1
|
Duan X, Liu R, Lan W, Liu S. The Essential Role of Mitochondrial Dynamics in Viral Infections. Int J Mol Sci 2025; 26:1955. [PMID: 40076578 PMCID: PMC11900267 DOI: 10.3390/ijms26051955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 02/17/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Mitochondria are dynamic organelles that play crucial roles in energy production, metabolic balance, calcium homeostasis, apoptosis, and innate immunity, and are key determinants of cell fate. They are also targets for viral invasion of the body. Many viral proteins target mitochondria, controlling mitochondrial morphology, metabolism, and immune response, thereby achieving immune evasion, promoting their proliferation, and accelerating the infection process. Mitochondrial quality control is key to maintaining normal physiological functions and mitochondrial homeostasis. Dysregulation of mitochondrial dynamics is closely related to the development of many diseases. New roles of mitochondrial dynamics in viral infection are constantly being discovered. Viruses change mitochondrial dynamics by targeting mitochondria to achieve a persistent state of infection. Currently, understanding of mitochondrial dynamics during viral infection is limited. Research on the impact of viral proteins on mitochondrial dynamics provides a foundation for investigating the pathogenesis of viral infections, the disease process, and identifying potential therapeutic targets. This review focuses on the connection between viral infection and mitochondrial dynamics and priority areas for research on virus-mediated mitochondrial immunity, provides insight into the regulation of mitochondrial dynamics by viruses targeting mitochondria, and explores potential means of mitochondrial-mediated control and treatment of viral diseases.
Collapse
Affiliation(s)
- Xujie Duan
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology for Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Rui Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology for Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Wenjing Lan
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology for Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Shuying Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Medicine, Inner Mongolia Agricultural University, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology for Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| |
Collapse
|
2
|
Aravind L, Nicastro GG, Iyer LM, Burroughs AM. The Prokaryotic Roots of Eukaryotic Immune Systems. Annu Rev Genet 2024; 58:365-389. [PMID: 39265037 DOI: 10.1146/annurev-genet-111523-102448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
Over the past two decades, studies have revealed profound evolutionary connections between prokaryotic and eukaryotic immune systems, challenging the notion of their unrelatedness. Immune systems across the tree of life share an operational framework, shaping their biochemical logic and evolutionary trajectories. The diversification of immune genes in the prokaryotic superkingdoms, followed by lateral transfer to eukaryotes, was central to the emergence of innate immunity in the latter. These include protein domains related to nucleotide second messenger-dependent systems, NAD+/nucleotide degradation, and P-loop NTPase domains of the STAND and GTPase clades playing pivotal roles in eukaryotic immunity and inflammation. Moreover, several domains orchestrating programmed cell death, ultimately of prokaryotic provenance, suggest an intimate link between immunity and the emergence of multicellularity in eukaryotes such as animals. While eukaryotes directly adopted some proteins from bacterial immune systems, they repurposed others for new immune functions from bacterial interorganismal conflict systems. These emerging immune components hold substantial biotechnological potential.
Collapse
Affiliation(s)
- L Aravind
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;
| | - Gianlucca G Nicastro
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;
| | - Lakshminarayan M Iyer
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;
| | - A Maxwell Burroughs
- Computational Biology Branch, National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, Maryland, USA;
| |
Collapse
|
3
|
Wang Y, Wang H, Yu X, Wu Q, Lv X, Zhou X, Chen Y, Geng S. Identification of metabolism related biomarkers in obesity based on adipose bioinformatics and machine learning. J Transl Med 2024; 22:986. [PMID: 39482740 PMCID: PMC11526509 DOI: 10.1186/s12967-024-05615-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/18/2024] [Indexed: 11/03/2024] Open
Abstract
BACKGROUND Obesity has emerged as a growing global public health concern over recent decades. Obesity prevalence exhibits substantial global variation, ranging from less than 5% in regions like China, Japan, and Africa to rates exceeding 75% in urban areas of Samoa. AIM To examine the involvement of metabolism-related genes. METHODS Gene expression datasets GSE110729 and GSE205668 were accessed from the GEO database. DEGs between obese and lean groups were identified through DESeq2. Metabolism-related genes and pathways were detected using enrichment analysis, WGCNA, Random Forest, and XGBoost. The identified signature genes were validated by real-time quantitative PCR (qRT-PCR) in mouse models. RESULTS A total of 389 genes exhibiting differential expression were discovered, showing significant enrichment in metabolic pathways, particularly in the propanoate metabolism pathway. The orangered4 module, which exhibited the highest correlation with propanoate metabolism, was identified using Weighted Correlation Network Analysis (WGCNA). By integrating the DEGs, WGCNA results, and machine learning methods, the identification of two metabolism-related genes, Storkhead Box 1 (STOX1), NACHT and WD repeat domain-containing protein 2(NWD2) was achieved. These signature genes successfully distinguished between obese and lean individuals. qRT-PCR analysis confirmed the downregulation of STOX1 and NWD2 in mouse models of obesity. CONCLUSION This study has analyzed the available GEO dataset in order to identify novel factors associated with obesity metabolism and found that STOX1 and NWD2 may serve as diagnostic biomarkers.
Collapse
Affiliation(s)
- Yanping Wang
- Department of Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, No. 1073 South Erhuan Road, Tangxiang Street, Dazu District, Chongqing, 402360, China
- Department of Endocrinology, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Honglin Wang
- Department of Orthopedic Surgery, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Xingrui Yu
- Institute of Information, Xiamen University, Xiamen, China
| | - Qinan Wu
- Department of Endocrinology, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Xinlu Lv
- Department of Endocrinology, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, 402360, China
| | - Xuelian Zhou
- The Affiliated Dazu Hospital of Chongqing Medical University, No. 1073 South Erhuan Road, Tangxiang Street, Dazu District, Chongqing, 402360, China
| | - Yong Chen
- The Affiliated Dazu Hospital of Chongqing Medical University, No. 1073 South Erhuan Road, Tangxiang Street, Dazu District, Chongqing, 402360, China.
| | - Shan Geng
- Department of Laboratory, The Affiliated Dazu Hospital of Chongqing Medical University, No. 1073 South Erhuan Road, Tangxiang Street, Dazu District, Chongqing, 402360, China.
- Department of Endocrinology, The Affiliated Dazu Hospital of Chongqing Medical University, Chongqing, 402360, China.
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
4
|
Boyd-Shiwarski CR, Shiwarski DJ, Subramanya AR. A New Phase for WNK Kinase Signaling Complexes as Biomolecular Condensates. Physiology (Bethesda) 2024; 39:0. [PMID: 38624245 PMCID: PMC11460533 DOI: 10.1152/physiol.00013.2024] [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: 02/22/2024] [Revised: 04/09/2024] [Accepted: 04/09/2024] [Indexed: 04/17/2024] Open
Abstract
The purpose of this review is to highlight transformative advances that have been made in the field of biomolecular condensates, with special emphasis on condensate material properties, physiology, and kinases, using the With-No-Lysine (WNK) kinases as a prototypical example. To convey how WNK kinases illustrate important concepts for biomolecular condensates, we start with a brief history, focus on defining features of biomolecular condensates, and delve into some examples of how condensates are implicated in cellular physiology (and pathophysiology). We then highlight how WNK kinases, through the action of "WNK droplets" that ubiquitously regulate intracellular volume and kidney-specific "WNK bodies" that are implicated in distal tubule salt reabsorption and potassium homeostasis, exemplify many of the defining features of condensates. Finally, this review addresses the controversies within this emerging field and questions to address.
Collapse
Affiliation(s)
- Cary R Boyd-Shiwarski
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | - Daniel J Shiwarski
- Vascular Medicine Institute, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Arohan R Subramanya
- Renal-Electrolyte Division, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Pittsburgh Heart, Lung, Blood Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
- VA Pittsburgh Healthcare System, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
5
|
Yamada S, Mizukoshi T, Sato A, Sakakibara SI. Purinosomes and Purine Metabolism in Mammalian Neural Development: A Review. Acta Histochem Cytochem 2024; 57:89-100. [PMID: 38988694 PMCID: PMC11231565 DOI: 10.1267/ahc.24-00027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 05/19/2024] [Indexed: 07/12/2024] Open
Abstract
Neural stem/progenitor cells (NSPCs) in specific brain regions require precisely regulated metabolite production during critical development periods. Purines-vital components of DNA, RNA, and energy carriers like ATP and GTP-are crucial metabolites in brain development. Purine levels are tightly controlled through two pathways: de novo synthesis and salvage synthesis. Enzymes driving de novo pathway are assembled into a large multienzyme complex termed the "purinosome." Here, we review purine metabolism and purinosomes as spatiotemporal regulators of neural development. Notably, around postnatal day 0 (P0) during mouse cortical development, purine synthesis transitions from the de novo pathway to the salvage pathway. Inhibiting the de novo pathway affects mTORC1 pathway and leads to specific forebrain malformations. In this review, we also explore the importance of protein-protein interactions of a newly identified NSPC protein-NACHT and WD repeat domain-containing 1 (Nwd1)-in purinosome formation. Reduced Nwd1 expression disrupts purinosome formation, impacting NSPC proliferation and neuronal migration, resulting in periventricular heterotopia. Nwd1 interacts directly with phosphoribosylaminoimidazole-succinocarboxamide synthetase (PAICS), an enzyme involved in de novo purine synthesis. We anticipate this review will be valuable for researchers investigating neural development, purine metabolism, and protein-protein interactions.
