1
|
Acharya D, Sayyad Z, Hoenigsperger H, Hirschenberger M, Zurenski M, Balakrishnan K, Zhu J, Gableske S, Kato J, Zhang SY, Casanova JL, Moss J, Sparrer KMJ, Gack MU. TRIM23 mediates cGAS-induced autophagy in anti-HSV defense. Nat Commun 2025; 16:4418. [PMID: 40360474 PMCID: PMC12075517 DOI: 10.1038/s41467-025-59338-5] [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: 07/16/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
The cGAS-STING pathway, well-known to elicit interferon (IFN) responses, is also a key inducer of autophagy upon virus infection or other stimuli. Whereas the mediators for cGAS-induced IFN responses are well characterized, much less is known about how cGAS elicits autophagy. Here, we report that TRIM23, a unique TRIM protein harboring both ubiquitin E3 ligase and GTPase activity, is crucial for cGAS-STING-dependent antiviral autophagy. Genetic ablation of TRIM23 impairs autophagic control of HSV-1 infection. HSV-1 infection or cGAS-STING stimulation induces TBK1-mediated TRIM23 phosphorylation at S39, which triggers TRIM23 autoubiquitination and GTPase activity and ultimately elicits autophagy. Fibroblasts from a patient with herpes simplex encephalitis heterozygous for a dominant-negative, kinase-inactivating TBK1 mutation fail to activate autophagy by TRIM23 and cGAS-STING. Our results thus identify the cGAS-STING-TBK1-TRIM23 axis as a key autophagy defense pathway and may stimulate new therapeutic interventions for viral or inflammatory diseases.
Collapse
Affiliation(s)
- Dhiraj Acharya
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Zuberwasim Sayyad
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | | | | | - Matthew Zurenski
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
| | - Kannan Balakrishnan
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Junji Zhu
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA
| | - Sebastian Gableske
- Department of Microbiology, The University of Chicago, Chicago, IL, USA
- Eisai GmbH, Frankfurt am Main, Germany
| | - Jiro Kato
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Shen-Ying Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Paris Cité University, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| | - Joel Moss
- The Critical Care Medicine and Pulmonary Branch; National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Konstantin M J Sparrer
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany
- German Center for Neurodegenerative Diseases (DZNE), Ulm, Germany
| | - Michaela U Gack
- Florida Research and Innovation Center, Cleveland Clinic, Port St. Lucie, FL, USA.
- Department of Microbiology, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Luo C, Ma C, Xu G, Lu C, Ma J, Huang Y, Nie L, Yu C, Xia Y, Liu Z, Zhu Y, Liu S. Hepatitis B surface antigen hijacks TANK-binding kinase 1 to suppress type I interferon and induce early autophagy. Cell Death Dis 2025; 16:304. [PMID: 40234418 PMCID: PMC12000394 DOI: 10.1038/s41419-025-07605-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
There are close links between innate immunity and autophagy. However, the crosstalk between innate immunity and autophagy in host cells infected with hepatitis B virus (HBV) remains unclear. Here, we reported that HBsAg suppressed type I interferon production and induced the accumulation of autophagosomes. HBsAg boosted TANK-binding kinase 1 (TBK1) phosphorylation and depressed interferon regulatory factor 3 (IRF3) phosphorylation ex vivo and in vivo. Mechanistic studies showed that HBsAg interaction with the kinase domain (KD) of TBK1 augmented its dimerization but disrupted TBK1-IRF3 complexes. Using the TBK1 inhibitor, BX795, we discovered that HBsAg-enhanced TBK1 dimerization, promoting sequestosome-1 (p62) phosphorylation, was necessary for HBV-induced autophagy and HBV replication. Moreover, HBsAg blocked autophagosome-lysosome fusion by inhibiting the synaptosomal-associated protein 29 (SNAP29) promoter. Notably, liver tissues from HBsAg transgenic mice or chronic HBV patients revealed that IFNβ signaling was inhibited and incomplete autophagy was induced. These findings suggest a novel mechanism by which HBsAg targets TBK1 to inhibit type I interferon and induce early autophagy, possibly leading to persistent HBV infection. Molecular mechanisms of HBsAg suppression of the IFNβ signaling pathway and triggering of early autophagy. HBsAg targets the kinase domain of TBK1, thereby disrupting the TBK1-IRF3 complex and inhibiting type I interferon production. On the other hand, HBsAg enhances TBK1 dimerization and phosphorylation, which upregulates the phosphorylation of p62 to induce p62-mediated autophagy. Furthermore, HBV infection causes the accumulation of autophagosomes. This is achieved by HBsAg suppressing the SNAP29 promoter activity, which blocks autophagosome-lysosome fusion.
Collapse
Affiliation(s)
- Chuanjin Luo
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Caijiao Ma
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Gang Xu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chengbo Lu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - June Ma
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Huang
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Longyu Nie
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Chen Yu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Yongfang Xia
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Zhiqiang Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China
| | - Ying Zhu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China.
| | - Shi Liu
- State Key Laboratory of Virology, Modern Virology Research Center, College of Life Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
3
|
Ko CY. Unveiling the role of TBK1 as a novel biomarker in obstructive sleep apnea syndrome. Sleep 2025; 48:zsaf001. [PMID: 39798071 PMCID: PMC11985379 DOI: 10.1093/sleep/zsaf001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Indexed: 01/13/2025] Open
Affiliation(s)
- Chih-Yuan Ko
- Department of Clinical Nutrition, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
4
|
Li Z, Yi H, Li Y, Yang J, Guo P, Han F. Identification and validation of a novel autophagy-related biomarker in obstructive sleep apnea syndrome. Sleep 2025; 48:zsae287. [PMID: 39665515 DOI: 10.1093/sleep/zsae287] [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: 05/07/2024] [Revised: 11/24/2024] [Indexed: 12/13/2024] Open
Abstract
STUDY OBJECTIVES Obstructive sleep apnea syndrome is closely associated with tumor growth. Chronic intermittent hypoxia promotes autophagy and is related to malignant tumor development. However, the role of autophagy in obstructive sleep apnea syndrome progression remains unclear. METHODS obstructive sleep apnea syndrome datasets (GSE135917 and GSE38792) from Gene Expression Omnibus were analyzed to identify differentially expressed genes and autophagy-related differentially expressed genes. Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and gene set enrichment analysis were conducted, and a protein-protein interaction network identified hub genes. Colorectal cancer datasets from The Cancer Genome Atlas were used for differential expression and survival analyses, along with gene set enrichment analysis and immune infiltration analysis. Chronic intermittent hypoxia-induced autophagy and oxidative stress were investigated in Sprague-Dawley rats using reactive oxygen species assays. Hub genes were validated in rats and obstructive sleep apnea syndrome patient samples. RESULTS Gene set enrichment analysis revealed significant differences in autophagy-related gene expression among obstructive sleep apnea syndrome patients. Hub genes ATG5, CASP1, MAPK8, EIF4G1, and TANK-binding kinase 1 were identified, with ATG5 and TANK-binding kinase 1 validated. Autophagy-related differentially expressed genes were predominantly upregulated in colorectal cancer tissues. TANK-binding kinase 1 expression in colorectal cancer patients was associated with enhanced sensitivity to immunotherapy and CD8 + T cell, macrophage, and regulatory T cell infiltration, potentially influencing the immune microenvironment. The animal experiments showed that chronic intermittent hypoxia increased reactive oxygen species levels, suggesting that chronic intermittent hypoxia plays a role in autophagy. TANK-binding kinase 1 expression was significantly higher in obstructive sleep apnea syndrome patients than in controls, and continuous positive airway pressure did not alter TANK-binding kinase 1 levels. CONCLUSIONS This study is the first to describe the potential contribution of TANK-binding kinase 1 to the development of obstructive sleep apnea syndrome and its potential as a novel biomarker and potential therapeutic target for obstructive sleep apnea syndrome.
Collapse
Affiliation(s)
- Zhiyong Li
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Huijie Yi
- Department of Sleep Medicine, Peking University People's Hospital, Xicheng, Beijing, China
| | - Yuxi Li
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Jie Yang
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Peng Guo
- Department of Emergency Surgery, Peking University People's Hospital, Xicheng, Beijing, China
| | - Fang Han
- Department of Sleep Medicine, Peking University People's Hospital, Xicheng, Beijing, China
| |
Collapse
|
5
|
Megat S, Marques C, Hernán-Godoy M, Sellier C, Stuart-Lopez G, Dirrig-Grosch S, Gorin C, Brunet A, Fischer M, Keime C, Kessler P, Mendoza-Parra MA, Zwamborn RAJ, Veldink JH, Scholz SW, Ferrucci L, Ludolph A, Traynor B, Chio A, Dupuis L, Rouaux C. CREB3 gain of function variants protect against ALS. Nat Commun 2025; 16:2942. [PMID: 40140376 PMCID: PMC11947196 DOI: 10.1038/s41467-025-58098-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal and rapidly evolving neurodegenerative disease arising from the loss of glutamatergic corticospinal neurons (CSN) and cholinergic motoneurons (MN). Here, we performed comparative cross-species transcriptomics of CSN using published snRNA-seq data from the motor cortex of ALS and control postmortem tissues, and performed longitudinal RNA-seq on CSN purified from male Sod1G86R mice. We report that CSN undergo ER stress and altered mRNA translation, and identify the transcription factor CREB3 and its regulatory network as a resilience marker of ALS, not only amongst vulnerable neuronal populations, but across all neuronal populations as well as other cell types. Using genetic and epidemiologic analyses we further identify the rare variant CREB3R119G (rs11538707) as a positive disease modifier in ALS. Through gain of function, CREB3R119G decreases the risk of developing ALS and the motor progression rate of ALS patients.
Collapse
Affiliation(s)
- Salim Megat
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France.
| | - Christine Marques
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Marina Hernán-Godoy
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Chantal Sellier
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Geoffrey Stuart-Lopez
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Sylvie Dirrig-Grosch
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Charlotte Gorin
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Aurore Brunet
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Mathieu Fischer
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Céline Keime
- Université de Strasbourg, Inserm UMR-S 1258, CNRS UMR-S 7104, Institut de Génétique, Biologie Moléculaire et Cellulaire, Illkirch-Graffenstaden, France
| | - Pascal Kessler
- Université de Strasbourg, Inserm UMS 38, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Marco Antonio Mendoza-Parra
- UMR 8030 Génomique Métabolique, Genoscope, Institut François Jacob, CEA, CNRS, University of Evry-val-d'Essonne, University Paris-Saclay, Evry, France
| | - Ramona A J Zwamborn
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Jan H Veldink
- Department of Neurology, Brain Centre Rudolf Magnus, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Sonja W Scholz
- Neurodegenerative Diseases Research Section, National Institute of Neurological Disorders and Stroke, Bethesda, MD, USA
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
| | - Luigi Ferrucci
- Intramural Research Program of the National Institute on Aging, NIH, Baltimore, MD, USA
| | | | - Bryan Traynor
- Department of Neurology, Johns Hopkins University Medical Center, Baltimore, MD, USA
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Therapeutic Development Branch, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, USA
| | - Adriano Chio
- ALS Center "Rita Levi Montalcini" Department of Neuroscience, University of Turin, Turin, Italy
| | - Luc Dupuis
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France
| | - Caroline Rouaux
- Université de Strasbourg, Inserm, Strasbourg Translational Neuroscience and Psychiatry, Inserm UMR-S 1329, Centre de Recherche en Biomédecine de Strasbourg, Strasbourg, France.
| |
Collapse
|
6
|
Hui L, Chen X, Huang M, Jiang Y, Liu T. TANK-Binding Kinase 1 in the Pathogenesis and Treatment of Inflammation-Related Diseases. Int J Mol Sci 2025; 26:1941. [PMID: 40076567 PMCID: PMC11900955 DOI: 10.3390/ijms26051941] [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: 01/13/2025] [Revised: 02/20/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
TANK-binding kinase 1 (TBK1) is a key signaling kinase involved in innate immune and inflammatory responses. TBK1 drives immune cells to participate in the inflammatory response by activating the NF-κB and interferon regulatory factor signaling pathways in immune cells, promoting the expression of pro-inflammatory genes, and regulating immune cell function. Thus, it plays a crucial role in initiating a signaling cascade that establishes an inflammatory environment. In inflammation-related diseases, TBK1 acts as a bridge linking inflammation to immunity, metabolism, or tumorigenesis, playing an important role in the pathogenesis of immune-mediated inflammatory diseases, metabolic, inflammatory syndromes, and inflammation-associated cancers by regulating the activation of inflammatory pathways and the production of inflammatory cytokines in cells. In this review, we focused on the mechanisms of TBK1 in immune cells and inflammatory-related diseases, providing new insights for further studies targeting TBK1 as a potential treatment for inflammation-related diseases. Thus, optimizing and investigating highly selective cell-specific TBK1 inhibitors is important for their application in these diseases.
Collapse
Affiliation(s)
- Lu Hui
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Xiaolin Chen
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Mengke Huang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
| | - Yongmei Jiang
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu 610041, China
| | - Ting Liu
- Department of Laboratory Medicine, West China Second University Hospital, Sichuan University, No. 20, Section 3, Renmin Road South, Chengdu 610041, China; (L.H.); (X.C.); (M.H.)
