1
|
Li Y, Chen Y, Tang Y, Yang T, Zhou P, Miao L, Chen H, Deng Y. Breaking the barriers in effective and safe Toll-like receptor stimulation via nano-immunomodulators for potent cancer immunotherapy. J Control Release 2025; 382:113667. [PMID: 40157608 DOI: 10.1016/j.jconrel.2025.113667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/20/2025] [Accepted: 03/26/2025] [Indexed: 04/01/2025]
Abstract
Immunotherapy is an emerging strategy that awakens the intrinsic immune system for cancer treatment. Generally, successful immunotherapy of malignant tumours relies on the effective production of tumour-associated antigens and their lymph node delivery, antigen processing and presentation for T-cell activation, and the dismantling of the immunosuppressive tumour microenvironment. Toll-like receptor (TLR) agonists are potent stimulants in cancer immunotherapy, which can directly activate antigen-presenting cells (APCs) and further induce T cell activation for antitumour immune response and convert immunosuppressive tumour microenvironment to an immunogenic one for cooperative tumour ablation. However, TLR agonists for effective cancer immunotherapy have encountered essential challenges, such as insufficient immune activation and systemic side effects. In recent years, nano-immunomodulators with TLR agonists have been employed for tumour- and/or lymph node-targeted immune activation to improve the antitumour immune response and alleviate their systemic toxicities, providing a promising strategy for enhanced cancer immunotherapy. Herein, we introduce the recent progress in developing various TLR nano-immunomodulators for cancer immunotherapy via APC activation and tumour microenvironment remodelling. Upon elucidating the rational design principles of nano-immunomodulators, we elucidate the advancement of TLR nanoagonists to break the barriers in effective and safe Toll-like receptor stimulation for potent cancer immunotherapy.
Collapse
Affiliation(s)
- Yaoqi Li
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yitian Chen
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Yong'an Tang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Tao Yang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Ping Zhou
- State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China
| | - Liyan Miao
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Institute for Interdisciplinary Drug Research and Translational Sciences, Soochow University, Suzhou 215006, China.
| | - Huabing Chen
- Department of Pharmacy, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou 215006, China; Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China.
| | - Yibin Deng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, and College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China; Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Soochow University, Suzhou 215123, China; State Key Laboratory of Molecular Engineering of Polymers, Fudan University, Shanghai 200438, China.
| |
Collapse
|
2
|
Griffin S, de Oliveira Mallia J, Psakis G, Attard J, Caruana M, Gatt R. Comparative analysis of N/TERT-1 and N/TERT-2G keratinocyte responses to oxidative stress and immune challenges. Cell Signal 2025; 132:111861. [PMID: 40355015 DOI: 10.1016/j.cellsig.2025.111861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 04/05/2025] [Accepted: 05/09/2025] [Indexed: 05/14/2025]
Abstract
The responses of N/TERT-1 and N/TERT-2G keratinocyte cell lines to oxidative stress and immune challenges were investigated to assess their suitability for dermatological testing. The cell lines were exposed to various stimuli, including PAMPs, DAMPs, H₂O₂, and menadione, to assess cytokine production, oxidative stress markers, cell viability, apoptosis, and membrane integrity. IL-1α, IL-6, IL-8, TNF-α, and TGF-β levels significantly increased in N/TERT-1 cells following exposure to LPS, while N/TERT-2G cells remained unaffected. Both cell lines showed increased production of IL-1α, IL-1β, TNF-α, IL-6, and IL-8 in response to dsDNA and LMW and HMW Poly I:C, although TGF-β significantly decreased only in N/TERT-1 cells. In response to H₂O₂, a dose-dependent increase in cytokine levels was observed in N/TERT-2G, whereas N/TERT-1 did not exhibit a clear dose-dependent response. Markers of oxidative stress, including SOD and GSH, displayed similar patterns in both cell lines, with N/TERT-2G showing slightly higher sensitivity. Lipid peroxidation and mitochondrial membrane potential fluctuations were more pronounced in N/TERT-2G, suggesting greater oxidative stress sensitivity. The baseline GSH levels were higher in N/TERT-1 cells, which may contribute towards the enhanced resilience to oxidative stress. Despite decreased viability in MTT assays following H₂O₂ exposure, the lack of significant changes in cleaved Caspase-3 levels indicated that apoptosis was not the primary mechanism of cell death. These findings highlight the distinct characteristics of N/TERT-1 and N/TERT-2G cells, with N/TERT-1 showing higher baseline resilience to oxidative stress and N/TERT-2G displaying greater sensitivity, particularly to H₂O₂. The study underscores the importance of selecting the appropriate cell line for specific research applications in skin biology and disease modelling, considering the differences in their responses to oxidative and immune challenges.
Collapse
Affiliation(s)
- Sholeem Griffin
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta.
| | - Jefferson de Oliveira Mallia
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| | - Georgios Psakis
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| | - Juan Attard
- Department of Food Sciences and Nutrition, Faculty of Health Sciences, University of Malta, Msida MSD2080, Malta
| | - Matthias Caruana
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta
| | - Ruben Gatt
- Metamaterials Unit, Faculty of Science, University of Malta, Msida MSD2080, Malta; Centre for Molecular Medicine and Biobanking, University of Malta, Msida MSD2080, Malta
| |
Collapse
|
3
|
Kumar S, Kahle AD, Keeler AB, Zunder ER, Deppmann CD. Characterizing Microglial Signaling Dynamics During Inflammation Using Single-Cell Mass Cytometry. Glia 2025; 73:1022-1035. [PMID: 39780484 PMCID: PMC11920681 DOI: 10.1002/glia.24670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 12/12/2024] [Accepted: 12/23/2024] [Indexed: 01/11/2025]
Abstract
Microglia play a critical role in maintaining central nervous system (CNS) homeostasis and display remarkable plasticity in their response to inflammatory stimuli. However, the specific signaling profiles that microglia adopt during such challenges remain incompletely understood. Traditional transcriptomic approaches provide valuable insights, but fail to capture dynamic post-translational changes. In this study, we utilized time-resolved single-cell mass cytometry (CyTOF) to measure distinct signaling pathways activated in microglia upon exposure to bacterial and viral mimetics-lipopolysaccharide (LPS) and polyinosinic-polycytidylic acid (Poly(I:C)), respectively. Furthermore, we evaluated the immunomodulatory role of astrocytes on microglial signaling in mixed cultures. Microglia or mixed cultures derived from neonatal mice were treated with LPS or Poly(I:C) for 48 h. Cultures were stained with a panel of 33 metal-conjugated antibodies targeting signaling and identity markers. High-dimensional clustering analysis was used to identify emergent signaling modules. We found that LPS treatment led to more robust early activation of pp38, pERK, pRSK, and pCREB compared to Poly(I:C). Despite these differences, both LPS and Poly(I:C) upregulated the classical reactivity markers CD40 and CD86 at later time points. Strikingly, the presence of astrocytes significantly blunted microglial responses to both stimuli, particularly dampening CD40 upregulation. Our studies demonstrate that single-cell mass cytometry effectively captures the dynamic signaling landscape of microglia under pro-inflammatory conditions. This approach may pave the way for targeted therapeutic investigations of various neuroinflammatory disorders. Moreover, our findings underscore the necessity of considering cellular context, such as astrocyte presence, in interpreting microglial behavior during inflammation.
Collapse
Affiliation(s)
- Sushanth Kumar
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate Program, School of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - August D. Kahle
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Austin B. Keeler
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Eli R. Zunder
- Department of Biomedical Engineering, School of EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Program in Fundamental Neuroscience, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| | - Christopher D. Deppmann
- Department of Biology, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
- Neuroscience Graduate Program, School of MedicineUniversity of VirginiaCharlottesvilleVirginiaUSA
- Department of Biomedical Engineering, School of EngineeringUniversity of VirginiaCharlottesvilleVirginiaUSA
- Program in Fundamental Neuroscience, College of Arts and SciencesUniversity of VirginiaCharlottesvilleVirginiaUSA
| |
Collapse
|
4
|
Keramati F, Leijte GP, Bruse N, Grondman I, Habibi E, Ruiz-Moreno C, Megchelenbrink W, Peters van Ton AM, Heesakkers H, Bremmers ME, van Grinsven E, Tesselaar K, van Staveren S, van der Velden WJ, Preijers FW, Te Pas B, van de Loop R, Gerretsen J, Netea MG, Stunnenberg HG, Pickkers P, Kox M. Systemic inflammation impairs myelopoiesis and interferon type I responses in humans. Nat Immunol 2025; 26:737-747. [PMID: 40251340 PMCID: PMC12043512 DOI: 10.1038/s41590-025-02136-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 03/17/2025] [Indexed: 04/20/2025]
Abstract
Systemic inflammatory conditions are classically characterized by an acute hyperinflammatory phase, followed by a late immunosuppressive phase that elevates the susceptibility to secondary infections. Comprehensive mechanistic understanding of these phases is largely lacking. To address this gap, we leveraged a controlled, human in vivo model of lipopolysaccharide (LPS)-induced systemic inflammation encompassing both phases. Single-cell RNA sequencing during the acute hyperinflammatory phase identified an inflammatory CD163+SLC39A8+CALR+ monocyte-like subset (infMono) at 4 h post-LPS administration. The late immunosuppressive phase was characterized by diminished expression of type I interferon (IFN)-responsive genes in monocytes, impaired myelopoiesis and a pronounced attenuation of the immune response on a secondary LPS challenge 1 week after the first. The infMono gene program and impaired myelopoiesis were also detected in patient cohorts with bacterial sepsis and coronavirus disease. IFNβ treatment restored type-I IFN responses and proinflammatory cytokine production and induced monocyte maturation, suggesting a potential treatment option for immunosuppression.
Collapse
Affiliation(s)
- Farid Keramati
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Guus P Leijte
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Niklas Bruse
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Inge Grondman
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ehsan Habibi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Québec, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Québec, Canada
| | - Cristian Ruiz-Moreno
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Wout Megchelenbrink
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Hidde Heesakkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Manita E Bremmers
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erinke van Grinsven
- Department of Respiratory Medicine and Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kiki Tesselaar
- Department of Respiratory Medicine and Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Selma van Staveren
- Department of Respiratory Medicine and Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- TmonoCOAST, Amsterdam, The Netherlands
| | | | - Frank W Preijers
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brigit Te Pas
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Raoul van de Loop
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands.
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
5
|
Blutt SE, Miller AD, Conner ME. Dendritic cell expression of MyD88 is required for rotavirus-induced B cell activation. J Virol 2025:e0065325. [PMID: 40304491 DOI: 10.1128/jvi.00653-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
Intestinal IgA, produced by local intestinal B cells, is thought to play a major role in protection against intestinal infections. Rotavirus, a well-characterized intestinal virus, induces a rapid viral-specific intestinal IgA response that occurs in the absence of T cells. Previous work has indicated that dendritic cells facilitate the early IgA response to rotavirus. To determine whether the early Peyer's patch B cell activation associated with rotavirus infection in mice requires dendritic cells, we depleted dendritic cells and assessed B cell activation. Depletion of CD11c+ cells in vivo prior to infection resulted in a complete abrogation of Peyer's patch B cell activation. With the use of in vitro cell-based assays, CD11c+, but not T or CD11b+ cells, was shown to be essential for rotavirus-induced activation of B cells. Investigation of several pathways of B cell activation revealed that dendritic cell expression of MyD88 and signaling through the type I interferon receptor were critical for the ability of the virus to induce B cell activation. These findings indicate that CD11c+ dendritic cells can modulate B cell responses to viruses through toll-like receptor and type I interferon signaling pathways.IMPORTANCEDendritic cells are key mediators of immune responses in the intestine. They can capture and process rotavirus antigens and present these antigens to B cells, which produce critical IgA antibody that is essential for clearance of rotavirus infection and protection from reinfection. In the work presented here, we demonstrate that dendritic cell expression of MyD88, a key component of pattern recognition pathways, and not classical IgA pathway molecules such as BAFF and APRIL, is critical for the ability of the dendritic cell to induce the activation of B cells. Our findings emphasize the important role that dendritic cells play in initiating and regulating immune responses including T cell-independent B cell activation. A consideration of the role of dendritic cells in B cell activation and antibody production is an important feature in the development of therapeutic and preventive modalities to combat intestinal viral infections.
Collapse
Affiliation(s)
- Sarah E Blutt
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| | - Amber D Miller
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
| | - Margaret E Conner
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, Texas, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
- Huffington Department of Education, Innovation, and Technology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
6
|
Li JX, Zhang J, Li CH, Li YF, Chen HM, Li T, Zhang Q, Kong BH, Wang PH. Human papillomavirus E1 proteins inhibit RIG-I/MDA5-MAVS, TLR3-TRIF, cGAS-STING, and JAK-STAT signaling pathways to evade innate antiviral immunity. Front Immunol 2025; 16:1549766. [PMID: 40330484 PMCID: PMC12052760 DOI: 10.3389/fimmu.2025.1549766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 05/08/2025] Open
Abstract
Human papillomavirus (HPV) is a major etiological agent of both malignant and benign lesions, with high-risk types, such as HPV16 and HPV18, being strongly linked to cervical cancer, while low-risk types like HPV11 are associated with benign conditions. While viral proteins such as E6 and E7 are well-established regulators of immune evasion, the role of E1 in modulating the host antiviral responses remains insufficiently characterized. This study investigates the immunomodulatory functions of HPV16 and HPV11 E1 in suppressing innate antiviral immune signaling pathways. Through a combination of RT-qPCR and luciferase reporter assays, we demonstrate that E1 suppresses the production of interferons and interferon-stimulated genes triggered by viral infections and the activation of RIG-I/MDA5-MAVS, TLR3-TRIF, cGAS-STING, and JAK-STAT pathways. Co-immunoprecipitation assays reveal that E1 interacts directly with key signaling molecules within these pathways. E1 also impairs TBK1 and IRF3 phosphorylation and obstructs the nuclear translocation of IRF3, thereby broadly suppressing IFN responses. Additionally, E1 disrupts the JAK-STAT pathway by binding STAT1, which prevents the assembly and nuclear localization of the ISGF3 complex containing STAT1, STAT2, and IRF9, thereby further diminishing antiviral response. These findings establish E1 as a pivotal regulator of immune evasion and suggest its potential as a novel therapeutic target to enhance antiviral immunity in HPV-associated diseases.