Collapse
Affiliation(s)
- Seiya Yamada
- Laboratory for Molecular Neurobiology, Faculty of Human Sciences, Waseda University, Saitama, Japan
- Neuroscience Center, HiLIFE-Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tomoya Mizukoshi
- Laboratory for Molecular Neurobiology, Faculty of Human Sciences, Waseda University, Saitama, Japan
| | - Ayaka Sato
- Laboratory for Molecular Neurobiology, Faculty of Human Sciences, Waseda University, Saitama, Japan
| | - Shin-Ichi Sakakibara
- Laboratory for Molecular Neurobiology, Faculty of Human Sciences, Waseda University, Saitama, Japan
| |
Collapse
|
6
|
Kanika, Khan R. Functionalized nanomaterials targeting NLRP3 inflammasome driven immunomodulation: Friend or Foe. NANOSCALE 2023; 15:15906-15928. [PMID: 37750698 DOI: 10.1039/d3nr03857b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2023]
Abstract
The advancement in drug delivery systems in recent times has significantly enhanced therapeutic effects by enabling site-specific targeting through nanocarriers. These nanocarriers serve as invaluable tools for pharmacotherapeutic advancements against various disorders that enhance the effectiveness of encapsulated drugs by reducing their toxicity and increasing the efficacy of less potent drugs, thereby improving the therapeutic index. Inflammasomes, protein complexes located in the activated immune cell cytoplasm, regulate the activation of caspases involved in inflammation. However, aberrant activation of inflammasomes can result in uncontrolled tissue responses, contributing to the development of various diseases. Therefore, achieving a precise balance between inflammasome inhibition and activation is crucial for effectively treating inflammatory disorders through targeted functionalized nanocarriers. Despite the wealth of available data on the relevance of functionalized nanocarriers in inflammatory disorders, the nanotechnological potential to modulate inflammasomes has not been adequately explored. In this comprehensive review, we highlight the latest research on the modulation of the inflammasome cascade, both upregulating and downregulating its function, using nanocarriers in the context of inflammatory disorders. The utilization of nanocarriers as a therapeutic strategy holds immense potential for researchers aiming to effectively target and modulate inflammasomes in the treatment of inflammatory disorders, thus improving disease severity outcomes.
Collapse
Affiliation(s)
- Kanika
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Knowledge City, Sector-81, 5 Sahibzada Ajit Singh Nagar, Punjab, Pin - 140306, India.
| |
Collapse
|
7
|
Liu F, Zhuang W, Song B, Yang Y, Liu J, Zheng Y, Liu B, Zheng J, Zhao W, Gao C. MAVS-loaded unanchored Lys63-linked polyubiquitin chains activate the RIG-I-MAVS signaling cascade. Cell Mol Immunol 2023; 20:1186-1202. [PMID: 37582970 PMCID: PMC10542333 DOI: 10.1038/s41423-023-01065-2] [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: 03/14/2023] [Accepted: 07/05/2023] [Indexed: 08/17/2023] Open
Abstract
The adaptor molecule MAVS forms prion-like aggregates to govern the RIG-I-like receptor (RLR) signaling cascade. Lys63 (K63)-linked polyubiquitination is critical for MAVS aggregation, yet the underlying mechanism and the corresponding E3 ligases and deubiquitinating enzymes (DUBs) remain elusive. Here, we found that the K63-linked polyubiquitin chains loaded on MAVS can be directly recognized by RIG-I to initiate RIG-I-mediated MAVS aggregation with the prerequisite of the CARDRIG-I-CARDMAVS interaction. Interestingly, many K63-linked polyubiquitin chains attach to MAVS via an unanchored linkage. We identified Ube2N as a major ubiquitin-conjugating enzyme for MAVS and revealed that Ube2N cooperates with the E3 ligase Riplet and TRIM31 to promote the unanchored K63-linked polyubiquitination of MAVS. In addition, we identified USP10 as a direct DUB that removes unanchored K63-linked polyubiquitin chains from MAVS. Consistently, USP10 attenuates RIG-I-mediated MAVS aggregation and the production of type I interferon. Mice with a deficiency in USP10 show more potent resistance to RNA virus infection. Our work proposes a previously unknown mechanism for the activation of the RLR signaling cascade triggered by MAVS-attached unanchored K63-linked polyubiquitin chains and establishes the DUB USP10 and the E2:E3 pair Ube2N-Riplet/TRIM31 as a specific regulatory system for the unanchored K63-linked ubiquitination and aggregation of MAVS upon viral infection.
Collapse
Affiliation(s)
- Feng Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Wanxin Zhuang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Bin Song
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, P.R. China
| | - Yuan Yang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Junqi Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Jie Zheng
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 201203, Shanghai, P.R. China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, 250012, Shandong, P.R. China.
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, 250012, Shandong, P.R. China.
| |
Collapse
|
8
|
Dai T, Zhang L, Ran Y, Zhang M, Yang B, Lu H, Lin S, Zhang L, Zhou F. MAVS deSUMOylation by SENP1 inhibits its aggregation and antagonizes IRF3 activation. Nat Struct Mol Biol 2023; 30:785-799. [PMID: 37188808 DOI: 10.1038/s41594-023-00988-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 04/06/2023] [Indexed: 05/17/2023]
Abstract
Mitochondrial antiviral signaling protein (MAVS) is an adapter that recruits and activates IRF3. However, the mechanisms underpinning the interplay between MAVS and IRF3 are largely unknown. Here we show that small ubiquitin-like modifier (SUMO)-specific protease 1 negatively regulates antiviral immunity by deSUMOylating MAVS. Upon virus infection, PIAS3-induced poly-SUMOylation promotes lysine 63-linked poly-ubiquitination and aggregation of MAVS. Notably, we observe that SUMO conjugation is required for MAVS to efficiently produce phase-separated droplets through association with a newly identified SUMO-interacting motif (SIM) in MAVS. We further identify a yet-unknown SIM in IRF3 that mediates its enrichment to the multivalent MAVS droplets. Conversely, IRF3 phosphorylation at crucial residues close to SIM rapidly disables SUMO-SIM interactions and releases activated IRF3 from MAVS. Our findings implicate SUMOylation in MAVS phase separation and suggest a thus far unknown regulatory process by which IRF3 can be efficiently recruited and released to facilitate timely activation of antiviral responses.
Collapse
Affiliation(s)
- Tong Dai
- Center for Infection & Immunity of International Institutes of Medicine, The Fourth Affiliated Hospital, ZheJiang University School of Medicine, Yiwu, China
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Lei Zhang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Ran
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Meirong Zhang
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China
| | - Bing Yang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
- Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Huasong Lu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Shixian Lin
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
| | - Fangfang Zhou
- Center for Infection & Immunity of International Institutes of Medicine, The Fourth Affiliated Hospital, ZheJiang University School of Medicine, Yiwu, China.
- Institutes of Biology and Medical Science, Soochow University, Suzhou, China.
| |
Collapse
|
9
|
Vargas JNS, Hamasaki M, Kawabata T, Youle RJ, Yoshimori T. The mechanisms and roles of selective autophagy in mammals. Nat Rev Mol Cell Biol 2023; 24:167-185. [PMID: 36302887 DOI: 10.1038/s41580-022-00542-2] [Citation(s) in RCA: 475] [Impact Index Per Article: 237.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2022] [Indexed: 11/09/2022]
Abstract
Autophagy is a process that targets various intracellular elements for degradation. Autophagy can be non-selective - associated with the indiscriminate engulfment of cytosolic components - occurring in response to nutrient starvation and is commonly referred to as bulk autophagy. By contrast, selective autophagy degrades specific targets, such as damaged organelles (mitophagy, lysophagy, ER-phagy, ribophagy), aggregated proteins (aggrephagy) or invading bacteria (xenophagy), thereby being importantly involved in cellular quality control. Hence, not surprisingly, aberrant selective autophagy has been associated with various human pathologies, prominently including neurodegeneration and infection. In recent years, considerable progress has been made in understanding mechanisms governing selective cargo engulfment in mammals, including the identification of ubiquitin-dependent selective autophagy receptors such as p62, NBR1, OPTN and NDP52, which can bind cargo and ubiquitin simultaneously to initiate pathways leading to autophagy initiation and membrane recruitment. This progress opens the prospects for enhancing selective autophagy pathways to boost cellular quality control capabilities and alleviate pathology.
Collapse
Affiliation(s)
- Jose Norberto S Vargas
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, University College London, London, UK
- UK Dementia Research Institute, University College London, London, UK
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Maho Hamasaki
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| | - Tsuyoshi Kawabata
- Department of Stem Cell Biology, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Richard J Youle
- Biochemistry Section, Surgical Neurology Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan.
- Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| |
Collapse
|
10
|
Wang J, Song W, Chai J. Structure, biochemical function, and signaling mechanism of plant NLRs. MOLECULAR PLANT 2023; 16:75-95. [PMID: 36415130 DOI: 10.1016/j.molp.2022.11.011] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/07/2022] [Accepted: 11/21/2022] [Indexed: 06/16/2023]
Abstract
To counter pathogen invasion, plants have evolved a large number of immune receptors, including membrane-resident pattern recognition receptors (PRRs) and intracellular nucleotide-binding and leucine-rich repeat receptors (NLRs). Our knowledge about PRR and NLR signaling mechanisms has expanded significantly over the past few years. Plant NLRs form multi-protein complexes called resistosomes in response to pathogen effectors, and the signaling mediated by NLR resistosomes converges on Ca2+-permeable channels. Ca2+-permeable channels important for PRR signaling have also been identified. These findings highlight a crucial role of Ca2+ in triggering plant immune signaling. In this review, we first discuss the structural and biochemical mechanisms of non-canonical NLR Ca2+ channels and then summarize our knowledge about immune-related Ca2+-permeable channels and their roles in PRR and NLR signaling. We also discuss the potential role of Ca2+ in the intricate interaction between PRR and NLR signaling.