- Key Laboratory of Obstetric & Gynecologic and Pediatric Diseases and Birth Defects of Ministry of Education, Sichuan University, Chengdu 610041, China
- State Key Laboratory of Biotherapy and Cancer Center/National Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu 610041, China
| |
Collapse
|
7
|
Hyde VR, Zhou C, Fernandez JR, Chatterjee K, Ramakrishna P, Lin A, Fisher GW, Çeliker OT, Caldwell J, Bender O, Sauer PJ, Lugo-Martinez J, Bar DZ, D'Aiuto L, Shemesh OA. Anti-herpetic tau preserves neurons via the cGAS-STING-TBK1 pathway in Alzheimer's disease. Cell Rep 2025; 44:115109. [PMID: 39753133 DOI: 10.1016/j.celrep.2024.115109] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 08/06/2024] [Accepted: 12/03/2024] [Indexed: 02/01/2025] Open
Abstract
Alzheimer's disease (AD) diagnosis relies on the presence of extracellular β-amyloid (Aβ) and intracellular hyperphosphorylated tau (p-tau). Emerging evidence suggests a potential link between AD pathologies and infectious agents, with herpes simplex virus 1 (HSV-1) being a leading candidate. Our investigation, using metagenomics, mass spectrometry, western blotting, and decrowding expansion pathology, detects HSV-1-associated proteins in human brain samples. Expression of the herpesvirus protein ICP27 increases with AD severity and strongly colocalizes with p-tau but not with Aβ. Modeling in human brain organoids shows that HSV-1 infection elevates tau phosphorylation. Notably, p-tau reduces ICP27 expression and markedly decreases post-infection neuronal death from 64% to 7%. This modeling prompts investigation into the cGAS-STING-TBK1 pathway products, nuclear factor (NF)-κB and IRF-3, which colocalizes with ICP27 and p-tau in AD. Furthermore, experimental activation of STING enhances tau phosphorylation, while TBK1 inhibition prevents it. Together, these findings suggest that tau phosphorylation acts as an innate immune response in AD, driven by cGAS-STING.
Collapse
Affiliation(s)
- Vanesa R Hyde
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Chaoming Zhou
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Juan R Fernandez
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Krishnashis Chatterjee
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Pururav Ramakrishna
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Amanda Lin
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Gregory W Fisher
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; Department of Chemistry, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Orhan Tunç Çeliker
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Jill Caldwell
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Omer Bender
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Peter Joseph Sauer
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Jose Lugo-Martinez
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Daniel Z Bar
- Department of Oral Biology, Goldschleger School of Dental Medicine, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Leonardo D'Aiuto
- Department of Psychiatry, Western Psychiatric Institute and Clinic, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Or A Shemesh
- School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112102, Israel; Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
8
|
Watanabe-Matsui M, Kadoya S, Segawa K, Shima H, Nakagawa T, Nagasawa Y, Hayashi S, Matsumoto M, Ikeda M, Muto A, Ochiai K, Nguyen LC, Doh-Ura K, Shirouzu M, Nakayama K, Murayama K, Igarashi K. Heme regulates protein interactions and phosphorylation of BACH2 intrinsically disordered region in humoral response. iScience 2025; 28:111529. [PMID: 39758820 PMCID: PMC11699347 DOI: 10.1016/j.isci.2024.111529] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 09/03/2023] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Heme is known to bind to the intrinsically disordered region (IDR) to regulate protein function. The binding of heme to the IDR of transcription factor BACH2 promotes plasma cell differentiation, but the molecular basis is unknown. Heme was found to increase BACH2 IDR interaction with TANK-binding kinase 1 (TBK1). TBK1 inactivated BACH2 by phosphorylation of its IDR, whereas BACH2 repressed TBK1 gene expression. BACH2 phosphorylation by TBK1 inhibited its interaction with the co-repressor NCOR1 and promoted plasma cell differentiation. Heme also induced BACH2 binding to ubiquitin E3 ligase adaptor FBXO22, which polyubiquitinated BACH2 only in the presence of heme in vitro. Mutations of some of the TBK1-mediated phosphorylation sites promoted BACH2-FBXO22 interaction, while additional mutations abrogated their interaction, suggesting that TBK1 can both inhibit and promote BACH2-FBXO22 interaction. Therefore, heme regulates phosphorylation of BACH2 IDR by TBK1 and its interaction with NCOR1 and FBXO22, leading to de-repression of BACH2 target genes in humoral immunity.
Collapse
Affiliation(s)
- Miki Watanabe-Matsui
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
- The Japan Society for the Promotion of Science (JSPS), Tokyo, Japan
| | - Shun Kadoya
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kei Segawa
- Pharmaceutical Discovery Research Laboratories, Teijin Pharma Limited, Tokyo, Japan
| | - Hiroki Shima
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tadashi Nakagawa
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Clinical Pharmacology, Sanyo-Onoda City University, Sanyo-Onoda, Japan
| | - Yuko Nagasawa
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Shuichiro Hayashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mariko Ikeda
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Akihiko Muto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kyoko Ochiai
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Long C. Nguyen
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Katsumi Doh-Ura
- Department of Neurochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Japan
| | - Keiko Nakayama
- Division of Cell Proliferation, ART, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kazutaka Murayama
- Division of Biomedical Measurements and Diagnostics, Tohoku University Graduate School of Biomedical Engineering, Sendai, Japan
| | - Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
9
|
Paul S, Biswas SR, Milner JP, Tomsick PL, Pickrell AM. Adaptor-Mediated Trafficking of Tank Binding Kinase 1 During Diverse Cellular Processes. Traffic 2025; 26:e70000. [PMID: 40047067 PMCID: PMC11883510 DOI: 10.1111/tra.70000] [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: 12/02/2024] [Revised: 02/11/2025] [Accepted: 02/14/2025] [Indexed: 03/09/2025]
Abstract
The serine/threonine kinase, Tank Binding Kinase 1 (TBK1), drives distinct cellular processes like innate immune signaling, selective autophagy, and mitosis. It is suggested that the translocation and activation of TBK1 at different subcellular locations within the cell, downstream of diverse stimuli, are driven by TBK1 adaptor proteins forming a complex directly or indirectly with TBK1. Various TBK1 adaptors and associated proteins like NAP1, TANK, SINTBAD, p62, optineurin (OPTN), TAX1BP1, STING, and NDP52 have been identified in facilitating TBK1 activation and recruitment with varying overlapping redundancy. This review focuses on what is known about these proteins, their interactions with TBK1, and the functional consequences of these associations. We shed light on underexplored areas of research on these TBK1 binding partners while emphasizing how future research is required to understand the function and flexibility of TBK1 signaling and crosstalk or regulation between different biological processes.
Collapse
Affiliation(s)
- Swagatika Paul
- Graduate Program in Biomedical and Veterinary SciencesVirginia‐Maryland College of Veterinary MedicineBlacksburgVirginiaUSA
| | - Sahitya Ranjan Biswas
- Translational Biology, Medicine, and Health Graduate ProgramVirginia Polytechnic Institute and State UniversityRoanokeVirginiaUSA
| | - Julia P. Milner
- School of NeuroscienceVirginia Polytechnic Institute and State UniversityBlacksburgVirginiaUSA
| | - Porter L. Tomsick
- School of NeuroscienceVirginia Polytechnic Institute and State UniversityBlacksburgVirginiaUSA
| | - Alicia M. Pickrell
- School of NeuroscienceVirginia Polytechnic Institute and State UniversityBlacksburgVirginiaUSA
| |
Collapse
|
10
|
Modica G, Tejeda-Valencia L, Sauvageau E, Yasa S, Maes J, Skorobogata O, Lefrancois S. Phosphorylation on serine 72 modulates Rab7A palmitoylation and retromer recruitment. J Cell Sci 2025; 138:jcs262177. [PMID: 39584231 PMCID: PMC11828465 DOI: 10.1242/jcs.262177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 11/20/2024] [Indexed: 11/26/2024] Open
Abstract
Rab7A has a key role in regulating membrane trafficking at late endosomes. By interacting with several different effectors, this small GTPase controls late endosome mobility, orchestrates fusion events between late endosomes and lysosomes, and participates in the formation of and regulates the fusion between autophagosomes and lysosomes. Rab7A is also responsible for the spatiotemporal recruitment of retromer, which is required for the endosome-to-trans-Golgi network retrieval of cargo receptors such as sortilin (SORT1) and CI-MPR (also known as IGF2R). Recently, several post-translational modifications have been shown to modulate Rab7A functions, including palmitoylation, ubiquitination and phosphorylation. Here, we show that phosphorylation of Rab7A at serine 72 is important to modulate its interaction with retromer, as the non-phosphorylatable Rab7AS72A mutant is not able to interact with and recruit retromer to late endosomes. We have previously shown that Rab7A palmitoylation is also required for efficient retromer recruitment. We found that palmitoylation of Rab7AS72A is reduced compared to that of the wild-type protein, suggesting an interplay between S72 phosphorylation and palmitoylation in regulating the Rab7A-retromer interaction. Finally, we identify NEK7 as a kinase required to phosphorylate Rab7A to promote retromer binding and recruitment.
Collapse
Affiliation(s)
- Graziana Modica
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Québec H7V 1B7, Canada
| | - Laura Tejeda-Valencia
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Québec H7V 1B7, Canada
| | - Etienne Sauvageau
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Québec H7V 1B7, Canada
| | - Seda Yasa
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Québec H7V 1B7, Canada
| | - Juliette Maes
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Québec H7V 1B7, Canada
| | - Olga Skorobogata
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Québec H7V 1B7, Canada
| | - Stephane Lefrancois
- Centre Armand-Frappier Santé Biotechnologie, Institut national de la recherche scientifique, Laval, Québec H7V 1B7, Canada
- Department of Anatomy and Cell Biology, McGill University, Montreal H3A 0C7, Canada
- Centre d'Excellence en Recherche sur les Maladies Orphelines - Fondation Courtois (CERMO-FC), Université du Québec à Montréal (UQAM), Montréal H2X 3Y7, Canada
| |
Collapse
|
11
|
Wang C, Luo H. Crosstalk Between Innate Immunity and Autophagy in Viral Myocarditis Leading to Dilated Cardiomyopathy. Rev Med Virol 2024; 34:e2586. [PMID: 39349889 DOI: 10.1002/rmv.2586] [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: 06/05/2024] [Revised: 09/02/2024] [Accepted: 09/12/2024] [Indexed: 11/08/2024]
Abstract
Viral myocarditis, characterised by inflammation of the heart muscle, presents a significant challenge to global public health, particularly affecting younger individuals and often progressing to dilated cardiomyopathy (DCM), a leading cause of heart failure. Despite ongoing research efforts, viable treatments for this condition remain elusive. Recent studies have shed light on the complex interplay between the innate immune response and autophagy mechanisms, revealing their pivotal roles in the pathogenesis of viral myocarditis and subsequent DCM development. This review aims to delve into the recent advancements in understanding the molecular mechanisms and pathways that intersect innate immunity and autophagy in the context of viral myocarditis. Furthermore, it explores the potential therapeutic implications of these findings, offering insights into promising avenues for the management and treatment of this debilitating condition.
Collapse
Affiliation(s)
- Chen Wang
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| | - Honglin Luo
- Centre for Heart Lung Innovation, St. Paul's Hospital-University of British Columbia, Vancouver, Canada
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, Canada
| |
Collapse
|
12
|
Yang X, Zheng R, Zhang H, Ou Z, Wan S, Lin D, Yan J, Jin M, Tan J. Optineurin regulates motor and learning behaviors by affecting dopaminergic neuron survival in mice. Exp Neurol 2024; 383:115007. [PMID: 39428042 DOI: 10.1016/j.expneurol.2024.115007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/17/2024] [Accepted: 10/16/2024] [Indexed: 10/22/2024]
Abstract
Optineurin (OPTN) is an autophagy receptor that participates in the degradation of damaged mitochondria, protein aggregates, and invading pathogens. OPTN is closely related to various types of neurodegenerative diseases. However, the role of OPTN in the central nervous system is unclear. Here, we found that OPTN dysregulation in the compact part of substantia nigra (SNc) led to motor and learning deficits in animal models. Knockdown of OPTN increased total and phosphorylated α-synuclein levels which induced microglial activation and dopaminergic neuronal loss in the SNc. Overexpression of OPTN can't reverse the motor and learning phenotypes. Mechanistic analysis revealed that upregulation of OPTN increased α-synuclein phosphorylation independent of its autophagy receptor activity, which further resulted in microglial activation and dopaminergic neuronal loss similar to OPTN downregulation. Our study uncovers the crucial role of OPTN in maintaining dopaminergic neuron survival and motor and learning functions which are disrupted in PD patients.
Collapse
Affiliation(s)
- Xianfei Yang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China
| | - Ruoling Zheng
- Shantou Longhu People's Hospital, Shantou 515041, China
| | - Hongyao Zhang
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China
| | - Zixian Ou
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China
| | - Sha Wan
- Department of Anatomy, College of Basic Medicine, Guilin Medical University, Guilin 541199, China
| | - Dongfeng Lin
- Shantou University Mental Health Center, Shantou University, Shantou 515063, China
| | - Jianguo Yan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China; Department of Physiology, College of Basic Medicine, Guilin Medical University, Guilin 541199, China
| | - Mingyue Jin
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China
| | - Jie Tan
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin 541199, China; Department of Physiology, College of Basic Medicine, Guilin Medical University, Guilin 541199, China; Clinical Research Center for Neurological Diseases of Guangxi Province, The Affiliated Hospital of Guilin Medical University, Guilin 541001, China.
| |
Collapse
|
13
|
Zelina P, de Ruiter AA, Kolsteeg C, van Ginneken I, Vos HR, Supiot LF, Burgering BMT, Meye FJ, Veldink JH, van den Berg LH, Pasterkamp RJ. ALS-associated C21ORF2 variant disrupts DNA damage repair, mitochondrial metabolism, neuronal excitability and NEK1 levels in human motor neurons. Acta Neuropathol Commun 2024; 12:144. [PMID: 39227882 PMCID: PMC11373222 DOI: 10.1186/s40478-024-01852-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 08/15/2024] [Indexed: 09/05/2024] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset neurodegenerative disease leading to motor neuron loss. Currently mutations in > 40 genes have been linked to ALS, but the contribution of many genes and genetic mutations to the ALS pathogenic process remains poorly understood. Therefore, we first performed comparative interactome analyses of five recently discovered ALS-associated proteins (C21ORF2, KIF5A, NEK1, TBK1, and TUBA4A) which highlighted many novel binding partners, and both unique and shared interactors. The analysis further identified C21ORF2 as a strongly connected protein. The role of C21ORF2 in neurons and in the nervous system, and of ALS-associated C21ORF2 variants is largely unknown. Therefore, we combined human iPSC-derived motor neurons with other models and different molecular cell biological approaches to characterize the potential pathogenic effects of C21ORF2 mutations in ALS. First, our data show C21ORF2 expression in ALS-relevant mouse and human neurons, such as spinal and cortical motor neurons. Further, the prominent ALS-associated variant C21ORF2-V58L caused increased apoptosis in mouse neurons and movement defects in zebrafish embryos. iPSC-derived motor neurons from C21ORF2-V58L-ALS patients, but not isogenic controls, show increased apoptosis, and changes in DNA damage response, mitochondria and neuronal excitability. In addition, C21ORF2-V58L induced post-transcriptional downregulation of NEK1, an ALS-associated protein implicated in apoptosis and DDR. In all, our study defines the pathogenic molecular and cellular effects of ALS-associated C21ORF2 mutations and implicates impaired post-transcriptional regulation of NEK1 downstream of mutant C21ORF72 in ALS.
