1,263 results on '"tbk1"'
Search Results
2. Unveiling the physiological impact of ESCRT-dependent autophagosome closure by targeting the VPS37A ubiquitin E2 variant-like domain
- Author
-
Hamamoto, Kouta, Liang, Xinwen, Ito, Ayako, Lanza, Matthew, Bui, Van, Zhang, Jiawen, Opozda, David M., Hattori, Tatsuya, Chen, Longgui, Haddock, David, Imamura, Fumiaki, Wang, Hong-Gang, and Takahashi, Yoshinori
- Published
- 2024
- Full Text
- View/download PDF
3. Schisandrin C enhances cGAS-STING pathway activation and inhibits HBV replication
- Author
-
Zhao, Jia, Xu, Guang, Hou, Xiaorong, Mu, Wenqing, Yang, Huijie, Shi, Wei, Wen, Jincai, Liu, Tingting, Wu, Zhixin, Bai, Jun, Zhang, Ping, Wang, Zhongxia, Xiao, Xiaohe, Zou, Wenjun, Bai, Zhaofang, and Zhan, Xiaoyan
- Published
- 2023
- Full Text
- View/download PDF
4. Mycobacterial peptidyl prolyl isomerase A activates STING‐TBK1‐IRF3 signaling to promote IFNβ release in macrophages.
- Author
-
Sharma, Arun Kumar, Mal, Soumya, Sahu, Sanjaya Kumar, Bagchi, Shreya, Majumder, Debayan, Chakravorty, Debangana, Saha, Sudipto, Kundu, Manikuntala, and Basu, Joyoti
- Abstract
Peptidyl prolyl isomerases (PPIases) are well‐conserved protein‐folding enzymes that moonlight as regulators of bacterial virulence. Peptidyl prolyl isomerase A, PPiA (Rv0009) is a secretory protein of Mycobacterium tuberculosis that possesses sequence and structural similarity to eukaryotic cyclophilins. In this study, we validated the interaction of PPiA with stimulator of interferon genes (STING) using both, Escherichia coli‐based and mammalian in vitro expression systems. In vitro pull‐down assays confirmed that the cytosolic domain of STING interacts with PPiA, and moreover, we found that PPiA could induce dimerization of STING in macrophages. In silico docking analyses suggested that the PXXP (PDP) motif of PPiA is crucial for interaction with STING, and concordantly, mutations in the PDP domain (PPiA MUT‐II) abrogated this interaction, as well as the ability of PPiA to facilitate STING dimerization. In agreement with these observations, fluorescence microscopy demonstrated that STING and wild‐type PPiA, but not PPiA MUT‐II, could colocalize when expressed in HEK293 cells. Highlighting the importance of the PDP domain further, PPiA, but not PPiA MUT‐II could activate Tank binding kinase 1 (TBK1)‐interferon regulatory factor 3 (IRF3) signaling to promote the release of interferon‐beta (IFNβ). PPiA, but not PPiA MUT‐II expressed in Mycobacterium smegmatis induced IFNβ release and facilitated bacterial survival in macrophages in a STING‐dependent manner. The PPiA‐induced release of IFNβ was c‐GAS independent. We conclude that PPiA is a previously undescribed mycobacterial regulator of STING‐dependent type I interferon production from macrophages. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
5. TBK1 is involved in M‐CSF‐induced macrophage polarization through mediating the IRF5/IRF4 axis.
- Author
-
Li, Yuanyuan, Ji, Le, Liu, Chang, Li, Juanjuan, Wen, Di, Li, Zhongyao, Yu, Lishuang, Guo, Moran, Zhang, Shaoran, Duan, Weisong, Yi, Le, Bi, Yue, Bu, Hui, Li, Chunyan, and Liu, Yakun
- Subjects
- *
MYELOID cells , *TRANSCRIPTION factors , *NATURAL immunity , *GLIOBLASTOMA multiforme , *MACROPHAGE activation , *MACROPHAGE colony-stimulating factor - Abstract
TANK binding kinase 1 (TBK1) is an important kinase that is involved in innate immunity and tumor development. Macrophage colony‐stimulating factor (M‐CSF) regulates the differentiation and function of macrophages towards the immunosuppressive M2 phenotype in the glioblastoma multiforme microenvironment. The role of TBK1 in macrophages, especially in regulating macrophage polarization in response to M‐CSF stimulation, remains unclear. Here, we found high TBK1 expression in human glioma‐infiltrating myeloid cells and that phosphorylated TBK1 was highly expressed in M‐CSF‐stimulated macrophages but not in granulocyte‐macrophage CSF‐induced macrophages (granulocyte‐macrophage‐CSF is involved in the polarization of M1 macrophages). Conditional deletion of TBK1 in myeloid cells induced M‐CSF‐stimulated bone marrow‐derived macrophages to exhibit a proinflammatory M1‐like phenotype with increased protein expression of CD86, interleukin‐1β and tumor necrosis factor‐α, as well as decreased expression of arginase 1. Mechanistically, TBK1 deletion or inhibition by amlexanox or GSK8612 reduced the expression of the transcription factor interferon‐regulatory factor (IRF)4 and increased the level of IRF5 activation in macrophages stimulated with M‐CSF, leading to an M1‐like profile with highly proinflammatory factors. IRF5 deletion reversed the effect of TBK1 inhibition on M‐CSF‐mediated macrophage polarization. Our findings suggest that TBK1 contributes to the regulation of macrophage polarization in response to M‐CSF stimulation partly through the IRF5/IRF4 axis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Porcine reproductive and respiratory syndrome virus degrades TANK-binding kinase 1 via chaperon-mediated autophagy to suppress type I interferon production and facilitate viral proliferation.
- Author
-
Zhao, Shuang-shuang, Qian, Qisheng, Wang, Yao, Qiao, Songlin, and Li, Rui
- Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has led to significant economic losses in the global swine industry. Type I interferon (IFN-I) plays a crucial role in the host's resistance to PRRSV infection. Despite extensive research showing that PRRSV employs multiple strategies to antagonise IFN-I induction, the underlying mechanisms remain to be fully elucidated. In this study, we have discovered that PRRSV inhibits the production of IFN-I by degrading TANK-binding kinase 1 (TBK1) through chaperon-mediated autophagy (CMA). From a mechanistic standpoint, PRRSV nonstructural protein 2 (Nsp2) increases the interaction between the heat shock protein member 8 (HSPA8) and TBK1. This interaction leads to the translocation of TBK1 into lysosomes for degradation, mediated by lysosomal-associated membrane protein 2A (LAMP2A). As a result, the downstream activation of IFN regulatory factor 3 (IRF3) and the production of IFN-I are hindered. Together, these results reveal a new mechanism by which PRRSV suppresses host innate immunity and contribute to the development of new antiviral strategies against the virus. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. TBK1-Zyxin signaling controls tumor-associated macrophage recruitment to mitigate antitumor immunity.
- Author
-
Zhou, Ruyuan, Wang, Mengqiu, Li, Xiao, Liu, Yutong, Yao, Yihan, Wang, Ailian, Chen, Chen, Zhang, Qian, Wu, Qirou, Zhang, Qi, Neculai, Dante, Xia, Bing, Shao, Jian-Zhong, Feng, Xin-Hua, Liang, Tingbo, Zou, Jian, Wang, Xiaojian, and Xu, Pinglong
- Subjects
- *
CELL migration , *FOCAL adhesions , *CELL motility , *MACROPHAGES , *IMMUNITY - Abstract
Mechanical control is fundamental for cellular localization within a tissue, including for tumor-associated macrophages (TAMs). While the innate immune sensing pathways cGAS-STING and RLR-MAVS impact the pathogenesis and therapeutics of malignant diseases, their effects on cell residency and motility remain incompletely understood. Here, we uncovered that TBK1 kinase, activated by cGAS-STING or RLR-MAVS signaling in macrophages, directly phosphorylates and mobilizes Zyxin, a key regulator of actin dynamics. Under pathological conditions and in STING or MAVS signalosomes, TBK1-mediated Zyxin phosphorylation at S143 facilitates rapid recruitment of phospho-Zyxin to focal adhesions, leading to subsequent F-actin reorganization and reduced macrophage migration. Intratumoral STING-TBK1-Zyxin signaling was evident in TAMs and critical in antitumor immunity. Furthermore, myeloid-specific or global disruption of this signaling decreased the population of CD11b+ F4/80+ TAMs and promoted PD-1-mediated antitumor immunotherapy. Thus, our findings identify a new biological function of innate immune sensing pathways by regulating macrophage tissue localization, thus providing insights into context-dependent mitigation of antitumor immunity. Synopsis: Although the role of innate immune sensing pathways is well-studied in the pathogenesis of malignancies, whether they influence immune cell migration is not known. This study shows that innate immune signaling via cGAS-STING or RLR-MAVS regulates tumor-associated macrophage tissue residency through activation of the TBK1-Zyxin axis. STING signalosomes recruit and directly phosphorylate Zyxin, thus promoting its localization to focal adhesions. TBK1-Zyxin-mediated chemo-mechanical signaling enhances macrophage adhesion in response to cGAS-STING or RLR-MAVS signaling. STING-TBK1-Zyxin signaling activation in mouse intratumoral tumor-associated macrophages detains TAMs in the tumor and reduces antitumor immunity responses. Myeloid-specific or global inhibition of cGAS-STING-TBK1-Zyxin signaling facilitates antitumor immunotherapy. Innate immune sensing pathways activate the TBK1 kinase for phosphorylation of the actin dynamics regulator Zyxin to trap macrophages in the tumor. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. TRIM27 Promotes Endothelial Progenitor Cell Apoptosis in Patients with In-Stent Restenosis by Ubiquitinating TBK1.
- Author
-
Liu, Bo, Wang, Huai, Xie, Wenhao, and Gong, Ting
- Abstract
Approximately 2–10% in-stent restenosis (ISR) may occur following percutaneous coronary intervention (PCI) despite the use of modern drug-eluting stents (DES); thus, our study aimed to explore the effects of tripartite motif-containing (TRIM) 27 on ISR and the underlying mechanism. For this purpose, a total of 42 patients undergoing coronary angiography who had prior coronary angiography with DES implantation were recruited. Endothelial progenitor cells (EPCs) markers (defined as CD34 and vascular endothelial growth factoreceptor-2 (VEGFR-2)) in peripheral blood were measured to asses the circulating EPC level. The TRIM family-related gene expressions were detected by reverse transcription-quantitative polymerase chain reaction. Results suggested that ISR patients had reduced CD34
+ VEGFR-2+ and increased apoptosis rate of EPCs, along with upregulated TRIM27 and TRIM37 and downregulated TRIM28. TRIM27 promoted and TBK1 inhibited the apoptosis rate of EPCs. Mechanically, TRIM27 interacted with TBK1 to ubiquitinate TBK1 in in vitro study. In summary, TRIM27 promoted the progression of ISR in patients after PCI by ubiquitinating TBK1, which might provide novel ideas for the clinical treatment of ISR. [ABSTRACT FROM AUTHOR]- Published
- 2024
- Full Text
- View/download PDF
9. Porcine reproductive and respiratory syndrome virus degrades TANK-binding kinase 1 via chaperon-mediated autophagy to suppress type I interferon production and facilitate viral proliferation
- Author
-
Shuang-shuang Zhao, Qisheng Qian, Yao Wang, Songlin Qiao, and Rui Li
- Subjects
PRRSV ,Nsp2 ,TBK1 ,IFN-I ,CMA ,Veterinary medicine ,SF600-1100 - Abstract
Abstract Porcine reproductive and respiratory syndrome virus (PRRSV) has led to significant economic losses in the global swine industry. Type I interferon (IFN-I) plays a crucial role in the host’s resistance to PRRSV infection. Despite extensive research showing that PRRSV employs multiple strategies to antagonise IFN-I induction, the underlying mechanisms remain to be fully elucidated. In this study, we have discovered that PRRSV inhibits the production of IFN-I by degrading TANK-binding kinase 1 (TBK1) through chaperon-mediated autophagy (CMA). From a mechanistic standpoint, PRRSV nonstructural protein 2 (Nsp2) increases the interaction between the heat shock protein member 8 (HSPA8) and TBK1. This interaction leads to the translocation of TBK1 into lysosomes for degradation, mediated by lysosomal-associated membrane protein 2A (LAMP2A). As a result, the downstream activation of IFN regulatory factor 3 (IRF3) and the production of IFN-I are hindered. Together, these results reveal a new mechanism by which PRRSV suppresses host innate immunity and contribute to the development of new antiviral strategies against the virus.