Collapse
Affiliation(s)
- Jizong Wang
- State Key Laboratory of Protein and Plant Gene Research, School of Advanced Agricultural Sciences, Peking University, Beijing 100871, China; Peking University Institute of Advanced Agricultural Sciences, Shandong Laboratory of Advanced Agricultural Sciences at Weifang, Weifang, Shandong 261000, China.
| | - Wen Song
- Institute of Biochemistry, University of Cologne, 50674 Cologne, Germany; Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany.
| | - Jijie Chai
- Tsinghua-Peking Joint Center for Life Sciences, Center for Plant Biology, School of Life Sciences, Tsinghua University, Beijing 100084, China; Institute of Biochemistry, University of Cologne, 50674 Cologne, Germany; Max Planck Institute for Plant Breeding Research, 50829 Cologne, Germany.
| |
Collapse
|
11
|
Li Y, Jiang Q. Uncoupled pyroptosis and IL-1β secretion downstream of inflammasome signaling. Front Immunol 2023; 14:1128358. [PMID: 37090724 PMCID: PMC10117957 DOI: 10.3389/fimmu.2023.1128358] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 03/24/2023] [Indexed: 04/25/2023] Open
Abstract
Inflammasomes are supramolecular platforms that organize in response to various damage-associated molecular patterns and pathogen-associated molecular patterns. Upon activation, inflammasome sensors (with or without the help of ASC) activate caspase-1 and other inflammatory caspases that cleave gasdermin D and pro-IL-1β/pro-IL-18, leading to pyroptosis and mature cytokine secretion. Pyroptosis enables intracellular pathogen niche disruption and intracellular content release at the cost of cell death, inducing pro-inflammatory responses in the neighboring cells. IL-1β is a potent pro-inflammatory regulator for neutrophil recruitment, macrophage activation, and T-cell expansion. Thus, pyroptosis and cytokine secretion are the two main mechanisms that occur downstream of inflammasome signaling; they maintain homeostasis, drive the innate immune response, and shape adaptive immunity. This review aims to discuss the possible mechanisms, timing, consequences, and significance of the two uncoupling preferences downstream of inflammasome signaling. While pyroptosis and cytokine secretion may be usually coupled, pyroptosis-predominant and cytokine-predominant uncoupling are also observed in a stimulus-, cell type-, or context-dependent manner, contributing to the pathogenesis and development of numerous pathological conditions such as cryopyrin-associated periodic syndromes, LPS-induced sepsis, and Salmonella enterica serovar Typhimurium infection. Hyperactive cells consistently release IL-1β without LDH leakage and pyroptotic death, thereby leading to prolonged inflammation, expanding the lifespans of pyroptosis-resistant neutrophils, and hyperactivating stimuli-challenged macrophages, dendritic cells, monocytes, and specific nonimmune cells. Death inflammasome activation also induces GSDMD-mediated pyroptosis with no IL-1β secretion, which may increase lethality in vivo. The sublytic GSDMD pore formation associated with lower expressions of pyroptotic components, GSDMD-mediated extracellular vesicles, or other GSDMD-independent pathways that involve unconventional secretion could contribute to the cytokine-predominant uncoupling; the regulation of caspase-1 dynamics, which may generate various active species with different activities in terms of GSDMD or pro-IL-1β, could lead to pyroptosis-predominant uncoupling. These uncoupling preferences enable precise reactions to different stimuli of different intensities under specific conditions at the single-cell level, promoting cooperative cell and host fate decisions and participating in the pathogen "game". Appropriate decisions in terms of coupling and uncoupling are required to heal tissues and eliminate threats, and further studies exploring the inflammasome tilt toward pyroptosis or cytokine secretion may be helpful.
Collapse
|
12
|
Yamada S, Furukawa R, Sakakibara SI. Identification and expression profile of novel STAND gene Nwd2 in the mouse central nervous system. Gene Expr Patterns 2022; 46:119284. [PMID: 36341976 DOI: 10.1016/j.gep.2022.119284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 10/18/2022] [Accepted: 10/22/2022] [Indexed: 11/04/2022]
Abstract
In the central nervous system (CNS), neurons need synaptic neurotransmitter release and cellular response for various cellular stress or environmental stimuli. To achieve these highly orchestrated cellular processes, neurons should drive the molecular mechanisms that govern and integrate complex signaling pathways. The signal transduction ATPases with numerous domains (STAND) family of proteins has been shown to play essential roles in diverse signal transduction mechanisms, including apoptosis and innate immunity. However, a comprehensive understanding of STAND genes remains lacking. Previously, we identified the NACHT and WD repeat domain-containing protein 1 (NWD1), a member of STAND family, in the regulation of the assembly of a giant multi-enzyme complex that enables efficient de novo purine biosynthesis during brain development. Here we identified the mouse Nwd2 gene, which is a paralog of Nwd1. A molecular phylogenetic analysis suggested that Nwd1 emerged during the early evolution of the animal kingdom, and that Nwd2 diverged in the process of Nwd1 duplication. RT-PCR and in situ hybridization analyses revealed the unique expression profile of Nwd2 in the developing and adult CNS. Unlike Nwd1, Nwd2 expression was primarily confined to neurons in the medial habenular nucleus, an essential modulating center for diverse psychological states, such as fear, anxiety, and drug addiction. In the adult brain, Nwd2 expression, albeit at a lower level, was also observed in some neuronal populations in the piriform cortex, hippocampus, and substantia nigra pars compacta. NWD2 might play a unique role in the signal transduction required for specific neuronal circuits, especially for cholinergic neurons in the habenula.
Collapse
Affiliation(s)
- Seiya Yamada
- Laboratory for Molecular Neurobiology, Faculty of Human Sciences, Waseda University, Tokorozawa, Saitama, 359-1192, Japan.
| | - Ryutaro Furukawa
- Laboratory of Life Science for Extremophiles, Faculty of Human Sciences, Waseda University, Tokorozawa, Saitama, 359-1192, Japan
| | - Shin-Ichi Sakakibara
- Laboratory for Molecular Neurobiology, Faculty of Human Sciences, Waseda University, Tokorozawa, Saitama, 359-1192, Japan.
| |
Collapse
|
13
|
Zhuang W, Zhang L, Zheng Y, Liu B, Ma C, Zhao W, Liu S, Liu F, Gao C. USP3 deubiquitinates and stabilizes the adapter protein ASC to regulate inflammasome activation. Cell Mol Immunol 2022; 19:1141-1152. [PMID: 36050480 PMCID: PMC9508167 DOI: 10.1038/s41423-022-00917-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 08/06/2022] [Indexed: 11/08/2022] Open
Abstract
Inflammasomes are essential components of the innate immune system and its defense against infections, whereas the dysregulation of inflammasome activation has a detrimental effect on human health. The activation of inflammasomes is subjected to tight regulation to maintain immune homeostasis, yet the underlying mechanism remains elusive. Here, we identify USP3 as a direct deubiquitinating enzyme (DUB) for ASC, the central adapter mediating the assembly and activation of most inflammasomes. USP3 removes the K48-linked ubiquitination on ASC and strengthens its stability by blocking proteasomal degradation. Additionally, USP3 promotes inflammasome activation, and this function was confirmed in mouse models of aluminum (Alum)-induced peritonitis, F. novicida infection and flagellin-induced pneumonia in vivo. Our work unveils that USP3 functions as a key regulator of ASC ubiquitination and maintains the physiological role of ASC in mediating inflammasome activation, and we propose a new mechanism by which the ubiquitination of ASC regulates inflammasome activation.
Collapse
Affiliation(s)
- Wanxin Zhuang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lei Zhang
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chunhong Ma
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Wei Zhao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Pathogenic Biology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Suxia Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Feng Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China.
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Key Laboratory for Experimental Teratology of Ministry of Education, Shandong University, Jinan, Shandong, 250012, PR China.
- Department of Immunology, School of Biomedical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
14
|
Essuman K, Milbrandt J, Dangl JL, Nishimura MT. Shared TIR enzymatic functions regulate cell death and immunity across the tree of life. Science 2022; 377:eabo0001. [DOI: 10.1126/science.abo0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the 20th century, researchers studying animal and plant signaling pathways discovered a protein domain shared across diverse innate immune systems: the Toll/Interleukin-1/Resistance-gene (TIR) domain. The TIR domain is found in several protein architectures and was defined as an adaptor mediating protein-protein interactions in animal innate immunity and developmental signaling pathways. However, studies of nerve degeneration in animals, and subsequent breakthroughs in plant, bacterial and archaeal systems, revealed that TIR domains possess enzymatic activities. We provide a synthesis of TIR functions and the role of various related TIR enzymatic products in evolutionarily diverse immune systems. These studies may ultimately guide interventions that would span the tree of life, from treating human neurodegenerative disorders and bacterial infections, to preventing plant diseases.
Collapse
Affiliation(s)
- Kow Essuman
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey Milbrandt
- Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- Needleman Center for Neurometabolism and Axonal Therapeutics, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
- McDonnell Genome Institute, Washington University School of Medicine in St. Louis, St. Louis, MO 63110, USA
| | - Jeffery L. Dangl
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Howard Hughes Medical Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Marc T. Nishimura
- Department of Biology, Colorado State University, Fort Collins, CO 80523, USA
| |
Collapse
|
15
|
Ren Z, Yu Y, Chen C, Yang D, Ding T, Zhu L, Deng J, Xu Z. The Triangle Relationship Between Long Noncoding RNA, RIG-I-like Receptor Signaling Pathway, and Glycolysis. Front Microbiol 2021; 12:807737. [PMID: 34917069 PMCID: PMC8670088 DOI: 10.3389/fmicb.2021.807737] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 11/09/2021] [Indexed: 12/11/2022] Open
Abstract
Long noncoding RNA (LncRNA), a noncoding RNA over 200nt in length, can regulate glycolysis through metabolic pathways, glucose metabolizing enzymes, and epigenetic reprogramming. Upon viral infection, increased aerobic glycolysis providzes material and energy for viral replication. Mitochondrial antiviral signaling protein (MAVS) is the only protein-specified downstream of retinoic acid-inducible gene I (RIG-I) that bridges the gap between antiviral immunity and glycolysis. MAVS binding to RIG-I inhibits MAVS binding to Hexokinase (HK2), thereby impairing glycolysis, while excess lactate production inhibits MAVS and the downstream antiviral immune response, facilitating viral replication. LncRNAs can also regulate antiviral innate immunity by interacting with RIG-I and downstream signaling pathways and by regulating the expression of interferons and interferon-stimulated genes (ISGs). Altogether, we summarize the relationship between glycolysis, antiviral immunity, and lncRNAs and propose that lncRNAs interact with glycolysis and antiviral pathways, providing a new perspective for the future treatment against virus infection, including SARS-CoV-2.
