1
|
Walter-Manucharyan M, Martin M, Pfützner J, Markert F, Rödel G, Deussen A, Hermann A, Storch A. Mitochondrial DNA replication is essential for neurogenesis but not gliogenesis in fetal neural stem cells. Dev Growth Differ 2024; 66:398-413. [PMID: 39436959 DOI: 10.1111/dgd.12946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/27/2024] [Accepted: 09/30/2024] [Indexed: 10/25/2024]
Abstract
Mitochondria are unique organelles that have their own genome (mtDNA) and perform various pivotal functions within a cell. Recently, evidence has highlighted the role of mitochondria in the process of stem cell differentiation, including differentiation of neural stem cells (NSCs). Here we studied the importance of mtDNA function in the early differentiation process of NSCs in two cell culture models: the CGR8-NS cell line that was derived from embryonic stem cells by a lineage selection technique, and primary NSCs that were isolated from embryonic day 14 mouse fetal forebrain. We detected a dramatic increase in mtDNA content upon NSC differentiation to adapt their mtDNA levels to their differentiated state, which was not accompanied by changes in mitochondrial transcription factor A expression. As chemical mtDNA depletion by ethidium bromide failed to generate living ρ° cell lines from both NSC types, we used inhibition of mtDNA polymerase-γ by 2'-3'-dideoxycytidine to reduce mtDNA replication and subsequently cellular mtDNA content. Inhibition of mtDNA replication upon NSC differentiation reduced neurogenesis but not gliogenesis. The mtDNA depletion did not change energy production/consumption or cellular reactive oxygen species (ROS) content in the NSC model used. In conclusion, mtDNA replication is essential for neurogenesis but not gliogenesis in fetal NSCs through as yet unknown mechanisms, which, however, are largely independent of energy/ROS metabolism.
Collapse
Affiliation(s)
- Meri Walter-Manucharyan
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Melanie Martin
- Department of Physiology, Technische Universität Dresden, Dresden, Germany
| | - Julia Pfützner
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
| | - Franz Markert
- Department of Neurology, University of Rostock, Rostock, Germany
| | - Gerhard Rödel
- Institute of Genetics, Technische Universität Dresden, Dresden, Germany
| | - Andreas Deussen
- Department of Physiology, Technische Universität Dresden, Dresden, Germany
| | - Andreas Hermann
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
- Center for Regenerative Therapies Dresden, Dresden, Germany
- Translational Neurodegeneration Section "Albrecht Kossel", Department of Neurology, University of Rostock, Rostock, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| | - Alexander Storch
- Division of Neurodegenerative Diseases, Department of Neurology, Technische Universität Dresden, Dresden, Germany
- Department of Neurology, University of Rostock, Rostock, Germany
- Center for Regenerative Therapies Dresden, Dresden, Germany
- German Center for Neurodegenerative Diseases (DZNE) Rostock/Greifswald, Rostock, Germany
| |
Collapse
|
2
|
Kate WD, Fanta M, Weinfeld M. Loss of the DNA repair protein, polynucleotide kinase/phosphatase, activates the type 1 interferon response independent of ionizing radiation. Nucleic Acids Res 2024; 52:9630-9653. [PMID: 39087523 PMCID: PMC11381348 DOI: 10.1093/nar/gkae654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 06/07/2024] [Accepted: 07/12/2024] [Indexed: 08/02/2024] Open
Abstract
DNA damage has been implicated in the stimulation of the type 1 interferon (T1IFN) response. Here, we show that downregulation of the DNA repair protein, polynucleotide kinase/phosphatase (PNKP), in a variety of cell lines causes robust phosphorylation of STAT1, upregulation of interferon-stimulated genes and persistent accumulation of cytosolic DNA, all of which are indicators for the activation of the T1IFN response. Furthermore, this did not require damage induction by ionizing radiation. Instead, our data revealed that production of reactive oxygen species (ROS) synergises with PNKP loss to potentiate the T1IFN response, and that loss of PNKP significantly compromises mitochondrial DNA (mtDNA) integrity. Depletion of mtDNA or treatment of PNKP-depleted cells with ROS scavengers abrogated the T1IFN response, implicating mtDNA as a significant source of the cytosolic DNA required to potentiate the T1IFN response. The STING signalling pathway is responsible for the observed increase in the pro-inflammatory gene signature in PNKP-depleted cells. While the response was dependent on ZBP1, cGAS only contributed to the response in some cell lines. Our data have implications for cancer therapy, since PNKP inhibitors would have the potential to stimulate the immune response, and also to the neurological disorders associated with PNKP mutation.
Collapse
Affiliation(s)
- Wisdom Deebeke Kate
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Mesfin Fanta
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - Michael Weinfeld
- Department of Oncology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
3
|
Tong Z, Zou JP, Wang SY, Luo WW, Wang YY. Activation of the cGAS-STING-IRF3 Axis by Type I and II Interferons Contributes to Host Defense. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308890. [PMID: 39004913 PMCID: PMC11425201 DOI: 10.1002/advs.202308890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 06/08/2024] [Indexed: 07/16/2024]
Abstract
Interferons (IFNs) activate JAK-STAT pathways to induce downstream effector genes for host defense against invaded pathogens and tumors. Here both type I (β) and II (γ) IFNs are shown that can activate the transcription factor IRF3 in parallel with STAT1. IRF3-deficiency impairs transcription of a subset of downstream effector genes induced by IFN-β and IFN-γ. Mechanistically, IFN-induced activation of IRF3 is dependent on the cGAS-STING-TBK1 axis. Both IFN-β and IFN-γ cause mitochondrial DNA release into the cytosol. In addition, IFNs induce JAK1-mediated tyrosine phosphorylation of cGAS at Y214/Y215, which is essential for its DNA binding activity and signaling. Furthermore, deficiency of cGAS, STING, or IRF3 impairs IFN-β- or IFN-γ-mediated antiviral and antitumor activities. The findings reveal a novel IRF3 activation pathway parallel with the canonical STAT1/2 activation pathways triggered by IFNs and provide an explanation for the pleiotropic roles of the cGAS-STING-IRF3 axis in host defense.
Collapse
Affiliation(s)
- Zhen Tong
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Bejing, 100049, China
| | - Jia-Peng Zou
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Bejing, 100049, China
| | - Su-Yun Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Wei-Wei Luo
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Bejing, 100049, China
- Hubei Jiangxia Laboratory, Wuhan, Hubei, 430200, China
| | - Yan-Yi Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Center for Biosafety Mega-Science, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Bejing, 100049, China
| |
Collapse
|
4
|
Huang X, Liang N, Zhang F, Lin W, Ma W. Lovastatin-Induced Mitochondrial Oxidative Stress Leads to the Release of mtDNA to Promote Apoptosis by Activating cGAS-STING Pathway in Human Colorectal Cancer Cells. Antioxidants (Basel) 2024; 13:679. [PMID: 38929118 PMCID: PMC11200898 DOI: 10.3390/antiox13060679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/27/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Statins are 3-hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase inhibitors widely used in the treatment of hyperlipidemia. The inhibition of HMG-CoA reductase in the mevalonate pathway leads to the suppression of cell proliferation and induction of apoptosis. The cyclic GMP-AMP synthase (cGAS) stimulator of the interferon genes (STING) signaling pathway has been suggested to not only facilitate inflammatory responses and the production of type I interferons (IFN), but also activate other cellular processes, such as apoptosis. It has not been studied, however, whether cGAS-STING activation is involved in the apoptosis induced by statin treatment in human colorectal cancer cells. In this study, we reported that lovastatin impaired mitochondrial function, including the depolarization of mitochondrial membrane potential, reduction of oxygen consumption, mitochondrial DNA (mtDNA) integrity, and mtDNA abundance in human colorectal cancer HCT116 cells. The mitochondrial dysfunction markedly induced ROS production in mitochondria, whereas the defect in mitochondria respiration or depletion of mitochondria eliminated reactive oxygen species (ROS) production. The ROS-induced oxidative DNA damage by lovastatin treatment was attenuated by mitochondrial-targeted antioxidant mitoquinone (mitoQ). Upon DNA damage, mtDNA was released into the cytosol and bound to DNA sensor cGAS, thus activating the cGAS-STING signaling pathway to trigger a type I interferon response. This effect was not activated by nuclear DNA (nuDNA) or mitochondrial RNA, as the depletion of mitochondria compromised this effect, but not the knockdown of retinoic acid-inducible gene-1/melanoma differentiation-associated protein 5 (RIG-I/MDA5) adaptor or mitochondrial antiviral signaling protein (MAVS). Moreover, lovastatin-induced apoptosis was partly dependent on the cGAS-STING signaling pathway in HCT116 cells as the knockdown of cGAS or STING expression rescued cell viability and mitigated apoptosis. Similarly, the knockdown of cGAS or STING also attenuated the antitumor effect of lovastatin in the HCT116 xenograft model in vivo. Our findings suggest that lovastatin-induced apoptosis is at least partly mediated through the cGAS-STING signaling pathway by triggering mtDNA accumulation in the cytosol in human colorectal cancer HCT116 cells.
Collapse
Affiliation(s)
- Xiaoming Huang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Ning Liang
- College of Physics and Optoelectronic Engineering, Shenzhen University, Shenzhen 518060, China
- School of Pharmacy, Shenzhen University Medical School, Shenzhen University, Shenzhen 518055, China
| | - Fuming Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Wanjun Lin
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| | - Wenzhe Ma
- State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 999078, China
| |
Collapse
|
5
|
Hagen JT, Montgomery MM, Aruleba RT, Chrest BR, Green TD, Kassai M, Zeczycki TN, Schmidt CA, Bhowmick D, Tan SF, Feith DJ, Chalfant CE, Loughran TP, Liles D, Minden MD, Schimmer AD, Cabot MC, Mclung JM, Fisher-Wellman KH. Mitochondria inside acute myeloid leukemia cells hydrolyze ATP to resist chemotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589110. [PMID: 38659944 PMCID: PMC11042215 DOI: 10.1101/2024.04.12.589110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Despite early optimism, therapeutics targeting oxidative phosphorylation (OxPhos) have faced clinical setbacks, stemming from their inability to distinguish healthy from cancerous mitochondria. Herein, we describe an actionable bioenergetic mechanism unique to cancerous mitochondria inside acute myeloid leukemia (AML) cells. Unlike healthy cells which couple respiration to the synthesis of ATP, AML mitochondria were discovered to support inner membrane polarization by consuming ATP. Because matrix ATP consumption allows cells to survive bioenergetic stress, we hypothesized that AML cells may resist cell death induced by OxPhos damaging chemotherapy by reversing the ATP synthase reaction. In support of this, targeted inhibition of BCL-2 with venetoclax abolished OxPhos flux without impacting mitochondrial membrane potential. In surviving AML cells, sustained polarization of the mitochondrial inner membrane was dependent on matrix ATP consumption. Mitochondrial ATP consumption was further enhanced in AML cells made refractory to venetoclax, consequential to downregulations in both the proton-pumping respiratory complexes, as well as the endogenous F1-ATPase inhibitor ATP5IF1. In treatment-naive AML, ATP5IF1 knockdown was sufficient to drive venetoclax resistance, while ATP5IF1 overexpression impaired F1-ATPase activity and heightened sensitivity to venetoclax. Collectively, our data identify matrix ATP consumption as a cancer-cell intrinsic bioenergetic vulnerability actionable in the context of mitochondrial damaging chemotherapy.
Collapse
Affiliation(s)
- James T Hagen
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Mclane M Montgomery
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Raphael T Aruleba
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Brett R Chrest
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Thomas D Green
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
| | - Miki Kassai
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Tonya N Zeczycki
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Cameron A Schmidt
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
- Department of Biology, East Carolina University, Greenville, NC
| | - Debajit Bhowmick
- Flow Cytometry Core Facility, Brody School of Medicine at East Carolina University, Greenville, NC
| | - Su-Fern Tan
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
- University of Virginia Cancer Center, Charlottesville, VA
| | - David J Feith
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
- University of Virginia Cancer Center, Charlottesville, VA
| | - Charles E Chalfant
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
- University of Virginia Cancer Center, Charlottesville, VA
- Department of Cell Biology, University of Virginia, Charlottesville, VA
- Research Service, Richmond Veterans Administration Medical Center, Richmond, VA
| | - Thomas P Loughran
- Department of Medicine, Hematology/Oncology, University of Virginia School of Medicine, Charlottesville, VA
- University of Virginia Cancer Center, Charlottesville, VA
| | - Darla Liles
- Department of Internal Medicine, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Mark D Minden
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Aaron D Schimmer
- Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Myles C Cabot
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
- Department of Biochemistry and Molecular Biology, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Joseph M Mclung
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
- Department of Cardiovascular Sciences, Brody School of Medicine, East Carolina University, Greenville, NC
| | - Kelsey H Fisher-Wellman
- Department of Physiology, Brody School of Medicine, East Carolina University, Greenville, NC
- East Carolina Diabetes and Obesity Institute, East Carolina University, Greenville, NC
- UNC Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC
| |
Collapse
|
6
|
Zhang Y, Guo D, Zhu Y, Liu L. Inhibition of mitochondrial function by approved drugs overcomes nasopharyngeal carcinoma chemoresistance. Anticancer Drugs 2024; 35:317-324. [PMID: 38215016 DOI: 10.1097/cad.0000000000001566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2024]
Abstract
The development of chemo-resistance in nasopharyngeal carcinoma (NPC) presents a significant therapeutic challenge, and its underlying mechanisms remain poorly understood. In our previous studies, we highlighted the association between isoprenylcysteine carboxylmethyltransferase (ICMT) and chemoresistance in NPC. In this current research, we revealed that both 5-FU and cisplatin-resistant NPC cells exhibited elevated mitochondrial function and increased expression of mitochondrial genes, independent of ICMT. Our investigations further showed that classic mitochondrial inhibitors, such as oligomycin, antimycin, and rotenone, were notably more effective in reducing viability in chemo-resistant NPC cells compared to parental cells. Moreover, we identified two antimicrobial drugs, tigecycline and atovaquone, recognized as mitochondrial inhibitors, as potent agents for decreasing chemo-resistant NPC cells by targeting mitochondrial respiration. Remarkably, tigecycline and atovaquone, administered at tolerable doses, inhibited chemo-resistant NPC growth in mouse models and extended overall survival rates. This work unveils the efficacy of mitochondrial inhibition as a promising strategy to overcome chemo-resistance in NPC. Additionally, our findings highlight the potential repurposing of clinically available drugs like tigecycline and atovaquone for treating NPC patients who develop chemoresistance.