Collapse
MESH Headings
- Humans
- Immunity, Innate
- Signal Transduction/immunology
- Interferon-Induced Helicase, IFIH1/metabolism
- Interferon-Induced Helicase, IFIH1/immunology
- DEAD Box Protein 58/metabolism
- DEAD Box Protein 58/immunology
- Membrane Proteins/metabolism
- Membrane Proteins/immunology
- Nucleotidyltransferases/metabolism
- Nucleotidyltransferases/immunology
- Toll-Like Receptor 3/metabolism
- Toll-Like Receptor 3/immunology
- Immune Evasion
- Papillomavirus Infections/immunology
- Papillomavirus Infections/virology
- Human papillomavirus 16/immunology
- Receptors, Immunologic
- Oncogene Proteins, Viral/immunology
- Oncogene Proteins, Viral/metabolism
- Adaptor Proteins, Vesicular Transport/metabolism
- Adaptor Proteins, Vesicular Transport/immunology
- Adaptor Proteins, Signal Transducing/metabolism
- Adaptor Proteins, Signal Transducing/immunology
- Human papillomavirus 11/immunology
- HEK293 Cells
- STAT Transcription Factors/metabolism
- Interferon Regulatory Factor-3
- Human Papillomavirus Viruses
Collapse
Affiliation(s)
- Jin-Xin Li
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Jing Zhang
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Cheng-Hao Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Yun-Fang Li
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Hui-Min Chen
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Tao Li
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| | - Qing Zhang
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
| | - Bei-Hua Kong
- Department of Obstetrics and Gynecology, Qilu Hospital, Shandong University, Jinan, China
- Gynecologic Oncology Key Laboratory of Shandong Province, Qilu Hospital, Shandong University, Jinan, China
| | - Pei-Hui Wang
- Department of Infectious Disease and Hepatology, The Second Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
- Key Laboratory for Experimental Teratology of Ministry of Education and Advanced Medical Research Institute, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, China
| |
Collapse
|
7
|
Li TF, Rothhaar P, Lang A, Grünvogel O, Colasanti O, Ugarte SMO, Traut J, Piras A, Acosta-Rivero N, Gonçalves Magalhães V, Springer E, Betz A, Huang HE, Park J, Qiu R, Gnouamozi GE, Mehnert AK, Thi VLD, Urban S, Muckenthaler M, Schlesner M, Wohlleber D, Binder M, Bartenschlager R, Pichlmair A, Lohmann V. RBM39 shapes innate immunity by controlling the expression of key factors of the interferon response. Front Immunol 2025; 16:1568056. [PMID: 40330464 PMCID: PMC12054253 DOI: 10.3389/fimmu.2025.1568056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/18/2025] [Indexed: 05/08/2025] Open
Abstract
Background and aims The contribution of innate immunity to clearance of viral infections of the liver, in particular sensing via Toll-like receptor 3 (TLR3), is incompletely understood. We aimed to identify the factors contributing to the TLR3 response in hepatocytes via CRISPR/Cas9 screening. Methods A genome-wide CRISPR/Cas9 screen on the TLR3 pathway was performed in two liver-derived cell lines, followed by siRNA knockdown validation. SiRNA knockdown and indisulam treatment were used to study the role of RNA-binding motif protein 39 (RBM39) in innate immunity upon poly(I:C) or cytokine treatment and viral infections. Transcriptome, proteome, and alternative splicing were studied via RNA sequencing and mass spectrometry upon depletion of RBM39. Results Our CRISPR/Cas9 screen identified RBM39, which is highly expressed in hepatocytes, as an important regulator of the TLR3 pathway. Knockdown of RBM39 or treatment with indisulam, an aryl sulfonamide drug targeting RBM39 for proteasomal degradation, strongly reduced the induction of interferon-stimulated genes (ISGs) in response to double-stranded RNA (dsRNA) or viral infections. RNA sequencing (seq) and mass spectrometry identified that transcription and/or splicing of the key pathway components IRF3, RIG-I, and MDA5 were affected by RBM39 depletion, along with multiple other cellular processes identified previously. RBM39 knockdown further restrained type I and type III IFN pathways by reducing the expression of individual receptor subunits and STAT1/2. The function of RBM39 was furthermore not restricted to hepatocytes. Conclusion We identified RBM39 as a regulatory factor of cell intrinsic innate immune signaling. Depletion of RBM39 impaired TLR3, RIG-I/MDA5, and IFN responses by affecting the basal expression of key pathway components.
Collapse
Affiliation(s)
- Teng-Feng Li
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Paul Rothhaar
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Arthur Lang
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Oliver Grünvogel
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Ombretta Colasanti
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Santa Mariela Olivera Ugarte
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Jannik Traut
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Antonio Piras
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Nelson Acosta-Rivero
- Department of Infectious Diseases, Molecular Virology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | | | - Emely Springer
- Institute of Molecular Immunology, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Andreas Betz
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Hao-En Huang
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Jeongbin Park
- Bioinformatics and Omics Data Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ruiyue Qiu
- Heidelberg University, Medical Faculty, Department of Pediatric Oncology, Hematology, Immunology and Pneumology, Heidelberg, Germany
| | - Gnimah Eva Gnouamozi
- Department of Infectious Diseases, Molecular Virology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Ann-Kathrin Mehnert
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Viet Loan Dao Thi
- Department of Infectious Diseases, Virology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Heidelberg, Germany
| | - Stephan Urban
- Department of Infectious Diseases, Molecular Virology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Heidelberg, Germany
| | - Martina Muckenthaler
- Heidelberg University, Medical Faculty, Department of Pediatric Oncology, Hematology, Immunology and Pneumology, Heidelberg, Germany
| | - Matthias Schlesner
- Bioinformatics and Omics Data Analysis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- Biomedical Informatics, Data Mining and Data Analytics, Faculty of Applied Computer Science and Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Dirk Wohlleber
- Institute of Molecular Immunology, University Hospital Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Marco Binder
- Division of Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ralf Bartenschlager
- Department of Infectious Diseases, Molecular Virology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- Division of Virus-Associated Carcinogenesis, German Cancer Research Center (DKFZ), Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Heidelberg, Germany
| | - Andreas Pichlmair
- Institute of Virology, School of Medicine, Technical University of Munich, Munich, Germany
- German Center for Infection Research (DZIF), Munich Partner Site, Munich, Germany
| | - Volker Lohmann
- Department of Infectious Diseases, Molecular Virology, Section Virus-Host-Interactions, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
- German Center for Infection Research (DZIF), Heidelberg Partner Site, Heidelberg, Germany
| |
Collapse
|
8
|
AlDaif BA, Fleming SB. Innate Immune Sensing of Parapoxvirus Orf Virus and Viral Immune Evasion. Viruses 2025; 17:587. [PMID: 40285029 PMCID: PMC12031380 DOI: 10.3390/v17040587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/14/2025] [Accepted: 04/17/2025] [Indexed: 04/29/2025] Open
Abstract
Orf virus (ORFV) is the type species of Parapoxvirus of the Poxviridae family that induces cutaneous pustular skin lesions in sheep and goats, and causes zoonotic infections in humans. Pattern recognition receptors (PRRs) sense pathogen-associated molecular patterns (PAMPs), leading to the triggering of the innate immune response through multiple signalling pathways involving type I interferons (IFNs). The major PAMPs generated during viral infection are nucleic acids, which are the most important molecules that are recognized by the host. The induction of type l IFNs leads to activation of the Janus kinase (JAK)-signal transducer activator of transcription (STAT) pathway, which results in the induction of hundreds of interferon-stimulated genes (ISGs), many of which encode proteins that have antiviral roles in eliminating virus infection and create an antiviral state. Genetic and functional analyses have revealed that ORFV, as found for other poxviruses, has evolved multiple immunomodulatory genes and strategies that manipulate the innate immune sensing response.
Collapse
Affiliation(s)
| | - Stephen B. Fleming
- Virus Research Unit, Department of Microbiology and Immunology, University of Otago, Dunedin 9016, New Zealand;
| |
Collapse
|
9
|
Zhang T, Pan Y, Sawa T, Akaike T, Matsunaga T. Supersulfide donors and their therapeutic targets in inflammatory diseases. Front Immunol 2025; 16:1581385. [PMID: 40308575 PMCID: PMC12040673 DOI: 10.3389/fimmu.2025.1581385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 03/31/2025] [Indexed: 05/02/2025] Open
Abstract
Inflammation is one defense mechanism of the body that has multiple origins, ranging from physical agents to infectious agents including viruses and bacteria. The resolution of inflammation has emerged as a critical endogenous process that protects host tissues from prolonged or excessive inflammation, which can become chronic. Failure of the inflammation resolution is a key pathological mechanism that drives the progression of numerous inflammatory diseases. Owing to the various side effects of currently available drugs to control inflammation, novel therapeutic agents that can prevent or suppress inflammation are needed. Supersulfides are highly reactive and biologically potent molecules that function as antioxidants, redox regulators, and modulators of cell signaling. The catenation state of individual sulfur atoms endows supersulfides with unique biological activities. Great strides have recently been made in achieving a molecular understanding of these sulfur species, which participate in various physiological and pathological pathways. This review mainly focuses on the anti-inflammatory effects of supersulfides. The review starts with an overview of supersulfide biology and highlights the roles of supersulfides in both immune and inflammatory responses. The various donors used to generate supersulfides are assessed as research tools and potential therapeutic agents. Deeper understanding of the molecular and cellular bases of supersulfide-driven biology can help guide the development of innovative therapeutic strategies to prevent and treat diseases associated with various immune and inflammatory responses.
Collapse
Affiliation(s)
- Tianli Zhang
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
| | - Yuexuan Pan
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takaaki Akaike
- Department of Redox Molecular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Shimadzu × Tohoku University Supersulfides Life Science Co-creation Research Center, Sendai, Japan
| | - Tetsuro Matsunaga
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita, Japan
- Shimadzu × Tohoku University Supersulfides Life Science Co-creation Research Center, Sendai, Japan
| |
Collapse
|
10
|
Kawakita T, Sekiya T, Kameda Y, Nomura N, Ohno M, Handabile C, Yamaya A, Fukuhara H, Anraku Y, Kita S, Toba S, Tsukamoto H, Sawa T, Oshiumi H, Itoh Y, Maenaka K, Sato A, Sawa H, Suzuki Y, Brown LE, Jackson DC, Kida H, Matsumoto M, Seya T, Shingai M. ARNAX is an ideal adjuvant for COVID-19 vaccines to enhance antigen-specific CD4 + and CD8 + T-cell responses and neutralizing antibody induction. J Virol 2025:e0229024. [PMID: 40231823 DOI: 10.1128/jvi.02290-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/02/2025] [Indexed: 04/16/2025] Open
Abstract
ARNAX is a synthetic nucleotide-based Toll-like receptor 3 (TLR3) ligand that specifically stimulates the TLR3/TIR domain-containing adaptor molecule 1 (TICAM-1) pathway without activating inflammatory responses. ARNAX activates cellular immunity via cross-presentation; hence, its practical application has been demonstrated in cancer immunotherapy. Given the importance of cellular immunity in virus infections, ARNAX is expected to be a more effective vaccine adjuvant for virus infections than alum, an adjuvant approved for human use that mainly enhances humoral immunity. In the present study, the trimeric recombinant spike protein of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was prepared as a vaccine antigen and formulated with ARNAX. When T-cell and neutralizing antibody responses were evaluated in immunized mice, antigen formulated with ARNAX generated significantly larger numbers of antigenspecific CD4+ and CD8+ T cells, as well as higher titers of neutralizing antibodies, compared to antigen alone or antigen formulated with alum. In experiments where immunized mice were challenged with a SARS-CoV-2 mouse-adapted virus derived from the ancestral strain, immunization with antigen formulated with ARNAX reduced virus titers in the lungs at 3 days post-infection to a much greater extent than did immunization with either antigen alone or that formulated with alum. These results show that ARNAX potently enhances the levels of both cellular and humoral immunity above those seen with alum, providing significantly greater viral clearing responses. Thus, ARNAX may act as a useful adjuvant for prophylactic vaccines, particularly for viral infectious diseases. IMPORTANCE Cellular immunity is a critical immunological defense system against virus infections. However, aluminum salts, the most widely used adjuvant for vaccines for human use, do not promote strong cellular immunity. To prepare for the next pandemic of viral origin, the development of Th1-type adjuvants with low adverse reactions that induce cellular immunity is necessary. ARNAX is a TLR3 agonist consisting of DNA-RNA hybrid nucleic acid, which is expected to be an adjuvant that induces cellular immunity. The present study using a coronavirus disease 2019 mouse model demonstrated that ARNAX potently induces cellular immunity in addition to humoral immunity with minimal induction of inflammatory cytokines. Therefore, ARNAX has the potential to be used as a potent and welltolerated adjuvant for vaccines against pandemic viruses emerging in the future.
Collapse
Affiliation(s)
- Tomomi Kawakita
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Toshiki Sekiya
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Yayoi Kameda
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Naoki Nomura
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Division of International Research Promotion, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Marumi Ohno
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
| | - Chimuka Handabile
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Akari Yamaya
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori, Japan
| | - Hideo Fukuhara
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yuki Anraku
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Shunsuke Kita
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
| | - Shinsuke Toba
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirotake Tsukamoto
- Division of Clinical Immunology and Cancer Immunotherapy, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroyuki Oshiumi
- Department of Immunology, Graduate School of Medical Sciences, Faculty of Life Sciences, Kumamoto University, Kumamoto, Japan
| | - Yasushi Itoh
- Division of Pathogenesis and Disease Regulation, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Katsumi Maenaka
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Pathogen Structure, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Laboratory of Biomolecular Science, and Center for Research and Education on Drug Discovery, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Japan
- Global Station for Biosurfaces and Drug Discovery, Hokkaido University, Sapporo, Japan
| | - Akihiko Sato
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Shionogi Pharmaceutical Research Center, Shionogi & Company, Limited, Toyonaka, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Hirofumi Sawa
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- One Health Research Center, Hokkaido University, Sapporo, Japan
- Division of Molecular Pathobiology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Yasuhiko Suzuki
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Bioresources, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Lorena E Brown
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - David C Jackson
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- The Department of Microbiology and Immunology, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Hiroshi Kida
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| | - Misako Matsumoto
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori, Japan
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Tsukasa Seya
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Nebuta Research Institute for Life Sciences, Aomori University, Aomori, Japan
- Department of Vaccine Immunology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masashi Shingai
- Division of Vaccine Immunology, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- Institute for Vaccine Research and Development (HU-IVReD), Hokkaido University, Sapporo, Japan
- Division of Biologics Development, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
- International Collaboration Unit, International Institute for Zoonosis Control, Hokkaido University, Sapporo, Japan
| |
Collapse
|
11
|
Wang W, Gong J. Identification of Key Nucleotide Metabolism Genes in Diabetic Retinopathy Based on Bioinformatics Analysis and Experimental Verification. BIOLOGY 2025; 14:409. [PMID: 40282274 PMCID: PMC12024606 DOI: 10.3390/biology14040409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/09/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
A dysregulated nucleotide metabolism has been implicated in the pathogenesis of diabetic retinopathy (DR). RNA sequencing datasets, GSE102485, GSE60436, and GSE165784, were downloaded from the GEO database. The differentially expressed genes (DEGs) between the DR and controls overlapped with nucleotide metabolism-related genes (NM-RGs), resulting in the differentially expressed NM-RGs (DE-NMRGs). Next, the core genes were identified by the five algorithms of the CytoHubba plugin. Receiver Operating Characteristic (ROC) curves and gene expression analysis were utilized to confirm the biomarkers. Then, the correlations between biomarker expression and the immune-related module were analyzed. The miRNA and transcription factor (TF) predictions, biomarker-targeting drugs, and molecular docking were implemented separately. The interaction between each subcluster of DR was elucidated through single-cell RNA (scRNA) analysis. Moreover, RT-PCR was applied to verify the expression of the biomarkers. In GSE102485, 48 DE-NMRGs were identified via the intersection of 1359 DEGs and 882 NM-RGs. Using the CytoHubba plugin, HMOX1, TLR4, and ACE were selected as core genes. As per the GSVA result, the interferon alpha response, IL6_JAK_STAT3 signaling, and apoptosis were activated in the DR group. The TF prediction identified TLR4 and HMOX1 as potential target genes of USF2. In conclusion, ACE and HMOX1 were possible diagnostic biomarkers related to nucleotide metabolism in DR.