Collapse
Affiliation(s)
- Zhihua Ren
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Yueru Yu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Chaoxi Chen
- College of Life Since and Technology, Southwest Minzu University, Chengdu, China
| | - Dingyong Yang
- College of Animal Husbandry and Veterinary Medicine, Chengdu Agricultural College, Chengdu, China
| | - Ting Ding
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Ling Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Junliang Deng
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| | - Zhiwen Xu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
16
|
Luciani A, Denley MCS, Govers LP, Sorrentino V, Froese DS. Mitochondrial disease, mitophagy, and cellular distress in methylmalonic acidemia. Cell Mol Life Sci 2021; 78:6851-6867. [PMID: 34524466 PMCID: PMC8558192 DOI: 10.1007/s00018-021-03934-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 08/18/2021] [Accepted: 08/30/2021] [Indexed: 01/09/2023]
Abstract
Mitochondria—the intracellular powerhouse in which nutrients are converted into energy in the form of ATP or heat—are highly dynamic, double-membraned organelles that harness a plethora of cellular functions that sustain energy metabolism and homeostasis. Exciting new discoveries now indicate that the maintenance of this ever changing and functionally pleiotropic organelle is particularly relevant in terminally differentiated cells that are highly dependent on aerobic metabolism. Given the central role in maintaining metabolic and physiological homeostasis, dysregulation of the mitochondrial network might therefore confer a potentially devastating vulnerability to high-energy requiring cell types, contributing to a broad variety of hereditary and acquired diseases. In this Review, we highlight the biological functions of mitochondria-localized enzymes from the perspective of understanding—and potentially reversing—the pathophysiology of inherited disorders affecting the homeostasis of the mitochondrial network and cellular metabolism. Using methylmalonic acidemia as a paradigm of complex mitochondrial dysfunction, we discuss how mitochondrial directed-signaling circuitries govern the homeostasis and physiology of specialized cell types and how these may be disturbed in disease. This Review also provides a critical analysis of affected tissues, potential molecular mechanisms, and novel cellular and animal models of methylmalonic acidemia which are being used to develop new therapeutic options for this disease. These insights might ultimately lead to new therapeutics, not only for methylmalonic acidemia, but also for other currently intractable mitochondrial diseases, potentially transforming our ability to regulate homeostasis and health.
Collapse
Affiliation(s)
- Alessandro Luciani
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8032, Zurich, Switzerland.
| | - Matthew C S Denley
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, 8032, Zurich, Switzerland
| | - Larissa P Govers
- Mechanisms of Inherited Kidney Diseases Group, Institute of Physiology, University of Zurich, 8032, Zurich, Switzerland
| | - Vincenzo Sorrentino
- Department of Musculo-Skeletal Health, Nestlé Institute of Health Sciences, Nestlé Research, 1015, Lausanne, Switzerland.
| | - D Sean Froese
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, 8032, Zurich, Switzerland.
| |
Collapse
|
17
|
Shaldzhyan AA, Zabrodskaya YA, Baranovskaya IL, Sergeeva MV, Gorshkov AN, Savin II, Shishlyannikov SM, Ramsay ES, Protasov AV, Kukhareva AP, Egorov VV. Old dog, new tricks: Influenza A virus NS1 and in vitro fibrillogenesis. Biochimie 2021; 190:50-56. [PMID: 34273416 DOI: 10.1016/j.biochi.2021.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 11/24/2022]
Abstract
The influenza NS1 protein is involved in suppression of the host immune response. Recently, there is growing evidence that prion-like protein aggregation plays an important role in cellular signaling and immune responses. In this work, we obtained a recombinant, influenza A NS1 protein and showed that it is able to form amyloid-like fibrils in vitro. Using proteolysis and subsequent mass spectrometry, we showed that regions resistant to protease hydrolysis highly differ between the native NS1 form (NS1-N) and fibrillar form (NS1-F); this indicates that significant structural changes occur during fibril formation. We also found a protein fragment that is capable of inducing the process of fibrillogenesis at 37 °C. The discovery of the ability of NS1 to form amyloid-like fibrils may be relevant to uncovering relationships between influenza A infection and modulation of the immune response.
Collapse
Affiliation(s)
- A A Shaldzhyan
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia; Petersburg Nuclear Physics Institute Named By B. P. Konstantinov of the National Research Center "Kurchatov Institute", 188300, mkr. Orlova Roshcha 1, Gatchina, Russia
| | - Y A Zabrodskaya
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia; Petersburg Nuclear Physics Institute Named By B. P. Konstantinov of the National Research Center "Kurchatov Institute", 188300, mkr. Orlova Roshcha 1, Gatchina, Russia; National Research Centre Kurchatov Institute, 123182, Akademika Kurchatova Sq. 1, Moscow, Russia; Peter the Great St. Petersburg Polytechnic University, 194064, Polyteknicheskaya 29, St. Petersburg, Russia.
| | - I L Baranovskaya
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia; Peter the Great St. Petersburg Polytechnic University, 194064, Polyteknicheskaya 29, St. Petersburg, Russia
| | - M V Sergeeva
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia
| | - A N Gorshkov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia
| | - I I Savin
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia
| | - S M Shishlyannikov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia; All-Russia Research Institute for Food Additives - Branch of V.M. Gorbatov Federal Research Center for Food Systems of RAS, 191014, Liteyny Av. 55, St. Petersburg, Russia
| | - E S Ramsay
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia
| | - A V Protasov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia; Peter the Great St. Petersburg Polytechnic University, 194064, Polyteknicheskaya 29, St. Petersburg, Russia
| | - A P Kukhareva
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia
| | - V V Egorov
- Smorodintsev Research Institute of Influenza, Russian Ministry of Health, 197376, Prof. Popov 15/17, St. Petersburg, Russia; Petersburg Nuclear Physics Institute Named By B. P. Konstantinov of the National Research Center "Kurchatov Institute", 188300, mkr. Orlova Roshcha 1, Gatchina, Russia; National Research Centre Kurchatov Institute, 123182, Akademika Kurchatova Sq. 1, Moscow, Russia; Federal State Budgetary Scientific Institution "Institute of Experimental Medicine", 197376, Akademika Pavlova 12, St. Petersburg, Russia
| |
Collapse
|
18
|
Kim SH, Shin HJ, Yoon CM, Lee SW, Sharma L, Dela Cruz CS, Kang MJ. PINK1 Inhibits Multimeric Aggregation and Signaling of MAVS and MAVS-Dependent Lung Pathology. Am J Respir Cell Mol Biol 2021; 64:592-603. [PMID: 33577398 PMCID: PMC8086043 DOI: 10.1165/rcmb.2020-0490oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Accepted: 01/20/2021] [Indexed: 12/16/2022] Open
Abstract
Mitochondria have emerged as important signaling organelles where intracellular perturbations are integrated and, consequently, intracellular signaling pathways are modulated to execute appropriate cellular functions. MAVS (mitochondrial antiviral signaling protein) represents such an example that functions as a platform molecule to mediate mitochondrial innate immune signaling. Recently, multimeric aggregation of MAVS has been identified as a key molecular process for its signaling. The underlying mechanisms to regulate this, however, are still incompletely understood. We hypothesized that PINK1 (PTEN-induced kinase 1) plays an important role in the regulation of multimeric MAVS aggregation and its consequent pathobiology. To test whether PINK1 interacts with MAVS, bimolecular fluorescence complementation analysis and IP were performed. RLH (RIG-I-like helicase) and NLRP3 inflammasome signaling were evaluated by in vitro assay. In vivo functional significance of PINK1 in the regulation of MAVS signaling was evaluated from both murine modeling of influenza viral infection and bleomycin-induced experimental pulmonary fibrosis, wherein MAVS plays important roles. Multimeric MAVS aggregation was induced by mitochondria dysfunction, and, during this event, the stabilized PINK1 interacted physically with MAVS and antagonized multimeric MAVS aggregation. Accordingly, the MAVS-mediated antiviral innate immune and NLRP3 inflammasome signaling were enhanced in PINK1 deficiency. In addition, in vivo studies revealed that MAVS-mediated pulmonary antiviral innate immune responses and fibrotic responses after bleomycin injury were enhanced in PINK1 deficiency. In conclusion, these results establish a new role of PINK1 in the regulation of MAVS signaling and the consequent pulmonary pathobiology.
Collapse
Affiliation(s)
- Sang-Hun Kim
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Hyeon Jun Shin
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Chang Min Yoon
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Sei Won Lee
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
- Department of Pulmonary and Critical Care Medicine, and Clinical Research Center for Chronic Obstructive Airway Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Lokesh Sharma
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Charles S. Dela Cruz
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| | - Min-Jong Kang
- Department of Internal Medicine, Section of Pulmonary, Critical Care and Sleep Medicine, Yale University School of Medicine, New Haven, Connecticut; and
| |
Collapse
|
19
|
Chen J, Zhang X, Li L, Ma X, Yang C, Liu Z, Li C, Fernandez-Cabezudo MJ, al-Ramadi BK, Wu C, Huang W, Zhang Y, Zhang Y, Liu W. Farnesyl pyrophosphate is a new danger signal inducing acute cell death. PLoS Biol 2021; 19:e3001134. [PMID: 33901180 PMCID: PMC8075202 DOI: 10.1371/journal.pbio.3001134] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 02/03/2021] [Indexed: 11/19/2022] Open
Abstract
Cell death is a vital event in life. Infections and injuries cause lytic cell death, which gives rise to danger signals that can further induce cell death, inflammation, and tissue damage. The mevalonate (MVA) pathway is an essential, highly conserved and dynamic metabolic pathway. Here, we discover that farnesyl pyrophosphate (FPP), a metabolic intermediate of the MVA pathway, functions as a newly identified danger signal to trigger acute cell death leading to neuron loss in stroke. Harboring both a hydrophobic 15-carbon isoprenyl chain and a heavily charged pyrophosphate head, FPP leads to acute cell death independent of its downstream metabolic pathways. Mechanistically, extracellular calcium influx and the cation channel transient receptor potential melastatin 2 (TRPM2) exhibit essential roles in FPP-induced cell death. FPP activates TRPM2 opening for ion influx. Furthermore, in terms of a mouse model constructing by middle cerebral artery occlusion (MCAO), FPP accumulates in the brain, which indicates the function of the FPP and TRPM2 danger signal axis in ischemic injury. Overall, our data have revealed a novel function of the MVA pathway intermediate metabolite FPP as a danger signal via transient receptor potential cation channels.