Collapse
Affiliation(s)
- Yunlong Zhang
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University
| | - Difeng Guo
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei Province, China
| | - Yongbo Zhu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University
| | - Lin Liu
- Department of Otolaryngology Head and Neck Surgery, The First Affiliated Hospital of Yangtze University
| |
Collapse
|
7
|
Shang R, Liao Y, Zheng X. Inhibition of Wnt Signaling by Atovaquone Inhibits Gastric Cancer and Enhances Chemotherapy Effectiveness Through Activation of Casein Kinase 1α. Nutr Cancer 2024; 76:452-462. [PMID: 38494910 DOI: 10.1080/01635581.2024.2328377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/03/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Abnormal activation of the Wnt/β-catenin signaling pathway is a driving force behind the progression of gastric cancer. Atovaquone, known as an antimalarial drug, has emerged as a potential candidate for anti-cancer therapy. This study investigated atovaquone's effects on gastric cancer and its underlying mechanisms. Using gastric cancer cell lines, we found that atovaquone, at concentrations relevant to clinical use, significantly reduced their viability. Notably, atovaquone exhibited a lower effectiveness in reducing the viability of normal gastric cells compared to gastric cancer cells. We further demonstrated that atovaquone inhibited gastric cancer growth and colony formation. Mechanism studies revealed that atovaquone inhibited mitochondrial respiration and induced oxidative stress. Experiments using ρ0 cells, deficient in mitochondrial respiration, indicated a slightly weaker effect of atovaquone on inducing apoptosis compared to wildtype cells. Atovaquone increased phosphorylated β-catenin at Ser45 and Ser33/37/Thr41, elevated Axin, and reduced β-catenin. The inhibitory effects of atovaquone on β-catenin were reversed upon depletion of CK1α. Furthermore, the combination of atovaquone with paclitaxel suppressed gastric cancer growth and improved overall survival in mice. Given that atovaquone is already approved for clinical use, these findings suggest its potential as a valuable addition to the drug arsenal available for treating gastric cancer.
Collapse
Affiliation(s)
- Rui Shang
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Yingying Liao
- Department of Gastroenterology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Xuejiao Zheng
- Department of Pharmacy, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| |
Collapse
|
8
|
Wang K, Du Y, Li P, Guan C, Zhou M, Wu L, Liu Z, Huang Z. Nanoplastics causes heart aging/myocardial cell senescence through the Ca 2+/mtDNA/cGAS-STING signaling cascade. J Nanobiotechnology 2024; 22:96. [PMID: 38448951 PMCID: PMC10918962 DOI: 10.1186/s12951-024-02375-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/26/2024] [Indexed: 03/08/2024] Open
Abstract
BACKGROUND Nanoplastics (NPs) are now a new class of pollutants widely present in the soil, atmosphere, freshwater and marine environments. Nanoplastics can rapidly penetrate cell membranes and accumulate in human tissues and organs, thus posing a potential threat to human health. The heart is the main power source of the body. But up to now, the toxicological effects of long-term exposure to nanoplastics on the heart has not been revealed yet. RESULTS We evaluated the effects of long term exposure of nanoplastics on cardiac cell/tissue in vitro and in vivo model. Furthermore, we explored the molecular mechanism by which nanoplastics exposure causes myocardial cell senescence. Immunohistochemistry, indirect immunofluorescence and ELISA were performed to detect the effects of nanoplastics on heart aging. We found that nanoplastics were able to induce significant cardiac aging through a series of biochemical assays in vivo. In vitro, the effects of nanoplastics on cardiac cell were investigated, and found that nanoplastics were able to internalize into cardiomyocytes in time and dose-dependant manner. Further biochemical analysis showed that nanoplastics induces cardiomyocytes senescence by detecting a series of senescence marker molecules. Molecular mechanism research shows that nanoplastics may cause mitochondrial destabilization by inducing oxidative stress, which leads to the leakage of mtDNA from mitochondria into the cytoplasm, and then cytoplasm-localized mt-DNA activates the cGAS-STING signaling pathway and promotes inflammation response, ultimately inducing cardiomyocytes senescence. CONCLUSIONS In this work, we found that nanoplastics exposure induces premature aging of heart. Current research also reveals the molecular mechanism by which nanoplastics induces cardiomyocyte senescence. This study laid the foundation for further studying the potential harm of nanoplastics exposure on heart.
Collapse
Affiliation(s)
- Kaihao Wang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yipeng Du
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Peixin Li
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Chang Guan
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Min Zhou
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Lanlan Wu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zengfu Liu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Zheng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
9
|
Zhou Y, Jiao J, Yang R, Wen B, Wu Q, Xu L, Tong X, Yan H. Temozolomide-based sonodynamic therapy induces immunogenic cell death in glioma. Clin Immunol 2023; 256:109772. [PMID: 37716612 DOI: 10.1016/j.clim.2023.109772] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 08/10/2023] [Accepted: 09/11/2023] [Indexed: 09/18/2023]
Abstract
BACKGROUND In our previous study, we found for the first time that temozolomide (TMZ), the first-line chemotherapeutic agent for glioblastoma (GBM), can generate a large amount of reactive oxygen species (ROS) under ultrasound irradiation. Sonodynamic therapy (SDT) using TMZ as the sonosensitizer produced more potent antitumor effects than TMZ alone. Here, we further evaluate the effects of TMZ-based SDT on subcellular structures and investigate the immunogenic cell death (ICD)-inducing capability of TMZ-based SDT. METHODS The sonotoxic effects of TMZ were explored in LN229 and GL261 glioma cells. The morphology of endoplasmic reticulum and mitochondria was observed by transmission electron microscopy. The nuclear DNA damage was represented by γ-H2AX staining. Bone marrow-derived dendritic cells (BMDCs) were employed to assess ICD-inducing capability of TMZ-based SDT. A cyclic arginine-glycine-aspartic (c(RGDyC))-modified nanoliposome drug delivery platform was used to improve the tumor targeting of SDT. RESULTS TMZ-based SDT had a greater inhibitory effect on glioma cells than TMZ alone. Transmission electron microscopy revealed that TMZ-based SDT caused endoplasmic reticulum dilation and mitochondrial swelling. In addition, endoplasmic reticulum stress response (ERSR), nuclear DNA damage and mitochondrial permeability transition pore (mPTP) opening were promoted in TMZ-based SDT group. Most importantly, we found that TMZ-based SDT could promote the "danger signals" produced by glioma cells and induce the maturation and activation of BMDCs, which was associated with the mitochondrial DNA released into the cytoplasm in glioma cells. In vivo experiments showed that TMZ-based SDT could remodel glioma immune microenvironment and provoke durable and powerful anti-tumor immune responses. What's more, the engineered nanoliposome vector of TMZ conferred SDT tumor targeting, providing an option for safer clinical application of TMZ in combination with SDT in the future. CONCLUSIONS TMZ-based SDT was capable of triggering ICD in glioma. The discovery of TMZ as a sonosensitizer have shown great promise in the treatment of GBM.
Collapse
Affiliation(s)
- Yan Zhou
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300350, China
| | - Jiji Jiao
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300350, China
| | - Rongyan Yang
- College of Environmental Science and Engineering of Nankai University, Tianjin 300350, China
| | - Binli Wen
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300350, China
| | - Qiaoli Wu
- Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China
| | - Lixia Xu
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.
| | - Xiaoguang Tong
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.
| | - Hua Yan
- Clinical College of Neurology, Neurosurgery and Neurorehabilitation, Tianjin Medical University, Tianjin 300350, China; Tianjin Key Laboratory of Cerebral Vascular and Neurodegenerative Diseases, Tianjin Neurosurgical Institute, Tianjin Huanhu Hospital, Tianjin 300350, China.
| |
Collapse
|
10
|
Konaka H, Kato Y, Hirano T, Tsujimoto K, Park J, Koba T, Aoki W, Matsuzaki Y, Taki M, Koyama S, Itotagawa E, Jo T, Hirayama T, Kawai T, Ishii KJ, Ueda M, Yamaguchi S, Akira S, Morita T, Maeda Y, Nishide M, Nishida S, Shima Y, Narazaki M, Takamatsu H, Kumanogoh A. Secretion of mitochondrial DNA via exosomes promotes inflammation in Behçet's syndrome. EMBO J 2023; 42:e112573. [PMID: 37661814 PMCID: PMC10577637 DOI: 10.15252/embj.2022112573] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 05/21/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Mitochondrial DNA (mtDNA) leakage into the cytoplasm can occur when cells are exposed to noxious stimuli. Specific sensors recognize cytoplasmic mtDNA to promote cytokine production. Cytoplasmic mtDNA can also be secreted extracellularly, leading to sterile inflammation. However, the mode of secretion of mtDNA out of cells upon noxious stimuli and its relevance to human disease remain unclear. Here, we show that pyroptotic cells secrete mtDNA encapsulated within exosomes. Activation of caspase-1 leads to mtDNA leakage from the mitochondria into the cytoplasm via gasdermin-D. Caspase-1 also induces intraluminal membrane vesicle formation, allowing for cellular mtDNA to be taken up and secreted as exosomes. Encapsulation of mtDNA within exosomes promotes a strong inflammatory response that is ameliorated upon exosome biosynthesis inhibition in vivo. We further show that monocytes derived from patients with Behçet's syndrome (BS), a chronic systemic inflammatory disorder, show enhanced caspase-1 activation, leading to exosome-mediated mtDNA secretion and similar inflammation pathology as seen in BS patients. Collectively, our findings support that mtDNA-containing exosomes promote inflammation, providing new insights into the propagation and exacerbation of inflammation in human inflammatory diseases.
Collapse
Affiliation(s)
- Hachiro Konaka
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
- Department of Internal MedicineNippon Life HospitalOsakaJapan
| | - Yasuhiro Kato
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
- Department of Advanced Clinical and Translational Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Toru Hirano
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Nishinomiya Municipal Central HospitalNishinomiyaJapan
| | - Kohei Tsujimoto
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
- Department of Advanced Clinical and Translational Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - JeongHoon Park
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Internal MedicineDaini Osaka Police HospitalOsakaJapan
| | - Taro Koba
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Wataru Aoki
- Division of Applied Life Sciences, Graduate School of AgricultureKyoto UniversityKyotoJapan
| | - Yusei Matsuzaki
- Division of Applied Life Sciences, Graduate School of AgricultureKyoto UniversityKyotoJapan
| | - Masayasu Taki
- Institute of Transformative Bio‐Molecules (WPI‐ITbM), Nagoya UniversityNagoyaJapan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
| | - Eri Itotagawa
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
| | - Tatsunori Jo
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
| | - Takehiro Hirayama
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
| | - Taro Kawai
- Laboratory of Molecular Immunobiology, Division of Biological Science, Graduate School of Science and TechnologyNara Institute of Science and Technology (NAIST)IkomaJapan
| | - Ken J Ishii
- Division of Vaccine ScienceThe Institute of Medical Science, The University of TokyoTokyoJapan
| | - Mitsuyoshi Ueda
- Division of Applied Life Sciences, Graduate School of AgricultureKyoto UniversityKyotoJapan
| | - Shigehiro Yamaguchi
- Institute of Transformative Bio‐Molecules (WPI‐ITbM), Nagoya UniversityNagoyaJapan
| | - Shizuo Akira
- Laboratory of Host Defense, Immunology Frontier Research Center (IFReC)Osaka UniversityOsakaJapan
| | - Takayoshi Morita
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
| | - Yuichi Maeda
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Masayuki Nishide
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
| | - Sumiyuki Nishida
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Yoshihito Shima
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Division of Thermo‐Therapeutics for Vascular Dysfunction, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Masashi Narazaki
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Advanced Clinical and Translational Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
| | - Hyota Takamatsu
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
- Department of Clinical Research CenterNational Hospital Organization Osaka Minami Medical CenterOsakaJapan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of MedicineOsaka UniversityOsakaJapan
- Department of Immunopathology, Immunology Frontier Research Center (iFReC)Osaka UniversityOsakaJapan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research InitiativesOsaka UniversityOsakaJapan
- Center for Infectious Disease for Education and Research (CiDER)Osaka UniversityOsakaJapan
| |
Collapse
|
11
|
Liu G, Chen ZG, Yang LR, Rong YX, Wang Q, Li L, Lu QW, Jiang MD, Qi HY. Z-ligustilide preferentially caused mitochondrial dysfunction in AML HL-60 cells by activating nuclear receptors NUR77 and NOR1. Chin Med 2023; 18:123. [PMID: 37735686 PMCID: PMC10512564 DOI: 10.1186/s13020-023-00808-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 07/18/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND Nuclear receptors NUR77 and NOR1 were identified as critical targets in acute myeloid leukemia (AML) therapy. Previously, we showed that Z-ligustilide (Z-LIG) selectively targeted AML by restoring NUR77 and NOR1. However, its downstream mechanisms are yet to be elucidated. METHODS SRB staining assay was used to measure cell viability. Cell apoptosis, mitochondrial membrane potential and mitochondrial reactive oxygen species were analyzed using flow cytometry. The potential targets of Z-LIG in AML HL-60 cells were evaluated by RNA sequencing. Changes in RNA levels were measured using quantitative RT-qPCR and western blot analysis was used to detect the expression of proteins. RESULTS Z-LIG preferentially induced mitochondrial dysfunction in HL-60 cells compared with 293T cells. Furthermore, RNA sequencing revealed that mitochondrial transcription and translation might be potential Z-LIG targets inhibiting HL-60 cells. NUR77/NOR1 overexpression significantly reduced the mitochondrial ATP and mitochondrial membrane potential and increased mitochondrial reactive oxygen species in HL-60 cells but not in 293T cells. Moreover, Z-LIG induced mitochondrial dysfunction by restoring NUR77 and NOR1 in HL-60 cells. Compared with HL-60 cells, the apoptosis-inducing activities of NUR77/NOR1 and Z-LIG were significantly reduced in HL-60 ρ0 cells depleted in mitochondrial DNA (mt-DNA). Moreover, NUR77/NOR1 and Z-LIG downregulated mitochondrial transcription and translation related proteins in HL-60 cells. Notably, Z-LIG remarkably reduced mitochondrial ATP in primary AML cells and showed anti-AML activity in mouse models of human AML. CONCLUSIONS Collectively, our findings suggested that Z-LIG selectively induces mitochondrial dysfunction in AML HL-60 cells by restoring NUR77 and NOR1, a process associated with interference in mtDNA transcription.