Collapse
Affiliation(s)
- Pavol Zelina
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Anna Aster de Ruiter
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Christy Kolsteeg
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Ilona van Ginneken
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Harmjan R Vos
- Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Laura F Supiot
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Boudewijn M T Burgering
- Center for Molecular Medicine, Oncode Institute, University Medical Center Utrecht, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Frank J Meye
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands
| | - Jan H Veldink
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - Leonard H van den Berg
- Department of Neurology and Neurosurgery, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, 3584 CX, Utrecht, The Netherlands
| | - R Jeroen Pasterkamp
- Department of Translational Neuroscience, University Medical Center Utrecht Brain Center, Utrecht University, Universiteitsweg 100, 3584 CG, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Liu Y, Wang A, Chen C, Zhang Q, Shen Q, Zhang D, Xiao X, Chen S, Lian L, Le Z, Liu S, Liang T, Zheng Q, Xu P, Zou J. Microglial cGAS-STING signaling underlies glaucoma pathogenesis. Proc Natl Acad Sci U S A 2024; 121:e2409493121. [PMID: 39190350 PMCID: PMC11388346 DOI: 10.1073/pnas.2409493121] [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: 05/17/2024] [Accepted: 07/24/2024] [Indexed: 08/28/2024] Open
Abstract
Characterized by progressive degeneration of retinal ganglion cells (RGCs) and vision loss, glaucoma is the primary cause of irreversible blindness, incurable and affecting over 78 million patients. However, pathogenic mechanisms leading to glaucoma-induced RGC loss are incompletely understood. Unexpectedly, we found that cGAS-STING (2'3'-cyclic GMP-AMP-stimulator of interferon genes) signaling, which surveils displaced double-stranded DNA (dsDNA) in the cytosol and initiates innate immune responses, was robustly activated during glaucoma in retinal microglia in distinct murine models. Global or microglial deletion of STING markedly relieved glaucoma symptoms and protected RGC degeneration and vision loss, while mice bearing genetic cGAS-STING supersensitivity aggravated retinal neuroinflammation and RGC loss. Mechanistically, dsDNA from tissue injury activated microglial cGAS-STING signaling, causing deleterious macroglia reactivity in retinas by cytokine-mediated microglia-macroglia interactions, progressively driving apoptotic death of RGCs. Remarkably, preclinical investigations of targeting cGAS-STING signaling by intraocular injection of TBK1i or anti-IFNAR1 antibody prevented glaucoma-induced losses of RGCs and vision. Therefore, we unravel an essential role of cGAS-STING signaling underlying glaucoma pathogenesis and suggest promising therapeutic strategies for treating this devastating disease.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou310058, China
| | - Ailian Wang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou310058, China
| | - Chen Chen
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou310058, China
| | - Qian Zhang
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou310058, China
| | - Qin Shen
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
| | - Dan Zhang
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
| | - Xueqi Xiao
- Eye Center of the Second Affiliated Hospital School of Medicine, Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Hangzhou310029, China
| | - Shasha Chen
- College of Life and Environmental Science, Wenzhou University, Wenzhou325035, China
| | - Lili Lian
- National Clinical Research Center for Ocular Diseases, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou325027, China
| | - Zhenmin Le
- National Clinical Research Center for Ocular Diseases, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou325027, China
| | - Shengduo Liu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou310058, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou310058, China
| | - Qinxiang Zheng
- National Clinical Research Center for Ocular Diseases, School of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou325027, China
| | - Pinglong Xu
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou310058, China
- Institute of Intelligent Medicine, Zhejiang University-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou310058, China
- Ministry of Education Laboratory of Biosystems Homeostasis and Protection, Zhejiang Provincial Key Laboratory for Cancer Molecular Cell Biology, Life Sciences Institute, Zhejiang University, Hangzhou310058, China
| | - Jian Zou
- Department of Hepatobiliary and Pancreatic Surgery and Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, University School of Medicine, Zhejiang University, Hangzhou310058, China
- Eye Center of the Second Affiliated Hospital School of Medicine, Center for Genetic Medicine, Zhejiang University International Institute of Medicine, Hangzhou310029, China
| |
Collapse
|
15
|
Ho PC, Hsieh TC, Tsai KJ. TDP-43 proteinopathy in frontotemporal lobar degeneration and amyotrophic lateral sclerosis: From pathomechanisms to therapeutic strategies. Ageing Res Rev 2024; 100:102441. [PMID: 39069095 DOI: 10.1016/j.arr.2024.102441] [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: 05/31/2024] [Revised: 07/12/2024] [Accepted: 07/24/2024] [Indexed: 07/30/2024]
Abstract
Proteostasis failure is a common pathological characteristic in neurodegenerative diseases. Revitalizing clearance systems could effectively mitigate these diseases. The transactivation response (TAR) DNA-binding protein 43 (TDP-43) plays a critical role as an RNA/DNA-binding protein in RNA metabolism and synaptic function. Accumulation of TDP-43 aggregates in the central nervous system is a hallmark of frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Autophagy, a major and highly conserved degradation pathway, holds the potential for degrading aggregated TDP-43 and alleviating FTLD/ALS. This review explores the causes of TDP-43 aggregation, FTLD/ALS-related genes, key autophagy factors, and autophagy-based therapeutic strategies targeting TDP-43 proteinopathy. Understanding the underlying pathological mechanisms of TDP-43 proteinopathy can facilitate therapeutic interventions.
Collapse
Affiliation(s)
- Pei-Chuan Ho
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Tsung-Chi Hsieh
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Kuen-Jer Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Research Center of Clinical Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
16
|
Talaia G, Bentley-DeSousa A, Ferguson SM. Lysosomal TBK1 responds to amino acid availability to relieve Rab7-dependent mTORC1 inhibition. EMBO J 2024; 43:3948-3967. [PMID: 39103493 PMCID: PMC11405869 DOI: 10.1038/s44318-024-00180-8] [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: 02/29/2024] [Revised: 05/22/2024] [Accepted: 06/24/2024] [Indexed: 08/07/2024] Open
Abstract
Lysosomes play a pivotal role in coordinating macromolecule degradation and regulating cell growth and metabolism. Despite substantial progress in identifying lysosomal signaling proteins, understanding the pathways that synchronize lysosome functions with changing cellular demands remains incomplete. This study uncovers a role for TANK-binding kinase 1 (TBK1), well known for its role in innate immunity and organelle quality control, in modulating lysosomal responsiveness to nutrients. Specifically, we identify a pool of TBK1 that is recruited to lysosomes in response to elevated amino acid levels. This lysosomal TBK1 phosphorylates Rab7 on serine 72. This is critical for alleviating Rab7-mediated inhibition of amino acid-dependent mTORC1 activation. Furthermore, a TBK1 mutant (E696K) associated with amyotrophic lateral sclerosis and frontotemporal dementia constitutively accumulates at lysosomes, resulting in elevated Rab7 phosphorylation and increased mTORC1 activation. This data establishes the lysosome as a site of amino acid regulated TBK1 signaling that is crucial for efficient mTORC1 activation. This lysosomal pool of TBK1 has broader implications for lysosome homeostasis, and its dysregulation could contribute to the pathogenesis of ALS-FTD.
Collapse
Affiliation(s)
- Gabriel Talaia
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06510, USA
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, 06510, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Amanda Bentley-DeSousa
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06510, USA
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, 06510, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Shawn M Ferguson
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Program in Cellular Neuroscience, Neurodegeneration and Repair, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Wu Tsai Institute, Yale University School of Medicine, New Haven, CT, 06510, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA.
- Kavli Institute for Neuroscience, Yale University School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
17
|
Jiao Y, Zhao P, Xu LD, Yu JQ, Cai HL, Zhang C, Tong C, Yang YL, Xu P, Sun Q, Chen N, Wang B, Huang YW. Enteric coronavirus nsp2 is a virulence determinant that recruits NBR1 for autophagic targeting of TBK1 to diminish the innate immune response. Autophagy 2024; 20:1762-1779. [PMID: 38597182 PMCID: PMC11262224 DOI: 10.1080/15548627.2024.2340420] [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: 09/22/2023] [Revised: 03/30/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
Non-structural protein 2 (nsp2) exists in all coronaviruses (CoVs), while its primary function in viral pathogenicity, is largely unclear. One such enteric CoV, porcine epidemic diarrhea virus (PEDV), causes high mortality in neonatal piglets worldwide. To determine the biological role of nsp2, we generated a PEDV mutant containing a complete nsp2 deletion (rPEDV-Δnsp2) from a highly pathogenic strain by reverse genetics, showing that nsp2 was dispensable for PEDV infection, while its deficiency reduced viral replication in vitro. Intriguingly, rPEDV-Δnsp2 was entirely avirulent in vivo, with significantly increased productions of IFNB (interferon beta) and IFN-stimulated genes (ISGs) in various intestinal tissues of challenged newborn piglets. Notably, nsp2 targets and degrades TBK1 (TANK binding kinase 1), the critical kinase in the innate immune response. Mechanistically, nsp2 induced the macroautophagy/autophagy process and recruited a selective autophagic receptor, NBR1 (NBR1 autophagy cargo receptor). NBR1 subsequently facilitated the K48-linked ubiquitination of TBK1 and delivered it for autophagosome-mediated degradation. Accordingly, the replication of rPEDV-Δnsp2 CoV was restrained by reduced autophagy and excess productions of type I IFNs and ISGs. Our data collectively define enteric CoV nsp2 as a novel virulence determinant, propose a crucial role of nsp2 in diminishing innate antiviral immunity by targeting TBK1 for NBR1-mediated selective autophagy, and pave the way to develop a new type of nsp2-based attenuated PEDV vaccine. The study also provides new insights into the prevention and treatment of other pathogenic CoVs.Abbreviations: 3-MA: 3-methyladenine; Baf A1: bafilomycin A1; CoV: coronavirus; CQ: chloroquine; dpi: days post-inoculation; DMVs: double-membrane vesicles; GABARAP: GABA type A receptor-associated protein; GFP: green fluorescent protein; GIGYF2: GRB10 interacting GYF protein 2; hpi: hours post-infection; IFA: immunofluorescence assay; IFIH1: interferon induced with helicase C domain 1; IFIT2: interferon induced protein with tetratricopeptide repeats 2; IFITM1: interferon induced transmembrane protein 1; IFNB: interferon beta; IRF3: interferon regulatory factor 3; ISGs: interferon-stimulated genes; mAb: monoclonal antibody; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MAVS: mitochondrial antiviral signaling protein; NBR1: NBR1 autophagy cargo receptor; nsp2: non-structural protein 2; OAS1: 2'-5'-oligoadenylate synthetase 1; PEDV: porcine epidemic diarrhea virus; PRRs: pattern recognition receptors; RIGI: RNA sensor RIG-I; RT-qPCR: reverse transcription quantitative polymerase chain reaction; SQSTM1: sequestosome 1; TBK1: TANK binding kinase 1; TCID50: 50% tissue culture infectious doses; VSV: vesicular stomatitis virus.
Collapse
Affiliation(s)
- Yajuan Jiao
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Pengwei Zhao
- Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ling-Dong Xu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Jia-Qi Yu
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Hou-Li Cai
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Chong Zhang
- Boehringer Ingelheim Vetmedica (China) Co. Ltd, Taizhou, China
| | - Chao Tong
- Boehringer Ingelheim Vetmedica (China) Co. Ltd, Taizhou, China
| | - Yong-Le Yang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
| | - Pinglong Xu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Qiming Sun
- Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Ning Chen
- Boehringer Ingelheim Vetmedica (China) Co. Ltd, Taizhou, China
| | - Bin Wang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
| | - Yao-Wei Huang
- Department of Veterinary Medicine, Zhejiang University, Hangzhou, China
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Veterinary Medicine, South China Agricultural University, Guangzhou, China
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, China
| |
Collapse
|
18
|
Song J, Guo Y, Wang D, Quan R, Wang J, Liu J. Seneca Valley virus 3C protease cleaves OPTN (optineurin) to Impair selective autophagy and type I interferon signaling. Autophagy 2024; 20:614-628. [PMID: 37930946 PMCID: PMC10936645 DOI: 10.1080/15548627.2023.2277108] [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: 02/15/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023] Open
Abstract
Seneca Valley virus (SVV) causes vesicular disease in pigs, posing a threat to global pork production. OPTN (optineurin) is a macroautophagy/autophagy receptor that restricts microbial propagation by targeting specific viral or bacterial proteins for degradation. OPTN is degraded and cleaved at glutamine 513 following SVV infection via the activity of viral 3C protease (3C[pro]), resulting in N-terminal and a C-terminal OPTN fragments. Moreover, OPTN interacts with VP1 and targets VP1 for degradation to inhibit viral replication. The N-terminal cleaved OPTN sustained its interaction with VP1, whereas the degradation capacity targeting VP1 decreased. The inhibitory effect of N-terminal OPTN against SVV infection was significantly reduced, C-terminal OPTN failed to inhibit viral replication, and degradation of VP1 was blocked. The knockdown of OPTN resulted in reduced TBK1 activation and phosphorylation of IRF3, whereas overexpression of OPTN led to increased TBK1-IRF3 signaling. Additionally, the N-terminal OPTN diminished the activation of the type I IFN (interferon) pathway. These results show that SVV 3C[pro] targets OPTN because its cleavage impairs its function in selective autophagy and type I IFN production, revealing a novel model in which the virus develops diverse strategies for evading host autophagic machinery and type I IFN response for survival.Abbreviations: Co-IP: co-immunoprecipitation; GFP-green fluorescent protein; hpi: hours post-infection; HRP: horseradish peroxidase; IFN: interferon; IFNB/IFN-β: interferon beta; IRF3: interferon regulatory factor 3; LIR: LC3-interacting region; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MOI: multiplicity of infection; OPTN: optineurin; PBS: phosphate-buffered saline; SVV: Seneca Valley virus; SQSTM1: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TBK1: TANK binding kinase 1; TCID50: 50% tissue culture infectious doses; UBAN: ubiquitin binding in TNIP/ABIN (TNFAIP3/A20 and inhibitor of NFKB/NF-kB) and IKBKG/NEMO; UBD: ubiquitin-binding domain; ZnF: zinc finger.