Collapse
Affiliation(s)
- Jing Chen
- School of Life Sciences, Institute for Immunology, Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
| | - Xiaochen Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of People’s Republic of China, IDG/McGovern Institute for Brain Research at Peking University, Beijing, China
| | - Liping Li
- Tsinghua-Peking Center for Life Sciences, School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing Advanced Innovation Center for Human Brain Protection, Beijing, China
| | - Xianqiang Ma
- Tsinghua-Peking Center for Life Sciences, School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing Advanced Innovation Center for Human Brain Protection, Beijing, China
| | - Chunxiao Yang
- School of Medicine, Nankai University, Tianjin, China
| | - Zhaodi Liu
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of People’s Republic of China, IDG/McGovern Institute for Brain Research at Peking University, Beijing, China
| | - Chenyang Li
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of People’s Republic of China, IDG/McGovern Institute for Brain Research at Peking University, Beijing, China
| | - Maria J. Fernandez-Cabezudo
- Department of Biochemistry and Molecular Biology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Basel K. al-Ramadi
- Department of Medical Microbiology & Immunology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, New York, United States of America
| | - Yong Zhang
- Neuroscience Research Institute and Department of Neurobiology, School of Basic Medical Sciences, Peking University, Key Laboratory for Neuroscience, Ministry of Education/National Health Commission of People’s Republic of China, IDG/McGovern Institute for Brain Research at Peking University, Beijing, China
| | - Yonghui Zhang
- Tsinghua-Peking Center for Life Sciences, School of Pharmaceutical Sciences, Ministry of Education Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing Advanced Innovation Center for Human Brain Protection, Beijing, China
| | - Wanli Liu
- School of Life Sciences, Institute for Immunology, Ministry of Education Key Laboratory of Protein Sciences, Beijing Advanced Innovation Center for Structural Biology, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
20
|
Targeting the NLRP3 inflammasome as new therapeutic avenue for inflammatory bowel disease. Biomed Pharmacother 2021; 138:111442. [PMID: 33667791 DOI: 10.1016/j.biopha.2021.111442] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The incidence and prevalence of inflammatory bowel disease (IBD) are increasing worldwide. Current approved medication for IBD treatment in the clinic mainly includes corticosteroids and neutralization antibodies to pro-inflammatory cytokines. However, drug resistance and severe side effect hinder long-term efficacy of these agents. The NOD-like receptor family pyrin domain containing protein 3 (NLRP3) is exclusively expressed in several inflammatory and autoimmune diseases. Excessive expression, aberrant activation, polymorphism, and gain-of-function mutation of the NLRP3 inflammasome contribute to IBD pathogenesis. In this article, we summarize the regulatory factors to NLRP3, and review recently developed NLRP3 inhibitors and their preclinical and clinical applications in treating inflammatory and autoimmune diseases. We present our views on the therapeutic potential of NLRP3 inhibitors as emerging therapeutic avenue for IBD.
Collapse
|
21
|
Wang HT, Hur S. Substrate recognition by TRIM and TRIM-like proteins in innate immunity. Semin Cell Dev Biol 2021; 111:76-85. [PMID: 33092958 PMCID: PMC7572318 DOI: 10.1016/j.semcdb.2020.09.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 09/28/2020] [Indexed: 12/23/2022]
Abstract
TRIM (Tripartite motif) and TRIM-like proteins have emerged as an important class of E3 ligases in innate immunity. Their functions range from activation or regulation of innate immune signaling pathway to direct detection and restriction of pathogens. Despite the importance, molecular mechanisms for many TRIM/TRIM-like proteins remain poorly characterized, in part due to challenges of identifying their substrates. In this review, we discuss several TRIM/TRIM-like proteins in RNA sensing pathways and viral restriction functions. We focus on those containing PRY-SPRY, the domain most frequently used for substrate recognition, and discuss emerging mechanisms that are commonly utilized by several TRIM/TRIM-like proteins to tightly control their interaction with the substrates.
Collapse
Affiliation(s)
- Hai-Tao Wang
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA
| | - Sun Hur
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
22
|
Wan WL, Kim ST, Castel B, Charoennit N, Chae E. Genetics of autoimmunity in plants: an evolutionary genetics perspective. THE NEW PHYTOLOGIST 2021; 229:1215-1233. [PMID: 32970825 DOI: 10.1111/nph.16947] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/12/2020] [Indexed: 05/14/2023]
Abstract
Autoimmunity in plants has been found in numerous hybrids as a form of hybrid necrosis and mutant panels. Uncontrolled cell death is a main cellular outcome of autoimmunity, which negatively impacts growth. Its occurrence highlights the vulnerable nature of the plant immune system. Genetic investigation of autoimmunity in hybrid plants revealed that extreme variation in the immune receptor repertoire is a major contributor, reflecting an evolutionary conundrum that plants face in nature. In this review, we discuss natural variation in the plant immune system and its contribution to fitness. The value of autoimmunity genetics lies in its ability to identify combinations of a natural immune receptor and its partner that are predisposed to triggering autoimmunity. The network of immune components for autoimmunity becomes instrumental in revealing mechanistic details of how immune receptors recognize cellular invasion and activate signaling. The list of autoimmunity-risk variants also allows us to infer evolutionary processes contributing to their maintenance in the natural population. Our approach to autoimmunity, which integrates mechanistic understanding and evolutionary genetics, has the potential to serve as a prognosis tool to optimize immunity in crops.
Collapse
Affiliation(s)
- Wei-Lin Wan
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Sang-Tae Kim
- Department of Life Sciences, The Catholic University of Korea, Bucheon, Gyeonggi-do, 14662, South Korea
| | - Baptiste Castel
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Nuri Charoennit
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| | - Eunyoung Chae
- Department of Biological Sciences, National University of Singapore, Singapore, 117558, Singapore
| |
Collapse
|
23
|
Sun B, Zeng H, Liang J, Zhang L, Hu H, Wang Q, Meng W, Li C, Ye F, Wang C, Zhu J. NSUN5 Facilitates Viral RNA Recognition by RIG-I Receptor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 205:3408-3418. [PMID: 33177158 DOI: 10.4049/jimmunol.1901455] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 10/14/2020] [Indexed: 11/19/2022]
Abstract
The RIG-I receptor induces the innate antiviral responses upon sensing RNA viruses. The mechanisms through which RIG-I optimizes the strength of the downstream signaling remain incompletely understood. In this study, we identified that NSUN5 could potentiate the RIG-I innate signaling pathway. Deficiency of NSUN5 enhanced RNA virus proliferation and inhibited the induction of the downstream antiviral genes. Consistently, NSUN5-deficient mice were more susceptible to RNA virus infection than their wild-type littermates. Mechanistically, NSUN5 bound directly to both viral RNA and RIG-I, synergizing the recognition of dsRNA by RIG-I. Collectively, to our knowledge, this study characterized NSUN5 as a novel RIG-I coreceptor, playing a vital role in restricting RNA virus infection.
Collapse
Affiliation(s)
- Boyue Sun
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Haoyang Zeng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Jiaqian Liang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Lele Zhang
- Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; and
| | - Haiyang Hu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Quanyi Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Wei Meng
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Chenhui Li
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Fuqiang Ye
- Department of Disease Control and Prevention, Center for Disease Control and Prevention of Eastern Theater Command, Nanjing 210002, China
| | - Chen Wang
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China;
| | - Juanjuan Zhu
- State Key Laboratory of Natural Medicines, School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China;
| |
Collapse
|
24
|
Su Z, Dhusia K, Wu Y. Understand the Functions of Scaffold Proteins in Cell Signaling by a Mesoscopic Simulation Method. Biophys J 2020; 119:2116-2126. [PMID: 33113350 DOI: 10.1016/j.bpj.2020.10.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/24/2020] [Accepted: 10/07/2020] [Indexed: 02/02/2023] Open
Abstract
Scaffold proteins are central players in regulating the spatial-temporal organization of many important signaling pathways in cells. They offer physical platforms to downstream signaling proteins so that their transient interactions in a crowded and heterogeneous environment of cytosol can be greatly facilitated. However, most scaffold proteins tend to simultaneously bind more than one signaling molecule, which leads to the spatial assembly of multimeric protein complexes. The kinetics of these protein oligomerizations are difficult to quantify by traditional experimental approaches. To understand the functions of scaffold proteins in cell signaling, we developed a, to our knowledge, new hybrid simulation algorithm in which both spatial organization and binding kinetics of proteins were implemented. We applied this new technique to a simple network system that contains three molecules. One molecule in the network is a scaffold protein, whereas the other two are its binding targets in the downstream signaling pathway. Each of the three molecules in the system contains two binding motifs that can interact with each other and are connected by a flexible linker. By applying the new simulation method to the model, we show that the scaffold proteins will promote not only thermodynamics but also kinetics of cell signaling given the premise that the interaction between the two signaling molecules is transient. Moreover, by changing the flexibility of the linker between two binding motifs, our results suggest that the conformational fluctuations in a scaffold protein play a positive role in recruiting downstream signaling molecules. In summary, this study showcases the capability of computational simulation in understanding the general principles of scaffold protein functions.
Collapse
Affiliation(s)
- Zhaoqian Su
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Kalyani Dhusia
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Yinghao Wu
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, New York.
| |
Collapse
|
25
|
Kazimirskii AN, Salmasi JM, Poryadin GV. Antiviral system of innate immunity: COVID-19 pathogenesis and treatment. BULLETIN OF RUSSIAN STATE MEDICAL UNIVERSITY 2020. [DOI: 10.24075/brsmu.2020.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Antiviral system of innate immunity includes two main components: the mitochondrial antiviral sensor — the mitochondrial outer membrane protein and peripheral blood neutrophils capable of forming neutrophilic extracellular traps. Depending on the activation pathway of the mitochondrial antiviral sensor (MAVS), two possible variants of cells death, apoptosis or cellular degeneration with necrotic changes, develop during cell infection with an RNA-containing virus. The development of virus-induced apoptosis of infected cells causes the formation of neutrophilic extracellular traps, the secretion of inflammatory cytokines, ROS generation, tissue damage, hemocoagulation and the development of an acute inflammatory process with the development of COVID-19 pneumonia. Violation of the prion-like reaction of MAVS in response to viral infection of the cell triggers an alternative pathway for activating autophagy. Cells under conditions of prolonged activation of autophagy experience necrotic changes and are eliminated from the organism by monocytes/macrophages that secrete anti-inflammatory cytokines. This type of reaction of the antiviral system of innate immunity corresponds to the asymptomatic course of the disease. From the most significant aspects of the pathogenesis of the coronavirus infection COVID-19 given, recommendations for the prophylactic treatment of this dangerous disease follow. The proposed treatment can significantly decrease the severity of COVID-19 disease and reduce mortality.