Collapse
Affiliation(s)
- Gen Liu
- College of Pharmaceutical Sciences, College of Chinese Medicine, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Zhi-Gang Chen
- College of Pharmaceutical Sciences, College of Chinese Medicine, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Li-Rong Yang
- College of Pharmaceutical Sciences, College of Chinese Medicine, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Yu-Xia Rong
- College of Pharmaceutical Sciences, College of Chinese Medicine, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Qin Wang
- College of Pharmaceutical Sciences, College of Chinese Medicine, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Li Li
- College of Pharmaceutical Sciences, College of Chinese Medicine, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing, 400715, China
| | - Qian-Wei Lu
- Radiotherapy Department, Chongqing Ninth People's Hospital, Chongqing, China
| | - Ming-Dong Jiang
- Radiotherapy Department, Chongqing Ninth People's Hospital, Chongqing, China
| | - Hong-Yi Qi
- College of Pharmaceutical Sciences, College of Chinese Medicine, Southwest University, 2 Tiansheng Road, Beibei District, Chongqing, 400715, China.
| |
Collapse
|
12
|
Ouyang W, Wang S, Yan D, Wu J, Zhang Y, Li W, Hu J, Liu Z. The cGAS-STING pathway-dependent sensing of mitochondrial DNA mediates ocular surface inflammation. Signal Transduct Target Ther 2023; 8:371. [PMID: 37735446 PMCID: PMC10514335 DOI: 10.1038/s41392-023-01624-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/21/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
The innate immune response is the main pathophysiological process of ocular surface diseases exposed to multiple environmental stresses. The epithelium is central to the innate immune response, but whether and how innate immunity is initiated by ocular epithelial cells in response to various environmental stresses in ocular surface diseases, such as dry eye, is still unclear. By utilizing two classic experimental dry eye models-a mouse ocular surface treated with benzalkonium chloride (BAC) and a mouse model with surgically removed extraorbital lachrymal glands, as well as dry eye patient samples-along with human corneal epithelial cells (HCE) exposed to hyperosmolarity, we have discovered a novel innate immune pathway in ocular surface epithelial cells. Under stress, mitochondrial DNA (mtDNA) was released into the cytoplasm through the mitochondrial permeability transition pore (mPTP) and further activated the cyclic GMP-AMP synthase (cGAS)-stimulator of interferon genes (STING) pathway, aggravating downstream inflammatory responses and ocular surface damage. Genetic deletion or pharmacological suppression of STING and inhibition of mtDNA release reduced inflammatory responses, whereas mtDNA transfection supported cytoplasmic mtDNA-induced inflammatory responses by activating the cGAS-STING pathway. Our study clarified the cGAS-STING pathway-dependent sensing of mitochondrial DNA-mediated ocular surface inflammation, which elucidated a new mechanism of ocular surface diseases in response to multiple environmental stresses.
Collapse
Affiliation(s)
- Weijie Ouyang
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China
| | - Shoubi Wang
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361005, China
| | - Dan Yan
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China
| | - Jieli Wu
- Changsha Aier Eye Hospital, Changsha, Hunan, 410016, China
| | - Yunuo Zhang
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China
| | - Wei Li
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China
| | - Jiaoyue Hu
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China.
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China.
| | - Zuguo Liu
- Xiamen University affiliated Xiamen Eye Center; Fujian Provincial Key Laboratory of Ophthalmology and Visual Science; Fujian Engineering and Research Center of Eye Regenerative Medicine; Eye Institute of Xiamen University; School of Medicine, Xiamen University, Xiamen, Fujian, 361005, China.
- Department of Ophthalmology, Xiang'an Hospital of Xiamen University, Xiamen, Fujian, 361005, China.
- Department of Ophthalmology, The First Affiliated Hospital of University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
13
|
Udeochu JC, Amin S, Huang Y, Fan L, Torres ERS, Carling GK, Liu B, McGurran H, Coronas-Samano G, Kauwe G, Mousa GA, Wong MY, Ye P, Nagiri RK, Lo I, Holtzman J, Corona C, Yarahmady A, Gill MT, Raju RM, Mok SA, Gong S, Luo W, Zhao M, Tracy TE, Ratan RR, Tsai LH, Sinha SC, Gan L. Tau activation of microglial cGAS-IFN reduces MEF2C-mediated cognitive resilience. Nat Neurosci 2023; 26:737-750. [PMID: 37095396 PMCID: PMC10166855 DOI: 10.1038/s41593-023-01315-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 03/20/2023] [Indexed: 04/26/2023]
Abstract
Pathological hallmarks of Alzheimer's disease (AD) precede clinical symptoms by years, indicating a period of cognitive resilience before the onset of dementia. Here, we report that activation of cyclic GMP-AMP synthase (cGAS) diminishes cognitive resilience by decreasing the neuronal transcriptional network of myocyte enhancer factor 2c (MEF2C) through type I interferon (IFN-I) signaling. Pathogenic tau activates cGAS and IFN-I responses in microglia, in part mediated by cytosolic leakage of mitochondrial DNA. Genetic ablation of Cgas in mice with tauopathy diminished the microglial IFN-I response, preserved synapse integrity and plasticity and protected against cognitive impairment without affecting the pathogenic tau load. cGAS ablation increased, while activation of IFN-I decreased, the neuronal MEF2C expression network linked to cognitive resilience in AD. Pharmacological inhibition of cGAS in mice with tauopathy enhanced the neuronal MEF2C transcriptional network and restored synaptic integrity, plasticity and memory, supporting the therapeutic potential of targeting the cGAS-IFN-MEF2C axis to improve resilience against AD-related pathological insults.
Collapse
Affiliation(s)
- Joe C Udeochu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| | - Yige Huang
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Eileen Ruth S Torres
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gillian K Carling
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Bangyan Liu
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Hugo McGurran
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Guillermo Coronas-Samano
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Grant Kauwe
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Gergey Alzaem Mousa
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Pearly Ye
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Ravi Kumar Nagiri
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Iris Lo
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Julia Holtzman
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Carlo Corona
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | - Allan Yarahmady
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Michael T Gill
- The Gladstone Institute of Neurological Disease, San Francisco, CA, USA
| | - Ravikiran M Raju
- The Picower Institute of Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Newborn Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sue-Ann Mok
- Department of Biochemistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shiaoching Gong
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Mingrui Zhao
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Tara E Tracy
- Buck Institute for Research on Aging, Novato, CA, USA
| | - Rajiv R Ratan
- Burke Neurological Institute at Weill Cornell Medicine, White Plains, NY, USA
| | - Li-Huei Tsai
- The Picower Institute of Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Subhash C Sinha
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Gan
- Helen and Robert Appel Alzheimer's Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
14
|
Zhang Y, Xu Q, Sun Q, Kong R, Liu H, Yi X, Liang Z, Letcher RJ, Liu C. Ustiloxin A inhibits proliferation of renal tubular epithelial cells in vitro and induces renal injury in mice by disrupting structure and respiratory function of mitochondria. JOURNAL OF HAZARDOUS MATERIALS 2023; 448:130791. [PMID: 36706486 DOI: 10.1016/j.jhazmat.2023.130791] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/08/2023] [Accepted: 01/12/2023] [Indexed: 06/18/2023]
Abstract
Recently, we found that Ustiloxin A (UA, a mycotoxin) was widely detected in paddy environment and rice samples from several countries, and was also detected in human urine samples from China. However, the current knowledge about the health risks of UA are limited. In this research, the cytotoxicity of UA in mice renal tubular epithelial cells (mRTECs) was evaluated, and the results indicated that UA arrested cell cycle in G2/M phase via altering cellular morphology and microtubule, and inhibited the proliferation and division of mRTECs. Furthermore, UA could inhibit mitochondrial respiration via binding to the CoQ-binding site in dihydro-orotate dehydrogenase (DHODH) protein, and resulted in mitochondrial damage. These adverse effects of UA on mitochondria might be responsible for the cytotoxicity observed in vitro. In vivo, UA at concentrations that were comparable to the realistic concentrations of human exposure induced renal insufficiency in mice, and this might be associated with the renal mitochondrial damage in mice. However, exposure to UA at those realistic concentrations did not promote the progression from renal insufficiency to renal fibrosis and chronic kidney disease was not observed in mice.
Collapse
Affiliation(s)
- Yongkang Zhang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qiaolin Xu
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Qian Sun
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Ren Kong
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China
| | - Hao Liu
- College of Plant Science & Technology, Huazhong Agricultural University, Wuhan 430070, China
| | - Xun'e Yi
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Zhengqi Liang
- College of Fisheries, Huazhong Agricultural University, Wuhan 430070, China
| | - Robert J Letcher
- Departments of Chemistry and Biology, Carleton University, Ottawa K1S 5B6, ON, Canada
| | - Chunsheng Liu
- School of Environmental Studies, China University of Geosciences, Wuhan 430074, China.
| |
Collapse
|
15
|
Jahun AS, Sorgeloos F, Chaudhry Y, Arthur SE, Hosmillo M, Georgana I, Izuagbe R, Goodfellow IG. Leaked genomic and mitochondrial DNA contribute to the host response to noroviruses in a STING-dependent manner. Cell Rep 2023; 42:112179. [PMID: 36943868 DOI: 10.1016/j.celrep.2023.112179] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 10/11/2022] [Accepted: 02/12/2023] [Indexed: 03/23/2023] Open
Abstract
The cGAS-STING pathway is central to the interferon response against DNA viruses. However, recent studies are increasingly demonstrating its role in the restriction of some RNA viruses. Here, we show that the cGAS-STING pathway also contributes to the interferon response against noroviruses, currently the commonest causes of infectious gastroenteritis worldwide. We show a significant reduction in interferon-β induction and a corresponding increase in viral replication in norovirus-infected cells after deletion of STING, cGAS, or IFI16. Further, we find that immunostimulatory host genome-derived DNA and mitochondrial DNA accumulate in the cytosol of norovirus-infected cells. Lastly, overexpression of the viral NS4 protein is sufficient to drive the accumulation of cytosolic DNA. Together, our data find a role for cGAS, IFI16, and STING in the restriction of noroviruses and show the utility of host genomic DNA as a damage-associated molecular pattern in cells infected with an RNA virus.
Collapse
Affiliation(s)
- Aminu S Jahun
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK.
| | - Frederic Sorgeloos
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK; Université catholique de Louvain, de Duve Institute, MIPA-VIRO 74-49, 74 Avenue Hippocrate, B-1200 Brussels, Belgium
| | - Yasmin Chaudhry
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Sabastine E Arthur
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Myra Hosmillo
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Iliana Georgana
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Rhys Izuagbe
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK
| | - Ian G Goodfellow
- Division of Virology, Department of Pathology, University of Cambridge, Addenbrooke's Hospital Level 5, Hills Road, Cambridge CB2 0QQ, UK.
| |
Collapse
|
16
|
Mitophagy-promoting miR-138-5p promoter demethylation inhibits pyroptosis in sepsis-associated acute lung injury. Inflamm Res 2023; 72:329-346. [PMID: 36538076 DOI: 10.1007/s00011-022-01675-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 07/25/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The present study was designed to explore the potential regulatory mechanism between mitophagy and pyroptosis during sepsis-associated acute lung injury (ALI). METHODS In vitro or in vivo models of sepsis-associated ALI were established by administering lipopolysaccharide (LPS) or performing caecal ligation and puncture (CLP) surgery. Pyroptosis levels were detected by electron microscopy, immunofluorescence, flow cytometry, western blotting and immunohistochemistry. Dual-luciferase reporter gene assay was applied to verify the targeting relationship between miR-138-5p and NLRP3. Methylation-specific PCR and chromatin immunoprecipitation assays were used to determine methylation of the miR-138-5p promoter. Mitophagy levels were examined by transmission electron microscopy and western blotting. RESULTS NLRP3 inflammasome silencing alleviated alveolar macrophage (AM) pyroptosis and septic lung injury. In addition, we confirmed the direct targeting relationship between miR-138-5p and NLRP3. Overexpressed miR-138-5p alleviated AM pyroptosis and the pulmonary inflammatory response. Moreover, the decreased expression of miR-138-5p was confirmed to depend on promoter methylation, while inhibition of miR-138-5p promoter methylation attenuated AM pyroptosis and pulmonary inflammation. Here, we discovered that an increased cytoplasmic mtDNA content in sepsis-induced ALI models induced the methylation of the miR-138-5p promoter, thereby decreasing miR-138-5p expression, which may activate the NLRP3 inflammasome and trigger AM pyroptosis. Mitophagy, a form of selective autophagy that clears damaged mitochondria, reduced cytoplasmic mtDNA levels. Furthermore, enhanced mitophagy might suppress miR-138-5p promoter methylation and relieve the pulmonary inflammatory response, changes that were reversed by treatment with isolated mtDNA. CONCLUSIONS In summary, our study indicated that mitophagy induced the demethylation of the miR-138-5p promoter, which may subsequently inhibit NLRP3 inflammasome, AM pyroptosis and inflammation in sepsis-induced lung injury. These findings may provide a promising therapeutic target for sepsis-associated ALI.
Collapse
|
17
|
cGAS-STING activation contributes to podocyte injury in diabetic kidney disease. iScience 2022; 25:105145. [PMID: 36176590 PMCID: PMC9513272 DOI: 10.1016/j.isci.2022.105145] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 08/25/2022] [Accepted: 09/12/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetic kidney disease (DKD) is the leading cause of end-stage renal diseases. DKD does not have efficacious treatment. The cGAS-STING pathway is activated in podocytes at the early stage of kidney dysfunction, which is associated with the activation of STING downstream effectors TBK1 and NF-κB but not IRF3. Lipotoxicity induces mitochondrial damage and mtDNA leakage to the cytosol through Bcl-2 associated X protein (BAX) in podocytes. BAX-mediated mtDNA cytosolic leakage can activate the cGAS-STING pathway in the absence of lipotoxicity and is sufficient to cause podocyte injury. Depletion of cytosolic mtDNA, genetic STING knockdown, or pharmacological inhibition of STING or TBK1 alleviates podocyte injury and improves renal functions in cultured podocytes or mouse models of diabetes and obesity. These results suggest that the mtDNA-cGAS-STING pathway promotes podocyte injury and is a potential therapeutic target for DKD or other obesity-related kidney diseases.