Collapse
Affiliation(s)
- Wei Wang
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China;
- Anhui Public Health Clinical Center, Hefei 230022, China
| | - Jianyang Gong
- Department of Ophthalmology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China;
- Anhui Public Health Clinical Center, Hefei 230022, China
| |
Collapse
|
12
|
Schultz TE, Mathmann CD, Domínguez Cadena LC, Muusse TW, Kim H, Wells JW, Ulett GC, Hamerman JA, Brooks AJ, Kobe B, Sweet MJ, Stacey KJ, Blumenthal A. TLR4 endocytosis and endosomal TLR4 signaling are distinct and independent outcomes of TLR4 activation. EMBO Rep 2025:10.1038/s44319-025-00444-2. [PMID: 40204912 DOI: 10.1038/s44319-025-00444-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/11/2025] Open
Abstract
Toll-like receptor 4 (TLR4) signaling at the plasma membrane and in endosomes results in distinct contributions to inflammation and host defence. Current understanding indicates that endocytosis of cell surface-activated TLR4 is required to enable subsequent signaling from endosomes. Contrary to this prevailing model, our data show that endosomal TLR4 signaling is not reliant on cell surface-expressed TLR4 or ligand-induced TLR4 endocytosis. Moreover, previously recognized requirements for the accessory molecule CD14 in TLR4 endocytosis and endosomal signaling are likely attributable to CD14 binding as well as trafficking and transferring lipopolysaccharide (LPS) to TLR4 at different subcellular localizations. TLR4 endocytosis requires the TLR4 intracellular signaling domain, contributions by phospholipase C gamma 2, spleen tyrosine kinase, E1/E2 ubiquitination enzymes, but not canonical TLR signaling adaptors and cascades. Thus, our study identifies independently operating TLR4 signaling modes that control TLR4 endocytosis, pro-inflammatory cell surface-derived, as well as endosomal TLR4 signaling. This revised understanding of how TLR4 functions within cells might be harnessed to selectively amplify or restrict TLR4 activation for the development of adjuvants, vaccines and therapeutics.
Collapse
Affiliation(s)
- Thomas E Schultz
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Carmen D Mathmann
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | | | - Timothy W Muusse
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Hyoyoung Kim
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - James W Wells
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Glen C Ulett
- School of Pharmacy and Medical Sciences and Menzies Health Institute Queensland, Griffith University, Gold Coast, QLD, 4215, Australia
| | - Jessica A Hamerman
- Immunology Program, Benaroya Research Institute, Seattle, WA, 98101, USA
| | - Andrew J Brooks
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Matthew J Sweet
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Katryn J Stacey
- School of Chemistry and Molecular Biosciences, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Antje Blumenthal
- Frazer Institute, The University of Queensland, Brisbane, QLD, 4102, Australia.
| |
Collapse
|
13
|
Tao M, Xue M, Zhou D, Zhang L, Hou X, Zhu X, Feng S, Yan H, Qian X, Wei L, Zong C, Yang X, Zhang L. Lipopolysaccharide Induces Resistance to CAR-T Cell Therapy of Colorectal Cancer Cells through TGF-β-Mediated Stemness Enhancement. Mol Pharm 2025; 22:1790-1803. [PMID: 40116228 DOI: 10.1021/acs.molpharmaceut.4c00264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2025]
Abstract
Chimeric antigen receptor-T (CAR-T) cell therapy is a cellular immunotherapy that has emerged in recent years, and increasing studies showed that therapeutic resistance to CAR-T cell therapy presents in colorectal cancer patients. Lipopolysaccharide (LPS), a component of the cell wall of Gram-negative bacteria, is known to preserve a high concentration in the colon. Whether LPS is a contributing factor to the development of resistance in colorectal cancer cells against CAR-T cell therapy remains unclear. For in vivo experiments, colorectal cancer cells COLO205 were pretreated with LPS for 24 h and then were injected into nude mice through the tail vein, followed by CAR-T cells transplantation one day later. Later, the number of tumors in the lung tissues of the mice was observed. The in vitro experiments were performed on COLO205 cells, which were treated with LPS for 24 h. The effect of LPS on the stemness of COLO205 and SW620 cells was observed by using the colony formation assay and spheroidization experiments. The effect of LPS on the expression of stemness-related genes, including CD44, SOX2, and NANOG, was observed by qRT-PCR assay, Western blotting assay, and immunofluorescence staining. Inhibitors of TGF-β and the MYD88 inhibitor were used to study the mechanisms by which LPS induces the stemness enhancement and resistance to CAR-T cell therapy of COLO205 cells. The correlation between MYD88 and TGFB1, as well as the correlation between TGFB1 and stemness-related genes was analyzed using the TCGA database. Both the in vivo assay of nude mice and the in vitro assay showed that LPS pretreatment could induce resistance to CAR-T cell therapy of colorectal cancer cells. LPS could enhance COLO205 and SW620 cells stemness presented by upregulation of CD44, SOX2, and NANOG. The reverse interfering assay using the TGF-β inhibitor indicated that the autosecretion of TGF-β induced by LPS played a critical role in the stemness enhancement of colorectal cancer cells. The TCGA database analysis revealed a strong positive correlation between MYD88 and TGFB1. Additionally, TGFB1 has been found to upregulate the expression of genes associated with stemness. Further mechanism studies showed that the TLR4/MYD88 pathway medicates LPS-induced TGF-β expression. Our results suggested that LPS-induced resistance to CAR-T cell therapy of colorectal cancer cells through stemness enhancement. TLR4/MYD88 signal pathway-dependent TGF-β expression was involved in stemness enhancement and CAR-T cell therapy resistance. In conclusion, our findings help us to understand the underlying mechanisms of CAR-T cell therapy resistance and indicate that inhibitors of TGF-β and MYD88 are promising targeting candidates to promote a therapeutic effect of CAR-T cell therapy in colorectal cancer in the clinic.
Collapse
Affiliation(s)
- Min Tao
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Mengmeng Xue
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Daoyu Zhou
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Luyao Zhang
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Xiaojuan Hou
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Xinyu Zhu
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Shiyao Feng
- Anhui Medical University, Hefei 230032, China
| | - Haixin Yan
- Department of Urology, The Second Hospital of Anhui Medical University, Hefei 230601, China
| | - Xiaofeng Qian
- Shanghai Putuo District Liqun Hospital, Shanghai 200061, China
| | - Lixin Wei
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Chen Zong
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Xue Yang
- Tumor Immunology and Gene Therapy Center, National Center for Liver Cancer, Naval Medical University, Shanghai 201805, China
| | - Li Zhang
- School of Pharmacy, Anhui Medical University, Hefei 230032, China
- Department of Clinical Pharmacology, The Second Hospital of Anhui Medical University, Hefei 230601, China
- Clinical Research Unit, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| |
Collapse
|
14
|
Chen S, Ye J, Lin Y, Chen W, Huang S, Yang Q, Qian H, Gao S, Hua C. Crucial Roles of RSAD2/viperin in Immunomodulation, Mitochondrial Metabolism and Autoimmune Diseases. Inflammation 2025; 48:520-540. [PMID: 38909344 DOI: 10.1007/s10753-024-02076-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/22/2024] [Accepted: 06/03/2024] [Indexed: 06/24/2024]
Abstract
Autoimmune diseases are typically characterized by aberrant activation of immune system that leads to excessive inflammatory reactions and tissue damage. Nevertheless, precise targeted and efficient therapies are limited. Thus, studies into novel therapeutic targets for the management of autoimmune diseases are urgently needed. Radical S-adenosyl methionine domain-containing 2 (RSAD2) is an interferon-stimulated gene (ISG) renowned for the antiviral properties of the protein it encodes, named viperin. An increasing number of studies have underscored the new roles of RSAD2/viperin in immunomodulation and mitochondrial metabolism. Previous studies have shown that there is a complex interplay between RSAD2/vipeirn and mitochondria and that binding of the iron-sulfur (Fe-S) cluster is necessary for the involvement of viperin in mitochondrial metabolism. Viperin influences the proliferation and development of immune cells as well as inflammation via different signaling pathways. However, the function of RSAD2/viperin varies in different studies and a comprehensive overview of this emerging theme is lacking. This review will describe the characteristics of RSAD2/viperin, decipher its function in immunometabolic processes, and clarify the crosstalk between RSAD2/viperin and mitochondria. Furthermore, we emphasize the crucial roles of RSAD2 in autoimmune diseases and its potential application value.
Collapse
Affiliation(s)
- Siyan Chen
- School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Jiani Ye
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Yinfang Lin
- School of the 1st Clinical Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Wenxiu Chen
- School of the 1st Clinical Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Shenghao Huang
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Qianru Yang
- School of the 1st Clinical Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Hengrong Qian
- School of the 2nd Clinical Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China
| | - Sheng Gao
- Laboratory Animal Center, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China.
| | - Chunyan Hua
- School of Basic Medical Sciences, Wenzhou Medical University, Zhejiang Province, Wenzhou, 325035, China.
| |
Collapse
|
15
|
Ji RL, Tao YX. Biased signaling in drug discovery and precision medicine. Pharmacol Ther 2025; 268:108804. [PMID: 39904401 DOI: 10.1016/j.pharmthera.2025.108804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 01/10/2025] [Accepted: 01/21/2025] [Indexed: 02/06/2025]
Abstract
Receptors are crucial for converting chemical and environmental signals into cellular responses, making them prime targets in drug discovery, with about 70% of drugs targeting these receptors. Biased signaling, or functional selectivity, has revolutionized drug development by enabling precise modulation of receptor signaling pathways. This concept is more firmly established in G protein-coupled receptor and has now been applied to other receptor types, including ion channels, receptor tyrosine kinases, and nuclear receptors. Advances in structural biology have further refined our understanding of biased signaling. This targeted approach enhances therapeutic efficacy and potentially reduces side effects. Numerous biased drugs have been developed and approved as therapeutics to treat various diseases, demonstrating their significant therapeutic potential. This review provides a comprehensive overview of biased signaling in drug discovery and disease treatment, highlighting recent advancements and exploring the therapeutic potential of these innovative modulators across various diseases.
Collapse
Affiliation(s)
- Ren-Lei Ji
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| | - Ya-Xiong Tao
- Department of Anatomy, Physiology and Pharmacology, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, United States.
| |
Collapse
|
16
|
Lv Z, Zhang M, Xu Y, Qin B, Yang H, Wei R, Xiao T. Structural and Functional Characteristics of TLR19 in Barbel Chub Compared to TLR19 in Grass Carp. Int J Mol Sci 2025; 26:3103. [PMID: 40243814 PMCID: PMC11988518 DOI: 10.3390/ijms26073103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2025] [Revised: 03/25/2025] [Accepted: 03/26/2025] [Indexed: 04/18/2025] Open
Abstract
The hybrid offspring of barbel chub Squaliobarbus curriculus and grass carp Ctenopharyngodon idella exhibit stronger resistance to the grass carp reovirus (GCRV) infection than grass carp. Toll-like receptors (TLRs) play indispensable roles in the antiviral immunity of fish. In this study, the structures and antiviral immune functions of barbel chub TLR19 (ScTLR19) and grass carp TLR19 (CiTLR19) were compared. The amino acid sequence of ScTLR19 shared high similarity (97.4%) and identity (94.0%) with that of CiTLR19, and a phylogenetic tree revealed the close evolutionary relationship between ScTLR19 and CiTLR19. Protein domain composition analyses showed that ScTLR19 possessed an additional leucine-rich repeat (designated as LRR9) located at amino acid positions 654-677 in the extracellular region, which was absent in CiTLR19. Multiple sequence alignment and three-dimensional structure comparison also indicated that the extracellular regions of ScTLR19 and CiTLR19 exhibited greater differences compared to their intracellular regions. Molecular docking revealed that the extracellular region of ScTLR19 (docking score = -512.31) showed a stronger tendency for binding with polyI:C, compared to the extracellular region of CiTLR19 (docking score = -474.90). Replacing LRR9 in ScTLR19 with the corresponding amino acid sequence from CiTLR19 reduced the binding activity of ScTLR19 to polyI:C, as confirmed by an ELISA. Moreover, overexpression experiments suggested that ScTLR19 could regulate both the IRF3-TRIF and IRF3-MyD88 signaling pathways during GCRV infection, while CiTLR19 only regulated the IRF3-MyD88 signaling pathway. Importantly, replacing LRR9 in ScTLR19 with the corresponding amino acid sequence from CiTLR19 altered the expression regulation on IRF3, MyD88, and TRIF during GCRV infection. These findings collectively reveal the structural and functional differences between ScTLR19 and CiTLR19, and they may provide data to support a deeper understanding of the molecular mechanisms underlying the differences in GCRV resistance between barbel chub and grass carp, as well as the genetic basis for the heterosis of GCRV resistance in their hybrid offspring.
Collapse
Affiliation(s)
- Zhao Lv
- Hunan Engineering Technology Research Center of Featured Aquatic Resources Utilization, Hunan Agricultural University, Changsha 410128, China; (M.Z.); (Y.X.); (B.Q.); (H.Y.); (R.W.); (T.X.)
| | | | | | | | | | | | | |
Collapse
|
17
|
Tanneti NS, Stillwell HA, Weiss SR. Human coronaviruses: activation and antagonism of innate immune responses. Microbiol Mol Biol Rev 2025; 89:e0001623. [PMID: 39699237 PMCID: PMC11948496 DOI: 10.1128/mmbr.00016-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2024] Open
Abstract
SUMMARYHuman coronaviruses cause a range of respiratory diseases, from the common cold (HCoV-229E, HCoV-NL63, HCoV-OC43, and SARS-CoV-2) to lethal pneumonia (SARS-CoV, SARS-CoV-2, and MERS-CoV). Coronavirus interactions with host innate immune antiviral responses are an important determinant of disease outcome. This review compares the host's innate response to different human coronaviruses. Host antiviral defenses discussed in this review include frontline defenses against respiratory viruses in the nasal epithelium, early sensing of viral infection by innate immune effectors, double-stranded RNA and stress-induced antiviral pathways, and viral antagonism of innate immune responses conferred by conserved coronavirus nonstructural proteins and genus-specific accessory proteins. The common cold coronaviruses HCoV-229E and -NL63 induce robust interferon signaling and related innate immune pathways, SARS-CoV and SARS-CoV-2 induce intermediate levels of activation, and MERS-CoV shuts down these pathways almost completely.