Collapse
Affiliation(s)
- AN Kazimirskii
- Pirogov Russian National Research Medical University. Moscow, Russia
| | - JM Salmasi
- Pirogov Russian National Research Medical University. Moscow, Russia
| | - GV Poryadin
- Pirogov Russian National Research Medical University. Moscow, Russia
| |
Collapse
|
26
|
Abstract
Amyloids are implicated in many protein misfolding diseases. Amyloid folds, however, also display a range of functional roles particularly in the microbial world. The templating ability of these folds endows them with specific properties allowing their self-propagation and protein-to-protein transmission in vivo. This property, the prion principle, is exploited by specific signaling pathways that use transmission of the amyloid fold as a way to convey information from a receptor to an effector protein. I describe here amyloid signaling pathways involving fungal nucleotide binding and oligomerization domain (NOD)-like receptors that were found to control nonself recognition and programmed cell death processes. Studies on these fungal amyloid signaling motifs stem from the characterization of the fungal [Het-s] prion protein and have led to the identification in fungi but also in multicellular bacteria of several distinct families of signaling motifs, one of which is related to RHIM [receptor-interacting protein (RIP) homotypic interaction motif], an amyloid motif regulating mammalian necroptosis.
Collapse
Affiliation(s)
- Sven J. Saupe
- Institut de Biochimie et de Génétique Cellulaire, UMR 5095 CNRS, Université de Bordeaux, 33077 Bordeaux CEDEX, France
| |
Collapse
|
27
|
Yamada S, Sato A, Sakakibara SI. Nwd1 Regulates Neuronal Differentiation and Migration through Purinosome Formation in the Developing Cerebral Cortex. iScience 2020; 23:101058. [PMID: 32344379 PMCID: PMC7186558 DOI: 10.1016/j.isci.2020.101058] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 03/09/2020] [Accepted: 04/08/2020] [Indexed: 12/17/2022] Open
Abstract
Engagement of neural stem/progenitor cells (NSPCs) into proper neuronal differentiation requires the spatiotemporally regulated generation of metabolites. Purines are essential building blocks for many signaling molecules. Enzymes that catalyze de novo purine synthesis are assembled as a huge multienzyme complex called “purinosome.” However, there is no evidence of the formation or physiological function of the purinosome in the brain. Here, we showed that a signal transduction ATPases with numerous domains (STAND) protein, NACHT and WD repeat domain-containing 1 (Nwd1), interacted with Paics, a purine-synthesizing enzyme, to regulate purinosome assembly in NSPCs. Altered Nwd1 expression affected purinosome formation and induced the mitotic exit and premature differentiation of NSPCs, repressing neuronal migration and periventricular heterotopia. Overexpression/knockdown of Paics or Fgams, other purinosome enzymes, in the developing brain resulted in a phenocopy of Nwd1 defects. These findings indicate that strict regulation of purinosome assembly/disassembly is crucial for maintaining NSPCs and corticogenesis. STAND protein Nwd1 interacts with Paics to regulate the purinosome formation Dysregulated expression of Nwd1 induced the premature differentiation of NSPCs Nwd1 KD repressed the neuronal migration, causing the periventricular heterotopia Tightly regulated assembly of purinosome components is crucial for corticogenesis
Collapse
Affiliation(s)
- Seiya Yamada
- Laboratory for Molecular Neurobiology, Graduate School of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
| | - Ayaka Sato
- Laboratory for Molecular Neurobiology, Graduate School of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan
| | - Shin-Ichi Sakakibara
- Laboratory for Molecular Neurobiology, Graduate School of Human Sciences, Waseda University, 2-579-15 Mikajima, Tokorozawa, Saitama 359-1192, Japan.
| |
Collapse
|
28
|
Abstract
The maintenance of a healthy and functional mitochondrial network is critical during development as well as throughout life in the response to physiological adaptations and stress conditions. Owing to their role in energy production, mitochondria are exposed to high levels of reactive oxygen species, making them particularly vulnerable to mitochondrial DNA mutations and protein misfolding. Given that mitochondria are formed from proteins encoded by both nuclear and mitochondrial genomes, an additional layer of complexity is inherent in the coordination of protein synthesis and the mitochondrial import of nuclear-encoded proteins. For these reasons, mitochondria have evolved multiple systems of quality control to ensure that the requisite number of functional mitochondria are present to meet the demands of the cell. These pathways work to eliminate damaged mitochondrial proteins or parts of the mitochondrial network by mitophagy and renew components by adding protein and lipids through biogenesis, collectively resulting in mitochondrial turnover. Mitochondrial quality control mechanisms are multi-tiered, operating at the protein, organelle and cell levels. Herein, we discuss mitophagy in different physiological contexts and then relate it to other quality control pathways, including the unfolded protein response, shedding of vesicles, proteolysis, and degradation by the ubiquitin-proteasome system. Understanding how these pathways contribute to the maintenance of mitochondrial homeostasis could provide insights into the development of targeted treatments when these systems fail in disease.
Collapse
|
29
|
Abstract
Recognition of bacterial peptidoglycan by the Drosophila IMD pathway triggers NF-κB activation and an associated immune response. In this issue of Immunity, Kleino et al. (2017) show that proteins in the IMD pathway form functional amyloids via a cryptic motif resembling the RHIM motif found in mammalian RIPK proteins. Amyloid formation can be negatively regulated, suggesting that it presents a regulatory point in multiple biological processes.
Collapse
|
30
|
Wickner RB, Son M, Edskes HK. Prion Variants of Yeast are Numerous, Mutable, and Segregate on Growth, Affecting Prion Pathogenesis, Transmission Barriers, and Sensitivity to Anti-Prion Systems. Viruses 2019; 11:v11030238. [PMID: 30857327 PMCID: PMC6466074 DOI: 10.3390/v11030238] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/28/2019] [Accepted: 03/02/2019] [Indexed: 02/07/2023] Open
Abstract
The known amyloid-based prions of Saccharomyces cerevisiae each have multiple heritable forms, called "prion variants" or "prion strains". These variants, all based on the same prion protein sequence, differ in their biological properties and their detailed amyloid structures, although each of the few examined to date have an in-register parallel folded β sheet architecture. Here, we review the range of biological properties of yeast prion variants, factors affecting their generation and propagation, the interaction of prion variants with each other, the mutability of prions, and their segregation during mitotic growth. After early differentiation between strong and weak stable and unstable variants, the parameters distinguishing the variants has dramatically increased, only occasionally correlating with the strong/weak paradigm. A sensitivity to inter- and intraspecies barriers, anti-prion systems, and chaperone deficiencies or excesses and other factors all have dramatic selective effects on prion variants. Recent studies of anti-prion systems, which cure prions in wild strains, have revealed an enormous array of new variants, normally eliminated as they arise and so not previously studied. This work suggests that defects in the anti-prion systems, analogous to immune deficiencies, may be at the root of some human amyloidoses.
Collapse
Affiliation(s)
- Reed B Wickner
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA.
| | - Moonil Son
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA.
| | - Herman K Edskes
- Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892-0830, USA.
| |
Collapse
|
31
|
Kerr N, Lee SW, Perez-Barcena J, Crespi C, Ibañez J, Bullock MR, Dietrich WD, Keane RW, de Rivero Vaccari JP. Inflammasome proteins as biomarkers of traumatic brain injury. PLoS One 2018; 13:e0210128. [PMID: 30596792 PMCID: PMC6312377 DOI: 10.1371/journal.pone.0210128] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 12/17/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The inflammasome plays an important role in the inflammatory innate immune response after central nervous system (CNS) injury. Inhibition of the inflammasome after traumatic brain injury (TBI) results in improved outcomes by lowering the levels of caspase-1 and interleukin (IL)-1b. We have previously shown that inflammasome proteins are elevated in the cerebrospinal fluid (CSF) of patients with TBI and that higher levels of these proteins were consistent with poorer outcomes after TBI when compared to patients that presented these inflammasome proteins at lower levels. METHODS AND FINDINGS Here we extend our work by analyzing serum from 21 TBI patients and CSF from 18 TBI patients compared to 120 serum samples and 30 CSF samples from no-TBI donor controls for the expression of caspase-1, apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC), interleukin(IL)-1b and IL-18. Analysis was carried out using the Ella Simple Plex system (Protein Simple) to determine the sensitivity and specificity of inflammasome proteins as biomarkers of TBI. Receiver operator characteristic (ROC) curves, confidence intervals and likelihood ratios for each biomarker was determined. ROC curves, confidence intervals, sensitivity and specificity for each biomarker examined revealed that caspase-1 (0.93 area under the curve (AUC)) and ASC (0.90 AUC) in serum and ASC (1.0 AUC) and IL-18 (0.84 AUC) in CSF are promising biomarkers of TBI pathology. Importantly, higher protein levels (above 547.6 pg/ml) of ASC (0.91 AUC) were consistent with poorer outcomes after TBI as determined by the Glasgow Outcome Scale-Extended (GOSE). CONCLUSION These findings indicate that inflammasome proteins are excellent diagnostic and predictive biomarkers of TBI.