Collapse
|
18
|
Yuan J, Mo L, Mo Y, Zhang Y, Zhang Y, Zhang Q. A protective role of autophagy in fine airborne particulate matter-induced apoptosis in LN-229 cells. Toxicology 2022; 477:153271. [PMID: 35872226 PMCID: PMC10825875 DOI: 10.1016/j.tox.2022.153271] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/15/2022] [Accepted: 07/20/2022] [Indexed: 01/16/2023]
Abstract
Air pollution is a public health threat and global epidemiological studies have shown that ambient air pollutants are closely related to various poor health conditions, including neurodegenerative diseases. Here, we evaluated the toxic effects and the underlying mechanisms of fine airborne particulate matter (PM2.5) on human glioblastoma LN-229 cells. Our results showed that exposure of LN-229 cells to PM2.5 (≥ 200 μg/mL) significantly reduced cell viability. PM2.5 exposure increased autophagy, apoptosis, and ROS production in the cells. Pre-treatment with a ROS scavenger, catalase, or depletion of mtDNA (ρ0 cells) abolished PM2.5-induced autophagy and apoptosis. PM2.5 exposure also activated MAPK signals in cells, which were blocked by catalase pre-treatment or mtDNA depletion. Furthermore, inhibition of JNK, but not ERK1/2 or p38, attenuated PM2.5-induced autophagy and apoptosis in cells. Finally, suppression of autophagy with Bafilomycin A1 or Beclin 1 siRNA exacerbated PM2.5-induced apoptosis, indicating a protective role of autophagy against PM2.5-induced apoptosis. Our results demonstrated that exposure of LN-229 cells to PM2.5 caused autophagy and apoptosis through PM2.5-induced ROS generation, mainly by mitochondria, and JNK activation. Autophagy may have a transient protective response in PM2.5-induced apoptosis. These findings have important implications for understanding the potential neurotoxicity of PM2.5.
Collapse
Affiliation(s)
- Jiali Yuan
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Luke Mo
- duPont Manual High School, Louisville, KY, USA
| | - Yiqun Mo
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yuanbao Zhang
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yue Zhang
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Epidemiology & Population Health, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
19
|
Tan HY, Yong YK, Xue YC, Liu H, Furihata T, Shankar EM, Ng CS. cGAS and DDX41-STING mediated intrinsic immunity spreads intercellularly to promote neuroinflammation in SOD1 ALS model. iScience 2022; 25:104404. [PMID: 35712074 PMCID: PMC9194172 DOI: 10.1016/j.isci.2022.104404] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 02/22/2022] [Accepted: 05/10/2022] [Indexed: 11/25/2022] Open
Abstract
Neuroinflammation exacerbates the progression of SOD1-driven amyotrophic lateral sclerosis (ALS), although the underlying mechanisms remain largely unknown. Herein, we demonstrate that misfolded SOD1 (SOD1Mut)-causing ALS results in mitochondrial damage, thus triggering the release of mtDNA and an RNA:DNA hybrid into the cytosol in an mPTP-independent manner to activate IRF3- and IFNAR-dependent type I interferon (IFN-I) and interferon-stimulating genes. The neuronal hyper-IFN-I and pro-inflammatory responses triggered in ALS-SOD1Mut were sufficiently robust to cause a strong physiological outcome in vitro and in vivo. cGAS/DDX41-STING-signaling is amplified in bystander cells through inter-neuronal gap junctions. Our results highlight the importance of a common DNA-sensing pathway between SOD1 and TDP-43 in influencing the progression of ALS. Constitutive basal activation of IFN-I was found in the SOD1-ALS animal model SOD1-ALS damaged mitochondria to release mtDNA and RNA:DNA to activate the STING-pathway Blocking cGAS and STING diminishes neurodegeneration in vivo in the SOD1-ALS model Connexin and pannexin channels are required to propagate neuroinflammation in SOD1-ALS
Collapse
Affiliation(s)
- Hong Yien Tan
- Laboratory Centre, Xiamen University Malaysia, Sepang, Selangor, Malaysia.,School of Traditional Chinese Medicine, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| | - Yean Kong Yong
- Laboratory Centre, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| | - Yuan Chao Xue
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.,Department of Pathology and Laboratory of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Huitao Liu
- Centre for Heart Lung Innovation, St Paul's Hospital, University of British Columbia, Vancouver, BC, Canada.,Department of Medicine, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tomomi Furihata
- Laboratory of Clinical Pharmacy and Experimental Therapeutics, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Esaki Muthu Shankar
- Infection Biology, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur, India
| | - Chen Seng Ng
- China-ASEAN College of Marine Sciences, Xiamen University Malaysia, Sepang, Selangor, Malaysia
| |
Collapse
|
20
|
Li Y, Sun Q, Chen S, Yu X, Jing H. Monensin inhibits anaplastic thyroid cancer via disrupting mitochondrial respiration and AMPK/mTOR signaling. Anticancer Agents Med Chem 2022; 22:2539-2547. [PMID: 35168524 DOI: 10.2174/1871520622666220215123620] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 01/12/2022] [Accepted: 01/12/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE The clinical management of anaplastic thyroid cancer (ATC) remains challenging and novel treatment methods are needed. Monensin is a carboxyl polyether ionophore that potently inhibits the growth of various cancer types. Our current work investigates whether monensin has selective anti-ATC activity and systematically explores its underlying mechanisms. METHODS Proliferation and apoptosis assays were performed using a panel of thyroid cancer cell lines. Mitochondrial biogenesis profiles, ATP levels, oxidative stress, AMPK and mTOR were examined in these cells after monensin treatment. RESULTS Monensin is effective to inhibit proliferation and induce apoptosis in a number of thyroid cancer cell lines. The results are consistent across cell lines of varying cellular origins and genetic mutations. Compared to other thyroid cancer cell types, ATC cell lines are the most sensitive to monensin. Of note, monensin used at our experimental concentration affects less of normal cells. Mechanistic studies reveal that monensin acts on ATC cells through disrupting mitochondrial function, inducing oxidative stress and damage, and AMPK activation-induced mTOR inhibition. We further show mitochondrial respiration is a critical target for monensin in ATC cells. CONCLUSIONS Our pre-clinical findings demonstrate the selective anti-ATC activities of monensin. This is supported by increasing evidence monensin can to be repurposed as a potential anti-cancer drug.
Collapse
Affiliation(s)
- Yanli Li
- Department of Endocrinology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Qianshu Sun
- Department of Endocrinology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Sisi Chen
- Department of Endocrinology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Xiongjie Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| | - Hongxia Jing
- Department of Ultrasound, Renmin Hospital, Hubei University of Medicine, Shiyan, Hubei Province, China
| |
Collapse
|
21
|
Davidson S, Yu CH, Steiner A, Ebstein F, Baker PJ, Jarur-Chamy V, Hrovat Schaale K, Laohamonthonkul P, Kong K, Calleja DJ, Harapas CR, Balka KR, Mitchell J, Jackson JT, Geoghegan ND, Moghaddas F, Rogers KL, Mayer-Barber KD, De Jesus AA, De Nardo D, Kile BT, Sadler AJ, Poli MC, Krüger E, Goldbach Mansky R, Masters SL. Protein kinase R is an innate immune sensor of proteotoxic stress via accumulation of cytoplasmic IL-24. Sci Immunol 2022; 7:eabi6763. [PMID: 35148201 PMCID: PMC11036408 DOI: 10.1126/sciimmunol.abi6763] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Proteasome dysfunction can lead to autoinflammatory disease associated with elevated type I interferon (IFN-αβ) and NF-κB signaling; however, the innate immune pathway driving this is currently unknown. Here, we identified protein kinase R (PKR) as an innate immune sensor for proteotoxic stress. PKR activation was observed in cellular models of decreased proteasome function and in multiple cell types from patients with proteasome-associated autoinflammatory disease (PRAAS). Furthermore, genetic deletion or small-molecule inhibition of PKR in vitro ameliorated inflammation driven by proteasome deficiency. In vivo, proteasome inhibitor-induced inflammatory gene transcription was blunted in PKR-deficient mice compared with littermate controls. PKR also acted as a rheostat for proteotoxic stress by triggering phosphorylation of eIF2α, which can prevent the translation of new proteins to restore homeostasis. Although traditionally known as a sensor of RNA, under conditions of proteasome dysfunction, PKR sensed the cytoplasmic accumulation of a known interactor, interleukin-24 (IL-24). When misfolded IL-24 egress into the cytosol was blocked by inhibition of the endoplasmic reticulum-associated degradation pathway, PKR activation and subsequent inflammatory signaling were blunted. Cytokines such as IL-24 are normally secreted from cells; therefore, cytoplasmic accumulation of IL-24 represents an internal danger-associated molecular pattern. Thus, we have identified a mechanism by which proteotoxic stress is detected, causing inflammation observed in the disease PRAAS.
Collapse
Affiliation(s)
- Sophia Davidson
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Chien-Hsiung Yu
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Annemarie Steiner
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
- Institute of Structural Biology, University Hospital Bonn, Bonn 53127, Germany
| | - Frédéric Ebstein
- University Medicine Greifswald, Institute of Medical Biochemistry and Molecular Biology, Greifswald 17475, Germany
| | - Paul J. Baker
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Valentina Jarur-Chamy
- Immunogenetics and Translational Immunology Program. Facultad de Medicina, Universidad del Desarrollo Clínica Alemana, Santiago, Chile
| | - Katja Hrovat Schaale
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Pawat Laohamonthonkul
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Klara Kong
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Dale J. Calleja
- Ubiquitin Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Cassandra R. Harapas
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Katherine R. Balka
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Jacob Mitchell
- Translational Autoinflammatory Disease Studies (TADS), Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Jacob T. Jackson
- Immunology Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Niall D. Geoghegan
- Centre for Dynamic Imaging, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Fiona Moghaddas
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| | - Kelly L. Rogers
- Centre for Dynamic Imaging, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Katrin D. Mayer-Barber
- Inflammation and Innate Immunity Unit, Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Adriana A. De Jesus
- Translational Autoinflammatory Disease Studies (TADS), Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Dominic De Nardo
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Benjamin T. Kile
- Department of Anatomy and Developmental Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Faculty of Health and Medical Sciences, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Anthony J. Sadler
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - M. Cecilia Poli
- Immunogenetics and Translational Immunology Program. Facultad de Medicina, Universidad del Desarrollo Clínica Alemana, Santiago, Chile
- Division of Pediatric Immunology, Allergy, and Rheumatology, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elke Krüger
- University Medicine Greifswald, Institute of Medical Biochemistry and Molecular Biology, Greifswald 17475, Germany
| | - Raphaela Goldbach Mansky
- Translational Autoinflammatory Disease Studies (TADS), Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, MD 20892, USA
| | - Seth L. Masters
- Inflammation Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
22
|
Kiyooka T, Ohanyan V, Yin L, Pung YF, Chen YR, Chen CL, Kang PT, Hardwick JP, Yun J, Janota D, Peng J, Kolz C, Guarini G, Wilson G, Shokolenko I, Stevens DA, Chilian WM. Mitochondrial DNA integrity and function are critical for endothelium-dependent vasodilation in rats with metabolic syndrome. Basic Res Cardiol 2022; 117:3. [PMID: 35039940 PMCID: PMC9030679 DOI: 10.1007/s00395-021-00908-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 01/31/2023]
Abstract
Endothelial dysfunction in diabetes is generally attributed to oxidative stress, but this view is challenged by observations showing antioxidants do not eliminate diabetic vasculopathy. As an alternative to oxidative stress-induced dysfunction, we interrogated if impaired mitochondrial function in endothelial cells is central to endothelial dysfunction in the metabolic syndrome. We observed reduced coronary arteriolar vasodilation to the endothelium-dependent dilator, acetylcholine (Ach), in Zucker Obese Fatty rats (ZOF, 34 ± 15% [mean ± standard deviation] 10-3 M) compared to Zucker Lean rats (ZLN, 98 ± 11%). This reduction in dilation occurred concomitantly with mitochondrial DNA (mtDNA) strand lesions and reduced mitochondrial complex activities in the endothelium of ZOF versus ZLN. To demonstrate endothelial dysfunction is linked to impaired mitochondrial function, administration of a cell-permeable, mitochondria-directed endonuclease (mt-tat-EndoIII), to repair oxidatively modified DNA in ZOF, restored mitochondrial function and vasodilation to Ach (94 ± 13%). Conversely, administration of a cell-permeable, mitochondria-directed exonuclease (mt-tat-ExoIII) produced mtDNA strand breaks in ZLN, reduced mitochondrial complex activities and vasodilation to Ach in ZLN (42 ± 16%). To demonstrate that mitochondrial function is central to endothelium-dependent vasodilation, we introduced (via electroporation) liver mitochondria (from ZLN) into the endothelium of a mesenteric vessel from ZOF and restored endothelium-dependent dilation to vasoactive intestinal peptide (VIP at 10-5 M, 4 ± 3% vasodilation before mitochondrial transfer and 48 ± 36% after transfer). Finally, to demonstrate mitochondrial function is key to endothelium-dependent dilation, we administered oligomycin (mitochondrial ATP synthase inhibitor) and observed a reduction in endothelium-dependent dilation. We conclude that mitochondrial function is critical for endothelium-dependent vasodilation.
Collapse
Affiliation(s)
- Takahiko Kiyooka
- Division of Cardiology, Tokai University Oiso Hospital, Oiso, Kanagawa, Japan
| | - Vahagn Ohanyan
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Liya Yin
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Yuh Fen Pung
- Division of Biomedical Sciences, University of Nottingham, Malaysia Campus, Selangor, Malaysia
| | - Yeong-Renn Chen
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Chwen-Lih Chen
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Patrick T Kang
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - James P Hardwick
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - June Yun
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Danielle Janota
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Joanna Peng
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Christopher Kolz
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA
| | - Giacinta Guarini
- Cardiovascular Unit, Spedali Riuniti Santa Maria Maddalena, Volterra, Italy
| | - Glenn Wilson
- Department of Biomedical Science, University of South Alabama, Mobile, USA
| | - Inna Shokolenko
- Department of Biomedical Science, University of South Alabama, Mobile, USA
| | - Donte A Stevens
- Division of Biological Sciences, University of California-San Diego, San Diego, USA
| | - William M Chilian
- Department of Integrative Medical Sciences, Northeast Ohio Medical University, Rootstown, OH, 44274, USA.
| |
Collapse
|
23
|
A novel mechanism for NLRP3 inflammasome activation. Metabol Open 2022; 13:100166. [PMID: 35198946 PMCID: PMC8844605 DOI: 10.1016/j.metop.2022.100166] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/24/2022] Open
Abstract
The NLRP3 inflammasome, as an important component of the innate immune system, plays vital roles in various metabolic disorders. It has been reported that the NLRP3 inflammasome can be activated by a broad range of distinct stimuli, such as K+ efflux, mitochondrial dysfunction, lysosomal disruption and trans-Golgi disassembly, etc. However, there has been no well-established model for NLRP3 inflammasome activation so far, especially the underlying mechanisms for mitochondria in NLRP3 inflammasome activation remain elusive. Given that K+ efflux is a widely accepted nexus for triggering activation of NLRP3 inflammasome in most previous studies, we sought to elucidate the role of mitochondria in K+ efflux-induced NLRP3 inflammasome activation. Here, we demonstrated that inflammation activation by LPS evoked the expression of genes that involved in mitochondrial biogenesis and mitophagy, subsequently mitochondrial mass and mitochondrial membrane potential were also elevated, suggesting the contribution of mitochondria in inflammatory responses. Moreover, we inhibited mitochondrial biogenesis by silencing Tfam and genetic ablation of Tfam abolished the NLRP3 inflammasome activation induced by K+ efflux via release of mitochondrial DNA (mtDNA), as deprivation of cellular mtDNA by EtBr treatment could reverse inflammasome activation induced by K+ efflux. Collectively, we reveal that mtDNA release induced by K+ efflux in macrophages activates NLRP3 inflammasome, and propose that mitochondria may serve as a potential therapeutic target for NLRP3 inflammasome-related diseases.