Collapse
Affiliation(s)
- Nikhila S. Tanneti
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Helen A. Stillwell
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Susan R. Weiss
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
18
|
Liu Z, Ou Y, He X, Yuan T, Li M, Long Y, Li Y, Tan Y. Guardians of the Lung: The Multifaceted Roles of Macrophages in Cancer and Infectious Disease. DNA Cell Biol 2025. [PMID: 40106386 DOI: 10.1089/dna.2024.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
Abstract
The lung as an organ that is fully exposed to the external environment for extended periods, comes into contact with numerous inhaled microorganisms. Lung macrophages are crucial for maintaining lung immunity and operate primarily through signaling pathways such as toll-like receptor 4 and nuclear factor-κB pathways. These macrophages constitute a diverse population with significant plasticity, exhibiting different phenotypes and functions on the basis of their origin, tissue residence, and environmental factors. During lung homeostasis, they are involved in the clearance of inhaled particles, cellular remnants, and even participate in metabolic processes. In disease states, lung macrophages transition from the inflammatory M1 phenotype to the anti-inflammatory M2 phenotype. These distinct phenotypes have varying transcriptional profiles and serve different functions, from combating pathogens to repairing inflammation-induced damage. However, macrophages can also exacerbate lung injury during prolonged inflammation or exposure to antigens. In this review, we delve into the diverse roles of pulmonary macrophages the realms in homeostasis, pneumonia, tuberculosis, and lung tumors.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
- Graduate Collaborative Training Base of Zhuzhou Central Hospital, Hengyang Medical School, University of South China, Zhuzhou, China
| | - Yangjing Ou
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Xiaojin He
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Ting Yuan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Miao Li
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yunzhu Long
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| | - Yingzheng Tan
- Department of Infectious Diseases, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, China
| |
Collapse
|
19
|
Salauddin M, Bhattacharyya D, Samanta I, Saha S, Xue M, Hossain MG, Zheng C. Role of TLRs as signaling cascades to combat infectious diseases: a review. Cell Mol Life Sci 2025; 82:122. [PMID: 40105962 PMCID: PMC11923325 DOI: 10.1007/s00018-025-05631-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Investigating innate immunity and its signaling transduction is essential to understand inflammation and host defence mechanisms. Toll-like receptors (TLRs), an evolutionarily ancient group of pattern recognition receptors, are crucial for detecting microbial components and initiating immune responses. This review summarizes the mechanisms and outcomes of TLR-mediated signaling, focusing on motifs shared with other immunological pathways, which enhances our understanding of the innate immune system. TLRs recognize molecular patterns in microbial invaders, activate innate immunity and promote antigen-specific adaptive immunity, and each of them triggers unique downstream signaling patterns. Recent advances have highlighted the importance of supramolecular organizing centers (SMOCs) in TLR signaling, ensuring precise cellular responses and pathogen detection. Furthermore, this review illuminates how TLR pathways coordinate metabolism and gene regulation, contributing to adaptive immunity and providing novel insights for next-generation therapeutic strategies. Ongoing studies hold promise for novel treatments against infectious diseases, autoimmune conditions, and cancers.
Collapse
Affiliation(s)
- Md Salauddin
- Department of Microbiology and Public Health, Faculty of Veterinary, Animal and Biomedical Sciences, Khulna Agricultural University, Khulna, 9202, Bangladesh
| | - Debaraj Bhattacharyya
- Department of Veterinary Biochemistry, West Bengal University of Animal and Fishery Sciences, 37, K.B. Sarani, Kolkata, West Bengal, 700037, India
| | - Indranil Samanta
- Department of Veterinary Microbiology, West Bengal University of Animal and Fishery Sciences, 37, K.B. Sarani, Kolkata, West Bengal, 700037, India
| | - Sukumar Saha
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China.
| | - Md Golzar Hossain
- Department of Microbiology and Hygiene, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh.
| | - Chunfu Zheng
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
20
|
Chen Z, Behrendt R, Wild L, Schlee M, Bode C. Cytosolic nucleic acid sensing as driver of critical illness: mechanisms and advances in therapy. Signal Transduct Target Ther 2025; 10:90. [PMID: 40102400 PMCID: PMC11920230 DOI: 10.1038/s41392-025-02174-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 01/14/2025] [Accepted: 02/11/2025] [Indexed: 03/20/2025] Open
Abstract
Nucleic acids from both self- and non-self-sources act as vital danger signals that trigger immune responses. Critical illnesses such as acute respiratory distress syndrome, sepsis, trauma and ischemia lead to the aberrant cytosolic accumulation and massive release of nucleic acids that are detected by antiviral innate immune receptors in the endosome or cytosol. Activation of receptors for deoxyribonucleic acids and ribonucleic acids triggers inflammation, a major contributor to morbidity and mortality in critically ill patients. In the past decade, there has been growing recognition of the therapeutic potential of targeting nucleic acid sensing in critical care. This review summarizes current knowledge of nucleic acid sensing in acute respiratory distress syndrome, sepsis, trauma and ischemia. Given the extensive research on nucleic acid sensing in common pathological conditions like cancer, autoimmune disorders, metabolic disorders and aging, we provide a comprehensive summary of nucleic acid sensing beyond critical illness to offer insights that may inform its role in critical conditions. Additionally, we discuss potential therapeutic strategies that specifically target nucleic acid sensing. By examining nucleic acid sources, sensor activation and function, as well as the impact of regulating these pathways across various acute diseases, we highlight the driving role of nucleic acid sensing in critical illness.
Collapse
Affiliation(s)
- Zhaorong Chen
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Rayk Behrendt
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Lennart Wild
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany
| | - Martin Schlee
- Institute of Clinical Chemistry and Clinical Pharmacology, University Hospital Bonn, 53127, Bonn, Germany
| | - Christian Bode
- Department of Anesthesiology and Intensive Care Medicine, University Hospital Bonn, 53127, Bonn, Germany.
| |
Collapse
|
21
|
Luo R, Yao Y, Chen Z, Sun X. An examination of the LPS-TLR4 immune response through the analysis of molecular structures and protein-protein interactions. Cell Commun Signal 2025; 23:142. [PMID: 40102851 PMCID: PMC11921546 DOI: 10.1186/s12964-025-02149-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/10/2025] [Indexed: 03/20/2025] Open
Abstract
The LPS-TLR4 immune response is a critical mechanism in the body's defense against Gram-negative bacterial infections, yet its dysregulation can lead to severe inflammatory diseases. Lipopolysaccharide (LPS), a pivotal pathogen-associated molecular pattern (PAMP) on the surface of gram-negative bacteria, is recognized by Toll-like receptor 4 (TLR4), initiating a complex cascade of immune responses. This review delves into the intricate molecular structures and protein-protein interactions that underpin the LPS-TLR4 signaling pathway, offering a comprehensive analysis of both extracellular recognition and intracellular signal transduction. We explore the roles of key molecules such as LBP, CD14, MD-2, and TLR4 in the initial recognition of LPS, followed by the downstream signaling pathways mediated by MyD88-dependent and MyD88-independent mechanisms. The MyD88-dependent pathway primarily activates NF-κB and AP-1, leading to macrophage M1 polarization and the release of pro-inflammatory cytokines, while the MyD88-independent pathway triggers IRF activation and type-I interferon production. By elucidating the structural basis and functional interactions of these signaling molecules, this review not only enhances our understanding of the LPS-TLR4 immune response but also highlights its implications in both infectious and non-infectious diseases. Our findings underscore the potential of targeting this pathway for therapeutic interventions, offering new avenues for the treatment of inflammatory and immune-related disorders.
Collapse
Affiliation(s)
- Ruiqin Luo
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Yuexin Yao
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China
| | - Zhuo Chen
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China.
| | - Xiaoming Sun
- School of Basic Medical Sciences, Hubei University of Medicine, Shiyan, 442000, China.
- Biomedical Research Institute, Hubei University of Medicine, Shiyan, 442000, China.
- Hubei Key Laboratory of Embryonic Stem Cell Research, Hubei University of Medicine, Shiyan, 442000, China.
| |
Collapse
|
22
|
Hu ZQ, Ma R, Sun JQ, Peng M, Yuan J, Lai N, Liu J, Xia D. Tenascin-C Facilitates Microglial Polarization via TLR4/MyD88/NF-κB Pathway Following Subarachnoid Hemorrhage. J Inflamm Res 2025; 18:3555-3570. [PMID: 40093948 PMCID: PMC11908393 DOI: 10.2147/jir.s511378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Purpose This study primarily aims to elucidate the underlying mechanism of Tenascin-C in neuroinflammation and microglia polarization in a mouse model of subarachnoid hemorrhage (SAH). Methods The subarachnoid hemorrhage model was constructed by injecting blood into the anterior chiasmatic cistern and stimulating primary microglia with hemoglobin in vitro. Then, Imatinib mesylate was used to inhibit Tenascin-C. Through neurological function scoring, brain edema, primary cell extraction, immunofluorescence staining, CCK8, Tunel staining, Elisa, Western blot and other methods, the potential mechanism of Tenascin-C induced microglia cell polarization was explored. Results The results of this study observed that the expression of Tenascin-C was up-regulated after subarachnoid hemorrhage. Inhibiting the increase of Tenascin-C by imatinib could significantly ameliorate neuroinflammation, neuronal apoptosis, blood brain barrier disruption and brain edema. When the level of Tenascin-C decreased, the numbers of TLR4 positive, MyD88 positive and NF-κB positive microglial cells decreased accordingly. Moreover, after subarachnoid hemorrhage, the number of microglial cells positive for M1-type markers increased significantly. After imatinib inhibited Tenascin-C, the number of M1-type microglial cells decreased and the number of M2-type microglial cells increased significantly. Conclusion In summary, the elevated level of Tenascin-C after subarachnoid hemorrhage induces the activation of microglia, releasing a large number of inflammatory factors and aggravating early brain injury.
Collapse
Affiliation(s)
- Zheng-Qing Hu
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241001, People's Republic of China
| | - Ruijie Ma
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241001, People's Republic of China
| | - Jia-Qing Sun
- Deparment of Neurosurgery, Nanjing DrumTower Hospital Clinical College of Xuzhou Medical University, Nanjing, 210008, People's Republic of China
| | - Min Peng
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241001, People's Republic of China
| | - Jinlong Yuan
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241001, People's Republic of China
| | - Niansheng Lai
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241001, People's Republic of China
| | - Jiaqiang Liu
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241001, People's Republic of China
| | - Dayong Xia
- The Translational Research Institute for Neurological Disorders of Wannan Medical College, Department of Neurosurgery, the First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241001, People's Republic of China
| |
Collapse
|
23
|
Lim PN, Cervantes MM, Pham LK, Doherty SR, Tufts A, Dubey D, Mai D, Aderem A, Diercks AH, Rothchild AC. Absence of c-Maf and IL-10 enables type I IFN enhancement of innate responses to LPS in alveolar macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkae029. [PMID: 40073087 PMCID: PMC11952875 DOI: 10.1093/jimmun/vkae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 11/19/2024] [Indexed: 03/14/2025]
Abstract
Alveolar macrophages (AMs) are lung-resident myeloid cells and airway sentinels for inhaled pathogens and environmental particles. While AMs can be highly inflammatory in response to respiratory viruses, they do not mount proinflammatory responses to all airborne pathogens. For example, we previously showed that AMs fail to mount a robust proinflammatory response to Mycobacterium tuberculosis. Here, we address this discrepancy by investigating the capacity of murine AMs for direct innate immune sensing, using LPS as a model. Use of LPS-coated fluorescent beads enabled us to distinguish between directly exposed and bystander cells to measure transcriptional responses, by RNA-sequencing after cell sorting, and cytokine responses, by flow cytometry. We find that AMs have decreased proinflammatory responses to low-dose LPS compared to other macrophage types (bone marrow-derived macrophages, peritoneal macrophages), as measured by TNF, IL-6, Ifnb, and Ifit3. The reduced response to low-dose LPS correlates with minimal TLR4 and CD14 surface expression, despite sufficient internal expression of TLR4. We also find that AMs do not produce IL-10 in response to a variety of stimuli due to low expression of the transcription factor c-Maf, while exogenous c-Maf expression restores IL-10 production in AMs. Lastly, we show that lack of IL-10 enables type I IFN enhancement of AM responses to LPS. Overall, we demonstrate AMs have a cell-intrinsic hyporesponsiveness to LPS, which makes them uniquely tolerant to low-dose exposure. Regulation of AM innate responses by distinct CD14, c-Maf, and IL-10 expression patterns has important implications for both respiratory infections and environmental airborne exposures.
Collapse
Affiliation(s)
- Pamelia N Lim
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| | - Maritza M Cervantes
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Linh K Pham
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Animal Biotechnology & Biomedical Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Sydney R Doherty
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Ankita Tufts
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| | - Divya Dubey
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
- Graduate Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA, United States
| | - Dat Mai
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alan Aderem
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alan H Diercks
- Center for Global Infectious Disease Research, Seattle Children’s Research Institute, Seattle, WA, United States
| | - Alissa C Rothchild
- Department of Veterinary and Animal Sciences, University of Massachusetts Amherst, Amherst, MA, United States
| |
Collapse
|
24
|
Gomez-Diaz C, Greulich W, Wefers B, Wang M, Bolsega S, Effern M, Varga DP, Han Z, Chen M, Bérouti M, Leonardi N, Schillinger U, Holzmann B, Liesz A, Roers A, Hölzel M, Basic M, Wurst W, Hornung V. RNase T2 restricts TLR13-mediated autoinflammation in vivo. J Exp Med 2025; 222:e20241424. [PMID: 39853306 PMCID: PMC11758920 DOI: 10.1084/jem.20241424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/18/2024] [Accepted: 12/20/2024] [Indexed: 01/26/2025] Open
Abstract
RNA-sensing TLRs are strategically positioned in the endolysosome to detect incoming nonself RNA. RNase T2 plays a critical role in processing long, structured RNA into short oligoribonucleotides that engage TLR7 or TLR8. In addition to its positive regulatory role, RNase T2 also restricts RNA recognition through unknown mechanisms, as patients deficient in RNase T2 suffer from neuroinflammation. Consistent with this, mice lacking RNase T2 exhibit interferon-dependent neuroinflammation, impaired hematopoiesis, and splenomegaly. However, the mechanism by which RNase T2 deficiency unleashes inflammation in vivo remains unknown. Here, we report that the inflammatory phenotype found in Rnaset2-/- mice is completely reversed in the absence of TLR13, suggesting aberrant accumulation of an RNA ligand for this receptor. Interestingly, this TLR13-driven inflammatory phenotype is also fully present in germ-free mice, suggesting a role for RNase T2 in limiting erroneous TLR13 activation by an as yet unidentified endogenous ligand. These results establish TLR13 as a potential self-sensor that is kept in check by RNase T2.
Collapse
Affiliation(s)
- Carlos Gomez-Diaz
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Wilhelm Greulich
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Benedikt Wefers
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- TUM School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen Site Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Meiyue Wang
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Maike Effern
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Daniel P. Varga
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Zhe Han
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Minyi Chen
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Marleen Bérouti
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Natascia Leonardi
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Ulrike Schillinger
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Bernhard Holzmann
- Department of Surgery, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Arthur Liesz
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
- Institute for Stroke and Dementia Research, University Hospital, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Axel Roers
- Institute for Immunology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Hölzel
- Institute of Experimental Oncology, Medical Faculty, University Hospital Bonn, Bonn, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Hannover, Germany
| | - Wolfgang Wurst
- Institute of Developmental Genetics, Helmholtz Zentrum München, Neuherberg, Germany
- TUM School of Life Sciences, Technische Universität München, Freising-Weihenstephan, Germany
- Deutsches Zentrum für Neurodegenerative Erkrankungen Site Munich, Munich, Germany
| | - Veit Hornung
- Gene Center and Department of Biochemistry, Ludwig-Maximilians-Universität München, Munich, Germany
| |
Collapse
|
25
|
Sato R, Liu K, Shibata T, Hoshino K, Yamaguchi K, Miyazaki T, Hiranuma R, Fukui R, Motoi Y, Fukuda-Ohta Y, Zhang Y, Reuter T, Ishida Y, Kondo T, Chiba T, Asahara H, Taoka M, Yamauchi Y, Isobe T, Kaisho T, Furukawa Y, Latz E, Nakatani K, Izumi Y, Nie Y, Taniguchi H, Miyake K. RNase T2 deficiency promotes TLR13-dependent replenishment of tissue-protective Kupffer cells. J Exp Med 2025; 222:e20230647. [PMID: 39853307 PMCID: PMC11758922 DOI: 10.1084/jem.20230647] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 08/18/2024] [Accepted: 12/04/2024] [Indexed: 01/26/2025] Open
Abstract
Lysosomal stress due to the accumulation of nucleic acids (NAs) activates endosomal TLRs in macrophages. Here, we show that lysosomal RNA stress, caused by the lack of RNase T2, induces macrophage accumulation in multiple organs such as the spleen and liver through TLR13 activation by microbiota-derived ribosomal RNAs. TLR13 triggered emergency myelopoiesis, increasing the number of myeloid progenitors in the bone marrow and spleen. Splenic macrophages continued to proliferate and mature into macrophages expressing the anti-inflammatory cytokine IL-10. In the liver, TLR13 activated monocytes/macrophages to proliferate and mature into monocyte-derived KCs (moKCs), in which, the liver X receptor (LXR) was activated. In accumulated moKCs, tissue clearance genes such as MerTK, AXL, and apoptosis inhibitor of macrophage (AIM) were highly expressed, while TLR-dependent production of proinflammatory cytokines was impaired. Consequently, Rnaset2-/- mice were resistant to acute liver injuries elicited by acetaminophen (APAP) and LPS with D-galactosamine. These findings suggest that TLR13 activated by lysosomal RNA stress promotes the replenishment of tissue-protective Kupffer cells.