Collapse
Affiliation(s)
- Jiangwei Song
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Yitong Guo
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Dan Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Rong Quan
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jing Wang
- Beijing Key Laboratory for Prevention and Control of Infectious Diseases in Livestock and Poultry, Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, China
| | - Jue Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
19
|
Yang X, Liu Z. Role of TBK1 Inhibition in Targeted Therapy of Cancer. Mini Rev Med Chem 2024; 24:1031-1045. [PMID: 38314681 DOI: 10.2174/0113895575271977231115062803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 09/16/2023] [Accepted: 09/16/2023] [Indexed: 02/06/2024]
Abstract
TANK-binding kinase 1 (TBK1) is a serine/threonine protein that plays a crucial role in various biological processes like immunity, autophagy, cell survival, and proliferation. The level and kinase activity of the TBK1 protein is regulated through post-translational modifications (PTMs). TBK1 mainly mediates the activation of IRF3/7 and NF-κB signaling pathways while also participating in the regulation of cellular activities such as autophagy, mitochondrial metabolism, and cell proliferation. TBK1 regulates immune, metabolic, inflammatory, and tumor occurrence and development within the body through these cellular activities. TBK1 kinase has emerged as a promising therapeutic target for tumor immunity. However, its molecular mechanism of action remains largely unknown. The identification of selective TBK1 small molecule inhibitors can serve as valuable tools for investigating the biological function of TBK1 protein and also as potential drug candidates for tumor immunotherapy. The current research progress indicates that some TBK1 inhibitors (compounds 15,16 and 21) exhibit certain antitumor effects in vitro culture systems. Here, we summarize the mechanism of action of TBK1 in tumors in recent years and the progress of small molecule inhibitors of TBK1.
Collapse
Affiliation(s)
- Xueqing Yang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| | - Zongliang Liu
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, China
| |
Collapse
|
20
|
Rizzuti M, Sali L, Melzi V, Scarcella S, Costamagna G, Ottoboni L, Quetti L, Brambilla L, Papadimitriou D, Verde F, Ratti A, Ticozzi N, Comi GP, Corti S, Gagliardi D. Genomic and transcriptomic advances in amyotrophic lateral sclerosis. Ageing Res Rev 2023; 92:102126. [PMID: 37972860 DOI: 10.1016/j.arr.2023.102126] [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: 06/01/2023] [Revised: 11/09/2023] [Accepted: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder and the most common motor neuron disease. ALS shows substantial clinical and molecular heterogeneity. In vitro and in vivo models coupled with multiomic techniques have provided important contributions to unraveling the pathomechanisms underlying ALS. To date, despite promising results and accumulating knowledge, an effective treatment is still lacking. Here, we provide an overview of the literature on the use of genomics, epigenomics, transcriptomics and microRNAs to deeply investigate the molecular mechanisms developing and sustaining ALS. We report the most relevant genes implicated in ALS pathogenesis, discussing the use of different high-throughput sequencing techniques and the role of epigenomic modifications. Furthermore, we present transcriptomic studies discussing the most recent advances, from microarrays to bulk and single-cell RNA sequencing. Finally, we discuss the use of microRNAs as potential biomarkers and promising tools for molecular intervention. The integration of data from multiple omic approaches may provide new insights into pathogenic pathways in ALS by shedding light on diagnostic and prognostic biomarkers, helping to stratify patients into clinically relevant subgroups, revealing novel therapeutic targets and supporting the development of new effective therapies.
Collapse
Affiliation(s)
- Mafalda Rizzuti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Sali
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Valentina Melzi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Simone Scarcella
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Gianluca Costamagna
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Linda Ottoboni
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy
| | - Lorenzo Quetti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Lorenzo Brambilla
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Federico Verde
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Antonia Ratti
- Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy; Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Nicola Ticozzi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Department of Neurology and Laboratory of Neuroscience, IRCCS Istituto Auxologico Italiano, Milan, Italy
| | - Giacomo Pietro Comi
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy; Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefania Corti
- Neurology Unit, Foundation IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| | - Delia Gagliardi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Università degli Studi di Milano, Milan, Italy.
| |
Collapse
|
21
|
Fox AR, Fingert JH. Familial normal tension glaucoma genetics. Prog Retin Eye Res 2023; 96:101191. [PMID: 37353142 DOI: 10.1016/j.preteyeres.2023.101191] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/25/2023]
Abstract
Glaucoma is defined by characteristic optic nerve damage and corresponding visual field defects and is the leading cause of irreversible blindness in the world. Elevated intraocular pressure (IOP) is a strong risk factor for developing glaucoma. However, glaucoma can occur at any IOP. Normal tension glaucoma (NTG) arises with IOPs that are within what has been defined as a normal range, i.e., 21 mm Hg or less, which may present challenges in its diagnosis and management. Identifying inheritance patterns and genetic mutations in families with NTG has helped elucidate mechanisms of NTG, however the pathophysiology is complex and not fully understood. Approximately 2% of NTG cases are caused primarily by mutations in single genes, optineurin (OPTN), TANK binding kinase 1 (TKB1), or myocilin (MYOC). Herein, we review pedigree studies of NTG and autosomal dominant NTG caused by OPTN, TBK1, and MYOC mutations. We review identified mutations and resulting clinical features of OPTN-associated and TBK1-associated NTG, including long-term follow up of these patients with NTG. In addition, we report a new four-generation pedigree of NTG caused by a Glu50Lys OPTN mutation, including six family members with a mean follow up of 17 years. Common features of OPTN -associated NTG due to Glu50Lys mutation included early onset of disease with an IOP <21 mm Hg, marked optic disc cupping, and progressive visual field loss which appeared to stabilize once an IOP of less than 10 mm Hg was achieved. Lastly, we review risk factor genes which have been identified to contribute to the complex inheritance of NTG.
Collapse
Affiliation(s)
- Austin R Fox
- Gavin Herbert Eye Institute, University of California, Irvine, CA, USA
| | - John H Fingert
- Institute for Vision Research, University of Iowa, Iowa City, IA, USA; Department of Ophthalmology and Visual Sciences, Carver College of Medicine, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
22
|
Du YX, Mamun AA, Lyu AP, Zhang HJ. Natural Compounds Targeting the Autophagy Pathway in the Treatment of Colorectal Cancer. Int J Mol Sci 2023; 24:7310. [PMID: 37108476 PMCID: PMC10138367 DOI: 10.3390/ijms24087310] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/03/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Autophagy is a highly conserved intracellular degradation pathway by which misfolded proteins or damaged organelles are delivered in a double-membrane vacuolar vesicle and finally degraded by lysosomes. The risk of colorectal cancer (CRC) is high, and there is growing evidence that autophagy plays a critical role in regulating the initiation and metastasis of CRC; however, whether autophagy promotes or suppresses tumor progression is still controversial. Many natural compounds have been reported to exert anticancer effects or enhance current clinical therapies by modulating autophagy. Here, we discuss recent advancements in the molecular mechanisms of autophagy in regulating CRC. We also highlight the research on natural compounds that are particularly promising autophagy modulators for CRC treatment with clinical evidence. Overall, this review illustrates the importance of autophagy in CRC and provides perspectives for these natural autophagy regulators as new therapeutic candidates for CRC drug development.
Collapse
Affiliation(s)
| | | | - Ai-Ping Lyu
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong SAR, China; (Y.-X.D.); (A.A.M.)
| | - Hong-Jie Zhang
- School of Chinese Medicine, Hong Kong Baptist University, 7 Baptist University Road, Kowloon Tong, Kowloon, Hong Kong SAR, China; (Y.-X.D.); (A.A.M.)
| |
Collapse
|
23
|
Zecca C, Tortelli R, Carrera P, Dell'Abate MT, Logroscino G, Ferrari M. Genotype-phenotype correlation in the spectrum of frontotemporal dementia-parkinsonian syndromes and advanced diagnostic approaches. Crit Rev Clin Lab Sci 2022; 60:171-188. [PMID: 36510705 DOI: 10.1080/10408363.2022.2150833] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The term frontotemporal dementia (FTD) refers to a group of progressive neurodegenerative disorders characterized mainly by atrophy of the frontal and anterior temporal lobes. Based on clinical presentation, three main clinical syndromes have traditionally been described: behavioral variant frontotemporal dementia (bvFTD), non-fluent/agrammatic primary progressive aphasia (nfPPA), and semantic variant PPA (svPPA). However, over the last 20 years, it has been recognized that cognitive phenotypes often overlap with motor phenotypes, either motor neuron diseases or parkinsonian signs and/or syndromes like progressive supranuclear palsy (PSP) and cortico-basal syndrome (CBS). Furthermore, FTD-related genes are characterized by genetic pleiotropy and can cause, even in the same family, pure motor phenotypes, findings that underlie the clinical continuum of the spectrum, which has pure cognitive and pure motor phenotypes as the extremes. The genotype-phenotype correlation of the spectrum, FTD-motor neuron disease, has been well defined and extensively investigated, while the continuum, FTD-parkinsonism, lacks a comprehensive review. In this narrative review, we describe the current knowledge about the genotype-phenotype correlation of the spectrum, FTD-parkinsonism, focusing on the phenotypes that are less frequent than bvFTD, namely nfPPA, svPPA, PSP, CBS, and cognitive-motor overlapping phenotypes (i.e. PPA + PSP). From a pathological point of view, they are characterized mainly by the presence of phosphorylated-tau inclusions, either 4 R or 3 R. The genetic correlate of the spectrum can be heterogeneous, although some variants seem to lead preferentially to specific clinical syndromes. Furthermore, we critically review the contribution of genome-wide association studies (GWAS) and next-generation sequencing (NGS) in disentangling the complex heritability of the FTD-parkinsonism spectrum and in defining the genotype-phenotype correlation of the entire clinical scenario, owing to the ability of these techniques to test multiple genes, and so to allow detailed investigations of the overlapping phenotypes. Finally, we conclude with the importance of a detailed genetic characterization and we offer to patients and families the chance to be included in future randomized clinical trials focused on autosomal dominant forms of FTLD.
Collapse
Affiliation(s)
- Chiara Zecca
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Rosanna Tortelli
- Neuroscience and Rare Diseases Discovery and Translational Area, Roche Pharma Research and Early Development, F. Hoffmann-La Roche Ltd, Basel, Switzerland
| | - Paola Carrera
- Unit of Genomics for Human Disease Diagnosis and Clinical Molecular Biology Laboratory, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Teresa Dell'Abate
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy
| | - Giancarlo Logroscino
- Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain, University of Bari "Aldo Moro", Pia Fondazione Card G. Panico Hospital, Tricase, Italy.,Department of Basic Medicine Sciences, Neuroscience, and Sense Organs, University of Bari "Aldo Moro", Bari, Italy
| | | |
Collapse
|
24
|
Kirola L, Mukherjee A, Mutsuddi M. Recent Updates on the Genetics of Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Mol Neurobiol 2022; 59:5673-5694. [PMID: 35768750 DOI: 10.1007/s12035-022-02934-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 06/16/2022] [Indexed: 10/17/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) primarily affect the motor and frontotemporal areas of the brain, respectively. These disorders share clinical, genetic, and pathological similarities, and approximately 10-15% of ALS-FTD cases are considered to be multisystemic. ALS-FTD overlaps have been linked to families carrying an expansion in the intron of C9orf72 along with inclusions of TDP-43 in the brain. Other overlapping genes (VCP, FUS, SQSTM1, TBK1, CHCHD10) are also involved in similar functions that include RNA processing, autophagy, proteasome response, protein aggregation, and intracellular trafficking. Recent advances in genome sequencing have identified new genes that are involved in these disorders (TBK1, CCNF, GLT8D1, KIF5A, NEK1, C21orf2, TBP, CTSF, MFSD8, DNAJC7). Additional risk factors and modifiers have been also identified in genome-wide association studies and array-based studies. However, the newly identified genes show higher disease frequencies in combination with known genes that are implicated in pathogenesis, thus indicating probable digenetic/polygenic inheritance models, along with epistatic interactions. Studies suggest that these genes play a pleiotropic effect on ALS-FTD and other diseases such as Alzheimer's disease, Ataxia, and Parkinsonism. Besides, there have been numerous improvements in the genotype-phenotype correlations as well as clinical trials on stem cell and gene-based therapies. This review discusses the possible genetic models of ALS and FTD, the latest therapeutics, and signaling pathways involved in ALS-FTD.
Collapse
Affiliation(s)
- Laxmi Kirola
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Ashim Mukherjee
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Institute of Science, Banaras Hindu University, Varanasi, Uttar Pradesh, India.
| |
Collapse
|
25
|
Chawla K, Subramanian G, Rahman T, Fan S, Chakravarty S, Gujja S, Demchak H, Chakravarti R, Chattopadhyay S. Autophagy in Virus Infection: A Race between Host Immune Response and Viral Antagonism. IMMUNO 2022; 2:153-169. [PMID: 35252965 PMCID: PMC8893043 DOI: 10.3390/immuno2010012] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Virus-infected cells trigger a robust innate immune response and facilitate virus replication. Here, we review the role of autophagy in virus infection, focusing on both pro-viral and anti-viral host responses using a select group of viruses. Autophagy is a cellular degradation pathway operated at the basal level to maintain homeostasis and is induced by external stimuli for specific functions. The degradative function of autophagy is considered a cellular anti-viral immune response. However, autophagy is a double-edged sword in viral infection; viruses often benefit from it, and the infected cells can also use it to inhibit viral replication. In addition to viral regulation, autophagy pathway proteins also function in autophagy-independent manners to regulate immune responses. Since viruses have co-evolved with hosts, they have developed ways to evade the anti-viral autophagic responses of the cells. Some of these mechanisms are also covered in our review. Lastly, we conclude with the thought that autophagy can be targeted for therapeutic interventions against viral diseases.