Collapse
|
24
|
Nguyen TTT, Shang E, Westhoff MA, Karpel-Massler G, Siegelin MD. Methodological Approaches for Assessing Metabolomic Changes in Glioblastomas. Methods Mol Biol 2022; 2445:305-328. [PMID: 34973000 DOI: 10.1007/978-1-0716-2071-7_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Glioblastoma (GBM), a highly malignant primary brain tumor, inevitably leads to death. In the last decade, a variety of novel molecular characteristics of GBMs were unraveled. The identification of the mutation in the IDH1 and less commonly IDH2 gene was surprising and ever since has nurtured research in the field of GBM metabolism. While initially thought that mutated IDH1 were to act as a loss of function mutation it became clear that it conferred the production of an oncometabolite that in turn substantially reprograms GBM metabolism. While mutated IDH1 represents truly the tip of the iceberg, there are numerous other related observations in GBM that are of significant interest to the field, including the notion that oxidative metabolism appears to play a more critical role than believed earlier. Metabolic zoning is another important hallmark of GBM since it was found that the infiltrative margin that drives GBM progression reveals enrichment of fatty acid derivatives. Consistently, fatty acid metabolism appears to be a novel therapeutic target for GBM. How metabolism in GBM intersects is another pivotal issue that appears to be important for its progression and response and resistance to therapies. In this review, we will summarize some of the most relevant findings related to GBM metabolism and cell death and how these observations are influencing the field. We will provide current approaches that are applied in the field to measure metabolomic changes in GBM models, including the detection of unlabeled and labeled metabolites as well as extracellular flux analysis.
Collapse
Affiliation(s)
- Trang T T Nguyen
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA
| | - Enyuan Shang
- Department of Biological Sciences, Bronx Community College, City University of New York, Bronx, NY, USA
| | - Mike-Andrew Westhoff
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, Ulm, Germany
| | | | - Markus D Siegelin
- Department of Pathology and Cell Biology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
25
|
Zhang Y, Mo Y, Yuan J, Zhang Y, Mo L, Zhang Q. MMP-3 activation is involved in copper oxide nanoparticle-induced epithelial-mesenchymal transition in human lung epithelial cells. Nanotoxicology 2021; 15:1380-1402. [PMID: 35108494 PMCID: PMC9484543 DOI: 10.1080/17435390.2022.2030822] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Copper oxide nanoparticles (Nano-CuO) are widely used in medical and industrial fields and our daily necessities. However, the biosafety assessment of Nano-CuO is far behind their rapid development. Here, we investigated the adverse effects of Nano-CuO on normal human bronchial epithelial BEAS-2B cells, especially determined whether Nano-CuO exposure would cause dysregulation of MMP-3, an important mediator in pulmonary fibrosis, and its potential role in epithelial-mesenchymal transition (EMT). Our results showed that exposure to Nano-CuO, but not Nano-TiO2, caused increased ROS generation, MAPKs activation, and MMP-3 upregulation. Nano-CuO-induced ROS generation was not observed in mitochondrial DNA-depleted BEAS-2B ρ0 cells, indicating that mitochondria may be the main source of Nano-CuO-induced ROS generation. Pretreatment of the cells with ROS scavengers or inhibitors or depleting mitochondrial DNA significantly attenuated Nano-CuO-induced MAPKs activation and MMP-3 upregulation, and pretreatment of cells with MAPKs inhibitors abolished Nano-CuO-induced MMP-3 upregulation, suggesting Nano-CuO-induced MMP-3 upregulation is through Nano-CuO-induced ROS generation and MAPKs activation. In addition, exposure of the cells to Nano-CuO for 48 h resulted in decreased E-cadherin expression and increased expression of vimentin, α-SMA, and fibronectin, which was ameliorated by MMP-3 siRNA transfection, suggesting an important role of MMP-3 in Nano-CuO-induced EMT. Taken together, our study demonstrated that Nano-CuO exposure caused mitochondrial ROS generation, MAPKs activation, and MMP-3 upregulation. Nano-CuO exposure also caused cells to undergo EMT, which was through Nano-CuO-induced dysregulation of ROS/MAPKs/MMP-3 pathway. Our findings will provide further understanding of the potential mechanisms involved in metal nanoparticle-induced various toxic effects including EMT and pulmonary fibrosis.
Collapse
Affiliation(s)
- Yuanbao Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yiqun Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Jiali Yuan
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Yue Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Luke Mo
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| | - Qunwei Zhang
- Department of Environmental and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, Louisville, KY, USA
| |
Collapse
|
26
|
Vowinckel J, Hartl J, Marx H, Kerick M, Runggatscher K, Keller MA, Mülleder M, Day J, Weber M, Rinnerthaler M, Yu JSL, Aulakh SK, Lehmann A, Mattanovich D, Timmermann B, Zhang N, Dunn CD, MacRae JI, Breitenbach M, Ralser M. The metabolic growth limitations of petite cells lacking the mitochondrial genome. Nat Metab 2021; 3:1521-1535. [PMID: 34799698 PMCID: PMC7612105 DOI: 10.1038/s42255-021-00477-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 09/10/2021] [Indexed: 12/25/2022]
Abstract
Eukaryotic cells can survive the loss of their mitochondrial genome, but consequently suffer from severe growth defects. 'Petite yeasts', characterized by mitochondrial genome loss, are instrumental for studying mitochondrial function and physiology. However, the molecular cause of their reduced growth rate remains an open question. Here we show that petite cells suffer from an insufficient capacity to synthesize glutamate, glutamine, leucine and arginine, negatively impacting their growth. Using a combination of molecular genetics and omics approaches, we demonstrate the evolution of fast growth overcomes these amino acid deficiencies, by alleviating a perturbation in mitochondrial iron metabolism and by restoring a defect in the mitochondrial tricarboxylic acid cycle, caused by aconitase inhibition. Our results hence explain the slow growth of mitochondrial genome-deficient cells with a partial auxotrophy in four amino acids that results from distorted iron metabolism and an inhibited tricarboxylic acid cycle.
Collapse
Affiliation(s)
- Jakob Vowinckel
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- Biognosys AG, Schlieren, Switzerland
| | - Johannes Hartl
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Biochemistry, Berlin, Germany
| | - Hans Marx
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Martin Kerick
- Sequencing Core Facility, Max Planck Institute for Molecular Genetics and Max Planck Unit for the Science of Pathogens, Berlin, Germany
- Institute of Parasitology and Biomedicine 'López-Neyra' (IPBLN, CSIC), Granada, Spain
| | - Kathrin Runggatscher
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Markus A Keller
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- Institute of Human Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Michael Mülleder
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Biochemistry, Berlin, Germany
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Jason Day
- Department of Earth Sciences, University of Cambridge, Cambridge, UK
| | - Manuela Weber
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Mark Rinnerthaler
- Department of Biosciences, University of Salzburg, Salzburg, Austria
| | - Jason S L Yu
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Simran Kaur Aulakh
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK
| | - Andrea Lehmann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Biochemistry, Berlin, Germany
| | - Diethard Mattanovich
- Department of Biotechnology, University of Natural Resources and Life Sciences Vienna, Vienna, Austria
| | - Bernd Timmermann
- Sequencing Core Facility, Max Planck Institute for Molecular Genetics and Max Planck Unit for the Science of Pathogens, Berlin, Germany
| | - Nianshu Zhang
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Cory D Dunn
- Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
- Department of Molecular Biology and Genetics, Koç University, İstanbul, Turkey
| | - James I MacRae
- Metabolomics Laboratory, The Francis Crick Institute, London, UK
| | | | - Markus Ralser
- Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK.
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Biochemistry, Berlin, Germany.
- The Molecular Biology of Metabolism Laboratory, The Francis Crick Institute, London, UK.
| |
Collapse
|
27
|
Weidling IW, Wilkins HM, Koppel SJ, Hutfles L, Wang X, Kalani A, Menta BW, Ryan B, Perez-Ortiz J, Gamblin TC, Swerdlow RH. Mitochondrial DNA Manipulations Affect Tau Oligomerization. J Alzheimers Dis 2021; 77:149-163. [PMID: 32804126 PMCID: PMC7962146 DOI: 10.3233/jad-200286] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND Mitochondrial dysfunction and tau aggregation occur in Alzheimer's disease (AD), and exposing cells or rodents to mitochondrial toxins alters their tau. OBJECTIVE To further explore how mitochondria influence tau, we measured tau oligomer levels in human neuronal SH-SY5Y cells with different mitochondrial DNA (mtDNA) manipulations. METHODS Specifically, we analyzed cells undergoing ethidium bromide-induced acute mtDNA depletion, ρ0 cells with chronic mtDNA depletion, and cytoplasmic hybrid (cybrid) cell lines containing mtDNA from AD subjects. RESULTS We found cytochrome oxidase activity was particularly sensitive to acute mtDNA depletion, evidence of metabolic re-programming in the ρ0 cells, and a relatively reduced mtDNA content in cybrids generated through AD subject mitochondrial transfer. In each case tau oligomer levels increased, and acutely depleted and AD cybrid cells also showed a monomer to oligomer shift. CONCLUSION We conclude a cell's mtDNA affects tau oligomerization. Overlapping tau changes across three mtDNA-manipulated models establishes the reproducibility of the phenomenon, and its presence in AD cybrids supports its AD-relevance.
Collapse
Affiliation(s)
- Ian W Weidling
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.,Molecular and Integrative Physiology, and University of Kansas Medical Center, Kansas City, KS, USA
| | - Heather M Wilkins
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Scott J Koppel
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.,Molecular and Integrative Physiology, and University of Kansas Medical Center, Kansas City, KS, USA
| | - Lewis Hutfles
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA
| | - Xiaowan Wang
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Anuradha Kalani
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Blaise W Menta
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.,Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Benjamin Ryan
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Judit Perez-Ortiz
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA
| | - T Chris Gamblin
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, USA
| | - Russell H Swerdlow
- University of Kansas Alzheimer's Disease Center; the University of Kansas Medical Center, Kansas City, KS, USA.,Departments of Neurology, University of Kansas Medical Center, Kansas City, KS, USA.,Molecular and Integrative Physiology, and University of Kansas Medical Center, Kansas City, KS, USA.,Biochemistry and Molecular Biology, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
28
|
Zhang C, Deng Q, Bao S, Zhu J. Anisomycin is active in preclinical models of pediatric acute myeloid leukemia via specifically inhibiting mitochondrial respiration. J Bioenerg Biomembr 2021; 53:693-701. [PMID: 34468904 DOI: 10.1007/s10863-021-09918-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/17/2021] [Indexed: 01/07/2023]
Abstract
The poor outcomes in acute myeloid leukemia (AML) necessitate new treatments. In this work, we identified that anisomycin is a potential selective anti-AML candidate, particularly for those with FLT3-ITD mutation. We found that anisomycin potently inhibited proliferation and induced apoptosis in multiple AML cell lines. Anisomycin was effective in targeting progenitor cells isolated from all tested pediatric AML patients, while sparing normal counterparts. Using AML xenograft mouse models, anisomycin exhibited inhibitory effect on tumor growth throughout the whole duration without causing toxicity in mice. The combination of anisomycin with standard of care drugs is synergistic and selective in AML cell culture system and mouse model. In addition, FLT3-ITD cells were more sensitive to anisomycin than FLT3 WT cells. Mechanistic studies revealed that anisomycin acted on AML in a p38-independent manner. We found that anisomycin decreased mitochondrial respiration by disrupting complex I activity, leading to intracellular oxidative stress. AML ρ0 cells that lack of mitochondrial respiration exhibited resistance to anisomycin. Finally, we showed that mitochondrial biogenesis contributes to differential sensitivity of FLT3-ITD and FLT3 WT cells to anisomycin. Our work is the first to systematically demonstrate that anisomycin is a useful addition to the treatment armamentarium for AML. Our findings highlight the therapeutic value of mitochondrial respiration inhibition in AML patients harboring FLT3-ITD mutation.
Collapse
Affiliation(s)
- Chuang Zhang
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, No.15 Jiefang Road, Fancheng District, Xiangyang, 441000, Hubei, China
| | - Qian Deng
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, No.15 Jiefang Road, Fancheng District, Xiangyang, 441000, Hubei, China
| | - Shiwei Bao
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, No.15 Jiefang Road, Fancheng District, Xiangyang, 441000, Hubei, China.
| | - Juanjuan Zhu
- Department of Pediatrics, Xiangyang No.1 People's Hospital, Hubei University of Medicine, No.15 Jiefang Road, Fancheng District, Xiangyang, 441000, Hubei, China.
| |
Collapse
|
29
|
Li J, Min Y. Pre-clinical evidence that salinomycin is active against retinoblastoma via inducing mitochondrial dysfunction, oxidative damage and AMPK activation. J Bioenerg Biomembr 2021; 53:513-523. [PMID: 34365583 DOI: 10.1007/s10863-021-09915-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/20/2021] [Indexed: 11/28/2022]
Abstract
The poor outcomes in retinoblastoma necessitate new treatments. Salinomycin is an attractive candidate, and has demonstrated selective anti-cancer properties in different cancer types. This work addressed the efficacy of salinomycin in retinoblastoma models and probe the associated mechanisms. Cellular functional assays were conducted to determine the effects salinomycin in vitro. Xenograft retinoblastoma mouse model was established to investigate the efficacy of salinomycin in vivo. Biochemical assays were conducted to analyze the mechanism of salinomycin's action focusing on mitochondrial functions, energy reduction-related signaling pathways. Salinomycin has positive effects towards retinoblastoma cells regardless of heterogeneity through suppressing growth and inducing apoptosis. Salinomycin also specifically inhibits cells displaying stemness and highly invasive phenotypes. Using retinoblastoma xenograft mouse model, we show that salinomycin at non-toxic dose effectively inhibits growth and induces apoptosis. Mechanistic studies show that salinomycin inhibits mitochondrial respiration via specifically suppressing complex I and II activities, reduces mitochondrial membrane potential and decreases energy reduction, followed by induction of oxidative stress and damage, AMPK activation and mTOR inhibition. Our study highlights that adding salinomycin to the existing treatment armamentarium for retinoblastoma is beneficial.