- Published
- 2024
- Full Text
- View/download PDF
10. STRAP upregulates antiviral innate immunity against PRV by targeting TBK1
- Author
-
Wenfeng He, Hongtao Chang, Chen Li, Chenlong Wang, Longxi Li, Guoqing Yang, Jing Chen, and Huimin Liu
- Subjects
STRAP ,TBK1 ,Pseudorabies virus ,Antiviral immunity ,Type I interferon ,Infectious and parasitic diseases ,RC109-216 - Abstract
Abstract Serine/threonine kinase receptor-associated protein (STRAP) serves as a scaffold protein and is engaged in a variety of cellular activities, although its importance in antiviral innate immunity is unknown. We discovered that STRAP works as an interferon (IFN)-inducible positive regulator, facilitating type I IFN signaling during pseudorabies virus infection. Mechanistically, STRAP interacts with TBK1 to activate type I IFN signaling. Both the CT and WD40 7 − 6 domains contribute to the function of STRAP. Furthermore, TBK1 competes with PRV-UL50 for binding to STRAP, and STRAP impedes the degradation of TBK1 mediated by PRV-UL50, thereby increasing the interaction between STRAP and TBK1. Overall, these findings reveal a previously unrecognized role for STRAP in innate antiviral immune responses during PRV infection. STRAP could be a potential therapeutic target for viral infectious diseases.
- Published
- 2024
- Full Text
- View/download PDF
11. Functional Involvement of TANK‐Binding Kinase 1 in the MyD88‐Dependent NF‐κB Pathway Through Syk.
- Author
-
Kim, Han Gyung, Kim, Ji Hye, Yu, Tao, Cho, Jae Youl, and Brzozowski, Tomasz
- Subjects
- *
IMMUNOREGULATION , *PROTEIN-tyrosine kinases , *MUTANT proteins , *POLYMERASE chain reaction , *GENETIC transcription - Abstract
Inflammation is a vital immune defense mechanism regulated by Toll‐like receptors (TLRs) and the nuclear factor‐kappa B (NF‐κB) pathway. TANK‐binding kinase 1 (TBK1) is central to immunity and inflammation and influences antiviral responses and cellular processes. However, the precise role of TBK1 in modulating the NF‐κB pathway through interactions with other proteins, such as spleen tyrosine kinase (Syk), remains poorly understood. As dysregulation of TBK1 and NF‐κB can lead to a variety of diseases, they are important therapeutic targets. In this work, inflammatory processes involving the TBK1‐Syk‐NF‐κB pathway were elucidated using lipopolysaccharide (LPS)‐induced macrophages; human embryonic kidney 293 (HEK293) cells overexpressing MyD88, TBK1, and Syk proteins and their mutants; and real‐time polymerase chain reaction (PCR), immunoblotting analyses, and kinase assays. TBK1 was activated in LPS‐, poly I:C‐, and Pam3CSK‐stimulated macrophages. Transcript levels of TNF, NOS2, and IL1B were increased in cells overexpressing TBK1 but not in cells overexpressing TBK1 K38A. The transcription of TNF, NOS2, and IL1B and NF‐κB luciferase activity were inhibited by silencing TBK1 in LPS‐stimulated RAW264.7 cells and MyD88‐transfected HEK293 cells. Syk was the key mediator of the TBK1‐dependent NF‐κB pathway and bound directly to the coiled coil domain of TBK1, which was necessary to activate Syk and the Syk‐p85 pathway. This research advances the understanding of the role of TBK1 in NF‐κB signaling, emphasizing Syk as a key mediator. The interaction between TBK1 and Syk has potential for precise immune modulation that can be applied to treat immune‐related disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. A TBK1-independent primordial function of STING in lysosomal biogenesis.
- Author
-
Lv, Bo, Dion, William A., Yang, Haoxiang, Xun, Jinrui, Kim, Do-Hyung, Zhu, Bokai, and Tan, Jay Xiaojun
- Subjects
- *
TRANSCRIPTION factors , *CELL survival , *ISOPRENYLATION , *CELLULAR immunity , *LYSOSOMES , *INTERFERON receptors - Abstract
Stimulator of interferon genes (STING) is activated in many pathophysiological conditions, leading to TBK1-dependent interferon production in higher organisms. However, primordial functions of STING independent of TBK1 are poorly understood. Here, through proteomics and bioinformatics approaches, we identify lysosomal biogenesis as an unexpected function of STING. Transcription factor EB (TFEB), an evolutionarily conserved regulator of lysosomal biogenesis and host defense, is activated by STING from multiple species, including humans, mice, and frogs. STING-mediated TFEB activation is independent of TBK1, but it requires STING trafficking and its conserved proton channel. GABARAP lipidation, stimulated by the channel of STING, is key for STING-dependent TFEB activation. STING stimulates global upregulation of TFEB-target genes, mediating lysosomal biogenesis and autophagy. TFEB supports cell survival during chronic sterile STING activation, a common condition in aging and age-related diseases. These results reveal a primordial function of STING in the biogenesis of lysosomes, essential organelles in immunity and cellular stress resistance. [Display omitted] • TFEB and TFE3 are conserved effectors of STING, predating type I interferon signaling • Noncanonical GABARAP lipidation, triggered by the channel of STING, activates TFEB/TFE3 • STING stimulates cytoprotective upregulation of lysosomal biogenesis and autophagy • The cGAS-STING-TFEB axis is a primordial form of the antiviral DNA-sensing pathway Lv et al. discover TFEB/TFE3 as evolutionarily ancient effectors of the cGAS-STING DNA-sensing innate immune pathway. TFEB/TFE3 activation via the channel of STING integrates cytosolic DNA sensing, a general cellular stress-sensing mechanism, with lysosomal biogenesis and autophagy upregulation, conserved and general strategies for host defense and stress clearance. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. Regulation of Type I Interferon and Autophagy in Immunity against Mycobacterium Tuberculosis: Role of CGAS and STING1.
- Author
-
Malik, Asrar Ahmad, Shariq, Mohd, Sheikh, Javaid Ahmad, Fayaz, Haleema, Srivastava, Gauri, Thakuri, Deeksha, Ahuja, Yashika, Ali, Saquib, Alam, Anwar, Ehtesham, Nasreen Z., and Hasnain, Seyed E.
- Subjects
TYPE I interferons ,MYCOBACTERIUM tuberculosis ,KNOCKOUT mice ,LUNG infections ,DENDRITIC cells ,NEUTROPHILS - Abstract
Mycobacterium tuberculosis (M. tb) is a significant intracellular pathogen responsible for numerous infectious disease‐related deaths worldwide. It uses ESX‐1 T7SS to damage phagosomes and to enter the cytosol of host cells after phagocytosis. During infection, M. tb and host mitochondria release dsDNA, which activates the CGAS‐STING1 pathway. This pathway leads to the production of type I interferons and proinflammatory cytokines and activates autophagy, which targets and degrades bacteria within autophagosomes. However, the role of type I IFNs in immunity against M. tb is controversial. While previous research has suggested a protective role, recent findings from cgas‐sting1 knockout mouse studies have contradicted this. Additionally, a study using knockout mice and non‐human primate models uncovered a new mechanism by which neutrophils recruited to lung infections form neutrophil extracellular traps. Activating plasmacytoid dendritic cells causes them to produce type I IFNs, which interfere with the function of interstitial macrophages and increase the likelihood of tuberculosis. Notably, M. tb uses its virulence proteins to disrupt the CGAS‐STING1 signaling pathway leading to enhanced pathogenesis. Investigating the CGAS‐STING1 pathway can help develop new ways to fight tuberculosis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
14. Lysosomal TBK1 responds to amino acid availability to relieve Rab7-dependent mTORC1 inhibition.
- Author
-
Talaia, Gabriel, Bentley-DeSousa, Amanda, and Ferguson, Shawn M
- Subjects
- *
AMYOTROPHIC lateral sclerosis , *LYSOSOMES , *CELL metabolism , *FRONTOTEMPORAL dementia , *AMINO acids , *CELL physiology - 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. Synopsis: In addition to their degradative functions, lysosomes play an important role in coordinating cellular responses to changes in nutrient availability. This study reveals that TBK1 is recruited to lysosomes when amino acids are abundant where it phosphorylates Rab7 and thus relieves Rab7-dependent suppression of mTORC1 signaling from lysosomes. TBK1 is recruited to lysosomes when amino acids are abundant. Rab7 (serine 72) is a substrate of TBK1 at lysosomes. Rab7 suppresses mTORC1 activity and this is relieved by TBK1-dependent phosphorylation of Rab7 on serine 72. The ALS-FTD associated E696K TBK1 mutant accumulates at lysosomes resulting in increased Rab7 phosphorylation and mTORC1 activity. TBK1 promotes mTORC1 activation by amino acids at the lysosome surface. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
15. Cardiomyocyte-specific Tbk1 deletion aggravated chronic doxorubicin cardiotoxicity via inhibition of mitophagy.
- Author
-
Yu, Wenjun, Deng, Dawei, Li, Yang, Ding, Kehan, Qian, Qiaofeng, Shi, Hongjie, Luo, Qiujie, Cai, Jie, and Liu, Jinping
- Subjects
- *
CARDIOTOXICITY , *DILATED cardiomyopathy , *MEMBRANE potential , *MITOCHONDRIAL membranes , *DOXORUBICIN - Abstract
Doxorubicin (Dox) use is limited by Dox-induced cardiotoxicity. TANK-blinding kinase 1 (TBK1) is an important kinase involved in the regulation of mitophagy, but the role of TBK1 in cardiomyocytes in chronic Dox-induced cardiomyopathy remains unclear. Cardiomyocyte-specific Tbk1 knockout (Tbk1 CKO) mice received Dox (6 mg/kg, injected intraperitoneally) once a week for 4 times, and cardiac assessment was performed 4 weeks after the final Dox injection. Adenoviruses encoding Tbk1 or containing shRNA targeting Tbk1 , or a TBK1 phosphorylation inhibitor were used for overexpression or knockdown of Tbk1 , or inhibit phosphorylation of TBK1 in isolated primary cardiomyocytes. Our results revealed that moderate Dox challenge decreased TBK1 phosphorylation (with no effect on TBK1 protein levels), resulting in compromised myocardial function, obvious mortality and overt interstitial fibrosis, and the effects were accentuated by Tbk1 deletion. Dox provoked mitochondrial membrane potential collapse and oxidative stress, the effects of which were exacerbated and mitigated by Tbk1 knockdown, specific inhibition of phosphorylation and overexpression, respectively. However, Tbk1 (Ser172A) overexpression did not alleviate these effects. Further scrutiny revealed that TBK1 exerted protective effects on mitochondria via SQSTM1/P62-mediated mitophagy. Tbk1 overexpression mediated cardioprotective effects on Dox-induced cardiotoxicity were cancelled off by Sqstm1/P62 knockdown. Moreover, TBK1-mitophagy-mitochondria cascade was confirmed in heart tissues from dilated cardiomyopathy patients. Taken together, our findings denoted a pivotal role of TBK1 in Dox-induced mitochondrial injury and cardiotoxicity possibly through its phosphorylation and SQSTM1/P62-mediated mitophagy. [Display omitted] • Cardiomyocyte-specific Tbk1 deletion aggravated chronic Doxorubicin-induced cardiotoxicity. • Specific inhibition of TBK1(Ser172) phosphorylation exacerbated the cardiomyocyte toxicity of Doxorubicin. • Overexpression of Tbk1 could mitigate the cardiotoxicity of Doxorubicin, while overexpression of Tbk1 (Ser172A) could not. • The TBK1-P62-mitophagy cascade may play a crucial role in the pathogenesis of Doxorubicin-induced cardiotoxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
