergoing liver transplantation was collected and analyzed. The results showed that the elderly recipients had much higher levels of TNF‐α, IL‐6, IL‐1β, and IL‐18 post‐transplantation, indicating increased NLRP3 activation in lR‐stressed livers of elderly recipients. In summary, our study demonstrated that the STING‐NLRP3 axis was critical for the proinflammatory response of aged macrophages and would be a novel therapeutic target to reduce IR injury in elderly patients.
Collapse
Affiliation(s)
- Weizhe Zhong
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| | - Zhuqing Rao
- Department of Anesthesiology The First Affiliated Hospital with Nanjing Medical University Nanjing China
| | - Jianhua Rao
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| | - Guoyong Han
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| | - Ping Wang
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| | - Tao Jiang
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| | - Xiongxiong Pan
- Department of Anesthesiology The First Affiliated Hospital with Nanjing Medical University Nanjing China
| | - Shun Zhou
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| | - Haoming Zhou
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| | - Xuehao Wang
- Hepatobiliary/Liver Transplantation Center The First Affiliated Hospital with Nanjing Medical University Nanjing China
- Research Unit of Liver Transplantation and Transplant Immunology Chinese Academy of Medical Sciences Nanjing China
- Key Laboratory of Liver Transplantation Chinese Academy of Medical Sciences Nanjing China
| |
Collapse
|
446
|
Hoong BYD, Gan YH, Liu H, Chen ES. cGAS-STING pathway in oncogenesis and cancer therapeutics. Oncotarget 2020; 11:2930-2955. [PMID: 32774773 PMCID: PMC7392626 DOI: 10.18632/oncotarget.27673] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 06/20/2020] [Indexed: 02/06/2023] Open
Abstract
The host innate immunity offers the first line of defense against infection. However, recent evidence shows that the host innate immunity is also critical in sensing the presence of cytoplasmic DNA derived from genomic instability events, such as DNA damage and defective cell cycle progression. This is achieved through the cyclic GMP-AMP synthase (cGAS)/Stimulator of interferon (IFN) genes (STING) pathway. Here we discuss recent insights into the regulation of this pathway in cancer immunosurveillance, and the downstream signaling cascades that coordinate immune cell recruitment to the tumor microenvironment to destroy transformed cells through cellular senescence or cell death programs. Its central role in immunosurveillance positions the cGAS-STING pathway as an attractive anti-cancer immunotherapeutic drug target for chemical agonists or vaccine adjuvants and suggests a key node to be targeted in a synthetic lethal approach. We also discuss adaptive mechanisms used by cancer cells to circumvent cGAS-STING signaling and present evidence linking chronic cGAS-STING activation to inflammation-induced carcinogenesis, cautioning against the use of activating the cGAS-STING pathway as an anti-tumor immunotherapy. A deeper mechanistic understanding of the cGAS-STING pathway will aid in the identification of potentially efficacious anti-cancer therapeutic targets.
Collapse
Affiliation(s)
- Brandon Yi Da Hoong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System (NUHS), Singapore
- Wong Hock Boon Society, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Yunn Hwen Gan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System (NUHS), Singapore
- NUS Graduate School of Integrative Sciences & Engineering (NGS), National University of Singapore, Singapore
| | - Haiyan Liu
- National University Health System (NUHS), Singapore
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Ee Sin Chen
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
- National University Health System (NUHS), Singapore
- NUS Graduate School of Integrative Sciences & Engineering (NGS), National University of Singapore, Singapore
| |
Collapse
|
447
|
Jena KK, Mehto S, Nath P, Chauhan NR, Sahu R, Dhar K, Das SK, Kolapalli SP, Murmu KC, Jain A, Krishna S, Sahoo BS, Chattopadhyay S, Rusten TE, Prasad P, Chauhan S, Chauhan S. Autoimmunity gene IRGM suppresses cGAS-STING and RIG-I-MAVS signaling to control interferon response. EMBO Rep 2020; 21:e50051. [PMID: 32715615 PMCID: PMC7507369 DOI: 10.15252/embr.202050051] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 06/27/2020] [Accepted: 07/02/2020] [Indexed: 12/25/2022] Open
Abstract
Activation of the type 1 interferon response is extensively connected to the pathogenesis of autoimmune diseases. Loss of function of Immunity Related GTPase M (IRGM) has also been associated to several autoimmune diseases, but its mechanism of action is unknown. Here, we found that IRGM is a master negative regulator of the interferon response. Several nucleic acid‐sensing pathways leading to interferon‐stimulated gene expression are highly activated in IRGM knockout mice and human cells. Mechanistically, we show that IRGM interacts with nucleic acid sensor proteins, including cGAS and RIG‐I, and mediates their p62‐dependent autophagic degradation to restrain interferon signaling. Further, IRGM deficiency results in defective mitophagy leading to the accumulation of defunct leaky mitochondria that release cytosolic DAMPs and mtROS. Hence, IRGM deficiency increases not only the levels of the sensors, but also those of the stimuli that trigger the activation of the cGAS‐STING and RIG‐I‐MAVS signaling axes, leading to robust induction of IFN responses. Taken together, this study defines the molecular mechanisms by which IRGM maintains interferon homeostasis and protects from autoimmune diseases.