Collapse
Affiliation(s)
- Jing Li
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 26 Shengli Street, Wuhan, 430014, Hubei, China
| | - Yao Min
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, 26 Shengli Street, Wuhan, 430014, Hubei, China.
| |
Collapse
|
30
|
Oresta B, Pozzi C, Braga D, Hurle R, Lazzeri M, Colombo P, Frego N, Erreni M, Faccani C, Elefante G, Barcella M, Guazzoni G, Rescigno M. Mitochondrial metabolic reprogramming controls the induction of immunogenic cell death and efficacy of chemotherapy in bladder cancer. Sci Transl Med 2021; 13:13/575/eaba6110. [PMID: 33408185 DOI: 10.1126/scitranslmed.aba6110] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 10/15/2020] [Indexed: 12/22/2022]
Abstract
Although chemotherapeutic agents have been used for decades, the mechanisms of action, mechanisms of resistance, and the best treatment schedule remain elusive. Mitomycin C (MMC) is the gold standard treatment for non-muscle-invasive bladder cancer (NMIBC). However, it is effective only in a subset of patients, suggesting that, aside from cytotoxicity, other mechanisms could be involved in mediating the success of the treatment. Here, we showed that MMC promotes immunogenic cell death (ICD) and in vivo tumor protection. MMC-induced ICD relied on metabolic reprogramming of tumor cells toward increased oxidative phosphorylation. This favored increased mitochondrial permeability leading to the cytoplasmic release of mitochondrial DNA, which activated the inflammasome for efficient IL-1β (interleukin-1β) secretion that promoted dendritic cell maturation. Resistance to ICD was associated with mitochondrial dysfunction related to low abundance of complex I of the respiratory chain. Analysis of complex I in patient tumors indicated that low abundance of this mitochondrial complex was associated with recurrence incidence after chemotherapy in patients with NMIBC. The identification of mitochondria-mediated ICD as a mechanism of action of MMC offers opportunities to optimize bladder cancer management and provides potential markers of treatment efficacy that could be used for patient stratification.
Collapse
Affiliation(s)
- Bianca Oresta
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Chiara Pozzi
- Department of Experimental Oncology, European Institute of Oncology-IRCCS, via Ripamonti 435, 20141 Milan, Italy
| | - Daniele Braga
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Rodolfo Hurle
- Department of Urology, Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Massimo Lazzeri
- Department of Urology, Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Piergiuseppe Colombo
- Department of Pathology, Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Nicola Frego
- Department of Urology, Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Marco Erreni
- Unit of Advanced Optical Microscopy, Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Cristina Faccani
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Grazia Elefante
- Department of Pathology, Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy
| | - Matteo Barcella
- Dipartimento di Scienze della Salute, Università degli Studi di Milano, 20142 Milan, Italy
| | - Giorgio Guazzoni
- Department of Urology, Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy.,Humanitas University, Department of Biomedical Sciences, via Rita Levi Montalcini 4, 20090 Pieve Emanuele (Milan), Italy
| | - Maria Rescigno
- Humanitas Clinical and Research Center-IRCCS, via Manzoni 56, 20089 Rozzano (Milan), Italy. .,Humanitas University, Department of Biomedical Sciences, via Rita Levi Montalcini 4, 20090 Pieve Emanuele (Milan), Italy
| |
Collapse
|
31
|
Huangfu N, Wang Y, Xu Z, Zheng W, Tao C, Li Z, Hu Y, Chen X. TDP43 Exacerbates Atherosclerosis Progression by Promoting Inflammation and Lipid Uptake of Macrophages. Front Cell Dev Biol 2021; 9:687169. [PMID: 34291051 PMCID: PMC8287832 DOI: 10.3389/fcell.2021.687169] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/07/2021] [Indexed: 12/18/2022] Open
Abstract
Objective Atherosclerosis (AS), characterized by cholesterol overloaded-macrophages accumulation and plaque formation in blood vessels, is the major cause of cardiovascular disease. Transactive response DNA-binding protein∼43 kDa (TDP43) has recently been identified as an independent driver of neurodegenerative diseases through triggering inflammatory response. This study investigated whether TDP43 is involved in AS development, especially in macrophages-mediated-foam cell formation and inflammatory responses. Methods Transactive response DNA-binding protein∼43 kDa expressions in oxidized low-density lipoprotein (oxLDL)-treated macrophages and peripheral blood mononuclear cells (PBMCs) from patients with coronary artery disease (CAD) were detected by real time-polymerase chain reaction (RT-PCR), Western blot, and immunofluorescence. Gene gain or loss of function was used to investigate the effects of TDP43 on macrophages-mediated lipid untake and inflammation with ELISA, protein immunoprecipitation, RT-PCR, Western blot, and immunofluorescence. Macrophage TDP43 specific knockout mice with ApoE-/- background were fed with western diet for 12 weeks to establish AS model, and used to explore the role of TDP43 on AS progression. Results Transactive response DNA-binding protein∼43 kDa expression increases in oxLDL-treated macrophages and PBMCs from patients with CAD. Furthermore, we find that TDP43 promotes activation of NF-κB to increase inflammatory factor expression in macrophages through triggering mitochondrial DNA release to activate cGAS-STING signaling. Moreover, TDP43 strengthens lipid uptake of macrophages through regulating β-catenin and PPAR-γ complex to promote scavenger receptor gene CD36 transcription. Finally, using macrophage TDP43 specific knockout mice with ApoE-/- background fed with western diet for 12 weeks to establish AS model, we find that specific knockout of TDP43 in macrophages obviously alleviates western diet-induced AS progression in mice. Conclusions Transactive response DNA-binding protein∼43 kDa exacerbates atherosclerosis progression by promoting inflammation and lipid uptake of macrophages, suggesting TDP43 as a potential target for developing atherosclerotic drug.
Collapse
Affiliation(s)
- Ning Huangfu
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Yong Wang
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Zhenyu Xu
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Wenyuan Zheng
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Chunlan Tao
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Zhenwei Li
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Yewen Hu
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| | - Xiaomin Chen
- Department of Cardiology, Ningbo First Hospital, Ningbo, China
| |
Collapse
|
32
|
Guo Y, Hu B, Fu B, Zhu H. Atovaquone at clinically relevant concentration overcomes chemoresistance in ovarian cancer via inhibiting mitochondrial respiration. Pathol Res Pract 2021; 224:153529. [PMID: 34174549 DOI: 10.1016/j.prp.2021.153529] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/09/2021] [Accepted: 06/14/2021] [Indexed: 12/24/2022]
Abstract
The poor outcomes in ovarian cancer necessitate new treatments. Strategies to interfere with oxidative phosphorylation have been recently highlighted for the treatment of ovarian tumors. Atovaquone, an approved antimicrobial drug, has demonstrated anti-cancer potential and ability in disrupting mitochondrial function. Here, we investigated the efficacy of atovaquone as single drug and its combination with cisplatin in ovarian cancer. We show that atovaquone at clinically achievable concentrations is active against ovarian cancer bulky and stem-cell like cells via inhibiting growth and colony formation, and inducing caspase-dependent apoptosis. In contrast, atovaquone either does not or inhibits normal cells in a less extent than in ovarian cancer cells. Mechanism studies using multiple independent approaches demonstrate that atovaquone acts on ovarian cancer cells via decreasing mitochondrial complex III which results in mitochondrial respiration inhibition, energy reduction and oxidative stress. In line with in vitro findings, atovaquone alone at non-toxic dose is effective in inhibiting ovarian cancer growth in vivo, and its combination with cisplatin is synergistic. Our study suggests that atovaquone is a promising candidate to the treatment of ovarian cancer. Our work also supports the notion that mitochondrial respiration is a therapeutic target in ovarian cancer.
Collapse
Affiliation(s)
- Yue Guo
- Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Bo Hu
- Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Bingbing Fu
- Department of Obstetrics and Gynaecology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, China.
| | - Hongyan Zhu
- Department of Oncology, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang, China.
| |
Collapse
|
33
|
Kim MC, Hwang SH, Yang Y, Kim NY, Kim Y. Reduction in mitochondrial oxidative stress mediates hypoxia-induced resistance to cisplatin in human transitional cell carcinoma cells. Neoplasia 2021; 23:653-662. [PMID: 34134082 PMCID: PMC8208898 DOI: 10.1016/j.neo.2021.05.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 05/16/2021] [Accepted: 05/18/2021] [Indexed: 12/04/2022] Open
Abstract
Tumor hypoxia is known to promote the acquisition of more aggressive phenotypes in human transitional cell carcinoma (TCC), including drug resistance. Accumulating evidence suggests that mitochondria play a central role in the chemoresistance of TCC. However, the role of mitochondria in the hypoxia-induced drug resistance in TCC remains elusive. The present study investigated the function of mitochondria in the drug resistance using a TCC cell line under hypoxic conditions. In vitro hypoxia (0.1% O2, 48 h) was achieved by incubating TCC cells in air chamber. Mitochondrial events involving hypoxia-induced drug resistance were assessed. Hypoxia significantly reduced the cisplatin-induced apoptosis of TCC cells. Additionally, hypoxia substantially decreased the level of mitochondrial reactive oxygen species (ROS) generated by cisplatin treatment. Analogously, elimination of mitochondrial ROS significantly rescued cells from cisplatin-induced apoptosis. Hypoxia enhanced mitochondrial hyperpolarization, which was not related to ATP production or the reversal of ATP synthase activity. The mitochondrial DNA (mtDNA) amplification efficiency data illustrated that hypoxia significantly prevented oxidative damage to the mitogenome. Moreover, transmission electron microscopy revealed that cisplatin-induced disruption of the mitochondrial ultrastructure was abated under hypoxic conditions. Notably, depletion of mtDNA by ethidium bromide abrogated hypoxia-induced resistance to cisplatin. Taken together, the present study demonstrated that TCC cells exposed to hypoxic conditions rendered mitochondria less sensitive to oxidative stress induced by cisplatin treatment, leading to enhanced drug resistance.
Collapse
Affiliation(s)
- Myung-Chul Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea; BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea; Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, USA
| | - Sung-Hyun Hwang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea; BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea
| | - Yeseul Yang
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea; BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea
| | - Na-Yon Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea; BK21 PLUS Program for Creative Veterinary Science Research, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea
| | - Yongbaek Kim
- Laboratory of Clinical Pathology, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea; Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul, The Republic of Korea.
| |
Collapse
|
34
|
Zhou L, Zhang YF, Yang FH, Mao HQ, Chen Z, Zhang L. Mitochondrial DNA leakage induces odontoblast inflammation via the cGAS-STING pathway. Cell Commun Signal 2021; 19:58. [PMID: 34016129 PMCID: PMC8136190 DOI: 10.1186/s12964-021-00738-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 04/13/2021] [Indexed: 12/22/2022] Open
Abstract
Background Mitochondrial DNA (mtDNA) is a vital driver of inflammation when it leaks from damaged mitochondria into the cytosol. mtDNA stress may contribute to cyclic GMP-AMP synthase (cGAS) stimulator of interferon genes (STING) pathway activation in infectious diseases. Odontoblasts are the first cells challenged by cariogenic bacteria and involved in maintenance of the pulp immune and inflammatory responses to dentine-invading pathogens. In this study, we investigated that mtDNA as an important inflammatory driver participated in defending against bacterial invasion via cGAS-STING pathway in odontoblasts. Methods The normal tissues, caries tissues and pulpitis tissues were measured by western blotting and immunohistochemical staining. Pulpitis model was built in vitro to evaluated the effect of the cGAS-STING pathway in odontoblast-like cell line (mDPC6T) under inflammation. Western blot and real-time PCR were performed to detect the expression of cGAS-STING pathway and pro-inflammatory cytokines. The mitochondrial function was evaluated reactive oxygen species (ROS) generated by mitochondria using MitoSOX Red dye staining. Cytosolic DNA was assessed by immunofluorescent staining and real-time PCR in mDPC6T cells after LPS stimulation. Furthermore, mDPC6T cells were treated with ethidium bromide (EtBr) to deplete mtDNA or transfected with isolated mtDNA. The expression of cGAS-STING pathway and pro-inflammatory cytokines were measured. Results The high expression of cGAS and STING in caries and pulpitis tissues in patients, which was associated with inflammatory progression. The cGAS-STING pathway was activated in inflamed mDPC6T. STING knockdown inhibited the nuclear import of p65 and IRF3 and restricted the secretion of the inflammatory cytokines CXCL10 and IL-6 induced by LPS. LPS caused mitochondrial damage in mDPC6T, which promoted mtDNA leakage into the cytosol. Depletion of mtDNA inhibited the cGAS-STING pathway and nuclear translocation of p65 and IRF3. Moreover, repletion of mtDNA rescued the inflammatory response, which was inhibited by STING knockdown. Conclusion Our study systematically identified a novel mechanism of LPS-induced odontoblast inflammation, which involved mtDNA leakage from damaged mitochondria into the cytosol stimulating the cGAS-STING pathway and the inflammatory cytokines IL-6 and CXCL10 secretion. The mtDNA-cGAS-STING axis could be a potent therapeutic target to prevent severe bacterial inflammation in pulpitis. Video Abstract
Graphic abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12964-021-00738-7.
Collapse
Affiliation(s)
- Lu Zhou
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yi-Fei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Fu-Hua Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Han-Qing Mao
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China.,Department of Endodontics, School and Hospital of Stomatology, Wuhan University, HongShan District, LuoYu Road No. 237, Wuhan, 430079, China
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei- MOST) and Key Laboratory of Oral Biomedicine Ministry of Education, School and Hospital of Stomatology, Wuhan University, Wuhan, China. .,Department of Endodontics, School and Hospital of Stomatology, Wuhan University, HongShan District, LuoYu Road No. 237, Wuhan, 430079, China.
| |
Collapse
|
35
|
Wang LQ, Liu T, Yang S, Sun L, Zhao ZY, Li LY, She YC, Zheng YY, Ye XY, Bao Q, Dong GH, Li CW, Cui J. Perfluoroalkyl substance pollutants activate the innate immune system through the AIM2 inflammasome. Nat Commun 2021; 12:2915. [PMID: 34006824 PMCID: PMC8131593 DOI: 10.1038/s41467-021-23201-0] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 04/15/2021] [Indexed: 02/03/2023] Open
Abstract
Perfluoroalkyl substances (PFAS) are widely used in various manufacturing processes. Accumulation of these chemicals has adverse effects on human health, including inflammation in multiple organs, yet how PFAS are sensed by host cells, and how tissue inflammation eventually incurs, is still unclear. Here, we show that the double-stranded DNA receptor AIM2 is able to recognize perfluorooctane sulfonate (PFOS), a common form of PFAS, to trigger IL-1β secretion and pyroptosis. Mechanistically, PFOS activates the AIM2 inflammasome in a process involving mitochondrial DNA release through the Ca2+-PKC-NF-κB/JNK-BAX/BAK axis. Accordingly, Aim2-/- mice have reduced PFOS-induced inflammation, as well as tissue damage in the lungs, livers, and kidneys in both their basic condition and in an asthmatic exacerbation model. Our results thus suggest a function of AIM2 in PFOS-mediated tissue inflammation, and identify AIM2 as a major pattern recognition receptor in response to the environmental organic pollutants.