16. STRAP upregulates antiviral innate immunity against PRV by targeting TBK1.
- Author
-
He, Wenfeng, Chang, Hongtao, Li, Chen, Wang, Chenlong, Li, Longxi, Yang, Guoqing, Chen, Jing, and Liu, Huimin
- Subjects
- *
SCAFFOLD proteins , *AUJESZKY'S disease virus , *VIRUS diseases , *NATURAL immunity , *COMMUNICABLE diseases , *TYPE I interferons - Abstract
Serine/threonine kinase receptor-associated protein (STRAP) serves as a scaffold protein and is engaged in a variety of cellular activities, although its importance in antiviral innate immunity is unknown. We discovered that STRAP works as an interferon (IFN)-inducible positive regulator, facilitating type I IFN signaling during pseudorabies virus infection. Mechanistically, STRAP interacts with TBK1 to activate type I IFN signaling. Both the CT and WD40 7 − 6 domains contribute to the function of STRAP. Furthermore, TBK1 competes with PRV-UL50 for binding to STRAP, and STRAP impedes the degradation of TBK1 mediated by PRV-UL50, thereby increasing the interaction between STRAP and TBK1. Overall, these findings reveal a previously unrecognized role for STRAP in innate antiviral immune responses during PRV infection. STRAP could be a potential therapeutic target for viral infectious diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
17. Post-Translational Variants of Major Proteins in Amyotrophic Lateral Sclerosis Provide New Insights into the Pathophysiology of the Disease.
- Author
-
Bedja-Iacona, Léa, Richard, Elodie, Marouillat, Sylviane, Brulard, Céline, Alouane, Tarek, Beltran, Stéphane, Andres, Christian R., Blasco, Hélène, Corcia, Philippe, Veyrat-Durebex, Charlotte, and Vourc'h, Patrick
- Subjects
- *
AMYOTROPHIC lateral sclerosis , *UBIQUITINATION , *PATHOLOGICAL physiology , *PHOSPHORYLATION , *ACETYLATION , *POST-translational modification - Abstract
Post-translational modifications (PTMs) affecting proteins during or after their synthesis play a crucial role in their localization and function. The modification of these PTMs under pathophysiological conditions, i.e., their appearance, disappearance, or variation in quantity caused by a pathological environment or a mutation, corresponds to post-translational variants (PTVs). These PTVs can be directly or indirectly involved in the pathophysiology of diseases. Here, we present the PTMs and PTVs of four major amyotrophic lateral sclerosis (ALS) proteins, SOD1, TDP-43, FUS, and TBK1. These modifications involve acetylation, phosphorylation, methylation, ubiquitination, SUMOylation, and enzymatic cleavage. We list the PTM positions known to be mutated in ALS patients and discuss the roles of PTVs in the pathophysiological processes of ALS. In-depth knowledge of the PTMs and PTVs of ALS proteins is needed to better understand their role in the disease. We believe it is also crucial for developing new therapies that may be more effective in ALS. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
18. TBK1 is paradoxical in tumor development: a focus on the pathway mediating IFN-I expression.
- Author
-
Banglu Wang, Fan Zhang, Xiaoyu Wu, and Mei Ji
- Subjects
TYPE I interferons ,CANCER invasiveness ,NATURAL immunity ,IMMUNE response ,CELLULAR signal transduction - Abstract
TANK-binding kinase 1 (TBK1) is a member of the IKK family and plays a crucial role in the activation of non-canonical NF-kB signaling and type I interferon responses. The aberrant activation of TBK1 contributes to the proliferation and survival of various types of tumor cells, particularly in specific mutational or tumorous contexts. Inhibitors targeting TBK1 are under development and application in both in vivo and in vitro settings, yet their clinical efficacy remains limited. Numerous literatures have shown that TBK1 can exhibit both tumor promoting and tumor inhibiting effects. TBK1 acts as a pivotal node within the innate immune pathway, mediating anti-tumor immunity through the activation of innate immune responses. Facilitating interferon-I (IFN-I) production represents a critical mechanism through which TBK1 bridges these processes. IFN has been shown to exert both beneficial and detrimental effects on tumor progression. Hence, the paradoxical role of TBK1 in tumor development may necessitate acknowledgment in light of its downstream IFNI signaling cascade. In this paper, we review the signaling pathways mediated by TBK1 in various tumor contexts and summarize the dual roles of TBK1 and the TBK1-IFN pathways in both promoting and inhibiting tumor progression. Additionally, we highlight the significance of the TBK1-IFN pathway in clinical therapy, particularly in the context of immune response. We anticipate further advancements in the development of TBK1 inhibitors as part of novel cancer treatment strategies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
19. Control of innate immunity and lipid biosynthesis in neurodegeneration.
- Author
-
Scoles, Daniel R. and Pulst, Stefan M.
- Subjects
NATURAL immunity ,NUCLEIC acids ,DOUBLE-stranded RNA ,NEURODEGENERATION ,ENDOPLASMIC reticulum - Abstract
The cGAS-STING innate immunity pathway and the SREBP-activated cholesterol and fatty acid synthesis pathway are abnormally co-regulated in neurodegenerative disease. Activation of STING signaling occurs at the endoplasmic reticulum (ER) membrane with STING anchored by INSIG1 along with SREBP and the sterol-bound SREBP cleavage activating protein (SCAP) when sterols are in abundance. When sterols are low, the INSIG-dependent STING pathway is inactivated and the SREBPSCAP complex is translocated to the Golgi where SREBP is cleaved and translocated to the nucleus to transactivate genes for cholesterol and fatty acid synthesis. Thus, there is inverse activation of STING vs. SREBP: when innate immunity is active, pathways for cholesterol and fatty acid synthesis are suppressed, and vice versa. The STING pathway is stimulated by foreign viral cytoplasmic nucleic acids interacting with the cyclic GMP-AMP synthase (cGAS) DNA sensor or RIG-I and MDA5 dsRNA sensors, but with neurodegeneration innate immunity is also activated by self-DNAs and double-stranded RNAs that accumulate with neuronal death. Downstream, activated STING recruits TBK1 and stimulates the transactivation of interferon stimulated genes and the autophagy pathway, which are both protective. However, chronic activation of innate immunity contributes to microglia activation, neuroinflammation and autophagy failure leading to neurodegeneration. STING is also a proton channel that when activated stimulates proton exit from STING vesicles leading to cell death. Here we review the salient features of the innate immunity and cholesterol and fatty acid synthesis pathways, observations of abnormal STING and SREBP signaling in neurodegenerative disease, and relevant therapeutic approaches. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
20. TBK1 pharmacological inhibition mitigates osteoarthritis through attenuating inflammation and cellular senescence in chondrocytes
- Author
-
Rui Lu, Yunkun Qu, Zhenggang Wang, Zhiyi He, Shimeng Xu, Peng Cheng, Zhengtao Lv, Hongbo You, Fengjing Guo, Anmin Chen, Jiaming Zhang, and Shuang Liang
- Subjects
BX795 ,Chondrocyte ,Inflammation ,Osteoarthritis ,Senescence ,TBK1 ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
Objectives: TANK-binding kinase 1 (TBK1) is pivotal in autoimmune and inflammatory diseases, yet its role in osteoarthritis (OA) remains elusive. This study sought to elucidate the effect of the TBK1 inhibitor BX795 on OA and to delineate the underlying mechanism by which it mitigates OA. Methods: Interleukin-1 Beta (IL-1β) was utilized to simulate inflammatory responses and extracellular matrix degradation in vitro. In vivo, OA was induced in 8-week-old mice through destabilization of the medial meniscus surgery. The impact of BX795 on OA was evaluated using histological analysis, X-ray, micro-CT, and the von Frey test. Additionally, Western blot, RT-qPCR, and immunofluorescence assays were conducted to investigate the underlying mechanisms of BX795. Results: Phosphorylated TBK1 (P-TBK1) levels were found to be elevated in OA knee cartilage of both human and mice. Furthermore, intra-articular injection of BX795 ameliorated cartilage degeneration and alleviated OA-associated pain. BX795 also counteracted the suppression of anabolic processes and the augmentation of catabolic activity, inflammation, and senescence observed in the OA mice. In vitro studies revealed that BX795 reduced P-TBK1 levels and reversed the effects of anabolism inhibition, catabolism promotion, and senescence induction triggered by IL-1β. Mechanistically, BX795 inhibited the IL-1β-induced activation of the cGAS–STING and TLR3–TRIF signaling pathways in chondrocytes. Conclusions: Pharmacological inhibition of TBK1 with BX795 protects articular cartilage by inhibiting the activation of the cGAS–STING and TLR3–TRIF signaling pathways. This action attenuates inflammatory responses and cellular senescence, positioning BX795 as a promising therapeutic candidate for OA treatment. The translational potential of this article: This study furnishes experimental evidence and offers a potential mechanistic explanation supporting the efficacy of BX795 as a promising candidate for OA treatment.
- Published
- 2024
- Full Text
- View/download PDF
21. Targeting TANK-binding kinase 1 attenuates painful diabetic neuropathy via inhibiting microglia pyroptosis
- Author
-
Qinming Liao, Yimei Yang, Yilu Li, Jun Zhang, Keke Fan, Yihao Guo, Jun Chen, Yinhao Chen, Pian Zhu, Lijin Huang, and Zhongjie Liu
- Subjects
Painful diabetic neuropathy ,TBK1 ,Pyroptosis ,Microglia ,Amlexanox ,Medicine ,Cytology ,QH573-671 - Abstract
Abstract Background Painful diabetic neuropathy (PDN) is closely linked to inflammation, which has been demonstrated to be associated with pyroptosis. Emerging evidence has implicated TANK-binding kinase 1 (TBK1) in various inflammatory diseases. However, it remains unknown whether activated TBK1 causes hyperalgesia via pyroptosis. Methods PDN mice model of type 1 or type 2 diabetic was induced by C57BL/6J or BKS-DB mice with Lepr gene mutation. For type 2 diabetes PDN model, TBK1-siRNA, Caspase-1 inhibitor Ac-YVAD-cmk or TBK1 inhibitor amlexanox (AMX) were delivered by intrathecal injection or intragastric administration. The pain threshold and plantar skin blood perfusion were evaluated through animal experiments. The assessments of spinal cord, dorsal root ganglion, sciatic nerve, plantar skin and serum included western blotting, immunofluorescence, ELISA, and transmission electron microscopy. Results In the PDN mouse model, we found that TBK1 was significantly activated in the spinal dorsal horn (SDH) and mainly located in microglia, and intrathecal injection of chemically modified TBK1-siRNA could improve hyperalgesia. Herein, we described the mechanism that TBK1 could activate the noncanonical nuclear factor κB (NF-κB) pathway, mediate the activation of NLRP3 inflammasome, trigger microglia pyroptosis, and ultimately induce PDN, which could be reversed following TBK1-siRNA injection. We also found that systemic administration of AMX, a TBK1 inhibitor, could effectively improve peripheral nerve injury. These results revealed the key role of TBK1 in PDN and that TBK1 inhibitor AMX could be a potential strategy for treating PDN. Conclusions Our findings revealed a novel causal role of TBK1 in pathogenesis of PDN, which raises the possibility of applying amlexanox to selectively target TBK1 as a potential therapeutic strategy for PDN.