Collapse
Affiliation(s)
- Nadine Kerr
- Department of Neurological Surgery, Neuroscience Program, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami FL, United States of America
| | - Stephanie W Lee
- Department of Neurological Surgery, Neuroscience Program, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami FL, United States of America
| | - Jon Perez-Barcena
- Intensive Care Department, Son Espases Hospital, Palma de Mallorca, Spain
| | - Catalina Crespi
- Fundacio Institut d'Investigacio Sanitaria Illes Balears (IdISBa), Son Espases Hospital, Palma de Mallorca, Spain
| | - Javier Ibañez
- Department of Neurological Surgery, Son Espases Hospital, Palma de Mallorca, Spain
| | - M Ross Bullock
- Department of Neurological Surgery, Neuroscience Program, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America.,Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - W Dalton Dietrich
- Department of Neurological Surgery, Neuroscience Program, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America.,Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
| | - Robert W Keane
- Department of Neurological Surgery, Neuroscience Program, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America.,Department of Physiology and Biophysics, University of Miami Miller School of Medicine, Miami FL, United States of America
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery, Neuroscience Program, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America.,Department of Neurological Surgery, The Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, United States of America
| |
Collapse
|
32
|
Galkin AP, Velizhanina ME, Sopova YV, Shenfeld AA, Zadorsky SP. Prions and Non-infectious Amyloids of Mammals - Similarities and Differences. BIOCHEMISTRY (MOSCOW) 2018; 83:1184-1195. [PMID: 30472956 DOI: 10.1134/s0006297918100048] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Amyloids are highly ordered aggregates of protein fibrils exhibiting cross-β structure formed by intermolecular hydrogen bonds. Pathological amyloid deposition is associated with the development of several socially significant incurable human diseases. Of particular interest are infectious amyloids, or prions, that cause several lethal neurodegenerative diseases in humans and can be transmitted from one organism to another. Because of almost complete absence of criteria for infectious and non-infectious amyloids, there is a lack of consensus, especially, in the definition of similarities and differences between prions and non-infectious amyloids. In this review, we formulated contemporary molecular-biological criteria for identification of prions and non-infectious amyloids and focused on explaining the differences between these two types of molecules.
Collapse
Affiliation(s)
- A P Galkin
- St. Petersburg Branch of Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, 199034, Russia. .,St. Petersburg State University, Department of Genetics and Biotechnology, St. Petersburg, 199034, Russia
| | - M E Velizhanina
- St. Petersburg State University, Department of Genetics and Biotechnology, St. Petersburg, 199034, Russia
| | - Yu V Sopova
- St. Petersburg Branch of Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, 199034, Russia.,St. Petersburg State University, Department of Genetics and Biotechnology, St. Petersburg, 199034, Russia
| | - A A Shenfeld
- St. Petersburg Branch of Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, 199034, Russia.,St. Petersburg State University, Department of Genetics and Biotechnology, St. Petersburg, 199034, Russia
| | - S P Zadorsky
- St. Petersburg Branch of Vavilov Institute of General Genetics, Russian Academy of Sciences, St. Petersburg, 199034, Russia. .,St. Petersburg State University, Department of Genetics and Biotechnology, St. Petersburg, 199034, Russia
| |
Collapse
|
33
|
Dai T, Wu L, Wang S, Wang J, Xie F, Zhang Z, Fang X, Li J, Fang P, Li F, Jin K, Dai J, Yang B, Zhou F, van Dam H, Cai D, Huang H, Zhang L. FAF1 Regulates Antiviral Immunity by Inhibiting MAVS but Is Antagonized by Phosphorylation upon Viral Infection. Cell Host Microbe 2018; 24:776-790.e5. [PMID: 30472208 DOI: 10.1016/j.chom.2018.10.006] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Revised: 09/04/2018] [Accepted: 10/15/2018] [Indexed: 11/20/2022]
Abstract
Mitochondrial antiviral signaling protein (MAVS) is an adaptor of the innate immune receptor retinoic acid-inducible gene 1 (RIG-I) that links recognition of viral RNA to antiviral signaling. Upon interacting with RIG-I, MAVS undergoes lysine 63-linked poly-ubiquitination by the E3 ligase TRIM31 and subsequently aggregates to activate downstream signaling effectors. We find that the scaffold protein FAF1 forms aggregates that negatively regulate MAVS. FAF1 antagonizes the poly-ubiquitination and aggregation of MAVS by competing with TRIM31 for MAVS association. FAF1 knockout mice are more resistant to RNA virus infection, and FAF1 deficiency in myeloid cells results in enhanced innate signaling and reduced viral load and morbidity in vivo. Upon virus infection, the kinase IKKɛ directly phosphorylates FAF1 at Ser556 and triggers FAF1 de-aggregation. Moreover, Ser556 phosphorylation promotes FAF1 lysosomal degradation, consequently relieving FAF1-dependent suppression of MAVS. These findings establish FAF1 as a modulator of MAVS and uncover mechanisms that regulate FAF1 to insure timely activation of antiviral defense.
Collapse
Affiliation(s)
- Tong Dai
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Liming Wu
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Shuai Wang
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Jing Wang
- The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Feng Xie
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Zhengkui Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Xiuwu Fang
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Jingxian Li
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Pengfei Fang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Fang Li
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Ke Jin
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China
| | - Jianfeng Dai
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Bing Yang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China; Department of Pharmaceutical Chemistry and the Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Fangfang Zhou
- Institutes of Biology and Medical Science, Soochow University, Suzhou 215123, China
| | - Hans van Dam
- Department of Molecular Cell Biology, Cancer Genomics Centre Netherlands, Leiden University Medical Center, Postbus 9600, 2300 RC Leiden, the Netherlands
| | - Dachuan Cai
- Department for Infectious Diseases, The Second Affiliated Hospital of Chonqing Medical University, Chongqing 400016, China
| | - Huizhe Huang
- Faculty of Basic Medical Sciences, Chonqing Medical University, Chongqing 400016, China
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
34
|
Zhu L, Xu J, Liu Y, Gong T, Liu J, Huang Q, Fischbach S, Zou W, Xiao X. Prion protein is essential for diabetic retinopathy-associated neovascularization. Angiogenesis 2018; 21:767-775. [DOI: 10.1007/s10456-018-9619-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Accepted: 04/30/2018] [Indexed: 12/13/2022]
|
35
|
AAA Proteases: Guardians of Mitochondrial Function and Homeostasis. Cells 2018; 7:cells7100163. [PMID: 30314276 PMCID: PMC6210556 DOI: 10.3390/cells7100163] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/04/2018] [Accepted: 10/09/2018] [Indexed: 12/30/2022] Open
Abstract
Mitochondria are dynamic, semi-autonomous organelles that execute numerous life-sustaining tasks in eukaryotic cells. Functioning of mitochondria depends on the adequate action of versatile proteinaceous machineries. Fine-tuning of mitochondrial activity in response to cellular needs involves continuous remodeling of organellar proteome. This process not only includes modulation of various biogenetic pathways, but also the removal of superfluous proteins by adenosine triphosphate (ATP)-driven proteolytic machineries. Accordingly, all mitochondrial sub-compartments are under persistent surveillance of ATP-dependent proteases. Particularly important are highly conserved two inner mitochondrial membrane-bound metalloproteases known as m-AAA and i-AAA (ATPases associated with diverse cellular activities), whose mis-functioning may lead to impaired organellar function and consequently to development of severe diseases. Herein, we discuss the current knowledge of yeast, mammalian, and plant AAA proteases and their implications in mitochondrial function and homeostasis maintenance.
Collapse
|
36
|
Johnston A, Wang Z. Necroptosis: MLKL Polymerization. JOURNAL OF NATURE AND SCIENCE 2018; 4:e513. [PMID: 30294675 PMCID: PMC6173486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Necroptosis is a subtype of regulated necrosis that occurs when caspases are inhibited or fail to activate. Stimulus of cell death receptors results in a signaling cascade that triggers caspase independent, immunogenic cell death. The core pathway relies on receptor interacting protein kinase (RIPK) 1 and 3, which interact through their receptor homotypic interacting motif (RHIM) domains, and form amyloid-like structures termed the necrosome. RIPK3 recruits and phosphorylates mixed lineage kinase domain-like pseudokinase (MLKL), the terminal mediator in the necroptotic pathway. MLKL polymerizes to form a second amyloid-like structure that causes cell membrane disruption resulting in cell death. Although the core necroptosis pathway has been elucidated, the details of MLKL membrane translocation and membrane disruption remain an open area of research.
Collapse
Affiliation(s)
- Andrea Johnston
- Department of Molecular Biology, UT Southwestern, 6000 Harry Hines Blvd., NA8.202, Dallas, Texas 75390, USA
| | - Zhigao Wang
- Department of Molecular Biology, UT Southwestern, 6000 Harry Hines Blvd., NA8.202, Dallas, Texas 75390, USA
| |
Collapse
|
37
|
Abstract
Pattern recognition receptors (PRRs) survey intra- and extracellular spaces for pathogen-associated molecular patterns (PAMPs) within microbial products of infection. Recognition and binding to cognate PAMP ligand by specific PRRs initiates signaling cascades that culminate in a coordinated intracellular innate immune response designed to control infection. In particular, our immune system has evolved specialized PRRs to discriminate viral nucleic acid from host. These are critical sensors of viral RNA to trigger innate immunity in the vertebrate host. Different families of PRRs of virus infection have been defined and reveal a diversity of PAMP specificity for wide viral pathogen coverage to recognize and extinguish virus infection. In this review, we discuss recent insights in pathogen recognition by the RIG-I-like receptors, related RNA helicases, Toll-like receptors, and other RNA sensor PRRs, to present emerging themes in innate immune signaling during virus infection.