Collapse
Affiliation(s)
- Li-Qiu Wang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Tao Liu
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Shuai Yang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Lin Sun
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhi-Yao Zhao
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Li-Yue Li
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yuan-Chu She
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Yan-Yan Zheng
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Xiao-Yan Ye
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Qing Bao
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guang-Hui Dong
- Guangdong Provincial Engineering Technology Research Center of Environmental Pollution and Health Risk Assessment, Department of Occupational and Environmental Health, School of Public Health, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chun-Wei Li
- Department of Otolaryngology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China.
| | - Jun Cui
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong, China.
| |
Collapse
|
36
|
Lutz-Bonengel S, Niederstätter H, Naue J, Koziel R, Yang F, Sänger T, Huber G, Berger C, Pflugradt R, Strobl C, Xavier C, Volleth M, Weiß SC, Irwin JA, Romsos EL, Vallone PM, Ratzinger G, Schmuth M, Jansen-Dürr P, Liehr T, Lichter P, Parsons TJ, Pollak S, Parson W. Evidence for multi-copy Mega-NUMTs in the human genome. Nucleic Acids Res 2021; 49:1517-1531. [PMID: 33450006 PMCID: PMC7897518 DOI: 10.1093/nar/gkaa1271] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 12/11/2020] [Accepted: 12/22/2020] [Indexed: 12/18/2022] Open
Abstract
The maternal mode of mitochondrial DNA (mtDNA) inheritance is central to human genetics. Recently, evidence for bi-parental inheritance of mtDNA was claimed for individuals of three pedigrees that suffered mitochondrial disorders. We sequenced mtDNA using both direct Sanger and Massively Parallel Sequencing in several tissues of eleven maternally related and other affiliated healthy individuals of a family pedigree and observed mixed mitotypes in eight individuals. Cells without nuclear DNA, i.e. thrombocytes and hair shafts, only showed the mitotype of haplogroup (hg) V. Skin biopsies were prepared to generate ρ° cells void of mtDNA, sequencing of which resulted in a hg U4c1 mitotype. The position of the Mega-NUMT sequence was determined by fluorescence in situ hybridization and two different quantitative PCR assays were used to determine the number of contributing mtDNA copies. Thus, evidence for the presence of repetitive, full mitogenome Mega-NUMTs matching haplogroup U4c1 in various tissues of eight maternally related individuals was provided. Multi-copy Mega-NUMTs mimic mixtures of mtDNA that cannot be experimentally avoided and thus may appear in diverse fields of mtDNA research and diagnostics. We demonstrate that hair shaft mtDNA sequencing provides a simple but reliable approach to exclude NUMTs as source of misleading results.
Collapse
Affiliation(s)
- Sabine Lutz-Bonengel
- Institute of Forensic Medicine, Medical Center, University of Freiburg and Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Harald Niederstätter
- Institute of Legal Medicine, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Jana Naue
- Institute of Forensic Medicine, Medical Center, University of Freiburg and Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Rafal Koziel
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck 6020, Austria
| | - Fengtang Yang
- Wellcome Sanger Institute, Hinxton, Cambridge CB10 1SA, UK
| | - Timo Sänger
- Institute of Forensic Medicine, Medical Center, University of Freiburg and Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Gabriela Huber
- Institute of Legal Medicine, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Cordula Berger
- Institute of Legal Medicine, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - René Pflugradt
- State Investigation Department of Lower Saxony, Hannover 30169, Germany
| | - Christina Strobl
- Institute of Legal Medicine, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Catarina Xavier
- Institute of Legal Medicine, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Marianne Volleth
- Magdeburg University Hospital, Institute of Human Genetics, Otto von Guericke University, Magdeburg 39120, Germany
| | - Sandra Carina Weiß
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Freiburg, Freiburg 79104, Germany
| | - Jodi A Irwin
- DNA Support Unit, FBI Laboratory, Quantico, VA 22135, USA
| | - Erica L Romsos
- U.S. National Institute of Standards and Technology, Biomolecular Measurement Division, Gaithersburg, MD 20899, USA
| | - Peter M Vallone
- U.S. National Institute of Standards and Technology, Biomolecular Measurement Division, Gaithersburg, MD 20899, USA
| | - Gudrun Ratzinger
- Department of Dermatology, Venereology and Allergy, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Matthias Schmuth
- Department of Dermatology, Venereology and Allergy, Medical University of Innsbruck, Innsbruck 6020, Austria
| | - Pidder Jansen-Dürr
- Institute for Biomedical Aging Research, University of Innsbruck, Innsbruck 6020, Austria
| | - Thomas Liehr
- Jena University Hospital, Institute of Human Genetics, Friedrich Schiller University, Jena 07747, Germany
| | - Peter Lichter
- German Cancer Research Center, Molecular Genetics, Heidelberg 69120, Germany
| | - Thomas J Parsons
- International Commission on Missing Persons, The Hague 2514 AA, Netherlands
- Forensic Science Program, The Pennsylvania State University, University Park, PA 16802, USA
| | - Stefan Pollak
- Institute of Forensic Medicine, Medical Center, University of Freiburg and Faculty of Medicine, University of Freiburg, Freiburg 79104, Germany
| | - Walther Parson
- Institute of Legal Medicine, Medical University of Innsbruck, Innsbruck 6020, Austria
- Forensic Science Program, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
37
|
Jauhari A, Baranov SV, Suofu Y, Kim J, Singh T, Yablonska S, Li F, Wang X, Oberly P, Minnigh MB, Poloyac SM, Carlisle DL, Friedlander RM. Melatonin inhibits cytosolic mitochondrial DNA-induced neuroinflammatory signaling in accelerated aging and neurodegeneration. J Clin Invest 2021; 130:3124-3136. [PMID: 32182222 DOI: 10.1172/jci135026] [Citation(s) in RCA: 96] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/05/2020] [Indexed: 12/19/2022] Open
Abstract
Chronic inflammation is a pathologic feature of neurodegeneration and aging; however, the mechanism regulating this process is not understood. Melatonin, an endogenous free radical scavenger synthesized by neuronal mitochondria, decreases with aging and neurodegeneration. We proposed that insufficient melatonin levels impair mitochondrial homeostasis, resulting in mitochondrial DNA (mtDNA) release and activation of cytosolic DNA-mediated inflammatory response in neurons. We found increased mitochondrial oxidative stress and decreased mitochondrial membrane potential, with higher mtDNA release in brain and primary cerebro-cortical neurons of melatonin-deficient aralkylamine N-acetyltransferase (AANAT) knockout mice. Cytosolic mtDNA activated the cGAS/STING/IRF3 pathway, stimulating inflammatory cytokine generation. We found that Huntington's disease mice had increased mtDNA release, cGAS activation, and inflammation, all inhibited by exogenous melatonin. Thus, we demonstrated that cytosolic mtDNA activated the inflammatory response in aging and neurodegeneration, a process modulated by melatonin. Furthermore, our data suggest that AANAT knockout mice are a model of accelerated aging.
Collapse
Affiliation(s)
| | | | | | - Jinho Kim
- Department of Neurological Surgery and
| | | | | | - Fang Li
- Department of Neurological Surgery and
| | | | - Patrick Oberly
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - M Beth Minnigh
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Samuel M Poloyac
- Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | | | | |
Collapse
|
38
|
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: 500] [Impact Index Per Article: 125.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [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
|
39
|
DNA-induced 2'3'-cGAMP enhances haplotype-specific human STING cleavage by dengue protease. Proc Natl Acad Sci U S A 2020; 117:15947-15954. [PMID: 32576686 PMCID: PMC7354927 DOI: 10.1073/pnas.1922243117] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Dengue virus (DENV) antagonizes the DNA sensing cGAS-STING pathway to subvert innate immunity, but how DENV protease-mediated human STING cleavage contributes to DENV pathogenesis remains obscure. Here, we found that STING haplotype frequency varies among different subhuman populations, and different haplotypes respond differently to DENV protease. The cleavage of a DENV protease-sensitive STING can be further enhanced by coculture with neighboring cells producing 2′3′-cGAMP, either by DNA transfection of cGAS or by reactivating Epstein–Barr virus from latent infection. Thus, DENV infection trims down human STING-mediated innate immunity in a haplotype-specific manner. The genetic background of host STING and bystander coinfection of pathogens triggering 2′3′-cGAMP production may be the missing link between STING cleavage and DENV pathogenesis. The cytosolic DNA sensor cGMP-AMP synthase (cGAS) synthesizes the noncanonical cyclic dinucleotide 2′3′-cGAMP to activate the adaptor protein stimulator of IFN genes (STING), thus awakening host immunity in response to DNA pathogen infection. However, dengue virus (DENV), an RNA virus without a DNA stage in its life cycle, also manipulates cGAS-STING–mediated innate immunity by proteolytic degradation of STING. Here, we found that the sensitivity of STING to DENV protease varied with different human STING haplotypes. Exogenous DNA further enhanced DENV protease’s ability to interact and cleave protease-sensitive STING. DNA-enhanced STING cleavage was reduced in cGAS-knockdown cells and triggered by the cGAS product 2′3′-cGAMP. The source of DNA may not be endogenous mitochondrial DNA but rather exogenous reactivated viral DNA. Cells producing 2′3′-cGAMP by overexpressing cGAS or with DNA virus reactivation enhanced STING cleavage in neighboring cells harboring DENV protease. DENV infection reduced host innate immunity in cells with the protease-sensitive STING haplotype, whose homozygote genotype frequency was found significantly reduced in Taiwanese people with dengue fever. Therefore, the human STING genetic background and DNA pathogen coinfection may be the missing links contributing to DENV pathogenesis.
Collapse
|
40
|
Artesunate targets oral tongue squamous cell carcinoma via mitochondrial dysfunction-dependent oxidative damage and Akt/AMPK/mTOR inhibition. J Bioenerg Biomembr 2020; 52:113-121. [PMID: 31965456 DOI: 10.1007/s10863-020-09823-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 01/13/2020] [Indexed: 12/12/2022]
Abstract
Although mitochondrial metabolism has recently gained attention as a promising therapeutic strategy in cancer, little is known on the impact of mitochondrial respiration inhibition on oral tongue squamous cell carcinoma (OTSCC). Using in vitro and in vivo OTSCC models, our work demonstrates that inducing mitochondrial dysfunction by anti-malarial drug artesunate is effective in targeting OTSCC stem-cell like and bulk cells. Artesunate inhibits anchorage-independent colony formation, proliferation and survival in all tested OTSCC cell lines although with varying efficacy. Artesunate displays preferential anti-OTSCC activity by sparing normal cells. Mechanism analysis indicates that artesunate inhibits mitochondrial respiration via suppressing mitochondrial complex I and II but not IV or V, resulting in oxidative stress and damage. Interestingly, OTSCC cells that are more sensitive to artesunate have higher level of basal mitochondrial respiration and reversed respiratory capacity compared to those with less sensitivity to artesunate, suggesting the varying dependence on mitochondrial respiration among OTSCC cell lines. In addition, artesunate induces oxidative stress and damage in cells with low sensitivity to a less extent than in those with high sensitivity. We confirm that mitochondrial respiration inhibition is required for the action of artesunate in OTSCC. Mitochondrial dysfunction by artesunate further activates AMPK and suppresses Akt/mTOR. Importantly, the in vitro observations are reproducible in vivo OTSCC xenograft mouse model. Our findings provide pre-clinical evidence on the efficacy of artesunate and emphasize the therapeutic value of targeting mitochondrial respiration in OTSCC.
Collapse
|
41
|
Shukla H, Chitrakar R, Bibi HA, Gaje G, Koucheki A, Trush MA, Zhu H, Li YR, Jia Z. Reactive oxygen species production by BP-1,6-quinone and its effects on the endothelial dysfunction: Involvement of the mitochondria. Toxicol Lett 2020; 322:120-130. [PMID: 31953210 DOI: 10.1016/j.toxlet.2020.01.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Revised: 01/08/2020] [Accepted: 01/13/2020] [Indexed: 01/11/2023]
Abstract
Strong epidemiological evidence supports the association between increased air pollution and the risk of developing atherosclerotic cardiovascular diseases (CVDs). However, the mechanism remains unclear. As an environmental air pollutant and benzo-a-pyrene (BP) metabolite, BP-1,6-quinone (BP-1,6-Q) is present in the particulate phase of air pollution. This study was undertaken to examine the redox activity of BP-1,6-Q and mechanisms associated with it using EA.hy926 endothelial cells. BP-1,6-Q at 0.01-1 μM significantly stimulated the production of reactive oxygen species (ROS)·in intact cells and isolated mitochondria. Furthermore, BP-1,6-Q-induced ROS was altered by mitochondrial electron transport chain (METC) inhibitors of complex I (rotenone) and complex III (antimycin A), denoting the involvement of mitochondrial electron transport chain (METC) in BP-1,6-Q mediated ROS production. In METC deficient cells, interestingly, BP-1,6-Q-mediated ROS production was enhanced, suggesting that overproduction of ROS by BP-1,6-Q is not only produced from mitochondria but can also be from the cell outside of mitochondria (extramitochondrial). BP-1,6-Q also triggered endothelial-monocyte interaction and stimulated expression of vascular adhesion molecule-1 (VCAM-1). In conclusion, these results demonstrate that BP-1,6-Q can generate ROS within both mitochondria and outside of mitochondria, resulting in stimulation of adhesion of monocytes to endothelial cells, a key event in the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Halley Shukla
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Rojin Chitrakar
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Humaira A Bibi
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Gabriella Gaje
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Ashkon Koucheki
- Department of Biology, University of North Carolina at Greensboro, NC, United States
| | - Michael A Trush
- Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, United States
| | - Hong Zhu
- Campbell University School of Osteopathic Medicine, Buies Creek, NC, United States
| | - Y Robert Li
- Department of Biology, University of North Carolina at Greensboro, NC, United States; Campbell University School of Osteopathic Medicine, Buies Creek, NC, United States.