Collapse
Affiliation(s)
- Karan Chawla
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Gayatri Subramanian
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Tia Rahman
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Shumin Fan
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Sukanya Chakravarty
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Shreyas Gujja
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Hayley Demchak
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Ritu Chakravarti
- Department of Physiology and Pharmacology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
| | - Saurabh Chattopadhyay
- Department of Medical Microbiology and Immunology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA
- Correspondence:
| |
Collapse
|
26
|
Mun DG, Vanderboom PM, Madugundu AK, Garapati K, Chavan S, Peterson JA, Saraswat M, Pandey A. DIA-Based Proteome Profiling of Nasopharyngeal Swabs from COVID-19 Patients. J Proteome Res 2021; 20:4165-4175. [PMID: 34292740 PMCID: PMC8315246 DOI: 10.1021/acs.jproteome.1c00506] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Indexed: 12/15/2022]
Abstract
Since the recent outbreak of COVID-19, there have been intense efforts to understand viral pathogenesis and host immune response to combat SARS-CoV-2. It has become evident that different host alterations can be identified in SARS-CoV-2 infection based on whether infected cells, animal models or clinical samples are studied. Although nasopharyngeal swabs are routinely collected for SARS-CoV-2 detection by RT-PCR testing, host alterations in the nasopharynx at the proteomic level have not been systematically investigated. Thus, we sought to characterize the host response through global proteome profiling of nasopharyngeal swab specimens. A mass spectrometer combining trapped ion mobility spectrometry (TIMS) and high-resolution QTOF mass spectrometer with parallel accumulation-serial fragmentation (PASEF) was deployed for unbiased proteome profiling. First, deep proteome profiling of pooled nasopharyngeal swab samples was performed in the PASEF enabled DDA mode, which identified 7723 proteins that were then used to generate a spectral library. This approach provided peptide level evidence of five missing proteins for which MS/MS spectrum and mobilograms were validated with synthetic peptides. Subsequently, quantitative proteomic profiling was carried out for 90 individual nasopharyngeal swab samples (45 positive and 45 negative) in DIA combined with PASEF, termed as diaPASEF mode, which resulted in a total of 5023 protein identifications. Of these, 577 proteins were found to be upregulated in SARS-CoV-2 positive samples. Functional analysis of these upregulated proteins revealed alterations in several biological processes including innate immune response, viral protein assembly, and exocytosis. To the best of our knowledge, this study is the first to deploy diaPASEF for quantitative proteomic profiling of clinical samples and shows the feasibility of adopting such an approach to understand mechanisms and pathways altered in diseases.
Collapse
Affiliation(s)
- Dong-Gi Mun
- Department of Laboratory Medicine and Pathology,
Mayo Clinic, 200 First Street Southwest, Rochester, Minnesota
55905, United States
| | - Patrick M. Vanderboom
- Department of Laboratory Medicine and Pathology,
Mayo Clinic, 200 First Street Southwest, Rochester, Minnesota
55905, United States
| | - Anil K. Madugundu
- Department of Laboratory Medicine and Pathology,
Mayo Clinic, 200 First Street Southwest, Rochester, Minnesota
55905, United States
- Center for Molecular Medicine, National
Institute of Mental Health and Neurosciences, Hosur Road, Bangalore,
560029, Karnataka India
- Institute of Bioinformatics, International
Technology Park, Bangalore, 560066, Karnataka
India
- Manipal Academy of Higher
Education, Manipal, 576104, Karnataka India
| | - Kishore Garapati
- Department of Laboratory Medicine and Pathology,
Mayo Clinic, 200 First Street Southwest, Rochester, Minnesota
55905, United States
- Institute of Bioinformatics, International
Technology Park, Bangalore, 560066, Karnataka
India
- Manipal Academy of Higher
Education, Manipal, 576104, Karnataka India
| | - Sandip Chavan
- Department of Laboratory Medicine and Pathology,
Mayo Clinic, 200 First Street Southwest, Rochester, Minnesota
55905, United States
| | - Jane A. Peterson
- Proteomics Core, Medical Genome Facility,
Mayo Clinic, Rochester, Minnesota 55905, United
States
| | - Mayank Saraswat
- Department of Laboratory Medicine and Pathology,
Mayo Clinic, 200 First Street Southwest, Rochester, Minnesota
55905, United States
- Institute of Bioinformatics, International
Technology Park, Bangalore, 560066, Karnataka
India
- Manipal Academy of Higher
Education, Manipal, 576104, Karnataka India
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology,
Mayo Clinic, 200 First Street Southwest, Rochester, Minnesota
55905, United States
- Center for Molecular Medicine, National
Institute of Mental Health and Neurosciences, Hosur Road, Bangalore,
560029, Karnataka India
- Center for Individualized Medicine, Mayo
Clinic, Rochester, Minnesota 55905, United States
| |
Collapse
|
27
|
Umair M, Khan S, Mohammad T, Shafie A, Anjum F, Islam A, Hassan MI. Impact of single amino acid substitution on the structure and function of TANK-binding kinase-1. J Cell Biochem 2021; 122:1475-1490. [PMID: 34237165 DOI: 10.1002/jcb.30070] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 12/14/2022]
Abstract
Tank-binding kinase 1 (TBK1) is a serine/threonine protein kinase involved in various signaling pathways and subsequently regulates cell proliferation, apoptosis, autophagy, antiviral and antitumor immunity. Dysfunction of TBK1 can cause many complex diseases, including autoimmunity, neurodegeneration, and cancer. This dysfunction of TBK1 may result from single amino acid substitutions and subsequent structural alterations. This study analyzed the effect of substituting amino acids on TBK1 structure, function, and subsequent disease using advanced computational methods and various tools. In the initial assessment, a total of 467 mutations were found to be deleterious. After that, in detailed structural and sequential analyses, 13 mutations were found to be pathogenic. Finally, based on the functional importance, two variants (K38D and S172A) of the TBK1 kinase domain were selected and studied in detail by utilizing all-atom molecular dynamics (MD) simulation for 200 ns. MD simulation, including correlation matrix and principal component analysis, helps to get deeper insights into the TBK1 structure at the atomic level. We observed a substantial change in variants' conformation, which may be possible for structural alteration and subsequent TBK1 dysfunction. However, substitution S172A shows a significant conformational change in TBK1 structure as compared to K38D. Thus, this study provides a structural basis to understand the effect of mutations on the kinase domain of TBK1 and its function associated with disease progression.
Collapse
Affiliation(s)
- Mohd Umair
- Department of Computer Science, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Shama Khan
- Drug Discovery and Development Centre (H3D), University of Cape Town, Rondebosch, South Africa
| | - Taj Mohammad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| |
Collapse
|
28
|
Moya GE, Rivera PD, Dittenhafer-Reed KE. Evidence for the Role of Mitochondrial DNA Release in the Inflammatory Response in Neurological Disorders. Int J Mol Sci 2021; 22:7030. [PMID: 34209978 PMCID: PMC8268735 DOI: 10.3390/ijms22137030] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/25/2021] [Accepted: 06/26/2021] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are regarded as the metabolic centers of cells and are integral in many other cell processes, including the immune response. Each mitochondrion contains numerous copies of mitochondrial DNA (mtDNA), a small, circular, and bacterial-like DNA. In response to cellular damage or stress, mtDNA can be released from the mitochondrion and trigger immune and inflammatory responses. mtDNA release into the cytosol or bloodstream can occur as a response to hypoxia, sepsis, traumatic injury, excitatory cytotoxicity, or drastic mitochondrial membrane potential changes, some of which are hallmarks of neurodegenerative and mood disorders. Released mtDNA can mediate inflammatory responses observed in many neurological and mood disorders by driving the expression of inflammatory cytokines and the interferon response system. The current understanding of the role of mtDNA release in affective mood disorders and neurodegenerative diseases will be discussed.
Collapse
Affiliation(s)
| | - Phillip D. Rivera
- Department of Chemistry and Biology, Hope College, Holland, MI 49423, USA;
| | | |
Collapse
|
29
|
Daussy CF, Pied N, Wodrich H. Understanding Post Entry Sorting of Adenovirus Capsids; A Chance to Change Vaccine Vector Properties. Viruses 2021; 13:1221. [PMID: 34202573 PMCID: PMC8310329 DOI: 10.3390/v13071221] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/25/2022] Open
Abstract
Adenovirus vector-based genetic vaccines have emerged as a powerful strategy against the SARS-CoV-2 health crisis. This success is not unexpected because adenoviruses combine many desirable features of a genetic vaccine. They are highly immunogenic and have a low and well characterized pathogenic profile paired with technological approachability. Ongoing efforts to improve adenovirus-vaccine vectors include the use of rare serotypes and non-human adenoviruses. In this review, we focus on the viral capsid and how the choice of genotypes influences the uptake and subsequent subcellular sorting. We describe how understanding capsid properties, such as stability during the entry process, can change the fate of the entering particles and how this translates into differences in immunity outcomes. We discuss in detail how mutating the membrane lytic capsid protein VI affects species C viruses' post-entry sorting and briefly discuss if such approaches could have a wider implication in vaccine and/or vector development.
Collapse
Affiliation(s)
| | | | - Harald Wodrich
- Microbiologie Fondamentale et Pathogénicité, MFP CNRS UMR 5234, University of Bordeaux, 146 rue Leo Saignat, CEDEX, 33076 Bordeaux, France; (C.F.D.); (N.P.)
| |
Collapse
|
30
|
Deng H, Zeng L, Chang K, Lv Y, Du H, Lu S, Liu Y, Zhou P, Mao H, Hu C. Grass carp (Ctenopharyngodon idellus) Cdc25a down-regulates IFN 1 expression by reducing TBK1 phosphorylation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 118:104014. [PMID: 33460677 DOI: 10.1016/j.dci.2021.104014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Revised: 01/11/2021] [Accepted: 01/11/2021] [Indexed: 02/07/2023]
Abstract
In vertebrates, TANK Binding Kinase 1 (TBK1) plays an important role in innate immunity, mainly because it can mediate production of interferon to resist the invasion of pathogens. In mammals, cell division cycle-25a (Cdc25a) is a member of the Cdc25 family of cell division cycle proteins. It is a phosphatase that plays an important role in cell cycle regulation by dephosphorylating its substrate proteins. Currently, many phosphatases are reported to play a role in innate immunity. This is because the phosphatases can shut down or reduce immune signaling pathways by down-regulating phosphorylation signals. However, there are no reports on fish Cdc25a in innate immunity. In this paper, we conducted a preliminary study on the involvement of grass carp Cdc25a in innate immunity. First, we cloned the full-length cDNA of grass carp Cdc25a (CiCdc25a), and found that it shares the highest genetic relationship with that of Anabarilius grahami through phylogenetic tree comparison. In grass carp tissues and CIK cells, the expression of CiCdc25a mRNA was up-regulated under poly (I:C) stimulation. Therefore, CiCdc25a can respond to poly (I:C). The subcellular localization results showed that CiCdc25a is distributed both in the cytoplasm and nucleus. We also found that CiCdc25a can down-regulate the expression of IFN 1 with or without poly (I:C) stimulation. In other words, the down-regulation of IFN1 by CiCdc25a is independent of poly (I:C) stimulation. Further functional studies have shown that the inhibition of IFN1 expression by CiCdc25a may be related to decrease of TBK1 activity. We also confirmed that the phosphorylation of TBK1 at Ser172 is essential for production of IFN 1. In short, CiCdc25a can interact with TBK1 and subsequently inhibits the phosphorylation of TBK1, thereby weakens TBK1 activity. These results indicated that grass carp Cdc25a down-regulates IFN 1 expression by reducing TBK1 phosphorylation.
Collapse
Affiliation(s)
- Hang Deng
- College of Life Science, Nanchang University, Nanchang 330031, China
| | - Liugen Zeng
- Nanchang Academy of Agricultural Sciences, Nanchang, 330038, China
| | - Kaile Chang
- College of Life Science, Nanchang University, Nanchang 330031, China
| | - Yangfeng Lv
- College of Life Science, Nanchang University, Nanchang 330031, China
| | - Hailing Du
- College of Life Science, Nanchang University, Nanchang 330031, China
| | - Shina Lu
- College of Life Science, Nanchang University, Nanchang 330031, China
| | - Yapeng Liu
- College of Life Science, Nanchang University, Nanchang 330031, China
| | - Pengcheng Zhou
- College of Life Science, Nanchang University, Nanchang 330031, China
| | - Huiling Mao
- College of Life Science, Nanchang University, Nanchang 330031, China.
| | - Chengyu Hu
- College of Life Science, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
31
|
Wang J, Luan Y, Fan EK, Scott MJ, Li Y, Billiar TR, Wilson MA, Jiang Y, Fan J. TBK1/IKKε Negatively Regulate LPS-Induced Neutrophil Necroptosis and Lung Inflammation. Shock 2021; 55:338-348. [PMID: 32925605 PMCID: PMC8183424 DOI: 10.1097/shk.0000000000001632] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
ABSTRACT Cell necroptosis, a form of regulated inflammatory cell death, is one of the mechanisms that controls cell release of inflammatory mediators from innate immune cells, such as polymorphonuclear neutrophils (PMNs), and critically regulates the progress of inflammation. Cell necroptosis features receptor-interacting protein (RIPK) 1 activation and necroptosome formation. This leads to loss of plasma membrane integrity, the release of cell contents into the extracellular space, and subsequent increased inflammation. Here, we report an intra-PMN mechanism of negative regulation of necroptosis mediated through TBK1/IKKε. Using an in vivo mouse model of intratracheal injection (i.t.) of LPS and in vitro LPS stimulation of mouse PMN, we found that LPS-TLR4 signaling in PMNs activates and phosphorylates TBK1 and IKKε, which in turn suppress LPS-induced formation of the RIPK1-RIPK3-MLKL (necrosome) complex. TBK1 dysfunction by knockdown or inhibitor significantly increases the phosphorylation of RIPK1 (∼67%), RIPK3 (∼68%), and MLKL (∼50%) and promotes RIPK1-RIPK3 and RIPK3-MLKL interactions and increases PMN necroptosis (∼83%) in response to LPS, with subsequent augmented lung inflammation. These findings suggest that the LPS-TLR4-TBK1 axis serves as a negative regulator for PMN necroptosis and might be a therapeutic target for modulating PMN death and inflammation.