Collapse
Affiliation(s)
- Ryota Sato
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Kaiwen Liu
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Takuma Shibata
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Katsuaki Hoshino
- Department of Immunology, Faculty of Medicine, Kagawa University, Miki, Japan
- Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Japan
| | - Kiyoshi Yamaguchi
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | | | - Ryosuke Hiranuma
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Ryutaro Fukui
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Yuji Motoi
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Yuri Fukuda-Ohta
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera, Japan
| | - Yun Zhang
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Tatjana Reuter
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Yuko Ishida
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Japan
| | - Toshikazu Kondo
- Department of Forensic Medicine, Wakayama Medical University, Kimiidera, Japan
| | - Tomoki Chiba
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Japan
| | - Hiroshi Asahara
- Department of Systems Biomedicine, Tokyo Medical and Dental University, Bunkyo-ku, Japan
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Masato Taoka
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Yoshio Yamauchi
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Toshiaki Isobe
- Department of Chemistry, Graduate School of Science, Tokyo Metropolitan University, Tokyo, Japan
| | - Tsuneyasu Kaisho
- Laboratory for Inflammatory Regulation, RIKEN Center for Integrative Medical Science (IMS-RCAI), Yokohama, Japan
- Department of Immunology, Institute of Advanced Medicine, Wakayama Medical University, Kimiidera, Japan
| | - Yoichi Furukawa
- Division of Clinical Genome Research, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn, Germany
- Deutsches Rheuma Forschungszentrum Berlin (DRFZ), Berlin, Germany
| | - Kohta Nakatani
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Japan
| | - Yoshihiro Izumi
- Division of Metabolomics, Medical Institute of Bioregulation, Kyushu University, Higashi-ku, Japan
| | - Yunzhong Nie
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Hideki Taniguchi
- Division of Regenerative Medicine, Center for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| | - Kensuke Miyake
- Division of Innate Immunity, The Institute of Medical Science, The University of Tokyo, Minato-ku, Japan
| |
Collapse
|
26
|
Yu H, Ren K, Jin Y, Zhang L, Liu H, Huang Z, Zhang Z, Chen X, Yang Y, Wei Z. Mitochondrial DAMPs: Key mediators in neuroinflammation and neurodegenerative disease pathogenesis. Neuropharmacology 2025; 264:110217. [PMID: 39557152 DOI: 10.1016/j.neuropharm.2024.110217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are increasingly linked to mitochondrial dysfunction and neuroinflammation. Central to this link are mitochondrial damage-associated molecular patterns (mtDAMPs), including mitochondrial DNA, ATP, and reactive oxygen species, released during mitochondrial stress or damage. These mtDAMPs activate inflammatory pathways, such as the NLRP3 inflammasome and cGAS-STING, contributing to the progression of neurodegenerative diseases. This review delves into the mechanisms by which mtDAMPs drive neuroinflammation and discusses potential therapeutic strategies targeting these pathways to mitigate neurodegeneration. Additionally, it explores the cross-talk between mitochondria and the immune system, highlighting the complex interplay that exacerbates neuronal damage. Understanding the role of mtDAMPs could pave the way for novel treatments aimed at modulating neuroinflammation and slowing disease progression, ultimately improving patient outcome.
Collapse
Affiliation(s)
- Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Ziheng Zhang
- College of Life Sciences, Xinjiang University, Urumqi, Xinjiang, 830046, PR China
| | - Xing Chen
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
27
|
Zou Y, Tian L, Pei L, Hao J, Chen T, Qi J, Qiu J, Xu Y, Hu X, Chen L, Dou X. SFAs facilitates ceramide's de novo synthesis via TLR4 and intensifies hepatocyte lipotoxicity. Int Immunopharmacol 2025; 147:114020. [PMID: 39793229 DOI: 10.1016/j.intimp.2025.114020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Non-alcoholic steatohepatitis (NASH), an advanced manifestation of non-alcoholic fatty liver disease (NAFLD), is characterized by hepatocyte injury, inflammation, and fibrosis. Saturated fatty acids (SFAs) have emerged as key contributors to hepatocyte lipotoxicity and disease progression. Toll-like receptor 4 (TLR4) acts as a sentinel for diverse ligands, including lipopolysaccharide (LPS) and endogenous molecules like palmitic acid (PA)-induced ceramide (CER) accumulation, promoting hepatocyte demise. However, the intricate mechanisms underlying TLR4's modulation of ceramide metabolism and their concerted effect on SFA-mediated hepatotoxicity remain elusive. METHODS A NASH mouse model with liver-specific TLR4 knockdown was established through palm oil feeding and AAV2/8 tail vein injection. Histological and biochemical assessments were conducted to evaluate the mice's condition and liver damage extent. Liquid chromatography-mass spectrometry (LC-MS) was employed to quantify ceramide levels in liver tissues, offering insights into NASH mechanisms. RESULTS The PO-fed model exhibited elevated serum ALT, AST, and liver TG levels, enhancing lipid accumulation and hepatocellular damage. TLR4 knock-down reduced liver mass and the liver-to-body weight ratio, signifying a decreased hepatic burden. Histopathological evaluations revealed substantial improvement in hepatic steatosis in TLR4-silenced PO-fed mice, with diminished lipid droplets and inflammatory infiltrates. LC-MS analysis showed a marked decrease in long-chain ceramides (C14, C16, C20) in TLR4-knockdown PO-fed mice. Furthermore, expression of MyD88, SPTLC1, SPTLC2, and inflammatory markers IL-1β, IL-6, TNF-α were significantly attenuated. CONCLUSION SFAs activate the TLR4 signaling pathway via MyD88, fostering ceramide de novo synthesis, which exacerbates hepatocyte lipotoxicity and accelerates NASH progression.
Collapse
Affiliation(s)
- Yuchao Zou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Lulu Tian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Liuhua Pei
- School of Public Health, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Jie Hao
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Tianhang Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Jiayu Qi
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Jiannan Qiu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Yinuo Xu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Xiaokai Hu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China
| | - Lin Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310053, PR China.
| |
Collapse
|
28
|
Kirby CS, Islam N, Wier E, Alphonse MP, Sweren E, Wang G, Liu H, Kim D, Li A, Lee SS, Overmiller AM, Xue Y, Reddy S, Archer NK, Miller LS, Yu J, Huang W, Jones JW, Kim S, Kane MA, Silverman RH, Garza LA. RNase L represses hair follicle regeneration through altered innate immune signaling. J Clin Invest 2025; 135:e172595. [PMID: 39903537 PMCID: PMC11910212 DOI: 10.1172/jci172595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Accepted: 01/24/2025] [Indexed: 02/06/2025] Open
Abstract
Mammalian injury responses are predominantly characterized by fibrosis and scarring rather than functional regeneration. This limited regenerative capacity in mammals could reflect a loss of proregeneration programs or active suppression by genes functioning akin to tumor suppressors. To uncover programs governing regeneration in mammals, we screened transcripts in human participants following laser rejuvenation treatment and compared them with mice with enhanced wound-induced hair neogenesis (WIHN), a rare example of mammalian organogenesis. We found that Rnasel-/- mice exhibit an increased regenerative capacity, with elevated WIHN through enhanced IL-36α. Consistent with RNase L's known role to stimulate caspase-1, we found that pharmacologic inhibition of caspases promoted regeneration in an IL-36-dependent manner in multiple epithelial tissues. We identified a negative feedback loop, where RNase L-activated caspase-1 restrains the proregenerative dsRNA-TLR3 signaling cascade through the cleavage of toll-like adaptor protein TRIF. Through integrated single-cell RNA-seq and spatial transcriptomic profiling, we confirmed OAS & IL-36 genes to be highly expressed at the site of wounding and elevated in Rnasel-/- mouse wounds. This work suggests that RNase L functions as a regeneration repressor gene, in a functional trade off that tempers immune hyperactivation during viral infection at the cost of inhibiting regeneration.
Collapse
Affiliation(s)
- Charles S. Kirby
- Department of Dermatology and
- Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Nasif Islam
- Department of Dermatology and
- Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | | | | | | | - Dongwon Kim
- Department of Dermatology and
- Department of Biochemical Engineering, College of Science and Technology, Dongseo University, Busan, South Korea
| | - Ang Li
- Department of Dermatology and
| | | | - Andrew M. Overmiller
- Laboratory of Skin Biology, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | | | - Sashank Reddy
- Department of Plastic Surgery, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | | | | | - Jianshi Yu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Weiliang Huang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Jace W. Jones
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Sooah Kim
- Department of Dermatology and
- Department of Environment Science and Biotechnology, College of Medical Science, Jeonju University, Jeonju, South Korea
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland, Baltimore, Maryland, USA
| | - Robert H. Silverman
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Luis A. Garza
- Department of Dermatology and
- Cell Biology, Johns Hopkins School of Medicine, Baltimore, Maryland, USA
- Department of Oncology, Johns Hopkins University, Baltimore, Maryland, USA
| |
Collapse
|
29
|
Khan S, Simsek R, Fuentes JDB, Vohra I, Vohra S. Implication of Toll-Like Receptors in growth and management of health and diseases: Special focus as a promising druggable target to Prostate Cancer. Biochim Biophys Acta Rev Cancer 2025; 1880:189229. [PMID: 39608622 DOI: 10.1016/j.bbcan.2024.189229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 11/18/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
Toll-like receptors (TLRs) are protein structures belonging to the pattern recognition receptors family. TLRs have the great potential that can directly recognize the specific molecular structures on the surface of pathogens, damaged senescent cells and apoptotic host cells. Available evidence suggests that TLRs have crucial roles in maintaining tissue homeostasis through control of the inflammatory and tissue repair responses during injury. TLRs are the player of first line of defense against different microbes and activate the signaling cascades which help to induce the immune system and inflammatory responses by affecting various signaling pathways, including nuclear factor-κB (NF-κB), interferon regulatory factors, and mitogen-activated protein kinases (MAPKs). TLRs have been identified to be over-expressed in different types of cancers and play an important role in control of health and management of diseases. The current review provides updated knowledge on the implication of TLRs in growth and management of cancers including prostate cancer.
Collapse
Affiliation(s)
- Shahanavaj Khan
- Department of Medical Lab Technology, Indian Institute of Health Technology (IIHT), Paramedical and Nursing College, Deoband, 247554 Saharanpur, India; Department of Health Sciences, Novel Global Community Educational Foundation, Australia.
| | - Rahime Simsek
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Hacettepe Unversity, 06100 Ankara, Turkey
| | - Javier David Benitez Fuentes
- Medical Oncology Department, Hospital General Universitario de Elche, Carrer Almazara, 11, 03203 Elche, Alicante, Spain
| | - Isra Vohra
- University of Houston Clear Lake Graduated with bachelors Physiology, Houston, TX, USA
| | - Saeed Vohra
- Department of Anatomy and Physiology, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
30
|
Chen S, Yu Y, Su Y, Lian X, Jiang L, Li Z, Zhang M, Gao Y, Zhang H, Zhu X, Ke J, Chen X. Screening and identification of host signaling pathways for alleviating influenza-induced production of pro-inflammatory cytokines, IP-10, IL-8, and MCP-1, using a U937 cell-based influenza model. Front Microbiol 2025; 16:1535002. [PMID: 39931380 PMCID: PMC11808136 DOI: 10.3389/fmicb.2025.1535002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
Influenza virus infection initiates an exaggerated inflammatory response, which may culminate in a fatal cytokine storm characterized by the excessive production of pro-inflammatory cytokines. Prior research indicates that IP-10, IL-8, and MCP-1, primarily produced by monocytes and macrophages, play a crucial role in influenza-induced inflammation. The lung injury from influenza virus infection can be mitigated by suppressing or inhibiting these cytokines through knockout, knockdown, or targeted intervention approaches. To identify the key host signaling pathways responsible for producing pro-inflammatory cytokines, we utilized a U937 cell model that secretes IP-10, IL-8, and MCP-1 in response to influenza infection. This model has been previously validated in our laboratory as an appropriate system for screening anti-inflammatory agents and potential drug targets. We conducted a screening assay employing an inhibitor library consisting of 2,138 compounds that target various known pathways and host factors. Our findings indicated that inhibitors targeting protein tyrosine kinases and mitogen-activated protein kinases demonstrated superior efficacy in suppressing cytokine production induced by influenza A virus infection compared to inhibitors aimed at other host factors. Notably, a substantial proportion of the identified hits capable of inhibiting the expression of all three cytokines in the secondary screening were classified as tyrosine kinase inhibitors. Validation experiments further established that Janus kinase/signal transducers and activators of transcription (JAK/STAT) pathways, along with p38 MAPK and Raf-MEK-ERK pathways, are the principal regulators of pro-inflammatory cytokine expression in monocytes and macrophages. Moreover, our results suggest that TKIs present promising opportunities as novel therapeutic agents against influenza-induced pneumonia.
Collapse
Affiliation(s)
- Si Chen
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
- State Key Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yang Yu
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Yue Su
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Xiaoqin Lian
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Lefang Jiang
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Zhuogang Li
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Mingxin Zhang
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Yarou Gao
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Haonan Zhang
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Xingjian Zhu
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Jiaxin Ke
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| | - Xulin Chen
- Department of Immunology and Microbiology, College of Life Science and Technology, Institute of Medical Microbiology, Jinan University, Guangzhou, China
| |
Collapse
|
31
|
Zhang Y, Yue Y, Cheng Y, Jiao H, Yan M. Antigen B from Echinococcus granulosus regulates macrophage phagocytosis by controlling TLR4 endocytosis in immune thrombocytopenia. Chem Biol Interact 2025; 406:111350. [PMID: 39674446 DOI: 10.1016/j.cbi.2024.111350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 12/16/2024]
Abstract
Immune thrombocytopenia (ITP) is characterized by a reduction in platelet counts, stemming from an autoimmune-mediated process where platelets are excessively cleared by macrophages. This enhanced phagocytosis is a cardinal pathogenic mechanism in ITP. Antigen B (AgB), a principal component of the Echinococcus granulosus cyst fluid, plays a pivotal role in safeguarding the parasite from host immune defenses by modulating macrophage activation. In this study, we explored the potential of AgB to regulate macrophage activation in the context of ITP. Our observations indicated a diminished presence of M1 macrophages and a reduced phagocytic capacity in patients infected with E. granulosus sensu stricto. We isolated AgB from E. granulosus cyst fluid (EgCF) and discovered that it could suppress the polarization of M1 macrophages and weaken their phagocytic activity via Fcγ receptors, consequently alleviating thrombocytopenia in an ITP mouse model. At the molecular level, AgB was found to suppress the activation of nuclear factor kappa B (NF-κB) and interferon regulatory factor 3 (IRF3) by impeding their nuclear translocation, leading to a reduction in the generation of inflammatory cytokines. Furthermore, AgB was shown to inhibit Toll-like receptor 4 (TLR4) endocytosis and the recycling of CD14. In aggregate, our findings uncover a novel immunomodulatory mechanism of AgB, which suppresses macrophage phagocytosis by regulating TLR4 endocytosis and the subsequent activation of NF-κB and IRF3 signaling pathways. These insights shed new light on the molecular intricacies of E. granulosus-induced immune evasion and suggest that AgB may serve as a promising therapeutic agent for ITP.