| | - Zhenquan Jia
- Department of Biology, University of North Carolina at Greensboro, NC, United States.
| |
Collapse
|
42
|
Gao Y, Dorn P, Liu S, Deng H, Hall SRR, Peng RW, Schmid RA, Marti TM. Cisplatin-resistant A549 non-small cell lung cancer cells can be identified by increased mitochondrial mass and are sensitive to pemetrexed treatment. Cancer Cell Int 2019; 19:317. [PMID: 31798346 PMCID: PMC6883680 DOI: 10.1186/s12935-019-1037-1] [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: 07/10/2019] [Accepted: 11/15/2019] [Indexed: 01/13/2023] Open
Abstract
Background Cisplatin plus pemetrexed combination therapy is considered the standard treatment for patients with advanced, non-squamous, non-small-cell lung cancer (NSCLC). However, advanced NSCLC has a 5-year survival rate of below 10%, which is mainly due to therapy resistance. We previously showed that the NSCLC cell line A549 harbors different subpopulations including a mesenchymal-like subpopulation characterized by increased chemo- and radiotherapy resistance. Recently, therapy resistance in hematological and solid tumors has been associated with increased mitochondrial activity. Thus, the aim of this study was to investigate the role of the mitochondrial activity in NSCLC chemotherapy resistance. Methods Based on MitoTracker staining, subpopulations characterized by the highest 10% (Mito-High) or lowest 10% (Mito-Low) mitochondrial mass content were sorted by FACS (Fluorescence-Activated Cell Sorting) from paraclonal cultures of the NSCLC A549 cell line . Mitochondrial DNA copy numbers were quantified by real-time PCR whereas basal cellular respiration was measured by high-resolution respirometry. Cisplatin and pemetrexed response were quantified by proliferation and colony formation assay. Results Pemetrexed treatment of parental A549 cells increased mitochondrial mass over time. FACS-sorted paraclonal Mito-High cells featured increased mitochondrial mass and mitochondrial DNA copy number compared to the Mito-Low cells. Paraclonal Mito-High cells featured an increased proliferation rate and were significantly more resistant to cisplatin treatment than Mito-Low cells. Interestingly, cisplatin-resistant, paraclonal Mito-High cells were significantly more sensitive to pemetrexed treatment than Mito-Low cells. We provide a working model explaining the molecular mechanism underlying the increased cisplatin- and decreased pemetrexed resistance of a distinct subpopulation characterized by high mitochondrial mass. Conclusions This study revealed that cisplatin resistant A549 lung cancer cells can be identified by their increased levels of mitochondrial mass. However, Mito-High cells feature an increased sensitivity to pemetrexed treatment. Thus, pemetrexed and cisplatin target reciprocal lung cancer subpopulations, which could explain the increased efficacy of the combination therapy in the clinical setting.
Collapse
Affiliation(s)
- Yanyun Gao
- 1Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Murtenstrasse 50, 3008 Bern, Switzerland.,2Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Patrick Dorn
- 1Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Murtenstrasse 50, 3008 Bern, Switzerland.,2Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Shengchen Liu
- 2Department of BioMedical Research, University of Bern, Bern, Switzerland.,3Department of Intensive Care Medicine, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Haibin Deng
- 1Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Murtenstrasse 50, 3008 Bern, Switzerland.,2Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Sean R R Hall
- 1Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Murtenstrasse 50, 3008 Bern, Switzerland.,2Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Ren-Wang Peng
- 1Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Murtenstrasse 50, 3008 Bern, Switzerland.,2Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Ralph A Schmid
- 1Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Murtenstrasse 50, 3008 Bern, Switzerland.,2Department of BioMedical Research, University of Bern, Bern, Switzerland
| | - Thomas M Marti
- 1Department of General Thoracic Surgery, Inselspital, Bern University Hospital, Murtenstrasse 50, 3008 Bern, Switzerland.,2Department of BioMedical Research, University of Bern, Bern, Switzerland
| |
Collapse
|
43
|
Maekawa H, Inoue T, Ouchi H, Jao TM, Inoue R, Nishi H, Fujii R, Ishidate F, Tanaka T, Tanaka Y, Hirokawa N, Nangaku M, Inagi R. Mitochondrial Damage Causes Inflammation via cGAS-STING Signaling in Acute Kidney Injury. Cell Rep 2019; 29:1261-1273.e6. [DOI: 10.1016/j.celrep.2019.09.050] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/01/2019] [Accepted: 09/18/2019] [Indexed: 12/19/2022] Open
|
44
|
Chloramphenicol Mitigates Oxidative Stress by Inhibiting Translation of Mitochondrial Complex I in Dopaminergic Neurons of Toxin-Induced Parkinson's Disease Model. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:4174803. [PMID: 31534621 PMCID: PMC6732590 DOI: 10.1155/2019/4174803] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 06/18/2019] [Indexed: 01/06/2023]
Abstract
Paraquat (PQ), an herbicide considered an environmental contributor to the development of Parkinson's disease (PD), induces dopaminergic neuronal loss through reactive oxygen species (ROS) production and oxidative stress by mitochondrial complex I. Most patients with PQ-induced PD are affected by chronic exposure and require a preventive strategy for modulation of disease progression. To identify drugs that are effective in preventing PD, we screened more than 1000 drugs that are currently used in clinics and in studies employing PQ-treated cells. Of these, chloramphenicol (CP) showed the most powerful inhibitory effect. Pretreatment with CP increased the viability of PQ-treated SN4741 dopaminergic neuronal cells and rat primary cultured dopaminergic neurons compared with control cells treated with PQ only. CP pretreatment also reduced PQ-induced ROS production, implying that mitochondrial complex I is a target of CP. This effect of CP reflected downregulation of the mitochondrial complex I subunit ND1 and diminished PQ recycling, a major mechanism of ROS production, and resulted in the prevention of cell loss. Notably, these effects of CP were not observed in rotenone-pretreated SN4741 cells and Rho-negative cells, in which mitochondrial function is defective. Consistent with these results, CP pretreatment of MPTP-treated PD model mice also ameliorated dopaminergic neuronal cell loss. Our findings indicate that the inhibition of mitochondrial complex I with CP protects dopaminergic neurons and may provide a strategy for preventing neurotoxin-induced PD.
Collapse
|
45
|
Oxidative Stress Induced by the Deubiquitinase Inhibitor b-AP15 Is Associated with Mitochondrial Impairment. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1659468. [PMID: 31281566 PMCID: PMC6590552 DOI: 10.1155/2019/1659468] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/05/2019] [Accepted: 04/30/2019] [Indexed: 12/17/2022]
Abstract
Inhibitors of the 20S proteasome such as bortezomib are cytotoxic to tumor cells and have been proven to be valuable for the clinical management of multiple myeloma. The therapeutic efficacy of bortezomib is, however, hampered by the emergence of acquired resistance. Available data suggest that blocking proteasome activity at the level of proteasome-associated deubiquitinases (DUBs) provides a mechanism to overcome resistance to bortezomib and also to other cancer therapies. The small molecule b-AP15 is an inhibitor of proteasome-associated DUB activity that induces both proteotoxic stress and increases in the levels of reactive oxygen species (ROS) in tumor cells. Antioxidants have been shown to decrease apoptosis induction by b-AP15 and we here addressed the question of the mechanism of redox perturbation by this compound. We show that oxidative stress induction by b-AP15 is abrogated in cells deprived of mitochondrial DNA (ρ0 cells). We also show associations between the level of proteotoxic stress, the degree of mitochondrial dysfunction, and the extent of induction of hemeoxygenase-1 (HO-1), a target of the redox-regulated Nrf-2 transcription factor. Decreased expression of COX5b (cytochrome c oxidase subunit 5b) and TOMM34 (translocase of outer mitochondrial membrane 34) was observed in b-AP15-treated cells. These findings suggest a mitochondrial origin of the increased levels of ROS observed in cells exposed to the DUB inhibitor b-AP15.
Collapse
|
46
|
Inhibition of mitochondrial translation selectively targets osteosarcoma. Biochem Biophys Res Commun 2019; 515:9-15. [PMID: 31118131 DOI: 10.1016/j.bbrc.2019.05.070] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 05/08/2019] [Indexed: 12/31/2022]
Abstract
The unique dependence of cancer cells on mitochondrial metabolism has been exploited therapeutically in various cancers but not osteosarcoma. In this work, we demonstrate that inhibition of mitochondrial translation is effective and selective in targeting osteosarcoma. We firstly showed that tigecycline at pharmacological achievable concentrations inhibited growth and induced apoptosis of multiple osteosarcoma cell lines while sparing normal osteoblast cells. Similarly, tigecycline at effective doses that delayed osteosarcoma growth did not cause significant toxicity to mice. We next showed that tigecycline specifically inhibits mitochondrial translation, resulting in defective mitochondrial respiration in both osteosarcoma and normal osteoblast cells. We further confirm mitochondrial respiration as the target of tigecycline using three independent approaches. In addition, we demonstrate that compared to normal osteoblasts, osteosarcoma cells have higher mitochondrial biogenesis. We finally show that specific inhibition of mitochondrial translation via EF-Tu depletion produces the similar anti-osteosarcoma effects of tigecycline. Our work highlights the therapeutic value of targeting mitochondrial metabolism in osteosarcoma and tigecycline as a useful addition to the treatment of osteosarcoma.
Collapse
|
47
|
Aarreberg LD, Esser-Nobis K, Driscoll C, Shuvarikov A, Roby JA, Gale M. Interleukin-1β Induces mtDNA Release to Activate Innate Immune Signaling via cGAS-STING. Mol Cell 2019; 74:801-815.e6. [PMID: 30952515 DOI: 10.1016/j.molcel.2019.02.038] [Citation(s) in RCA: 209] [Impact Index Per Article: 41.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Revised: 01/11/2019] [Accepted: 02/27/2019] [Indexed: 12/11/2022]
Abstract
Interleukin-1 beta (IL-1β) is a pleiotropic mediator of inflammation and is produced in response to a wide range of stimuli. During infection, IL-1β production occurs in parallel with the onset of innate antimicrobial defenses, but the contribution of IL-1β signaling to cell-intrinsic immunity is not defined. Here, we report that exogenous IL-1β induces interferon regulatory factor 3 (IRF3) activation in human myeloid, fibroblast, and epithelial cells. IRF3 activation by IL-1β is dependent upon the DNA-sensing pathway adaptor, stimulator of interferon genes (STING), through the recognition of cytosolic mtDNA by cyclic guanosine monophosphate (GMP)-AMP synthase (cGAS). IL-1β treatment results in interferon (IFN) production and activation of IFN signaling to direct a potent innate immune response that restricts dengue virus infection. This study identifies a new function for IL-1β in the onset or enhancement of cell-intrinsic immunity, with important implications for cGAS-STING in integrating inflammatory and microbial cues for host defense.
Collapse
Affiliation(s)
- Lauren D Aarreberg
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Katharina Esser-Nobis
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Connor Driscoll
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Andrey Shuvarikov
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Justin A Roby
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, University of Washington School of Medicine, Seattle, WA 98109, USA.
| |
Collapse
|
48
|
Chen Z, Zhang F, Xu H. Human mitochondrial DNA diseases and Drosophila models. J Genet Genomics 2019; 46:201-212. [DOI: 10.1016/j.jgg.2019.03.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 03/05/2019] [Accepted: 03/25/2019] [Indexed: 01/06/2023]
|
49
|
Apoptotic Caspases Suppress Type I Interferon Production via the Cleavage of cGAS, MAVS, and IRF3. Mol Cell 2019; 74:19-31.e7. [PMID: 30878284 DOI: 10.1016/j.molcel.2019.02.013] [Citation(s) in RCA: 193] [Impact Index Per Article: 38.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 12/13/2018] [Accepted: 02/08/2019] [Indexed: 01/12/2023]
Abstract
Viral infection triggers host defenses through pattern-recognition receptor-mediated cytokine production, inflammasome activation, and apoptosis of the infected cells. Inflammasome-activated caspases are known to cleave cyclic GMP-AMP synthase (cGAS). Here, we found that apoptotic caspases are critically involved in regulating both DNA and RNA virus-triggered host defenses, in which activated caspase-3 cleaved cGAS, MAVS, and IRF3 to prevent cytokine overproduction. Caspase-3 was exclusively required in human cells, whereas caspase-7 was involved only in murine cells to inactivate cGAS, reflecting distinct regulatory mechanisms in different species. Caspase-mediated cGAS cleavage was enhanced in the presence of dsDNA. Alternative MAVS cleavage sites were used to ensure the inactivation of this critical protein. Elevated type I IFNs were detected in caspase-3-deficient cells without any infection. Casp3-/- mice consistently showed increased resistance to viral infection and experimental autoimmune encephalomyelitis. Our results demonstrate that apoptotic caspases control innate immunity and maintain immune homeostasis against viral infection.
Collapse
|
50
|
Xiao Y, Xiong T, Meng X, Yu D, Xiao Z, Song L. Different influences on mitochondrial function, oxidative stress and cytotoxicity of antibiotics on primary human neuron and cell lines. J Biochem Mol Toxicol 2018; 33:e22277. [PMID: 30597674 DOI: 10.1002/jbt.22277] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 10/28/2018] [Accepted: 10/31/2018] [Indexed: 02/06/2023]
Abstract
Although antibiotics are generally well tolerated, their toxic effects on the central nervous system have been gained attention. In this study, we systematically investigated the neuron toxicity of antibiotics from six different classes. We show that clinically relevant concentrations of metronidazole, tigecycline, azithromycin and clindamycin but not ampicillin or sulfamethoxazole induce apoptosis of human primary neuron cells and lines. Notably, tigecycline, azithromycin and clindamycin cause neuron cell oxidative damage whereas metronidazole has no effect on reactive oxygen species (ROS) production, suggesting that metronidazole induces neuron death via ROS-independent mechanism. Tigecycline, azithromycin and clindamycin induce mitochondrial dysfunctions via targeting different mitochondrial respiratory complexes, leading to mitochondrial membrane potential disruption and energy crisis. The deleterious effects of antibiotics are reversed by pretreatment of neuron cells with antioxidant. Our work highlights the different influences of antibiotics on mitochondrial dysfunction, oxidative damage and cytotoxicity in neuron cells. We also provide a strategy to prevent the neurotoxicity.
Collapse
Affiliation(s)
- Yao Xiao
- Department of Neurology, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ting Xiong
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Xiangping Meng
- Department of Integrated Medicine, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Danfang Yu
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| | - Zheng Xiao
- Department of Emergency Medicine, Wuhan Fourth Hospital, Puai Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lin Song
- Department of Neurology, Hubei Provincial Hospital of Integrated Chinese and Western Medicine, Wuhan, China
| |
Collapse
|