Collapse
Affiliation(s)
- Jieyan Wang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yingyi Luan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Trauma Research Center, Fourth Medical Center of the Chinese PLA General Hospital, Beijing China
| | - Erica K. Fan
- Department of Epidemiology, University of Pittsburgh Graduate School of Public Health, Pittsburgh, Pennsylvania
| | - Melanie J. Scott
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yuehua Li
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
| | - Timothy R. Billiar
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Mark A. Wilson
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yong Jiang
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Jie Fan
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Research and Development, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, Pennsylvania
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
32
|
Chang R, Chu Q, Zheng W, Zhang L, Xu T. The Sp1-Responsive microRNA-15b Negatively Regulates Rhabdovirus-Triggered Innate Immune Responses in Lower Vertebrates by Targeting TBK1. Front Immunol 2021; 11:625828. [PMID: 33584728 PMCID: PMC7873567 DOI: 10.3389/fimmu.2020.625828] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 12/07/2020] [Indexed: 01/07/2023] Open
Abstract
As is known to all, the production of type I interferon (IFN) plays pivotal roles in host innate antiviral immunity, and its moderate production play a positive role in promoting the activation of host innate antiviral immune response. However, the virus will establish a persistent infection model by interfering with the production of IFN, thereby evading the organism inherent antiviral immune response. Therefore, it is of great necessity to research the underlying regulatory mechanisms of type I IFN appropriate production under viral invasion. In this study, we report that a Sp1–responsive miR-15b plays a negative role in siniperca chuatsi rhabdovirus (SCRV)-triggered antiviral response in teleost fish. We found that SCRV could dramatically upregulate miiuy croaker miR-15b expression. Enhanced miR-15b could negatively regulate SCRV-triggered antiviral genes and inflammatory cytokines production by targeting TANK-binding kinase 1 (TBK1), thereby accelerating viral replication. Importantly, we found that miR-15b feedback regulates antiviral innate immune response through NF-κB and IRF3 signaling pathways. These findings highlight that miR-15b plays a crucial role in regulating virus–host interactions, which outlines a new regulation mechanism of fish’s innate immune responses.
Collapse
Affiliation(s)
- Renjie Chang
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai, China
| | - Qing Chu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China
| | - Weiwei Zheng
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai, China.,National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
| | - Lei Zhang
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China
| | - Tianjun Xu
- Laboratory of Fish Molecular Immunology, College of Fisheries and Life Science, Shanghai Ocean University, Shanghai, China.,Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources (Shanghai Ocean University), Ministry of Education, Shanghai, China.,Laboratory of Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, China.,National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai, China
| |
Collapse
|
33
|
Khan KA, Marineau A, Doyon P, Acevedo M, Durette É, Gingras AC, Servant MJ. TRK-Fused Gene (TFG), a protein involved in protein secretion pathways, is an essential component of the antiviral innate immune response. PLoS Pathog 2021; 17:e1009111. [PMID: 33411856 PMCID: PMC7790228 DOI: 10.1371/journal.ppat.1009111] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Accepted: 10/30/2020] [Indexed: 12/15/2022] Open
Abstract
Antiviral innate immune response to RNA virus infection is supported by Pattern-Recognition Receptors (PRR) including RIG-I-Like Receptors (RLR), which lead to type I interferons (IFNs) and IFN-stimulated genes (ISG) production. Upon sensing of viral RNA, the E3 ubiquitin ligase TNF Receptor-Associated Factor-3 (TRAF3) is recruited along with its substrate TANK-Binding Kinase (TBK1), to MAVS-containing subcellular compartments, including mitochondria, peroxisomes, and the mitochondria-associated endoplasmic reticulum membrane (MAM). However, the regulation of such events remains largely unresolved. Here, we identify TRK-Fused Gene (TFG), a protein involved in the transport of newly synthesized proteins to the endomembrane system via the Coat Protein complex II (COPII) transport vesicles, as a new TRAF3-interacting protein allowing the efficient recruitment of TRAF3 to MAVS and TBK1 following Sendai virus (SeV) infection. Using siRNA and shRNA approaches, we show that TFG is required for virus-induced TBK1 activation resulting in C-terminal IRF3 phosphorylation and dimerization. We further show that the ability of the TRAF3-TFG complex to engage mTOR following SeV infection allows TBK1 to phosphorylate mTOR on serine 2159, a post-translational modification shown to promote mTORC1 signaling. We demonstrate that the activation of mTORC1 signaling during SeV infection plays a positive role in the expression of Viperin, IRF7 and IFN-induced proteins with tetratricopeptide repeats (IFITs) proteins, and that depleting TFG resulted in a compromised antiviral state. Our study, therefore, identifies TFG as an essential component of the RLR-dependent type I IFN antiviral response. Antiviral innate immune response is the first line of defence against the invading viruses through type I interferon (IFN) signaling. However, viruses have devised ways to target signaling molecules for aberrant IFN response and worsen the disease outcome. As such, deciphering the roles of new regulators of innate immunity could transform the antiviral treatment paradigm by introducing novel panviral therapeutics designed to reinforce antiviral host responses. This could be of great use in fighting recent outbreaks of severe acute respiratory syndrome coronavirus (SARS-CoV), Middle East respiratory syndrome MERS-CoV, and the more recent SARS-CoV-2 causing the COVID-19 pandemic. However, aberrant activation of such pathways can lead to detrimental consequences, including autoimmune diseases. Regulation of type I IFN responses is thus of paramount importance. To prevent an uncontrolled response, signaling events happen in discrete subcellular compartments, therefore, distinguishing sites involved in recognition of pathogens and those permitting downstream signaling. Here, we show TFG as a new regulator of type I IFN response allowing the efficient organization of signaling molecules. TFG, thus, further substantiates the importance of the protein trafficking machinery in the regulation of optimal antiviral responses. Our findings have implications for both antiviral immunity and autoimmune diseases.
Collapse
Affiliation(s)
| | | | - Priscilla Doyon
- Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - Mariana Acevedo
- Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - Étienne Durette
- Faculty of Pharmacy, Université de Montréal, Montréal, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Marc J. Servant
- Faculty of Pharmacy, Université de Montréal, Montréal, Canada
- * E-mail:
| |
Collapse
|
34
|
Abreha MH, Ojelade S, Dammer EB, McEachin ZT, Duong DM, Gearing M, Bassell GJ, Lah JJ, Levey AI, Shulman JM, Seyfried NT. TBK1 interacts with tau and enhances neurodegeneration in tauopathy. J Biol Chem 2021; 296:100760. [PMID: 33965374 PMCID: PMC8191334 DOI: 10.1016/j.jbc.2021.100760] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/29/2021] [Accepted: 05/05/2021] [Indexed: 12/12/2022] Open
Abstract
One of the defining pathological features of Alzheimer's disease (AD) is the deposition of neurofibrillary tangles (NFTs) composed of hyperphosphorylated tau in the brain. Aberrant activation of kinases in AD has been suggested to enhance phosphorylation and toxicity of tau, making the responsible tau kinases attractive therapeutic targets. The full complement of tau-interacting kinases in AD brain and their activity in disease remains incompletely defined. Here, immunoaffinity enrichment coupled with mass spectrometry (MS) identified TANK-binding kinase 1 (TBK1) as a tau-interacting partner in human AD cortical brain tissues. We validated this interaction in human AD, familial frontotemporal dementia and parkinsonism linked to chromosome 17 (FTDP-17) caused by mutations in MAPT (R406W & P301L) and corticobasal degeneration (CBD) postmortem brain tissues as well as human cell lines. Further, we document increased TBK1 activation in both AD and FTDP-17 and map TBK1 phosphorylation sites on tau based on in vitro kinase assays coupled to MS. Lastly, in a Drosophila tauopathy model, activating expression of a conserved TBK1 ortholog triggers tau hyperphosphorylation and enhanced neurodegeneration, whereas knockdown had the reciprocal effect, suppressing tau toxicity. Collectively, our findings suggest that increased TBK1 activation may promote tau hyperphosphorylation and neuronal loss in AD and related tauopathies.
Collapse
Affiliation(s)
- Measho H Abreha
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Shamsideen Ojelade
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA
| | - Eric B Dammer
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Zachary T McEachin
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Duc M Duong
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Marla Gearing
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Gary J Bassell
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Cell Biology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - James J Lah
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Allan I Levey
- Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Joshua M Shulman
- Department of Neurology, Baylor College of Medicine, Houston, Texas, USA; Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, Texas, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas, USA; Department of Neuroscience, Baylor College of Medicine, Houston, Texas, USA.
| | - Nicholas T Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, Georgia, USA; Center for Neurodegenerative Diseases, Emory University School of Medicine, Atlanta, Georgia, USA; Department of Neurology, Emory University School of Medicine, Atlanta, Georgia, USA.
| |
Collapse
|
35
|
Chen J, Zhou XY, Li P, Li ZC, Zhang C, Sun YH, Wang GY, Chen DD, Lu LF, Li S. Molecular characterization of a cyprinid fish (Ancherythroculter nigrocauda) TBK1 and its kinase activity in IFN regulation. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103805. [PMID: 32755617 DOI: 10.1016/j.dci.2020.103805] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 07/19/2020] [Accepted: 07/19/2020] [Indexed: 06/11/2023]
Abstract
TANK-binding kinase 1 (TBK1) plays a vital role in activating interferon (IFN) production and positively regulating antiviral response in mammals. Research on more species of fish is necessary to clarify whether the function of fish TBK1 is conserved compared to that in mammals. Here, a cyprinid fish (Ancherythroculter nigrocauda) TBK1 (AnTBK1) was functionally identified and characterized. The full-length open reading frame (ORF) of AnTBK1 consists of 2184 nucleotides encoding 727 amino acids and contains a conserved Serine/Threonine protein kinase catalytic domain (S_TKc) in the N-terminal, similar to TBK1 in other species. The transcripts of AnTBK1 were found in all the tissues evaluated and the cellular distribution indicated that AnTBK1 was localized in the cytoplasm. In terms of functional identification, AnTBK1 induced a variety of IFN promoter activities as well as the expression of downstream IFN-stimulated genes (ISGs). In addition, AnTBK1 interacted with and significantly phosphorylated IFN regulatory factor 3 (IRF3), exhibiting the canonical kinase activity of TBK1. Finally, AnTBK1 presented strong antiviral activity against spring viremia of carp virus (SVCV) infection. Taken together, our research on the features and functions of AnTBK1 demonstrated that AnTBK1 plays a central role in IFN induction against SVCV infection.
Collapse
Affiliation(s)
- Jian Chen
- Fisheries Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Xiao-Yu Zhou
- College of Fisheries and Life Science, Dalian Ocean University, Dalian, China; Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Pei Li
- Fisheries Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Zhuo-Cong Li
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Can Zhang
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China; University of Chinese Academy of Sciences, Beijing, China
| | - Yan-Hong Sun
- Fisheries Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Gui-Ying Wang
- Fisheries Research Institute, Wuhan Academy of Agricultural Sciences, Wuhan, China
| | - Dan-Dan Chen
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Long-Feng Lu
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.
| | - Shun Li
- Key Laboratory of Aquaculture Disease Control, Ministry of Agriculture, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.
| |
Collapse
|
36
|
Inagaki S, Kawase K, Funato M, Seki J, Kawase C, Ohuchi K, Kameyama T, Ando S, Sato A, Morozumi W, Nakamura S, Shimazawa M, Iejima D, Iwata T, Yamamoto T, Kaneko H, Hara H. Effect of Timolol on Optineurin Aggregation in Transformed Induced Pluripotent Stem Cells Derived From Patient With Familial Glaucoma. ACTA ACUST UNITED AC 2020; 59:2293-2304. [DOI: 10.1167/iovs.17-22975] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Satoshi Inagaki
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Kazuhide Kawase
- Department of Ophthalmology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Michinori Funato
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Junko Seki
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Chizuru Kawase
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Kazuki Ohuchi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Tsubasa Kameyama
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Shiori Ando
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Arisu Sato
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Wataru Morozumi
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Shinsuke Nakamura
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Masamitsu Shimazawa
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| | - Daisuke Iejima
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Takeshi Iwata
- Division of Molecular and Cellular Biology, National Institute of Sensory Organs, National Hospital Organization Tokyo Medical Center, Tokyo, Japan
| | - Tetsuya Yamamoto
- Department of Ophthalmology, Gifu University Graduate School of Medicine, Gifu, Japan
| | - Hideo Kaneko
- Department of Clinical Research, National Hospital Organization, Nagara Medical Center, Gifu, Japan
| | - Hideaki Hara
- Molecular Pharmacology, Department of Biofunctional Evaluation, Gifu Pharmaceutical University, Gifu, Japan
| |
Collapse
|
37
|
Abstract
Deregulated phosphatidylinositide 3-kinase (PI3K) signaling plays a crucial role in the biology of different lymphoma entities leading to the proliferation and survival of the malignant cells. Due to novel treatment options and modern supportive care, the outcome of patients with lymphomas has significantly improved in the past years. However, patients with relapsed or refractory disease still have a limited prognosis. PI3K inhibitors represent a modern and effective therapeutic option for patients with different types of lymphoma. However, the efficacy of PI3K inhibitors varies among lymphoma entities. Additionally, severe toxicity including infectious and autoimmune complications leading to therapy-related deaths has been observed. Next-generation PI3K inhibitors show promising efficacy and manageable toxicity profiles. Future research might identify effective combinatorial therapy approaches for PI3K inhibitors to further improve response rates. This review discusses the most recent developments in the field of PI3K inhibition in different subtypes of lymphoma.