Collapse
Affiliation(s)
- Kautilya Kumar Jena
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India.,School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Subhash Mehto
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Parej Nath
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India.,School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Nishant Ranjan Chauhan
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Rinku Sahu
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Kollori Dhar
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Saroj Kumar Das
- Centre for Biotechnology, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, India
| | | | - Krushna C Murmu
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Ashish Jain
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Sivaram Krishna
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| | | | - Soma Chattopadhyay
- Molecular Virology Lab, Department of Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Tor Erik Rusten
- Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.,Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Punit Prasad
- Epigenetic and Chromatin Biology Unit, Institute of Life Sciences, Bhubaneswar, India
| | | | - Santosh Chauhan
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
448
|
Barillé-Nion S, Lohard S, Juin PP. Targeting of BCL-2 Family Members during Anticancer Treatment: A Necessary Compromise between Individual Cell and Ecosystemic Responses? Biomolecules 2020; 10:E1109. [PMID: 32722518 PMCID: PMC7464802 DOI: 10.3390/biom10081109] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/22/2020] [Indexed: 01/07/2023] Open
Abstract
The imbalance between BCL-2 homologues and pro-death counterparts frequently noted in cancer cells endows them with a cell autonomous survival advantage. To eradicate ectopic cells, inhibitors of these homologues (BH3 mimetics) were developed to trigger, during anticancer treatment, full activation of the canonical mitochondrial apoptotic pathway and related caspases. Despite efficiency in some clinical settings, these compounds do not completely fulfill their initial promise. We herein put forth that a growing body of evidence indicates that mitochondrial integrity, controlled by BCL-2 family proteins, and downstream caspases regulate other cell death modes and influence extracellular signaling by committed cells. Moreover, intercellular communications play a key role in spreading therapeutic response across cancer cell populations and in engaging an immune response. We thus advocate that BH3 mimetics administration would be more efficient in the long term if it did not induce apoptosis in all sensitive cells at the same time, but if it could instead allow (or trigger) death signal production by non-terminally committed dying cell populations. The development of such a trade-off strategy requires to unravel the effects of BH3 mimetics not only on each individual cancer cell but also on homotypic and heterotypic cell interactions in dynamic tumor ecosystems.
Collapse
Affiliation(s)
- Sophie Barillé-Nion
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- SIRIC ILIAD, 44000 Nantes, France
| | - Steven Lohard
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- Radiation Oncology Branch, National Cancer Institute, Bethesda, MD 20892, USA
| | - Philippe P. Juin
- Centre de Recherche en Cancérologie et Immunologie Nantes Angers (CRCINA), INSERMU1232, Université de Nantes, F-44000 Nantes, France; (S.B.-N.); (S.L.)
- SIRIC ILIAD, 44000 Nantes, France
- Institut de Cancérologie de l’Ouest, 15 Rue André Boquel, 49055 Angers, France
| |
Collapse
|
449
|
Papakyrikos AM, Kim MJ, Wang X. Drosophila PTPMT1 Has a Function in Tracheal Air Filling. iScience 2020; 23:101285. [PMID: 32629421 PMCID: PMC7334580 DOI: 10.1016/j.isci.2020.101285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 05/28/2020] [Accepted: 06/14/2020] [Indexed: 01/02/2023] Open
Abstract
The fly trachea is the equivalent of the mammalian lung and is a useful model for human respiratory diseases. However, little is known about the molecular mechanisms underlying tracheal air filling during larval development. In this study, we discover that PTPMT1 has a function in tracheal air filling. PTPMT1 is a widely conserved, ubiquitously expressed mitochondrial phosphatase. To reveal PTPMT1's functions in genetically tractable invertebrates and whether those functions are tissue specific, we generate a Drosophila model of PTPMT1 depletion. We find that fly PTPMT1 mutants show impairments in tracheal air filling and subsequent activation of innate immune responses. On a cellular level, these defects are preceded by aggregation of mitochondria within the tracheal epithelial cells. Our work demonstrates a cell-type-specific role for PTPMT1 in fly tracheal epithelial cells to support air filling and to prevent immune activation. The establishment of this model will facilitate exploration of PTPMT1's physiological functions in vivo.
Collapse
Affiliation(s)
- Amanda M Papakyrikos
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA; Graduate Program in Developmental Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Min Joo Kim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Xinnan Wang
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
450
|
Abstract
Type I interferon (IFN) release by irradiated cancer cells is paramount for radiation therapy to elicit anticancer immunity. Our findings demonstrate that mitochondrial outer membrane permeabilization (MOMP) triggered by RT enables exposure of mitochondrial DNA to the cytosol, hence setting off CGAS-driven type I IFN synthesis. These data point to the existence of a therapeutically actionable mitochondrial checkpoint that restricts innate immune signaling in irradiated cancer cells.
Collapse
Affiliation(s)
- Takahiro Yamazaki
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA.,Sandra and Edward Meyer Cancer Center, New York, NY, USA.,Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.,Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA.,Université Paris Descartes/Paris V, Paris, France
| |
Collapse
|