Collapse
Affiliation(s)
- Yunfei Zhang
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Yingbin Yue
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Yongfeng Cheng
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Hongjie Jiao
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China
| | - Mei Yan
- Department of Pediatrics, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| |
Collapse
|
32
|
Chaudhari A, Axelsson C, Mattsson Hultén L, Rotter Sopasakis V. Toll-like Receptors 1, 3 and 7 Activate Distinct Genetic Features of NF-κB Signaling and γ-Protocadherin Expression in Human Cardiac Fibroblasts. Inflammation 2025:10.1007/s10753-025-02238-z. [PMID: 39828779 DOI: 10.1007/s10753-025-02238-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/31/2024] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Fibroblasts play a pivotal role in key processes within the heart, particularly in cardiac remodeling that follows both ischemic and non-ischemic injury. During remodeling, fibroblasts drive fibrosis and inflammation by reorganizing the extracellular matrix and modulating the immune response, including toll-like receptor (TLR) activation, to promote tissue stabilization. Building on findings from our prior research on heart tissue from patients with advanced coronary artery disease and aortic valve disease, this study sought to explore specific effects of TLR1, TLR3, and TLR7 activation on NF-κB signaling, proinflammatory cytokine production, and γ-protocadherin expression in cardiac fibroblasts. Human cardiac fibroblasts were exposed to agonists for TLR1, TLR3, or TLR7 for 24 h, followed by an analysis of NF-κB signaling, cytokine production, and γ-protocadherin expression. The activation of these TLRs triggered distinct responses in the NF-κB signaling pathway, with TLR3 showing a stronger activation profile compared to TLR1 and TLR7, particularly in downregulating γ-protocadherin expression. These findings highlight a potential role for TLR3 in amplifying inflammatory responses and reducing γ-protocadherin levels in cardiac fibroblasts, correlating with the enhanced inflammation and lower γ-protocadherin expression observed in diseased myocardium from patients with coronary artery disease and aortic valve disease. Consequently, TLR3 represents a potential therapeutic target for modulating immune responses in cardiovascular diseases.
Collapse
Affiliation(s)
- Aditi Chaudhari
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Camila Axelsson
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
| | - Lillemor Mattsson Hultén
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
- Wallenberg Laboratory, Department of Molecular and Clinical Medicine, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Victoria Rotter Sopasakis
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden.
| |
Collapse
|
33
|
Zhu YX, Li ZY, Yu ZL, Lu YT, Liu JX, Chen JR, Xie ZZ. The underlying mechanism and therapeutic potential of IFNs in viral-associated cancers. Life Sci 2025; 361:123301. [PMID: 39675548 DOI: 10.1016/j.lfs.2024.123301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 11/28/2024] [Accepted: 12/06/2024] [Indexed: 12/17/2024]
Abstract
Interferons (IFNs) are a diverse family of cytokines secreted by various cells, including immune cells, fibroblasts, and certain viral-parasitic cells. They are classified into three types and encompass 21 subtypes based on their sources and properties. The regulatory functions of IFNs closely involve cell surface receptors and several signal transduction pathways. Initially investigated for their antiviral properties, IFNs have shown promise in combating cancer-associated viruses, making them a potent therapeutic approach. Most IFNs have been identified for their role in inhibiting cancer; however, they have also demonstrated cancer-promoting effects under specific conditions. These mechanisms primarily rely on immune regulation and cytotoxic effects, significantly impacting cancer progression. Despite widespread use of IFN-based therapies in viral-related cancers, ongoing research aims to develop more effective treatments. This review synthesizes the signal transduction pathways and regulatory capabilities of IFNs, highlighting their connections with viruses, cancers, and emerging clinical treatments.
Collapse
Affiliation(s)
- Yu-Xin Zhu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Zi-Yi Li
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Zi-Lu Yu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Yu-Tong Lu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Jia-Xiang Liu
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Jian-Rui Chen
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, PR China; Queen Mary School, Medical Department, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Zhen-Zhen Xie
- School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi 330031, PR China.
| |
Collapse
|
34
|
Li W, Saeki H, Yang B, Shimizu Y, Joe GH. Enhanced anti-inflammatory effect of fish myofibrillar protein by introducing pectin oligosaccharide and its molecular mechanisms. Food Chem 2025; 463:141082. [PMID: 39276689 DOI: 10.1016/j.foodchem.2024.141082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/08/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024]
Abstract
This study investigated the efficacy of glycation with edible uronic acid-containing oligosaccharides via the Maillard reaction to enhance the anti-inflammatory effect of fish myofibrillar protein (Mf). Lyophilized Mf was reacted with pectin oligosaccharide (PO, half of the total protein weight) at 60 °C and 35 % relative humidity for up to 12 h to produce glycated Mf (Mf-PO). After pepsin and trypsin digestion, the anti-inflammatory effect was assessed by measuring the secretions of proinflammatory cytokines in LPS-stimulated RAW 264.7 macrophages, and the anti-inflammatory effect of Mf was enhanced by PO-glycation without marked lysine loss and browning. The effects on the expressions of genes related to the LPS-stimulated signaling pathway in macrophages were also examined. PO-glycation suppressed LPS-stimulated inflammation by suppressing expression of cd14 and enhancing suppressive effect of Mf on the TLR4-MyD88-dependent inflammatory signaling pathway. Therefore, as an edible reducing sugar, PO could be an effective bioindustrial material for developing anti-inflammatory Mf.
Collapse
Affiliation(s)
- Wenzhao Li
- Laboratory of Marine Food Science and Technology, Faculty of Fisheries Sciences, Hokkaido University, Minato 3, Hakodate, Hokkaido 041-8611, Japan
| | - Hiroki Saeki
- Laboratory of Marine Food Science and Technology, Faculty of Fisheries Sciences, Hokkaido University, Minato 3, Hakodate, Hokkaido 041-8611, Japan
| | - Boxue Yang
- Laboratory of Marine Food Science and Technology, Faculty of Fisheries Sciences, Hokkaido University, Minato 3, Hakodate, Hokkaido 041-8611, Japan; Liaoning Ocean and Fisheries Science Research Institute, Dalian, Liaoning 116023, PR China
| | - Yutaka Shimizu
- Laboratory of Marine Food Science and Technology, Faculty of Fisheries Sciences, Hokkaido University, Minato 3, Hakodate, Hokkaido 041-8611, Japan
| | - Ga-Hyun Joe
- Laboratory of Marine Food Science and Technology, Faculty of Fisheries Sciences, Hokkaido University, Minato 3, Hakodate, Hokkaido 041-8611, Japan.
| |
Collapse
|
35
|
Ni C, Zhou L, Yang S, Ran M, Luo J, Cheng K, Huang F, Tang X, Xie X, Qin D, Mei Q, Wang L, Xiao J, Wu J. Oxymatrine, a novel TLR2 agonist, promotes megakaryopoiesis and thrombopoiesis through the STING/NF-κB pathway. J Pharm Anal 2025; 15:101054. [PMID: 39906691 PMCID: PMC11791361 DOI: 10.1016/j.jpha.2024.101054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 02/06/2025] Open
Abstract
Radiation-induced thrombocytopenia (RIT) faces a perplexing challenge in the clinical treatment of cancer patients, and current therapeutic approaches are inadequate in the clinical settings. In this research, oxymatrine, a new molecule capable of healing RIT was screened out, and the underlying regulatory mechanism associated with magakaryocyte (MK) differentiation and thrombopoiesis was demonstrated. The capacity of oxymatrine to induce MK differentiation was verified in K-562 and Meg-01 cells in vitro. The ability to induce thrombopoiesis was subsequently demonstrated in Tg (cd41:enhanced green fluorescent protein (eGFP)) zebrafish and RIT model mice. In addition, we carried out network pharmacological prediction, drug affinity responsive target stability assay (DARTS) and cellular thermal shift assay (CETSA) analyses to explore the potential targets of oxymatrine. Moreover, the pathway underlying the effects of oxymatrine was determined by Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, Western blot (WB), and immunofluorescence. Oxymatrine markedly promoted MK differentiation and maturation in vitro. Moreover, oxymatrine induced thrombopoiesis in Tg (cd41:eGFP) zebrafish and accelerated thrombopoiesis and platelet function recovery in RIT model mice. Mechanistically, oxymatrine directly binds to toll-like receptor 2 (TLR2) and further regulates the downstream pathway stimulator of interferon genes (STING)/nuclear factor-kappaB (NF-κB), which can be blocked by C29 and C-176, which are specific inhibitors of TLR2 and STING, respectively. Taken together, we demonstrated that oxymatrine, a novel TLR2 agonist, plays a critical role in accelerating MK differentiation and thrombopoiesis via the STING/NF-κB axis, suggesting that oxymatrine is a promising candidate for RIT therapy.
Collapse
Affiliation(s)
- Chengyang Ni
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Ling Zhou
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Shuo Yang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Mei Ran
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jiesi Luo
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Kui Cheng
- Guangdong Provincial Key Laboratory of New Drug Screening and NMPA Key Laboratory for Research and Evaluation of Drug Metabolism, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Feihong Huang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiaoqin Tang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Xiang Xie
- Public Center of Experimental Technology, Model Animal and Human Disease Research of Luzhou Key Laboratory, School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Dalian Qin
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Qibing Mei
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Long Wang
- Sichuan Key Medical Laboratory of New Drug Discovery and Druggability, Luzhou Key Laboratory of Activity Screening and Druggability Evaluation for Chinese Materia Medica, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Juan Xiao
- Department of Cardiovascular Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jianming Wu
- School of Basic Medical Sciences, Southwest Medical University, Luzhou, Sichuan, 646000, China
- Education Ministry Key Laboratory of Medical Electrophysiology, Southwest Medical University, Luzhou, Sichuan, 646000, China
| |
Collapse
|
36
|
Skrtic M, Yusuf B, Patel S, Reddy EC, Ting KKY, Cybulsky MI, Freeman SA, Robinson LA. The neurorepellent SLIT2 inhibits LPS-induced proinflammatory signaling in macrophages. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:141-152. [PMID: 40073268 PMCID: PMC11844144 DOI: 10.1093/jimmun/vkae009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 10/30/2024] [Indexed: 03/14/2025]
Abstract
Macrophages are important mediators of immune responses with critical roles in the recognition and clearance of pathogens, as well as in the resolution of inflammation and wound healing. The neuronal guidance cue SLIT2 has been widely studied for its effects on immune cell functions, most notably directional cell migration. Recently, SLIT2 has been shown to directly enhance bacterial killing by macrophages, but the effects of SLIT2 on inflammatory activation of macrophages are less known. Using RNA sequencing analysis, quantitative polymerase chain reaction, and enzyme-linked immunosorbent assay, we determined that in murine bone marrow-derived macrophages challenged with the potent proinflammatory mediator lipopolysaccharide (LPS), exposure to the bioactive N-terminal fragment of SLIT2 (NSLIT2) suppressed production of proinflammatory cytokines interleukin (IL)-6 and IL-12 and concurrently increased the anti-inflammatory cytokine IL-10. We found that NSLIT2 inhibited LPS-induced MyD88- and TRIF-mediated signaling cascades and did not inhibit LPS-induced internalization of Toll-like receptor 4 (TLR4), but instead inhibited LPS-induced upregulation of macropinocytosis. Inhibition of macropinocytosis in macrophages attenuated LPS-induced production of proinflammatory IL-6 and IL-12 and concurrently enhanced anti-inflammatory IL-10. Taken together, our results indicate that SLIT2 can selectively modulate macrophage response to potent proinflammatory stimuli, such as LPS, by attenuating proinflammatory activation and simultaneously enhancing anti-inflammatory activity. Our results highlight the role of macropinocytosis in proinflammatory activation of macrophages exposed to LPS. Given that LPS-producing bacteria cause host illness through synergistic direct bacterial infection and excessive LPS-induced systemic inflammation, our work suggests a novel therapeutic role for SLIT2 in combatting the significant morbidity and mortality of patients with Gram-negative bacterial sepsis.
Collapse
Affiliation(s)
- Marko Skrtic
- Division of Nephrology, Kingston Health Sciences Centre, Queen’s University, Kingston, ON, Canada
| | - Bushra Yusuf
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Sajedabanu Patel
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Emily C Reddy
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Kenneth K Y Ting
- Department of Immunology, University of Toronto, Toronto, ON, Canada
| | - Myron I Cybulsky
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Spencer A Freeman
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Lisa A Robinson
- Program in Cell Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
37
|
Rughetti A, Bharti S, Savai R, Barmpoutsi S, Weigert A, Atre R, Siddiqi F, Sharma R, Khabiya R, Hirani N, Baig MS. Imperative role of adaptor proteins in macrophage toll-like receptor signaling pathways. Future Sci OA 2024; 10:2387961. [PMID: 39248050 PMCID: PMC11385170 DOI: 10.1080/20565623.2024.2387961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 07/30/2024] [Indexed: 09/10/2024] Open
Abstract
Macrophages are integral part of the body's defense against pathogens and serve as vital regulators of inflammation. Adaptor molecules, featuring diverse domains, intricately orchestrate the recruitment and transmission of inflammatory responses through signaling cascades. Key domains involved in macrophage polarization include Toll-like receptors (TLRs), Src Homology2 (SH2) and other small domains, alongside receptor tyrosine kinases, crucial for pathway activation. This review aims to elucidate the enigmatic role of macrophage adaptor molecules in modulating macrophage activation, emphasizing their diverse roles and potential therapeutic and investigative avenues for further exploration.