Collapse
Affiliation(s)
- Philipp Berning
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| | - Georg Lenz
- Department of Medicine A, Hematology, Oncology, and Pneumology, University Hospital Münster, Münster, Germany
| |
Collapse
|
38
|
Revach OY, Liu S, Jenkins RW. Targeting TANK-binding kinase 1 (TBK1) in cancer. Expert Opin Ther Targets 2020; 24:1065-1078. [PMID: 32962465 PMCID: PMC7644630 DOI: 10.1080/14728222.2020.1826929] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 09/18/2020] [Indexed: 12/16/2022]
Abstract
INTRODUCTION TANK-binding kinase 1 (TBK1) is a Ser/Thr kinase with a central role in coordinating the cellular response to invading pathogens and regulating key inflammatory signaling cascades. While intact TBK1 signaling is required for successful anti-viral signaling, dysregulated TBK1 signaling has been linked to a variety of pathophysiologic conditions, including cancer. Several lines of evidence support a role for TBK1 in cancer pathogenesis, but the specific roles and regulation of TBK1 remain incompletely understood. A key challenge is the diversity of cellular processes that are regulated by TBK1, including inflammation, cell cycle, autophagy, energy homeostasis, and cell death. Nevertheless, evidence from pre-clinical cancer models suggests that targeting TBK1 may be an effective strategy for anti-cancer therapy in specific settings. AREAS COVERED This review provides an overview of the roles and regulation of TBK1 with a focus on cancer pathogenesis and drug targeting of TBK1 as an anti-cancer strategy. Relevant literature was derived from a PubMed search encompassing studies from 1999 to 2020. EXPERT OPINION TBK1 is emerging as a potential target for anti-cancer therapy. Inhibition of TBK1 alone may be insufficient to restrain the growth of most cancers; hence, combination strategies will likely be necessary. Improved understanding of tumor-intrinsic and tumor-extrinsic TBK1 signaling will inform novel therapeutic strategies.
Collapse
Affiliation(s)
- Or-yam Revach
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Shuming Liu
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Russell W. Jenkins
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
- Laboratory of Systems Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Lu LF, Li ZC, Zhang C, Zhou XY, Zhou Y, Jiang JY, Chen DD, Li S, Zhang YA. Grass Carp Reovirus (GCRV) Giving Its All to Suppress IFN Production by Countering MAVS Signaling Transduction. Front Immunol 2020; 11:545302. [PMID: 33193312 PMCID: PMC7649419 DOI: 10.3389/fimmu.2020.545302] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 10/06/2020] [Indexed: 01/05/2023] Open
Abstract
Viruses typically target host RIG-I-like receptors (RLRs), a group of key factors involved in interferon (IFN) production, to enhance viral infection. To date, though immune evasion methods to contradict IFN production have been characterized for a series of terrestrial viruses, the strategies employed by fish viruses remain unclear. Here, we report that all grass carp reovirus (GCRV) proteins encoded by segments S1 to S11 suppress mitochondrial antiviral signaling protein (MAVS)-mediated IFN expression. First, the GCRV viral proteins blunted the MAVS-induced expression of IFN, and impair MAVS antiviral capacity significantly. Interestingly, subsequent co-immunoprecipitation experiments demonstrated that all GCRV viral proteins interacted with several RLR cascades, especially with TANK-binding kinase 1 (TBK1) which was the downstream factor of MAVS. To further illustrate the mechanisms of these interactions between GCRV viral proteins and host RLRs, two of the viral proteins, NS79 (S4) and VP3 (S3), were selected as representative proteins for two distinguished mechanisms. The obtained data demonstrated that NS79 was phosphorylated by gcTBK1, leading to the reduction of host substrate gcIRF3/7 phosphorylation. On the other hand, VP3 degraded gcMAVS and the degradation was significantly reversed by 3-MA. The biological effects of both NS79 and VP3 were consistently found to be related to the suppression of IFN expression and the promotion of viral evasion. Our findings shed light on the special evasion mechanism utilized by fish virus through IFN regulation, which might differ between fish and mammals.
Collapse
Affiliation(s)
- Long-Feng Lu
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Zhuo-Cong Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Can Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiao-Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries and Life Science, Dalian Ocean University, Dalian, China
| | - Yu Zhou
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jing-Yu Jiang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Dan-Dan Chen
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Shun Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Yong-An Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China.,College of Fisheries, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
40
|
Zhao P, Saltiel AR. Interaction of Adipocyte Metabolic and Immune Functions Through TBK1. Front Immunol 2020; 11:592949. [PMID: 33193441 PMCID: PMC7606291 DOI: 10.3389/fimmu.2020.592949] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 09/30/2020] [Indexed: 12/19/2022] Open
Abstract
Adipocytes and adipose tissue play critical roles in the regulation of metabolic homeostasis. In obesity and obesity-associated metabolic diseases, immune cells infiltrate into adipose tissues. Interaction between adipocytes and immune cells re-shapes both metabolic and immune properties of adipose tissue and dramatically changes metabolic set points. Both the expression and activity of the non-canonical IKK family member TBK1 are induced in adipose tissues during diet-induced obesity. TBK1 plays important roles in the regulation of both metabolism and inflammation in adipose tissue and thus affects glucose and energy metabolism. Here we review the regulation and functions of TBK1 and the molecular mechanisms by which TBK1 regulates both metabolism and inflammation in adipose tissue. Finally, we discuss the potential of a TBK1/IKKε inhibitor as a new therapy for metabolic diseases.
Collapse
Affiliation(s)
- Peng Zhao
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Alan R Saltiel
- Department of Medicine, University of California San Diego, La Jolla, CA, United States.,Department of Pharmacology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
41
|
Sieverding K, Ulmer J, Bruno C, Satoh T, Tsao W, Freischmidt A, Akira S, Wong PC, Ludolph AC, Danzer KM, Lobsiger CS, Brenner D, Weishaupt JH. Hemizygous deletion of Tbk1 worsens neuromuscular junction pathology in TDP-43 G298S transgenic mice. Exp Neurol 2020; 335:113496. [PMID: 33038415 DOI: 10.1016/j.expneurol.2020.113496] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/26/2020] [Accepted: 10/04/2020] [Indexed: 12/12/2022]
Abstract
Mutations in the genes TARDBP (encoding the TDP-43 protein) and TBK1 can cause familial ALS. Neuronal cytoplasmatic accumulations of the misfolded, hyperphosphorylated RNA-binding protein TDP-43 are the pathological hallmark of most ALS cases and have been suggested to be a key aspect of ALS pathogenesis. Pharmacological induction of autophagy has been shown to reduce mutant TDP-43 aggregates and alleviate motor deficits in mice. TBK1 is exemplary for several other ALS genes that regulate autophagy. Consequently, we employed double mutant mice with both a heterozygous Tbk1 deletion and transgenic expression of human TDP-43G298S to test the hypothesis that impaired autophagy reduces intracellular clearance of an aggregation-prone protein and enhances toxicity of mutant TDP-43. The heterozygous deletion of Tbk1 did not change expression or cellular distribution of TDP-43 protein, motor neuron loss or reactive gliosis in the spinal cord of double-mutant mice at the age of 19 months. However, it aggravated muscle denervation and, albeit to a small and variable degree, motor dysfunction in TDP-43G298S transgenic mice, as similarly observed in the SOD1G93A transgenic mouse model for ALS before. Conclusively, our findings suggest that TBK1 mutations can affect the neuromuscular synapse.
Collapse
Affiliation(s)
| | - Johannes Ulmer
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Clara Bruno
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Takashi Satoh
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - William Tsao
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, United States
| | | | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Philip C Wong
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, United States; Cellular and Molecular Medicine Program, Johns Hopkins University School of Medicine, Baltimore, United States
| | | | - Karin M Danzer
- Department of Neurology, University of Ulm, Ulm, Germany
| | - Christian S Lobsiger
- Institut du Cerveau et de la Moelle Épinière, Institut National de la Santé et de la Recherche Médicale Unité 1127, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 7225, Sorbonne Université, Paris, France
| | - David Brenner
- Department of Neurology, University of Ulm, Ulm, Germany; Division of Neurodegenerative Disorders, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Germany
| | - Jochen H Weishaupt
- Department of Neurology, University of Ulm, Ulm, Germany; Division of Neurodegenerative Disorders, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim, Heidelberg University, Germany.
| |
Collapse
|
42
|
Häkkinen S, Chu SA, Lee SE. Neuroimaging in genetic frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2020; 145:105063. [PMID: 32890771 DOI: 10.1016/j.nbd.2020.105063] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) have a strong clinical, genetic and pathological overlap. This review focuses on the current understanding of structural, functional and molecular neuroimaging signatures of genetic FTD and ALS. We overview quantitative neuroimaging studies on the most common genes associated with FTD (MAPT, GRN), ALS (SOD1), and both (C9orf72), and summarize visual observations of images reported in the rarer genes (CHMP2B, TARDBP, FUS, OPTN, VCP, UBQLN2, SQSTM1, TREM2, CHCHD10, TBK1).
Collapse
Affiliation(s)
- Suvi Häkkinen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie A Chu
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Suzee E Lee
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
43
|
Bruno C, Sieverding K, Freischmidt A, Satoh T, Walther P, Mayer B, Ludolph AC, Akira S, Yilmazer-Hanke D, Danzer KM, Lobsiger CS, Brenner D, Weishaupt JH. Haploinsufficiency of TANK-binding kinase 1 prepones age-associated neuroinflammatory changes without causing motor neuron degeneration in aged mice. Brain Commun 2020; 2:fcaa133. [PMID: 33005894 PMCID: PMC7519725 DOI: 10.1093/braincomms/fcaa133] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/21/2020] [Accepted: 07/27/2020] [Indexed: 12/13/2022] Open
Abstract
Loss-of-function mutations in TANK-binding kinase 1 cause genetic amyotrophic lateral sclerosis and frontotemporal dementia. Consistent with incomplete penetrance in humans, haploinsufficiency of TANK-binding kinase 1 did not cause motor symptoms in mice up to 7 months of age in a previous study. Ageing is the strongest risk factor for neurodegenerative diseases. Hypothesizing that age-dependent processes together with haploinsufficiency of TANK-binding kinase 1 could create a double hit situation that may trigger neurodegeneration, we examined mice with hemizygous deletion of Tbk1 (Tbk1 +/- mice) and wild-type siblings up to 22 months. Compared to 4-month old mice, aged, 22-month old mice showed glial activation, deposition of motoneuronal p62 aggregates, muscular denervation and profound transcriptomic alterations in a set of 800 immune-related genes upon ageing. However, we did not observe differences regarding these measures between aged Tbk1 +/- and wild-type siblings. High age did also not precipitate TAR DNA-binding protein 43 aggregation, neurodegeneration or a neurological phenotype in Tbk1+/ - mice. In young Tbk1+/ - mice, however, we found the CNS immune gene expression pattern shifted towards the age-dependent immune system dysregulation observed in old mice. Conclusively, ageing is not sufficient to precipitate an amyotrophic lateral sclerosis or frontotemporal dementia phenotype or spinal or cortical neurodegeneration in a model of Tbk1 haploinsufficiency. We hypothesize that the consequences of Tbk1 haploinsufficiency may be highly context-dependent and require a specific synergistic stress stimulus to be uncovered.
Collapse
Affiliation(s)
- Clara Bruno
- Department of Neurology, University of Ulm, 89081 Ulm, Germany
| | | | | | - Takashi Satoh
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Paul Walther
- Central Facility for Electron Microscopy, University of Ulm, 89081 Ulm, Germany
| | - B Mayer
- Institute of Epidemiology and Medical Biometry, Ulm University, Ulm, Germany
| | | | - Shizuo Akira
- Department of Host Defense, Research Institute for Microbial Diseases, Osaka University, Osaka 565-0871, Japan
| | - Deniz Yilmazer-Hanke
- Department of Neurology, Clinical Neuroanatomy, Neurology, University of Ulm, 89081 Ulm, Germany
| | - Karin M Danzer
- Department of Neurology, University of Ulm, 89081 Ulm, Germany
| | - Christian S Lobsiger
- Institut du Cerveau et de la Moelle Épinière, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, Sorbonne Université, 75013 Paris, France
| | - David Brenner
- Department of Neurology, University of Ulm, 89081 Ulm, Germany.,Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 61867 Mannheim, Germany
| | - Jochen H Weishaupt
- Department of Neurology, University of Ulm, 89081 Ulm, Germany.,Division of Neurodegenerative Disorders, Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neurosciences, Heidelberg University, 61867 Mannheim, Germany
| |
Collapse
|
44
|
Mingione A, Ottaviano E, Barcella M, Merelli I, Rosso L, Armeni T, Cirilli N, Ghidoni R, Borghi E, Signorelli P. Cystic Fibrosis Defective Response to Infection Involves Autophagy and Lipid Metabolism. Cells 2020; 9:cells9081845. [PMID: 32781626 PMCID: PMC7463682 DOI: 10.3390/cells9081845] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/31/2020] [Accepted: 08/04/2020] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) is a hereditary disease, with 70% of patients developing a proteinopathy related to the deletion of phenylalanine 508. CF is associated with multiple organ dysfunction, chronic inflammation, and recurrent lung infections. CF is characterized by defective autophagy, lipid metabolism, and immune response. Intracellular lipid accumulation favors microbial infection, and autophagy deficiency impairs internalized pathogen clearance. Myriocin, an inhibitor of sphingolipid synthesis, significantly reduces inflammation, promotes microbial clearance in the lungs, and induces autophagy and lipid oxidation. RNA-seq was performed in Aspergillusfumigatus-infected and myriocin-treated CF patients’ derived monocytes and in a CF bronchial epithelial cell line. Fungal clearance was also evaluated in CF monocytes. Myriocin enhanced CF patients’ monocytes killing of A. fumigatus. CF patients’ monocytes and cell line responded to infection with a profound transcriptional change; myriocin regulates genes that are involved in inflammation, autophagy, lipid storage, and metabolism, including histones and heat shock proteins whose activity is related to the response to infection. We conclude that the regulation of sphingolipid synthesis induces a metabolism drift by promoting autophagy and lipid consumption. This process is driven by a transcriptional program that corrects part of the differences between CF and control samples, therefore ameliorating the infection response and pathogen clearance in the CF cell line and in CF peripheral blood monocytes.