- Published
- 2024
- Full Text
- View/download PDF
22. MARCH1 negatively regulates TBK1-mTOR signaling pathway by ubiquitinating TBK1
- Author
-
Xiao Li, Kai Cheng, Meng-Di Shang, Yong Yang, Bin Hu, Xi Wang, Xiao-Dan Wei, Yan-Chun Han, Xiao-Gang Zhang, Meng-Hua Dong, Zhen-Lin Yang, and Jiu-Qiang Wang
- Subjects
MARCH1 ,STING ,TBK1 ,mTOR ,Growth factors ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background TBK1 positively regulates the growth factor-mediated mTOR signaling pathway by phosphorylating mTOR. However, it remains unclear how the TBK1-mTOR signaling pathway is regulated. Considering that STING not only interacts with TBK1 but also with MARCH1, we speculated that MARCH1 might regulate the mTOR signaling pathway by targeting TBK1. The aim of this study was to determine whether MARCH1 regulates the mTOR signaling pathway by targeting TBK1. Methods The co-immunoprecipitation (Co-IP) assay was used to verify the interaction between MARCH1 with STING or TBK1. The ubiquitination of STING or TBK1 was analyzed using denatured co-immunoprecipitation. The level of proteins detected in the co-immunoprecipitation or denatured co-immunoprecipitation samples were determined by Western blotting. Stable knocked-down cells were constructed by infecting lentivirus bearing the related shRNA sequences. Scratch wound healing and clonogenic cell survival assays were used to detect the migration and proliferation of breast cancer cells. Results We showed that MARCH1 played an important role in growth factor-induced the TBK1- mTOR signaling pathway. MARCH1 overexpression attenuated the growth factor-induced activation of mTOR signaling pathway, whereas its deficiency resulted in the opposite effect. Mechanistically, MARCH1 interacted with and promoted the K63-linked ubiquitination of TBK1. This ubiquitination of TBK1 then attenuated its interaction with mTOR, thereby inhibiting the growth factor-induced mTOR signaling pathway. Importantly, faster proliferation induced by MARCH1 deficiency was weakened by mTOR, STING, or TBK1 inhibition. Conclusion MARCH1 suppressed growth factors mediated the mTOR signaling pathway by targeting the STING-TBK1-mTOR axis.
- Published
- 2024
- Full Text
- View/download PDF
23. Genetic and clinical landscape of Chinese frontotemporal dementia: dominance of TBK1 and OPTN mutations
- Author
-
Haitian Nan, Yeon-Jeong Kim, Min Chu, Dan Li, Jieying Li, Deming Jiang, Yiming Wu, Toshihisa Ohtsuka, and Liyong Wu
- Subjects
Frontotemporal dementia ,Genetic spectrum ,TBK1 ,OPTN ,Autophagy ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 ,Neurology. Diseases of the nervous system ,RC346-429 - Abstract
Abstract Background Our study aims to evaluate the genetic and phenotypic spectrum of Frontotemporal dementia (FTD) gene variant carriers in Chinese populations, investigate mutation frequencies, and assess the functional properties of TBK1 and OPTN variants. Methods Clinically diagnosed FTD patients underwent genetic analysis through exome sequencing, repeat-primed polymerase chain reaction, and Sanger sequencing. TBK1 and OPTN variants were biologically characterized in vitro using immunofluorescence, immunoprecipitation, and immunoblotting analysis. The frequencies of genes implicated in FTD in China were analyzed through a literature review and meta-analysis. Results Of the 261 Chinese FTD patients, 61 (23.4%) carried potential causative variants in FTD-related genes, including MAPT (n = 17), TBK1 (n = 7), OPTN (n = 6), GRN (n = 6), ANXA11 (n = 4), CHMP2B (n = 3), C9orf72 GGGGCC repeats (n = 2), CYLD (n = 2), PRNP (n = 2), SQSTM1 (n = 2), TARDBP (n = 2), VCP (n = 1), CCNF (n = 1), CHCHD10 (n = 1), SIGMAR1 (n = 1), CHCHD2 (n = 1), FUS (n = 1), TMEM106B (n = 1), and UBQLN2 (n = 1). 29 variants can be considered novel, including the MAPT p.D54N, p.E342K, p.R221P, p.T263I, TBK1 p.E696G, p.I37T, p.E232Q, p.S398F, p.T78A, p.Q150P, p.W259fs, OPTN p.R144G, p.F475V, GRN p.V473fs, p.C307fs, p.R101fs, CHMP2B p.K6N, p.R186Q, ANXA11 p.Q155*, CYLD p.T157I, SQSTM1 p.S403A, UBQLN2 p.P509H, CCNF p.S160N, CHCHD10 p.A8T, SIGMAR1 p.S117L, CHCHD2 p.P53fs, FUS p.S235G & p.S236G, and TMEM106B p.L144V variants. Patients with TBK1 and OPTN variants presented with heterogeneous clinical phenotypes. Functional analysis demonstrated that TBK1 I37T and E232Q mutants showed decreased autophosphorylation, and the OPTN phosphorylation was reduced by the TBK1 I37T mutant. The OPTN-TBK1 complex formation was enhanced by the TBK1 E696G mutant, while OPTN R144G and F475V mutants exhibited reduced recruitment to autophagosomes compared to the wild-type. The overall frequency of TBK1 and OPTN in Chinese FTD patients was 2.0% and 0.3%, respectively. Conclusions Our study demonstrates the extensive genetic and phenotypic heterogeneity of Chinese FTD patients. TBK1 mutations are the second most frequent cause of clinical FTD after MAPT in the Chinese.
- Published
- 2024
- Full Text
- View/download PDF
24. MARCH1 negatively regulates TBK1-mTOR signaling pathway by ubiquitinating TBK1.
- Author
-
Li, Xiao, Cheng, Kai, Shang, Meng-Di, Yang, Yong, Hu, Bin, Wang, Xi, Wei, Xiao-Dan, Han, Yan-Chun, Zhang, Xiao-Gang, Dong, Meng-Hua, Yang, Zhen-Lin, and Wang, Jiu-Qiang
- Subjects
- *
CELLULAR signal transduction , *CANCER cell proliferation , *GROWTH factors - Abstract
Background: TBK1 positively regulates the growth factor-mediated mTOR signaling pathway by phosphorylating mTOR. However, it remains unclear how the TBK1-mTOR signaling pathway is regulated. Considering that STING not only interacts with TBK1 but also with MARCH1, we speculated that MARCH1 might regulate the mTOR signaling pathway by targeting TBK1. The aim of this study was to determine whether MARCH1 regulates the mTOR signaling pathway by targeting TBK1. Methods: The co-immunoprecipitation (Co-IP) assay was used to verify the interaction between MARCH1 with STING or TBK1. The ubiquitination of STING or TBK1 was analyzed using denatured co-immunoprecipitation. The level of proteins detected in the co-immunoprecipitation or denatured co-immunoprecipitation samples were determined by Western blotting. Stable knocked-down cells were constructed by infecting lentivirus bearing the related shRNA sequences. Scratch wound healing and clonogenic cell survival assays were used to detect the migration and proliferation of breast cancer cells. Results: We showed that MARCH1 played an important role in growth factor-induced the TBK1- mTOR signaling pathway. MARCH1 overexpression attenuated the growth factor-induced activation of mTOR signaling pathway, whereas its deficiency resulted in the opposite effect. Mechanistically, MARCH1 interacted with and promoted the K63-linked ubiquitination of TBK1. This ubiquitination of TBK1 then attenuated its interaction with mTOR, thereby inhibiting the growth factor-induced mTOR signaling pathway. Importantly, faster proliferation induced by MARCH1 deficiency was weakened by mTOR, STING, or TBK1 inhibition. Conclusion: MARCH1 suppressed growth factors mediated the mTOR signaling pathway by targeting the STING-TBK1-mTOR axis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
25. Targeting TANK-binding kinase 1 attenuates painful diabetic neuropathy via inhibiting microglia pyroptosis.
- Author
-
Liao, Qinming, Yang, Yimei, Li, Yilu, Zhang, Jun, Fan, Keke, Guo, Yihao, Chen, Jun, Chen, Yinhao, Zhu, Pian, Huang, Lijin, and Liu, Zhongjie
- Subjects
- *
DIABETIC neuropathies , *PYROPTOSIS , *MICROGLIA , *DORSAL root ganglia , *PERIPHERAL nerve injuries , *TYPE 2 diabetes - Abstract
Background: Painful diabetic neuropathy (PDN) is closely linked to inflammation, which has been demonstrated to be associated with pyroptosis. Emerging evidence has implicated TANK-binding kinase 1 (TBK1) in various inflammatory diseases. However, it remains unknown whether activated TBK1 causes hyperalgesia via pyroptosis. Methods: PDN mice model of type 1 or type 2 diabetic was induced by C57BL/6J or BKS-DB mice with Lepr gene mutation. For type 2 diabetes PDN model, TBK1-siRNA, Caspase-1 inhibitor Ac-YVAD-cmk or TBK1 inhibitor amlexanox (AMX) were delivered by intrathecal injection or intragastric administration. The pain threshold and plantar skin blood perfusion were evaluated through animal experiments. The assessments of spinal cord, dorsal root ganglion, sciatic nerve, plantar skin and serum included western blotting, immunofluorescence, ELISA, and transmission electron microscopy. Results: In the PDN mouse model, we found that TBK1 was significantly activated in the spinal dorsal horn (SDH) and mainly located in microglia, and intrathecal injection of chemically modified TBK1-siRNA could improve hyperalgesia. Herein, we described the mechanism that TBK1 could activate the noncanonical nuclear factor κB (NF-κB) pathway, mediate the activation of NLRP3 inflammasome, trigger microglia pyroptosis, and ultimately induce PDN, which could be reversed following TBK1-siRNA injection. We also found that systemic administration of AMX, a TBK1 inhibitor, could effectively improve peripheral nerve injury. These results revealed the key role of TBK1 in PDN and that TBK1 inhibitor AMX could be a potential strategy for treating PDN. Conclusions: Our findings revealed a novel causal role of TBK1 in pathogenesis of PDN, which raises the possibility of applying amlexanox to selectively target TBK1 as a potential therapeutic strategy for PDN. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
26. Regulation of Mitochondria-Derived Immune Activation by 'Antiviral' TRIM Proteins.
- Author
-
Oh, Seeun and Mandell, Michael A.