Collapse
Affiliation(s)
- Aurelia Rughetti
- Laboratory of Tumor Immunology & Cell Therapy, Department of Experimental Medicine, Policlinico Umberto I, University of Rome "Sapienza", Rome, Italy
| | - Shreya Bharti
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rajkumar Savai
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, D-35390, Germany
- Max Planck Institute for Heart & Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, D-61231, Germany
- Institute of Biochemistry, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, D-60590, Germany
| | - Spyridoula Barmpoutsi
- Lung Microenvironmental Niche in Cancerogenesis, Institute for Lung Health (ILH), Justus Liebig University, Giessen, D-35390, Germany
- Max Planck Institute for Heart & Lung Research, Member of the German Center for Lung Research (DZL), Member of the Cardio-Pulmonary Institute (CPI), Bad Nauheim, D-61231, Germany
| | - Andreas Weigert
- Institute of Biochemistry, Faculty of Medicine, Goethe University Frankfurt, Frankfurt, D-60590, Germany
- Frankfurt Cancer Institute (FCI), Goethe University Frankfurt, Frankfurt, D-60323, Germany
| | - Rajat Atre
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Faaiza Siddiqi
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rahul Sharma
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Rakhi Khabiya
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| | - Nik Hirani
- MRC Centre for Inflammation Research, Queen's Medical Research Institute, University of Edinburgh, Edinburgh, EH164TJ, UK
| | - Mirza S Baig
- Department of Biosciences & Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Indore, India
| |
Collapse
|
38
|
Chung KY, Kim S, Yoon HT, Kwon SH, Park HS, Im JP, Kim JS, Kim JW, Han YM, Koh SJ. Toll-like receptor 3 signaling attenuated colitis-associated cancer development in mice. Sci Rep 2024; 14:30308. [PMID: 39639064 PMCID: PMC11621332 DOI: 10.1038/s41598-024-76954-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 10/18/2024] [Indexed: 12/07/2024] Open
Abstract
Inflammatory bowel disease is associated with a high risk of colitis-associated cancer (CAC). We evaluated the role of TLR3 in CAC using a murine model. Wild-type (WT) and TLR3-knockout (TLR3-/-) mice received azoxymethane (AOM) 12.5 mg/kg intraperitoneally on day zero, followed by three cycles of 2% dextran sulfate sodium (DSS) for five days and free water for two weeks. We evaluated clinical indices, such as weight change, colon length, histological severity of colitis, and tumor number. We performed immunofluorescence assays for phospho-IκB kinase and β-catenin in colon tissues. To elucidate the antitumorigenic mechanism of TLR3 signaling, we injected poly(I: C) or phosphate-buffered saline intraperitoneally into an AOM/DSS-induced tumorigenesis model in WT mice. We also evaluate the direct antitumor effect of TLR signaling in AOM-treated WT and TLR3-/- mice without DSS. TLR3 deficiency increased tumor burden and colitis severity in the colon tissue than in the WT mice. β-catenin immunoreactivity was higher in TLR3-/- mice, while phospho-IκB kinase expression was similar. TLR3 activation by poly(I: C) did not reduce tumor burden in WT mice, but long-term AOM administration without DSS significantly increased tumor burden in TLR3-/- mice. TLR3 signaling attenuates CAC development, suggesting it may be a target for preventing CAC in inflammatory bowel disease.
Collapse
Affiliation(s)
- Kee Young Chung
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea
- Laboratory of Intestinal Mucosa and Skin Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Seulji Kim
- Division of Gastroenterology, Department of Internal Medicine, Korea Cancer Center Hospital, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
- Laboratory of Intestinal Mucosa and Skin Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Hee Tae Yoon
- Laboratory of Intestinal Mucosa and Skin Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - So Hyun Kwon
- Laboratory of Intestinal Mucosa and Skin Immunology, Seoul National University College of Medicine, Seoul, Korea
| | - Hyun Sun Park
- Laboratory of Intestinal Mucosa and Skin Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Dermatology, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Jong Pil Im
- Division of Gastroenterology, Department of Internal medicine, Seoul National University Hospital, Seoul, Korea
| | - Joo Sung Kim
- Division of Gastroenterology, Department of Internal medicine, Seoul National University Hospital, Seoul, Korea
| | - Ji Won Kim
- Division of Gastroenterology, Department of Internal medicine, SMG-SNU Boramae Medical Center, Seoul, Korea
| | - Yoo Min Han
- Laboratory of Intestinal Mucosa and Skin Immunology, Seoul National University College of Medicine, Seoul, Korea
- Department of Internal Medicine and Healthcare Research Institute, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Korea
| | - Seong-Joon Koh
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Korea.
- Laboratory of Intestinal Mucosa and Skin Immunology, Seoul National University College of Medicine, Seoul, Korea.
- Division of Gastroenterology, Department of Internal medicine, Seoul National University Hospital, Seoul, Korea.
| |
Collapse
|
39
|
Bryant CE. Rethinking Toll-like receptor signalling. Curr Opin Immunol 2024; 91:102460. [PMID: 39288726 DOI: 10.1016/j.coi.2024.102460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/23/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024]
Abstract
Since the discovery of Toll and Toll-like receptors (TLRs) in the 90s, an extensive body of research has been performed to determine how Pattern Recognition Receptors (PRRs) recognise 'ligands' and signal. The families of PRRs now include membrane and cytosolic proteins, which broadly signal by forming large protein platforms or supramolecular organising centres (SMOCs). The concept of SMOC-driven signalling has led to the development of a set of assumptions, particularly for TLRs, based on experimental data, to explain the physiological consequences of PRR activation. Recent research suggests that at least some of these assumptions should be reconsidered, especially as many of these receptors are important therapeutic targets for drug development, so understanding the mechanisms by which they signal is critical.
Collapse
Affiliation(s)
- Clare E Bryant
- Department of Medicine, University of Cambridge, Cambridge CB2 0PY, UK; Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge CB3 0ES, UK.
| |
Collapse
|
40
|
Zhang X, Zhang Y, Wei F. Research progress on the nonstructural protein 1 (NS1) of influenza a virus. Virulence 2024; 15:2359470. [PMID: 38918890 PMCID: PMC11210920 DOI: 10.1080/21505594.2024.2359470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/19/2024] [Indexed: 06/27/2024] Open
Abstract
Influenza A virus (IAV) is the leading cause of highly contagious respiratory infections, which poses a serious threat to public health. The non-structural protein 1 (NS1) is encoded by segment 8 of IAV genome and is expressed in high levels in host cells upon IAV infection. It is the determinant of virulence and has multiple functions by targeting type Ι interferon (IFN-I) and type III interferon (IFN-III) production, disrupting cell apoptosis and autophagy in IAV-infected cells, and regulating the host fitness of influenza viruses. This review will summarize the current research on the NS1 including the structure and related biological functions of the NS1 as well as the interaction between the NS1 and host cells. It is hoped that this will provide some scientific basis for the prevention and control of the influenza virus.
Collapse
Affiliation(s)
- Xiaoyan Zhang
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| | - Yuying Zhang
- School of Biological Science and Technology, University of Jinan, Jinan, China
| | - Fanhua Wei
- College of Animal Science and Technology, Ningxia University, Yinchuan, China
| |
Collapse
|
41
|
Tian H, Ling N, Guo C, Gao M, Wang Z, Liu B, Sun Y, Chen Y, Ji C, Li W. Immunostimulatory activity of sea buckthorn polysaccharides via TLR2/4-mediated MAPK and NF-κB signaling pathways in vitro and in vivo. Int J Biol Macromol 2024; 283:137678. [PMID: 39566757 DOI: 10.1016/j.ijbiomac.2024.137678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 10/14/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
SP0.1-1, derived from Sea buckthorn (Hippophae rhamnoides L.), has been discovered to exhibit unique antioxidant activity. In this study, we investigated the immunomodulatory activity and mechanisms of SP0.1-1 on macrophage RAW 264.7 cells in vitro and immunosuppressive mice induced by cyclophosphamide in vivo. The results indicated SP0.1-1 strengthened the immune functions via promoting the proliferation of RAW264.7 cells and phagocytic activity, along with stimulating the release of NO, ROS and cytokines including TNF-α, IL-6, IL-1β and IFN-γ. Western blot and molecular docking analysis demonstrated that SP0.1-1 attached to the prime receptors TLR2 and TLR4 in RAW264.7 cells, and triggered the activation of MyD88-mediated MAPK and NF-κB signaling pathways, thereby exerting the immune response in RAW264.7 cells. However, the intervention of specific inhibitors against TLR2, TLR4, JNK, ERK, p38 and NF-κB blocked the TLR-mediated MAPK and NF-κB signaling pathways and downregulated the levels of NO and the aforementioned cytokines, thus suppressing the activation of macrophages. Therefore, it can be speculated that SP0.1-1 activated the macrophages principally via the TLR2/4-MyD88-mediated MAPK and NF-κB signaling pathways. Additionally, SP0.1-1 could protect against the cyclophosphamide-induced immunosuppression in mice, manifested by the improvement of body weight, immune organ indices, phagocytic index, and the relievement of spleen damage, along with the enhancement of cytokines TNF-α, IL-6, IFN-γ and immunoglobulin IgG and IgM. These findings will shed light on the molecular mechanism of SP0.1-1 on the immunoregulatory effect, and lay the foundation for exploiting a potential immunostimulatory agent of SP0.1-1.
Collapse
Affiliation(s)
- Haiyan Tian
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Na Ling
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| | - Chunqiu Guo
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Mingze Gao
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Zihao Wang
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Bing Liu
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Yuan Sun
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China
| | - Yin Chen
- School of Pharmacy, Zhejiang Ocean University, Zhoushan 316022, China
| | - Chenfeng Ji
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| | - Wenlan Li
- Pharmaceutical Engineering Technology Research Center, Harbin University of Commerce, Harbin 150076,China; Engineering Research Center for Natural Antitumor Drugs, Ministry of Education, Harbin University of Commerce, Harbin 150076, China.
| |
Collapse
|
42
|
He Z, Li F, Yan J, Liu M, Chen Y, Guo C. The dual role of autophagy during porcine reproductive and respiratory syndrome virus infection: A review. Int J Biol Macromol 2024; 282:136978. [PMID: 39471930 DOI: 10.1016/j.ijbiomac.2024.136978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/12/2024] [Accepted: 10/25/2024] [Indexed: 11/01/2024]
Abstract
Autophagy is a highly conserved catabolic process that transports cellular components to lysosomes for degradation and reuse. It impacts various cellular functions, including innate and adaptive immunity. It can exhibit a dual role in viral infections, either promoting or inhibiting viral replication depending on the virus and the stage of the infection cycle. Porcine reproductive and respiratory syndrome virus (PRRSV) is a significant pathogen impacting the sustainable development of the global pork industry. Recent research has shown that PRRSV has evolved specific mechanisms to facilitate or impede autophagosome maturation, thereby evading innate and adaptive immune responses. These primary mechanisms involve viral proteins that target multiple regulators of autophagosome formation, including autophagy receptors, tethering proteins, autophagy-related (ATG) genes, as well as the functional proteins of autophagosomes and late endosomes/lysosomes. Additionally, these mechanisms are related to the post-translational modification of key components, viral antigens for presentation to T lymphocytes, interferon production, and the biogenesis and function of lysosomes. This review discusses the specific mechanisms by which PRRSV targets autophagy in host defence and virus survival, summarizes the role of viral proteins in subverting the autophagic process, and examines how the host utilizes the antiviral functions of autophagy to prevent PRRSV infection.
Collapse
Affiliation(s)
- Zhan He
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Fangfang Li
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Jiecong Yan
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Min Liu
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Yongjie Chen
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China
| | - Chunhe Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, State Key Laboratory for Animal Disease Control and Prevention, Key Laboratory of Zoonosis Prevention and Control of Guangdong Province, College of Veterinary Medicine, South China Agricultural University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
43
|
Shrestha R, Johnson PM, Ghimire R, Whitley CJ, Channappanavar R. Differential TLR-ERK1/2 Activity Promotes Viral ssRNA and dsRNA Mimic-Induced Dysregulated Immunity in Macrophages. Pathogens 2024; 13:1033. [PMID: 39770293 PMCID: PMC11676137 DOI: 10.3390/pathogens13121033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 01/11/2025] Open
Abstract
RNA virus-induced excessive inflammation and impaired antiviral interferon (IFN-I) responses are associated with severe disease. This innate immune response, also referred to as "dysregulated immunity" is caused by viral single-stranded RNA (ssRNA)- and double-stranded-RNA (dsRNA)-mediated exuberant inflammation and viral protein-induced IFN antagonism. However, key host factors and the underlying mechanism driving viral RNA-mediated dysregulated immunity are poorly defined. Here, using viral ssRNA and dsRNA mimics, which activate toll-like receptor 7 (TLR7) and TLR3, respectively, we evaluated the role of viral RNAs in causing dysregulated immunity. We observed that murine bone marrow-derived macrophages (BMDMs), when stimulated with TLR3 and TLR7 agonists, induced differential inflammatory and antiviral cytokine response. TLR7 activation triggered a robust inflammatory cytokine/chemokine induction compared to TLR3 activation, whereas TLR3 stimulation induced significantly increased IFN/IFN stimulated gene (ISG) response relative to TLR7 activation. To define the mechanistic basis for dysregulated immunity, we examined cell-surface and endosomal TLR levels and downstream mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NF-kB) activation. We identified significantly higher cell-surface and endosomal TLR7 levels compared to TLR3, which were associated with early and robust MAPK (p-ERK1/2, p-P38, and p-JNK) and NF-kB activation in TLR7-stimulated macrophages. Furthermore, blocking EKR1/2 and NF-kB activity reduced TLR3/7-induced inflammatory cytokine/chemokine levels, whereas only ERK1/2 inhibition enhanced viral RNA mimic-induced IFN/ISG responses. Collectively, our results illustrate that high cell-surface and endosomal TLR7 expression and robust ERK1/2 activation drive viral ssRNA mimic-induced excessive inflammatory and reduced IFN/ISG response and blocking ERK1/2 activity would likely mitigate viral-RNA/TLR-induced dysregulated immunity.
Collapse
Affiliation(s)
- Rakshya Shrestha
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (R.S.); (P.M.J.); (R.G.); (C.J.W.)
| | - Paige Marie Johnson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (R.S.); (P.M.J.); (R.G.); (C.J.W.)
| | - Roshan Ghimire
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (R.S.); (P.M.J.); (R.G.); (C.J.W.)
| | - Cody John Whitley
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (R.S.); (P.M.J.); (R.G.); (C.J.W.)
| | - Rudragouda Channappanavar
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Oklahoma State University, Stillwater, OK 74078, USA; (R.S.); (P.M.J.); (R.G.); (C.J.W.)
- Oklahoma Center for Respiratory and Infectious Diseases, Oklahoma State University, Stillwater, OK 74078, USA
| |
Collapse
|
44
|
Holze J, Lauber F, Soler S, Kostenis E, Weindl G. Label-free biosensor assay decodes the dynamics of Toll-like receptor signaling. Nat Commun 2024; 15:9554. [PMID: 39532846 PMCID: PMC11558003 DOI: 10.1038/s41467-024-53770-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The discovery of Toll-like receptors (TLRs) represented a significant breakthrough that paved the way for the study of host-pathogen interactions in innate immunity. However, there are still major gaps in understanding TLR function, especially regarding the early dynamics of downstream TLR pathways. Here, we present a label-free optical biosensor-based assay as a method for detecting TLR activation in a native and label-free environment and defining the dynamics of TLR pathway activation. This technology is sufficiently sensitive to detect TLR signaling and readily discriminates between different TLR signaling pathways. We define pharmacological modulators of cell surface and endosomal TLRs and downstream signaling molecules and uncover TLR signaling signatures, including potential biased receptor signaling. These findings highlight that optical biosensor assays complement traditional assays that use a single endpoint and have the potential to facilitate the future design of selective drugs targeting TLRs and their downstream effector cascades.