Collapse
Affiliation(s)
- Alessandra Mingione
- Biochemistry and Molecular Biology Laboratory, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (A.M.); (R.G.)
| | - Emerenziana Ottaviano
- Laboratory of Clinical Microbiology, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (E.O.); (M.B.); (E.B.)
| | - Matteo Barcella
- Laboratory of Clinical Microbiology, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (E.O.); (M.B.); (E.B.)
| | - Ivan Merelli
- National Research Council of Italy, Institute for Biomedical Technologies, Via Fratelli Cervi 93, 20090 Milan, Italy;
| | - Lorenzo Rosso
- Health Sciences Department, University of Milan, Thoracic surgery and transplantation Unit, Fondazione IRCCS Ca Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy;
| | - Tatiana Armeni
- Department of Clinical Sciences, Section of Biochemistry, Biology and Physics, Polytechnic University of Marche, 60131 Ancona, Italy;
| | - Natalia Cirilli
- Cystic Fibrosis Referral Care Center, Mother-Child Department, United Hospitals Le Torrette, 60126 Ancona, Italy;
| | - Riccardo Ghidoni
- Biochemistry and Molecular Biology Laboratory, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (A.M.); (R.G.)
- “Aldo Ravelli” Center for Neurotechnology and Experimental Brain Therapeutics, via Antonio di Rudinì 8, 20142 Milan, Italy
| | - Elisa Borghi
- Laboratory of Clinical Microbiology, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (E.O.); (M.B.); (E.B.)
| | - Paola Signorelli
- Biochemistry and Molecular Biology Laboratory, Health Science Department, University of Milan, San Paolo Hospital, 20142 Milan, Italy; (A.M.); (R.G.)
- Correspondence:
| |
Collapse
|
45
|
Guo Q, Wang J, Weng Q. The diverse role of optineurin in pathogenesis of disease. Biochem Pharmacol 2020; 180:114157. [PMID: 32687832 DOI: 10.1016/j.bcp.2020.114157] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/11/2020] [Accepted: 07/14/2020] [Indexed: 02/07/2023]
Abstract
Optineurin is a widely expressed protein that possesses multiple functions. Growing evidence suggests that mutation or dysregulation of optineurin can cause several neurodegenerative diseases, including amyotrophic lateral sclerosis, primary open-angle glaucoma, and Huntington's disease, as well as inflammatory digestive disorders such as Crohn's disease. Optineurin engages in vesicular trafficking, receptor regulation, immune reactions, autophagy, and distinct signaling pathways including nuclear factor kappa beta, by which optineurin contributes to cellular death and related diseases, indicating its potential as a therapeutic target. In this review, we discuss the major functions and signaling pathways of optineurin. Furthermore, we illustrate the influence of optineurin mutation or dysregulation to region-specific pathogenesis as well as potential applications of optineurin in therapeutic strategies.
Collapse
Affiliation(s)
- Qingyi Guo
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jincheng Wang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Qinjie Weng
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
46
|
Yamashiro LH, Wilson SC, Morrison HM, Karalis V, Chung JYJ, Chen KJ, Bateup HS, Szpara ML, Lee AY, Cox JS, Vance RE. Interferon-independent STING signaling promotes resistance to HSV-1 in vivo. Nat Commun 2020; 11:3382. [PMID: 32636381 PMCID: PMC7341812 DOI: 10.1038/s41467-020-17156-x] [Citation(s) in RCA: 126] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Accepted: 06/10/2020] [Indexed: 12/16/2022] Open
Abstract
The Stimulator of Interferon Genes (STING) pathway initiates potent immune responses upon recognition of DNA. To initiate signaling, serine 365 (S365) in the C-terminal tail (CTT) of STING is phosphorylated, leading to induction of type I interferons (IFNs). Additionally, evolutionary conserved responses such as autophagy also occur downstream of STING, but their relative importance during in vivo infections remains unclear. Here we report that mice harboring a serine 365-to-alanine (S365A) mutation in STING are unexpectedly resistant to Herpes Simplex Virus (HSV)-1, despite lacking STING-induced type I IFN responses. By contrast, resistance to HSV-1 is abolished in mice lacking the STING CTT, suggesting that the STING CTT initiates protective responses against HSV-1, independently of type I IFNs. Interestingly, we find that STING-induced autophagy is a CTT- and TBK1-dependent but IRF3-independent process that is conserved in the STING S365A mice. Thus, interferon-independent functions of STING mediate STING-dependent antiviral responses in vivo.
Collapse
Affiliation(s)
- Lívia H Yamashiro
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Howard Hughes Medical Institute, University of California, Berkeley, CA, 94720, USA
| | - Stephen C Wilson
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Bristol Myers Squibb, 200 Cambridge Park Dr, Cambridge, MA, 02140, USA
| | - Huntly M Morrison
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Vasiliki Karalis
- Division of Neurobiology, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Jing-Yi J Chung
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Katherine J Chen
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
| | - Helen S Bateup
- Division of Neurobiology, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, CA, 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA, 94158, USA
| | - Moriah L Szpara
- Departments of Biology and Biochemistry & Molecular Biology, Center for Infectious Disease Dynamics, Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, PA, 16801, USA
| | - Angus Y Lee
- Cancer Research Laboratory, University of California, Berkeley, CA, 94720, USA
| | - Jeffery S Cox
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA
- Henry Wheeler Center for Emerging and Neglected Diseases, University of California, Berkeley, CA, 94720, USA
| | - Russell E Vance
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, University of California, Berkeley, CA, 94720, USA.
- Howard Hughes Medical Institute, University of California, Berkeley, CA, 94720, USA.
- Cancer Research Laboratory, University of California, Berkeley, CA, 94720, USA.
- Henry Wheeler Center for Emerging and Neglected Diseases, University of California, Berkeley, CA, 94720, USA.
| |
Collapse
|
47
|
Lv P, Li C, Wang M, Ren J, Zhang Y, Fu G. TANK-binding kinase 1 alleviates myocardial ischemia/reperfusion injury through regulating apoptotic pathway. Biochem Biophys Res Commun 2020; 528:574-579. [PMID: 32505355 DOI: 10.1016/j.bbrc.2020.05.143] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 05/20/2020] [Indexed: 11/25/2022]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury, a complicated pathophysiological process, is regulated by lots of signaling pathways. Here in our present study, we identified TANK-binding kinase 1 (TBK1), an IKK-related serine/threonine kinase, as a protective regulator in MI/R injury. Our results indicated that TBK1 was decreased in MI/R injury in mice. However, after overexpressing TBK1 through an intramyocardial injection of TBK1 adenovirus, TBK1 overexpression improved cardiac function detected by echocardiography, decreased infarct size detected by Evans Blue and TTC staining, reduced cardiomyocyte apoptosis measured by TUNEL staining and alleviated disruption of mitochondria and cardiac muscle fibers detected by TEM in response to MI/R injury. Consistently, TBK1 overexpression ameliorated mitochondrial oxygen consumption rate (OCR) in neonatal rat cardiomyocytes (NRCMs) in response to hypoxia/reoxygenation (H/R) injury. Mechanistically, TBK1 overexpression upregulated Bcl-2 (an anti-apoptotic protein) but downregulated Bax (a pro-apoptotic protein) in vivo and in vitro. Collectively, our findings uncovered a pivotal function of TBK1 in MI/R injury through regulating the levels of apoptotic proteins for the first time, which might represent a promising target in treating MI/R patients in the future.
Collapse
Affiliation(s)
- Ping Lv
- Department of Cardiology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, 310020, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Congye Li
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Meihui Wang
- Department of Cardiology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, 310020, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China
| | - Jun Ren
- Department of Cardiology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, China; Center for Cardiovascular Research and Alternative Medicine, University of Wyoming College of Health Sciences, Laramie, WY, 82071, USA
| | - Yingmei Zhang
- Department of Cardiology, Zhongshan Hospital, Fudan University, 200032, Shanghai, China; Shanghai Institute of Cardiovascular Diseases, Shanghai, China.
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Medical School of Zhejiang University, 310020, Hangzhou, Zhejiang, China; Key Laboratory of Cardiovascular Intervention and Regenerative Medicine of Zhejiang Province, Hangzhou, Zhejiang, China.
| |
Collapse
|
48
|
Hoffpauir CT, Bell SL, West KO, Jing T, Wagner AR, Torres-Odio S, Cox JS, West AP, Li P, Patrick KL, Watson RO. TRIM14 Is a Key Regulator of the Type I IFN Response during Mycobacterium tuberculosis Infection. THE JOURNAL OF IMMUNOLOGY 2020; 205:153-167. [PMID: 32404352 DOI: 10.4049/jimmunol.1901511] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/20/2020] [Indexed: 01/05/2023]
Abstract
Tripartite motif-containing proteins (TRIMs) play a variety of recently described roles in innate immunity. Although many TRIMs regulate type I IFN expression following cytosolic nucleic acid sensing of viruses, their contribution to innate immune signaling and gene expression during bacterial infection remains largely unknown. Because Mycobacterium tuberculosis is an activator of cGAS-dependent cytosolic DNA sensing, we set out to investigate a role for TRIM proteins in regulating macrophage responses to M. tuberculosis In this study, we demonstrate that TRIM14, a noncanonical TRIM that lacks an E3 ubiquitin ligase RING domain, is a critical negative regulator of the type I IFN response in Mus musculus macrophages. We show that TRIM14 interacts with both cGAS and TBK1 and that macrophages lacking TRIM14 dramatically hyperinduce IFN stimulated gene (ISG) expression following M. tuberculosis infection, cytosolic nucleic acid transfection, and IFN-β treatment. Consistent with a defect in resolution of the type I IFN response, Trim14 knockout macrophages have more phospho-Ser754 STAT3 relative to phospho-Ser727 and fail to upregulate the STAT3 target Socs3, which is required to turn off IFNAR signaling. These data support a model whereby TRIM14 acts as a scaffold between TBK1 and STAT3 to promote phosphorylation of STAT3 at Ser727 and resolve ISG expression. Remarkably, Trim14 knockout macrophages hyperinduce expression of antimicrobial genes like Nos2 and are significantly better than control cells at limiting M. tuberculosis replication. Collectively, these data reveal an unappreciated role for TRIM14 in resolving type I IFN responses and controlling M. tuberculosis infection.
Collapse
Affiliation(s)
- Caitlyn T Hoffpauir
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Samantha L Bell
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Kelsi O West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Tao Jing
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77807; and
| | - Allison R Wagner
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Sylvia Torres-Odio
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Jeffery S Cox
- Department of Molecular and Cell Biology, University of California Berkeley, Berkeley, CA 94720
| | - A Phillip West
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Pingwei Li
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX 77807; and
| | - Kristin L Patrick
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807
| | - Robert O Watson
- Department of Microbial Pathogenesis and Immunology, Texas A&M Health Science Center, Bryan, TX 77807;
| |
Collapse
|
49
|
Wu S, Shen Y, Zhang S, Xiao Y, Shi S. Salmonella Interacts With Autophagy to Offense or Defense. Front Microbiol 2020; 11:721. [PMID: 32390979 PMCID: PMC7188831 DOI: 10.3389/fmicb.2020.00721] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Accepted: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
Autophagy is an important component of the innate immune system in mammals. Low levels of basic autophagy are sustained in normal cells, to help with the clearance of aging organelles and misfolded proteins, thus maintaining their structural and functional stability. However, when cells are faced with challenges, such as starvation or pathogenic infection, their level of autophagy increases significantly. Salmonella is a facultative intracellular pathogen, which imposes an economic burden on the poultry farming industry and human public health. Previous studies have shown that Salmonella can induce the autophagy of cells following invasion, which to a certain extent helps to protect the cells from bacterial colonization. This review summarizes the latest research in the field of Salmonella-induced autophagy, including: (i) the autophagy induction and escape mechanisms employed by Salmonella during the infection of host cells; (ii) the effect of autophagy on intracellular Salmonella; (iii) the important autophagy adaptors that recognize intracellular Salmonella in host cells; and (iv) the effect of autophagy-modulating drugs on Salmonella infection.
Collapse
Affiliation(s)
- Shu Wu
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China.,Institute of Effective Evaluation of Feed and Feed Additive (Poultry institute), Ministry of Agriculture, Yangzhou, China.,College of Animal Science and Technology, Hunan Agricultural University, Changsha, China
| | - Yiru Shen
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China.,Institute of Effective Evaluation of Feed and Feed Additive (Poultry institute), Ministry of Agriculture, Yangzhou, China
| | - Shan Zhang
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China.,Institute of Effective Evaluation of Feed and Feed Additive (Poultry institute), Ministry of Agriculture, Yangzhou, China
| | - Yunqi Xiao
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China.,Institute of Effective Evaluation of Feed and Feed Additive (Poultry institute), Ministry of Agriculture, Yangzhou, China
| | - Shourong Shi
- Department of Feed and Nutrition, Poultry Institute, Chinese Academy of Agricultural Sciences, Yangzhou, China.,Institute of Effective Evaluation of Feed and Feed Additive (Poultry institute), Ministry of Agriculture, Yangzhou, China.,Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, China
| |
Collapse
|
50
|
Tao Y, Yang Y, Zhou R, Gong T. Golgi Apparatus: An Emerging Platform for Innate Immunity. Trends Cell Biol 2020; 30:467-477. [PMID: 32413316 DOI: 10.1016/j.tcb.2020.02.008] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/24/2020] [Accepted: 02/24/2020] [Indexed: 12/23/2022]
Abstract
The Golgi apparatus serves as a receiving station where proteins from the endoplasmic reticulum (ER) are further processed before being sent to other cellular compartments. In addition to its well-appreciated roles in vesicular trafficking and protein/lipid secretion, recent studies have demonstrated that the Golgi acts as a signaling platform to facilitate multiple innate immune pathways. Moreover, the membranous networks that connect the Golgi with the ER, mitochondria, endosomes, and autophagosomes provide convenient access to innate immune signal transduction and subsequent effector responses. Here, we review the emerging knowledge about the roles of the Golgi in the initiation and activation of innate immune signaling. Moreover, microbial hijacking strategies that inhibit Golgi-associated innate immune responses will also be discussed.
Collapse
Affiliation(s)
- Ye Tao
- Department of Otolaryngology-Head and Neck Surgery, the First Affiliated Hospital of Anhui Medical University, Hefei 230022, China; Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230027, China
| | - Yanqing Yang
- Department of Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Bengbu 233004, China
| | - Rongbin Zhou
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230027, China.
| | - Tao Gong
- Hefei National Laboratory for Physical Sciences at Microscale, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Science, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230027, China.
| |
Collapse
|