- Subjects
- *
TRIM proteins , *VIRUS diseases , *MITOCHONDRIA , *NATURAL immunity , *INTERFERONS - Abstract
Mitochondria are key orchestrators of antiviral responses that serve as platforms for the assembly and activation of innate immune-signaling complexes. In response to viral infection, mitochondria can be triggered to release immune-stimulatory molecules that can boost interferon production. These same molecules can be released by damaged mitochondria to induce pathogenic, antiviral-like immune responses in the absence of infection. This review explores how members of the tripartite motif-containing (TRIM) protein family, which are recognized for their roles in antiviral defense, regulate mitochondria-based innate immune activation. In antiviral defense, TRIMs are essential components of immune signal transduction pathways and function as directly acting viral restriction factors. TRIMs carry out conceptually similar activities when controlling immune activation related to mitochondria. First, they modulate immune-signaling pathways that can be activated by mitochondrial molecules. Second, they co-ordinate the direct removal of mitochondria and associated immune-activating factors through mitophagy. These insights broaden the scope of TRIM actions in innate immunity and may implicate TRIMs in diseases associated with mitochondria-derived inflammation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
27. Nonstructural Protein A238L of the African Swine Fever Virus (ASFV) Enhances Antiviral Immune Responses by Activating the TBK1-IRF3 Pathway.
- Author
-
Liu, Wei, Yang, Lanlan, Di, Chuanyuan, Sun, Jing, Liu, Penggang, and Liu, Huisheng
- Subjects
AFRICAN swine fever virus ,HUMAN herpesvirus 1 ,IMMUNE response ,TYPE I interferons ,DNA viruses ,SENDAI virus - Abstract
Simple Summary: A238L, a non-structural protein of the African swine fever virus (ASFV), inhibits the activation of NF-κB by suppressing the HAT activity of p300. Whether A238L also affects the transcriptional activity of IRF3 remains unexplored. Here we first confirmed the ability of A238L to suppress NF-κB-activity in L929 cells. In contrast, A238L did not inhibit but rather increased TBK1 and IRF3 phosphorylation and enhanced innate antiviral immunity in the absence or presence of poly d (A:T) or poly (I:C) stimulation, or herpes simplex virus type 1 (HSV-1) or Sendai virus (SeV) infection. This study reveals an unrecognized role for A238L in promoting antiviral immune responses by activating the TBK1-IRF3 pathway. African swine fever virus (ASFV) is a double-stranded DNA virus with an envelope. ASFV has almost the largest genome among all DNA viruses, and its mechanisms of immune evasion are complex. Better understanding of the molecular mechanisms of ASFV genes will improve vaccine design. A238L, a nonstructural protein of ASFV, inhibits NF-κB activation by suppressing the HAT activity of p300. Whether A238L also affects the transcriptional activity of IRF3 remains unexplored. Here we first confirmed the ability of A238L to suppress NF-κB-activity in L929 cells. A238L inhibits the expression of proinflammatory cytokine genes. In contrast, A238L increased the phosphorylation levels of TBK1 and IRF3 in three different cell lines. A238L increases the IRF3-driven promoter activity and induces IRF3 nuclear translocation. Furthermore, A238L enhanced innate antiviral immunity in the absence or presence of poly d (A:T) or poly (I:C) stimulation, or herpes simplex virus type 1 (HSV-1) or Sendai virus (SeV) infection. This study reveals a previously unrecognized role of A238L in promoting antiviral immune responses by TBK1-IRF3 pathway activation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
28. Molecular genetics of inherited normal tension glaucoma.
- Author
-
Yang Pan and Takeshi Iwata
- Subjects
- *
GLAUCOMA , *RETINAL ganglion cells , *INTRAOCULAR pressure , *MUTANT proteins , *VISUAL fields , *MOLECULAR genetics , *AUDITORY neuropathy - Abstract
Normal tension glaucoma (NTG) is a complex optic neuropathy characterized by progressive retinal ganglion cell death and glaucomatous visual field loss, despite normal intraocular pressure (IOP). This condition poses a unique clinical challenge due to the absence of elevated IOP, a major risk factor in typical glaucoma. Recent research indicates that up to 21% of NTG patients have a family history of glaucoma, suggesting a genetic predisposition. In this comprehensive review using PubMed studies from January 1990 to December 2023, our focus delves into the genetic basis of autosomal dominant NTG, the only known form of inheritance for glaucoma. Specifically exploring optineurin (OPTN), TANK binding kinase 1 (TBK1), methyltransferase-like 23 (METTL23), and myocilin (MYOC) mutations, we summarize their clinical manifestations, mutant protein behaviors, relevant animal models, and potential therapeutic pathways. This exploration aims to illuminate the intricate pathogenesis of NTG, unraveling the contribution of these genetic components to its complex development. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
29. TBK1 对 NLRC4 炎症小体的作用及其机制研究.
- Author
-
曾 强, 张在奎, 孙乃双, and 陈允梓
- Abstract
Objective: To investigate the mechanism by which TANK binding kinase 1 (TBK1) regulates the activation of nucleotide⁃ binding oligomerization domain ⁃like receptor 4 (NLRC4) inflammasome. Methods: Western blot was used to detect the activation of NLRC4 inflammasome and their downstream molecules cysteine aspartic acid-specific protease 1 (Caspase-1) and Gasdermin D (GSDMD) in immortalized bone marrow-derived macrophages (IBMDM) infected with Salmonella typhimurium (S.T) . A lactate dehydrogenase detection kit was used to detect the content of lactate dehydrogenase in the supernatant of cell culture medium. The interaction between TBK1 and NLRC4 and their specific interaction domain was determined through protein co ⁃immunoprecipitation experiments. Cellular immunofluorescence assay was used to determine the spatial localization of TBK1 and NLRC4. The GST pull⁃down experiment confirmed the direct interaction between TBK1 and NLRC4. The assembly of NLRC4 inflammasome was verified using apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization detection experiments. The S.T infected animal model of C57BL/6 mice was built and the survival of mice was observed. The bacterial load of lung tissues and peritoneal cavity-flushed fluid was analyzed through smear analysis. ELISA was used to detect the content of tunor necrosis factor (TNF)-α and interleukin (IL)⁃1β in peritoneal cavity ⁃flushed fluid and serum. Flow cytometry was used to detect the proportion of neutrophils in peritoneal cavity-flushed fluid. Results: In S.T infected IBMDM, inhibiting TBK1 led to a weakened activation of NLRC4 inflammasomes, decreased phosphorylation levels of NLRC4, and reduced cleavage of Caspase-1 and GSDMD. There was an interaction between TBK1 and NLRC4, and the N⁃terminal of TBK1 interaced with the NACHT domain of NLRC4. TBK1 and NLRC4 had spatial co⁃localization. TBK1 phosphorylated the NLRC4 Ser533 site. S.T animal model experiments showed that inhibiting TBK1 activity significantly improved the survival rate of mice, weakened the bacterial load in the peritoneal cavity-flushed fluid and lung tissues of mice, reduced the content of IL⁃1β and TNF⁃α in serum and peritoneal cavity-flushed fluid, and reduced the proportion of neutrophils in peritoneal cavity-flushed fluid. Conclusion: TBK1 interacts with NLRC4, phosphorylates the NLRC4 Ser533 site, and promotes the activation of NLRC4 inflammasome, which providing a theoretical basis and new potential targets for treating related diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
30. The Ca2+-dependent phosphatase calcineurin dephosphorylates TBK1 to suppress antiviral innate immunity.
- Author
-
Yang Qu, Siyuan Wang, Hui Jiang, Qingyi Wang, Ying Liao, Xusheng Qiu, Lei Tan, Cuiping Song, Chan Ding, Yingjie Sun, and Zengqi Yang
- Subjects
- *
NATURAL immunity , *TYPE I interferons , *CALCINEURIN , *INTERFERON receptors , *TUMOR necrosis factors , *LIFE cycles (Biology) , *NEWCASTLE disease virus - Abstract
Tumor necrosis factor receptor-associated factor family member-associated NF-κB activator-binding kinase 1 (TBK1) plays a key role in the induction of the type 1 interferon (IFN-I) response, which is an important component of innate antiviral defense. Viruses target calcium (Ca2+) signaling networks, which participate in the regulation of the viral life cycle, as well as mediate the host antiviral response. Although many studies have focused on the role of Ca2+ signaling in the regulation of IFN-I, the relationship between Ca2+ and TBK1 in different infection models requires further elucidation. Here, we examined the effects of the Newcastle disease virus (NDV)-induced increase in intracellular Ca2+ levels on the suppression of host antiviral responses. We demonstrated that intracellular Ca2+ increased significantly during NDV infection, leading to impaired IFN-I production and antiviral immunity through the activation of calcineurin (CaN). Depletion of Ca²+ was found to lead to a significant increase in virus-induced IFN-I production resulting in the inhibition of viral replication. Mechanistically, the accumulation of Ca2+ in response to viral infection increases the phosphatase activity of CaN, which in turn dephosphorylates and inactivates TBK1 in a Ca2+-dependent manner. Furthermore, the inhibition of CaN on viral replication was counteracted in TBK1 knockout cells. Together, our data demonstrate that NDV hijacks Ca2+ signaling networks to negatively regulate innate immunity via the CaN-TBK1 signaling axis. Thus, our findings not only identify the mechanism by which viruses exploit Ca2+ signaling to evade the host antiviral response but also, more importantly, highlight the potential role of Ca2+ homeostasis in the viral innate immune response. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
31. Mechanism of TBK1 activation in cancer cells
- Author
-
Lianxin Hu and Qing Zhang
- Subjects
TBK1 ,Ser172 phosphorylation ,Cancer ,Biology (General) ,QH301-705.5 ,Medicine (General) ,R5-920 - Abstract
TANK-binding kinase 1 (TBK1) is a serine/threonine kinase with well-established roles as a central player in innate immune signaling. Dysregulation of TBK1 activity has been implicated in a variety of pathophysiologic conditions, including cancer. Generally, TBK1 acts as an oncogene and increased TBK1 activity, indicated by increased phosphorylation at the Ser172 residue, can be observed in multiple human cancers. TBK1 can be activated either by autophosphorylation of Ser172 or transphosphorylation at this site by upstream kinases. Serving as a hub for integrating numerous extracellular and intracellular signals, TBK1 can be activated through multiple signaling pathways. However, the direct upstream kinase responsible for TBK1 activation remains elusive, which limits our comprehensive understanding of its activation mechanism and potential therapeutic application targeting TBK1-related signaling especially in cancer. In this review, we summarize the findings on mechanisms of TBK1 activation in cancer cells and recent discoveries that shed light on the direct upstream kinases promoting TBK1 activation.