Collapse
Affiliation(s)
- Janine Holze
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Felicitas Lauber
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany
| | - Sofía Soler
- Institute of Experimental Haematology and Transfusion Medicine, University Hospital Bonn, Bonn, Germany
| | - Evi Kostenis
- Institute for Pharmaceutical Biology, Molecular, Cellular and Pharmacobiology Section, University of Bonn, Bonn, Germany
| | - Günther Weindl
- Pharmaceutical Institute, Section Pharmacology and Toxicology, University of Bonn, Bonn, Germany.
| |
Collapse
|
45
|
Zhang QB, Wang H, Xu F, Song Y, Jiang RD, Li Q, Liu EY. TLR3 activation enhances antitumor effects of sorafenib in hepatocellular carcinoma by activating NK cell functions through ERK and NF-κB pathways. Sci Rep 2024; 14:26422. [PMID: 39488569 PMCID: PMC11531577 DOI: 10.1038/s41598-024-78316-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 10/30/2024] [Indexed: 11/04/2024] Open
Abstract
Background Sorafenib is a standard therapeutic agent for advanced hepatocellular carcinoma (HCC). However, its efficacy is moderate, as the survival of patients is prolonged for only a few months, and the response rate is low. The mechanism of low efficacy remains unclear. In this study, we investigated the effect of Toll-like receptor 3 (TLR3) on the effects of sorafenib on HCC. Methods Polyinosinic-polycytidylic acid [poly(I: C)] was used as a double-stranded RNA analog and TLR3 agonist in subsequent experiments. After orthotopic implantation of HCC tumors in BALBc nu/nu or C57BL/6 mice, survival time, tumor growth, and metastasis in the abdomen and lungs were analyzed. Flow cytometry and cytotoxicity assays were used to analyze NK cells isolated from the spleen or peripheral blood. ELISA was used to detect the expression of plasma interferon (IFN)-γ and monocyte chemoattractant protein (MCP)-1. In addition, the expression of phosphorylated-extracellular regulated kinase 1/2 (pERK1/2), phosphorylated-protein kinase B (pAKT), ERK1/2 and AKT was analyzed by Western blotting. Results Sorafenib reduced the number and activity of NK cells in tumor-bearing mice and simultaneously decreased the levels of MCP-1 and IFN-γ in the plasma. The combination of sorafenib and poly(I: C) synergistically inhibited tumor growth and metastasis in tumor xenograft mice and prolonged survival. Poly(I: C) not only exerts a direct inhibitory effect on tumor growth and metastasis by targeting the TLR3 receptor on tumor cells but also facilitates the proliferation and activation of NK cells, indirectly impeding tumor progression. Mechanistically, poly(I: C) decreased the sorafenib-induced inhibition of ERK phosphorylation and increased the phosphorylation of IκB in NK cells, thereby enhancing NK cell function. Conclusion Activation of TLR3 can enhance the antitumor effect of sorafenib on HCC. The combination of a TLR3 activator and sorafenib may be a new strategy for the treatment of HCC.
Collapse
Affiliation(s)
- Qiang-Bo Zhang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Hong Wang
- Department of Anesthesiology, Yidu Central Hospital, Weifang Medical University, Qingzhou, 262500, Shandong Province, China
| | - Fei Xu
- Department of Anesthesiology, Yidu Central Hospital, Weifang Medical University, Qingzhou, 262500, Shandong Province, China
| | - Yan Song
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Run-de Jiang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - Qi Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China
| | - En-Yu Liu
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, 250012, Shandong Province, China.
- General Surgery Qilu Hospital, Shandong University, 107 Wenhua West Road, Jinan, 250012, China.
| |
Collapse
|
46
|
Liu H, Sheng Q, Dan J, Xie X. Crosstalk and Prospects of TBK1 in Inflammation. Immunol Invest 2024; 53:1205-1233. [PMID: 39194013 DOI: 10.1080/08820139.2024.2392587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
BACKGROUND TANK-binding kinase 1 (TBK1) is a pivotal mediator of innate immunity, activated by receptors such as mitochondrial antiviral signaling protein (MAVS), stimulator of interferon genes (STING), and TIR-domain-containing adaptor inducing interferon-β (TRIF). It modulates immune responses by exerting influence on the type I interferons (IFN-Is) signaling and the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathways, Over the past few years, TBK1 multifaceted role in both immune and inflammatory responses is increasingly recognized. METHODS AND RESULTS This review aims to scrutinize how TBK1 operates within the NF-κB pathway and the interferon regulatory transcription factor 3 (IRF3)-dependent IFN-I pathways, highlighting the kinases and other molecules involved in these processes. This analysis reveals the distinctive characteristics of TBK1's involvement in these pathways. Furthermore, it has been observed that the role of TBK1 in exerting anti-inflammatory or pro-inflammatory effects is contingent upon varying pathological conditions, indicating a multifaceted role in immune regulation. DISCUSSION TBK1's evolving role in various diseases and the potential of TBK1 inhibitors as therapeutic agents are explored. Targeting TBK1 may provide new strategies for treating inflammatory disorders and autoimmune diseases associated with IFN-Is, warranting further investigation.
Collapse
Affiliation(s)
- Huan Liu
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Qihuan Sheng
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Juhua Dan
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| | - Xiaoli Xie
- Laboratory of Molecular Genetics of Aging and Tumor, Medical School, Kunming University of Science and Technology, Kunming, China
| |
Collapse
|
47
|
Yan YR, Sun YH. Genotypic diversity and immunological implications of porcine circovirus: Inspiration from PCV1 to PCV4. Microb Pathog 2024; 196:106997. [PMID: 39369754 DOI: 10.1016/j.micpath.2024.106997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 09/16/2024] [Accepted: 10/02/2024] [Indexed: 10/08/2024]
Abstract
Porcine circovirus (PCV) is a group of DNA viruses that cause diseases in pigs, with multiple genotypes ranging from PCV1 to PCV4. PCV1 is generally considered non-pathogenic, while PCV2 can cause severe immune system damage, especially associated with porcine multisystemic wasting syndrome (PMWS). PCV2 has a genetic homology of about 68 % but differs from PCV1 in antigenicity and phenotype. PCV3 and PCV4 have lower genetic homology with PCV1 and PCV2, with limited research available on their pathogenicity. During virus infection, the host's innate immune system detects PCVs through pattern recognition receptors (PRRs) like TLRs and NLRs. PCV disrupts immune pathways, including interferon and NF-κB pathways, aiding viral replication and causing immunosuppression. This review systematically compares the characteristics and pathogenicity of different genotypes of PCV and their interactions with the host's immune system, aiming to better understand the mechanisms of PCV infection and provide a theoretical basis for prevention and treatment.
Collapse
Affiliation(s)
- You-Rong Yan
- Jiangsu Agri-animal Husbandry Vocational College, No. 8 Fenghuang East Road, Hailing District, Taizhou City, Jiangsu Province, 225300, China.
| | - Ying-Hui Sun
- Shanghai Academy of Agricultural Sciences, No.2901 Beidi Road, Minhang District, Shanghai, 201106, China
| |
Collapse
|
48
|
Wei J, Ji Y, Bai Y, Cheng R, Zhang J, Hu X, Zhang C. MiR-130c-5p targets the SHVV n gene and upregulates immune cytokines (IL-6, IL-22, IL-1β) to inhibit viral replication. Front Immunol 2024; 15:1486816. [PMID: 39555085 PMCID: PMC11563963 DOI: 10.3389/fimmu.2024.1486816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/10/2024] [Indexed: 11/19/2024] Open
Abstract
Background Snakehead vesiculovirus (SHVV) has led to huge economic losses in snakehead aquaculture, and its pathogenic mechanisms is still not fully understood. MicroRNAs (miRNAs), as an important class of non-coding RNAs, play a key regulatory role in the process of viral infection. Methods We examined the effect of SHVV infection on the expression of miR-130c-5p and the effect of overexpression of miR-130c-5p on the proliferation of SHVV. Cotransfection of viral N protein and miR-130c-5p, and the effect of miR-130c-5p on the expression of N protein was detected. Meanwhile, the effect of overexpression of miR-130c-5p on the expression of various immune factors in the case of viral infection were also tested. Results In this study, SHVV infection significantly upregulated the expression of miR-130c-5p in channel catfish ovary (CCO) cells in a time- and dose-dependent manner. The further research revealed that miR-130c-5p mimic significantly inhibited, while its inhibitors promoted SHVV replication. In addition, miR-130c-5p could directly target the viral mRNA of n gene, and overexpression of miR-130c-5p could significantly decrease, and conversely, downregulation of miR-130c-5p could increase the mRNA and protein expression of the viral n gene. Meanwhile, overexpression of miR-130c-5p also upregulated the expression of immune-related genes, such as nucleotide-oligomerization domain (NOD)-like receptor subfamily C3 (NLRC3), myeloid differentiation factor 88 (MyD88), nuclear factor kappa-B (NF-κB), interleukin-6 (IL-6), interleukin-22 (IL-22), and interleukin-1beta (IL-1β) in host cells. Conclusion miR-130c-5p was upregulated in the host during SHVV infection, and the upregulated miR-130c-5p directly inhibited viral replication by targeting the n gene of SHVV and promoting viral nucleoprotein degradation. The up-regulated miR-130c-5p also activated the expression of immune-related genes such as NLRC3, MyD88, NF-κB, IL-6, IL-22, and IL-1β, which were involved in the regulation of the signaling pathways including NF-κB, MyD88, Toll-like receptor (TLR), NLR, and janus tyrosine kinase-signal converter and activator of transcription (JAK-STAT), to enhance the host's antiviral immune response, and thus indirectly inhibited the viral proliferation.
Collapse
Affiliation(s)
- Jin Wei
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Yan Ji
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Yaqian Bai
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Rui Cheng
- Key Laboratory of Ecological Impacts of Hydraulic-Projects and Restoration of Aquatic Ecosystem of Ministry of Water Resources, Institute of Hydroecology, Ministry of Water Resources and Chinese Academy of Sciences, Wuhan, China
| | - Jiaqi Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Xianqin Hu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| | - Chi Zhang
- Hubei Key Laboratory of Animal Nutrition and Feed Science, School of Animal Science and Nutritional Engineering, Wuhan Polytechnic University, Wuhan, China
| |
Collapse
|
49
|
Lee H, Park SK, Lim J. Dual Roles of Host Zinc Finger Proteins in Viral RNA Regulation: Decay or Stabilization. Int J Mol Sci 2024; 25:11138. [PMID: 39456919 PMCID: PMC11508327 DOI: 10.3390/ijms252011138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Host defense mechanisms against viral infections have been extensively studied over the past few decades and continue to be a crucial area of research in understanding human diseases caused by acute and chronic viral infections. Among various host mechanisms, recent findings have revealed that several host RNA-binding proteins play pivotal roles in regulating viral RNA to suppress viral replication and eliminate infection. We have focused on identifying host proteins that function as regulators of viral RNA, specifically targeting viral components without adversely affecting host cells. Interestingly, these proteins exhibit dual roles in either restricting viral infections or promoting viral persistence by interacting with cofactors to either degrade viral genomes or stabilize them. In this review, we discuss RNA-binding zinc finger proteins as viral RNA regulators, classified into two major types: ZCCCH-type and ZCCHC-type. By highlighting the functional diversity of these zinc finger proteins, this review provides insights into their potential as therapeutic targets for the development of novel antiviral therapies.
Collapse
Affiliation(s)
- Hyokyoung Lee
- Department of Pharmacy, School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju 54896, Republic of Korea
| | - Sung-Kyun Park
- Infectious Disease Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Functional Genomics, KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Junghyun Lim
- Department of Pharmacy, School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju 54896, Republic of Korea
| |
Collapse
|
50
|
Gibson SA, Liu Y, Li R, Hurst BL, Fan Z, Siddharthan V, Larson DP, Sheesley AY, Stewart R, Kunzler M, Polejaeva IA, Van Wettere AJ, Moisyadi S, Morrey JD, Tarbet EB, Wang Z. Differences in Susceptibility to SARS-CoV-2 Infection Among Transgenic hACE2-Hamster Founder Lines. Viruses 2024; 16:1625. [PMID: 39459957 PMCID: PMC11512293 DOI: 10.3390/v16101625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Animal models that are susceptible to SARS-CoV-2 infection and develop clinical signs like human COVID-19 are desired to understand viral pathogenesis and develop effective medical countermeasures. The golden Syrian hamster is important for the study of SARS-CoV-2 since hamsters are naturally susceptible to SARS-CoV-2. However, infected hamsters show only limited clinical disease and resolve infection quickly. In this study, we describe development of human angiotensin-converting enzyme 2 (hACE2) transgenic hamsters as a model for COVID-19. During development of the model for SARS-CoV-2, we observed that different hACE2 transgenic hamster founder lines varied in their susceptibility to SARS-CoV-2 lethal infection. The highly susceptible hACE2 founder lines F0F35 and F0M41 rapidly progress to severe infection and death within 6 days post-infection (p.i.). Clinical signs included lethargy, weight loss, dyspnea, and mortality. Lethality was observed in a viral dose-dependent manner with a lethal dose as low as 1 × 100.15 CCID50. In addition, virus shedding from highly susceptible lines was detected in oropharyngeal swabs on days 2-5 p.i., and virus titers were observed at 105.5-6.5 CCID50 in lung and brain tissue by day 4 p.i.. Histopathology revealed that infected hACE2-hamsters developed rhinitis, tracheitis, bronchointerstitial pneumonia, and encephalitis. Mortality in highly susceptible hACE2-hamsters can be attributed to neurologic disease with contributions from the accompanying respiratory disease. In contrast, virus challenge of animals from less susceptible founder lines, F0M44 and F0M51, resulted in only 0-20% mortality. To demonstrate utility of this SARS-CoV-2 infection model, we determined the protective effect of the TLR3 agonist polyinosinic-polycytidylic acid (Poly (I:C)). Prophylactic treatment with Poly (I:C) significantly improved survival in highly susceptible hACE2-hamsters. In summary, our studies demonstrate that hACE2 transgenic hamsters differ in their susceptibility to SARS-CoV-2 infection, based on the transgenic hamster founder line, and that prophylactic treatment with Poly (I:C) was protective in this COVID-19 model of highly susceptible hACE2-hamsters.
Collapse
Affiliation(s)
- Scott A. Gibson
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Yanan Liu
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| | - Rong Li
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| | - Brett L. Hurst
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Zhiqiang Fan
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| | - Venkatraman Siddharthan
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Deanna P. Larson
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| | - Ashley Y. Sheesley
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - Rebekah Stewart
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| | - Madelyn Kunzler
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| | - Irina A. Polejaeva
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| | - Arnaud J Van Wettere
- Department of Veterinary, Clinical, and Life Sciences, Utah State University, Logan, UT 84322, USA;
- Utah Veterinary Diagnostic Laboratory, Utah State University, Logan, UT 84322, USA
| | - Stefan Moisyadi
- Institute of Biogenesis Research, John A. Burns School of Medicine, University of Hawaii, Honolulu, HI 96822, USA;
| | - John D. Morrey
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
| | - E. Bart Tarbet
- Institute for Antiviral Research, Utah State University, Logan, UT 84322, USA
- Department of Veterinary, Clinical, and Life Sciences, Utah State University, Logan, UT 84322, USA;
| | - Zhongde Wang
- Department of Animal, Diary and Veterinary Sciences, Utah State University, Logan, UT 84322, USA; (S.A.G.); (Y.L.); (R.L.); (B.L.H.); (Z.F.); (V.S.); (D.P.L.); (A.Y.S.); (R.S.); (M.K.); (I.A.P.); (J.D.M.); (Z.W.)
| |
Collapse
|