Collapse
Affiliation(s)
- Kwan T Chow
- Center for Innate Immunity and Immune Disease and Department of Immunology, University of Washington, Seattle, Washington 98109, USA; , ,
| | - Michael Gale
- Center for Innate Immunity and Immune Disease and Department of Immunology, University of Washington, Seattle, Washington 98109, USA; , ,
| | - Yueh-Ming Loo
- Center for Innate Immunity and Immune Disease and Department of Immunology, University of Washington, Seattle, Washington 98109, USA; , ,
| |
Collapse
|
38
|
Ramachandran B, Stabley JN, Cheng SL, Behrmann AS, Gay A, Li L, Mead M, Kozlitina J, Lemoff A, Mirzaei H, Chen Z, Towler DA. A GTPase-activating protein-binding protein (G3BP1)/antiviral protein relay conveys arteriosclerotic Wnt signals in aortic smooth muscle cells. J Biol Chem 2018; 293:7942-7968. [PMID: 29626090 DOI: 10.1074/jbc.ra118.002046] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
In aortic vascular smooth muscle (VSM), the canonical Wnt receptor LRP6 inhibits protein arginine (Arg) methylation, a new component of noncanonical Wnt signaling that stimulates nuclear factor of activated T cells (viz NFATc4). To better understand how methylation mediates these actions, MS was performed on VSM cell extracts from control and LRP6-deficient mice. LRP6-dependent Arg methylation was regulated on >500 proteins; only 21 exhibited increased monomethylation (MMA) with concomitant reductions in dimethylation. G3BP1, a known regulator of arteriosclerosis, exhibited a >30-fold increase in MMA in its C-terminal domain. Co-transfection studies confirm that G3BP1 (G3BP is Ras-GAP SH3 domain-binding protein) methylation is inhibited by LRP6 and that G3BP1 stimulates NFATc4 transcription. NFATc4 association with VSM osteopontin (OPN) and alkaline phosphatase (TNAP) chromatin was increased with LRP6 deficiency and reduced with G3BP1 deficiency. G3BP1 activation of NFATc4 mapped to G3BP1 domains supporting interactions with RIG-I (retinoic acid inducible gene I), a stimulus for mitochondrial antiviral signaling (MAVS) that drives cardiovascular calcification in humans when mutated in Singleton-Merten syndrome (SGMRT2). Gain-of-function SGMRT2/RIG-I mutants increased G3BP1 methylation and synergized with osteogenic transcription factors (Runx2 and NFATc4). A chemical antagonist of G3BP, C108 (C108 is 2-hydroxybenzoic acid, 2-[1-(2-hydroxyphenyl)ethylidene]hydrazide CAS 15533-09-2), down-regulated RIG-I-stimulated G3BP1 methylation, Wnt/NFAT signaling, VSM TNAP activity, and calcification. G3BP1 deficiency reduced RIG-I protein levels and VSM osteogenic programs. Like G3BP1 and RIG-I deficiency, MAVS deficiency reduced VSM osteogenic signals, including TNAP activity and Wnt5-dependent nuclear NFATc4 levels. Aortic calcium accumulation is decreased in MAVS-deficient LDLR-/- mice fed arteriosclerotic diets. The G3BP1/RIG-I/MAVS relay is a component of Wnt signaling. Targeting this relay may help mitigate arteriosclerosis.
Collapse
Affiliation(s)
- Bindu Ramachandran
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - John N Stabley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Su-Li Cheng
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Abraham S Behrmann
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Austin Gay
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Li Li
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Megan Mead
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Julia Kozlitina
- Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Hamid Mirzaei
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Zhijian Chen
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas 75390
| | - Dwight A Towler
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390.
| |
Collapse
|
39
|
Lugrin J, Martinon F. The AIM2 inflammasome: Sensor of pathogens and cellular perturbations. Immunol Rev 2017; 281:99-114. [DOI: 10.1111/imr.12618] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jérôme Lugrin
- Service of Adult Intensive Care Medicine; Lausanne University Hospital; Epalinges Switzerland
| | - Fabio Martinon
- Department of Biochemistry; University of Lausanne; Epalinges Switzerland
| |
Collapse
|
40
|
Kuri P, Schieber NL, Thumberger T, Wittbrodt J, Schwab Y, Leptin M. Dynamics of in vivo ASC speck formation. J Cell Biol 2017; 216:2891-2909. [PMID: 28701426 PMCID: PMC5584180 DOI: 10.1083/jcb.201703103] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Revised: 05/31/2017] [Accepted: 06/13/2017] [Indexed: 12/18/2022] Open
Abstract
The inflammasome adaptor ASC forms enormous intracellular complexes called specks. Live imaging of endogenous ASC in keratinocytes reveals speck formation dynamics and their lethal effects, as well as macrophages’ engulfment and digestion of the specks left behind by dead cells. Activated danger or pathogen sensors trigger assembly of the inflammasome adaptor ASC into specks, large signaling platforms considered hallmarks of inflammasome activation. Because a lack of in vivo tools has prevented the study of endogenous ASC dynamics, we generated a live ASC reporter through CRISPR/Cas9 tagging of the endogenous gene in zebrafish. We see strong ASC expression in the skin and other epithelia that act as barriers to insult. A toxic stimulus triggered speck formation and rapid pyroptosis in keratinocytes in vivo. Macrophages engulfed and digested that speck-containing, pyroptotic debris. A three-dimensional, ultrastructural reconstruction, based on correlative light and electron microscopy of the in vivo assembled specks revealed a compact network of highly intercrossed filaments, whereas pyrin domain (PYD) or caspase activation and recruitment domain alone formed filamentous aggregates. The effector caspase is recruited through PYD, whose overexpression induced pyroptosis but only after substantial delay. Therefore, formation of a single, compact speck and rapid cell-death induction in vivo requires a full-length ASC.
Collapse
Affiliation(s)
- Paola Kuri
- Directors' Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Nicole L Schieber
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Thomas Thumberger
- Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Joachim Wittbrodt
- Centre for Organismal Studies, Heidelberg University, Heidelberg, Germany
| | - Yannick Schwab
- Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, Heidelberg, Germany.,Electron Microscopy Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Maria Leptin
- Directors' Research Unit, European Molecular Biology Laboratory, Heidelberg, Germany .,Institute of Genetics, University of Cologne, Cologne, Germany.,European Molecular Biology Organization, Heidelberg, Germany
| |
Collapse
|
41
|
Multiple truncated isoforms of MAVS prevent its spontaneous aggregation in antiviral innate immune signalling. Nat Commun 2017; 8:15676. [PMID: 28607490 PMCID: PMC5474743 DOI: 10.1038/ncomms15676] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/19/2017] [Indexed: 12/18/2022] Open
Abstract
In response to virus infection, RIG-I-like receptors (RLRs) sense virus RNA and induce MAVS to form prion-like aggregates to further propagate antiviral signalling. Although monomeric MAVS recombinant protein can assemble into prion-like filaments spontaneously in vitro, endogenous MAVS in cells is prevented from aggregation until viral infection. The mechanism preventing cellular MAVS from spontaneous aggregation is unclear. Here we show that multiple N-terminal truncated isoforms of MAVS are essential in preventing full-length MAVS from spontaneous aggregation through transmembrane domain-mediated homotypic interaction. Without these shorter isoforms, full-length MAVS is prone to spontaneous aggregation and Nix-mediated mitophagic degradation. In the absence of N-terminally truncated forms, blocking Nix-mediated mitophagy stabilizes full-length MAVS, which aggregates spontaneously and induces the subsequent expression of type I interferon and other proinflammatory cytokines. Our data thus uncover an important mechanism preventing spontaneous aggregation of endogenous MAVS to avoid accidental activation of antiviral innate immune signalling.
Collapse
|
42
|
Rayman JB, Kandel ER. TIA-1 Is a Functional Prion-Like Protein. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a030718. [PMID: 28003185 DOI: 10.1101/cshperspect.a030718] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Prions are self-propagating protein conformations that are traditionally regarded as agents of neurodegenerative disease in animals. However, it has become evident that prion-like aggregation of endogenous proteins can also occur under normal physiological conditions (e.g., during memory storage or activation of the immune response). In this review, we focus on the functional prion-related protein TIA-1, an RNA-binding protein that is involved in multiple aspects of RNA metabolism but is best understood in terms of its role in stress granule assembly during the cellular stress response. We propose that stress granule formation provides a useful conceptual framework with which to address the positive role of TIA-1 prion-like aggregation. Elucidating the function of TIA-1 prion-like aggregation will advance our understanding of how prion-based molecular switches are used in normal physiological settings.
Collapse
Affiliation(s)
- Joseph B Rayman
- Department of Neuroscience, College of Physicians and Surgeons of Columbia University, New York, New York 10032
| | - Eric R Kandel
- Department of Neuroscience, College of Physicians and Surgeons of Columbia University, New York, New York 10032.,Department of Psychiatry, College of Physicians and Surgeons of Columbia University, New York, New York 10032.,Howard Hughes Medical Institute at Columbia University, New York, New York 10032.,Zuckerman Mind Brain Behavior Institute, Columbia University, New York, New York 10032.,Kavli Institute for Brain Science, Columbia University, New York, New York 10032
| |
Collapse
|
43
|
Loquet A, Saupe SJ. Diversity of Amyloid Motifs in NLR Signaling in Fungi. Biomolecules 2017; 7:biom7020038. [PMID: 28406433 PMCID: PMC5485727 DOI: 10.3390/biom7020038] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 04/10/2017] [Accepted: 04/10/2017] [Indexed: 01/11/2023] Open
Abstract
Amyloid folds not only represent the underlying cause of a large class of human diseases but also display a variety of functional roles both in prokaryote and eukaryote organisms. Among these roles is a recently-described activity in signal transduction cascades functioning in host defense and programmed cell death and involving Nod-like receptors (NLRs). In different fungal species, prion amyloid folds convey activation signals from a receptor protein to an effector domain by an amyloid templating and propagation mechanism. The discovery of these amyloid signaling motifs derives from the study of [Het-s], a fungal prion of the species Podospora anserina. These signaling pathways are typically composed of two basic components encoded by adjacent genes, the NLR receptor bearing an amyloid motif at the N-terminal end and a cell death execution protein with a HeLo pore-forming domain bearing a C-terminal amyloid motif. Activation of the NLR receptor allows for amyloid folding of the N-terminal amyloid motifs which then template trans-conformation of the homologous motif in the cell death execution protein. A variety of such motifs, which differ by their sequence signature, have been described in fungi. Among them, the PP-motif bears resemblance with the RHIM amyloid motif involved in the necroptosis pathway in mammals suggesting an evolutionary conservation of amyloid signaling from fungi to mammals.
Collapse
Affiliation(s)
- Antoine Loquet
- Institute of Chemistry and Biology of Membranes and Nanoobjects, UMR 5248 CBMN-CNRS Université de Bordeaux, Allée Geoffroy Saint-Hillaire, 33600 Pessac, France.
| | - Sven J Saupe
- Non-Self Recognition in Fungi, Institut de Biochimie et de Génétique Cellulaire, UMR 5095 CNRS Université de Bordeaux, 1 rue Camille Saint Saëns, 33077 Bordeaux CEDEX, France.
| |
Collapse
|