- Published
- 2024
- Full Text
- View/download PDF
32. TBK1, a prioritized drug repurposing target for amyotrophic lateral sclerosis: evidence from druggable genome Mendelian randomization and pharmacological verification in vitro
- Author
-
Qing-Qing Duan, Han Wang, Wei-Ming Su, Xiao-Jing Gu, Xiao-Fei Shen, Zheng Jiang, Yan-Ling Ren, Bei Cao, Guo-Bo Li, Yi Wang, and Yong-Ping Chen
- Subjects
Amyotrophic lateral sclerosis ,Mendelian randomization ,Drug repurposing ,Druggable gene ,TBK1 ,Medicine - Abstract
Abstract Background There is a lack of effective therapeutic strategies for amyotrophic lateral sclerosis (ALS); therefore, drug repurposing might provide a rapid approach to meet the urgent need for treatment. Methods To identify therapeutic targets associated with ALS, we conducted Mendelian randomization (MR) analysis and colocalization analysis using cis-eQTL of druggable gene and ALS GWAS data collections to determine annotated druggable gene targets that exhibited significant associations with ALS. By subsequent repurposing drug discovery coupled with inclusion criteria selection, we identified several drug candidates corresponding to their druggable gene targets that have been genetically validated. The pharmacological assays were then conducted to further assess the efficacy of genetics-supported repurposed drugs for potential ALS therapy in various cellular models. Results Through MR analysis, we identified potential ALS druggable genes in the blood, including TBK1 [OR 1.30, 95%CI (1.19, 1.42)], TNFSF12 [OR 1.36, 95%CI (1.19, 1.56)], GPX3 [OR 1.28, 95%CI (1.15, 1.43)], TNFSF13 [OR 0.45, 95%CI (0.32, 0.64)], and CD68 [OR 0.38, 95%CI (0.24, 0.58)]. Additionally, we identified potential ALS druggable genes in the brain, including RESP18 [OR 1.11, 95%CI (1.07, 1.16)], GPX3 [OR 0.57, 95%CI (0.48, 0.68)], GDF9 [OR 0.77, 95%CI (0.67, 0.88)], and PTPRN [OR 0.17, 95%CI (0.08, 0.34)]. Among them, TBK1, TNFSF12, RESP18, and GPX3 were confirmed in further colocalization analysis. We identified five drugs with repurposing opportunities targeting TBK1, TNFSF12, and GPX3, namely fostamatinib (R788), amlexanox (AMX), BIIB-023, RG-7212, and glutathione as potential repurposing drugs. R788 and AMX were prioritized due to their genetic supports, safety profiles, and cost-effectiveness evaluation. Further pharmacological analysis revealed that R788 and AMX mitigated neuroinflammation in ALS cell models characterized by overly active cGAS/STING signaling that was induced by MSA-2 or ALS-related toxic proteins (TDP-43 and SOD1), through the inhibition of TBK1 phosphorylation. Conclusions Our MR analyses provided genetic evidence supporting TBK1, TNFSF12, RESP18, and GPX3 as druggable genes for ALS treatment. Among the drug candidates targeting the above genes with repurposing opportunities, FDA-approved drug-R788 and AMX served as effective TBK1 inhibitors. The subsequent pharmacological studies validated the potential of R788 and AMX for treating specific ALS subtypes through the inhibition of TBK1 phosphorylation.
- Published
- 2024
- Full Text
- View/download PDF
33. Novel role of macrophage TXNIP-mediated CYLD-NRF2-OASL1 axis in stress-induced liver inflammation and cell death.
- Author
-
Zhan, Yongqiang, Xu, Dongwei, Tian, Yizhu, Qu, Xiaoye, Sheng, Mingwei, Lin, Yuanbang, Ke, Michael, Jiang, Longfeng, Xia, Qiang, Kaldas, Fady M, Farmer, Douglas G, and Ke, Bibo
- Subjects
ALT ,alanine aminotransferase ,APAF1 ,apoptotic peptidase activating factor 1 ,ASK1 ,apoptosis signal-regulating kinase 1 ,AST ,aspartate aminotransferase ,Apoptosis ,BMM ,bone marrow-derived macrophage ,CXCL-10 ,C-X-C motif chemokine ligand 10 ,CYLD ,cyclindromatosis ,ChIP ,chromatin immunoprecipitation ,DAMP ,damage-associated molecular pattern ,DUB ,deubiquitinating enzyme ,ER ,endoplasmic reticulum ,ES ,embryonic stem ,G3BP1 ,G3BP1 ,Ras GTPase-activating protein-binding protein 1 ,GCLC ,glutamate-cysteine ligase catalytic subunit ,GCLM ,glutamate-cysteine ligase regulatory subunit ,IHC ,immunohistochemistry ,INF-β ,interferon-β ,IR ,ischaemia/reperfusion ,IRF3 ,IRF3 ,interferon regulatory factor 3 ,IRF7 ,IFN-regulating transcription factor 7 ,IRI ,ischaemia/reperfusion injury ,Innate immunity ,KO ,knockout ,LPS ,lipopolysaccharide ,Liver inflammation ,Lyz2 ,Lysozyme 2 ,MCP-1 ,monocyte chemoattractant protein 1 ,NOX2 ,NADPH oxidase 2 ,NOX4 ,NADPH oxidase 4 ,NQO1 ,NAD(P)H quinone dehydrogenase 1 ,NRF2 ,nuclear factor (erythroid-derived 2)-like 2 ,NS ,non-specific ,Necroptosis ,OASL1 ,2′ ,5′oligoadenylate synthetase-like 1 ,PAMP ,pathogen-derived molecular pattern ,RIPK3 ,receptor-interacting serine/threonine-protein kinase 3 ,ROS ,reactive oxygen species ,STING ,STING ,stimulator of interferon genes ,TBK1 ,TANK-binding kinase 1 ,TLR4 ,Toll-like receptor 4 ,TNF-α ,tumour necrosis factor-alpha ,TRX ,thioredoxin ,TSS ,transcription start sites ,TXNIP ,thioredoxin-interacting protein ,TXNIPFL/FL ,floxed TXNIP ,TXNIPM-KO ,myeloid-specific TXNIP KO ,UTR ,untranslated region ,sALT ,serum ALT ,sAST ,serum AST ,siRNA ,small interfering RNA ,Liver Disease ,Digestive Diseases ,Chronic Liver Disease and Cirrhosis ,Aetiology ,2.1 Biological and endogenous factors ,Good Health and Well Being - Abstract
Background & aimsThe stimulator of interferon genes (STING)/TANK-binding kinase 1 (TBK1) pathway is vital in mediating innate immune and inflammatory responses during oxidative/endoplasmic reticulum (ER) stress. However, it remains unknown whether macrophage thioredoxin-interacting protein (TXNIP) may regulate TBK1 function and cell death pathways during oxidative/ER stress.MethodsA mouse model of hepatic ischaemia/reperfusion injury (IRI), the primary hepatocytes, and bone marrow-derived macrophages were used in the myeloid-specific TXNIP knockout (TXNIPM-KO) and TXNIP-proficient (TXNIPFL/FL) mice.ResultsThe TXNIPM-KO mice were resistant to ischaemia/reperfusion (IR) stress-induced liver damage with reduced serum alanine aminotransferase (ALT)/aspartate aminotransferase (AST) levels, macrophage/neutrophil infiltration, and pro-inflammatory mediators compared with the TXNIPFL/FL controls. IR stress increased TXNIP, p-STING, and p-TBK1 expression in ischaemic livers. However, TXNIPM-KO inhibited STING, TBK1, interferon regulatory factor 3 (IRF3), and NF-κB activation with interferon-β (IFN-β) expression. Interestingly, TXNIPM-KO augmented nuclear factor (erythroid-derived 2)-like 2 (NRF2) activity, increased antioxidant gene expression, and reduced macrophage reactive oxygen species (ROS) production and hepatic apoptosis/necroptosis in IR-stressed livers. Mechanistically, macrophage TXNIP deficiency promoted cylindromatosis (CYLD), which colocalised and interacted with NADPH oxidase 4 (NOX4) to enhance NRF2 activity by deubiquitinating NOX4. Disruption of macrophage NRF2 or its target gene 2',5' oligoadenylate synthetase-like 1 (OASL1) enhanced Ras GTPase-activating protein-binding protein 1 (G3BP1) and TBK1-mediated inflammatory response. Notably, macrophage OASL1 deficiency induced hepatocyte apoptotic peptidase activating factor 1 (APAF1), cytochrome c, and caspase-9 activation, leading to increased caspase-3-initiated apoptosis and receptor-interacting serine/threonine-protein kinase 3 (RIPK3)-mediated necroptosis.ConclusionsMacrophage TXNIP deficiency enhances CYLD activity and activates the NRF2-OASL1 signalling, controlling IR stress-induced liver injury. The target gene OASL1 regulated by NRF2 is crucial for modulating STING-mediated TBK1 activation and Apaf1/cytochrome c/caspase-9-triggered apoptotic/necroptotic cell death pathway. Our findings underscore a novel role of macrophage TXNIP-mediated CYLD-NRF2-OASL1 axis in stress-induced liver inflammation and cell death, implying the potential therapeutic targets in liver inflammatory diseases.Lay summaryLiver inflammation and injury induced by ischaemia and reperfusion (the absence of blood flow to the liver tissue followed by the resupply of blood) is a significant cause of hepatic dysfunction and failure following liver transplantation, resection, and haemorrhagic shock. Herein, we uncover an underlying mechanism that contributes to liver inflammation and cell death in this setting and could be a therapeutic target in stress-induced liver inflammatory injury.
- Published
- 2022
34. Azamollugin, a mollugin derivative, has inhibitory activity on MyD88- and TRIF-dependent pathways
- Author
-
Nakajima, Yuki, Nishino, Hitomi, Takahashi, Kazunori, Nugroho, Alfarius Eko, Hirasawa, Yusuke, Kaneda, Toshio, and Morita, Hiroshi
- Published
- 2024
- Full Text
- View/download PDF
35. Genetic and clinical landscape of Chinese frontotemporal dementia: dominance of TBK1 and OPTN mutations
- Author
-
Nan, Haitian, Kim, Yeon-Jeong, Chu, Min, Li, Dan, Li, Jieying, Jiang, Deming, Wu, Yiming, Ohtsuka, Toshihisa, and Wu, Liyong
- Published
- 2024
- Full Text
- View/download PDF
36. TBK1, a prioritized drug repurposing target for amyotrophic lateral sclerosis: evidence from druggable genome Mendelian randomization and pharmacological verification in vitro
- Author
-
Duan, Qing-Qing, Wang, Han, Su, Wei-Ming, Gu, Xiao-Jing, Shen, Xiao-Fei, Jiang, Zheng, Ren, Yan-Ling, Cao, Bei, Li, Guo-Bo, Wang, Yi, and Chen, Yong-Ping
- Published
- 2024
- Full Text
- View/download PDF
37. TANK Binding Kinase 1 Promotes BACH1 Degradation through Both Phosphorylation-Dependent and -Independent Mechanisms without Relying on Heme and FBXO22.
- Author
-
Liu, Liang, Matsumoto, Mitsuyo, Watanabe-Matsui, Miki, Nakagawa, Tadashi, Nagasawa, Yuko, Pang, Jingyao, Callens, Bert K. K., Muto, Akihiko, Ochiai, Kyoko, Takekawa, Hirotaka, Alam, Mahabub, Nishizawa, Hironari, Shirouzu, Mikako, Shima, Hiroki, Nakayama, Keiko, and Igarashi, Kazuhiko
- Subjects
- *
HEME , *IRON metabolism , *REACTIVE oxygen species , *PROTEIN kinases , *B cells - Abstract
BTB and CNC homology 1 (BACH1) represses the expression of genes involved in the metabolism of iron, heme and reactive oxygen species. While BACH1 is rapidly degraded when it is bound to heme, it remains unclear how BACH1 degradation is regulated under other conditions. We found that FBXO22, a ubiquitin ligase previously reported to promote BACH1 degradation, polyubiquitinated BACH1 only in the presence of heme in a highly purified reconstitution assay. In parallel to this regulatory mechanism, TANK binding kinase 1 (TBK1), a protein kinase that activates innate immune response and regulates iron metabolism via ferritinophagy, was found to promote BACH1 degradation when overexpressed in 293T cells. While TBK1 phosphorylated BACH1 at multiple serine and threonine residues, BACH1 degradation was observed with not only the wild-type TBK1 but also catalytically impaired TBK1. The BACH1 degradation in response to catalytically impaired TBK1 was not dependent on FBXO22 but involved both autophagy-lysosome and ubiquitin-proteasome pathways judging from its suppression by using inhibitors of lysosome and proteasome. Chemical inhibition of TBK1 in hepatoma Hepa1 cells showed that TBK1 was not required for the heme-induced BACH1 degradation. Its inhibition in Namalwa B lymphoma cells increased endogenous BACH1 protein. These results suggest that TBK1 promotes BACH1 degradation in parallel to the FBXO22- and heme-dependent pathway, placing BACH1 as a downstream effector of TBK1 in iron metabolism or innate immune response. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
38. Phosphorylation of AGO2 by TBK1 Promotes the Formation of Oncogenic miRISC in NSCLC.
- Author
-
Zhao, Xian, Cao, Yingting, Lu, Runhui, Zhou, Zihan, Huang, Caihu, Li, Lian, Huang, Jiayi, Chen, Ran, Wang, Yanli, Huang, Jian, Cheng, Jinke, Zheng, Junke, Fu, Yujie, and Yu, Jianxiu
- Subjects
- *
NON-small-cell lung carcinoma , *PHOSPHORYLATION - Abstract
Non‐small‐cell lung cancer (NSCLC) is a highly lethal tumor that often develops resistance to targeted therapy. It is shown that Tank‐binding kinase 1 (TBK1) phosphorylates AGO2 at S417 (pS417‐AGO2), which promotes NSCLC progression by increasing the formation of microRNA‐induced silencing complex (miRISC). High levels of pS417‐AGO2 in clinical NSCLC specimens are positively associated with poor prognosis. Interestingly, the treatment with EGFR inhibitor Gefitinib can significantly induce pS417‐AGO2, thereby increasing the formation and activity of oncogenic miRISC, which may contribute to NSCLC resistance to Gefitinib. Based on these, two therapeutic strategies is developed. One is jointly to antagonize multiple oncogenic miRNAs highly expressed in NSCLC and use TBK1 inhibitor Amlexanox reducing the formation of oncogenic miRISC. Another approach is to combine Gefitinib with Amlexanox to inhibit the progression of Gefitinib‐resistant NSCLC. This findings reveal a novel mechanism of oncogenic miRISC regulation by TBK1‐mediated pS417‐AGO2 and suggest potential therapeutic approaches for NSCLC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
39. ZO-1 Regulates Hippo-Independent YAP Activity and Cell Proliferation via a GEF-H1- and TBK1-Regulated Signalling Network.
- Author
-
Haas, Alexis J., Karakus, Mert, Zihni, Ceniz, Balda, Maria S., and Matter, Karl
- Subjects
- *
YAP signaling proteins , *CELL proliferation , *FOCAL adhesions , *TIGHT junctions , *CELL junctions , *CELL adhesion - Abstract
Tight junctions are a barrier-forming cell–cell adhesion complex and have been proposed to regulate cell proliferation. However, the underlying mechanisms are not well understood. Here, we used cells deficient in the junction scaffold ZO-1 alone or together with its paralog ZO-2, which disrupts the junctional barrier. We found that ZO-1 knockout increased cell proliferation, induced loss of cell density-dependent proliferation control, and promoted apoptosis and necrosis. These phenotypes were enhanced by double ZO-1/ZO-2 knockout. Increased proliferation was dependent on two transcriptional regulators: YAP and ZONAB. ZO-1 knockout stimulated YAP nuclear translocation and activity without changes in Hippo-dependent phosphorylation. Knockout promoted TANK-binding kinase 1 (TBK1) activation and increased expression of the RhoA activator GEF-H1. Knockdown of ZO-3, another paralog interacting with ZO1, was sufficient to induce GEF-H1 expression and YAP activity. GEF-H1, TBK1, and mechanotransduction at focal adhesions were found to cooperate to activate YAP/TEAD in ZO-1-deficient cells. Thus, ZO-1 controled cell proliferation and Hippo-independent YAP activity by activating a GEF-H1- and TBK1-regulated mechanosensitive signalling network. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
40. TRIM28 facilitates type I interferon activation by targeting TBK1.
- Author
-
Fang Hua, Nass Jr, Tim, and Parvatiyar, Kislay
- Subjects
TYPE I interferons ,PATTERN perception receptors ,TRANSCRIPTION factors ,VIRUS diseases ,UBIQUITIN ,POST-translational modification - Abstract
Type I interferons play a fundamental role in innate host defense against viral infections by eliciting the induction of an antiviral gene program that serves to inhibit viral replication. Activation of type I interferon is regulated by the IRF3 transcription factor, which undergoes phosphorylation-dependent activation by the upstream kinase, TBK1, during viral infection. However, the mechanisms by which TBK1 achieves activation to support signaling to IRF3 remain incompletely understood. Here we identified the E3 ubiquitin ligase, tripartite motif containing 28 (TRIM28), as a positive regulator of type I interferon activation by facilitating TBK1 signaling. Genetic deletion of TRIM28 via CRISPR-Cas9 editing resulted in impaired type I interferon activation upon both RNA and DNA virus challenge, corresponding with increased susceptibility to virus infections in TRIM28 knockout cells. Mechanistically, TRIM28 interacted with TBK1 and mediated the assembly of K63-linked ubiquitin chains onto TBK1, a post-translational modification shown to augment TBK1 signal transmission events. TRIM28 knockout cells further displayed defective TBK1 phosphorylation and complex assembly with IRF3, resulting in impaired IRF3 phosphorylation. Altogether, our data demonstrate TBK1 to be a novel substrate for TRIM28 and identify TRIM28 as an essential regulatory factor in controlling innate antiviral immune responses. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
41. ASFV pA151R negatively regulates type I IFN production via degrading E3 ligase TRAF6.
- Author
-
You Li, Li Huang, Hui Li, Yingqi Zhu, Zilong Yu, Xiaojie Zheng, Changjiang Weng, and Wen-hai Feng
- Subjects
UBIQUITIN ligases ,AFRICAN swine fever virus ,TYPE I interferons ,AFRICAN swine fever ,ALVEOLAR macrophages - Abstract
African swine fever (ASF) caused by African swine fever virus (ASFV) is a highly mortal and hemorrhagic infectious disease in pigs. Previous studies have indicated that ASFV modulates interferon (IFN) production. In this study, we demonstrated that ASFV pA151R negatively regulated type I IFN production. Ectopic expression of pA151R dramatically inhibited K63-linked polyubiquitination and Ser172 phosphorylation of TANK-binding kinase 1 (TBK1). Mechanically, we demonstrated that E3 ligase TNF receptor-associated factor 6 (TRAF6) participated in the ubiquitination of TBK1 in cGAS-STING signaling pathway. We showed that pA151R interacted with TRAF6 and degraded it through apoptosis pathway, leading to the disruption of TBK1 and TRAF6 interaction. Moreover, we clarified that the amino acids H102, C109, C132, and C135 in pA151R were crucial for pA151R to inhibit type I interferon production. In addition, we verified that overexpression of pA151R facilitated DNA virus Herpes simplex virus 1 (HSV-1) replication by inhibiting IFN-b production. Importantly, knockdown of pA151R inhibited ASFV replication and enhanced IFN-b production in porcine alveolar macrophages (PAMs). Our findings will help understand how ASFV escapes host antiviral immune responses and develop effective ASFV vaccines. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
42. STING upregulation mediates ferroptosis and inflammatory response in lupus nephritis by upregulating TBK1 and activating NF-κB signal pathway.
- Author
-
Chen, Jinshu, Chen, Pihou, Song, Yijin, Wei, Jiaxin, Wu, Fan, Sun, Jing, and Xu, Zhiquan
- Abstract
Accumulated evidence implicates lipid peroxidation as a key mechanism contributing to the pathogenesis of lupus nephritis (LN). Ferroptosis is a specialized form of cell death induced by loss or deficient activity of the glutathione peroxidase 4 (GPX4) and decreased clearance of polyunsaturated fatty acid hydroperoxides. STING production may lead to the occurrence of intracellular lipid peroxidation, ultimately triggering ferroptosis, but it has not been clarified whether STING can aggravate LN via ferroptosis. The adjacent normal kidney tissues from renal cell carcinoma and biopsied kidney tissue samples from LN patients were used for research, and the expression of STING protein in kidney tissue was detected by immunohistochemistry and RT-qPCR. MRL/lpr mice, a model of LN, were used to detect STING expression in kidney tissue. STING expression in the kidney tissue of MRL/lpr mice was knocked down by sh-STING-AAV, and then levels of 4-HNE, MDA, ROS, iron ion, blood urea nitrogen and serum creatinine, IL-6, IL-1β, and TNF-α, and the protein expression of STING, TBK1, NF-κB, GPX4, ACSL4, and SLC7A11 were subsequently examined. STING was elevated in the kidney tissue of LN patients and MRL/lpr mice. Compared with the MRL/lpr group, liproxstatin-1 or ferrostatin-1 treatment alleviated ferroptosis-related indicators 4-HNE, MDA, ROS, iron ion release, and GPX4 and SLC7A1 expression, whereas the treatment enhanced ACSL4 expression. STING interference observably decreased 4-HNE, ROS, MDA, iron ion, STING, and ACSL4 levels, and increased GPX4 and SLC7A11 expression in MRL/lpr mice kidney tissues. Besides, inhibition of STING reduced kidney tissue damage and inflammatory cell infiltration in MRL/lpr mice, and levels of serum creatinine, blood urea nitrogen, serum anti-double-stranded DNA antibody, inflammatory factors IL-6, IL-1β, and TNF-α, as well as phosphorylation of NF-κB were all significantly decreased in MRL/lpr mice. TBK1 overexpression reversed the impact of STING inhibition on ferroptosis and inflammatory response. STING contributed to ferroptosis and inflammatory response by activating the TBK1/NF-κB pathway, suggesting that STING may be a potent therapeutic target in LN. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
43. Lumpy skin disease virus ORF127 protein suppresses type I interferon responses by inhibiting K63‐linked ubiquitination of tank binding kinase 1.
- Author
-
Liang, Zhengji, Wang, Shasha, Yao, Kaishen, Ren, Shanhui, Cheng, Pengyuan, Qu, Min, Ma, Xiaoqin, Gao, Xin, Yin, Xiangping, Wang, Xiangwei, and Sun, Yuefeng
- Abstract
Lumpy skin disease (LSD) is a severe animal infectious disease caused by lumpy skin disease virus (LSDV), inducing extensive nodules on the cattle mucosa or the scarfskin. LSDV genome encodes multiple proteins to evade host innate immune response. However, the underlying molecular mechanisms are poorly understood. In this study, we found that LSDV could suppress the expression of IFN‐β and interferon‐stimulated genes (ISGs) in MDBK cells during the early stage of infection. Subsequently, an unbiased screen was performed to screen the LSDV genes with inhibitory effects on the type I interferon (IFN‐I) production. ORF127 protein was identified as one of the strongest inhibitory effectors on the expression of IFN‐β and ISGs, meanwhile, the 1–43 aa of N‐terminal of ORF127 played a vital role in suppressing the expression of IFN‐β. Overexpression of ORF127 could significantly promote LSDV replication through inhibiting the production of IFN‐β and ISGs in MDBK cells. Mechanism study showed that ORF127 specifically interacted with TBK1 and decreased the K63‐linked polyubiquitination of TBK1 which suppressed the phosphorylation of TBK1 and ultimately decreased the production of IFN‐β. In addition, truncation mutation analysis indicated that the 1–43 aa of N‐terminal of ORF127 protein was the key structural domain for its interaction with TBK1. In short, these results validated that ORF127 played a negative role in regulating IFN‐β expression through cGAS‐STING signaling pathway. Taken together, this study clarified the molecular mechanism of ORF127 gene antagonizing IFN‐I‐mediated antiviral, which will helpfully provide new strategies for the treatment and prevention of LSD. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
44. Porcine Sapovirus Protease Controls the Innate Immune Response and Targets TBK1.
- Author
-
Georgana, Iliana, Hosmillo, Myra, Jahun, Aminu S., Emmott, Edward, Sorgeloos, Frederic, Cho, Kyoung-Oh, and Goodfellow, Ian G.
- Subjects
- *
IMMUNE response , *CELL lines , *NOROVIRUSES , *GASTROENTERITIS , *JEJUNUM - Abstract
Human sapoviruses (HuSaVs) and noroviruses are considered the leading cause of acute gastroenteritis worldwide. While extensive research has focused on noroviruses, our understanding of sapoviruses (SaVs) and their interactions with the host's immune response remains limited. HuSaVs have been challenging to propagate in vitro, making the porcine sapovirus (PSaV) Cowden strain a valuable model for studying SaV pathogenesis. In this study we show, for the first time, that PSaV Cowden strain has mechanisms to evade the host's innate immune response. The virus 3C-like protease (NS6) inhibits type I IFN production by targeting TBK1. Catalytically active NS6, both during ectopic expression and during PSaV infection, targets TBK1 which is then led for rapid degradation by the proteasome. Moreover, deletion of TBK1 from porcine cells led to an increase in PSaV titres, emphasizing its role in regulating PSaV infection. Additionally, we successfully established PSaV infection in IPEC-J2 cells, an enterocytic cell line originating from the jejunum of a neonatal piglet. Overall, this study provides novel insights into PSaV evasion strategies, opening the way for future investigations into SaV–host interactions, and enabling the use of a new cell line model for PSaV research. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
45. Suppression of Interferon Response and Antiviral Strategies of Bunyaviruses
- Author
-
Yingying He, Min Shen, Xiaohe Wang, Anqi Yin, Bingyan Liu, Jie Zhu, and Zhenhua Zhang
- Subjects
bunyavirus ,interferon ,NSs ,TBK1 ,STAT ,targeted therapy ,Medicine - Abstract
The order Bunyavirales belongs to the class of Ellioviricetes and is classified into fourteen families. Some species of the order Bunyavirales pose potential threats to human health. The continuously increasing research reveals that various viruses within this order achieve immune evasion in the host through suppressing interferon (IFN) response. As the types and nodes of the interferon response pathway are continually updated or enriched, the IFN suppression mechanisms and target points of different virus species within this order are also constantly enriched and exhibit variations. For instance, Puumala virus (PUUV) and Tula virus (TULV) can inhibit IFN response through their functional NSs inhibiting downstream factor IRF3 activity. Nevertheless, the IFN suppression mechanisms of Dabie bandavirus (DBV) and Guertu virus (GTV) are mostly mediated by viral inclusion bodies (IBs) or filamentous structures (FSs). Currently, there are no effective drugs against several viruses belonging to this order that pose significant threats to society and human health. While the discovery, development, and application of antiviral drugs constitute a lengthy process, our focus on key targets in the IFN response suppression process of the virus leads to potential antiviral strategies, which provide references for both basic research and practical applications.
- Published
- 2024
- Full Text
- View/download PDF
46. Phosphorylation of AGO2 by TBK1 Promotes the Formation of Oncogenic miRISC in NSCLC
- Author
-
Xian Zhao, Yingting Cao, Runhui Lu, Zihan Zhou, Caihu Huang, Lian Li, Jiayi Huang, Ran Chen, Yanli Wang, Jian Huang, Jinke Cheng, Junke Zheng, Yujie Fu, and Jianxiu Yu
- Subjects
AGO2 ,Gefitinib ,miRISC ,non‐small‐cell lung cancer (NSCLC) ,TBK1 ,Science - Abstract
Abstract Non‐small‐cell lung cancer (NSCLC) is a highly lethal tumor that often develops resistance to targeted therapy. It is shown that Tank‐binding kinase 1 (TBK1) phosphorylates AGO2 at S417 (pS417‐AGO2), which promotes NSCLC progression by increasing the formation of microRNA‐induced silencing complex (miRISC). High levels of pS417‐AGO2 in clinical NSCLC specimens are positively associated with poor prognosis. Interestingly, the treatment with EGFR inhibitor Gefitinib can significantly induce pS417‐AGO2, thereby increasing the formation and activity of oncogenic miRISC, which may contribute to NSCLC resistance to Gefitinib. Based on these, two therapeutic strategies is developed. One is jointly to antagonize multiple oncogenic miRNAs highly expressed in NSCLC and use TBK1 inhibitor Amlexanox reducing the formation of oncogenic miRISC. Another approach is to combine Gefitinib with Amlexanox to inhibit the progression of Gefitinib‐resistant NSCLC. This findings reveal a novel mechanism of oncogenic miRISC regulation by TBK1‐mediated pS417‐AGO2 and suggest potential therapeutic approaches for NSCLC.
- Published
- 2024
- Full Text
- View/download PDF
47. Elucidating the binding mechanism of SARS-CoV-2 NSP6-TBK1 and structure-based designing of phytocompounds inhibitors for instigating the host immune response.
- Author
-
Suleman, Muhammad, Ishaq, Iqra, Khan, Haji, khan, Safir Ullah, Masood, Rehana, Albekairi, Norah A., Alshammari, Abdulrahman, Crovella, Sergio, Khadim, Adeeba, and Abbas, Syed Qamar
- Subjects
- *
SARS-CoV-2 , *IMMUNE response , *MOLECULAR docking , *PROTEIN kinases , *BETACORONAVIRUS - Abstract
SARS-CoV-2, also referred to as severe acute respiratory syndrome coronavirus 2, is the virus responsible for causing COVID-19, an infectious disease that emerged in Wuhan, China, in December 2019. Among its crucial functions, NSP6 plays a vital role in evading the human immune system by directly interacting with a receptor called TANK-binding kinase (TBK1), leading to the suppression of IFNp production. Consequently, in the present study we used the structural and biophysical approaches to analyze the effect of newly emerged mutations on the binding of NSP6 and TBK1. Among the identified mutations, four (F35G, L37F, L125F, and I162T) were found to significantly destabilize the structure of NSP6. Furthermore, the molecular docking analysis highlighted that the mutant NSP6 displayed its highest binding affinity with TBK1, exhibiting docking scores of-1436.2 for the wildtype and-1723.2,-1788.6,-1510.2, and-1551.7 for the F35G, L37F, L125F, and I162T mutants, respectively. This suggests the potential for an enhanced immune system evasion capability of NSP6. Particularly, the F35G mutation exhibited the strongest binding affinity, supported by a calculated binding free energy of-172.19 kcal/mol. To disrupt the binding between NSP6 and TBK1, we conducted virtual drug screening to develop a novel inhibitor derived from natural products. From this screening, we identified the top 5 hit compounds as the most promising candidates with a docking score of-6.59 kcal/mol,-6.52 kcal/mol,-6.32 kcal/mol,-6.22 kcal/mol, and-6.21 kcal/mol. The molecular dynamic simulation of top 3 hits further verified the dynamic stability of drugs-NSP6 complexes. In conclusion, this study provides valuable insight into the higher infectivity of the SARS-CoV-2 new variants and a strong rationale for the development of novel drugs against NSP6. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
48. The genetic puzzle of a SOD1-patient with ocular ptosis and a motor neuron disease: a case report.
- Author
-
Vacchiano, Veria, Palombo, Flavia, Ormanbekova, Danara, Fiorini, Claudio, Fiorentino, Alessia, Caporali, Leonardo, Mastrangelo, Andrea, Valentino, Maria Lucia, Capellari, Sabina, Liguori, Rocco, and Carelli, Valerio
- Subjects
MOTOR neuron diseases ,AMYOTROPHIC lateral sclerosis ,GENETIC variation ,MONOGENIC & polygenic inheritance (Genetics) ,MITOCHONDRIAL DNA ,HEREDITY ,OLANZAPINE - Abstract
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease with a complex genetic architecture, showing monogenic, oligogenic, and polygenic inheritance. In this study, we describe the case of a 71 years-old man diagnosed with ALS with atypical clinical features consisting in progressive ocular ptosis and sensorineural deafness. Genetic analyses revealed two heterozygous variants, in the SOD1 (OMIM*147450) and the TBK1 (OMIM*604834) genes respectively, and furthermore mitochondrial DNA (mtDNA) sequencing identified the homoplasmic m.14484T>C variant usually associated with Leber’s Hereditary Optic Neuropathy (LHON). We discuss how all these variants may synergically impinge on mitochondrial function, possibly contributing to the pathogenic mechanisms which might ultimately lead to the neurodegenerative process, shaping the clinical ALS phenotype enriched by adjunctive clinical features. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
49. Mutant p53R211* ameliorates inflammatory arthritis in AIA rats via inhibition of TBK1-IRF3 innate immune response.
- Author
-
Zeng, Yaling, Ng, Jerome P. L., Wang, Linna, Xu, Xiongfei, Law, Betty Yuen Kwan, Chen, Guobing, Lo, Hang Hong, Yang, Lijun, Yang, Jiujie, Zhang, Lei, Qu, Liqun, Yun, Xiaoyun, Zhong, Jing, Chen, Ruihong, Zhang, Dingqi, Wang, Yuping, Luo, Weidan, Qiu, Congling, Huang, Baixiong, and liu, Wenfeng
- Subjects
- *
EXPERIMENTAL arthritis , *IMMUNE response , *T helper cells , *KNEE joint , *RHEUMATOID arthritis , *ARTHRITIS - Abstract
Background: Rheumatoid arthritis (RA) is an autoimmune inflammation disease characterized by imbalance of immune homeostasis. p53 mutants are commonly described as the guardian of cancer cells by conferring them drug-resistance and immune evasion. Importantly, p53 mutations have also been identified in RA patients, and this prompts the investigation of its role in RA pathogenesis. Methods: The cytotoxicity of disease-modifying anti-rheumatic drugs (DMARDs) against p53 wild-type (WT)/mutant-transfected RA fibroblast-like synoviocytes (RAFLSs) was evaluated by MTT assay. Adeno-associated virus (AAV) was employed to establish p53 WT/R211* adjuvant-induced arthritis (AIA) rat model. The arthritic condition of rats was assessed by various parameters such as micro-CT analysis. Knee joint samples were isolated for total RNA sequencing analysis. The expressions of cytokines and immune-related genes were examined by qPCR, ELISA assay and immunofluorescence. The mechanistic pathway was determined by immunoprecipitation and Western blotting in vitro and in vivo. Results: Among p53 mutants, p53R213* exhibited remarkable DMARD-resistance in RAFLSs. However, AAV-induced p53R211* overexpression ameliorated inflammatory arthritis in AIA rats without Methotrexate (MTX)-resistance, and our results discovered the immunomodulatory effect of p53R211* via suppression of T-cell activation and T helper 17 cell (Th17) infiltration in rat joint, and finally downregulated expressions of pro-inflammatory cytokines. Total RNA sequencing analysis identified the correlation of p53R211* with immune-related pathways. Further mechanistic studies revealed that p53R213*/R211* instead of wild-type p53 interacted with TANK-binding kinase 1 (TBK1) and suppressed the innate immune TBK1–Interferon regulatory factor 3 (IRF3)–Stimulator of interferon genes (STING) cascade. Conclusions: This study unravels the role of p53R213* mutant in RA pathogenesis, and identifies TBK1 as a potential anti-inflammatory target. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
50. African swine fever virus S273R protein antagonizes type I interferon production by interfering with TBK1 and IRF3 interaction.
- Author
-
Li, Hui, Zheng, Xiaojie, Li, You, Zhu, Yingqi, Xu, Yangyang, Yu, Zilong, and Feng, Wen-Hai
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.